251
Identification of PPP1CC2 Interacting Proteins in the Mouse Testis by George Graham MacLeod A thesis submitted in conformity with the requirements for the degree of Doctor of Philosophy Department of Cell & Systems Biology University of Toronto © Copyright by George Graham MacLeod 2013

Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

Identification of PPP1CC2 Interacting Proteins in the Mouse

Testis

by

George Graham MacLeod

A thesis submitted in conformity with the requirements

for the degree of Doctor of Philosophy

Department of Cell & Systems Biology

University of Toronto

© Copyright by George Graham MacLeod 2013

Page 2: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

ii

Identification of PPP1CC2 Interacting Proteins in the Mouse Testis

George Graham MacLeod

Doctor of Philosophy

Department of Cell & Systems Biology

University of Toronto

2013

Abstract

Protein phosphorylation is a central regulatory mechanism in countless cellular

processes. Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male

infertility due to a severe impairment in spermatogenesis. This disruption in spermatogenesis is

hypothesized to arise due to a deficiency of the testis specific Ppp1cc isoform PPP1CC2. To

learn more about the function of PPP1CC2 in spermatogenesis, we have employed several

proteomic approaches aimed at identifying both regulatory proteins and substrates that interact

with PPP1CC2 in the testis. First, we created transgenic mouse embryonic stem cell lines

expressing a tandem affinity tagged version of PPP1CC2. Tandem affinity purification using

these cell lines identified a number of known PP1 interacting proteins, and one novel interactor

DDOST (dolichyl-di-phosphooligosaccharide-protein glycotransferase) which we hypothesize

to have a role in spermatogenesis. In a second approach, we conducted GST pull down assays

from mouse testis lysates to identify PPP1CC2 interacting proteins. TSSK1 (testis-specific

serine kinase 1) was identified as a novel PPP1CC2 interacting protein. We then demonstrated

that TSSK1 interacts with PPP1CC2 in an indirect manner via a common interacting protein

TSKS (testis-specific serine kinase substrate). Binding of TSKS to PPP1CC2 is regulated via

Page 3: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

iii

phosphorylation of a PP1 docking motif on the TSKS surface, and localization of TSSK1 and

TSKS in the testis is disrupted in Ppp1cc mutants. Finally, to identify candidate substrates of

PPP1CC2 in the testis, we conducted a comparative phosphoproteomic analysis and identified

33 different peptides that were hyperphosphorylated in the testis of 3 week old Ppp1cc knockout

mice. Amongst these candidate substrates are several proteins essential for mouse

spermatogenesis—HMGA1 (high mobility group AT-hook 1), HSPA4 (heat shock protein 4),

YBX2 (Y box protein 2) and SYCP2 (synaptonemal complex protein 2). Taken together, our

results suggest that PPP1CC2 interacts with a number of different proteins in the testis, and is

likely to play a role at several different stages of spermatogenesis, in both meiotic and post-

meiotic spermatogenic cells.

Page 4: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

iv

Acknowledgments

I would like to first thank my supervisor, Dr. Sue Varmuza for her support and guidance

throughout my graduate studies. Dr. Varmuza has taught me a great deal about what it takes to be a

successful researcher, and her advice and encouragement were instrumental in the successful completion

of this work. Dr. Varmuza has always allowed me the independence to follow my own ideas, and for that

I feel I have become a much stronger researcher. I am also grateful for the advice and support of my

committee members Dr. Darrell Desveaux and Dr. Tim Westwood, who provided continual support and

helpful advice throughout the past several years; and Dr. Dinesh Christendat and Dr. Greg Moorhead for

serving on my thesis examination committee.

I would like to thank all of the current and past members of the Varmuza Lab, who have made

my stay in the lab thoroughly enjoyable. I would especially like to thank Hannah Henderson, who was

extremely helpful to me in getting started as a graduate student, and Kamelia Miri who has been an

excellent and helpful colleague throughout my time in the lab. I would also like to acknowledge the

contributions of all of the talented undergraduate students who supported my work, especially Richard

Cheng, Lucas Mastropaolo, Gregory Booth and Niloufar Manafpoursakha. In addition I would like to

thank all of the member of the Department of Cell & Systems Biology who helped me in this work,

especially Daniel Rivero for his assistance with maintaining our mouse colony, and Jacky Jhingree and

Dr. Pauline Wang for their help and advice in LC-MS/MS analysis procedures.

This work would not have been possible without the help of all of the researchers mentioned in

the coming chapters who generously provided technical advice and reagents used in my thesis research.

For this, I would particularly like to thank Peng Shang and Dr. J Anton Grootegoed for their

contributions to the study detailed in Chapter 3. I would like to emphasize my gratitude to Paul Taylor

for many hours of help and advice in the LC-MS/MS analysis of phosphopeptide samples, without which

this project would not have been possible.

Finally, I would like to thank all of my friends and family for their continuous encouragement

over the last few years. Particularly I would like to thank my parents for providing me with the

upbringing and education that made all of this possible; as well as my wife, Kristy, whose unwavering

support and patience was pivotal in allowing me to reach this goal.

Page 5: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

v

TABLE OF CONTENTS

CHAPTER 1: INTRODUCTION 1

1.1 MAMMALIAN SPERMATOGENESIS .......................................................................................................... 1

1.1.1 Organization of the testis ............................................................................................................... 1

1.1.2 Spermatogenesis ............................................................................................................................ 2

1.1.3 The cycle of the seminiferous epithelium ........................................................................................ 7

1.1.4 Mouse models of male infertility .................................................................................................. 10

1.1.5 Male factor infertility .................................................................................................................. 10

1.2PROTEIN PHOSPHATASE PPP1CC2 ........................................................................................................ 11

1.2.1 Protein phosphatases ................................................................................................................... 11

1.2.2 The PP1 family of protein phosphatases ..................................................................................... 12

1.2.3 Regulation of PP1 catalytic subunits .......................................................................................... 14

1.2.4 The Ppp1cc knockout mouse ....................................................................................................... 22

1.2.5 PPP1CC2 protein – protein interactions in the testis ................................................................. 25

1.2.6 Identification of PPP1CC2 substrates in the testis ..................................................................... 27

1.3 HYPOTHESIS AND RESEARCH OBJECTIVE ............................................................................................. 28

CHAPTER 2: TANDEM AFFINITY PURIFICATION IN TRANSGENIC MOUSE EMBRYONIC STEM

CELLS IDENTIFIES DDOST AS A NOVEL PPP1CC2 INTERACTING PROTEIN 30

2.1 INTRODUCTION .................................................................................................................................... 31

2.2 MATERIALS AND METHODS ................................................................................................................. 32

2.3 RESULTS .............................................................................................................................................. 40

2.4 DISCUSSION ......................................................................................................................................... 63

CHAPTER 3: PPP1CC2 CAN FORM A KINASE/PHOSPHATASE COMPLEX WITH THE TESTIS

SPECIFIC PROTEINS TSSK1 AND TSKS DURING MOUSE SPERMIOGENESIS 69

3.1 INTRODUCTION .................................................................................................................................... 70

3.2 MATERIALS AND METHODS ................................................................................................................ 72

3.3 RESULTS .............................................................................................................................................. 77

3.4 DISCUSSION ......................................................................................................................................... 92

CHAPTER 4: PHOSPHOPROTEOMIC ANALYSIS OF THE PPP1CC MUTANT TESTES 102

4.1 INTRODUCTION .................................................................................................................................. 103

4.2 MATERIALS AND METHODS ............................................................................................................... 105

4.3 RESULTS ............................................................................................................................................ 113

4.4 DISCUSSION ....................................................................................................................................... 137

CHAPTER 5: CONCLUSION 147

5.1 THE APPLICATION OF PROTEOMIC APPROACHES TO THE STUDY OF MAMMALIAN SPERMATOGENESIS . 147

5.2 SIGNIFICANCE AND FUTURE DIRECTIONS ........................................................................................... 154

REFERENCES 158

APPENDIX A.1: MOUSE MUTATIONS RESULTING IN MALE INFERTILITY ................................................... 174

APPENDIX A.2: YEAST 3-HYBRID SCREEN FOR SUBSTRATES OF THE SH3GLB1T-PPP1CC2 HOLOENZYME

..................................................................................................................................................................... 191

APPENDIX A.3: THE EFFECT OF PPP1CC DELETION ON THE LOCALIZATION OF CANDIDATE SUBSTRATES HSPA2

AND SH3GLB1T IN THE MOUSE SPERMATOGENIC CELL NUCLEI ............................................................ 201

APPENDIX A.4: SUPPLEMENTAL MATERIAL ............................................................................................. 210

Page 6: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

vi

List of Tables

Table 2.1: Protein-protein interactions detected via tandem affinity purification 52

Table 3.1: Testis proteins identified in an SDS-PAGE gel band after sedimentation by

GST-PPP1CC1 and GST-PPP1CC2 78

Table 3.2: Quantitative evaluation of TSKS and TSSK1 staining patterns in wild-type

and Ppp1cc mutant seminiferous tubules 95

Table 4.1: Primers used for qPCR analysis of PP1 isoform expression in the post-

natal mouse testis 107

Table 4.2: Candidate PPP1CC2 substrates 130

Table 4.3: GO biological processes and molecular functions enriched in

hyperphosphorylated testis proteins 138

Page 7: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

vii

List of Figures

Figure 1.1 The organisation of the seminiferous epithelium 3

Figure 1.2 The mouse spermatogenic cycle 8

Figure 1.3 The PP1 structure and catalytic mechanism 15

Figure 1.4 Regulation of PP1 catalytic subunits via interaction with regulatory PIPs. 19

Figure 2.1 Strategy for the generation of SBP-3XFLAG-PPP1CC knock-in

embryonic stem cells 41

Figure 2.2 Cre-mediated excision of second loxP site in pGT0lxf gene-trap line

RRR804 44

Figure 2.3 Verification of Cre-mediated insertion of transgene into RRR804-SA ES

cell genome 46

Figure 2.4 Expression of the SBP-3XFLAG-PPP1CC1/2 transgenes in knock-in ES

cells at the cDNA and protein level 49

Figure 2.5 Validation of PPP1CC2-DDOST interaction in mouse testis lysate 56

Figure 2.6 Localization of DDOST in dissociated spermatogenic cells 58

Figure 3.1 PPP1CC2 interacts with both TSSK1 and TSKS in the testis 80

Figure 3.2 The interaction between PPP1CC2 and TSSK1 is not direct 83

Figure 3.3 The TSKS RVxF motif is required for interaction with PPP1CC2 87

Figure 3.4 TSKS localization is impaired, but not abolished in Ppp1cc mutant

seminiferous tubules 90

Figure 3.5 TSSK1 localization is impaired but not abolished in Ppp1cc mutant

seminiferous tubules 93

Figure 4.1 Expression of mouse Ppp1c isoforms in the testis during the first wave of

spermatogenesis 114

Figure 4.2 Phenotypic abnormalities are present in the seminiferous tubules of 3

week old Ppp1cc mutant mice 117

Figure 4.3 Reproducible phosphopeptide enrichment and identification in wild-type

and Ppp1cc mutant testis samples 120

Page 8: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

viii

Figure 4.4 Strategy for quantitative analysis of wild-type and Ppp1cc mutant 3

week testis phosphoproteomes 124

Figure 4.5 Distribution of spectral counting and MS/MS peak XIC data for wild-

type and Ppp1cc mutant testis phosphopeptides 126

Figure 4.6 Example of Skyline MS1 filtering output 133

Figure 4.7 Quantitative comparison of XIC peak areas of hyperphosphorylated

phosphopeptides mapping to genes essential for spermatogenesis and

known/predicted PP1 interacting proteins 135

Page 9: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

ix

List of Appendices

Appendix A.1 Mouse mutations causing male infertility 174

Appendix A.2 Yeast 3-hybrid screen for substrates of the SH3GLB1T-PPP1CC2

holoenzyme 191

Appendix A.3 The effect of Ppp1cc deletion on the localization of candidate

substrates HSPA2 and β-tubulin in mouse spermatogenic cell nuclei 201

Appendix A.4 Supplemental material 210

A.4.1 Proteins identified via tandem affinity purification 210

A.4.2 Peptides identified by LC-MS/MS of ~45 kDa gel slice 214

A.4.3 TSKS phosphopeptide site assignment data 217

A.4.4 TSKS testis phosphopeptide site assignment charge

tables/annotated spectra 218

A.4.5 3 week testis phosphoproteins of interest for quantitative analysis 221

A.4.6 DAVID GO enrichment analysis of 3 week testis phosphoproteins 223

A.4.7 Peptides identified via preliminary semi-quantitative analysis as

more abundant in Ppp1cc knockout testes 235

A4.8 Quantitative comparison of XIC peak areas of Ppp1cc

hyperphosphorylated peptides assigned to indicated proteins 238

Appendix References 239

Page 10: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

x

List of Abbreviations

(E)GFP (enhanced) green fluorescent protein

2DE two-dimensional electrophoresis

AMPR ampicillin resistance gene

ANOVA analysis of variance

AP-MS affinity purification mass spectrometry

BCA bicinchoninic acid

BGH bovine growth hormone

bp base pair

cDNA complementary DNA

Cre cre-recombinase

DAPI 4' 6-diamidino-2-phenylindole

DMEM Dulbecco’s modified eagle medium

DRM detergent resistant membrane

ECMV encephalomyocarditis virus

En2 engrailed 2

ES embryonic stem

FACS fluorescence activated cell sorting

FBS fetal bovine serum

FRT Flp recombinase target

FTMS fourier transform mass spectrometry

GO gene ontology

GST Glutathione S-transferase

HA human influenza hemagglutinin

HRP horseradish peroxidase

IMAC immobilized metal ion affinity chromatography

IP immunoprecipitation

IRES internal ribosomal entry site

LC liquid chromatography

MS mass spectrometry

MS/MS tandem mass spectrometry

MS1 mass spectrometer analyzer 1

pA polyadenylation signal

PAGE polyacrylamide gel electrophoresis

PAS-H periodic acid schiff's-hematoxylin

PBS phosphate buffered saline

PCR polymerase chain reaction

PIPs PP1 interacting proteins

PMSF phenylmethanesulfonylfluoride

PP1 protein phosphatase 1

Page 11: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

xi

PP1c protein phosphatase 1 catalytic subunit

PTM post-translational modification

qPCR quantitative polymerase chain reaction

RT reverse transcriptase

SBP streptavidin binding peptide

SDS sodium dodecyl-sulphate

SIMAC sequential elution form IMAC

TAP tandem affinity purification

TFA trifluoroacetic acid

XIC extracted ion chromatogram

Page 12: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

1

Chapter 1: Introduction

1.1 Mammalian Spermatogenesis

Spermatogenesis is the process by which the male gamete, the spermatozoan, is generated. What

begins as a pluripotent germline stem cell (Guan et al. 2006) eventually differentiates into what is

perhaps the most specialized cell type known, through a process that involves the coordination between

hundreds, if not thousands, of genes. Being the cell responsible for the transmission of the paternal

genome, the spermatozoan is well characterized at the cellular level as is the spermatogenic processes.

The following sections will give a brief overview of spermatogenesis in mammals and highlight the

importance of related research to human health.

1.1.1 Organization of the Testis

The mammalian testis contains two structurally distinct compartments that are required for

spermatogenesis: the interstitium and the seminiferous tubules. The interstitial compartment is

permeated by both blood, and lymphatic vessels (Russell et al. 1990). The most frequently observed cell

type in the interstitium is the Leydig cell (Hess & de Franca. 2009). The primary function of the Leydig

cells is the production and secretion of testosterone, for which they are the principal source in the

systemic circulation of males (Ge et al. 2009). The interstitial compartment, although essential for

fertility, will not be a point of focus for this thesis. Instead, I will focus on the seminiferous tubules, the

site of spermatogenesis.

The seminiferous tubules are highly convoluted such that a cross section of the testis would

show a cross section of many individual tubules. The seminiferous tubules are bounded by myoid cells,

and a basal lamina which provide structural support for the most basal cells of the seminiferous

epithelium, and contractile forces for moving mature spermatozoa through the tubules (Russell et al.

1990). Within the seminiferous tubules, cells are arranged into an epithelial configuration, known as the

seminiferous epithelium.

Page 13: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

2

The seminiferous epithelium has a stratified organization, with the germ cells traversing the

epithelium outward towards the central lumen as they mature (Figure 1.1). In addition to the developing

germ cells, there is also a somatic cell component to the seminiferous epithelium, the Sertoli cells. These

cells, which cease to divide during pubertal development (Griswold & McLean. 2006), serve to support

the developing germ cells through spermatogenesis. Sertoli cells are attached in a single layer to the basal

lamina and span the entire width of the seminiferous epithelium. Developing germ cells are found

tightly bound between adjacent Sertoli cells, with a single Sertoli cell forming cell-cell contacts with

numerous germ cells at various developmental stages. The Sertoli cells’ extensive cell-cell contacts serve

to maintain the integrity of the epithelium, and also to compartmentalize it, via the formation of the

blood-testis barrier (Kopera et al. 2010). During spermatogenesis, the Sertoli cells have a number of

critical functions, including regulation of the spermatogenic cycle via communicating junctions

(Griswold & McLean. 2006). Sertoli cells aid in the movement of germ cells through the seminiferous

epithelium, and provide them with nutrients needed to complete spermatogenesis (Russell et al. 1990,

Kopera et al. 2010). Additional roles of the Sertoli cells include, but are not limited to participation in

spermiation, and phagocytosis of residual bodies (Russell et al. 1990).

1.1.2 Spermatogenesis

Spermatogenesis is a complex differentiation processes in which cells that begin as pluripotent

stem cells undergo a series of cell divisions, genome reorganization, and dramatic morphological change

to become spermatozoa. Spermatogenesis can be divided into three key functional phases—proliferative,

meiotic, and spermiogenesis, all of which are occurring simultaneously in the adult testis.

The proliferative phase refers to the mitotic division of the spermatogonia, the most immature

germ cells found in adults. These cells are located along the basement membrane of the seminiferous

epithelium. Spermatogonia are responsible for both providing the large number of cells required to

support the production of upwards of millions of sperm per day, and maintaining a pool of stem cells to

Page 14: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

3

Figure 1.1: The organization of the seminiferous epithelium. A diagram depicting the general

architecture of the mouse seminiferous epithelium (based on a diagram by Russell et al., 1990; not to

scale). Shown are the developing spermatogenic cells from the least mature spermatogonia, which are

localized in the basal region, to the elongate spermatids adjacent to the epithelial lumen. All stages of

spermatogenic cell development are associated with the somatic Sertoli Cells.

Page 15: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

4

Page 16: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

5

ensure that this production will endure for the sexual lifespan of the animal. There are three major types

of spermatogonia—stem cell, proliferative and differentiating, as well as several more sub-types (Russell

et al. 1990). The spermatogonial stem cells represent only an estimated 0.03% of the total germ cell

population (Tegelenbosch & de Rooij. 1993) and can undergo both self-renewal cell divisions and

differentiation cell divisions (into proliferative spermatogonia) (Huckins. 1971, Oakberg 1971, Phillips et

al. 2010). During proliferation, cytokinesis is incomplete, producing intercellular bridges between

adjacent cells, which subsequently undergo additional rounds of mitosis to produce large numbers of

spermatogonia. Eventually these cells will cease to divide and then undergo several stages of

differentiation (differentiating spermatogonia) into a number of spermatogonial subtypes; they ultimately

divide to form spermatocytes, which then enter into meiosis with a round of DNA replication (Russell et

al. 1990).

In the meiotic phase of spermatogenesis the spermatocytes undergo two successive meiotic cell

divisions to produce haploid spermatids. The most immature spermatocytes are the preleptotene cells,

which are the last spermatogenic cells to reside in the S-phase of the cell cycle, and are classified as

leptotene spermatocytes upon entry to prophase (Russell et al. 1990). In mammalian spermatogenesis,

prophase of the first meiotic division is of a particularly long duration, lasting almost two weeks in the

mouse (Hess & de Franca. 2009). Spermatocytes in prophase I go through successive morphological

changes from leptotene spermatocytes into zygotene, pachytene and finally diplotene spermatocytes.

During this time the nuclei greatly increase in size, the synaptonemal complex is formed, linking

homologous chromosomes, and genetic recombination occurs (Russell et al. 1990). After this

exceptionally long meiotic prophase, the first and second meiotic divisions are rapidly completed, with

the intermediate secondary spermatocytes only visible in the seminiferous epithelium for a brief period

(Russell et al. 1990). After the completion of the second meiotic division, the germ cells now contain a

haploid DNA complement and are known as spermatids.

Page 17: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

6

The final phase of spermatogenesis is spermiogenesis, a phase of dramatic morphological

differentiation from a round cell to the characteristic shape of a spermatozoan and is carried out in the

absence of cell division. There are four key aspects to this transformation—formation of the flagellum,

formation of the acrosome, nuclear shaping/condensation and elimination of the cytoplasm—all of which

are simultaneously occurring in the seminiferous epithelium (Russell et al. 1990). The flagellum or

“tail” of the sperm is required for forward motility via transit through the female tract, and the acrosome

is an exocytotic vesicle that surrounds the sperm head and allows for penetration of the oocyte

(Guyonnet et al. 2012). The shaping and condensation of the nucleus observed in spermatogenesis from

a rounded to its characteristic falciform shape involves factors both interior and exterior to the nucleus.

The manchette is a transient cytoplasmic tubulin structure that envelopes the nucleus during

spermiogenesis and is thought to play a mechanical role in its shaping and condensation (reviewed in

Kierszenbaum 2002). Inside the nucleus a major remodelling of the chromatin occurs, allowing for a

large reduction in size. At the beginning of spermiogenesis, the chromatin is packaged in nucleosomes

by histones as in somatic tissues. During spermiogenesis nearly all of the histones are replaced by basic

proteins known as transition proteins (TPs) and subsequently by protamines resulting in a more compact

orientation (reviewed in Lewis et al. 2003) and transcriptional silencing. The elimination of cytoplasm

to ~25% of the original volume during spermiogenesis serves to make the spermatozoan more

streamlined for motility (Russell et al. 1990). This is accomplished by several different mechanisms

including the bulk removal of a large portion of cytoplasm containing RNA and organelles, termed the

residual body, which is phagocytosed by the Sertoli cells (Russell et al. 1990).

After the completion of spermiogenesis, spermatozoa are released from the seminiferous tubules

via a process termed spermiation. However, the spermatozoa are still not competent for fertilization and

require further development in the epididymis and female tract (capacitation) before they are able to

fertilize an oocyte.

Page 18: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

7

1.1.3 The Cycle of the Seminiferous Epithelium

As mentioned above, the seminiferous epithelium has a stratified organization such that several

generations of spermatogenic cells are simultaneously present in any cross-section of the epithelium.

This organization is highly synchronous and tightly regulated, which is necessary to support the

continuous generation of spermatozoa (Hess & de Franca. 2009). Mammalian spermatogenesis can be

morphologically divided into a number of different “stages” based on the cellular associations present in

a given cross section. This system was first developed in the rat by Leblond and Clermont (1952), with

the first detailed classification in the mouse following later (Oakberg. 1956). Mouse spermatogenesis

was divided into 12 stages, and was classified in even further detail by Russell (1990), and represents the

staging scheme that will be used throughout this thesis (Figure 1.2). It should be noted, that while

staging seminiferous tubules is a very useful tool, the stages described represent artificial delineations in

a continuous process and intermediates between stages are commonly found in the seminiferous tubules.

Developing germ cells do not move laterally in the seminiferous tubule; thus, a given cross-sectional

plane in a seminiferous tubule would move temporally through each of the 12 stages before returning to

stage 1 and repeating the cycle. With the exception of the first wave of spermatogenesis in juvenile

mice, the spermatogenic cycle is not synchronized along the entire length of a seminiferous tubule.

Instead, the spermatogenic cycle presents a wave-like pattern of stages along the longitudinal axis such

that successive segments are found to be at sequential stages of the cycle, in descending order moving

distally from the rete testis (Russell et al. 1990). During the first wave of spermatogenesis, there is a

much higher degree of synchronization throughout the seminiferous tubules; however, this

synchronization is not absolute, as is frequently assumed to be the case (Varmuza &Ling 2003).

Page 19: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

8

Figure 1.2: The mouse spermatogenic cycle. A diagram depicting the cell types present at each of the

12 stages of mouse spermatogenesis (based on diagram in Russell et al., 1990). Vertical columns depict

the cell types associated with each stage, numbered by Roman numerals. As cells progress through

spermatogenesis they move closer to the tubule lumen, which is indicated by vertical position in the

individual columns. Steps in spermatid development are indicated by Arabic numbers. Spermatogonia

and Spermatocytes are labelled as follows: mIn

= mitosis of Type A4 spermatogonia/ intermediate

spermatogonia, In

m = Intermediate spermatogonia/mitosis of intermediate spermatogonia, B = type B

spermatogonia, Bm =type B spermatogonia/mitosis of type B spermatogonia, Pl = preleptotene

spermatocyte, L = leptotene spermatocyte, Z = zygotene spermatocyte, P = pachytene spermatocyte, D =

diplotene spermatocyte, m20m = secondary spermatocyte.

Page 20: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

9

Page 21: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

10

1.1.4 Mouse Models of Male Infertility

Advances in mouse (Mus musculus) genetic manipulation techniques have provided a wealth of

information to male infertility research. The ability to directly alter specific genetic loci by targeted,

deletion, tissue/time specific deletions, knock-in, point mutation, etc. has led to the identification of at

least 459 genes involved in spermatogenesis (Matzuk & Lamb 2008, Kuzmin et al. 2009) (See complete

list in appendix A.1). In addition, the process of spermatogenesis is highly conserved between mice and

humans at the developmental, tissue organization, and molecular levels (Russell et al. 1990, Bonilla &

Xu 2008). Together, these factors make mouse models a very powerful tool in the study of mammalian

spermatogenesis.

1.1.5 Male factor infertility

Infertility is a prominent medical condition, seriously affecting quality of life for those afflicted.

Recent studies have shown that currently, infertility affects between 11-16% of Canadian couples

(Bushnik et al. 2012). Of these couples, the cause of infertility is equally shared between males and

females (Nieschlag et al., 2010). One common class of male factor infertility is azoospermia—the

complete absence of spermatozoa in the semen. There are two types of azoospermia: obstructive, and

nonobstructive. The absence of sperm in nonobstructive azoospermia is due to a failure of

spermatogenesis (also known as testicular failure), as opposed to a physical blocking of sperm

progression via factors such as the congenital bilateral absence or blockage of the vas deferens or

epididymis (Batruch et al., 2012). Nonobstructive azoospermia is prevalent in the human population and

is displayed in 10-15% of infertile men, or roughly 1% of the general population (Hu et al. 2012).

Despite its prevalence, male infertility is rarely treatable and is more often bypassed using

assisted reproductive technologies (ARTs) such as intercytoplasmic sperm injection (ICSI). While these

technologies often satisfy the ultimate goal of successful reproduction, their consequences to future

generations are an issue of considerable debate. A recent large scale study of over 300,000 cases

Page 22: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

11

comparing children conceived naturally to children from assisted conception showed a significant

increase in the rate of birth defects in children conceived via ICSI (Davies et al. 2012). However, these

statistics do not necessarily indicate defects that are a product of the ICSI technique itself, but more

likely are reflective of genetic abnormalities that contributed to the initial infertility. Although a much

smaller amount of data is available, children conceived via ICSI from men exhibiting testicular failure

show no increased likelihood of birth defects or perinatal lethality when compared to children conceived

via ICSI from ejaculated spermatozoa; however there is a significant decrease in the chance of a

successful pregnancy being produced (Tournaye 2012). These studies suggest that bypassing male

infertility as opposed to treating it, results in the genetic causes of male infertility to be simply passed on,

which could negatively affect the fertility of future generations.

Presently a large proportion of male infertility cases are classified as idiopathic, and given a

description that classifies their infertility based on phenotype, but does not provide a specific cause for

the defect. Similarly, even if a genetic cause for male infertility can be diagnosed, there are very few

effective treatments available. It is clear from these facts that more basic research into the process of

spermatogenesis is needed. To date hundreds of genes that are required for fertility have been identified

(Matzuk & Lamb. 2008, Kuzmin et al. 2009), however in most cases their specific roles are poorly

defined. Furthering our understanding of the function of genes and gene networks in spermatogenesis is

the first step in improving the efficacy of diagnosing and treating infertility in males.

One gene that has been shown to be essential for spermatogenesis in mice is the protein

phosphatase Ppp1cc, which when deleted results in non-obstructive azoospermia (Varmuza et al. 1999).

By learning more about the function of this gene in spermatogenesis, we hope to better understand the

causes of idiopathic male infertility in humans.

1.2 Protein Phosphatase PPP1CC2

1.2.1 Protein phosphatases

Page 23: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

12

Protein phosphorylation is an ancient mechanism of protein modification that plays a role in the

regulation of countless cellular processes. There is currently debate over the percentage of proteins that

are phosphorylated in at least some context, but estimates range from 33 % (Cohen 1999) to as high as

70 % (Olsen et al. 2010). The status of protein phosphorylation is regulated by the opposing activities of

protein kinases and protein phosphatases. This regulatory mechanism is so prevalent that kinases and

phosphatases constitute between 2-4% of all genes in a typical eukaryotic genome (Moorhead et al.

2009). Although proteins can be phosphorylated on nine different residues, serine, threonine and

tyrosine phosphorylation constitute the vast majority of these events, representing 86.4, 11.8 and 1.8% of

phosphorylation events respectively (Moorhead et al. 2009, Olsen et al. 2006). The human genome

encodes approximately 90 phosphatases that either act on Tyr residues exclusively (PTPs or protein

tyrosine phosphatases), or act on Tyr, as well as Ser and Thr resides (DSPs or dual specificity

phosphatases) (Brautigan 2012). Phosphatases that act only on Ser and Thr residues fall into three

different protein superfamilies—PPP, PPM and DxDxT phosphatases (Brautigan 2012) which together

number approximately 40 in a typical mammalian genome (Moorhead et al. 2007, Bollen et al. 2010).

The PPP (phosphoprotein phosphatase) superfamily includes 13 different genes in humans throughout

the PP1, PP2A, PP2B, PP4, PP5, PP6 and PP7 subclasses (Moorhead et al. 2009, Kerk et al. 2008), and

account for over 90% of eukaryotic Ser/Thr dephosphorylation reactions (Heroes et al. 2012).

1.2.2 The PP1 family of protein phosphatases

The PPP superfamily is defined by a structurally conserved ~280 amino acid catalytic domain

containing 3 signature motifs (-GDXHG-, -GDXDRG- and –GNHC-) (Moorhead et al. 2009). All

eukaryotic and most bacterial and archeal genomes surveyed to date contain at least one PPP member

(Moorhead et al. 2009). Within this superfamily, the type-1 protein phosphatase family (PP1) is defined

by its biochemical activity—its preferential dephosphorylation of the β subunit of phosphorylase kinase,

and its inhibition by two heat-stable proteins known as Inhibitor-1 and Inhibitor-2 (Ingebritsen & Cohen,

1983). PP1s are some of the most highly conserved eukaryotic proteins known (Ceulemans & Bollen.

Page 24: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

13

2004) with mammalian catalytic domains being 76-88% identical to those of plants, and 90% identical to

those of fungi (Moorhead et al. 2009). In fact, the coding sequences for mouse PP1 isoforms Ppp1cc1

and Ppp1cc2 are capable of rescuing a cold-sensitivity phenotype found in a mutation of the S. pombe

homologue dis2 (Okano et al. 1997).

The mammalian genome contains three PP1 genes, Ppp1ca (PP1cα), Ppp1cb (PP1cβ) and

Ppp1cc (PP1cγ) the latter of which has two splice isoforms Ppp1cc1 (PP1cγ1) and Ppp1cc2 (PP1cγ2).

The Ppp1cc1 and Ppp1cc2 transcripts are identical until the final intron, which is retained in the Ppp1cc1

transcript, and contains a stop codon closely following the skipped splice junction (Okano et al. 1997).

All four mammalian PP1 isoforms are ubiquitously expressed with the exception of PPP1CC2 which is

testis-specific (Takizawa et al. 1994). Targeted deletion of mouse Ppp1cb results in preweaning lethality

(Lexicon Genetics. 2005); there is no published account of a Ppp1ca knockout phenotype, although one

paper has referred to unpublished data suggesting that Ppp1ca knockouts are viable (Cheng et al. 2009),

and personal communication from Shirish Shenolikar confirms this observation. Ppp1cc knockout mice,

which lack both the PPP1CC1 and PPP1CC2 isoforms, are phenotypically normal with the exception that

homozygous males are infertile (Varmuza et al. 1999). This mutant phenotype will be discussed in

greater detail in a later section.

Structurally, the PP1 catalytic subunits consist of a single, compact elliptical domain with a

central β sandwich from which the disordered C-terminus and extreme N-terminus emanate (Egloff et al.

1995, Ceulemans & Bollen 2004). The PP1 active site is located at the bifurcation point of a series of 3

shallow surface grooves in a Y-shaped conformation (Egloff et al. 1995, Goldberg et al. 1995). Each of

these 3 grooves has distinct amino acid side chain compositions and are termed the hydrophobic, acidic

and C-terminal grooves, the latter of which runs towards the C-terminus of the catalytic subunit

(Goldberg et al. 1995). At the active site two metal ions, Fe+2

and Zn+2

(Mn+2

when bacterially

expressed) are positioned and are utilized in a single-step metal-assisted catalysis mechanism for

substrate dephosphorylation (Egloff et al. 1995, Goldberg et al. 1995) (Figure 1.3). In this mechanism,

Page 25: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

14

the positively charged metal ions stabilize the negative charges in the phosphate ester and make them

susceptible to nucleophilic attack by an activated water molecule, resulting in hydrolysis of the

phosphate group from the substrate (Egloff et al. 1995, Goldberg et al. 1995). Association of proteins and

phosphate analogues to the active site does not appear to significantly alter the structure of the PP1

catalytic subunits (Egloff et al. 1995, Hurley et al. 2007).

1.2.3 Regulation of PP1 catalytic subunits

Mammalian genomes encode a roughly equivalent number of tyrosine kinases and phosphatases

(approximately 100 each) (Ceulemans & Bollen 2004). Conversely, mammals have approximately 10

times as many Ser/Thr kinases as Ser/Thr phosphatases (Moorhead et al. 2007, Bollen et al. 2010). This

fact has prompted the question of how so few phosphatases oppose the activity of so many kinases.

Historically, it was thought that phosphatases were far less specific than kinases, and simply removed

phosphate groups from proteins indiscriminately, passively undoing the work of more specific kinases.

This view was due in large part to the fact that in vitro bacterially expressed protein phosphatases such as

PP1s exhibit very broad substrate specificity and can even dephosphorylate tyrosine residues (Egloff et

al. 1995). However, it is now accepted that Ser/Thr phosphatases like PP1 are actually subject to very

precise regulation via interaction with a large and diverse range of “regulatory subunits” (Hubbard &

Cohen. 1993). These regulatory subunits are generally unrelated in both origin and structure. To date

there are approximately 200 known PP1 interacting proteins (PIPs), many of which act as regulatory

subunits. This represents a very large pool of PP1 holoenzymes, each with its own target(s) and

regulatory properties (Heroes et al. 2012). Despite the large number of PIPs currently known, it is

hypothesized that there are significantly more that remain to be discovered, perhaps as many as 650 in

total (Bollen et al. 2010). When considering this holoenzyme view of PP1 diversity it is appreciated how

so few phosphatase catalytic subunits could oppose the activity of hundreds of kinases.

Page 26: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

15

Figure 1.3: The PP1 structure and catalytic mechanism. (A) The structure of the PP1 catalytic subunit.

Circled is the active site. (B) A diagram depicting the PP1 active site with catalytically important amino

acids shown (function indicated by color on left). The phosphate group and leaving group are also

shown. Green circles labelled “M” indicate metal ions, and arrows indicate the course of the

nucleophillic attach mechanism. PPP1CC structure image modified from original version downloaded

from the RCSB PDB (www.pdb.org) of PDB208A (Hurley et al., 2007).

Page 27: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

16

A

B

Page 28: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

17

Almost 90% of known PIPs contain a short degenerate motif commonly referred to as the RVxF

motif (Bollen et al. 2010). One caveat to this statistic is that the presence of the RVxF motif has been

utilized successfully in several studies to bioinformatically search for novel PIPs (Meiselbach et al. 2006,

Hendrickx et al. 2009), and thus the current set of proteins may be biased in this respect. Regardless of

the exact number, many regulatory subunit-PP1 interactions are mediated by this motif. Several different

definitions of the RVxF motif have been offered, from the sensitive, but relatively non-specific [RK]-

X(0,1)-[VI]-{P}-[FW] (Wakula et al. 2003), to the less sensitive but more specific [ACHKMNQRSTV]-

[V]-[CHKNQRST]-[FW] (Meiselbach et al. 2006). Based on the most recently published catalog of

validated PIPs, the RVxF motif follows the consensus sequence [K55R34]-[K28R26]-[V94I6]-{FIMYDP}-

[F83W17] (subscripts represent % occurrence) (Heroes et al. 2012). The RVxF motif binding site is a

shallow hydrophobic groove on the surface of the PP1 catalytic subunit, remote from the active site

(Egloff et al. 1997). There are numerous examples of PIPs that do not contain RVxF motifs, and thus its

presence or absence is not deterministic of interaction with PP1. As is the case for substrates, interaction

with an RVxF motif does not result in any large conformational change to the catalytic subunit (Egloff et

al. 1997). However, when present the RVxF motif is thought to serve as an anchoring point for

interaction, allowing for weaker secondary interaction motifs to bind to the PP1 surface (Hendrickx et al.

2009). Several less common PP1 interaction motifs have been identified, including the G/SILK motif

(found in seven known PIPs) which is thought to function as an additional PP1 anchoring motif located

N-terminally to the RVxF motif, and the MyPhoNE motif (found in six known PIPs) which is thought to

play a role in substrate selection (Hendrickx et al. 2009, Heroes et al. 2012). One recent study

demonstrated that mutation of either the G/SILK motif or the adjacent RVxF motif in the S. Pombe PIP

Cut12 reduced binding efficiency to the PP1 catalytic subunit Dis2, while simultaneous mutation of both

docking motifs completely abolished interaction (Grallert et al. 2013). An emerging mechanism for the

regulation of the interaction between PP1 catalytic subunits and PIPs is the reversible phosphorylation of

the region in and around the RVxF motif, which a number of studies have demonstrated to decrease

Page 29: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

18

binding between the two proteins (Beullens et al. 1999, McAvoy et al. 1999, Liu & Brautigan 2000,

Bollen 2001, Grallert et al.2013).

The RVxF motif is also thought to have played an instrumental role in the evolution of PP1

catalytic subunits (Moorhead et al. 2009). There exist key RVxF containing PP1c regulatory subunits

that are functionally conserved in other eukaryotic genomes. Therefore it is theorized that the existence

of these ancient regulatory subunits hindered further evolutionary change in the catalytic subunits and

thus instead of evolving new domains to specify new targets, PP1 catalytic subunits instead “exploited”

this short degenerate motif in new proteins.

PIPs in general fall into one of four categories: substrates, inhibitors, subcellular targeting

subunits, or substrate specifying subunits (Figure 1.4). Substrates of PP1 have traditionally been difficult

to identify, due to the generally transient nature of the enzyme-substrate interaction. Unlike PTPs, PPPs

do not form a stable intermediate with substrate proteins. Despite these difficulties a number of PP1

substrates such as PTK2 and SRSF10 have been identified (Bianchi et al. 2005, Shi & Manley 2007).

Inhibitory PIPs bind to the PP1c active site and prevent the binding of substrates. One well known

example of a PP1 inhibitor is PPP1R2 (Inhibitor-2) which interacts with PP1c at the acidic and

hydrophobic grooves of the active site, preventing the binding of other PP1 substrates. In addition,

PPP1R2 either prevents the binding or actively displaces metal ions from the PP1c active site, further

inactivating the phosphatase (Hurley et al. 2007). Subcellular targeting subunits direct PP1c to specific

subcellular locations or compartments bringing it into contact with specific pools of substrates. One

recently discovered example of this mode of regulation is Repo-man (CDCA2) which recruits PPP1CC1

onto mitotic chromatin during anaphase (Trinkle-Mulcahy et al. 2006). Substrate specifying PIPs direct

PP1c activity towards a specific substrate or set of substrates. As mentioned previously, binding of

regulatory subunits via the RVxF motif does not significantly alter the confirmation of the PP1 catalytic

subunit. Instead, the binding of the regulatory subunit changes the available surface area of PP1, which

can sterically exclude certain interactions, and/or extend the binding surface for others (Heroes et al.

Page 30: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

19

Figure 1.4: Regulation of PP1 catalytic subunits via interaction with regulatory PIPs. PIPs which can act

as substrate specifying subunits (A, D), inhibitors (B), or subcellular targeting subunits (C). Substrate

specification can occur via partial blocking of the active site, restricting access for specific substrates (D)

or by providing additional surfaces for interaction (A). “RVXF” indicates the location on the PP1c

surface to which the RVxF docking motifs bind, “A” indicates the location of the PP1c active site. Based

on a figure from Heroes et al., 2012.

Page 31: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

20

Page 32: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

21

2012). One well characterized example of a substrate specifying PIP is that of Spinophilin (PPP1R9b),

resolved by a crystal structure of this holoenzyme complex (Ragusa et al. 2010). An RVxF motif found

in Spinophilin interacts with PP1c, allowing for several secondary points of interaction, which obstruct

both the acidic and hydrophobic grooves of the PP1c active site. This steric hindrance prevents PP1c

from dephosphorylating phosphorylase a (a known substrate), but is permissive to dephosphorylation of

the Spinophilin-PP1c substrate GLUR1. Although PIPs typically fall into one of the four aforementioned

categories it should be noted that this is somewhat of a simplified view, and some PIPs could be

considered to fall into more than one category. As well, the functional nature of many PIP-PP1

interactions remains unknown.

Another aspect of the fine-tuning of PP1 activity lies in the isoform selectivity of some PIPs.

While most PIPs studied appear to be capable of interacting with all PP1c isoforms, a growing number

seem to interact preferentially with certain isoforms in vivo. This fact is not surprising considering that

although the PP1c isoforms are very similar, they do diverge significantly at their C-termini. For

example, Repo-man although capable of interacting with PPP1CA, preferentially recruits PPP1CC1 to

chromatin during mitosis (Trinkle-Mulcahy et al. 2006). Similarly, studies of Neurabin-1 (PPP1R9a)

have indicated a binding preference of PPP1CC1 over PPP1CA and only a weak interaction with

PPP1CB (Terry-Lorenzo et al. 2002). There are also examples of proteins that are completely isoform

specific such as SPZ1 and a testis-specific Endophilin B1 isoform (SH3GLB1) that can only interact

with the testis-specific isoform PPP1CC2 (Hrabchak & Varmuza. 2004, Hrabchak et al. 2007). The

isoform selectivity of MYPT1 (PPP1R12A) for PPP1CB was illustrated structurally by Terrak et al.,

(2004) when they showed that MYPT1 interacted with a specific region of the PPP1CB C-terminus.

Research in the last two decades has shifted our view of phosphatases as relatively few

indiscriminate phosphorylation “reset buttons” to a large number of specifically regulated holoenzyme

complexes. Now that we have a better understanding of how PP1s function, our attention shifts to the

functional roles of specific PP1c isoforms. Overwhelmingly, PP1c research has focused on the 3

Page 33: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

22

isoforms expressed throughout somatic tissues: PPP1CA, PPP1CB and PPP1CC1. However, several labs

including ours have focused on the testis-specific PP1 isoform PPP1CC2.

1.2.4 Ppp1cc knockout mouse

Targeted deletion of the Ppp1cc gene (loss of PPP1CC1 and PPP1CC2) results in no observable

defects aside from infertility in homozygous male mice (Varmuza et al. 1999). These mice display a

prominent loss of germ cells, especially spermatids, from the seminiferous epithelium, indicating a

breakdown in spermatogenesis. The loss of cells appears to stem from both an increase in germ cell

apoptosis, and the premature sloughing of germ cells from the epithelium. Subsequently, Ppp1cc

knockout epididymides contain few germ cells, most of which are prematurely sloughed round and

elongating spermatids, and a small number of immotile and malformed spermatozoa. This phenotype

resembles a condition commonly found in humans, known as non-obstructive azoospermia, or testicular

failure.

Closer examination of the Ppp1cc knockout seminiferous epithelium shows a large number of

defects. The general architecture of the seminiferous epithelium is preserved and all stages of

spermatogenesis can be observed, which suggests no absolute block at any one developmental timepoint

(Varmuza et al. 1999). The first wave of spermatogenesis proceeds at a rate similar to that of wild-type,

but the first phenotypic effects become visible at 3 weeks of age, with the premature sloughing of germ

cells (late pachytene spermatocytes) first observed (Varmuza & Ling. 2003). As spermatogenesis

progresses in the adult testis, a general breakdown in the integrity of the spermatogenic cycle is

observed, with cell associations becoming asynchronous, making the staging of seminiferous tubules

challenging (Oppedisano-Wells & Varmuza. 2003). A quantitative analysis of seminiferous tubule

staging revealed a developmental bottleneck with an increased number of tubules found to be in stages

VII/VIII of spermatogenesis (corresponding to tubules containing mid-pachytene spermatocytes, see

Figure 1.2) and a reduced number of late stage tubules (Forgione et al. 2010). Further evidence of loss of

Page 34: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

23

epithelial integrity is found in the displacement of Sertoli cell nuclei from the basement membrane, and

the presence of large vacuoles in the epithelium (Varmuza et al. 1999). As mice age, the breakdown of

spermatogenesis becomes increasingly severe, eventually to the point of a Sertoli cell only (SCO)

phenotype in older mice (Oppedisano-Wells & Varmuza. 2003). This loss of epithelial integrity does not

appear to be due to defective cell junctions, as they are normal at the ultrastructural level (Forgione et al.

2010).

Morphologic defects appear throughout the seminiferous epithelium in Ppp1cc knockout mice,

affecting all germ cell types, especially round and elongating spermatids. Evidence of depletion of

spermatogonia can be observed, increasingly in older mice (Forgione et al. 2010) but with no apparent

defect in the mitotic rate of these cells (Varmuza et al. 1999). In spermatocytes, electron microscopy

revealed subtle abnormalities in chromatin condensation with unusual dense inclusions found in the

nuclei (Forgione et al. 2010). Spermatocytes are also found at a lower frequency in Ppp1cc mutant testes

(Varmuza & Ling. 2003). Although there is no block in meiotic progression, this process does appear to

be affected by the Ppp1cc deletion as evidenced by the presence of cells with >2 spindle poles or

multiple nucleolus-like structures as well as multinucleated spermatids (Varmuza et al. 1999) and an

increased frequency of recombination (Varmuza & Ling. 2003).

The most severe phenotypic consequences of Ppp1cc deletion are found in spermatids. As

mentioned above, a severe reduction in the quantity of post-meiotic cells is found in mutant tubules, and

those that are present are frequently abnormal. The majority of these defects seem to relate to nuclear

shaping and chromatin condensation. Elongating spermatid nuclei rarely display the characteristic

“hook-like” shape found in wild-type testes, and instead display a number of different shapes ranging

from globular to oblong “bowling pin” like shapes, and feature frequent indentations and abnormal

dorsal angles (Varmuza et al. 1999, Chakrabarti et al. 2007a, Forgione et al. 2010). Chromatin

condensation in Ppp1cc mutant spermatids is non-uniform at the round and elongating phases (Forgione

et al. 2010). Several processes known to play a role in chromatin condensation and nuclear shaping are

Page 35: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

24

disrupted in Ppp1cc knockout testes. Histones H1 and H3, which normally are largely replaced by

transition proteins, and later protamines during condensation of the spermatid nucleus, are retained in

Ppp1cc mutant spermatids, which offers a biochemical clue to the source of this phenotype (Varmuza et

al. 1999). As well, the manchette, which is thought to play a role in nuclear shaping, develops

abnormally in knockout spermatids, and is often ectopically positioned around the spermatid nucleus

(Forgione et al. 2010, Henderson et al. 2011). Unsurprisingly, given the morphological abnormalities

described above, there is an increase in DNA damage in round and elongating spermatids and their

improperly remodelled chromatin is more fragile, and thus unsuitable for fertilization even by in vitro

techniques such as ICSI (Jurisicova et al. 1999, Davies & Varmuza. 2003). Other key structures in the

spermatid are also abnormal upon deletion of Ppp1cc. The initiation of acrosome biogenesis is

unimpeded; however defects begin to appear in step 7 spermatids resulting in the presence of large or

multiple vacuoles around the nuclear surface (Forgione et al. 2010). Flagellar abnormalities such as

poorly developed mitochondrial sheaths and disorganized outer dense fibers (Chakrabarti et al. 2007b)

are present, which is consistent with the immotile nature of the few surviving epididymal spermatozoa

(Varmuza et al. 1999).

The numerous defects throughout the seminiferous epithelium in Ppp1cc knockout mice suggest

the possibility of a pleiotropic function for this gene in spermatogenesis. This is not surprising given the

functional diversity and multitude of protein-protein interactions typical of PP1s. In fact, there are

several known examples of proteins that play different roles in both meiotic and post-meiotic

spermatogenic cells, such as PARP2 and HSPA2 (Dantzer et al. 2006, Govin et al. 2006). Conversely, it

is possible that the loss of a single PPP1CC holoenzyme complex renders a single substrate

hyperphosphorylated in the mutant testis, producing a cascade of secondary effects. This scenario is not

unprecedented, as due to the highly regulated nature of the spermatogenic cycle, a single defect can

result in widespread germ cell degeneration throughout the testis as is seen in the mutation of

transcription factor CREM (Blendy et al. 1996, Yan. 2009).

Page 36: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

25

The Ppp1cc deletion in this study results in a loss of both the ubiquitous PPP1CC1 and testis-

specific PPP1CC2, but the phenotype is restricted to the testis. Other PP1 isoforms, PPP1CA and

PPP1CB, are able to compensate for a loss of the Ppp1cc gene in every tissue except the testis, despite

being expressed in that tissue (Varmuza et al. 1999). This has led our laboratory to hypothesize that there

is an essential role(s) for the PPP1CC2 isoform in spermatogenesis. This fact is supported by the

conservation of this testis-specific isoform throughout mammals (Shima et al. 1993, Takizawa et al.

1994, Smith et al. 1996, Vijayaraghavan et al. 1996). Alternatively, there are two additional

possibilities—that both the PPP1CC1 and PPP1CC2 each have separate essential roles in

spermatogenesis, or that the Ppp1cc mutant phenotype is a result of a decreased total PP1 dosage in the

testis. The former can be ruled out, as recent data has shown that ectopically expressed PPP1CC2 can

rescue the infertility found in Ppp1cc mutant males (Sinha et al. 2012). Whether the mutant phenotype is

due to a dose dependent, or isoform specific mechanism remains to be determined, and will be discussed

in greater detail later.

1.2.5 PPP1CC2 protein-protein interactions in the testis

As mentioned in the preceding sections, PP1s primarily function as holoenzymes, being directed

towards specific substrates by a large and diverse array of regulatory subunits. Thus, if we wish to

understand the function of PPP1CC2 in spermatogenesis, we must characterize its interactome in the

testis. The Varmuza lab, as well as several others, has conducted a number of different studies over the

past several years to that aim. Yeast 2-hybrid analysis using mouse testis cDNA libraries has identified

two testis-specific proteins that interact with only the PPP1CC2 isoform—SPZ1 and a novel testis-

specific splice isoform of Endophilin B1 (SH3GLB1) (Hrabchak & Varmuza 2004, Hrabchak et al.

2007). While SH3GLB1 is not essential for male fertility (Takahashi et al. 2007), SPZ1, when

overexpressed, results in over-proliferation of germ cells which causes a breakdown in spermatogenesis

and subsequent male infertility (Hsu et al. 2001). To date, the biological implications of these

interactions remain unclear. A similar study using a human testis cDNA library identified a number of

Page 37: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

26

candidate PPP1CC2 interacting proteins in the testis and confirmed an interaction with ANKRD42

(SARP2) via coimmunoprecipitation in human sperm extracts (Fardilha et al. 2011b). ANKRD42 had

been previously identified as a PIP in other tissues, and thus, interaction is not isoform specific (Browne

et al. 2007). The functional consequences of this interaction are still unknown, although ANKRD42 is a

DNA binding protein and may function to bring PP1 to that subcellular locale (Browne et al. 2007).

Another protein implicated in spermatogenesis, PPP1R42, otherwise known as TLRR (testis leucine-rich

repeat) was validated as a PPP1CC2 interactor via coimmunoprecipitation in mouse testis lysate (Wang

et al. 2010). PPP1R42 is a testis-specific protein that also interacts with the molecular motor kinesin 1B

(KIF1B) and β-tubulin in the testis (Wang et al. 2010). In early elongating spermatids, PPP1R42

localizes to the manchette and is later transported to the centrosome (Wang et al. 2010). PPP1CC2 forms

a complex with PPP1R42 that has active phosphatase activity, and is most abundant early in

spermiogenesis (likely round spermatids) prior to its association with the manchette (Wang & Sperry

2011). While the function of this holoenzyme remains unknown, the link between PPP1R42 and the

cytoskeleton suggest a role in the complex cytoskeletal rearrangements that occur throughout

spermiogenesis (Wang & Sperry 2011).

While the focus of this thesis is primarily the role of PPP1CC2 in spermatogenesis, there also

appears to be a prominent role for PPP1CC2 in the regulation of sperm motility. After release from the

seminiferous epithelium, spermatozoa enter the caput epididymis, at which point they are still immotile.

As spermatozoa transit to the caudal epididymis, known as epididymal maturation, they become motile.

Enzymatic PP1 activity in spermatozoa is associated with the inhibition of motility (Smith et al. 1996).

This facet of PPP1CC2 activity in no way explains the infertility phenotype found in Ppp1cc mutant

males, as the defects with the seminiferous epithelium occur long before the process of epididymal

maturation. Nevertheless, understanding sperm motility is an important aspect of reproductive biology,

and thus a number of groups have studied PPP1CC2 interactions during this process. When results from

a number of studies are considered together, a general mechanism of PPP1CC2 regulation in the

Page 38: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

27

acquisition of sperm motility begins to emerge. PPP1CC2 has been shown to interact with a number of

classical PP1 regulators, which are well characterized PP1 inhibitors, such as PPP1R2, PPP1R7 and

PPP1R11 as a part of inactive complexes in motile caudal spermatozoa, but not (or to a lesser degree) in

immotile caput spermatozoa (Vijayaraghavan et al. 1996, Huang et al. 2002, Cheng et al. 2009). This

suggests that active PPP1CC2 complexes participate in one or more processes in spermatogenesis whilst

also serving to keep the developing spermatids from becoming prematurely motile prior to the

completion of development. Then, when the mature spermatozoa are “ready” for motility, strong PP1

inhibitors bind PPP1CC2 at various regions throughout the cell (Fardilha et al. 2011a), initiating sperm

motility. However, this is a somewhat simplified picture of PPP1CC2 regulation in spermatozoa, as each

individual complex will have its own regulatory mechanism. One recently described example is that of

the OAZ3-PPP1R16A (MYPT3)-PPP1CC2 complex. In Oaz3 knockout mice, male infertility is

produced due to the separation of sperm heads from tails despite both being ultrastructurally normal

(Tokuhiro et al. 2009). The headless tails of Oaz3 knockout sperm appear to display increased vigorous

flagellar beating (Ruan et al. 2011). Further analysis revealed that PPP1R16A binds directly to both

OAZ3 and PPP1CC2 (Ruan et al. 2011). When PPP1R16A and PPP1CC2 alone are in complex, there is

a lack of phosphatase activity; however, the presence of OAZ3 in the complex renders PPP1CC2 active

(Ruan et al. 2011). Therefore, a possible molecular explanation for the Oaz3 knockout phenotype is

premature and overly vigorous motility brought on by the ablation of PPP1CC2 activity (Ruan et al.

2011). Several other proteins have been implicated as PPP1CC2 interactors in spermatozoa such as

YWHAZ (14-3-3zeta) (Puri et al. 2008) and several A-kinase anchoring proteins (AKAPs) (Reinton et

al. 2000, Huang et al. 2005, Fardilha et al. 2011a) but their regulation and effect on sperm motility are

poorly understood.

1.2.6 Identification of PPP1CC2 Substrates in the testis

Substrates of Ser/Thr phosphatases such as PPP1CC2 have typically been more challenging to

identify than regulatory PIPs as their interaction with the PP1 catalytic subunit is frequently weak and/or

Page 39: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

28

transient. Thus, as an alternate means of identifying candidate substrates of PPP1CC2 in the testis our

lab capitalized on the fact that substrates should be hyperphosphorylated when the Ppp1cc gene is

deleted (Henderson et al. 2011). In this experiment, testis lysates from adult wild-type and Ppp1cc

knockout mice were subjected to two-dimensional electrophoresis followed by ProQ phosphoprotein

staining. Comparative analysis of replicate gels identified 10 reproducible protein spots that indicated

increased phosphorylation in Ppp1cc mutant testis lysate, with no change in general protein abundance.

Protein content of the hyperphosphorylated spots was identified via mass spectrometry and amongst the

identified proteins were the testis specific heat shock protein HSPA2 and several isoforms of α and β

tubulin (Henderson et al. 2011). Both of these proteins were subsequently shown to interact with

PPP1CC2 in the testis. This work will be discussed further in chapter 4 and appendix A.3.

1.3 Hypothesis and Research Objective

The PP1 protein phosphatase gene Ppp1cc is essential for completion of spermatogenesis in

mice. Additional highly similar PP1c isoforms PPP1CA and PPP1CB are capable of compensating for

the loss of Ppp1cc in every tissue with the exception of the testis, despite their expression in that tissue.

This suggests that the predominant Ppp1cc isoform in the testis, PPP1CC2 has an isoform specific

function in spermatogenesis. As PP1s are known to be incorporated into multi-protein complexes to

dephosphorylate substrate proteins, my research objective is to identify novel PPP1CC2 interacting

proteins in the mouse testis to gain a better understanding of PPP1CC2’s regulation and function(s) in

spermatogenesis. This objective encompasses the identification of both PPP1CC2 regulatory proteins

and putative substrates.

To this end, I have used a number of proteomic and biochemical approaches the results of which

are detailed in the forthcoming sections. Each of these studies utilized LC-MS/MS analysis as a means

for protein identification. For the first approach, I have created a transgenic mouse embryonic stem cell

line expressing tandem affinity tagged versions of PPP1CC1 and PPP1CC2 which were used in tandem

Page 40: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

29

affinity purification experiments to identify both previously characterized, and novel PIPs via LC-

MS/MS. In my second approach, I performed sedimentation assays using recombinant GST-PPP1CC1

and GST-PPP1CC2 as bait in mouse testis lysate to identify novel interacting proteins in the mouse testis.

Finally, in a third approach, I performed a phosphoproteomic analysis to identify proteins that were

hyperphosphorylated in Ppp1cc mutant testes, which represent candidate substrates of PPP1CC2.

Page 41: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

30

Chapter 2: Tandem affinity purification in transgenic mouse embryonic stem cells identifies

DDOST as a novel PPP1CC2 interacting protein

Author’s note: A modified version of this chapter was published as: MacLeod G and Varmuza S (2012).

Tandem affinity purification in transgenic mouse embryonic stem cells identifies DDOST as a novel

PPP1CC2 interacting protein. Biochemistry, 51 (48), 9678–9688.

Experiments were designed and conceived by myself and S. Varmuza. I was responsible for performing

all experiments, as well as writing the paper with editing from S. Varmuza.

Abstract

The PP1 family of protein phosphatases achieve functional diversity through numerous and varied

protein-protein interactions. In mammals there are four PP1 isoforms, the ubiquitously expressed

PPP1CA, PPP1CB and PPP1CC1, and the testis specific splice isoform PPP1CC2. When the mouse

Ppp1cc gene is deleted, the only phenotypic consequence is a failure of spermatogenesis in homozygous

males. To elucidate the function of the Ppp1cc gene we sought to identify novel protein-protein

interactions. To this aim, we have created SBP-3XFLAG-PPP1CC1 and SBP-3XFLAG-PPP1CC2

knock-in mouse embryonic stem cell lines using a gene trap based system. Tandem affinity purification

using our knock-in cell lines identified 11 significant protein-protein interactions, including 9 known PP1

interacting proteins and two additional proteins—ATP5C1 and DDOST. Reciprocal in vitro

sedimentation assays confirmed the interaction between PPP1CC2 and DDOST which may have

physiological implications in spermatogenesis. Immunolocalization studies revealed that DDOST

localized to the nuclear envelope in dissociated spermatogenic cells, and persists throughout

spermatogenesis. The knock-in system described in this paper is applicable for creating tandem affinity

tagged knock-in embryonic stem cell lines with any gene for which a compatible gene trap line is

available.

Page 42: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

31

2.1 Introduction

The current consensus is that the PP1 family of protein phosphatases achieve functional diversity

via interaction with a large number of regulatory proteins termed PP1 interacting proteins (PIPs). To

date, approximately 200 PIPs have been identified but emerging evidence suggests that there could be

many more, perhaps as many as 650 (Bollen et al. 2010). Therefore, to fully understand the role of a PP1

isoform, such as PPP1CC2 (Serine/threonine-protein phosphatase PP1-gamma catalytic subunit, isoform

2), it is important to determine to the fullest extent possible the interactome of that protein. This can be a

challenge for PIPs, as they are generally unrelated and difficult to identify based on sequence alone.

Previous attempts to identify PIPs computationally based on the presence of the RVxf motif that is

present in many known interactors have been moderately successful (Hendrickx et al. 2009); however

they cannot identify the complete interactome, as not all PIPs contain an RVxf motif.

The difficulty of identifying PIPs in silico means that to fully describe the interactome of PP1s,

experimental approaches are necessary. One powerful system of identifying protein-protein interactions

is Tandem Affinity Purification (TAP) (Rigaut et al. 1999). The use of two affinity purification steps

significantly reduces the amount of contaminating proteins in the final product making the technique

more sensitive in identification of legitimate interactors. The use of LC-MS/MS in protein identification

aids in this sensitivity allowing for the simultaneous identification of many interactors, even those in

relatively low stoichiometry. Additional advantages of TAP over other methods such as yeast 2-hybrid

include the ability to identify protein complexes that have more than two members, and the ability to

target expression of the bait protein to a variety of locations within the cell. Numerous TAP systems

have been developed that feature improved expression in mammalian systems, employing combinations

of affinity tags such as the FLAG-tag, and Streptavidin-binding peptide (SBP) (Angers et al. 2006, Chen

& Gingras 2007). In addition to being more readily expressed in mammalian cells, these tags are also

smaller in size, and thus less likely to interfere with the function of the tagged protein. TAP has now

been used in many studies, most frequently using cell-culture with transiently expressed TAP-tagged

Page 43: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

32

proteins. Such techniques are able to give great insight into the interactomes of many proteins, but they

do have limitations based on the artificial nature of the system, including false-positive interactions

based on over/mis-expression of the bait protein. In addition, not all cell types/tissue can be modelled in

culture, and thus cell or tissue specific interactions may be missed. In an effort to combat these

limitations, some groups have begun to utilize TAP in transgenic mice. One such example utilized FLAG

and HA tandem affinity tagged Cyclin D1 knock-in mice to identify Cyclin D1 interacting proteins in

multiple mouse tissues, including brain and eyes (Bienvenu et al. 2010).

PPP1CC2 is a testis specific splice isoform of the PP1 family member Ppp1cc that is essential

for spermatogenesis (Varmuza et al. 1999). Deletion of Ppp1cc results in a wide range of morphological

abnormalities in the testis, most prominently a severe depletion of developing germ cells. However the

precise mechanism by which deletion of the Ppp1cc gene contributes to failure of spermatogenesis

remains unknown. In an effort to gain insight into the role of PPP1CC2 in spermatogenesis our aim is to

characterize its interactome. It is currently not possible to model the testis in cell culture due to its

complex architecture, and the fact that most of the relevant cells are post-meiotic. Therefore we have

sought to generate a system for performing PPP1CC2 TAP in transgenic mouse cells. In an effort to

recapitulate the wild-type expression pattern of the bait protein, and avoid artifacts associated with over-

expression, we have relied on gene-trap technology to ensure that expression is under the control of the

endogenous promoter. In addition to PPP1CC2, this system is amenable to any gene for which a gene-

trap ES cell line is available, making it a powerful tool for determining protein-protein interactions in a

transgenic setting.

2.2 Materials and Methods

Knock-in Vector Construction

The Streptavidin binding peptide (SBP)-3xFLAG knock-in vector (Figure 2.1A) was built using

pFloxin as a backbone (Singla et al. 2010). The pFloxin plasmid contains a beta-actin promoter sequence

Page 44: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

33

followed by a lox66 site, and was generously provided by Dr. Bill Skarnes (Wellcome Trust Sanger

Institute). Cre-mediated recombination between the lox66 site in the knock-in vector and lox71 in the

pGT0lxf genetrap vector creates a double-mutant Lox site which shows a low affinity for Cre-

recombinase, and thus increases the efficiency of Cre-mediated insertion of the knock-in vector (Zhang

& Lutz 2002). The Engrailed2 (En2) splice-acceptor sequence was cloned from the pGT0lxf gene trap

line RRR804 (International Gene Trap Consortium) and inserted 3’ of the lox66 site. The internal

ribosomal entry site (IRES) of the encephalomyocarditis virus (ECMV) was cloned from the pGLUE

plasmid (Angers et al. 2006) which was generously provided by Dr. Stephane Angers (University of

Toronto, Faculty of Pharmacy). Immediately 3’ of the IRES, the SBP and 3xFLAG tandem affinity tags

were inserted, having been cloned from the pGLUE and pTFHW plasmids respectively. The SBP and

3xFLAG affinity tags were separated by a spacer sequence encoding the amino acids GGSPGGT.

Following the 3xFLAG affinity tag, an oligonucleotide sequence that included the coding region for the

spacer peptide sequence GGSPGG, as well as SphI and AflII restriction sites, was inserted. Finally, a

bovine growth hormone polyA signal fragment was cloned from the pGLUE plasmid and inserted 3’ of

the multiple cloning site.

The coding sequences for Ppp1cc1 and Ppp1cc2 were PCR amplified from pGEM7z vectors

containing their respective full-length cDNA sequences (Mann et al. 1995) to generate sticky-ended PCR

products according to the protocol of Walker et al. (Walker 2008). Primer sequences are as follows: for

Ppp1cc1, forward reaction 1, 5’-CATGGCGGATATCGACAAACTC-3’ (primer A); reverse reaction 1,

5’- TTAACTATTTCT TTGCTTGCTTTGTGATC-3’ (primer B), forward reaction 2, 5’-

GCGATATCGACAAACTCAAC-3’ (primer C); reverse reaction 2 5’-

CTATTTCTTTGCTTGCTTTGTGATC-3’ (primer D). For Ppp1cc2, forward reaction 1 primer A;

reverse reaction 1 5’-TTAATCGGTCACTCGTAAGGA-3’ (primer E); forward reaction 2 primer C;

reverse reaction 2 5’- TCGGTCCACTCGTATAGGA-3’ (primer F). The sticky-ended PCR products

Page 45: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

34

were then ligated into the SphI (5’) and AflII (3’) sites of the Strep-3xFLAG knock-in vector. Vector

sequence was verified by sequencing.

Cell Culture

ES cells were cultured on 0.1% gelatin in high glucose DMEM supplemented with 15% FBS,

nonessential amino acids, 1 mM sodium pyruvate, 2 mM glutamine, penicillin-streptomycin, 10 µM β-

mercapto-ethanol, and 8 µg/mL leukemia inhibitory factor.

Cre-mediated Excision

RRR804 (Bay Genomics) ES cells were grown to ~70% confluence in a single well of a

gelatinized 24-well tissue culture dish and subsequently transfected with 800 ng pCMV-Cre-EGFP using

the Lipofectamine 2000 reagent (Invitrogen) according to the manufacturer’s protocol. The pCMV-Cre-

EGFP plasmid encodes a Cre-EGFP fusion protein and was a generous gift from Dr. Sabine Cordes

(Samuel Lunenfeld Research Institute). Successful transfection was verified by viewing EGFP

fluorescence. Proper cre-mediated excision eliminates expression of the β-geo reporter sequence,

resulting in G418 sensitivity. Thus to aid in the selection of cells that have undergone Cre-mediated

excision pCMV-Cre-EGFP treated RRR804 ES cells were sorted into GFP+ and GFP- fractions via

fluorescence activated cell sorting (FACS). Transfected cells were harvested and resuspended in 1 X

PBS, 5mM EDTA, 25mM HEPES and 1% FBS, followed by filtration through a 40µM nylon mesh cell

strainer. FACS was carried out using a FACS Aria machine at the University of Toronto, Faculty of

Medicine Flow Cytometry Facility. Sorted cells were collected in DMEM with 50% FBS, and GFP+

fraction was plated on 10cm tissue culture dishes at low density in standard culture media. Cells were

grown for approximately 6 days until ES colonies became visible, and were subsequently picked.

Cre-mediated Insertion

Page 46: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

35

RRR804-SA ES cells were grown to approximately 70% confluence in 10cm tissue culture

plates and were then trypsinized, harvested and washed in PBS. After resuspension in PBS cells were

counted using a hemacytometer. Cell concentration was then adjusted to approximately 107 cells per 800

µL PBS. 107 RRR804-SA cells were added to a chilled electroporation cuvette along with 60 µg each of

pCX-nlsCre and either Ppp1cc1 or Ppp1cc2 knock-in vector. pCX-nlsCre expresses Cre-recombinase

with a nuclear localization signal, under the control of a β-actin promoter and was a generous gift from

Dr. Andras Nagy (University of Toronto, Samuel Lunenfeld Research Institute, Toronto). Cells were

electroporated at 240V, 500 µF using a BioRad GenePulser. After electroporation, cells were incubated

at room temperature for approximately 20 minutes and then plated evenly between six 10cm tissue

culture plates. G418 selection was started on the second day after electroporation, and was conducted at

a concentration of 300 µg/mL. Colonies were picked between days 8 and 12 of G418 selection.

RT-PCR

RNA extraction and cDNA synthesis were performed using standard protocols. Primer sequences

utilized are as follows: 1F 5’-CGCACAGTCTAGGTGGGTATTGC-3’; 1R 5’-

CTGCAAAGGGTCGCTACAGACG-3’; 2F 5’-ACGTGGTGGAGGGCCTGG-3’; 2R 5’-

GTGGTGTGACAGGTCTCGTG-3’; 3F 5’-GTTGCCTTTTATGGCTCGAGCGG-3’; 3R 5’-

TAACCGTGCATCTGCCAGTTTGAGG-3’; 4F 5’-TGAGAGGGACAGGCCACCAAGG-3’. IRES

forward primer 5’-GGACGTGGTTTTCCTTTGAA-3’; Ppp1cc coding sequence 3’ region 5’-

TGTGACAGGTCTCGTGGC-3’.

Western Blotting

SDS-PAGE and western blotting were performed using standard protocols. PPP1CC antibody

(N-19, Santa Cruz Biotechnology) was used at a concentration of 1:500, with Donkey anti-goat HRP

secondary (Santa Cruz Biotechnology) at a concentration of 1:5000. Anti-FLAG M2 antibody (Sigma)

Page 47: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

36

was used at a concentration of 1:5000, with Goat anti-mouse HRP secondary (Zymed) at a concentration

of 1:5000.

Tandem Affinity Purification

TAP protocol was adapted from Chen and Gingras (2007) and Angers et al. (2006). SBP-

3xFLAG-PPP1CC knock-in ES cells were harvested by trypsinization and washed once with PBS. Cells

were pelleted and flash frozen in liquid nitrogen, and stored at -80°C until processed. Cells were thawed

and resuspended in approximately 9 volumes of TAP lysis buffer (10% glycerol, 50mM HEPES-NaOH

pH 8.0, 150 mM NaCl, 2 mM EDTA, 0.1% NP-40, 1mM DTT, 10mM NaF, 0.25 mM sodium

orthovanadate, 50 mM beta-glycerolphosphate, 1mM PMSF, 1X sigma protease inhibitor), and incubated

a 4°C for 15 minutes with rocking. Lysate was then frozen in liquid nitrogen, and thawed at 4°C,

followed by centrifugation for 10 minutes at 10,000 x g to remove insoluble material. Cleared lysate was

then added to washed (3 x with TAP lysis buffer) Anti-FLAG-M2 resin (Sigma) at a ratio of

approximately 2.5 µL of packed resin for each 10cm plate of ES cells harvested. FLAG pull-down was

incubated overnight at 4°C with rocking, and then washed 3 times with TAP lysis buffer. Protein was

eluted from Anti-FLAG-M2 resin using 3XFLAG Peptide (Sigma) at a concentration of 100 µg/mL in

400 µL of TAP lysis buffer for 30 minutes at 4 °C with rocking. This elution step was repeated one

additional time and eluates were pooled. FLAG elution was then added to 50 µL of packed Streptavidin-

Sepharose 4B (U. S. Biological), that had been washed 3 times in TAP lysis buffer, and incubated

overnight at 4°C with rocking. Streptavidin-Sepharose resin and bound proteins were washed 3 times

with TAP lysis buffer, and twice with Streptavidin Wash Buffer (10 mM β-mercaptoethanol, 50 mM

HEPES-NaOH pH 8.0, 150 mM NaCl, 1mM MgOAc, 1mM Imidazole, 0.1% NP-40, 2 mM CaCl2).

Bound proteins were eluted from Streptavidin-Sepharose using Streptavidin Wash Buffer supplemented

with 50 mM D-Biotin. Elution was performed with gentle vortex agitation at 4 °C, twice in 200 µL of

elution buffer and once in 100 µL with all elutions pooled. Eluted proteins were precipitated via

treatment with 30% trichloroacetic acid for 30 minutes on ice followed by centrifugation at ~16,000 x g

Page 48: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

37

for 30 minutes at 4 °C. Protein pellet was then washed twice with 500 μL of ice-cold acetone. Protein

pellet was resuspended in 6M Urea, and then reduced with 10 mM DTT in a final volume of 100 µL of

50 mM ammonium bicarbonate for 45 minutes at 50°C. Next, to alkylate the sample iodoacetamide was

added to a concentration of 33.3 mM and incubated at room temperature for 1 hour in darkness. To

digest the sample 1.2 µg of proteomics grade trypsin (Sigma) was added, and incubation at 37°C was

performed overnight. Trypsin was inactivated with 1% formic acid, and the sample was lyophilized.

Chimera Production

SBP-3XFLAG-PPP1CC2 knock-in ES cell clones were submitted to the Transgenic Core

Facility at the Toronto Centre for Phenogenomics (http://www.phenogenomics.ca; TCP) for chimera

generation via blastocyst microinjection with C57BL/6 hosts. ES cells were tested for both ploidy and

presence of contaminating viruses and bacteria (mycoplasma) before submission to TCP. Chimeric mice

were identified via coat colour. Chimeric mice were bred with both C57BL6 and CD1 mice and offspring

were identified by coat colour.

LC-MS/MS Analysis and Database Searching of TAP experiments

Peptide mixtures were subjected to LC-MS/MS analysis using a nanoLC (Eksigent) attached to a

LTQ-Orbitrap mass spectrometer (ThermoFisher) which was performed by the Centre for the Analysis of

Genome Evolution and Function (University of Toronto, Toronto, ON). LC-MS/MS data files were

processed into peak lists using Mascot Daemon and submitted to Mascot (Matrix Science, London, UK;

version 2.3.02) for database searching. The SwissProt mouse protein database 57.15 (515203 entries)

was queried, using the following parameters: carboxymethyl (C) set as a fixed modification, and

oxidation (M) as a variable modification, fragment ion mas tolerance of 0.80 Da and a parent ion

tolerance of 7 ppm.

His-tag sedimentation assay

Page 49: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

38

A recombinant His-tagged DDOST fragment (Abcam), corresponding to amino acids 144-360 of

human DDOST was bound to Ni-NTA resin. The His-DDOST resin was incubated with 1 mg of mouse

testis lysate in TAP lysis buffer for 2 hours at 4°C with rocking, followed by 3 washes with TAP-lysis

buffer. His-DDOST resin was boiled in 1X SDS-PAGE Sample loading buffer and submitted to SDS-

PAGE followed by western blotting.

Immunohistochemistry

Wild-type and Ppp1cc mutant testes were harvested and placed in dMEM after removal of the

tunica albuginea and homogenized using a razor blade to release germ cells from the seminiferous

tubules. To enhance germ cell release, tubules were squeezed repeatedly with square-tipped forceps. The

resultant homogenate was mixed and allowed to settle for several minutes. The developing germ cells

were then pelleted by centrifugation at 2,000 rpm for 2 minutes. Following removal of the supernatant

the germ cells were re-suspended in PBS and dropped onto poly-l-lysine coated slides and fixed in 4%

paraformaldehyde. Slides containing sections and dissociated cells were permeabilized with 0.01%

Triton-X in PBS, and blocked in 10% goat serum, 1% BSA, 0.01% Tween20, PBS solution, followed by

a second blocking step in 0.1 mg/mL AffiniPure F(ab’)2 fragment goat anti-mouse IgG (Jackson

ImmunoResearch). Primary antibody incubations were performed in antibody dilution buffer (5% goat

serum, 1% BSA, 0.01% Tween20, PBS) over night. Anti-DDOST (Santa Cruz Biotechnology, H-300)

was used at a concentration of 1:250 and Anti-MAb414 (Covance) was used at a concentration of 1:250.

Secondary antibody incubations were performed in antibody dilution buffer for 2 hours in darkness using

Cy3 conjugated secondary antibodies either AffiniPure Goat Anti-Rabbit IgG (DDOST, 1:2000) and/or

Dylight® 488 goat anti-mouse (Abcam). Slides were then stained with DAPI and mounted in 50%

glycerol for viewing with an Olympus BX60 microscope using the appropriate filters. Images were

captured using Cool Snap software and a CCD camera (RSPhotometrics). Images were merged using

ImagePro Plus version 4.1 and adjusted for brightness and contrast using Photoshop 6.0 (Adobe).

Page 50: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

39

Phosphopeptide enrichment and identification

Adult mouse testes were decapsulated and lysed in 400μL of 7M Urea, 2M Thiourea, 4%

CHAPs, 40mM Tris, reduced with 20mM DTT for 1 hour at room temperature and alkylated with 40 mM

iodoacetaminde for 35 minutes at room temperature in darkness. Testis proteins were precipitated with

acetone and dried using a speedvac, followed by resuspension in a 1M urea, 50 mM ammonium

bicarbonate solution containing 10μg of proteomics grade trypsin. Digestion was carried out over night

at 37°C, and at completion the solution was acidified with 1% formic acid and centrifuged to remove

insoluble material. 500 μg of testis protein was used for phosphopeptide enrichment via sequential

elution from IMAC (SIMAC) using the protocol from Thingholm et al. (Thingholm et al. 2009), but with

50μL of TiO2 Mag Sepharose (GE Healthcare) substituted for TiO2 phosphopeptide enrichment steps.

LC-MS/MS analysis and phosphopeptide identification was performed by the Advanced Protein

Technology Centre (Toronto, ON, Canada). All MS/MS samples were analyzed using Sequest (Thermo

Fisher Scientific, San Jose, CA, USA; version 1.3.0.339) and X! Tandem (The GPM, thegpm.org;

version CYCLONE (2010.12.01.1)). Sequest was set up to search MOUSE-Uniprot-Sep-05-12.fasta

(55250 entries) assuming the digestion enzyme trypsin. X! Tandem was set up to search the MOUSE-

Uniprot-Sep-05-12 database (55270 entries) also assuming trypsin. Sequest and X! Tandem were

searched with a fragment ion mass tolerance of 0.60 Da and a parent ion tolerance of 10.0 PPM.

Iodoacetamide derivative of cysteine was specified in Sequest and X! Tandem as a fixed modification.

Oxidation of methionine and phosphorylation of serine, threonine and tyrosine were specified in Sequest

and X! Tandem as variable modifications. For protein identification Scaffold (version Scaffold_3.4.3,

Proteome Software Inc., Portland, OR) was used to validate MS/MS based peptide and protein

identifications. Peptide identifications were accepted if they exceeded specific database search engine

thresholds. Sequest identifications required at least deltaCn scores of greater than 0.10 and XCorr scores

of greater than 1.8, 2.5, 3.5 and 3.5 for singly, doubly, triply and quadruply charged peptides. X! Tandem

identifications required at least -Log(Expect Scores) scores of greater than 2.0. Protein identifications

Page 51: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

40

were accepted if they contained at least 1 identified peptide. Proteins that contained similar peptides and

could not be differentiated based on MS/MS analysis alone were grouped to satisfy the principles of

parsimony. Statistical analysis of phospho-site localizations were performed using ScaffoldPTM

(version 2.0.0, Proteome Software Inc., Portland, OR).

2.3 Results

Generation of SBP-3XFLAG-PPP1CC1 and SBP-3XFLA-PPP1CC2 knock-in ES cell lines

In an effort to identify interactors of PPP1CC isoforms from transgenic mouse cells, we created

mouse embryonic stem (ES) cell lines carrying knock-in alleles of either PPP1CC1 or PPP1CC2 with N-

terminal Streptavidin-binding peptide (SBP) and 3XFLAG tandem affinity tags. A two-step strategy

based on that originally proposed by Hardouin and Nagy (2000) and also utilized by Singla et al. (2010)

was used (Figure 2.1B). The first step is the Cre-mediated excision of the additional loxP site in the

pGT0lxf based gene-trap line. pGT0lxf gene trap ES cell lines feature a gene-trap cassette containing an

En2 splice-acceptor sequence flanked by Lox71 (5’) and LoxP (3’) sites. Following the floxed splice-

acceptor is the coding sequence for the β-geo reporter gene. Recombination between the lox71 and loxP

sites results in the removal of the splice-acceptor sequence and should in theory restore the expression of

the endogenous Ppp1cc transcript(s).

The Ppp1cc gene trap ES cell line RRR804, obtained through the International Gene Trap

Consortium (Nord et al. 2006) and Mutant Mouse Regional Resource Centres, contains a gene trap

insertion at nucleotide 458 of intron 2, as verified by genomic DNA sequencing. RRR804 ES cells were

transiently transfected with pCMV-Cre-EGFP which expresses a Cre-recombinase-EGFP fusion protein.

Transfected cells were subjected to FACS, and the GFP positive fraction was isolated. To assay for

successful Cre-mediated excision, genomic DNA was isolated from GFP positive colonies, followed by

PCR using primers flanking the lox71-splice acceptor- loxP region. All GFP positive colonies screened

Page 52: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

41

Figure 2.1: Strategy for the generation of SBP-3XFLAG-PPP1CC knock-in embryonic stem cells. (A)

Schematic diagram of the knock-in vector used in this study. Actb = β-actin promoter, pA = bovine

growth hormone polyadenylation signal, AmpR = ampicillin resistance gene, SBP= Streptavidin binding

peptide, SA = splice acceptor sequence, FRT = Flp recombinase site. (B) Two-step strategy for

integration of the knock-in plasmid into pGTl0xf gene-trap lines.

Page 53: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

42

A

B

Page 54: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

43

had successfully undergone Cre-mediated excision, as evidenced by the smaller PCR product in Figure

2.2. If the FACS step was omitted, the percentage of colonies that showed successful Cre-mediated

excision was 25%. RRR804 cell lines with an excised splice-acceptor region were designated RRR804-

SA.

The second step is the integration of the knock-in plasmid at the remaining lox71 site. This was

accomplished via Cre-mediated insertion resulting from recombination between the gene-trap lox71 site

and a lox66 site in our knock-in plasmid. The backbone for our knock-in vector is the pFloxin plasmid

which was used in the generation of similar constructs by Singla et al. (2010). To this backbone we

added an En2 splice-acceptor sequence, followed by an IRES. Immediately following the IRES the

coding sequences for a SBP, a 3XFLAG tag, and either PPP1CC1 or PPP1CC2, as well as a BGH poly-

adenylation signal (Figure 2.1A). Proper integration of the knock-in vector should lead to expression of

an N-terminal SBP-3XFLAG-tagged version of the relevant transgene, under the control of the natural

promoter for the gene-trap locus. This vector was designed such that a tandem affinity tagged version of

any transgene can be integrated into any pGT0lxf gene-trap line. RRR804-SA ES cells were

simultaneously electroporated with both a Cre-recombinase expressing plasmid (pCX-nls-Cre) and either

the PP1CC1 or PPP1CC2 SBP-3XFLAG knock-in vector. To select for knock-in positive ES cells,

transformants were grown in the presence of G418, as proper integration of the knock-in plasmid will

expose the β-geo reporter gene to a β-actin promoter. A total of 48 G418 resistant ES cell colonies were

isolated (19 PPP1CC1 and 29 PPP1CC2).

Verification of SBP-3XFLAG-PPP1CC1 and SBP-3XFLAG-PPP1CC2 knock-in ES cell lines

To verify the presence of the knock-in allele, genomic DNA of 15 randomly selected G418 resistant

colonies was assayed via PCR using a series of primer pairs (Figure 2.3). All but one of the 15 assayed

G418 resistant ES cell colonies was positive for the presence of the knock-in construct. To verify the

expression of the SBP-3XFLAG-Ppp1cc transcripts, G418 resistant clones were subjected to

Page 55: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

44

Figure2.2: Cre-mediated excision of second loxP site in pGT0lxf gene-trap line RRR804. Depicted is

genomic DNA PCR product amplified using primers flanking the Lox71 and LoxP sites in the gene-trap

cassette. –Cre/+Cre indicates samples prior to or after treatment with Cre-recombinase respectively; L

indicates DNA ladder. Expected size of the original genomic DNA fragment is 861 bp, while fragments

having undergone Cre-mediated excision are expected to be 570 bp.

Page 56: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

45

Page 57: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

46

Figure 2.3: Verification of Cre-mediated insertion of transgene into RRR804-SA ES cell genome. (A)

Schematic diagram depicting the genomic DNA sequence features of a transgene positive clone at the

locus of insertion. Sequence features are coloured as in Figure 2.1. Location and direction of forward

(F) and reverse (R) primers used in the following section are shown. (B) Genomic DNA PCR product of

four different PCR reactions using the primer pairs indicated on the left in RRR804 gene-trap ES cells,

and representative SBP-3XFLAG-PPP1CC1 and SBP-3XFLAG-PPP1CC2 knock-in clones. Results

demonstrate the presence of the transgene in knock-in clones but not the original gene-trap cell line.

Page 58: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

47

Page 59: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

48

RT-PCR using a forward primer in the IRES sequence and a reverse primer in a 3’ region common to the

Ppp1cc1 and Ppp1cc2 transcripts (Figure 2.4A). Expression of the SBP-3XFLAG-Ppp1cc1 and SBP-

3XFLAG-Ppp1cc2 transgenes was detectable in respective knock-in ES cell lines at a size consistent

with the predicted sequence. Finally, to verify the effective translation of the knock-in constructs, ES

cell lysates were subjected to western blotting with an antibody against the FLAG epitope. As seen in

Figure 2.4B, both SBP-3XFLAG-PPP1CC1 and SBP-3XFLAG-PPP1CC2 knock-in ES cell lines showed

a robust FLAG signal, while no signal was detected in lysate from the original gene-trap ES cell line.

Curiously, western blotting with anti-PPP1CC showed only the <40 kDa band of the endogenous

PPP1CC1 signal, and no band with the expected molecular weight shift corresponding to the TAP tag.

This could possibly be due to the fact that the PPP1CC antibody used is directed against the N-terminus,

which may be sterically hidden by the N-terminal TAP tag.

PPP1CC1 and PPP1CC2 Tandem Affinity Purification in ES cells

To verify the functionality of the TAP tag in our SBP-3XFLAG-PPP1CC knock-in ES cells, TAP

experiments were carried out in ES cell culture. Optimization of our ES cell TAP protocol resulted in a

recovery of approximately 30% of our bait protein. This recovery is comparable with other mammalian

TAP systems (Li et al. 2011b). One note of particular interest is that initial experiments which utilized

Streptavidin-Sepharose as the first affinity purification step yielded very little capture of the bait protein.

Conversely, binding efficiency of the bait protein to the Streptavidin-sepharose resin, after elution from

the anti-FLAG-M2 affinity resin was nearly 100%.

After optimization, TAP studies were performed on cell lysate from SBP-3XFLAG-PP1CC ES

cells, and the isolated proteins were digested with trypsin and subjected to LC-MS/MS. The resultant

mass spectra were searched against the SwissProt mouse protein database using the MASCOT search

tool, and unique peptides exceeding the threshold score calculated by MASCOT (p<0.05; absolute value

Page 60: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

49

Figure 2.4: Expression of the SBP-3XFLAG-PPP1CC1/2 transgenes in knock-in ES cells at the cDNA

and protein level. (A) Product from RT-PCR reaction performed on ES cell RNA using a forward primer

which anneals to the IRES sequence and a reverse primer which anneals to a region in the Ppp1cc coding

sequence common to Ppp1cc1 and Ppp1cc2 demonstrates expression of the SBP-3XFLAG-PPP1CC1/2

transgenes. “Plasmid” indicates the use of the SBP-3XFLAG-PPP1CC knock-in plasmid as a template

for the PCR reaction, set to serve as a positive control. (B) Western blot of ES cell proteins using anti-

FLAG-M2 antibody (Sigma) shows expression of the SBP-3XFLAG-PPP1CC1/2 proteins.

Page 61: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

50

A

B

Page 62: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

51

calculated independently in each experiment) were recorded. In total, approximately sixty 10-cm tissue

culture dishes each of SBP-3XFLAG-PPP1CC1 and SBP-3XFLAG-PPP1CC2 ES cells were subjected to

TAP throughout 3-5 replicate experiments. Additionally, 4 negative control experiments were conducted

using ES cells that did not express our knock-in construct. Lists of proteins identified in our TAP

experiments as potential PPP1CC interacting proteins were subjected to a number of filtering criteria,

designed to eliminate contaminants and identify bona-fide interactors. First, proteins that were identified

in our negative control experiments were discounted, as they were likely to be contaminants. Remaining

proteins were then compared to three lists of likely contaminant proteins generated by other labs; a list of

common background contaminants from FLAG IPs (Chen & Gingras 2007), a dataset from a series of

FLAG IPs conducted in HEK293T cells expressing the FLAG-tag (A.C. Gingras, unpublished data), and

a TAP dataset from experiments using the GS-tag (includes SBP) that identifies non-specific interactors

(Burckstummer et al. 2006).

The top protein hit in all runs was identified as PPP1CC (average MASCOT protein score =

807), indicating that our transgenic lines were in fact expressing the expected bait protein, despite the

lack of anti-PPP1CC signal in western blotting. After application of the above-mentioned filtering

parameters, significant unique peptides corresponding to 76 different proteins were identified (Appendix

A.4.1/Supplementary table S1). However, identification of a protein via a single unique peptide can

result in a high incidence of false-positive interactions. Eleven proteins in our dataset were identified by

at least two unique and non-overlapping peptides, representing a more confident set of PPP1CC

interacting proteins (See Table 2.1). Amongst the identified PPP1CC interacting proteins, 9 were known

PP1 interacting proteins, thus validating the correct folding of our SBP-3XFLAG-PPP1CC fusion

proteins in our knock-in system. Two of the known PP1 interacting proteins, WDR82 and LMTK2 had

previously not been shown to bind to the PPP1CC2 isoform.

Page 63: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

52

Table 2.1: Protein-protein interactions detected via tandem affinity purification. “X”

indicates protein was identified in a tandem affinity purification experiment with the indicated bait

protein. * indicates a protein shown to interact indirectly with PP1, via an intermediate protein. +

indicates an interaction that did not meet our requirement of 2 unique peptide hits, but was included as it

is a known PPP1CC2 interacting protein in the testis.

Page 64: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

53

Gene

symbol

Uniprot

Accession

Number

Known

PIP?

RVxF

motif?

SBP-

3XFLAG

PPP1CC1

TAP

SBP-

3XFLAG

PPP1CC2

TAP

Best

MASCOT

protein

score

Unique

Peptides

Spectra

l Count

%

Coverage

PPP1R7 Q3UM45 Yes No X X 681 17 126 61.5

PPP1R8 Q8R3G1 Yes Yes X X 110 8 20 37.9

PPP1R2 Q9DCL8 Yes Yes X X 151 5 35 24.3

WDR82* Q8BFQ4 Yes No X X 68 4 8 16.3

DDOST O54734 No Yes X 56 3 3 6.3

PPP1R10 Q80W00 Yes Yes X 43 3 5 6.1

PPP1R11 Q8K1L5 Yes Yes X X 75 2 11 39.7

RRP1B Q91YK2 Yes Yes X 29 3 8 6.9

ATP2A2 O55143 Yes Yes X 85 2 3 3.8

ATP5C1 Q91VR2 No No X 41 2 2 7.4

LMTK2 Q3TYD6 Yes Yes X X 48 2 13 4.6

PPP1R42+ Q8R1Z4 Yes Yes X 25 1 1 2

Page 65: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

54

Aside from known PP1 interacting proteins we have identified two novel PPP1CC interactors in

our ES-cell TAP experiments. However, one of these proteins, ATP5C1, is a member of the ATP

synthase,

H+ transporting, mitochondrial F1 complex, to which 3 other proteins found in negative control

pull-down datasets belong, putting its assignment as a legitimate PPP1CC interacting protein into

question. The remaining putative PPP1CC interacting protein identified was Dolichyl-

diphosphooligosaccharide--protein glycosyltransferase 48 kDa subunit (DDOST). Examination of the

amino acid sequences of this protein revealed the presence of the classical PP1 binding RVxF. DDOST

contains the sequence RVIF at amino acids 239-242.

While identification of protein-protein interactions based on a single unique peptide is

challenging, and has a high rate of false positives, it is still possible to glean useful information from

these cases. This is particularly true of low abundance interactions which may be masked by the high

abundance of previously described PP1 interaction partners such as PPP1R7. In fact, one known

PPP1CC2 interacting protein PPP1R42 was found in our list of proteins identified by a single unique

peptide. One indication that there may be more legitimate PPP1CC interacting proteins amongst the low

confidence hit is the prevalence of the classical PP1 binding RVxF motif ([KR]-X(0,1)-[VI]-{P}-[FW]);

found in up to 25% of all proteins (Ceulemans &Bollen. 2006) but found in 38% (25 of 65) in our single

peptide list, a significant enrichment (p < 0.025 via χ2). One RVxF motif containing protein that stands

out as a candidate for PPP1CC2 interaction is BRWD1 (Bromodomain and WD repeat containing 1),

which when deleted, results in a male infertility phenotype very similar to that of the Ppp1cc knockout

(Philipps et al. 2008).

Validation of PPP1CC2-DDOST interaction in Testis

To validate DDOST as a PPP1CC2 interacting protein we conducted a sedimentation assay using

a recombinant his-tagged DDOST construct (Figure 2.5). The assay was conducted in mouse testis

Page 66: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

55

lysate, as this is the only tissue in which both proteins are abundantly expressed. The His-DDOST

construct utilized corresponds to amino acids 144-360 of Human DDOST with an N-terminal His tag,

which contains the PP1 docking motif; this region is highly conserved between human and mouse, with

only 5 amino acid substitutions in 217 residues. His-DDOST, bound to Ni-NTA resin, was able to

precipitate PPP1CC2 from mouse testis lysate, while the Ni-NTA resin alone was not. This reciprocal

pull-down validates the interaction between DDOST and PPP1CC2.

Immunolocalization of DDOST in mouse spermatogenic cells

DDOST is a member of the ER membrane localized Oligosaccharyltransferase (OST) complex.

To our knowledge the distribution of DDOST throughout spermatogenesis has not been characterized.

Therefore immunolocalization studies on dissociated mouse spermatogenic cells were performed. Anti-

DDOST staining displayed a punctate pattern in a range of germ cell types (Figure 2.6) that was not

visible when omitting the primary antibody (Figure 2.6C). While DDOST puncta were visible

throughout the cytoplasm, the staining was primarily concentrated around the nucleus, most likely on the

nuclear envelope as puncta were visible just outside the chromatin stained region (Figure 2.6A, arrows).

The rough endoplasmic reticulum is known to be associated with the nuclear surface in spermatogenesis

(Chemes et al. 1978) which is consistent with the predicted ER localization of DDOST. This staining

pattern was especially prevalent in spermatocytes, whereas round spermatids displayed an increase in

cytoplasmic staining (Figure 2.6A). In elongating spermatids, nuclear envelope staining was reduced

significantly especially in the dorsal and apical regions (Figure 2.6A). In a small subset of elongating

spermatids a large degree of cytoplasmic accumulation of DDOST signal into a region distant from the

nucleus was observed, which likely corresponds to the residual body in which excess cytoplasm and

organelles are sequestered for elimination from the spermatid (Figure 2.6A, asterisk). The observed

DDOST staining pattern was preserved in Ppp1cc knockout spermatogenic cells, from spermatocytes

through to the small number of morphologically abnormal elongating spermatids present in these

mutants (Figure 2.6B). However, we did not observe the residual body-like staining pattern seen in a

Page 67: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

56

Figure 2.5: Validation of PPP1CC2-DDOST interaction in mouse testis lysate. Western blot using anti-

PPP1CC probe on eluates from His-DDOST and mock pull-downs demonstrates that PPP1CC2 and

DDOST can interact in the testis. Input represents roughly 5% of total testis protein used.

Page 68: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

57

Page 69: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

58

Figure 2.6: Localization of DDOST in dissociated spermatogenic cells. Shown are spermatocytes (Sc),

round spermatids (RS) and elongating spermatids (ES) with DDOST signal localizing primarily to the

nuclear envelope region. Nuclei are indicated using DAPI. Anti-DDOST was visualized with goat anti-

rabbit Cy3 in wild-type (A) and Ppp1cc knockout (B) spermatogenic cells. Arrows indicate DDOST

puncta falling outside the nuclear stained region. Asterisk indicates cytoplasmic accumulation of DDOST

in residual body. (C) Localization of DDOST to the nuclear envelope, but not nuclear pore complexes in

wild-type spermatocytes, evidenced via comparison with a nuclear pore complex marker MAb414. +Ab

indicates inclusion of primary antibodies; -Ab indicates no primary antibody was used.

Page 70: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

59

Figure

2.6A

Page 71: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

60

Figure 2.6B

Page 72: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

61

Figure 2.6C

Page 73: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

62

small number of wild-type elongating spermatids, in any Ppp1cc spermatids. This is likely due to the

small number of these cells in present in the mutants, as the amount of DDOST present in later

spermatids appears similar to wild type cells (Figure 2.6A-B bottom rows). These results suggest that

PPP1CC2 is not required for proper DDOST localization in the testis. It should be noted that PPP1CC2 is

strongly expressed throughout the developing spermatogenic cells particularly in the cytoplasm of late

spermatocytes and spermatids (Hrabchack & Varmuza, 2007) to such a degree that co-localization

studies with DDOST would not be informative.

To confirm that DDOST localized to the nuclear envelope, spermatogenic cells were stained with

MAb414 which recognizes a family of nuclear pore complex (NPC) proteins on the nuclear envelope.

MAb414 and DDOST stained the same region of the cell around the nucleus confirming localization of

DDOST to the nuclear envelope region (Figure 2.6C). Closer examination of DDOST and MAb414

puncta shows DDOST puncta are next to, but not completely co-localized with MAb414 puncta (Figure

2.6C, inset) indicating that while localized to the nuclear envelope region, DDOST does not localize to

the NPC.

Phosphorylation of OST complex member STT3B in the mouse testis

DDOST localization in the testis does not appear to be dependent of PPP1CC2 activity. This fact

and the presence of the RVxF motif common to PP1 regulators suggests the possibility that DDOST

serves to recruit PPP1CC2 to its subcellular locale during spermatogenesis, allowing for the

dephosphorylation of similarly localized substrates. As DDOST is a member of the multi-protein OST

complex, we sought to determine if any of the other members of OST complex are Ser/Thr

phosphorylated in the testis, making them potential PPP1CC2 substrates. To test this, we performed a

phosphoproteomic analysis of the adult mouse testis, using sequential elution from IMAC (SIMAC)

phosphopeptide enrichment. Amongst the recovered peptides we identified two different overlapping

doubly phosphorylated peptides ENPPVEDpSpSDEDDKR and ENPPVEDpSpSDEDDKRNPGNLYDK

Page 74: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

63

corresponding to the STT3B gene, a member of the OST complex. The phosphorylated serine residues

map to S495 and S496 of mouse STT3B. The localization probability of both phosphosites is 100%

according to the ScaffoldPTM software, with Ascores of 1,000 being assigned to each phospho-site. In

total, these two peptides were identified 13 times throughout 3 SIMAC fractions, further strengthening

confidence in the phospho-site localization (See supplemental data for complete statistical data,

annotated spectra and fragmentation tables).

Generation of SBP-3XFLAG-PPP1CC2 chimeric mice

The original aim of the experiment was to identify PPP1CC2 interactions in a more biologically

relevant setting – the testis - by creating SBP-3XFLAG-PPP1CC2 knock-in mice, using our engineered

ES cells. Chimera production was performed by the Toronto Centre for Phenogenomics, using blastocyst

microinjection into C57BL6 host embryos. Two male and 5 female chimeras, all with a weak

contribution from the knock-in cell lineage (evidenced by coat colour), were generated from two

different cell lines. One male chimera died before the commencement of breeding, and the second died

after producing several litters. Breeding of the male chimera resulted in the production of 11 offspring

derived from the ES cells, as judged by coat colour, indicating that the ES cells were able to populate the

germline; however all were wild type and none carried the SBP-3XFLAG-PPP1CC2 allele.

2.4 Discussion

To truly understand the function of a protein in a given tissue it is critical to define its

interactome—the full complement of protein-protein interactions. Due to the recent advancements in

proteomic technologies, high throughput studies are making great inroads into defining the interactomes

of many proteins. However, one major drawback of these studies is the fact that the vast majority are

conducted in cell-lines that may not be physiologically relevant to the question at hand and as such are

not directly translatable to a protein's interactome in a specific cell type or tissue. One example of such a

tissue is the testis, which is not possible to model in tissue culture due to its complex architecture and its

Page 75: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

64

complement of post-meiotic cells. For this reason we undertook to produce a system that would be

useful in generating affinity tagged knock-in ES cells that could theoretically be used to generate

transgenic mice. The SBP-3XFLAG knock-in vector described in this paper, based on the floxin system,

will allow for the creation of transgenic tandem affinity tagged versions of any gene for which a pGT0lxf

gene trap line exists (over 24,000 different lines are currently available (Singla et al. 2010)).

The PP1 isoform PPP1CC2 is a testis-specific splice isoform that is essential in mouse

spermatogenesis (Varmuza et al. 1999). While approximately 200 PP1 interacting proteins have been

identified (Heroes et al. 2012), the interactome of PPP1CC2 in the testis requires further study. Previous

studies from our lab have sought to identify PPP1CC2 interacting proteins via approaches such as the

yeast 2-hybrid (Hrabchak & Varmuza 2004, Hrabchak et al. 2007). While these approaches allowed us

to identify several novel PPP1CC2 specific interacting proteins, to get a complete picture of the

PPP1CC2 interactome, we require a means of performing experiments directly in the testis. As shown

above, we were able to successfully produce mouse ES cell lines expressing functional tandem affinity

tagged versions of our genes of interest. The recombinant proteins expressed by our transgenic ES cell

lines not only expressed SBP-3XFLAG-PPP1CC1 and SBP-3XFLAG-PPP1CC2, but were able to

interact with a number of known PP1 interacting proteins. This demonstrates the utility of our system in

creating such modified ES cell lines, which can easily be extended to many other genes by varying only

a single cloning step. Our attempt to generate a line of transgenic mice expressing SBP-3XFLAG-

PPP1CC2 was foreshortened by the early demise of the founder male. However, the fact the chimera

sired 11 ES derived offspring (all wild type), based on coat colour, indicates that the ES line is capable of

populating the germline. Singla et al. (2010) utilized the same vector backbone and knock-in strategy,

and found that pluripotency was not affected by the process. Although the numbers are too low to draw

any definitive conclusions, the possibility that the TAP-tagged PPP1CC2 has dominant negative

properties cannot be ruled out.

Page 76: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

65

In ES cell TAP experiments we have identified and validated a novel PP1 interacting protein,

DDOST. This protein contains the classical PP1 docking motif that is found in approximately 90% of all

identified PP1 binding proteins (Hendrickx et al. 2009). This study has demonstrated that DDOST can

bind to PPP1CC2; however its ability to bind to other PP1 isoforms has yet to be tested. DDOST is a

member of the oligosaccharyltransferase complex, and catalyzes the transfer of high-mannose

oligosaccharides to nascent polypeptide chains across the ER membrane (Yamagata et al. 1997). DDOST

plays a non-catalytic role, and is essential for assembly of the OST complex (Roboti & High. 2012). One

recent clinical study found that a patient’s congenital disorder of glycosylation, with symptoms including

severe dysfunction of multiple organs and cognitive impairment was found to be due to a 22 bp deletion

and missense mutation in DDOST (Jones et al. 2012). In the rat testis, DDOST has been shown to be

expressed throughout spermatogenesis, with its highest expression found at a time point corresponding to

the beginning of spermatid formation (Luk et al. 2003). Additionally, DDOST has recently been

identified in the detergent resistant membrane fraction of both human and mouse spermatozoa, and has a

putative role in sperm-oocyte interaction and/or cell adhesion (Asano et al. 2010, Nixon et al. 2011) as

are other members of the OST complex RPN1 and RPN2. Despite this expression data, whether or not

DDOST plays a critical role in spermatogenesis remains untested.

Our results show that DDOST is expressed in a range of spermatogenic cell types and shows a

prominent punctate localization to the nuclear envelope which is especially prevalent in spermatocytes.

This is consistent with the known rough endoplasmic reticulum (RER) localization of DDOST, as the

nuclear envelope is a specific domain of the ER (Lynes & Simmen 2011) and spermatocyte nuclei are

known to be covered with a mantle of RER that can cover up to 50% of the nuclear surface (Chemes et

al. 1978). Later in spermatogenesis, localization of DDOST to the nuclear envelope appears to be

reduced, with an accumulation in the cytoplasm evident, most likely corresponding to removal via the

residual body, along with other excess cytoplasmic organelles and components being shed by the

maturing spermatid. However, even in late elongating spermatids a significant amount of DDOST

Page 77: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

66

remains present around the spermatid nucleus, and even in mature spermatozoa (Asano et al. 2010,

Nixon et al. 2011), suggesting a potential role in late spermatogenesis, or fertility.

The presence of a PP1 docking motif, its membrane bound status and the fact that its localization

is unaffected by loss of PPP1CC2 in the testis, suggests the possibility that DDOST functions as a

substrate targeting PIP. By sequestering a pool of PPP1CC2 to a specific region of the nuclear envelope,

DDOST may bring the phosphatase into regions of close contact with potential substrates. A number of

other PIPs with similar functions have been identified, including another ER protein PPP1R15A

(GADD34) (Brush et al. 2003), which has been shown to localize PP1 isoforms to the ER. If this is the

case, obvious candidate substrates for PPP1CC2 dephosphorylation are the other members of the OST

complex, such as STT3B which we showed to be phosphorylated in the testis, and has also been reported

by another study of testis phosphoproteins (Huttlin et al. 2010). However, at this stage the experiments

detailed in this study are preliminary in nature and further inquiry is necessary to determine if DDOST

plays an essential role in spermatogenesis, and to determine the biological nature of the PPP1CC2-

DDOST interaction.

In addition to the identification of DDOST as a novel PPP1CC2 interacting protein, our data

indicate a significant enrichment of the classical PP1 RVxF docking motif amongst proteins identified by

a single unique peptide in our TAP studies. While at least two unique and non-overlapping peptides are

generally required for confident identification of a protein in such datasets, the enrichment of the RVxF

motif suggests there are likely more legitimate PP1 interacting proteins within this dataset. However, as

a caveat this subset of proteins will contain a high rate of false-positives and any candidate interactors

require validation by other means. A total of 4 genes within this dataset are known to be required for

fertility in males—Brwd1, Fancl, Gapdhs, and Tial1. All of these proteins contain the classical PP1

docking RVxF motif. Brwd1 stands out as a particularly interesting candidate as its knockout phenotype

includes a decrease in post meiotic cell numbers, malformed sperm heads, acrosomal defects and

Page 78: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

67

aberrant chromatin condensation, all of which are also seen in Ppp1cc mutants (Varmuza et al. 1999,

Philipps et al. 2008, Forgione et al. 2010).

The high number of proteins identified in our dataset by single-unique peptides could be a

reflection of several factors. First, many other affinity purification-mass spectrometry (AP-MS) studies

utilize vector based expression systems for affinity tagged genes which often results in a significant

overexpression of the bait protein and thus a larger number of interactions detected. Our system utilized

the Ppp1cc promoter coupled to an IRES which will result in a lower expression level of our bait

construct. However this should reflect a more endogenous expression pattern and interactions detected

are less likely to be due to ectopic gene expression. Another potential contributing factor is our

utilization of a tandem affinity purification strategy. The use of two purification steps results in less

contaminant proteins being identified, but at a cost of fewer legitimate interactors identified, as weaker

and lower abundance interactions may be lost during the additional purification step (Chen & Gingras

2007). A more physiological explanation could lie in the nature of ES cells. Being primed for

differentiation, ES cells are thought to express more proteins, but at a lower abundance than

differentiated cell types. This could highlight an interesting utility for ES cells in interactome study, and

comparison of the relative abundance of protein-protein interactions between different cell types, could

yield considerable insight into gene function.

Supporting Information Available Online:

Supplementary Table S1 - List of all proteins identified in this study by at least one significant peptide,

excluding contaminant proteins. Table includes summary results of LC-MS/MS data, as well as presence

of RVxF motif and known infertility KO phenotypes. Table S2 - Further data on single unique peptide

LC-MS/MS hits. Table S3 - Annotated MS/MS spectrum and fragment assignments of known PPP1CC2

interacting protein PPP1R42, which was identified by a single unique peptide. Table S4 – Annotated

MS/MS spectrum and fragment assignments of BRWD1 protein, which was identified by a single unique

Page 79: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

68

peptide. Table S5 – ScaffoldPTM protein report displaying statistical information regarding STT3B

phosphosite localization. Supp File S6 – Annotated MS/MS spectra and representative fragment

assignments for STT3B phosphopeptides. This material is available free of charge via the Internet at

http://pubs.acs.org.

Page 80: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

69

Chapter 3: PPP1CC2 can form a kinase/phosphatase complex with the testis specific proteins

TSSK1 and TSKS in the mouse testis.

Author’s note: A manuscript featuring a modified version of this chapter is currently in preparation as:

MacLeod G, Shang P, Booth GT, Mastropaolo LA, Manafpoursakha N and Varmuza S (2013). PPP1CC2

can form a kinase/phosphatase complex with the testis specific proteins TSSK1 and TSKS in the mouse

testis.

Experiments were designed and conceived by myself and S. Varmuza. LA Mastropaolo, and GT Booth

were responsible for cloning GST-TSKS, GST-TSSK1, and GST-TSSK6 constructs. Production of

bacterial expressed fusion proteins was performed by me, GT Booth and N Manafpoursakha. GST-

PPP1CC2 sedimentation assay with SDS-PAGE, silver staining, and in-gel digestion was conducted by

LA Mastropaolo and me. Replicates of GST-TSKS pull down with anti-PPP1CC western blotting were

performed by me and GT Booth. Replicates of GST-TSSK6 sedimentation assay with anti-PPP1CC

western blotting were conducted by GT Booth and N Manafpoursakha. Replicates of GST-PPP1CC2

sedimentation assay with anti-TSSK2 western blotting were conducted by GT Booth and N

Manafpoursakha. P Shang generated TSKS, TSSK1 and TSSK2 antibodies and provided technical

advice for immunohistochemistry experiments. I performed all remaining experiments and was

responsible for authorship of the manuscript and figure generation with editing by S. Varmuza.

Abstract:

The mouse protein phosphatase gene Ppp1cc is essential for male fertility, with mutants

displaying a failure in spermatogenesis including a widespread loss of post-meiotic germ cells. This

phenotype is hypothesized to be attributed to the loss of the testis specific isoform PPP1CC2. To identify

PPP1CC2 interacting proteins with a function in spermatogenesis, we performed GST pull down assays

in mouse testis lysate. Amongst the identified candidate interactors was the testis specific protein kinase

TSSK1, which is also essential for male fertility. Subsequent interaction experiments have confirmed the

Page 81: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

70

capability of PPP1CC2 to form a complex with TSSK1 mediated by the kinase substrate protein TSKS.

Interaction between PPP1CC2 and TSKS is mediated through an RVxF docking motif on the TSKS

surface. Phosphoproteomic analysis of the mouse testis identified a novel serine phosphorylation site

within the TSKS RVxF motif that appears to regulate binding to PPP1CC2. Immunohistochemical

analysis of TSSK1 and TSKS in Ppp1cc mutant testis shows reduced accumulation to distinct

cytoplasmic foci and other abnormalities in their distribution consistent with the Ppp1cc mutant

phenotype. This data demonstrates a novel kinase/phosphatase complex in the testis that could play a

critical role in the completion of spermatogenesis.

3.1 Introduction

Protein phosphorylation is a key post-translational regulatory mechanism that plays a role in

countless cellular processes. Precise regulation of protein phosphorylation is carried out by the opposing

activities of protein kinases and protein phosphatases. While the mammalian genome encodes for

approximately 400 Ser/Thr kinases, it encodes only approximately 40 Ser/Thr phosphatases (Moorhead

et al. 2007, Bollen et al. 2010). Thus, many Ser/Thr phosphatases, including PP1s, obtain substrate

specificity by functioning as holoenzymes via interactions with a large and diverse array of regulatory

subunits (Hubbard &Cohen. 1993). To date almost 200 distinct PP1 interacting proteins have been

identified (Bollen et al. 2010), and it is hypothesized that many more exist.

Spermatogenesis is no exception to the importance of protein phosphorylation based regulatory

processes. By searching the Gene Ontology database, it is observed that 14 genes are linked to both the

Ser/Thr Protein Kinase molecular function (GO:0004674) and the spermatogenesis biological process

(GO:0007283), including all six members of the testis-specific Ser/Thr Kinase (TSSK) family. Using a

similar database search for Ser/Thr Protein Phosphatase molecular function (GO:0004722), no genes are

found to overlap with the spermatogenesis biological process; however, at least 3 Ser/Thr protein

phosphatase genes, Ppp1cc (Varmuza et al. 1999) Mtmr2 (Bolino et al., 2004) and Ppm1d (Choi et al.

Page 82: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

71

2002), produce male infertility phenotypes when deleted, indicating that functional annotation in this

database is not complete.

Ppp1cc is a member of the PP1 family of protein phosphatases that encodes two splice isoforms,

the ubiquitous Ppp1cc1 and the testis-specific Ppp1cc2 (Okano et al. 1997). When Ppp1cc is deleted by

targeted mutagenesis, the only observable phenotypic consequence is homozygous male infertility, due to

a failure of spermatogenesis, reminiscent of the common human condition non-obstructive azoospermia

(Varmuza et al. 1999). Inside the seminiferous tubules a widespread loss of germ cells is evident, most

prominently in post-meiotic spermatids, leading to a breakdown of the spermatogenic cycle (Varmuza et

al. 1999, Forgione et al. 2010). The few surviving germ cells feature a range of morphological

abnormalities, including those involving meiosis, chromatin condensation, acrosome formation and

mitochondrial organization (Varmuza et al. 1999, Chakrabarti et al. 2007, Forgione et al. 2010). In the

mouse testis, a number of different proteins have been shown to interact with PPP1CC2. These include

both isoform specific interactions such as SPZ1 and Endophilin B1t (SH3GLB1, testis specific isoform)

(Hrabchak & Varmuza 2004, Hrabchak et al. 2007) and more numerously, proteins that have the ability

to interact with multiple PP1 isoforms, such as PPP1R11 (Cheng et al., 2009). In addition, another

testis-specific protein, Testis Specific Ser/Thr Kinase Substrate (TSKS) was bioinformatically predicted

to interact with PP1 based on the presence of a high affinity PP1 docking motif and in vitro experiments

confirmed that a TSKS fragment was capable of interacting with PPP1CA (Hendrickx et al. 2009). The

ability of full length TSKS to bind to other PP1 isoforms such as PPP1CC2 was not tested.

In a search for additional PPP1CC2 interacting proteins, we performed pull down assays in

mouse testis protein extracts using GST-PPP1CC1 and GST-PPP1CC2 as bait. Among the identified

proteins was the testis-specific Ser/Thr kinase TSSK1, which is essential for spermatogenesis in the

mouse (Xu et al. 2008, Shang et al. 2010). We describe experiments which demonstrate a clear link

between TSSK1, TSKS and PPP1CC2 in mouse testis.

Page 83: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

72

3.2 Materials and Methods

Mouse Testis Protein Extract Preparation

Whole mouse testes were homogenized in cold protein extraction buffer (10% (v/v) glycerol,

50mM HEPES-NaOH pH 8.0, 150 mM NaCl, 2 mM EDTA, 0.1% (v/v) NP-40, 1 mM dithioreitol, 10

mM NaF, 0.25 mM sodium orthovanadate, 50 mM beta-glycerol phosphate) supplemented with Sigma

protease inhibitor cocktail) using a Dounce homogenizer. After homogenization, samples were incubated

on ice for 10 minutes, followed by centrifugation at a speed of 10,000 x g for 10 minutes at 4°C to

remove non-soluble material. All animal protocols were approved by the Canadian Council on Animal

Care.

GST and His-tag Pull Down Assays

The PPP1CC1 coding sequence was PCR amplified from the pGEM7zf plasmid using the

forward primer 5’-GGCGGATCCGCGATGGC`-3’ and the reverse primer 5’-

GCTATGTTAGAATTCCCAACCAGGC-3’ and ligated into the BamHI and EcoRI sites of the pGEX-

6P-2 plasmid (Amersham). GST-PPP1CC2, and GST containing plasmids were cloned previously and

fusion proteins produced as previously described (Hrabchak & Varmuza 2004, Hrabchak et al. 2007).

500 μg mouse testis lysates were incubated with approximately 2 μg GST-fusion proteins for 2-4 hours at

4° C with gentle rocking. Samples were centrifuged at 1,500 x g for 2 minutes at 4° C, and GST-fusion

protein beads were washed 3 times with 500 μL lysis buffer, after centrifugation steps. For pull down

experiments with LC-MS/MS analysis, testis lysate was initially pre-cleared via incubation with

glutathione agarose beads, and recombinant proteins were subjected to additional washes, both before

and after incubation with testis lysate. Recombinant human His-TSSK1 (Millipore cat. number 14-670)

was bound to Ni-NTA resin and incubated with mouse testis lysates using the same protocol as for GST-

fusions. In vitro, lysate-free pull down assays were based on a previously a described protocol (DeWever

et al. 2012), where 500 ng of unbound His-TSSK1 was incubated with GST, GST-PPP1CC2 or GST-

Page 84: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

73

TSKS bound to glutathione agarose beads in 250 μL binding buffer (25 mM Tris pH 7.5, 5% glycerol

(v/v), 150 mM NaCl, 0.5% NP-40 (v/v) and 10 mM imidazole) for 2 hours at 4°C with rocking. Beads

were then spun down at 1,500 x g for 1 min at 4°C and washed 3 times with 1 mL of binding buffer. For

all experiments, bound proteins were eluted by boiling in SDS-PAGE sample buffer (50mM Tris, 2%

(w/v) SDS, 0.1% (w/v) bromophenol blue, 10% (v/v) glycerol, 25mM β-mercaptoethanol), and analyzed

by SDS-PAGE followed by silver staining (FOCUS-FASTsilver™, G Biosciences) or western blotting

using standard protocols.

In-gel digestion of silver stained gel slices

Gel slices were excised from silver stained gel washed with HPLC grade water. 200 μL of

acetonitrile was added to each gel slice and incubated at room temperature for 15 minutes with mixing.

Slices were then reduced with 10 mM dithioreitol in 100 mM ammonium bicarbonate for 30 minutes at

50°C, followed by removal of the reduction solution and wash with acetonitrile. Alkylation was

performed using 55 mM iodoacetic acid in 100 mM ammonium bicarbonate for 20 minutes in darkness

at room temperature. Alkylation solution was removed and gel slices washed in ammonium bicarbonate

and dried. Gel slices were then incubated with proteomics grade trypsin (Sigma-Aldrich, T6567) in 50

mM ammonium bicarbonate, 5 mM CaCl2 on ice for 45 minutes, then overnight at 37°C. After

deactivating trypsin with trifluoroacetic acid the supernatant was collected and 100 μL of 60% (v/v)

acetonitrile added to the gel slices and incubated with rocking for 10 minutes. The supernatant was then

collected and combined with that of the previous step and dried in a speed vac.

LC-MS/MS analysis and protein/peptide identification

LC-MS/MS analysis was performed by the Advanced Protein Technology Centre (Toronto, ON,

Canada http://www.sickkids.ca/Research/APTC/index.html). Peptides were loaded onto a 150 μm ID

pre-column (Magic C18, Michrom Biosciences) at 4 μL/min and separated over a 75 μm ID analytical

column packed into an emitter tip containing the same packing material. The peptides were eluted over

Page 85: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

74

60 min. at 300 nl/min using a 0 to 40% (v/v) acetonitrile gradient in 0.1% (v/v) formic acid using an

EASY n-LC nano-chromatography pump (Proxeon Biosystems, Odense Denmark). The peptides were

eluted into a LTQ-Orbitrap hybrid mass spectrometer (Thermo-Fisher, Bremen, Germany) operated in a

data dependant mode. MS was acquired at 60,000 FWHM resolution in the FTMS and MS/MS was

carried out in the linear ion trap. 6 MS/MS scans were obtained per MS cycle. The Raw data was

searched using Mascot (Matrix Sciences, London UK). Tandem mass spectra were extracted, charge state

deconvoluted and deisotoped by BioWorks version 3.3. All MS/MS samples were analyzed using Mascot

(Matrix Science, London, UK; version Mascot). Mascot was set up to search the NCBInr_20110515

database for gel slices peptide samples and NCBInr_20110813 database for phosphopeptide samples

(both selected for Mus musculus) for trypsin digestion. Mascot was searched with a fragment ion mass

tolerance of 0.40 Da and a parent ion tolerance of 20 PPM for phosphopeptide samples, and a fragment

ion mass tolerance of 0.50 Da and 3.0 Da peptide tolerance for gel slice peptide samples. Iodoacetamide

derivative of cysteine was specified as a fixed modification, with the following variable modifications:

Pyro-glu from E of the N-terminus, s-carbamoylmethylcysteine cyclization of the N-terminus,

deamidation of asparagine and glutamine, oxidation of methionine, acetylation of the n-terminus for gel

slice peptide samples with phosphorylation of serine, threonine and tyrosine were as additional variable

modifications in phosphopeptide samples. Scaffold (version Scaffold_3.1.2, Proteome Software Inc.,

Portland, OR) was used to validate MS/MS based peptide and protein identifications. Peptide

identifications were accepted if they could be established at greater than 95.0% probability as specified

by the Peptide Prophet algorithm (Keller et al. 2002). Protein identifications were accepted if they could

be established at greater than 95.0% probability and contained at least 2 identified peptides (or 1 for

phosphopeptide samples). Protein probabilities were assigned by the Protein Prophet algorithm

(Nesvizhskii et al. 2003). Proteins that contained similar peptides and could not be differentiated based

on MS/MS analysis alone were grouped to satisfy the principles of parsimony.

PCR Mutagenesis

Page 86: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

75

For generation of RVXF motif mutants, PCR mutagenesis was performed using primers

complementary to the relevant region of the TSKS coding sequence with the exception of required non-

synonymous base pair substitutions. Primer sequences were as follows: for the KAASA mutation

forward primer: 5’-CGAAAAAAAAGAAGGCTGCGTCCGCCCATGGGTGGAGCCCCG-3’ and

reverse primer: 5’-CGGGGCTCCACCCCTAGGGCGGACGCAGCCTTCTTTTTTTTCG-3’, for the

KAVEF mutation forward primer: 5’-

CGAAAAAAAAGAAGGCTGTGGAGTTCCATGGGGTGGAGCCCCG-3’ and reverse primer: 5’-

CGGGGCTCCACCCCATGGAACTCCACAGCCTTCTTTTTTTTCG-3’. Primers were used in PCR

amplification of the pGEX-TSKS plasmid with PFU polymerase (BioBasic) and resulting reaction

product was purified and digested with DpnI restriction enzyme to digest template plasmid. Digested

DNA was transformed into DH5α for selection, and the presence of mutations was verified via plasmid

sequencing. Plasmids containing TSKS coding sequences with mutated RVxF motifs were then used to

produce GST-fusion proteins as outlined above.

Antibodies

Goat anti-PPP1CC (N-19, Santa Cruz Biotechnology), which recognises both PPP1CC1 and

PPP1CC2 was used at a dilution of 1:500 for western blotting, with donkey anti-goat HRP secondary

(Santa Cruz Biotechnology) at a 1:5000 dilution. Guinea pig anti-TSKS and TSSK1 antibodies (Shang

et al. 2010) were used at dilutions of 1:500 for western blotting in cell lysate samples, and 1:5000 using

purified fusion proteins, with goat anti-guinea pig HRP secondary (Jackson Immunoreagents) at a

dilution of 1:5000. For immunohistochemistry, the same anti-TSKS and TSSK1 primary antibodies were

used at a 1:500 dilution, with Cy3 conjugated AffiniPure goat anti-guinea pig IgG (Jackson

Immunoreagents) at a 1:5000 dilution. Guinea pig anti-TSSK2 (Shang et al. 2010) was used at a dilution

of 1:2000 for western blotting with the same secondary antibody conditions as other Guinea pig

antibodies listed.

Page 87: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

76

Phosphopeptide Enrichment

Adult mouse testes were decapsulated and germ cell suspensions were produced as previously

described (Henderson et al. 2011, MacLeod & Varmuza 2012). Germ cells were lysed in 400μL of 7M

Urea, 2M Thiourea, 4% CHAPs (w/v), 40mM Tris, reduced with 20mM dithioreitol for 1 hour at room

temperature and alkylated with 40 mM iodoacetic acid for 35 minutes at room temperature in darkness.

Germ cell proteins were precipitated with acetone and dried using a speed vac, followed by resuspension

in a 1M urea, 50 mM ammonium bicarbonate solution containing 10μg of proteomics grade trypsin.

Digestion was carried out over night at 37°C, and at completion the solution was acidified with 1% (v/v)

formic acid and centrifuged to remove insoluble material. One quarter of the sample was used for

phosphopeptide enrichment via sequential elution from IMAC (SIMAC) using the protocol from

Thingholm et al. (2009), but with 50μL of TiO2 Mag Sepharose (GE Healthcare) substituted for TiO2

phosphopeptide enrichment steps. Phosphopeptide enriched samples were then analyzed by LC-MS/MS

(see above).

Immunohistochemistry

After removal of the tunica albuginea, wild-type and Ppp1cc mutant testes were fixed in 4%

(w/v) paraformaldehyde overnight at 4°C and dehydrated using a graded series of ethanol solutions and

embedded in paraffin. 7μm sections were dewaxed, hydrated and subjected to antigen retrieval by

heating in 10 mM Sodium Citrate, 0.05% (v/v) Tween20, 1XPBS. Sections were permeabilized with

0.01% (v/v) Triton-X in PBS, and blocked in 10% (w/v) goat serum, 1% BSA (w/v), 0.01% (v/v)

Tween20, PBS solution. Primary antibody incubations were performed in antibody dilution buffer (5%

(w/v) goat serum, 1% (w/v) BSA, 0.01% (v/v) Tween20, PBS) overnight at using the conditions

indicated above. Secondary antibody incubations were performed in antibody dilution buffer for 2 hours

in the dark. Nuclei were then stained with DAPI and sections were mounted in 50% (v/v) glycerol for

viewing with an Olympus BX60 microscope. Images were captured using Cool Snap software and a

Page 88: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

77

CCD camera (RSPhotometrics). Images were adjusted for brightness and contrast using Photoshop 6.0

(Adobe) and then merged using ImagePro 4.1.

3.3 Results

Testis-specific Ser/Thr kinase TSSK1 interacts with GST-PP1CC1 and GST-PPP1CC2 in mouse testis

lysate.

In exploratory assays designed to identify PPP1CC interacting proteins in the mouse testis,

recombinant GST-PPP1CC1 and GST-PPP1CC2 were purified and used in pull down assays with mouse

testis protein lysates. Proteins were then subjected to SDS-PAGE, followed by silver staining. Selected

gel slices were excised from both the GST-PPP1CC1 and GST-PPP1CC2 lanes. In one such experiment,

a band of approximately 45 kDa containing several visible bands was excised. After trypsin digestion,

proteins present in the gel slices were identified via LC-MS/MS with MASCOT database searching.

Common LC-MS/MS contaminant proteins such as keratins, actins, and tubulins were removed from the

list of candidate interactors. Remaining proteins for which ≥ 2 unique peptides were identified are shown

in Table 3.1 (Appendix A.4.2). All four of the remaining proteins, UQCRC2, FADS2, SCCPDH and

TSSK1, have been shown to be expressed in the mouse testis (Kueng et al. 1997, Stoffel et al. 2008, Guo

et al. 2011) and deletions of Fads2 and Tssk1/Tssk2 have been shown to disrupt spermatogenesis (Stoffel

et al. 2008, Xu et al. 2008, Shang et al. 2010). FADS2 was the only protein that appeared in only the

GST-PPP1CC2 gel band, however fewer peptides were assigned to this protein compared to the other

identified proteins. Experiments to determine if FADS2 is a legitimate PPP1CC2 interacting protein in

the testis remain at a preliminary stage. Tssk1 is a testis-specific kinase gene that plays a role in

spermatogenesis, and thus stood out to us as a particularly interesting candidate PPP1CC2 interactor in

the testis. To test if TSSK1 actually bound specifically to the GST-PPP1CC2, the pull down assay in

testis lysate was repeated, this time followed by western blotting for TSSK1. The results of this

experiment confirmed the initial pull-down, with the GST-PPP1CC2 bait, but not GST alone, being able

Page 89: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

78

Table 3.1: Testis proteins identified in a SDS-PAGE gel band after sedimentation by GST-

PPP1CC1 and GST-PPP1CC2. Only proteins identified by at least 2 significant unique peptides are

included. Common contaminant proteins i.e. keratin and tubulin were removed from the list. Protein IDs

are from UniProtKB/Swiss-Prot. Sequence coverage combines all unique peptides from GST-PPP1CC1

and GST-PPP1CC2 pull downs.

Page 90: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

79

Unique Peptides

Gene

Symbol Name

Protein

ID

GST-

PPP1CC1

GST-

PPP1CC2

Sequence

Coverage

Uqcrc2 Cytochrome b-c1 complex subunit 2, mitochondrial Q9DB77 12 10 35.30%

Tssk1 Testis-specific serine/threonine-protein kinase 1 Q61241 10 2 25%

Sccpdh Saccharopine dehydrogenase-like oxidoreductase Q8R127 7 5 16%

Fads2 Fatty acid desaturase 2 Q9Z0R9 0 2 5%

Page 91: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

80

Figure 3.1: PPP1CC2 interacts with both TSSK1 and TSKS in the testis. GST and His-tag pull down

assays followed by SDS-PAGE and western blotting using the indicated antibodies. (A) Recombinant

mouse GST-PPP1CC2 successfully precipitates TSSK1 from mouse testis protein extract, while GST

alone does not. (B) Ni-NTA bound 6His-tagged human TSSK1 successfully precipitates PPP1CC2 from

mouse testis protein extract, while Ni-NTA resin alone does not. (C) Ni-NTA bound 6His-tagged human

TSSK1 precipitates its substrate TSKS from mouse testis protein extract, while Ni-NTA resin alone does

not. (D) Recombinant mouse GST-PPP1CC2 successfully precipitates full length TSKS from mouse

testis protein extract, while GST alone does not. (E) Recombinant mouse GST-TSKS successfully

precipitates PPP1CC2 from mouse testis protein extract, while GST alone does not.

Page 92: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

81

Page 93: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

82

to pull down TSSK1 (Figure 3.1a). Reciprocal pull down experiments were performed using His-TSSK1

bait, which successfully precipitated PPP1CC2 from mouse testis lysate (Figure 3.1b). Additionally a

known substrate of TSSK1, testis-specific kinase substrate (TSKS) was successfully

precipitated by His-TSSK1, verifying the functionality of the fusion protein (Figure 3.1c).

Testis-specific kinase substrate, TSKS interacts with PPP1CC2 via an RVxF docking motif

In a previously published study, the TSSK1 substrate TSKS was bioinformatically predicted to

be a PP1 interacting protein, and subsequent in vitro experiments showed that a TSKS fragment was

capable of interacting with PPP1CA (Hendrickx et al. 2009), the only PP1 isoform assayed. This

prediction was based on the presence of a PP1 docking motif, known as an RVxF motif, in the TSKS

amino acid sequence. In the mouse, this motif has the sequence KAVSF in amino acid positions 51-55.

This suggests that PPP1CC2 and TSSK1 could potentially share a common interactor in the testis, which

may mediate their association. To test whether the PPP1CC2 isoform could interact with the full-length

native TSKS in the testis, proteins pulled down by GST-PPP1CC2 were subjected to western blotting for

TSKS. The results of this experiment indicate that the PPP1CC2 isoform can bind to full-length TSKS

in the testis (Figure 3.1d). The anti-TSKS signal observed via western blot showed a doublet which is

consistent with previously published data (Shang et al. 2010). Furthermore, a reciprocal pull down assay

showed that GST-TSKS was able to bind native PPP1CC2 in mouse testis lysate (Figure 3.1e),

confirming the interaction between these two testis proteins. The GST tag on its own was unable to

precipitate either PPP1CC2 or TSKS (Figure 3.1d-e). To verify that interaction with PPP1CC2 was

dependent upon the RVxF motif on the TSKS surface, we produced fusion proteins containing TSKS

with a mutated RVxF motif, changing the sequence from KAVSF to KAASA (GST-KAASA). When

GST-KAASA was incubated with mouse testis lysate, it was unable to precipitate a detectable quantity of

PPP1CC2 as compared to unaltered GST-TSKS (Figure 3.2). Furthermore, GST-KAASA was still able

to precipitate the known TSKS interactor TSSK2, indicating that the general structure of the TSKS

protein was not affected by the RVxF motif mutation (Figure 3.2). A second mutation KAVEF, gave

Page 94: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

83

Figure 3.2: The TSKS RVxF motif is required for interaction with PPP1CC2. GST-fusion proteins

corresponding to unaltered TSKS protein, RVxF mutant KAASA and RVxF phosphoserine mimic

KAVEF were incubated with mouse testis lysate and assayed via western blotting for interaction with

PPP1CC2 (top) and TSSK2 (bottom). Unaltered TSKS interacts with PPP1CC2 in the testis, while

KAASA and KAVEF constructs do not. All TSKS constructs interact with TSSK2, demonstrating no

major change in protein architecture in mutant constructs.

Page 95: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

84

Page 96: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

85

similar results, which will be discussed further below. This experiment confirms that TSKS and

PPP1CC2 interact through the RVxF docking motif. To date, attempts at detecting an interaction between

PPP1CC2 and two additional TSSK isoforms, TSSK2 and TSSK6 via similar pull down assays have

been unsuccessful (data not shown), indicating that possibility that PPP1CC2 may only interact with the

TSSK1 isoform.

TSKS is phosphorylated on at least two different serine residues in the mouse testis including the PP1

docking motif

Previous experiments have shown TSKS to be phosphorylated on the serine 281 residue in the

mouse testis (Xu et al. 2008) which is hypothesized to be the target site of TSSK1 phosphorylation. In

an experiment aimed at identifying novel phosphorylation sites in the mouse testis, we performed

sequential elution from IMAC (Thingholm et al. 2008) (SIMAC) phosphopeptide enrichment followed

by LC-MS/MS on proteins extracted from adult germ cell suspensions (see Appendix A.4.3 and A.4.4 for

further data on site assignment). MASCOT database searching of the resultant tandem mass spectra was

used to map phosphorylation sites. Amongst the identified phosphopeptides were two unique peptides

that mapped to TSKS with greater than 95% confidence. The first peptide, which was identified in both

the IMAC and TiO2 fractions was the previously known TSKS phosphopeptide

HGLSPATPIQGcSGPPGS*PEEPPR which was phosphorylated on the serine 281 residue (best

Mascot delta score = 26.6, Ascore = 94.9). The second phosphopeptide mapped to TSKS was

AVS*FHGVEPR, which represents a novel TSKS testis phosphorylation site at the serine 54 residue

(Mascot delta score = 30.9, Ascore = 1,000). This novel phosphopeptide was identified in the IMAC

fraction only. According to the Phosphosite plus database (Hornbeck et al. 2012), this phosphopeptide

has previously been identified in human Jurkat cell, T cell leukemia model, but not in the mouse, or

specifically the testis. Interestingly, this phosphorylated serine residue is found within the PP1 docking

motif KAVSF.

Page 97: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

86

Phosphorylation of the PP1 docking motif in TSKS likely inhibits interaction with PPP1CC2

Previous research with other PP1 interacting proteins has indicated that phosphorylation within

and next to the RVxF docking motif can inhibit interaction with the PP1 catalytic subunit (Beullens et al.

1999, McAvoy et al. 1999, Liu & Brautigan 2000, Bollen 2001, Grallert et al. 2013). To test whether

this was the case for TSKS we produced a GST fusion protein in which the serine residue of the RVxF

docking motif (KAVSF) was mutated to glutamate (KAVEF) to mimic serine phosphorylation. When

this phospho-mimic fusion protein (GST-KAVEF) was incubated with mouse testis lysate, no pull down

of PPP1CC2 was observed, although there was no reduction in binding to TSSK2, indicating

preservation of protein structure (Figure 3.2). This experiment indicates that phosphorylation of the

RVxF motif in TSKS likely inhibits interaction with PPP1CC2 in the testis.

The interaction between PPP1CC2 and TSSK1 is not direct

In vitro experiments have previously demonstrated direct interactions between TSKS and PP1

fragments, as well as between TSKS and TSSK1 (Kueng et al. 1997, Hendrickx et al. 2009). Thus, we

sought to determine if there was also a direct interaction between PPP1CC2 and TSSK1. To do this

purified His-TSSK1 was incubated with GST-PPP1CC2 coupled to glutathione agarose in an in vitro

setting without the presence of any cell lysate (Figure 3.3). GST-PPP1CC2 was not able to precipitate

His-TSSK1 in our experiment, nor was GST alone. Conversely, a direct interaction was detected

between GST-TSKS and His-TSSK1. This experiment indicates that the interaction between PPP1CC2

and TSSK1 is not direct, and is therefore likely mediated by another factor, such as TSKS which directly

interacts with both.

TSKS and TSSK1 localization is impaired in Ppp1cc mutant seminiferous tubules

In the seminiferous epithelium both TSSK1 and TSKS are expressed in the cytoplasm of

elongating spermatids, with prominent accumulation of both at distinct cytoplasmic foci (Shang et al.

Page 98: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

87

Figure 3.3: The interaction between PPP1CC2 and TSSK1 is not direct. In vitro pull-down assay

wherein recombinant GST, GST-PPP1CC2 or GST-TSKS bound to glutathione Sepharose was incubated

in the presence (+) or absence (-) of purified His-TSSK1 followed by sedimentation, SDS-PAGE and

western blotting with a polyclonal antibody directed against TSSK1. Only GST-TSKS successfully

precipitated His-TSSK1 demonstrating no direct interaction between PPP1CC2 and TSSK1.

Page 99: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

88

Page 100: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

89

2010). In response to targeted deletion of Tssk1 and Tssk2, the level of TSKS expression remains similar

in the cytoplasm of elongating spermatids, but its accumulation in distinct foci is lost (Shang et al. 2010)

suggesting regulation by TSSK1 (and/or TSSK2) is required for this punctate expression pattern. To test

whether PPP1CC2 also plays a role in regulating TSKS localization, as well as TSSK1 localization, we

performed immunohistochemistry on testis sections from wild-type and Ppp1cc knockout mice. Mouse

spermatogenesis can be divided into twelve different stages that arise in a cyclical fashion in the

seminiferous tubules. It should be noted that the loss of Ppp1cc results in a bottleneck in

spermatogenesis, as well as a prominent loss of spermatids, making precise staging of seminiferous

tubules challenging. In wild-type seminiferous tubules, the previously described punctate expression

pattern was observed for TSKS (Figure 3.4a-d). This punctate expression pattern is stage specific, as the

level of accumulation into distinct foci varies between seminiferous tubules. Strong TSKS expression

throughout the cytoplasm begins in stage IX (Figure 3.4d), with emergence of foci amongst the

cytoplasmic staining by stage I (Figure 3.4a). By stage IV, only distinct cytoplasmic foci are evident

(Figure 3.4b), which are no longer visible by stage VII (Figure 3.4c). In Ppp1cc mutant seminiferous

tubules all of these localization patterns can be found; however there are statistically significant

differences in their frequencies (Figure 3.4e-h, Table 3.2). In our analysis, wild-type tubules exhibited

punctate staining with limited general cytoplasmic staining (Figure 3.4b) 39% of the time and an absence

of staining 24% of the time (n=100; Figure 3.4c). In contrast only 9% of Ppp1cc mutant tubules

displayed the punctate staining pattern (Figure 3.4f) and 51% displayed an absence of TSKS signal,

which represent statistically significant differences from wild-type (p < 0.001; Figure 3.4g). In addition

to the quantitative differences, there were several qualitative changes in TSKS localization commonly

observed in Ppp1cc mutant tubules. These defects included isolated elongating spermatids showing

strong staining throughout the cytoplasm in tubules that otherwise contained a punctate expression

pattern (Figure 3.4f, inset), as well as tubules showing a large number of cytoplasmic foci instead of

diffuse staining seen in wild-type counterparts (Figure 3.4a, e, inset).

Page 101: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

90

Figure 3.4: TSKS localization is impaired, but not abolished in Ppp1cc mutant seminiferous

tubules. Immunohistochemical analysis was performed on wild-type (A-D) and Ppp1cc mutant (E-H)

testes using a polyclonal antibody for TSKS (red) and nuclear staining using DAPI (blue). Shown are

representative seminiferous tubules at four different approximate time points of the spermatogenic cycle.

For wild-type tubules approximate stages depicted are: (A) stage I, (B) stage IV, (C) stage VII, (D) stage

IX. Degeneration of seminiferous tubules in Ppp1cc mutant tubules makes staging challenging but

depicted mutant tubules were matched as closely as possible to wild-type counterparts. Numbers in the

bottom right corner of each panel indicate the percentage of tubules that exhibit the depicted staining

pattern. Asterisks indicates values significantly different than wild-type (p < 0.05). Arrow indicates

paired TSKS puncta in wild-type spermatids that are abnormal in Ppp1cc mutant samples.

Page 102: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

91

Page 103: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

92

As was previously reported, the expression pattern for TSSK1 in the wild-type testis is very similar to

that of TSKS (Shang et al. 2010) (Figure 3.5a-d). We observed strong cytoplasmic staining beginning in

stage VIII (step 8 spermatids; Figure 3.5c), one stage before the onset of TSKS expression. Foci were

evident amongst the cytoplasmic staining by stage X (Figure 3.5d), with only cytoplasmic foci visible by

stage II (Figure 3.5a). TSSK1 staining was not visible in wild-type spermatids at stage VII (Figure 3.5b).

Again, similar to TSKS we observed all of these staining patterns in Ppp1cc mutant seminiferous tubules

(Figure 3.5e-h, Table 3.2), but frequencies differed with the fully developed punctate staining pattern

observed in only 11% of mutant tubules (44% in wild-type, p<0.001; Figure 3.5a, e) and 41% of tubules

lacked TSSK1 signal (14% in wild-type, p<0.001; Figure 3.5b, f). There was also a statistically

significant increase in the proportion of tubules displaying TSSK1 foci with accompanying signal

throughout the cytoplasm with this pattern being visible in 33% of Ppp1cc mutant tubules compared to

16% in wild-type (p<0.01; Figure 3.5d,h). Qualitative abnormalities in TSSK1 staining pattern similar to

those described for TSKS were also observed in Ppp1cc mutant seminiferous tubules. Clouds of

cytoplasmic staining were evident alongside developed foci (Figure 3.5e, inset), and aggregates of

numerous foci (Figure 3.5h, inset) as opposed to pairs of foci observed in wild-type tubules (Figure 3.5d,

inset). The results of this experiment indicate that both TSKS and TSSK1 are able to achieve correct

localization in the absence of PPP1CC isoforms; however they do so at a lower frequency and with

several observable defects. As was mentioned in the previous chapter, anti-PPP1CC staining in the testis

is very strong and found throughout the cytoplasm (Hrabchack et al., 2007) to the extent that

examination of PPP1CC2 localization with TSSK1 and TSKS would not be informative.

3.4 Discussion

The necessity of the protein phosphatase gene Ppp1cc for completion of spermatogenesis in mice

is well established (Varmuza et al. 1999). However, the precise role in this process, particularly that of

the testis specific isoform PPP1CC2, remains unknown. The multitude of defects within the Ppp1cc

Page 104: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

93

Figure 3.5: TSSK1 localization is impaired but not abolished in Ppp1cc mutant seminiferous

tubules. Immunohistochemical analysis was performed on wild-type (A-D) and Ppp1cc mutant (E-H)

testes using a polyclonal antibody for TSSK1 (red) and nuclear staining using DAPI (blue). Shown are

representative seminiferous tubules at four different approximate time points of the spermatogenic cycle.

For wild-type tubules approximate stages depicted are: (A) stage II, (B) stage VII, (C) stage VIII, (D)

stage X. Degeneration of seminiferous tubules in Ppp1cc mutant tubules makes staging challenging but

depicted mutant tubules were matched as closely as possible to wild-type counterparts. Numbers in the

bottom right corner of each panel indicate the percentage of tubules that exhibit the depicted staining

pattern. Asterisks indicates values significantly different than wild-type (p < 0.05). Arrow indicates

paired TSSK1 puncta in wild-type spermatids that are abnormal in Ppp1cc mutants.

Page 105: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

94

Page 106: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

95

Table 3.2 Quantitative evaluation of TSKS and TSSK1 staining patterns in wild-type and Ppp1cc

mutant seminiferous tubules. One hundred randomly selected seminiferous tubules were classified into

one of four staining patterns for both wild-type and Ppp1cc mutant testis samples. Asterisk indicates

values significantly different (p ≤ 0.05) from wild-type.

Page 107: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

96

TSKS Staining Pattern

Genotype Blank Cytoplasmic Cytoplasmic + Puncta Puncta

Wild-type 24 21 16 39

Ppp1cc KO *51 18 22 *9

TSSK1 Staining Pattern

Genotype Blank Cytoplasmic Cytoplasmic + Puncta Puncta

Wild-type 14 26 16 44

Ppp1cc KO *41 15 *33 *11

Page 108: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

97

mutant seminiferous epithelium suggests the possibility of pleiotropic functions, with defects in both

meiotic and post-meiotic germ cells being evident (Varmuza et al. 1999, Forgione et al. 2010). In an

effort to learn more about the function(s) of PPP1CC2 in spermatogenesis we performed GST pull down

assays to identify PPP1CC2 interacting proteins in the testis. Amongst the identified proteins was the

testis-specific kinase TSSK1, which is also required for male fertility, although with a phenotype quite

different from that of the Ppp1cc deletion (Xu et al. 2008, Shang et al. 2010). While loss of Ppp1cc

results in a severe impairment in spermatogenesis including the widespread loss of post-meiotic germ

cells and a bottleneck in the spermatogenic cycle (Varmuza et al. 1999, Forgione et al. 2010), the loss of

the Tssk1 and Tssk2 genes results in a much milder phenotype with little/no loss of spermatogenic cells

(Shang et al. 2010). This difference in phenotypic severity is consistent with a requirement for Ppp1cc

earlier in spermatogenesis than Tssk1/2, which is reflected by the expression patterns of these genes in

the developing spermatogenic cells. The PPP1CC2 is expressed throughout spermatogenesis and at a

high level from the meiotic pachytene spermatocyte stage onward (Hrabchak &Varmuza. 2004).

Conversely TSSK1 and TSSK2 are only expressed in post-meiotic spermatids (Li et al. 2011). Despite

these differences, the time point of peak PPP1CC2 expression levels in the testis overlap with that of

TSSK1/2 and there are several key similarities between the knockout phenotypes. Both mutants exhibit

a significant reduction in the number and motility of epididymal spermatozoa as well as prominent

defects in the organization of the mitochondrial sheaths (Varmuza et al. 1999, Chakrabarti et al. 2007,

Shang et al. 2010). The mitochondrial sheath is a tight helical bundle of mitochondria that surrounds the

sperm flagellum providing energy for motility (Fawcett, 1975). In Tssk1/2 knockout spermatids,

assembly of the mitochondrial sheath is impaired with the mitochondria forming clusters in the proximal

region of the sperm tail, and failing to form a compact, elongated sheath (Shang et al., 2010). In the few

remaining Ppp1cc knockout spermatozoa similar defects are visible; mitochondrial sheaths generally fail

to form a compact helical structure and often do not extend to the distal region of the sperm tail

(Chakrabarti et al., 2007b, Soler et al., 2009). Ppp1cc2 knock-in mice with low levels of transgene

expression (>50% of heterozygous level) are able to rescue the loss of post-meiotic germ cells found in

Page 109: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

98

Ppp1cc knockouts, but are still infertile and mitochondrial sheath abnormalities are still visible (Soler et

al., 2009, Sinha et al., 2012). Taken together all of this evidence points to a functional link between

PPP1CC2 and TSSK1 late in spermiogenesis, after the time point where PPP1CC2 is first required, and

suggests that PPP1CC2 does play a role in multiple events in spermatogenesis. This pleiotropic function

is not out of character for a protein like PPP1CC2, as the PP1 family of protein phosphatases are

involved in numerous cellular functions and interact with a multitude of different proteins.

The results of this study demonstrate that the interaction between PPP1CC2 and TSSK1 in the

testis is indirect, and likely mediated by a common interactor, TSKS. This multiprotein complex thus

contains both a protein kinase and phosphatase, and all three proteins are known to be phosphorylated in

the testis (Kueng et al. 1997, Huang & Vijayaraghavan 2004, Jaleel et al. 2005). We have shown

conclusively that interaction with PPP1CC2 is dependent on the presence of a PP1 docking RVxF motif

in the N-terminal region of TSKS (amino acids 51-55), which was not surprising given that TSKS was

bioinformatically predicted to be a PP1 interacting protein due to the presence of this motif (Hendrickx et

al. 2009). Previous studies have demonstrated that the N-terminal region of TSKS is also required for

interaction with TSSK proteins (Xu et al. 2008), indicating that both proteins are likely in close contact

while bound to their common interactor. Future studies will be needed to determine enzyme/substrate

relationships between any of these proteins aside from the known phosphorylation of TSKS by TSSK1.

Currently our laboratory is conducting an intensive phosphoproteomic analysis of the wild-type and

Ppp1cc knockout testes in a search for candidate PPP1CC2 substrates, which may shed some light on

this question.

Kinase activity of TSSK1 towards TSKS has been demonstrated previously, and is thought to

occur on the serine 281 residue (Kueng et al. 1997, Xu et al. 2008). During the course of this study, we

identified a second phosphorylated residue, serine 54, which interestingly lies within the PP1 docking

RVxF motif. Previous studies have shown phosphorylation in and around RVxF motifs to inhibit

interaction with PP1 isoforms (Beullens et al. 1999, McAvoy et al. 1999, Liu & Brautigan 2000, Bollen.

Page 110: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

99

2001), which our experiments confirmed for TSKS. These data suggest that within the testis, there exists

a pool of TSKS that is incapable of interacting with PPP1CC2 (phosphorylated S54), while another

portion is permissive to interaction (unphosphorylated S54), given our ability to demonstrate this

interaction in the testis. Precisely how this is regulated remains an open question. PPP1CC2 itself is

unlikely to dephosphorylate this residue as the RVxF binding surface is distant from the active site on the

surface of the phosphatase (Egloff et al. 1997). Moreover, phosphorylation of the RVxF motif inhibits

binding of PPP1CC2. This indicates the involvement of another protein phosphatase in the regulation of

this interaction, the identity of which remains unknown, although a number of other non-type 1 protein

phosphatases are known to be expressed in the testis (Fardilha et al. 2011). Similarly, the kinase

responsible for this phosphorylation remains in question. It is tempting to suggest that TSSK1

phosphorylates S54 of TSKS as opposed to, or in addition to S281. However, if this were the case it is

difficult to envision how TSSK1 and PPP1CC2 could simultaneously exist in the same complex, even in

a transient manner, as occupancy of the RVxF motif by either protein would make it inaccessible to the

other. Therefore, there may be another kinase involved in regulation of this interaction as well, adding a

further layer of complexity. The kinase prediction tools Scansite (Obenauer et al. 2003) and KinasePhos

(Huang et al. 2005) both suggest PKC kinases as the top candidate based on the sequence surrounding

the phosphorylation site; however this would require further testing before any definitive conclusions are

drawn.

Our immunohistochemical analysis of TSKS and TSSK1 expression in the Ppp1cc knockout

testis shows reduced localization of both proteins to distinct puncta as well as a number of qualitative

defects. The puncta correspond to previously described structures proposed to originate in the

chromatoid body; a ring-shaped structure and a satellite (Shang et al. 2010). These abnormalities are

consistent with the phenotype of the Ppp1cc knockout as the TSKS/TSSK1 expressing elongating

spermatids are frequently missing in mutants, resulting in tubules lacking any observable staining for

these proteins. As well, loss of Ppp1cc causes in a bottleneck in the spermatogenic cycle, resulting in an

Page 111: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

100

increased number of tubules in stages VII/VIII (stages that are for the most part devoid of staining) and a

disorganization of the seminiferous epithelium resulting in tubules displaying a seeming mixture of

stages (Forgione et al. 2010). Taken together this suggests that the abnormalities in TSKS/TSSK1

staining are secondary to the initial effects of the Ppp1cc mutation early in spermatogenesis, but places

these proteins upstream of PPP1CC2 in a biochemical pathway later in spermatogenesis. A previous

analysis of TSKS expression in Tssk1/Tssk2 mutant tubules has shown that TSSK1 or TSSK2 or both are

required for the formation of chromatoid body derived ring and satellite structures as well as the correct

localization of TSKS to these structures during spermiogenesis (Shang et al. 2010). As well, as recent

analysis of the evolutionary history of the Tssk1 and Tssk2 genes in primates suggests that the two genes

may have not completely overlapping functions (Shang et al. 2013). One can thus envision a scenario

where TSSK1 binds and phosphorylates TSKS resulting in localization to specific puncta in the

developing elongating spermatid, whereupon it binds to PPP1CC2, forming at least transiently, a trimeric

(or higher oligomer) complex. The biological consequences of this interaction have yet to be uncovered,

but a critical role in spermiogenesis seems likely. TSSK1 and TSKS play a role in the formation and/or

function of chromatoid body derived structures in elongating spermatids (Shang et al. 2010). The

chromatoid body is a centre of RNA processing in developing round spermatids, but this may not be the

only role for this structure (Meikar et al. 2011). The role of the structures derived from the chromatoid

body in further developed elongating spermatids is less clearly defined, but it has been hypothesized that

it may have a role in assembly of the mitochondrial sheath, a compact helix of mitochondria that wrap

around the midpiece and connecting piece of the sperm flagellum (Shang et al. 2010). The fact that the

mitochondrial sheath is abnormal in Ppp1cc mutant spermatids as well leaves open the possibility that

PPP1CC2 is important for post-translational regulation of proteins during these processes. Another

known PPP1CC2 interactor in the testis, PPP1R42 (TLRR) has been shown to have a remarkably similar

localization to TSKS in elongating spermatids, with distinct PPP1R42 puncta located adjacent to the

centrosome, near the site of the chromatoid body (Shang et al. 2010, Wang et al. 2010). At other time

Page 112: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

101

points, PPP1R42 localization shows little similarity to that of TSKS/TSSK1; however this period of

overlap further suggests that PPP1CC2 may have some local concentration at this site.

In conclusion, the results of this study have shown interaction between the testis-specific Ser/Thr

phosphatase PPP1CC2, and two additional testis-specific proteins TSKS and the testis-specific Ser/Thr

kinase TSSK1. Furthermore, we have shown that the interaction between TSSK1 and PPP1CC2 is

indirect and likely mediated by TSKS, which binds to PPP1CC2 via the classical PP1 docking RVxF

motif. The RVxF motif on the TSKS surface can be phosphorylated in the testis, which is inhibitory to

PPP1CC2 interaction. The biological function of this interaction is unknown, but our data suggests that

TSKS and TSSK1 function upstream of PPP1CC2, and that regulation of this complex may include

several other proteins. Future experiments will seek to both identify these proteins, and determine the

precise regulatory mechanisms involved. It should also be noted that the results described in this chapter

arise from LC-MS/MS analysis of a single gel slice from GST-PPP1CC2 pull downs in the testis, and is

by no means an exhaustive survey of PPP1CC2 interactors in the testis. Analysis of additional bands can

reasonably be expected to identify more PPP1CC2 interacting proteins in the testis, some of which may

play a role in spermatogenesis.

Page 113: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

102

Chapter 4: Comparative phosphoproteomic analysis of the mouse testis reveals candidate

substrates of PPP1CC2

Author’s Note: A manuscript based containing a modified version of this chapter is currently in

preparation as: MacLeod G, Taylor P, Mastropaolo LA and Varmuza S. Comparative phosphoproteomic

analysis of the mouse testis reveals candidate substrates of PPP1CC2 (2013).

Except where indicated, all experiments were designed and conceived by myself and S. Varmuza. LA

Mastropaolo performed 50% of replicates for β-actin, Ppp1ca, Ppp1cc1 and Ppp1cc2 qPCR

experiments. P. Taylor designed and conducted all LC-MS/MS analysis of phosphopeptide samples,

conducted database searches and produced output files for analysis. P. Taylor also advised on

quantitative analysis strategy and writing the materials and methods for the LC-MS/MS analysis. I

performed all remaining experiments and was responsible for authorship of the manuscript and figure

generation, with editing by S. Varmuza.

Abstract

Deletion of the serine/threonine phosphatase gene Ppp1cc in the mouse results in male infertility

due to a failure of spermatogenesis. Ppp1cc is a member of the PP1 family of protein phosphatases, and

encodes two splice isoforms: the ubiquitous Ppp1cc1 and the testis-specific Ppp1cc2. As Ppp1cc mutant

mice are deficient for a protein phosphatase gene, we hypothesize that substrates of PPP1CC2 will be

hyperphosphorylated in mutant testes. Thus, we conducted a comparative phosphoproteomic analysis of

the Ppp1cc mutant testis. To limit the influence of secondary effects, we identified the time point in

spermatogenesis where PPP1CC2 is first required. Quantitative reverse-transcriptase PCR analysis of all

PP1 isoforms identified specific upregulation of Ppp1cc2 beginning at 3 weeks post-natal in the testis.

Histological analysis of the Ppp1cc mutant testis at this age confirmed the presence of morphological

abnormalities. Phosphopeptides were enriched using IMAC and TiO2 chromatography and compared

using both spectral counting and extracted ion chromatogram quantitation. Our data identified 755

different proteins phosphorylated in the 3 week mouse testis, and confidently assigned 1,026 unique

Page 114: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

103

phosphorylation sites. 33 different peptides corresponding to 32 different proteins were found to be more

abundant in Ppp1cc mutant samples than in wild-type, making these proteins candidate substrates of

PPP1CC2 in the testis. Amongst these proteins were several known to be essential for

spermatogenesis—HSPA4, HMGA1, YBX2 and SYCP2. In conclusion, our study identified a large pool

of testis phosphoproteins including a number of candidate PPP1CC2 substrates which will form the basis

of future studies on the role of PPP1CC2 in spermatogenesis.

4.1 Introduction

Protein phosphorylation is a critical intracellular signalling mechanism that plays a role in the

regulation of countless cellular processes. Spermatogenesis is no exception; in fact, next to the brain, the

testis contains more tissue specific phosphorylation sites than any other tissue studied (Huttlin et al.

2010, Lundby et al. 2012b). There exist examples of both kinases (such as TSSK1/TSSK2 (Xu et al.

2008, Shang et al. 2010), TSSK6 (Spiridonov et al. 2005), LMTK2 (Kawa et al. 2006)) and

phosphatases (PPP1CC2 (Varmuza et al. 1999), MTMR2 (Bolino et al. 2004), PPM1D (Choi et al. 2002)

which are indispensable to spermatogenesis. Thus, in the case of the Ppp1cc knockout mouse, it is

hypothesized that a loss of phosphatase activity towards a specific substrate or substrates causes failure

of spermatogenesis. Targeted deletion of Ppp1cc in mice results in the widespread loss of developing

germ cells most prominently from the round spermatid stage onwards (Varmuza et al. 1999). Ppp1cc

mutant seminiferous tubules appear to display a bottleneck in spermatogenesis that results in a

disorganised seminiferous epithelium (Forgione et al. 2010). Surviving germ cells display a wide range

of abnormalities including defects in chromatin condensation and acrosomal biogenesis (Varmuza et al.

1999, Forgione et al. 2010). Ppp1cc is a member of the PP1 family of Ser/Thr protein phosphatases,

which includes three different genes in mammals, Ppp1ca, Ppp1cb and Ppp1cc. The Ppp1cc knockout

mouse lacks the expression of two different splice isoforms, the ubiquitous Ppp1cc1 and the testis-

specific Ppp1cc2. Studies have indicated that PPP1CC2 is the predominant isoform in the mouse testis

although other PP1 isoforms are expressed (Takizawa et al. 1994, Varmuza et al. 1999). A recent

Page 115: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

104

transgenic rescue experiment has established that expression of the PPP1CC2 isoform alone is able to

rescue the Ppp1cc mutant infertility phenotype, provided expression levels surpass a threshold amount

(Sinha et al. 2012).

While the requirement of PPP1CC2 for completion of mammalian spermatogenesis is well

established, its exact role(s) has yet to be elucidated. To date, a number of PPP1CC2 interacting proteins

in the testis have been identified (Vijayaraghavan et al. 1996, Huang et al. 2002, Hrabchak & Varmuza

2004, Huang et al. 2004, Hrabchak et al. 2007, Cheng et al. 2009, Fardilha et al. 2011b, Henderson et al.

2011, Ruan et al. 2011, Wang & Sperry 2011), including most recently, DDOST (MacLeod & Varmuza

2012, chapter 2), TSKS and TSSK1 (MacLeod et al., 2013, in preparation, chapter 3); however the

identification of bona fide PPP1CC2 substrates has proven to be challenging, as the interaction between

PP1 isoforms and their substrates is typically weak, and short-lived. Ppp1cc mutant testes are lacking

expression of a protein phosphatase gene, meaning that PPP1CC2 is unavailable for substrate

dephosphorylation. Therefore, by identifying hyperphosphorylated testis expressed proteins in a Ppp1cc

mutant background we can identify candidate substrates of PPP1CC2 that may play a crucial role in

spermatogenesis. Previous research from our laboratory performed 2-dimensional electrophoresis (2DE)

coupled with phosphoprotein staining to identify hyperphosphorylated proteins in the Ppp1cc mutant

testis (Henderson et al. 2011). This approach identified several candidate PPP1CC2 substrates, including

HSPA2 and β-Tubulin. In the present study we have expanded upon this line of inquiry by making

several key changes to our methodology. First, although the use of 2DE is a dependable method of

protein separation, its use in proteomics is compromised by the fact that it requires the excision of

individual spots on a gel. Another limitation of 2DE is that larger changes in phosphorylation can affect

the pI of a protein, and alter its migration on a gel making comparison between phosphorylated and

unphosphorylated proteins difficult. In contrast, LC-MS/MS based methods coupled with

phosphopeptide enrichment can simultaneously identify numerous phosphopeptides from a single

sample, and can also identify the specific site of phosphorylation. Phosphopeptide enrichment strategies

Page 116: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

105

such as immobilized metal ion chromatography (IMAC), and TiO2 chromatography have been

demonstrated to be highly effective in identifying phosphorylation sites via LC-MS/MS from rodent

testis samples (Huttlin et al. 2010, Lundby et al. 2012b). Similar approaches have been used successfully

to analyze phosphopeptide changes during rat sperm capacitation, and epididymal maturation, with as

little as 150 µg of starting material (Baker et al. 2010, Baker et al. 2011).

A second major methodological change relates to the tissue samples analyzed. Our previous

testis phosphoproteomics study (Henderson et al. 2011) used testis proteins from adult mice for their

analysis. However, the composition of the wild-type adult testis is very different from the Ppp1cc

mutant, as the latter suffers from a near complete lack of cells from the round spermatid phase onwards

(Varmuza et al. 1999). The proteomes of different spermatogenic cell types are quite different (Huang et

al. 2008, Guo et al. 2011), and by extension their phosphoproteomes. Thus, many observed changes in

protein phosphorylation status are likely due to secondary effects. To ensure that phosphoproteomic

analysis reflects changes due specifically to a loss of PPP1CC activity, and not secondary effects, we

have opted to focus on the time-point that the Ppp1cc gene is first required in the testis. Previous work

from our laboratory has suggested that this time point occurs at approximately 3 weeks of age,

corresponding to the first observed defects in the seminiferous tubules of Ppp1cc knockout mice

(Varmuza & Ling 2003). To verify this observation we first performed an analysis of the expression of all

four PP1 isoforms in the mouse testis to test for the earliest incidence of Ppp1cc2 specific upregulation

and performed a phenotypic comparison of wild-type and Ppp1cc mutant seminiferous tubules at this

time point prior to conducting a comparative phosphoproteomic analysis.

4.2 Materials and Methods

RNA extraction and cDNA synthesis

Testis and brain samples were homogenized in Trizol® Reagent (Invitrogen) and RNA was

extracted using the manufacturer`s suggested protocol. RNA quality was assayed via gel electrophoresis

Page 117: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

106

with examination of the 16S and 18S rRNA bands. RNA purity and yield were determined using a

nanodrop spectrophotometer (Fisher Scientific). Following RNA extraction, samples were diluted to 1

µg/µL in ultrapure H2O. For cDNA synthesis 5 µg of RNA was treated with DNAse I in the presence of

RiboLock™ RNAse inhibitor (Thermo Scientific), followed by reverse transcription using 20 units of M-

MuLV (Fermentas) with 100 ng random hexamer primers (Fermentas) for 1 hour at 50°C. Resultant

cDNA was diluted in ultrapure H2O.

Quantitative PCR

Two primer pairs were utilized in qPCR experiments for each transcript tested; an “outside”

primer pair for production of template DNA for standard curve production, and an “inside” primer pair

for the qPCR reaction itself. Primer pairs used in this experiment are listed in Table 4.1. qPCR was

performed using SYBR® Advantage® qPCR Premix (Clontech) according to the provided protocol.

Reactions were carried out in a Rotorgene 3000 Light Cycler (Corbett Research) and analysed using the

accompanying software. Following an initial denaturation period of 30 seconds at 95°C, PCR cycling

conditions were as follows: denaturation for 5 seconds at 95°C; annealing for 20 seconds at 55°C for

actin, 53°C for Ppp1ca and Ppp1cb, 56.1°C for Ppp1cc1, 53.7°C for Ppp1cc2; extension for 30 seconds

at 72°C for a total of 40 cycles. Normalized Ppp1c expression levels were calculated by dividing raw

Ppp1c values by the average of 2 Actb qPCR runs for a given cDNA sample. Analysis was performed on

cDNA samples from 3 different mice at each age, with duplicate qPCR runs performed on each sample.

Exceptions to this sample size include Ppp1cc2 1 week sample, where an outlier (~100X all other

samples at that age point) was discarded and Ppp1cb 5 week sample where only two samples were

available. Relative Ppp1c expression levels from duplicates were averaged prior to statistical analysis.

Statistical analysis of qPCR data

Page 118: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

107

Table 4.1: Primers used for qPCR analysis of PP1 isoform expression in the post-natal mouse testis

Page 119: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

108

Transcript Outside/Inside Forward Primer Sequence Reverse Primer Sequence

Actb outside ATGAAGATCCTGACCGAGCG TACTTGCGCTCAGGAGGAGC

Actb inside GACGGCCAGGTCATCACTAT GCACTGTGTTGGCATAGAGG

Ppp1ca outside CAGCCATTGTGGATGAGAAG TTTCTTGGCTTTGGCAGAAT

Ppp1ca inside TTGCCAAGAGACAGTTGGTG ACTGCCCATACTTGCCCTTA

Ppp1cc1 outside GTTTTAACTGCTTGCCGATAGC CTGAATGGACGGGTTCAGG

Ppp1cc1 inside CCCATCAGGTGGTTGAAGAT CTGAATGGACGGGTTCAGG

Ppp1cc2 outside GTTTTAACTGCTTGCCGATAGC GCCTGATCCAACCCGTGG

Ppp1cc2 inside CCCATCAGGTGGTTGAAGAT GCCTGATCCAACCCGTGG

Ppp1cb outside TGCTAGCATCAAATCGCATTT TCGGTGGATTAGCTGTTCG

Ppp1cb inside CTAAACGACAGTTGGTAACCTT TCGGTGGATTAGCTGTTCG

Page 120: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

109

To test for significant changes in Ppp1c isoform expression levels throughout post-natal

development a one-way analysis of variance (ANOVA) was performed with the aid of Microsoft Excel.

ANOVAs were followed by Tukey’s post-tests, performed manually, to conduct pairwise comparisons of

expression data from different age groups. Newman-Kuels multiple range test, and pairwise t-tests were

also performed on selected samples where indicated.

Histology

Testes from 3 week old wild-type and Ppp1cc mutant mice were dissected and fixed in Bouin’s

solution overnight at 4°C followed by dehydration in a graded series of ethanol solutions and embedding

in TissuePrep® (Fisher Scientific). Seven μm sections were dewaxed, hydrated and stained using the

Periodic Acid-Schiff (PAS) staining system (Sigma-Aldrich) with haematoxylin counterstaining. Slides

were mounted in 50% glycerol for viewing with an Olympus BX60 microscope and images were

captured using Cool Snap software and a CCD camera (RSPhotometrics). Images were adjusted for

brightness and contrast using Photoshop 6.0 (Adobe).

Phosphopeptide Enrichment

Mouse testes were decapsulated, homogenized in 400μL of 7M Urea, 4% CHAPs (w/v), 40mM

Tris and DNAseI and incubated with rocking at 4°C for 30 minutes. Extracted proteins were reduced

with 20mM dithioreitol for 1 hour at room temperature followed by alkylation with 40 mM iodoacetic

acid for 35 minutes at room temperature in darkness. Testis proteins were precipitated with acetone and

dried using a speed vac. Dried acetone pellets were resuspended in 1M urea, 50 mM ammonium

bicarbonate and concentration was determined using the BCA Protein Assay Kit from Bio Basic Inc.

Trypsin digestion of testis protein was carried out overnight with 2% (w/w) of proteomics grade trypsin

(Sigma-Aldrich T6765) at room temperature with end over end rotation. Digestion was subsequently

acidified with 1% (v/v) formic acid and centrifuged to remove insoluble material. 120 μg testis peptides

were then was used for phosphopeptide enrichment via sequential elution from IMAC (SIMAC) using

Page 121: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

110

the protocol modified from Thingholm et al. (Thingholm et al. 2009, Thingholm et al. 2008). Briefly,

peptides were resuspended in ~300 μL of IMAC loading buffer (50% acetonitrile, 0.1% TFA) and

incubated with 40 μL of washed PHOS-select™ IMAC resin (Sigma-Aldrich) for 30 minutes with end

over end rotation. IMAC resin was packed into a 200 μL gel loading pipette tip, and washed with 150 μL

of IMAC loading buffer. A first phosphopeptide elution step was conducted with 140 μL of acidic

elution buffer (1% TFA, 20% acetonitrile) followed by a second elution with 160 μL of basic elution

buffer (ammonia water). The basic elution fraction was acidified with 10 μL of formic acid and

lyophilized. IMAC flow through (and wash) and acidic elution fraction were also lyophilized, followed

by resuspension in 250 μL TiO2 loading buffer (80% acetonitrile, 5% TFA, 1M glycolic acid).

Resuspended peptides were incubated with 50 μL of washed TiO2 Mag Sepharose™ (GE Healthcare) for

30 minutes with end over end rotation. TiO2 resin was washed sequentially with 500 μL TiO2 loading

buffer, 80 μL of 80% acetonitrile with 1% TFA and 80 μL of 10% acetonitrile with 0.2% TFA. After 10

minutes of air drying phosphopeptides were eluted from TiO2 resin by incubation with 50 μL ammonia

water for 5 minutes with constant mixing. Eluted phosphopeptide solution was acidified with 2 μL of

10% TFA and 7 μL of formic acid. For each individual mouse the TiO2 enriched IMAC flow through and

acidic elution fractions were then combined and lyophilized.

LC-MS/MS analysis

The digested peptides were loaded onto a 100 μm ID pre-column (Thermo-Fisher Scientific) at 4

μl/min and separated over a 75 μm ID X 15 cm analytical column (EASY-Spray, Thermo-Fisher

Scientific, Odense Denmark). The peptides were eluted over 60 min. at 300 nl/min. using a 0 to 40%

acetonitrile gradient in 0.1% formic acid using an EASY nLC 1000 nano-chromatography pump

(Thermo-Fisher Scientific, Odense Denmark). The peptides were eluted into a LTQ Velos-Orbitrap Elite

hybrid mass spectrometer (Thermo-Fisher, Bremen, Germany) operated in a data dependant mode. MS

was acquired at 240,000 FWHM resolution in the FTMS and MS/MS was carried out in the linear ion

trap. 10 MS/MS scans were obtained per MS cycle. The multistage activation option of the analyzer

Page 122: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

111

was selected to reduce the effect of neutral loss fragmentation. The raw data files were searched using

Proteome Discover 1.3 (Thermo-Fisher Scientific) using a parent ion accuracy of 10 ppm and a fragment

accuracy of 0.6 Da. A fixed modification of carbamidomethyl cysteine and variable modifications of

phosphorylated serine, threonine and tyrosine residues, as well as oxidized methionine were considered

in the search

Database searching

All MS/MS samples were analyzed using Sequest (Thermo Fisher Scientific, San Jose, CA,

USA; version 1.3.0.339) and X! Tandem (The GPM, thegpm.org; version CYCLONE (2010.12.01.1)).

Sequest was set up to search MOUSE-Uniprot-Sep-05-12.fasta (unknown version, 55250 entries)

assuming the digestion enzyme trypsin. X! Tandem was set up to search the MOUSE-Uniprot-Sep-05-12

database (unknown version, 55270 entries) also assuming trypsin. Sequest and X! Tandem were searched

with a fragment ion mass tolerance of 0.60 Da and a parent ion tolerance of 10.0 PPM. Carbamidomethyl

of cysteine was specified in Sequest and X! Tandem as a fixed modification. Glu->pyro-Glu of the n-

terminus, ammonia-loss of the n-terminus, gln->pyro-Glu of the n-terminus, oxidation of methionine and

phospho of serine, threonine and tyrosine were specified in X! Tandem as variable modifications.

Oxidation of methionine and phospho of serine, threonine and tyrosine were specified in Sequest as

variable modifications.

Criteria for protein identification

Scaffold (version Scaffold_3.4.3, Proteome Software Inc., Portland, OR) was used to validate

MS/MS based peptide and protein identifications. Peptide identifications were accepted if they exceeded

specific database search engine thresholds. Sequest identifications required at least deltaCn scores of

greater than 0.10 and XCorr scores of greater than 1.8, 2.5, 3.5 and 3.5 for singly, doubly, triply and

quadruply charged peptides. X! Tandem identifications required at least -Log (Expect Scores) scores of

greater than 1.00. Protein identifications were accepted if they contained at least 1 identified peptide.

Page 123: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

112

Proteins that contained similar peptides and could not be differentiated based on MS/MS analysis alone

were grouped to satisfy the principles of parsimony.

Bioinformatic and semi-quantitative analysis of testis phosphoproteins

Enrichment analysis was performed on identified testis phosphoproteins using the DAVID

Functional Annotation Chart tool from the DAVID Bioinformatics Resources 6.7 (Huang et al. 2008,

Huang et al. 2009) using the Mus musculus background and queried for GO cellular component,

biological process and molecular function. Known and predicted PP1 interacting proteins were

identified using PSICQUIC (Aranda et al. 2011) and I2D (Brown & Jurisica 2005, Brown & Jurisica

2007) (mouse as target organism) search tools. Annotation of identified phosphopeptides to the GO

biological process reproduction (GO:0000003) was performed using the Scaffold viewer software.

Assignment of phosphorylation to specific sites was performed using Scaffold PTM (version 2.0.0,

Proteome Software) which is based on the Ascore algorithm (Beausoleil et al. 2006). Spectral counting

of individual phosphorylation sites was performed using data from Scaffold PTM assigned sites only,

which were present in at least 2 of 3 replicates of at least one genotype. A cut-off value of 2 was used as

a minimal fold-change required for further analysis.

MS1 filtering and quantitative analysis of testis phosphopeptides

MS1 filtering quantitative analysis of extracted ion chromatograms was performed using Skyline

(Schilling et al. 2012) according to the step by step instructions available on the Skyline website

(http://proteome.gs.washington.edu/software/Skyline/tutorials/ms1filtering.html). A spectral library was

first created using .DAT files from all six replicate MS/MS runs described above using a cut-off score of

0.8. The Skyline peptide tree was populated with candidate hyperphosphorylated peptides identified in

the preliminary analysis described above by either importing specific peptide sequences, or full protein

FASTA sequences retrieved using the Uniprot retrieval tool. Results for MS1 filtering were imported

from raw files for all six samples using an import filter resolution of 100,000. Skyline automated peak

Page 124: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

113

selection was manually adjusted when clear outliers in retention time were observed in replicate results.

XIC peak area values were exported from Skyline into Microsoft Excel and were log2 transformed and

subjected to statistical analysis using a student’s t-test (p ≤ 0.05).

4.3 Results

Quantitative analysis of PP1 isoform expression during the first wave of spermatogenesis

Expression of Ppp1ca, Ppp1cb, Ppp1cc1, and Ppp1cc2 was detectable in mouse testis cDNA

ranging from 1 to 5 weeks of age. In addition, Ppp1ca and Ppp1cc1 were detectable in brain cDNA,

whereas little or no Ppp1cc2 expression was evident (level of expression was outside the range of the

standard curve) and Ppp1cb was not assayed. The levels of Ppp1ca, Ppp1cb and Ppp1cc1 transcripts

displayed no significant difference in expression level throughout the first wave of spermatogenesis, or

between brain and testis cDNA samples (Figure 4.1). In contrast, ANOVA analysis of Ppp1cc2 transcript

levels showed a clear and statistically significant increase in expression level throughout the first wave of

spermatogenesis (p=0.001) (Figure 4.1). Ppp1cc2 expression level was low at 1 and 2 weeks of age.

Subsequently, at three weeks of age the level of Ppp1cc2 increased approximately six-fold, which is a

statistically significant increase relative to the 2 week time point (p<0.001). By four weeks of age

Ppp1cc2 transcript levels had again significantly increased (p=0.02) a further 2.5-fold. By the four week

time point Ppp1cc2 expression level had reached its maximum level in the first-wave of

spermatogenesis, as no significant change was evidenced at the 5 week time point. This data shows

conclusively that the Ppp1cc2 transcript specifically is upregulated in the mouse testis by 3 weeks of age.

The seminiferous epithelium of 3 week old Ppp1cc mutant mice displays impaired spermatogenesis

To test if the upregulation of Ppp1cc2 in the mouse testis at 3 weeks of age corresponds to a

requirement of PPP1CC2 for normal spermatogenesis, we examined the seminiferous tubules of wild-

type and Ppp1cc mutant mice. Previous research has observed the first phenotypic effects of the Ppp1cc

mutant to be visible at this stage (Varmuza & Ling 2003), which we sought to confirm before progressing

Page 125: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

114

Figure 4.1: Expression of mouse Ppp1c isoforms in the testis during the first wave of spermatogenesis

demonstrates specific upregulation of only Ppp1cc2. Time points listed are weeks post-natal. Expression

levels from brain were from whole adult tissue. Values are relative to β-actin x 1000. * indicates a

statistically significant (α= 0.05) change in expression level from the previous time point. Note that in

the brain, Ppp1cb levels were not tested, and Ppp1cc2 levels were tested but below the level of

quantitation.

Page 126: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

115

*

*

Page 127: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

116

with further experiments. While the first wave of spermatogenesis is commonly referred to as

synchronous, our observations suggest that this is not entirely true, consistent with previous work from

our lab (Varmuza &Ling. 2003). By three weeks of age, most wild-type mouse seminiferous tubules are

in the final stages of meiosis and the remainder contain the first round spermatids (Figure 4.2A). Wild-

type seminiferous tubules generally display a uniform thickness with very few observed instances of cell

sloughing (Figure 4.2C). Progression of spermatogenesis does not appear to be impaired in Ppp1cc

mutant seminiferous tubules as both tubules containing late spermatocytes and round spermatids were

visible (Figure 4.2B). While a substantial proportion of Ppp1cc mutant seminiferous tubules display

normal morphology, many others are abnormal in appearance including an increase in cell sloughing and

vacuolation (Figure 4.2B). A closer examination of the seminiferous epithelium inside Ppp1cc mutant

seminiferous tubules reveals a number of reoccurring defects including cell sloughing (Figure 4.2D),

joined round spermatid nuclei within a single cytoplasm (Figure 4.2E, arrows), and prominent “blobs” of

cellular material projecting into the tubule lumen (Figure 4.2F, arrowhead). These results indicate that

while there is no block in the progression of spermatogenesis, there is impairment in the process by three

weeks of age in response to the loss of Ppp1cc. Furthermore, while there is evidence of cell loss at this

age, the cellular composition of wild-type and Ppp1cc mutant seminiferous tubules is very similar.

Phosphoproteomic analysis of the 3 week mouse testis.

After determining that Ppp1cc is first required in the testis by 3 weeks of age we sought to

achieve two goals via phosphoproteomic analysis. First, we wanted to identify as many phosphoproteins

in the 3 week testis as possible. These proteins would serve as a resource to others interested in PTMs

during testis development, as well as representing a large pool of potential PPP1CC1/PPP1CC2

substrates for further bioinformatic analysis. Our second and ultimate goal was to identify proteins that

were differentially phosphorylated in wild-type and Ppp1cc mutant 3 week testes. Those proteins that

are hyperphosphorylated on Ser/Thr residues in Ppp1cc mutants would represent strong candidate

PPP1CC2 testis substrates.

Page 128: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

117

Figure 4.2: Phenotypic abnormalities are present in the seminiferous tubules of 3 week old Ppp1cc

mutant mice. Heterozygous seminiferous tubules (A and C) retain the vast majority of developing germ

cells and present a consistent and normal morphology. In Ppp1cc mutant testes phenotypically normal

and abnormal seminiferous tubule phenotypes are often evident within the same cross-section (B). A

number of phenotypic abnormalities are visible in Ppp1cc mutant seminiferous tubules including germ

cell sloughing (B, asterisk and D)multiple spermatid nuclei joined closely together (E, arrows) and large

“blobs” of cellular material projecting into the tubule lumen (F, arrowhead).

Page 129: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

118

Page 130: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

119

Phosphopeptide enrichment of 3 week mouse testis peptide samples was performed using a

modified sequential elution from IMAC (SIMAC) (Thingholm et al. 2009, Thingholm et al. 2008)

strategy. The SIMAC method includes an initial phosphopeptide enrichment using IMAC, in which

bound peptides are sequentially eluted with an acidic and basic solution. Next the IMAC flow-

through and acidic elution are subjected to a second phosphopeptide enrichment step using TiO2

chromatography. Using our conditions, the TiO2 enriched fractions yielded 95% phosphopeptides.

However, the IMAC basic elution fraction contained only 15% phosphopeptides. For this reason, we

proceeded using only the combined TiO2 enriched fractions.

Phosphopeptide enriched samples from three biological replicates each of 3 week old wild-type and

Ppp1cc mutant testis were then analyzed via LC-MS/MS using an LTQ-Orbitrap elite to identify the

phosphopeptides present. This data has been uploaded to the Global Proteome Machine website

(http://review.thegpm.org/review/index.html and can be accessed using the following accession numbers-

Mut1: GPM77700020781, Mut 2: GPM00300024842, Mut 3: GPM77700020782, WT1:

GPM77700020783, WT2: GPM77700020784, WT3: GPM77700020785) and will be made publicly

available following publication. In total 12,651 phosphopeptides were identified, corresponding to 755

different proteins and 1,026 different phosphorylation sites. Searching identified spectra against a decoy

database revealed 0 false identifications. Fifteen percent more phosphopeptides were detected in the

Ppp1cc mutant samples when compared to the wild-type (6741 vs. 5912), however this increase was not

statistically significant (p=0.17). Our phosphopeptide enrichment strategy was highly reproducible as

was evidenced at the protein level with a high degree of overlap between wild-type and Ppp1cc mutant

samples, as well as between biological replicates within the same group. Six hundred and four (604)

different proteins were identified in wild-type samples and 666 were identified in Ppp1cc mutant

samples. Of these proteins, 515 were identified in at least one biological replicate of each genotype, 89

were unique to wild-type and 151 were unique to Ppp1cc mutants (Figure 4.3A). Fifty-five percent of

phosphorylated proteins detected in wild-type samples were detected in all 3 biological replicates and

Page 131: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

120

Figure 4.3: Reproducible phosphopeptide enrichment and identification in wild-type and Ppp1cc mutant

testis samples. (A) Venn diagram depicting the number of unique phosphoproteins identified in each

genotype examined. (B) Venn diagram depicting the number of unique phosphopeptides identified in

each genotype examined. (C) Venn diagram depicting the inter-sample reproducibility at the protein

level in wild-type testis samples. Triangle represents proteins found in at least 2 of 3 replicates, which

were subsequently considered for quantitative analysis via spectral counting. (D) A similar Venn diagram

depicting inter-sample reproducibility in Ppp1cc mutant samples.

Page 132: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

121

Page 133: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

122

72% were found in more than one replicate (Figure 4.3C). In Ppp1cc mutant samples, 47% of

phosphorylated proteins detected were found in all 3 biological replicates, and 70% were found in at least

2 replicates (Figure 4.3D). At the peptide level 1261 unique phosphopeptides (meaning amino acid

sequence, ignoring phosphorylation site assignment) were identified, 786 of which were found in both

wild-type and Ppp1cc mutant testis samples (Figure 4.3B). Amongst the identified testis phosphoproteins

were 40 proteins encoded by genes for which male infertility mouse models exist (Appendix A.4.5). As

well, we have found 21 known PP1 interacting proteins (or approximately 10% of all known interactors)

and 8 predicted PP1 interacting proteins to be phosphorylated in the 3 week mouse testis (Appendix

A.4.5), including at least 3 known PP1 substrates: RB1, SRSF10 and PPP1R2. Eleven different proteins

that were identified in SBP-3XFLAG-PPP1CC2 ES cell tandem affinity purification (Chapter 2), as well

as the OST complex protein STT3B were also identified in our phosphoproteomic dataset (Appendix

A.4.5). None of the proteins identified in GST-PPP1CC2 adult testis pull down assays (Chapter 3) were

present in the phosphoproteomic dataset. The absence of TSSK1 and TSKS from this dataset is not

surprising as they are not expressed in the testis by 3 weeks of age.

To further characterize the overall characteristics of the 3 week testis phosphoproteome, we

performed a bioinformatic analysis using the DAVID Functional Annotation Tool to identify GO Cellular

Compartment, Functional Annotation, and Biological Process terms enriched in our dataset (Full list in

Appendix A.4.6). GO cellular compartment analysis showed enrichment for a number of compartments

including chromosome/chromatin, ribonucleoprotein complexes and the cytoskeleton. DNA, RNA and

chromatin binding proteins, serine/threonine kinases and transcription factor binding proteins were all

highly enriched via GO molecular function analysis. The most highly enriched biological processes in

the 3 week mouse testis phosphoproteome included RNA processing, chromatin organisation and the cell

cycle, with meiosis and spermatogenesis also being significantly enriched. These functional

characteristics are not unexpected given our knowledge of this tissue at this particular point in

development, where intensive reorganization of the nuclear material is underway in the majority of cells,

Page 134: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

123

and cytoskeletal reorganization is primed to begin in newly emerging cells. However, the large

proportion of phosphoproteins identified does serve to underscore the importance of protein

phosphorylation in regulating these processes.

Semi-quantitative comparison of wild-type and Ppp1cc mutant testis phosphoproteomes

In order to examine changes in protein phosphorylation status in response to the Ppp1cc deletion

it is necessary to examine our results at the peptide level, as it is the phosphorylation stoichiometry at

specific sites that will yield candidate substrate identification. Label-free quantitative analysis was

performed on the three biological replicates of each genotype and subjected to both spectral counting and

extracted ion chromatogram quantitation. Our quantitative analysis strategy consisted of two stages, a

preliminary stage where candidate peptides were identified for closer inspection, followed by a thorough

quantitative analysis of the candidate peptides using MS1 filtering of extracted ion chromatograms

across all six replicates (Figure 4.4).

Spectral counting is a semi-quantitative measure based on the correlation between the abundance of a

protein and the frequency of sampling of precursor ions in an LC-MS/MS experiment (Liu et al. 2004).

As a first approach in our preliminary quantitative analysis of the 3 week testis phosphoproteome we

used the Scaffold PTM software (version 2.0.0, Proteome Software Inc., Portland, OR) which reanalyzes

MS/MS results using an extension of the Ascore algorithm to produce confident PTM site assignments.

Hence, we were able to confidently assign 1,026 unique phosphorylation sites (923 serine, 97 threonine

and 6 tyrosine) in 572 different proteins throughout our 6 replicates. To identify candidate

hyperphosphorylated proteins in 3 week Ppp1cc mutant testes we identified phosphorylation sites that

were a) present in at least 2 of 3 biological replicates of at least one genotype, and b) exhibited at least a

2-fold increase in spectral count in Ppp1cc mutant replicates (Figure 4.5A). This analysis yielded

phosphorylation sites in 145 different proteins that were more abundant in Ppp1cc mutants (Appendix

A.4.7). Amongst these proteins were at least two known PP1 interacting proteins; MAP1B (Ulloa et al.

Page 135: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

124

Figure 4.4: Strategy for quantitative analysis of wild-type and Ppp1cc mutant 3 week testis

phosphoproteomes. After phosphopeptide enrichment via IMAC and TiO2 chromatography, peptides

were subjected to LC-MS/MS analysis. MS/MS data was used for preliminary identification of candidate

hyperphosphorylated peptides via spectral counting, MS/MS peak XIC quantitation as well as peptides

from genes essential for male fertility, associated with the GO biological process Reproduction, and

encoding known or predicted PP1 interacting proteins (PIPs). Candidate peptides were then subjected to

MS1 filtering using Skyline to compare XIC quantitative measurements from MS data across all six

replicates.

Page 136: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

125

Page 137: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

126

Figure 4.5: Distribution of spectral counting and MS/MS peak XIC data for wild-type and Ppp1cc

mutant testis phosphopeptides. A) Spectral counting of individual phosphorylation sites. The solid line

indicates an equal spectral count in each genotype, while dashed lines indicate a 2-fold increase in either

genotype. B) XIC of MS/MS peaks of individual phosphopeptides. X-axis represents a log2

transformation of the Ppp1cc mutant/wild-type ratio of extracted ion chromatogram area under the curve

values. Y-axis is the untransformed sum of the average extracted ion chromatogram area under the curve

values for wild-type and Ppp1cc samples for a given peptide observed in at least 2 of 3 replicates of each

genotype.

Page 138: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

127

A

B

Page 139: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

128

1993, Hendrickx et al. 2009) and NCL (Nucleolin) (Morimoto et al. 2002) as well as 12 proteins encoded

by genes which when deleted result in infertility in mice (ESPL1, TDRD1, SYCP3, AGFG1, CCNE2,

ADAM2, HMGA1, CLGN, CSDA, SYCP2, RAD18 and HSPA4). As mentioned above, spectral

counting is considered to be a semi-quantitative analytical method is subject to an increased amount of

variation when dealing with low spectral count numbers as is the case for many of the phosphopeptides

observed in this study. Therefore, these peptides require further analysis to determine if they are truly

hyperphosphorylated in Ppp1cc mutants.

To complement our preliminary spectral counting analysis of the 3 week mouse testis

phosphoproteome, we next performed a second preliminary quantitative analysis approach, this time

based on the extracted ion chromatograms (XIC) of phosphopeptides. The signal intensity of a given

peptide in an LC-MS run is directly proportional to its abundance, thus by extracting the area under to

curve of a peptide peak (XIC value) in a number of samples we can compare its abundance (Chelius &

Bondarenko. 2002). For this approach, we compared the average log2 transformed XIC values for all

phosphopeptides detected in the SEQUEST database search between wild-type and Ppp1cc mutant testis

samples (Figure 4.5B). To reduce the effect of outliers and allow for statistical analysis we only

considered peptides that were present in at least 2 of 3 biological replicates for each genotype. Both a

two sample student’s t-test (when 3 replicate values of each genotype were available) and a local-pooled

error test (LPE) (Jain et al. 2003, Colinge et al. 2005) were conducted to detect peptides that differed in

abundance when Ppp1cc was deleted in the 3 week mouse testis. This analysis identified 40 different

phosphopeptides that were more abundant in Ppp1cc mutant samples, corresponding to 36 unique

proteins (Appendix A.4.7), seven of which were also identified via spectral counting (ACIN1, ENTHD1,

ESPL1, HNRNPU, LARP1, RALY, and SYCP2). The preliminary quantitative analysis described above

suffers from one key limitation; the fact that the XIC values for only those peptide ions that were

subsequently selected for MS/MS fragmentation in that sample are considered. Thus, a large amount of

Page 140: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

129

information remains unexplored in the LC-MS data, and peptides identified as more abundant in mutant

testes by these means require further analysis as confirmation.

Identification of hyperphosphorylated proteins in the 3 week Ppp1cc mutant testis via label-free

quantitative analysis

Preliminary quantitative analysis of the 3 week Ppp1cc mutant testis phosphoproteome provided

a list of candidate hyperphosphorylated peptides that is suitable for further label-free quantitative

analysis. In addition to the candidate hyperphosphorylated peptides, we also further analyzed all known

or predicted PP1 interacting proteins (29 proteins), proteins encoded by genes essential for male fertility

(40 proteins), and proteins associated with the GO biological process Reproduction (33 proteins) that

were identified in our phosphoproteomic dataset, as well as proteins identified in SBP-3XFLAG-

PPP1CC2 ES cell tandem affinity purification (Chapter 2). In total phosphopeptides from 233 different

proteins were subjected to extracted ion chromatogram quantitative analysis across all six replicates

using the Skyline MS1 filtering tool (Schilling et al. 2012). Skyline was used to select and align MS

peaks with integrated MS/MS and retention time data from which area under the curve (AUC) values

were calculated. Log2 transformed AUC values were then subjected to statistical analysis via a student’s

t-test and peptides displaying a statistically significant (p ≤ 0.05) increase in abundance were identified.

This yielded the identification of 33 different peptides in 32 proteins that were significantly more

abundant in the 3 week Ppp1cc mutant testis (Table 4.2). A representative example of a peptide from

ARHGEF11 is depicted in Figure 4.6. The average fold-change in these peptides is 4.8 with a lowest

significant fold-change of 1.6 and a highest of 26.9. Amongst the identified hyperphosphorylated

proteins, 4 are encoded by genes which are essential for male fertility—HMGA1, HSPA4, SYCP2 and

YBX2 (Figure 4.7). Additionally one known PP1 interacting protein MAP1B, and two predicted PP1

interacting proteins HSPA4 and SIRT2 were hyperphosphorylated (Figure 4.7, full set in Appendix

A.4.8). As all of these proteins have phosphorylation sites that are more abundant upon deletion of the

phosphatase gene Ppp1cc they all represent candidate PPP1CC2 substrates in the testis. Gene ontology

Page 141: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

130

Table 4.2: Candidate PPP1CC2 substrates. Shown is a list of identified phosphopeptides significantly

more abundant in 3 week old Ppp1cc knockout testes than in wild-type. Gene names followed by *

indicate an infertility phenotype in knockout mice. pS and pT reflect phosphorylated serine and threonine

residues respectively. Residues surrounded by parentheses represent cases of ambiguous phosphorylation

site assignment. Ascore values are given for only those phosphorylation sites confidently assigned by

Scaffold PTM software.

Page 142: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

131

Accession

Gene

Name PeptideSequence

GO Biological Process Localized

Phosphorylation

site Ascore

Mut/

WT

XIC

AUC t-test

F6RJ39 Acin1 HLpSHPEPEQQHVIQR apoptotic process S656 1,000 2.9 0.002

Q68FM7 Arhgef11 NSVLSDPGLDpSPQTpSPVILAR cytokinesis S251, S255 36.46, 28.50 3.7 0.023

Q8K019 Bclaf1 QKFHDpSEGDDTEETEDYR transcription S395 56.76 1.6 0.036

Q9CXS4 Cenpv pSGATGGLSGGESR cell cycle S18 26.79 1.9 0.016

P60904 Dnajc5 p(SLSTSGESLYHVLGLDK) protein folding 2.4 0.040

Q8JZQ9 Eif3b AKPAAQSEEETATpSPAApSPTPQSAER translation S75, S79 51.64, 20.17 2.3 0.039

E9Q0C6 Gm14569 p(SISHDSVFCLDPEPEKGAGK) N/A 2.3 0.001

P17095 Hmga1*

KLEKEEEEGIpSQEpSpSEEEQ transcription S99, S102, S103 1,000, 1,000, 1,000 3.2 0.010

Q3U2G2 Hspa4*

HAEQNGPVDGQGDNPGpSQAAEHGADTAVPSDGDK response to stress S817 23.91 2.0 0.046

Q920Q8 Ivns1abp NpSPQp(SSPTSTPK)

negative regulation of

apoptotic process S282 58.33 6.3 0.034

Q6ZQ58 Larp1 DGAEREpSPRPPAAAEAPAGpSDGEDGGRR macroautophagy S68, S81 1,000, 1,000 2.9 0.036

Q05CL8 Larp7 pp(TASEGSEAETPEAPKQPAK) RNA processing 4.4 0.048

P14873 Map1b VLpSPLRpSPPLLGSESPYEDFLSADSK

cytoskeletal

organization S1391, S1395 81.66, 77.60 8.1 0.009

E9PVG7 Map4k4 RDpSPLQGGGQQNSQAGQR

protein

phosphorylation S587 78.73 2.2 0.027

E9QKD1 Nol8 SSMpSDDDVDpSEDELK DNA replication S318, S324 17.01, 106.20 16.8 0.010

Q9R0L6 Pcm1 pp(NVRSDVSDQEEDEESERCPVSINLSK)

Centrosome

organization 26.9 0.004

Q3UHX2 Pdap1 SLDpSDEpSEDEDDDYQQK N/A S60, S63 20.05, 63.94 5.3 0.034

Q99JF8 Psip1 p(QSNAS)pSDVEVEEK Transcription S106 32.40 3.2 0.023

Q8K094 Pvr ENVQYSpSVNGDCR cell-cell adhesion S393 20.35 3.0 0.042

Q64012 Raly LPAPQEDpTASEAGpTPQGEVQTR mRNA processing T268, T274 55.44, 26.42 2.8 0.018

Q9ERU9 Ranbp2 pQNQPTSCVSAPAp(SSETSRSPK) protein transport 2.2 0.032

Q5U4C3 Scaf1 APpSPAPAVpSPK mRNA processing S676, S682 1,000, 1,000 13.8 0.046

Q8BP27 Sfr1 pp(ENPPSPPTSPAAPQPR) DNA repair 2.5 0.039

Page 143: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

132

Q8VDQ8 Sirt2 VQEAQDpSDpSDTEGGATGGEAEMDFLR cell cycle S23, S25 12.57, 12.74 7.7 0.038

Q52KI8 Srrm1 pp(RLSPSASPPR) mRNA processing 6.5 0.042

Q52KI8 Srrm1 KEpTEpSEAEDDNLDDLER mRNA processing T876, S878 1,000, 1,000 4.0 0.037

Q8BTI8 Srrm2 ARpSRpTPPSAPSQSR mRNA processing S2264, T2266 103.22, 126.91 2.0 0.005

Q9CUU3 Sycp2*

p(SLS)PYPKAPpSPEFLNGNNSVVGR meiosis S1124 84.11 3.3 0.012

Q8BYJ6 Tbc1d4 HAp(SAPS)HVQPSDSEK

vesicle-mediated

transport 2.3 0.006

P39447 Tjp1 pSREDLSAQPVQTK

negative regulator of

vascular permeability S617 17.48 1.8 0.049

P83510 Tnik ANpSKp(SEGSPVLPHEPSK)

protein

phosphorylation S735, ?? 21.77 5.0 0.048

G5E870 Trip12 VREDDEDpSDDDGpSDEEIDESLAAQFLNSGNVR DNA repair S1350, S1355 27.86, 43.65 3.9 0.019

B2RUF0 Ybx2*

pTPGNQATAASGpTPAPPAR spermatid development T66, T77 40.19, 40.28 2.9 0.045

Page 144: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

133

Figure 4.6: Example of Skyline MS1 filtering output. Skyline MS1 filtering identified a peptide

NSVLSDPGLDpSPQTpSPVILAR (which maps to ARHGEF11) that was more abundant in Ppp1cc

mutant testis samples than in wild-type. A) Histogram showing XIC peak area calculations across all six

replicates reveals that this peptide was more abundant in all Ppp1cc knockout testis samples. B)

Retention times for selected peaks are consistent across all 6 replicates giving confidence in Skyline

MS1 peak selection.

Page 145: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

134

Page 146: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

135

Figure 4.7: Quantitative comparison of XIC peak areas of hyperphosphorylated phosphopeptides

mapping to genes essential for spermatogenesis and known/predicted PP1 interacting proteins. HMGA1,

YBX2, HSPA4, SYCP2 are essential for mouse spermatogenesis, MAP1B is a known PP1 interacting

protein. HSPA4 and SIRT2 are predicted PP1 interacting proteins. Peptides from table 4.2 mapping to

these proteins were more abundant in Ppp1cc mutant testis samples than in wild-type. Bars represent

average XIC peak areas across 3 biological replicates of each genotype +/- standard error.

Page 147: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

136

Page 148: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

137

analysis revealed several biological processes and molecular functions to be significantly enriched in this

set of genes, primarily involving RNA processing (Table 4.3).

A large portion (approximately 90%) of all currently known PIPs contain a short degenerate RVxF

docking motif ([KR]-X(0,1)-[VI]-{P}-[FW]) which is the principal site of interaction with

the phosphatase. However, as a caveat, the presence of this motif does not prove interaction with PP1

isoforms, as the motif is frequently observed throughout the proteome (~25% of all proteins). In many

instances, PIPs are thought to act as regulatory subunits and by binding to the RVxF motif target PP1

catalytic subunits to dephosphorylate specific substrates. However, this is not always the case, as many

known PP1 substrates also contain RVxF motifs (Bollen et al. 2010). Seventeen of the 32 (53%)

candidate PPP1CC2 substrate proteins identified in this study contain RVxF motifs (34 instances). PP1

docking motifs often reside in flexible, exposed surface regions of proteins as opposed to inside globular

domains (Egloff et al. 1997, Hendrickx et al. 2009). Ignoring instances of RVxF motifs inside globular

domains, there remain six candidate PPP1CC2 substrate proteins with predicted accessible docking

motifs—PVR, RANBP2, SIRT2, SRRM2, TBC1D4, and the known interactor MAP1B. The presence of

this motif in an accessible region of a substrate protein may be indicative of a stronger association with

the phosphatase catalytic subunit than is typical of PP1 substrates.

4.4 Discussion

In a landmark study, Bellve et al. (1977) isolated and characterized the cell type composition of

the prepubertal mouse testis throughout the first 20 days postnatal. The first wave of mouse

spermatogenesis begins one week post-natal (p.n.). Until that point, the only germ cells present in the

testis are Spermatogonia (Type A at six days p.n. and Types A and B at eight days p.n.). By day 10,

meiosis has begun, with preleptotene and leptotene spermatocytes first being observed. Subsequently,

zygotene and early pachytene spermatocytes are first found at days 12 and 14 respectively. Developing

germ cells reach the late pachytene stage between days 18 and 20. Day 20 marks the completion of the

Page 149: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

138

Table 4.3: GO biological processes and molecular functions enriched in the hyperphosphorylated dataset

relative to a full mouse background dataset. Data from DAVID Functional Annotation Tool.

Page 150: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

139

Biological Process/Molecular Function Proteins

(no.) P-value

mRNA metabolic process 5 1.10E-03

RNA binding 6 2.70E-03

RNA splicing 4 3.00E-03

RNA processing 5 4.10E-03

mRNA processing 4 7.30E-03

small GTPase regulator activity 3 4.40E-02

Page 151: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

140

first wave of meiosis, with secondary spermatocytes and the first round spermatids beginning to appear

at this date. By four weeks of age the mice are sexually mature, and a full complement of germ cell

types are found in the testis. To date, several studies have examined the expression pattern of Ppp1cc1

and Ppp1cc2 in the mouse testis, primarily at the protein level. An early study by Takizawa and co-

workers (Takizawa et al. 1994) performed western blots on various mouse tissues for PP1 isoforms and

found that both PPP1CC isoforms, as well as PPP1CA and PPP1CB were expressed in the mature mouse

testis, with PPP1CC2 being expressed at a very high level. Another study, by Chakrabarti and co-

workers (Chakrabarti et al. 2007b) found that both PPP1CC1 and PPP1CC2 were both detectable in the

mouse testis at eight days p.n. via western blot (the first time point examined) and that the PPP1CC2

protein level increased between the ages of 8 and 30 days p.n. indicating a higher degree of PPP1CC2

expression in later stage germ cells. This is consistent with our qPCR data, which is important

confirmation as the correlation between mRNA and protein levels can often be poor in the testis (Cagney

et al. 2005). Immunohistochemical (IHC) analysis of PPP1CC2 in the mouse testis shows high PPP1CC2

expression in all germ cell stages from primary spermatocytes onwards, with lower expression levels in

spermatogonia as well as in the somatic Sertoli cells (Hrabchak &Varmuza. 2004). PPP1CC1 has been

observed via IHC to be expressed at low levels in all germ cell stages, as well as in at higher levels in

interstitial cells (Chakrabarti et al. 2007b). Analysis of PPP1CA expression in the testis has shown

expression throughout spermatogenesis, including in condensing spermatids (Varmuza et al. 1999). In

the existing body of literature of PP1s in the testis, there have been multiple claims that PPP1CC2 is the

only PP1 isoform present in mammalian spermatozoa; however, several proteomic studies indicate that

this is not actually the case (Baker et al. 2007, Baker et al. 2008b, de Mateo et al. 2011). It is apparent

from these previous works that PP1 isoforms are differentially expressed in the mouse testis (Takizawa et

al. 1994, Varmuza et al., 1999, Chakrabarti et al., 2007b). However, studies such as

immunohistochemistry and western blots are only semi-quantitative at best, and can lead to discrepancies

in descriptions in expression patterns. The results of our study prove definitively that the expression of

the Ppp1cc2 isoform is specifically upregulated beginning at 3 weeks post-natal.

Page 152: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

141

Histological examination of the 3 week old Ppp1cc mutant testis has confirmed previous

observations (Varmuza & Ling 2003) that the earliest phenotypic effects of this mutation are visible by 3

weeks of age, corresponding to upregulation of the Ppp1cc2 isoform. At this age, many seminiferous

tubules retain normal architecture, however, others are beginning to prematurely shed germ cells, and

show signs of seminiferous tubule degeneration. To date, a large portion of the research regarding

PPP1CC2’s role in mammalian spermatogenesis has focused on a later established role in motility, but it

is clear that PPP1CC2 is first required much earlier in spermatogenesis. In order to learn more about the

function(s) of PPP1CC2 at this stage in spermatogenesis, this study sought to identify candidate

PPP1CC2 substrates via a comparative phosphoproteomic analysis at a stage when minimal cellular

differences between mutant and wild type would be expected to confound results. For an informative

comparison to be made, this time point was pivotal.

The experiments described in this study represent to our knowledge, the largest comparative

analysis of the mouse testis phosphoproteome to date. Previous large-scale analyses of different mouse

tissues has produced two very large testis phosphoproteome datasets (Huttlin et al. 2010, Lundby et al.

2012b), including one of the three week testis, however, these experiments focused on tissue-tissue

variation as opposed to an examination of the testis under pathological (i.e. mutant) conditions. Despite

the size of these datasets almost ¼ of the phosphoprotein gene symbols (179/755) identified in our

dataset were not listed in the 3 week testis by Huttlin et al. including four proteins which we found to be

more abundant in Ppp1cc mutant testes (SFR1, MAP1B, PVR and GM14569). Differences in the

formatting of results likely accounts for some of the variation, as conversion between different types of

gene/protein identifiers can be problematic. At the peptide level of 1261 unique peptides in our dataset

(ignoring phosphorylation site assignment) 27.6% were not identified by Huttlin et al. including 7 of the

33 hyperphosphorylated testis peptides (mapping to BCLAF1, GM14569, HSPA4, ACIN1, LARP1,

TBC1D4 and PVR). One potential reason for this is differences in the phosphopeptide enrichment

strategy; Huttlin employed only IMAC, while we used both IMAC and TiO2 chromatography. It has been

Page 153: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

142

established experimentally that each phosphopeptide enrichment strategy isolates a distinct overlapping

fraction of the total phosphoproteome (Bodenmiller et al., 2007). This illustrates that, while

phosphoproteomic datasets continue to increase in size, covering the entire phosphoproteome by any one

method, is currently not technically feasible. When our dataset is compared to the previous

phosphoproteomic dataset from our lab, only 7 proteins were common to both sets, and none identified

as being more abundant in 3 week Ppp1cc mutant testes (Henderson et al. 2011, Henderson 2007). This

lack of overlap is likely due to several factors. First the previous dataset is small (only 57 proteins) and

many of the proteins were identified from gel spots picked from 2-D gels after phospho-specific staining,

that contained multiple proteins, and thus may not even be phosphorylated. In addition, this experiment

was conducted using adult testes which contain cell types and therefore proteins/phosphorylation events

not present in the 3 week testis.

Our testis phosphoproteome data, the product of efficient phosphopeptide enrichment, displays a

high degree of reproducibility, making it possible to perform an informative comparison of the wild-type

and Ppp1cc mutant phosphoproteomes. To identify phosphopeptides that were more abundant in the

Ppp1cc mutant testis, we utilized a combination of multiple, label-free quantitation methods. While

many quantitative proteomic and phosphoproteomic studies use metabolic labelling strategies such as

SILAC, label-free methods can provide a powerful alternative, that is often more suitable to tissue

samples from mammalian models. Our analysis strategy consisted of two stages (Figure 4.4) in which a

preliminary semi-quantitative analysis was used to develop a list of peptides for further, more rigorous

quantitative analysis. The use of a two-stage analytical strategy was helpful in reducing the amount of

data processing in the MS1 filtering stage. While Skyline automatically assigns and aligns MS peaks,

manual inspection of this data for each peptide was performed to ensure that accurate retention times

were selected for alignment. By only performing a semi-quantitative analysis on a large portion of the

data, it could be argued that a significant amount phosphoproteomic data was not thoroughly analyzed

quantitatively. However, we feel that our two stage analytical strategy allowed us to extract the strongest

Page 154: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

143

candidate PPP1CC2 substrates from our dataset whilst reducing the amount of data processing needed to

a manageable amount.

Our quantitative phosphoproteomic analysis of the mouse testis identified 33 different peptides

in 32 proteins that were more abundant in 3 week old Ppp1cc mutants. These mutants lack the

expression of a phosphatase gene and thus, the identified proteins represent candidate PPP1CC2

substrates. As a caveat, our data does not demonstrate direct dephosphorylation of any of these proteins

by PPP1CC2, but instead provides a strong list of candidates for further study. Previous

phosphoproteomic studies have demonstrated that there are thousands of phosphorylated proteins in the

mouse testis, even at 3 weeks of age (Huttlin et al. 2010). Furthermore, interactions between PP1

catalytic subunits and their substrates are often weak and transient, making substrate identification based

on interaction studies challenging. For these reasons, we feel that our study represents an important step

in the substrate identification process by developing a list of candidate PPP1CC2 substrates that is of a

suitable length to test individually over the course of future studies. As is the case for many quantitative

phosphoproteomic studies, it is difficult to discern instances of true hyperphosphorylation from

differences in protein abundance due to the fact that so few unique peptides are isolated for each

individual protein. It is likely that some of these observed changes in phosphopeptide abundance derive

from indirect effects secondary to the loss of Ppp1cc in the testis. However, by performing our

phosphoproteomic analysis at the time point where the earliest phenotypic changes are observed, we

have kept these instances to a minimum. Most of the candidate PPP1CC2 substrates identified (21/32)

were represented by more than one unique phosphopeptide in our dataset. For all but two of these

proteins (TJP1 and LARP7) these additional peptides did not appear to show increased abundance in

Ppp1cc mutants, indicating the likelihood of hyperphosphorylation at the specific site(s) indicated.

Eleven of the candidate PPP1CC2 substrate proteins (ARHGEF11, CENPV, DNAJC5, IVNS1ABP,

LARP1, MAP4K4, PVR, RANBP2, SCAF1, SIRT2, and TBC1D4) were represented by a single unique

peptide and we were therefore unable to rule out changes in protein abundance. Future studies evaluating

Page 155: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

144

all of these candidate substrates should first aim to rule out this alternate explanation using techniques

such as western blotting or quantitative mass spectrometry of non-phosphopeptide enriched samples.

Nonetheless, it is worth noting that even indirect misregulation of the phosphorylation status or

abundance of proteins can provide information regarding pathways or processes affected by the loss of

Ppp1cc in the testis.

Amongst the identified candidate testis substrates is one known PP1 interacting protein (PIP),

MAP1B a microtubule binding protein to which PP1 isoforms have been demonstrated to bind and even

dephosphorylate in vitro (Ulloa et al. 1993, Hendrickx et al. 2009). MAP1B has no known role in

spermatogenesis, but an interaction between PPP1CC2 and another microtubule associated protein

PPP1R42 (TLRR) has been established and was shown to be most active at 3 weeks in the testis (Wang

et al. 2010, Wang & Sperry 2011). Furthermore, a previous study from our lab has demonstrated

hyperphosphorylation and disrupted localization of β-tubulin in the Ppp1cc knockout testis as well as an

association with PPP1CC2 (Henderson et al. 2011). Several additional candidate substrates have links to

PP1 via shared interacting proteins including predicted PIP, SIRT2 (HDAC6; Nahhas et al., 2007)),

SRRM1 (TRA2B; Long & Caceres 2009), and TJP1 (OCLN; Li et al., 2004). TJP1, which is important

in the regulation and maintenance of the Sertoli-cell junctions in the testis (Byers et al. 1991) also

associates with two of the other hyperphosphorylated proteins identified in this study—HSPA4 and

ARHGEF11 (data from Uniprot database www.uniprot.org).

Several of the candidate PPP1CC2 substrates identified in this study are known to be essential

for spermatogenesis. Targeted deletion of Hspa4 results in male specific infertility due to a partial arrest

in prophase I of meiosis, resulting in widespread apoptosis in late pachytene spermatocytes (Held et al.

2011). This phenotype has many similarities to the Ppp1cc mutant phenotype with widespread loss of

developing germ cell populations, the presence of immature germ cells in the epididymis and defects in

chromatin condensation, however, the most severe loss of germ cells occurs slightly earlier in Hspa4

knockouts than in Ppp1cc (Varmuza et al. 1999, Held et al. 2011). The Hspa4 mutant phenotype is also

Page 156: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

145

highly similar to that of Hspa2 (Dix et al. 1996) which was previously identified as a candidate

PPP1CC2 substrate and interactor and was shown to be abnormally distributed in Ppp1cc mutant

spermatogenic cells (Henderson et al. 2011). Chimeric mice harbouring a deletion of another

hyperphosphorylated testis protein HMGA1 were also observed to have disrupted spermatogenesis, with

widespread loss of germ cells beginning with spermatocytes, resulting in azoospermia (Liu et al. 2003).

SYCP2 is a component of the synaptonemal complex during meiosis, and targeted deletion of this gene

results in a complete meiotic arrest (Yang et al. 2006). Recent research has demonstrated that many

proteins in the meiotic chromosome axis, including SYCP2 become phosphorylated during prophase I of

spermatogenesis and it has been hypothesized that phosphorylation is important in regulating

chromosomal events at this juncture (Fukuda et al. 2012). Deletion of another candidate PPP1CC2

substrate gene, Ybx2 (Msy2) results in the complete absence of mature sperm in male mice due to

abnormalities in both meiotic and post-meiotic cells (Yang et al. 2005). Ybx2 knockout seminiferous

tubules display an increase in apoptosis in spermatocytes, and later a block in spermatid elongation.

Multinucleated spermatids were frequently observed, as were abnormally shaped and variably condensed

spermatid nuclei, features that are also observed in Ppp1cc mutants (Varmuza et al. 1999, Yang et al.

2005). Aside from the above-mentioned proteins that are encoded by genes shown to be essential for

spermatogenesis, several other candidate PPP1CC2 substrates identified in this study have also been

linked to spermatogenesis: LARP7 (Okamura et al. 2012), PSIP1 (Sutherland et al. 2006), PVR

(Wakayama et al. 2007) and RANBP2 (Nagai et al. 2011).

Spermatogenesis is a complex developmental process involving the coordination of hundreds, if

not thousands, of proteins. While the identities of many of these proteins are known, the specific roles

they play, and how they are regulated remains, in most cases, unknown. The results of this study will

help to shed some light on the role of one of these proteins, the testis specific protein phosphatase

PPP1CC2. Our comparative phosphoproteomic analysis of the Ppp1cc mutant testis has produced a list

of candidate substrates of a practical length for future analysis, including a number of proteins with

Page 157: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

146

known or potential links to spermatogenesis. Future studies will seek to verify which of these candidates

are direct PPP1CC2 substrates, and determine their roles in the regulation of mammalian

spermatogenesis.

Page 158: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

147

Chapter 5: Conclusion

Authors note: Portions of this section are included in a mini-review article currently in review for

publication in FEBS Journal. I was responsible for writing this section, with editing from S. Varmuza.

Full citation: G. MacLeod and S. Varmuza (2013). The application of proteomic approaches to the study

of mammalian spermatogenesis and sperm function. FEBS Journal [In review].

5.1 The Application of Proteomic Approaches to the study of mammalian spermatogenesis

To better understand the process of spermatogenesis, we must uncover which genes are involved,

what roles they play and how they are regulated. Emerging proteomic technologies can provide a number

of useful tools for studying mammalian spermatogenesis. The use of proteomics is particularly important

for spermatogenesis, because the correlation between RNA and protein expression is lower in the testis

than in other tissues (Cagney et al. 2005), indicating that microarray studies are less informative in this

context. Compounding these difficulties further are transcriptional silencing and storage of earlier

produced transcripts in the later stages of spermatogenesis, and well as the abundance of tissue-specific

alternative splicing observed in the testis (Yeo et al. 2004). Despite its importance, we have only recently

begun to scratch the surface of the potential of proteomic research in application to spermatogenesis.

Throughout the course of my thesis research, I have applied several of these proteomic techniques aimed

at studying the role of PPP1CC2 in mouse spermatogenesis.

Each spermatogenic cell type represents a step towards the production of spermatozoa, and thus,

by characterizing the proteome of the different cell types we can gain insight into the genes and proteins

involved in each step. Current estimates suggest that the human sperm proteome contains roughly

2500-3000 proteins (Baker and Aitken 2009); however, less differentiated spermatogenic cells may

contain a much higher number. All of the major types of spermatogenic cells—spermatogonia

(Guillaume et al. 2000, Com et al. 2003, Dihazi et al. 2009), spermatocytes (Guo et al. 2011), spermatids

(Guo et al. 2010) and spermatozoa (Baker et al. 2008a) have been isolated and subjected to analysis in

Page 159: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

148

order to characterize their proteomes. Some experiments have examined even narrower classes of

spermatogenic cells, such as those described by Delbes et al., (2011) who performed a proteomic

analysis of elongated spermatids. The utility of proteomic analysis of whole testes or isolated germ cell

populations goes beyond simply cataloging the proteome of the cell types. Proteomic approaches have

also been used to ask specific biological questions by an increasing number of groups. For example, the

previously mentioned elongating spermatid proteome investigation by Delbes et al., compared the

proteomic profiles of wild-type and Paip2a/Paip2b double knockouts to identify 29 differentially

expressed proteins (Delbes et al. 2011).

By breaking the cell down into its constituent parts (be they fractions, structural elements or

organelles) we can gain an even better coverage of the proteome and learn about the localization of

proteins within the spermatogenic cell types, which will help us to gain functional insight. In fact, the

spermatozoa are uniquely suited to this form of analysis. Being arguably the most differentiated cell type

in the body, they contain a number of different component parts that can be readily isolated and subjected

to proteomic analysis. To date, subcellular spermatozoa fractions examined in proteomic studies include

the sperm surface (Nixon et al. 2009, Asano et al. 2010, Belleannee et al. 2011, Gu et al. 2011, Nixon et

al. 2011, Byrne et al. 2012), the acrosomal matrix (Guyonnet et al. 2012), the sperm tail (Amaral et al.

2013), the sperm nucleus (de Mateo et al. 2011) and sperm chromatin binding proteins (Govin et al.

2012).

Post-translational modifications in spermatogenic cells

Another area of proteomics that has garnered increased interest over the past several years is the

study of post-translational modifications (PTMs). Once translated in the cell, proteins can be covalently

modified in a number of different ways, which can govern the activity of proteins and signalling

networks. Identifying PTMs of testis proteins gives us a more detailed picture of how those proteins exist

in the tissue, and can offer clues as to their functions. As well, by characterizing changes in PTM status

Page 160: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

149

in response to different stimuli or at different time points in spermatogenesis we can gain even further

insight into the regulation of a protein’s activity. The analysis of PTMs is especially relevant in mature

spermatozoa, as they are both transcriptionally and translationally silent, meaning the entirety of cellular

functions must be governed by post-translational activity.

Phosphorylation is by far the most studied PTM in spermatogenesis. Following recent

technological advances in the isolation of phosphorylated peptides such as those by Larsen et al. (2005),

the field of phosphoproteomics has experienced significant growth over the past several years. The study

of spermatogenesis has been no exception to this, as an increasing number of groups have performed

phosphoproteome analysis in sperm and spermatogenic cells. However, the largest existing testis

phosphoproteomic datasets were not produced from labs focusing directly on spermatogenesis, but on

large scale studies aiming to map phosphorylation sites across multiple tissues. One large scale analysis

of nine tissues from 3 week old mice identified over 2,500 phosphoproteins in the testis corresponding to

over 10,000 phosphorylation sites after immobilized metal ion affinity chromatography (IMAC)

phosphopeptide enrichment (Huttlin et al. 2010). Another phosphoproteomic study utilized titanium

dioxide (TiO2) phosphopeptide enrichment on 14 different tissues and organs in the rat, and also

identified over 10,000 phosphorylation sites in the testis, including over 200 testis specific

phosphorylation sites (Lundby et al. 2012b). These studies, while not directly focused on

spermatogenesis, represent a wealth of data to the research community and have provided insight into

post-translational regulation of spermatogenesis. As an example, both studies found that the testis was

second only to the brain in the number of tissue specific phosphorylation sites (17% of all identified sites

in one experiment (Huttlin et al. 2010)), further emphasizing the importance of protein phosphorylation

in spermatogenesis.

Aside from these large scale analyses of the mammalian testis, most groups have chosen to focus

on spermatozoa for phosphoproteomic analyses due to their ease of retrieval and

transcriptional/translational silent status. Baker et al., have published 2 separate examinations of

Page 161: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

150

changes in protein phosphorylation during rat epididymal maturation using TiO2 phosphopeptide

enrichment, identifying 53 (Baker et al. 2011) and 22 (Baker et al. 2012) differentially phosphorylated

proteins. Perhaps the most extensively studied aspect of sperm maturation via phosphoproteomic

analysis is capacitation, which has been examined in mouse (Platt et al. 2009), rat (Baker et al. 2010) and

human samples (Ficarro et al. 2003). Another investigation has even performed phosphoproteomic

analysis on clinical samples, comparing sperm from fertile individuals with those suffering from

asthenozoospermia and identified 66 differentially phosphorylated peptides (Parte et al. 2012).

In contrast to those groups examining spermatozoa, our group has focused on the response of

developing spermatogenic cells in the testis to the loss of the protein phosphatase Ppp1cc, and in one

previous study identified 10 proteins hyperphosphorylated in response to this deletion (Henderson et al.

2011). Furthering this line of inquiry, I conducted a phosphoproteomic analysis (Chapter 4) of the 3 week

old mouse testis. To our knowledge, this dataset represents the largest comparative phosphoproteomic

analysis of the mouse testis under pathologic (i.e. mutant) conditions conducted to date. This study

identified 755 different phosphorylated proteins in the 3 week old mouse testis, and confidently assigned

1,026 unique phosphorylation sites. This dataset represents one of the largest collections of mouse testis

phosphoproteomic data available, and could provide a wealth of information to researchers interested in

spermatogenesis. Quantitative analysis of our data identified 33 different phosphorylated peptides in 32

different proteins that were more abundant in Ppp1cc mutant samples than in wild-type. These

hyperphosphorylated proteins represent candidate PPP1CC2 substrates in the testis. This combination of

shotgun phosphoproteomics with a specific biological question has yielded an excellent set of candidate

genes that will form the basis of future studies regarding the role of PPP1CC2 in spermatogenesis. In

addition to this large scale phosphoproteomic study, we used phosphoproteomic analysis in other studies

to look at specific proteins of interest, identifying a novel phosphorylation site important in the

regulation of the interaction between PPP1CC2 and TSKS (Chapter 3), and showing that a protein in

complex with newly identified PIP DDOST is phosphorylated in the testis (Chapter 2).

Page 162: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

151

In addition to the growing number of phosphoproteomic studies relating to spermatogenesis and

sperm function, a series of other types of PTMs have also been catalogued including lysine acetylation

(Lundby et al. 2012a), glycosylation (Kaji et al. 2012, Tian et al. 2010), S-nitrosylation (Lefièvre et al.

2007) and most recently SUMOylation (Vigodner et al. 2012). It is clear from these studies that the

nature of a protein does not end when it is translated—PTMs confer an extremely high diversity of

protein forms within a given cell type; a complexity that must be better understood for deciphering a

protein’s function. Fortunately, technological improvements in mass spectrometry instruments, data

analysis tools, and protein enrichment are moving us closer to this goal.

Protein-protein interaction networks (interactomes) in spermatogenesis

Proteins rarely if ever function in isolation—it is the interaction between numerous protein

species that carries out functions. Thus, to fully understand a process as complex as spermatogenesis, we

must not only look to what proteins are present in a given space and how they are modified, but to which

proteins they interact with—the interactome. By characterizing interactomes, that is the full complement

of protein-protein interactions for a given protein, we have a much better chance of understanding a

protein’s function than if we looked at each in isolation. In the past, the most common approach to

interactome studies was the yeast 2-hybrid assay. While this approach has been quite successful in

identifying protein-protein interactions, even with regard to genes important to spermatogenesis (for

example Hrabchak & Varmuza 2004, Hrabchak et al. 2007, Fardilha et al. 2011b) it has several

prominent limitations due to the artificial nature of the system (for instance, the absence of species/tissue

specific PTMs) and the fact that it only looks at binary interactions. Currently, the yeast 2-hybrid system

has been largely replaced by affinity-purification/mass spectrometry (AP-MS) based approaches,

featuring either single, or tandem affinity (TAP) tags. These approaches are typically applied to

mammalian tissue culture systems and can simultaneously identify a large number of protein-protein

interactions including multi-protein complexes. AP-MS when applied with the appropriate controls, also

results in the identification of fewer false positive interactions, and use of a mammalian system is for

Page 163: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

152

obvious reasons preferable to a yeast-based system (see Chen &Gingras. 2007) for an overview of such

systems).

As outlined in the preceding sections, the testis is particularly abundant in tissue specific protein

expression as well as PTMs, and the complex architecture of the testis cannot be modelled in culture.

Thus, if looking to define the interactome of a protein involved in spermatogenesis using tissue culture, it

is likely that a large amount of information is being missed. For this reason it is important to conduct

interactome studies directly in the testis when possible. This prospect is significantly more demanding

than tissue culture based approaches due to the need to either have highly specific antibodies, or the

ability to generate a transgenic line with an affinity-tagged version of the gene of interest. Despite its

technological demands, a number of groups have successfully performed interactome studies in the

testis. In 2009 Chen et al., used immunoprecipitation followed by gel-free LC-MS/MS to characterize

the interactomes of germline-specific Argonaute family members MIWI and MILI in the mouse testis

(Chen et al. 2009). The authors then went on to perform a reciprocal experiment using one of their

identified interactions partners, TDRKH, and characterized a multi-protein interaction network in the

testis. Similarly, another experiment used a modified immunoprecipitation and mass spectrometry

approach to identify an additional member of the CATSPER complex in the mouse testis (Chung et al.

2011). The authors of this study had undertaken this approach due to their inability to successfully

express the CATSPER complex in any other system, which underscores the importance of examining

protein interactions in their natural environment. Other groups have generated transgenic mouse lines in

order to perform tandem affinity purification directly in the testis. To identify novel interactors involved

in Bardet-Biedl syndrome, Seo et al. generated a mouse line expressing a green fluorescent protein

(GFP) and S-tag coupled BBS4 construct, and performed tandem affinity purification in transgenic

testes, successfully identifying a novel protein complex member (Seo et al. 2011). Another TAP

experiment in the testis using a mouse expressing TAP-tagged 14-3-3ζ identified a large number of novel

Page 164: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

153

protein-protein interactions, although the lack of appropriate control experiments leaves the total number

of true interactors in question (Puri et al. 2011).

To further define the interactome of PPP1CC2 in the testis, we endeavored to produce a TAP-

tagged PPP1CC2 knock-in mouse model (Chapter 2). To do this, we first generated a vector system for

producing SBP-3XFLAG tagged versions of any gene using gene-trap ES cell lines. Our method was

based on the Floxin system (Singla et al. 2010) from which we obtained our plasmid vector backbone.

This system can serve as a useful resource for other groups that wish to conduct interactome studies in

transgenic tissues. Via this method SBP-3XFLAG-PPP1CC1 and PPPCC2 transgenic ES cell lines were

produced. Subsequently, we used these cells in an attempt to create a transgenic mouse line, but chimeric

mice were unable to transmit the transgene through the germline, possibly due to dominant negative

effects. However, TAP studies in the transgenic ES cells successfully identified a novel PIP, DDOST

with a potential role in spermatogenesis. To further study the interactome of PPP1CC2 directly in the

testis, we performed GST pull down assays and identified several novel candidate PPP1CC2 interacting

proteins, two of which, TSSK1 and TSKS, were validated and studied further (Chapter 3). The different

proteins identified in each approach underlines the importance of conducting multiple approaches to

studying the interactome of any protein, as no one approach is likely to identify all interactors.

Furthermore, the fact that testis specific proteins were identified as PPP1CC2 interactors in our pull

down assays highlights why performing interactome studies in a biologically relevant context is the ideal

experiment.

The utilization of proteomics in clinical studies of male infertility

The ultimate aim in furthering our understanding of spermatogenesis is to improve our ability to

diagnose and treat male infertility. In recent years a number of investigators have applied proteomic

analysis to clinical samples in an effort to go beyond basic research. By analyzing the sperm proteomes

of infertile men, we may be able to detect aberrations to explain impairments, and evaluate the feasibility

Page 165: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

154

and course of future treatment. While it is difficult to obtain a sufficient amount of starting material from

spermatogenic cells in the testes of infertile men, ejaculated spermatozoa and seminal fluid are much

easier to obtain, and thus have constituted the bulk of the studies to date.

The proteome of sperm from infertile donors has been examined by a number of groups, to date.

Experiments have identified proteins differentially expressed in patients exhibiting generalized infertility

(de Mateo et al. 2011, Xu et al. 2012), low sperm count and motility (Thacker et al. 2011),

asthenozoospermia (Zhao et al. 2007, Martínez-Heredia et al. 2008, Siva et al. 2010), Sertoli Cell Only

Syndrome (Li et al. 2012), in vitro fertilization failure (Pixton et al. 2004), diabetes and obesity (Kriegel

et al. 2009). The seminal plasma provides a protective and facilitative environment for sperm transit that

is critical in a number of ways for proper sperm function. As such, a number of clinical studies have

applied proteomic analysis in the hopes of finding useful biomarkers for defects of the male reproductive

system. (Pilch & Mann 2006, Wang et al. 2009, Batruch et al. 2011, Batruch et al. 2012, Davalieva et al.

2012, Kagedan et al. 2012, Milardi et al. 2012). From these studies a large number of biomarkers have

been identified, including those useful in discrimination between obstructive and nonobstructive

azoospermia which provide a non-invasive diagnostic alternative to the current practices of biopsy and

histology. The routine use of proteomic analysis in a clinical setting may be practical in the near future.

Alternatively, experiments such as those listed above can be used as discovery tools to produce smaller

biomarker panels which are simpler to use and interpret, less expensive, and thus easier to institute in a

clinical setting.

5.2 Significance and future directions

The experiments described in this thesis have greatly increased our knowledge of PPP1CC2

protein-protein interactions in the mouse testis. Three novel PPP1CC2 interactors in the testis have been

validated (DDOST, TSSK1 and TSKS) and four more potential interactors were identified (ATP5C1,

UQCRC2, SCCPDH and FADS2). In another experiment, from thousands of phosphorylated proteins in

Page 166: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

155

the mouse testis, we have identified 32 that are hyperphosphorylated in response to deletion of Ppp1cc

making them excellent candidate substrates. In addition to this new information regarding protein-protein

interactions, this thesis contains a number of additional contributions to the body of literature regarding

the role of PPP1CC2 in spermatogenesis, as well as to the research community in general. qPCR

analysis has characterized the expression levels of all four PP1c isoforms in the first wave of

spermatogenesis, and demonstrated specific upregulation of Ppp1cc2 at both the transcriptional and

splicing levels beginning at 3 weeks of age. Histological analysis of the mouse testis has verified that the

first phenotypic effects of Ppp1cc deletion are present by 3 weeks of age in the testis, and further

characterized this phenotype. Apart from the identification of candidate PPP1CC2 substrates, our testis

phosphoproteomic dataset represents one of the largest of its kind, and contains a large amount of data

that will be useful to the scientific community. Finally, the SBP-3XFLAG knock-in vector produced for

TAP experiments represents a useful tool for the research community to explore the interactome of any

gene.

Taking all of this information together, we can draw several important conclusions regarding the

function of PPP1CC2 in spermatogenesis. First, we have proven definitively that PPP1CC2 has a role

early in spermatogenesis, by 3 weeks of age. Isoform specific upregulation of the Ppp1cc2 transcript and

the presence of a knockout phenotype by three weeks of age show conclusively that PPP1CC2 is

required at this stage of spermatogenesis. At this point, the late spermatocyte is the predominant germ

cell type present and only the very first round spermatids have appeared. Second, this early role for

PPP1CC2 in spermatogenesis is unlikely to be its only function. Previous studies have demonstrated a

role for PPP1CC2 in the acquisition of spermatozoan motility, which occurs long after its first

requirement in the testis (reviewed in Fardilha et al. 2011a). Furthermore, our identification of a complex

containing PPP1CC2, TSKS, and TSSK1 suggests an additional role in the mid to late stages of

spermiogenesis, as TSKS and TSSK1are not even expressed in the testis at 3 weeks of age. Further

support for this model is found in transgenic Ppp1cc2 knock-in mice. Transgenic expression of Ppp1cc2

Page 167: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

156

in a Ppp1cc knockout background is only able to completely rescue the infertility phenotype if

expression levels reach a certain threshold; ~50% of heterozygous protein levels (Sinha et al., 2012).

Below this expression level only a partial rescue is of the Ppp1cc mutant phenotype is observed (Soler et

al., 2009, Sinha et al., 2012). A low level of PPP1CC2 expression in the testis rescues extensive

depletion of spermatids but mice remain infertile due to abnormal spermatid morphology with defective

nuclear shaping and mitochondrial sheath morphology. This suggests the possibility of different

threshold levels of PPP1CC2 required at different points in spermatogenesis. One can envision a scenario

where a lower level of PPP1CC2 expression is required at 3 weeks of age for a function in early

spermatogenesis, but a higher level is required for an additional function in late spermiogenesis. This is

also consistent with the expression levels observed in our qPCR analysis of Ppp1cc2 in the first wave of

spermatogenesis. A pleiotropic nature of PPP1CC2 in the testis is not surprising, given the fact that PP1s

are involved in numerous and varied biological processes, and our research (and that of others) suggests

that this is the most likely scenario.

The knowledge gained from the research described in this thesis also produces a large number of

new questions regarding PPP1CC2’s role in spermatogenesis. These questions were discussed in detail

in previous chapter, but a few central ideas warrant future mention. Does the PPP1CC2-DDOST complex

play a role in spermatogenesis? The first step in determining this is to perform a targeted knockout or

tissue-specific deletion of Ddost to determine if it is essential for spermatogenesis, and if so characterize

the nature of the infertility phenotype. Another key question posed is what is the biochemical relationship

between members of the PPP1CC2-TSKS-TSSK1 complex, and how do they regulate one another?

Given that this complex contains both a Ser/Thr kinase and phosphatase it is logical to hypothesize that,

aside from the known phosphorylation of TSKS by TSSK1 there may be additional enzyme-substrate

relationships present. This could be tested by examining the phosphorylation status of the three proteins

in Tssk1/2 and Ppp1cc mutant backgrounds as well as using in vitro phosphatase and kinase assays. The

biological role of the PPP1CC2-TSKS-TSSK1 complex also requires further study. It appears that this

Page 168: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

157

complex may play a role in organizing the formation of the mitochondrial sheath. One interesting

experiment would be to perform a cross between the Ppp1cc and Tssk1/2 knockout models. If the

phosphatase and kinase play opposing roles in this process perhaps the double-knockout would result in

restoration of spermiogenesis. However, due to the more severe and early acting phenotype of the

Ppp1cc knockout, the use of a line with hypomorphic PPP1CC2 expression, such as those that resulted in

partial rescue of the spermatogenic impairment (Soler et al., 2009, Sinha et al., 2012) may be more

appropriate. Additional experiments should also include a more thorough examination of the

interactomes of all three proteins to determine whether there are additional members of this complex,

which could offer further clues as to its function in spermatogenesis. It remains to be determined, which

of the proteins hyperphosphorylated in the Ppp1cc mutant testis are true substrates of PPP1CC2? This

could be tested via in vitro phosphatase assays and/or interaction studies that show a direct interaction

between the phosphatase and the candidate substrates. And finally, is there an isoform specific role for

PPP1CC2 in spermatogenesis, or is there a threshold total PP1 dosage that is required for completion of

spermatogenesis? These central questions will need to be tested in future studies if we are to truly

understand the function of PPP1CC2. Developing a better understanding of mammalian spermatogenesis

including which genes are involved, and how they are regulated is needed in order to increase the

efficacy of the diagnosis and treatment of male infertility. As infertility affects the quality of life in

approximately 15% of all couples, basic research such as that which is described in this thesis is an

important step towards this goal.

Page 169: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

158

References

Amaral A, Castillo J, Estanyol JM, Ballesca JL, Ramalho-Santos J & Oliva R 2013 Human sperm

tail proteome suggests new endogenous metabolic pathways. Molecular & Cellular Proteomics 12 330-

342.

Angers S, Thorpe CJ, Biechele TL, Goldenberg SJ, Zheng N, MacCoss MJ & Moon RT 2006 The

KLHL12-Cullin-3 ubiquitin ligase negatively regulates the Wnt-[beta]-catenin pathway by targeting

Dishevelled for degradation. Nature cell biology 8 348-357.

Aranda B, Blankenburg H, Kerrien S, Brinkman FSL, Ceol A, Chautard E, Dana JM, De LR,

Dumousseau M, Galeota E, Gaulton A, Goll J, Hancock REW, Isserlin R, Jimenez RC,

Kerssemakers J, Khadake J, Lynn DJ, Michaut M, O'Kelly G, Ono K, Orchard S, Prieto C, Razick

S, Rigina O, Salwinski L, Simonovic M, Velankar S, Winter A, Wu G, Bader GD, Cesareni G,

Donaldson IM, Eisenberg D, Kleywegt GJ, Overington J, Ricard-Blum S, Tyers M, Albrecht M &

Hermjakob H 2011 PSICQUIC and PSISCORE: accessing and scoring molecular interactions. Nature

Methods 8 528-529.

Asano A, Nelson JL, Zhang S & Travis AJ 2010 Characterization of the proteomes associating with

three distinct membrane raft sub-types in murine sperm. Proteomics 10 3494-3505.

Baker MA, Hetherington L, Reeves GM & Aitken RJ 2008a The mouse sperm proteome

characterized via IPG strip prefractionation and LC-MS/MS identification. Proteomics 8 1720-1730.

Baker MA, Hetherington L, Reeves G, Müller J & Aitken RJ 2008b The rat sperm proteome

characterized via IPG strip prefractionation and LC-MS/MS identification. Proteomics 8 2312-2321.

Baker MA, Hetherington L, Weinburg A, Naumovski N, Velkov T, Pelzing M, Dolman S, Condina

MR & Aitken RJ 2012 Analysis of Phosphopeptide Changes as Spermatozoa Acquire Functional

Competence in the Epididymis Demonstrates Changes in the Post-Translational Modification of Izumo1.

Journal of Proteome Research 11 5252-5264.

Baker MA, Reeves G, Hetherington L, Müller J, Baur I & Aitken RJ 2007 Identification of gene

products present in Triton X-100 soluble and insoluble fractions of human spermatozoa lysates using LC-

MS/MS analysis. Proteomics - Clinical Applications 1 524-532.

Baker MA, Smith ND, Hetherington L, Pelzing M, Condina MR & Aitken RJ 2011 Use of Titanium

Dioxide To Find Phosphopeptide and Total Protein Changes During Epididymal Sperm Maturation.

Journal of Proteome Research 10 1004-1017.

Baker MA, Smith ND, Hetherington L, Taubman K, Graham ME, Robinson PJ & Aitken RJ 2010

Label-Free Quantitation of Phosphopeptide Changes During Rat Sperm Capacitation. Journal of

Proteome Research 9 718-729.

Batruch I, Lecker I, Kagedan D, Smith CR, Mullen BJ, Grober E, Lo KC, Diamandis EP & Jarvi

KA 2011 Proteomic Analysis of Seminal Plasma from Normal Volunteers and Post-Vasectomy Patients

Identifies over 2000 Proteins and Candidate Biomarkers of the Urogenital System. Journal of Proteome

Research 10 941-953.

Page 170: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

159

Batruch I, Smith CR, Mullen BJ, Grober E, Lo KC, Diamandis EP & Jarvi KA 2012 Analysis of

Seminal Plasma from Patients with Non-obstructive Azoospermia and Identification of Candidate

Biomarkers of Male Infertility. Journal of Proteome Research 11 1503-1511.

Beausoleil SA, Villen J, Gerber SA, Rush J & Gygi SP 2006 A probability-based approach for high-

throughput protein phosphorylation analysis and site localization. 24 1285-1292.

Belleannee C, Belghazi M, Labas V, Teixeira-Gomes A, Gatti JL, Dacheux J & Dacheux F 2011

Purification and identification of sperm surface proteins and changes during epididymal maturation.

Proteomics 11 1952-1964.

Bellve A, Cavicchia J, Millette C, O'Brien D, Bhatnagar Y & Dym M 1977 Spermatogenic cells of

the prepuberal mouse: isolation and morphological characterization. The Journal of cell biology 74 68-

85.

Beullens M, Van Eynde A, Vulsteke V, Connor J, Shenolikar S, Stalmans W & Bollen M 1999

Molecular Determinants of Nuclear Protein Phosphatase-1 Regulation by NIPP-1. Journal of Biological

Chemistry 274 14053-14061.

Bianchi M, De Lucchini S, Marin O, Turner DL, Hanks SK & -Villa-Moruzzi E 2005 Regulation of

FAK Ser-722 phosphorylation and kinase activity by GSK3 and PP1 during cell spreading and migration.

Biochemical Journal 391 359-- 370.

Bienvenu F, Jirawatnotai S, Elias JE, Meyer CA, Mizeracka K, Marson A, Frampton GM, Cole

MF, Odom DT, Odajima J, Geng Y, Zagozdzon A, Jecrois M, Young RA, Liu XS, Cepko CL, Gygi

SP & Sicinski P 2010 Transcriptional role of cyclin D1 in development revealed by a genetic-proteomic

screen. Nature 463 374-378.

Blendy JA, Kaestner KH, Weinbauer GF, Nieschlag E & Schutz G 1996 Severe impairment of

spermatogenesis in mice lacking the CREM gene. Nature 380 162-165.

Bodenmiller B, Mueller LN, Mueller M, Domon B & Aebersold R 2007 Reproducible isolation of

distinct, overlapping segments of the phosphoproteome. Nature Methods 4 231-237.

Bolino A, Bolis A, Previtali SC, Dina G, Bussini S, Dati G, Amadio S, Del Carro U, Mruk DD, Feltri

ML, Cheng CY, Quattrini A & Wrabetz L 2004 Disruption of Mtmr2 produces CMT4B1-like

neuropathy with myelin outfolding and impaired spermatogenesis. The Journal of cell biology 167 711-

721.

Bollen M 2001 Combinatorial control of protein phosphatase-1. Trends in biochemical sciences 26 426-

431.

Bollen M, Peti W, Ragusa MJ & Beullens M 2010 The extended PP1 toolkit: designed to create

specificity. Trends in biochemical sciences 35 450-458.

Bonilla E &Xu EY 2008 Identification and characterization of novel mammalian spermatogenic genes

conserved from fly to human. Molecular human reproduction 14 137-142.

Brautigan DL 2012 Protein Ser/Thr phosphatases -the ugly ducklings of cell signalling. FEBS Journal

280 324-325.

Brown KR &Jurisica I 2005 Online Predicted Human Interaction Database. Bioinformatics 21 2076-

2082.

Page 171: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

160

Brown K &Jurisica I 2007 Unequal evolutionary conservation of human protein interactions in

interologous networks. Genome biology 8 R95.

Browne GJ, Fardilha M, Oxenham SK, Wu W, Helps NR, da cruz e Silva, Odete A.B., Cohen PTW

& da Cruz e Silva EF 2007 SARP, a new alternatively spliced protein phosphatase 1 and DNA

interacting protein. - Biochemical Journal 402 187-196.

Brush MH, Weiser DC & Shenolikar S 2003 Growth Arrest and DNA Damage-Inducible Protein

GADD34 Targets Protein Phosphatase 1α to the Endoplasmic Reticulum and Promotes

Dephosphorylation of the α Subunit of Eukaryotic Translation Initiation Factor 2. Molecular and cellular

biology 23 1292-1303.

Burckstummer T, Bennett KL, Preradovic A, Schutze G, Hantschel O, Superti-Furga G & Bauch A 2006 An efficient tandem affinity purification procedure for interaction proteomics in mammalian cells. 3

1013-1019.

Bushnik T, Cook JL, Yuzpe AA, Tough S & Collins J 2012 Estimating the prevalence of infertility in

Canada. Human Reproduction 27 738-746.

Byers S, Graham R, Dai HN & Hoxter B 1991 Development of Sertoli cell junctional specializations

and the distribution of the tight-junction-associated protein ZO-1 in the mouse testis. American Journal

of Anatomy 191 35-47.

Byrne K, Leahy T, McCulloch R, Colgrave ML & Holland MK 2012 Comprehensive mapping of the

bull sperm surface proteome. Proteomics 12 3559-3579.

Cagney G, Park S, Chung C, Tong B, O'Dushlaine C, Shields DC & Emili A 2005 Human Tissue

Profiling with Multidimensional Protein Identification Technology. Journal of Proteome Research 4

1757-1767.

Ceulemans H &Bollen M 2004 Functional Diversity of Protein Phosphatase-1, a Cellular Economizer

and Reset Button. Physiological reviews 84 1-39.

Ceulemans H &Bollen M 2006 A Tighter RVxF Motif Makes a Finer Sift. Chemistry & biology 13 6-8.

Chakrabarti R, Cheng L, Puri P, Soler D & Vijayaraghavan S 2007a Protein phosphatase

PP1[gamma]2 in sperm morphogenesis and epididymal initiation of sperm motility. Asian Journal of

Andrology 9 445-452.

Chakrabarti R, Kline D, Lu J, Orth J, Pilder S & Vijayaraghavan S 2007b Analysis of Ppp1cc-Null

Mice Suggests a Role for PP1gamma2 in Sperm Morphogenesis. Biology of reproduction 76 992-1001.

Chelius D &Bondarenko PV 2002 Quantitative Profiling of Proteins in Complex Mixtures Using

Liquid Chromatography and Mass Spectrometry. Journal of Proteome Research 1 317-323.

Chemes H, E., Fawcett D, W. & Dym M 1978 Unusual features of the nuclear envelope in human

spermatogenic cells. 192 493-512.

Chen C, Jin J, James DA, Adams-Cioaba MA, Park JG, Guo Y, Tenaglia E, Xu C, Gish G, Min J &

Pawson T 2009 Mouse Piwi interactome identifies binding mechanism of Tdrkh Tudor domain to

arginine methylated Miwi. Proceedings of the National Academy of Sciences 106 20336-20341.

Chen GI &Gingras A 2007 Affinity-purification mass spectrometry (AP-MS) of serine/threonine

phosphatases. Methods 42 298-305.

Page 172: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

161

Cheng L, Pilder S, Nairn AC, Ramdas S & Vijayaraghavan S 2009 PP1Cγ2 and PPP1R11 Are Parts

of a Multimeric Complex in Developing Testicular Germ Cells in which their Steady State Levels Are

Reciprocally Related. PLoS ONE 4 e4861.

Choi J, Nannenga B, Demidov ON, Bulavin DV, Cooney A, Brayton C, Zhang Y, Mbawuike IN,

Bradley A, Appella E & Donehower LA 2002 Mice Deficient for the Wild-Type p53-Induced

Phosphatase Gene (Wip1) Exhibit Defects in Reproductive Organs, Immune Function, and Cell Cycle

Control. Molecular and cellular biology 22 1094-1105.

Chung J, Navarro B, Krapivinsky G, Krapivinsky L & Clapham DE 2011 A novel gene required for

male fertility and functional CATSPER channel formation in spermatozoa. Nature Communications 2

153.

Cohen P 1999 The Croonian Lecture 1998. Identification of a protein kinase cascade of major

importance in insulin signal transduction. Philosophical transactions of the Royal Society of London.

Series B, Biological sciences 354 485-495.

Colinge J, Chiappe D, Lagache S, Moniatte M & Bougueleret L 2005 Differential Proteomics via

Probabilistic Peptide Identification Scores. Analytical Chemistry 77 596-606.

Com E, Evrard B, Roepstorff P, Aubry F & Pineau C 2003 New Insights into the Rat Spermatogonial

Proteome. Molecular & Cellular Proteomics 2 248-261.

Dantzer F, Mark M, Quenet D, Scherthan H, Huber A, Liebe B, Monaco L, Chicheportiche A,

Sassone-Corsi P, de Murcia G & Ménissier-de Murcia J 2006 Poly(ADP-ribose) polymerase-2

contributes to the fidelity of male meiosis I and spermiogenesis. Proceedings of the National Academy of

Sciences 103 14854-14859.

Davalieva K, Kiprijanovska S, Noveski P, Plaseski T, Kocevska B, Broussard C & Plaseska-

Karanfilska D 2012 Proteomic analysis of seminal plasma in men with different spermatogenic

impairment. Andrologia 44 256-264.

Davies MJ, Moore VM, Willson KJ, Van Essen P, Priest K, Scott H, Haan EA & Chan A 2012

Reproductive Technologies and the Risk of Birth Defects. N Engl J Med 366 1803-1813.

Davies T &Varmuza S 2003 Development to Blastocyst Is Impaired When Intracytoplasmic Sperm

Injection Is Performed with Abnormal Sperm from Infertile Mice Harboring a Mutation in the Protein

Phosphatase 1cγ Gene. Biology of reproduction 68 1470-1476.

de Mateo S, Castillo J, Estanyol JM, Ballescà JL & Oliva R 2011 Proteomic characterization of the

human sperm nucleus. Proteomics 11 2714-2726.

De Wever V, Lloyd DC, Nasa I, Nimick M, Trinkle-Mulcahy L, Gourlay R, Morrice N & Moorhead

GBG 2012 Isolation of Human Mitotic Protein Phosphatase Complexes: Identification of a Complex

between Protein Phosphatase 1 and the RNA Helicase Ddx21. PLoS ONE 7 e39510.

Delbes G, Yanagiya A, Sonenberg N & Robaire B 2011 PABP Interacting Protein 2A (PAIP2A)

Regulates Specific Key Proteins During Spermiogenesis in the Mouse. Biology of reproduction.

Dihazi H, Dihazi GH, Nolte J, Meyer S, Jahn O, Müller GA & Engel W 2009 Multipotent Adult

Germline Stem Cells and Embryonic Stem Cells: Comparative Proteomic Approach. Journal of

Proteome Research 8 5497-5510.

Page 173: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

162

Dix DJ, Allen JW, Collins BW, Mori C, Nakamura N, Poorman-Allen P, Goulding EH & Eddy EM 1996 Targeted gene disruption of Hsp70-2 results in failed meiosis, germ cell apoptosis, and male

infertility. Proceedings of the National Academy of Sciences 93 3264-3268.

Egloff MP, Johnson DF, Moorhead G, Cohen PT, Cohen P & Barford D 1997 Structural basis for the

recognition of regulatory subunits by the catalytic subunit of protein phosphatase 1.The EMBO Journal

16 1876-1887.

Egloff M, Cohen PTW, Reinemer P & Barford D 1995 Crystal Structure of the Catalytic Subunit of

Human Protein Phosphatase 1 and its Complex with Tungstate. Journal of Molecular Biology 254 942-

959.

Fardilha M, Esteves SLC, Korrodi-Gregório L, Pelech S, da Cruz e Silva,Odete A.B. & da Cruz e

Silva E 2011a Protein phosphatase 1 complexes modulate sperm motility and present novel targets for

male infertility. Molecular human reproduction 17 466-477.

Fardilha M, Esteves SLC, Korrodi-Gregório L, Vintém AP, Domingues SC, Rebelo S, Morrice N,

Cohen PTW, Silva OA & da Cruz e Silva EF 2011b Identification of the human testis protein

phosphatase 1 interactome. Biochemical pharmacology 82 1403-1415.

Fawcett, DW 1975 The mammalian spermatozoa. Developmental Biology 44 394-436.

Ficarro S, Chertihin O, Westbrook VA, White F, Jayes F, Kalab P, Marto JA, Shabanowitz J, Herr

JC, Hunt DF & Visconti PE 2003 Phosphoproteome Analysis of Capacitated Human Sperm. Journal of

Biological Chemistry 278 11579-11589.

Forgione N, Vogl AW & Varmuza S 2010 Loss of protein phosphatase 1c{gamma} (PPP1CC) leads to

impaired spermatogenesis associated with defects in chromatin condensation and acrosome development:

an ultrastructural analysis. Reproduction 139 1021-1029.

Fukuda T, Pratto F, Schimenti JC, Turner JMA, Camerini-Otero R & Höög C 2012

Phosphorylation of Chromosome Core Components May Serve as Axis Marks for the Status of

Chromosomal Events during Mammalian Meiosis. PLoS Genet 8 e1002485.

Ge R, Chen G & Hardy MP 2009 The Role of the Leydig Cell in Spermatogenic Function. In

Molecular Mechanisms in Spermatogenesis, pp 255. Cheng CY (Ed), New York: Springer.

Goldberg J, Huang H, Kwon Y, Greengard P, Nairn AC & Kuriyan J 1995 Three-dimensional

structure of the catalytic subunit of protein serine/threonine phosphatase-1. Nature 376 745-753.

Govin J, Caron C, Escoffier E, Ferro M, Kuhn L, Rousseaux S, Eddy EM, Garin J & Khochbin S 2006 Post-meiotic Shifts in HSPA2/HSP70.2 Chaperone Activity during Mouse Spermatogenesis.

Journal of Biological Chemistry 281 37888-37892.

Govin J, Gaucher J, Ferro M, Debernardi A, Garin J, Khochbin S & Rousseaux S 2012 Proteomic

strategy for the identification of critical actors in reorganization of the post-meiotic male genome.

Molecular human reproduction 18 1-13.

Grallert A, Chan KY, Alonso- Nuñez ML, Madrid M, Biswas A, Alvarez- Tabarés, Connolly Y,

Tanaka K, Robertson A, Ortiz JM, Smith DL & Hagan IM 2013 Removal of Centrosomal PP1 by

NIMA Kinase Unlocks the MPF Feedback Loop to Promote Mitotic Commitment in S. pombe. Current

Biology 23 213-222.Griswold MD &McLean D 2006 The Sertoli Cell. In The Physiology of

Reproduction, edn 3, pp 949-975. Eds E Knobil and JD Neill. St Louis: Academic Press.

Page 174: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

163

Gu, Bin AND Zhang, Jiarong AND Wu, Ying AND Zhang, Xinzong AND Tan, Zhou AND Lin,

Yuanji AND Huang, Xiao AND Chen, Liangbiao AND Yao, Kangshou AND Zhang,Ming 2011

Proteomic Analyses Reveal Common Promiscuous Patterns of Cell Surface Proteins on Human

Embryonic Stem Cells and Sperms. PLoS ONE 6 e19386.

Guan K, Nayernia K, Maier LS, Wagner S, Dressel R, Lee JH, Nolte J, Wolf F, Li M, Engel W &

Hasenfuss G 2006 Pluripotency of spermatogonial stem cells from adult mouse testis. Nature 440 1199-

1203.

Guillaume E, Dupaix A, Moertz E, Courtens J, Jégou, B & Pineau C 2000 Proteome analysis of

spermatogonia: identification of a first set of 53 proteins. Proteome 1 1-20.

Guo X, Shen J, Xia Z, Zhang R, Zhang P, Zhao C, Xing J, Chen L, Chen W, Lin M, Huo R, Su B,

Zhou Z & Sha J 2010 Proteomic Analysis of Proteins Involved in Spermiogenesis in Mouse. Journal of

Proteome Research 9 1246-1256.

Guo X, Zhang P, Qi Y, Chen W, Chen X, Zhou Z & Sha J 2011 Proteomic analysis of male 4C germ

cell proteins involved in mouse meiosis. Proteomics 11 298-308.

Guyonnet B, Zabet-Moghaddam M, SanFrancisco S & Cornwall GA 2012 Isolation and proteomic

characterization of the mouse sperm acrosomal matrix. Molecular & Cellular Proteomics 11 758-774.

Hardouin N &Nagy A 2000 Gene-trap-based target site for Cre-mediated transgenic insertion. Genesis

26 245-252.

Held T, Barakat AZ, Mohamed BA, Paprotta I, Meinhardt A, Engel W & Adham IM 2011 Heat-

shock protein HSPA4 is required for progression of spermatogenesis. Reproduction 142 133-144.

Henderson H. 2006 Identifying the targets of Protein Phosphatase PP1cγ2 in the Mouse Testis. Thesis

(M.Sc.), University of Toronto.

Henderson H, MacLeod G, Hrabchak C & Varmuza S 2011 New candidate targets of protein

phosphatase-1c-gamma-2 in mouse testis revealed by a differential phosphoproteome analysis.

International Journal of Andrology 34 339-351.

Hendrickx A, Beullens M, Ceulemans H, Den Abt T, Van Eynde A, Nicolaescu E, Lesage B &

Bollen M 2009 Docking Motif-Guided Mapping of the Interactome of Protein Phosphatase-1. Chemistry

& biology 16 365-371.

Heroes E, Lesage B, Görnemann J, Beullens M, Van Meervelt L & Bollen M 2012 The PP1 binding

code: a molecular-lego strategy that governs specificity. FEBS Journal 280 584-595.

Hess RA &de Franca LR 2009 Spermatogenesis and Cycle of the Seminiferous Epithelium. In

Molecular Mechanisms in Spermatogenesis, pp 1. Ed. CY Cheng, New York: Springer.

Hornbeck PV, Kornhauser JM, Tkachev S, Zhang B, Skrzypek E, Murray B, Latham V & Sullivan

M 2012 PhosphoSitePlus: a comprehensive resource for investigating the structure and function of

experimentally determined post-translational modifications in man and mouse. Nucleic acids research 40

D261-D270.

Hrabchak C &Varmuza S 2004 Identification of the spermatogenic zip protein Spz1 as a putative

protein phosphatase-1 (PP1) regulatory protein that specifically binds the PP1cgamma2 splice variant in

the mouse testis. Journal of Biological Chemistry 279 37079-37086.

Page 175: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

164

Hrabchak C, Henderson H & Varmuza S 2007 A Testis Specific Isoform of Endophilin B1,

Endophilin B1t, Interacts Specifically with Protein Phosphatase-1cgamma2 in Mouse Testis and Is

Abnormally Expressed in PP1cgamma Null Mice. Biochemistry 46 4635-4644.

Hsu S, Shyu H, Hsieh-Li H & Li H 2001 Spz1, a novel bHLH-Zip protein, is specifically expressed in

testis. Mechanisms of development 100 177-187.

Hu Z, Xia Y, Guo X, Dai J, Li H, Hu H, Jiang Y, Lu F, Wu Y, Yang X, Li H, Yao B, Lu C, Xiong C,

Li Z, Gui Y, Liu J, Zhou Z, Shen H, Wang X & Sha J 2012 A genome-wide association study in

Chinese men identifies three risk loci for non-obstructive azoospermia. Nature genetics 44 183-186.

Huang DW, Sherman BT & Lempicki RA 2008 Systematic and integrative analysis of large gene lists

using DAVID bioinformatics resources. 4 44-57.

Huang DW, Sherman BT & Lempicki RA 2009 Bioinformatics enrichment tools: paths toward the

comprehensive functional analysis of large gene lists. Nucleic acids research 37 1-13.

Huang H, Lee T, Tzeng S & Horng J 2005 KinasePhos: a web tool for identifying protein kinase-

specific phosphorylation sites. Nucleic acids research 33 W226-W229.

Huang X, Guo X, Shen J, Wang Y, Chen L, Xie J, Wang N, Wang F, Zhao C, Huo R, Lin M, Wang

X, Zhou Z & Sha J 2008 Construction of a Proteome Profile and Functional Analysis of the Proteins

Involved in the Initiation of Mouse Spermatogenesis. Journal of Proteome Research 7 3435-3446.

Huang Z, Khatra B, Bollen M, Carr DW & Vijayaraghavan S 2002 Sperm PP1γ2 Is Regulated by a

Homologue of the Yeast Protein Phosphatase Binding Protein sds221. Biology of reproduction 67 1936-

1942.

Huang Z, Myers K, Khatra B & Vijayaraghavan S 2004 Protein 14-3-3ζ Binds to Protein Phosphatase

PP1γ2 in Bovine Epididymal Spermatozoa. Biology of reproduction 71 177-184.

Huang Z, Somanath PR, Chakrabarti R, Eddy EM & Vijayaraghavan S 2005 Changes in

Intracellular Distribution and Activity of Protein Phosphatase PP1γ2 and Its Regulating Proteins in

Spermatozoa Lacking AKAP4. Biology of reproduction 72 384-392.

Huang Z &Vijayaraghavan S 2004 Increased Phosphorylation of a Distinct Subcellular Pool of Protein

Phosphatase, PP1γ2, During Epididymal Sperm Maturation. Biology of reproduction 70 439-447.

Hubbard MJ &Cohen P 1993 On target with a new mechanism for the regulation of protein

phosphorylation. Trends in biochemical sciences 18 172-177.

Huckins C 1971 The spermatogonial stem cell population in adult rats. I. Their morphology,

proliferation and maturation. The Anatomical Record 169 533-557.

Hurley TD, Yang J, Zhang L, Goodwin KD, Zou Q, Cortese M, Dunker AK & DePaoli-Roach AA 2007 Structural Basis for Regulation of Protein Phosphatase 1 by Inhibitor-2. Journal of Biological

Chemistry 282 28874-28883.

Huttlin EL, Jedrychowski MP, Elias JE, Goswami T, Rad R, Beausoleil SA, Villén J, Haas W, Sowa

ME & Gygi SP 2010 A Tissue-Specific Atlas of Mouse Protein Phosphorylation and Expression. Cell

143 1174-1189.

Ingebritsen TS &Cohen P 1983 The Protein Phosphatases Involved in Cellular Regulation. European

Journal of Biochemistry 132 255-261.

Page 176: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

165

Jain N, Thatte J, Braciale T, Ley K, O'Connell M & Lee JK 2003 Local-pooled-error test for

identifying differentially expressed genes with a small number of replicated microarrays. Bioinformatics

19 1945-1951.

Jaleel M, McBride A, Lizcano JM, Deak M, Toth R, Morrice NA & Alessi DR 2005 Identification of

the sucrose non-fermenting related kinase SNRK, as a novel LKB1 substrate. FEBS letters 579 1417-

1423.

Jones M, Ng B, Bhide S, Chin E, Rhodenizer D, He P, Losfeld M, He M, Raymond K, Berry G,

Freeze H & Hegde M 2012 DDOST Mutations Identified by Whole-Exome Sequencing Are Implicated

in Congenital Disorders of Glycosylation. The American Journal of Human Genetics 90 363-368.

Jurisicova A, Lopes S, Meriano J, Oppedisano L, Casper RF & Varmuza S 1999 DNA damage in

round spermatids of mice with a targeted disruption of the Pp1c{gamma} gene and in testicular biopsies

of patients with non-obstructive azoospermia. Molecular human reproduction 5 323-330.

Kagedan D, Lecker I, Batruch I, Smith C, Kaploun I, Lo K, Grober E, Diamandis E & Jarvi K 2012 Characterization of the seminal plasma proteome in men with prostatitis by mass spectrometry.

Clinical Proteomics 9 2.

Kaji H, Shikanai T, Sasaki-Sawa A, Wen H, Fujita M, Suzuki Y, Sugahara D, Sawaki H, Yamauchi

Y, Shinkawa T, Taoka M, Takahashi N, Isobe T & Narimatsu H Large-scale Identification of N-

Glycosylated Proteins of Mouse Tissues and Construction of a Glycoprotein Database, GlycoProtDB.

Journal of Proteome Research 11 4553-4566.

Kawa S, Ito C, Toyama Y, Maekawa M, Tezuka T, Nakamura T, Nakazawa T, Yokoyama K,

Yoshida N, Toshimori K & Yamamoto T 2006 Azoospermia in mice with targeted disruption of the

Brek/Lmtk2 (brain-enriched kinase/lemur tyrosine kinase 2) gene. Proceedings of the National Academy

of Sciences 103 19344-19349.

Keller A, Nesvizhskii AI, Kolker E & Aebersold R 2002 Empirical Statistical Model To Estimate the

Accuracy of Peptide Identifications Made by MS/MS and Database Search. Analytical Chemistry 74

5383-5392.

Kerk D, Templeton G & Moorhead GBG February 2008 Evolutionary Radiation Pattern of Novel

Protein Phosphatases Revealed by Analysis of Protein Data from the Completely Sequenced Genomes of

Humans, Green Algae, and Higher Plants. Plant physiology 146 351-367.

Kierszenbaum AL 2002 Intramanchette transport (IMT): Managing the making of the spermatid head,

centrosome, and tail. Molecular reproduction and development 63 1-4.

Kopera IA, Bilinska B, Cheng CY & Mruk DD 2010 Sertoli–germ cell junctions in the testis: a review

of recent data. Philosophical Transactions of the Royal Society B: Biological Sciences 365 1593-1605.

Kriegel TM, Heidenreich F, Kettner K, Pursche T, Hoflack B, Grunewald S, Poenicke K, Glander

H & Paasch U 2009 Identification of diabetes- and obesity-associated proteomic changes in human

spermatozoa by difference gel electrophoresis. Reproductive BioMedicine Online 19 660-670.

Kueng P, Nikolova Z, Djonov V, Hemphill A, Rohrbach V, Boehlen D, Zuercher G, Andres A &

Ziemiecki A 1997 A Novel Family of Serine/Threonine Kinases Participating in Spermiogenesis. The

Journal of cell biology 139 1851-1859.

Page 177: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

166

Kuzmin A, Jarvi K, Lo K, Spencer L, Chow GYC, Macleod G, Wang Q & Varmuza S 2009

Identification of Potentially Damaging Amino Acid Substitutions Leading to Human Male Infertility.

Biology of reproduction 81 319-326.

Larsen MR, Thingholm TE, Jensen ON, Roepstorff P & Jørgensen TJD 2005 Highly Selective

Enrichment of Phosphorylated Peptides from Peptide Mixtures Using Titanium Dioxide Microcolumns.

Molecular & Cellular Proteomics 4 873-886.

Leblond CP &Clermont Y 1952 Spermiogenesis of rat, mouse, hamster and guinea pig as revealed by

the “periodic acid-fuchsin sulfurous acid” technique. American Journal of Anatomy 90 167-215.

Lefièvre L, Chen Y, Conner SJ, Scott JL, Publicover SJ, Ford WCL & Barratt CLR 2007 Human

spermatozoa contain multiple targets for protein S-nitrosylation: An alternative mechanism of the

modulation of sperm function by nitric oxide? Proteomics 7 3066-3084.

Lewis JD, Abbott DW & Ausió J 2003 A haploid affair: core histone transitions during

spermatogenesis. Biochemistry and Cell Biology 81 131-140.

Lexicon Genetics 2005 NIH initiative supporting placement of Lexicon Genetics, Inc. mice into public

repositories. MGI: J:103485.

Li J, Guo W, Li F, He J, Yu Q, Wu X, Li J & Mao X 2012 HnRNPL as a key factor in

spermatogenesis: Lesson from functional proteomic studies of azoospermia patients with sertoli cell only

syndrome. Journal of Proteomics 75 2879-2891.

Li X, Ionescu AV, Lynn BD, Kamasawa N, Morita M, Davidson KG, Yasumura T, Rash JE, Nagy JI

2004 Connexin47, connexin29 and connexin32 co-expression in oligodendrocytes and Cx47 association

with zonula occludens-1 (ZO-1) in mouse brain. Neuroscience 126 611-630.

Li Y, Sosnik J, Brassard L, Reese M, Spiridonov NA, Bates TC, Johnson GR, Anguita J, Visconti

PE & Salicioni AM 2011a Expression and localization of five members of the testis-specific serine

kinase (Tssk) family in mouse and human sperm and testis. Molecular human reproduction 17 42-56.

Li Y, Franklin S, Zhang MJ & Vondriska TM 2011b Highly efficient purification of protein

complexes from mammalian cells using a novel streptavidin-binding peptide and hexahistidine tandem

tag system: Application to Bruton's tyrosine kinase. Protein Science 20 140-149.

Liu H, Sadygov RG & Yates JR 2004 A Model for Random Sampling and Estimation of Relative

Protein Abundance in Shotgun Proteomics. Analytical Chemistry 76 4193-4201.

Liu J &Brautigan DL 2000 Glycogen Synthase Association with the Striated Muscle Glycogen-

targeting Subunit of Protein Phosphatase-1: Synthase activation involves scaffolding regulated by β-

adrenergic signalling. Journal of Biological Chemistry 275 26074-26081.

Liu J, Schiltz JF, Ashar HR & Chada KK 2003 Hmga1 is required for normal sperm development.

Molecular reproduction and development 66 81-89.

Long JC, Caceres JF 2009 The SR protein family of splicing factors: master regulators of gene

expression. Biochemical Journal 417 15-27.

Luk JM, Mok BW, Shum CK, Yeung WS, Tam PC, Tse JY, Chow JF, Woo J, Kam K & Lee KF 2003 Identification of novel genes expressed during spermatogenesis in stage-synchronized rat testes by

differential display. Biochemical and biophysical research communications 307 782-790.

Page 178: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

167

Lundby A, Lage K, Weinert B, Bekker-Jensen D, Secher A, Skovgaard T, Kelstrup C, Dmytriyev

A, Choudhary C, Lundby C & Olsen J 2012a Proteomic Analysis of Lysine Acetylation Sites in Rat

Tissues Reveals Organ Specificity and Subcellular Patterns. Cell Reports 2 419-431.

Lundby A, Secher A, Lage K, Nordsborg NB, Dmytriyev A, Lundby C & Olsen JV 2012b

Quantitative maps of protein phosphorylation sites across 14 different rat organs and tissues. Nature

Communications 3 876.

Lynes EM &Simmen T 2011 Urban planning of the endoplasmic reticulum (ER): How diverse

mechanisms segregate the many functions of the ER. Biochimica et Biophysica Acta (BBA) - Molecular

Cell Research 1813 1893-1905.

MacLeod G &Varmuza S 2012 Tandem Affinity Purification in Transgenic Mouse Embryonic Stem

Cells Identifies DDOST as a Novel PPP1CC2 Interacting Protein. Biochemistry 51 9678-9688.

Mann M, Latham K, E. & Varmuza S 1995 Identification of Genes Showing Altered Expression in

Preimplantation and Early Postimplantation Parthenogenetic Embryos. Developmental Genetics 17 223-

232.

Martínez-Heredia J, de Mateo S, Vidal-Taboada JM, Ballescà JL & Oliva R 2008 Identification of

proteomic differences in asthenozoospermic sperm samples. Human Reproduction 23 783-791.

Matzuk MM &Lamb DJ 2008 The biology of infertility: research advances and clinical challenges.

Nature medicine 14 1197-1213.

McAvoy T, Allen PB, Obaishi H, Nakanishi H, Takai Y, Greengard P, Nairn AC & Hemmings HC 1999 Regulation of Neurabin I Interaction with Protein Phosphatase 1 by Phosphorylation. Biochemistry

38 12943-12949.

Meikar O, Da Ros M, Korhonen H & Kotaja N 2011 Chromatoid body and small RNAs in male germ

cells. Reproduction 142 195-209.

Meiselbach H, Sticht H & Enz R 2006 Structural Analysis of the Protein Phosphatase 1 Docking Motif:

Molecular Description of Binding Specificities Identifies Interacting Proteins. Chemistry & biology 13

49-59.

Milardi D, Grande G, Vincenzoni F, Messana I, Pontecorvi A, De Marinis L, Castagnola M &

Marana R 2012 Proteomic approach in the identification of fertility pattern in seminal plasma of fertile

men. Fertility and sterility 97 67-73.e1.

Moorhead GBG, Trinkle-Mulcahy L & Ulke-Lemee A 2007 Emerging roles of nuclear protein

phosphatases. Nature reviews. Molecular cell biology 8 234-244.

Moorhead GBG, De wever V, Templeton G & Kerk D 2009 Evolution of protein phosphatases in

plants and animals. Biochemical Journal 417 401-409.

Morimoto H, Okamura H & Haneji T 2002 Interaction of Protein Phosphatase 1 Delta with Nucleolin

in Human Osteoblastic Cells. Journal of Histochemistry & Cytochemistry 50 1187-1193.

Nagai M, Moriyama T, Mehmood R, Tokuhiro K, Ikawa M, Okabe M, Tanaka H & Yoneda Y 2011

Mice lacking Ran binding protein 1 are viable and show male infertility. FEBS letters 585 791-796.7.

Page 179: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

168

Nahhas F, Dryden SC, Abrams J, Tainsky MA 2007 Mutations in SIRT2 deacetylase which regulate

enzymatic activity but not its interaction with DHAC6 and tubulin. Molecular and Cellular Biochemistry

303 221-230.

Nesvizhskii AI, Keller A, Kolker E & Aebersold R 2003 A Statistical Model for Identifying Proteins by

Tandem Mass Spectrometry. Analytical Chemistry 75 4646-4658.

Nieschlag E, Behre HM &Nieschlag S (Eds) 2010 Andrology: Male Reproductive Health and

Dysfunction, edn 3, New York: Springer.

Nixon B, Bielanowicz A, Mclaughlin EA, Tanphaichitr N, Ensslin MA & Aitken RJ 2009

Composition and significance of detergent resistant membranes in mouse spermatozoa. Journal of

cellular physiology 218 122-134.

Nixon B, Mitchell LA, Anderson AL, Mclaughlin EA, O'bryan MK & Aitken RJ 2011a Proteomic

and functional analysis of human sperm detergent resistant membranes. Journal of cellular physiology

226 2651-2665.

Nord AS, Chang PJ, Conklin BR, Cox AV, Harper CA, Hicks GG, Huang CC, Johns SJ, Kawamoto

M, Liu S, Meng EC, Morris JH, Rossant J, Ruiz P, Skarnes WC, Soriano P, Stanford WL, Stryke

D, von Melchner H, Wurst W, Yamamura K, Young SG, Babbitt PC & Ferrin TE 2006 The

International Gene Trap Consortium Website: a portal to all publicly available gene trap cell lines in

mouse. Nucleic acids research 34 D642-D648.

Oakberg EF 1971 Spermatogonial stem-cell renewal in the mouse. The Anatomical Record 169 515-

531.

Oakberg EF 1956 A description of spermiogenesis in the mouse and its use in analysis of the cycle of

the seminiferous epithelium and germ cell renewal. American Journal of Anatomy 99 391-413.

Obenauer JC, Cantley LC & Yaffe MB 2003 Scansite 2.0: proteome-wide prediction of cell signaling

interactions using short sequence motifs. Nucleic acids research 31 3635-3641.

Okamura D, Maeda I, Taniguchi H, Tokitake Y, Ikeda M, Ozato K, Mise N, Abe K, Noce T, Izpisua

Belmonte JC & Matsui Y 2012 Cell cycle gene-specific control of transcription has a critical role in

proliferation of primordial germ cells. Genes & development 26 2477-2482.

Okano K, Heng H, Trevisanato S, Tyers M & Varmuza S 1997 - Genomic Organization and

Functional Analysis of the Murine Protein Phosphatase 1c γ (Ppp1cc) Gene. 45 215.

Olsen JV, Blagoev B, Gnad F, Macek B, Kumar C, Mortensen P, Mann M 2006 Global, in vivo, and

site-specific phosphorylation dynamics in signalling networks. Cell 127 635-648.

Olsen JV, Vermeulen M, Santamaria A, Kumar C, Miller ML, Jensen LJ, Gnad F, Cox J, Jensen

TS, Nigg EA, Brunak S & Mann M 2010 Quantitative Phosphoproteomics Reveals Widespread Full

Phosphorylation Site Occupancy During Mitosis. Science Signaling 3 ra3.

Oppedisano-Wells L &Varmuza S 2003 Protein phosphatase 1cgamma is required in germ cells in

murine testis. Molecular reproduction and development 65 157-166.

Parte PP, Rao P, Redij S, Lobo V, D'Souza SJ, Gajbhiye R & Kulkarni V 2012 Sperm

phosphoproteome profiling by ultra performance liquid chromatography followed by data independent

analysis (LC–MSE) reveals altered proteomic signatures in asthenozoospermia. Journal of Proteomics 75

5861-5871.

Page 180: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

169

Philipps DL, Wigglesworth K, Hartford SA, Sun F, Pattabiraman S, Schimenti K, Handel M,

Eppig JJ & Schimenti JC 2008 The dual bromodomain and WD repeat-containing mouse protein

BRWD1 is required for normal spermiogenesis and the oocyte–embryo transition. Developmental

biology 317 72-82.

Phillips BT, Gassei K & Orwig KE 2010 Spermatogonial stem cell regulation and spermatogenesis.

Philosophical Transactions of the Royal Society B: Biological Sciences 365 1663-1678.

Pilch B &Mann M 2006 Large-scale and high-confidence proteomic analysis of human seminal plasma.

Genome biology 7 R40.

Pixton KL, Deeks ED, Flesch FM, Moseley FLC, Björndahl L, Ashton PR, Barratt CLR & Brewis

IA 2004 Sperm proteome mapping of a patient who experienced failed fertilization at IVF reveals altered

expression of at least 20 proteins compared with fertile donors: Case report. Human Reproduction 19

1438-1447.

Platt MD, Salicioni AM, Hunt DF & Visconti PE 2009 Use of Differential Isotopic Labeling and Mass

Spectrometry To Analyze Capacitation-Associated Changes in the Phosphorylation Status of Mouse

Sperm Proteins. Journal of Proteome Research 8 1431-1440.

Puri P, Acker-Palmer A, Stahler R, Chen Y, Kline D & Vijayaraghavan S 2011 Identification of testis

14-3-3 binding proteins by tandem affinity purification. Spermatogenesis 1 354-365.

Puri P, Myers K, Kline D & Vijayaraghavan S 2008 Proteomic Analysis of Bovine Sperm YWHA

Binding Partners Identify Proteins Involved in Signaling and Metabolism. Biology of reproduction 79

1183-1191.

Ragusa MJ, Dancheck B, Critton DA, Nairn AC, Page R & Peti W 2010 Spinophilin directs protein

phosphatase 1 specificity by blocking substrate binding sites. Nature structural & molecular biology 17

459-464.

Reinton N, Collas P, Haugen TB, Skålhegg BS, Hansson V, Jahnsen T & Taskén K 2000

Localization of a Novel Human A-Kinase-Anchoring Protein, hAKAP220, during Spermatogenesis.

Developmental biology 223 194-204.

Rigaut G, Shevchenko A, Rutz B, Wilm M, Mann M & Seraphin B 1999 A generic protein

purification method for protein complex characterization and proteome exploration. Nature

Biotechnology 17 1030-1032.

Roboti P &High S 2012 The oligosaccharyltransferase subunits OST48, DAD1 and KCP2 function as

ubiquitous and selective modulators of mammalian N-glycosylation. Journal of cell science 125 3474-

3484.

Ruan Y, Cheng M, Ou Y, Oko R & van der Hoorn FA 2011 Ornithine Decarboxylase Antizyme Oaz3

Modulates Protein Phosphatase Activity. Journal of Biological Chemistry 286 29417-29427.

Russell LD, Ettlin RA, Sinha Hikim AP & Clegg EG 1990 Histological and Histopathological

Evaluation of the Testis. Clearwater, Fl: Cache River Press.

Schilling B, Rardin MJ, MacLean BX, Zawadzka AM, Frewen BE, Cusack MP, Sorensen DJ,

Bereman MS, Jing E, Wu CC, Verdin E, Kahn CR, MacCoss MJ & Gibson BW 2012 Platform-

independent and Label-free Quantitation of Proteomic Data Using MS1 Extracted Ion Chromatograms in

Page 181: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

170

Skyline: APPLICATION TO PROTEIN ACETYLATION AND PHOSPHORYLATION. Molecular &

Cellular Proteomics 11 202-214.

Seo, Seongjin AND Zhang, Qihong AND Bugge, Kevin AND Breslow, David K. AND Searby,

Charles C. AND Nachury, Maxence V. AND Sheffield,Val C. 2011 A Novel Protein LZTFL1

Regulates Ciliary Trafficking of the BBSome and Smoothened. PLoS Genet 7 e1002358.

Shang P, Hoogerbrugge J, Baarends WM & Grootegoed JA 2013 Evolution of testis-specific kinases

TSSK1B and TSSK2 in primates. Andrology 1 160-168.

Shang P, Baarends WM, Hoogerbrugge J, Ooms MP, van Cappellen WA, de Jong AAW, Dohle GR,

van Eenennaam H, Gossen JA & Grootegoed JA 2010 Functional transformation of the chromatoid

body in mouse spermatids requires testis-specific serine/threonine kinases. Journal of cell science 123

331-339.

Shi Y &Manley JL 2007 A Complex Signaling Pathway Regulates SRp38 Phosphorylation and Pre-

mRNA Splicing in Response to Heat Shock. Molecular cell 28 79-90.

Shima H, Haneji T, Hatano Y, Kasugai I, Sugimura T & Nagao M 1993 Protein Phosphatase 1γ2 Is

Associated with Nuclei of Meiotic Cells in Rat Testis. Biochemical and biophysical research

communications 194 930-937.

Singla V, Hunkapiller J, Santos N, Seol AD, Norman AR, Wakenight P, Skarnes WC & Reiter JF 2010 Floxin, a resource for genetically engineering mouse ESCs. Nature Methods 7 50-52.

Sinha N, Pilder S & Vijayaraghavan S 2012 Significant Expression Levels of Transgenic PPP1CC2 in

Testis and Sperm Are Required to Overcome the Male Infertility Phenotype of Ppp1cc Null Mice. PLoS

ONE 7 e47623.

Siva AB, Kameshwari DB, Singh V, Pavani K, Sundaram CS, Rangaraj N, Deenadayal M &

Shivaji S 2010 Proteomics-based study on asthenozoospermia: differential expression of proteasome

alpha complex. Molecular human reproduction 16 452-462.

Smith GD, Wolf DP, Trautman KC, da Cruz e Silva,E F., Greengard P & Vijayaraghavan S 1996

Primate sperm contain protein phosphatase 1, a biochemical mediator of motility. Biology of

reproduction 54 719-727.

Soler DC, Kadunganattil S, Ramdas S, Myers K, Roca J, Slaughter T, Pilder SH, Vijayaraghavan S

2009 Expression of Transgenic PPP1CC2 in the Testis of Ppp1cc-Null Mice Rescues Spermatid Viability

and Spermiation but does not Restore Normal Sperm Tail Ultrastructure, Sperm Motility or Fertility.

Biology of Reproduction 81 343-352.

Spiridonov NA, Wong L, Zerfas PM, Starost MF, Pack SD, Paweletz CP & Johnson GR 2005

Identification and Characterization of SSTK, a Serine/Threonine Protein Kinase Essential for Male

Fertility. Molecular and cellular biology 25 4250-4261.

Stoffel W, Holz B, Jenke B, Binczek E, Gunter RH, Kiss C, Karakesisoglou I, Thevis M, Weber A,

Arnhold S & Addicks K 2008 [Delta]6-Desaturase (FADS2) deficiency unveils the role of [omega]3-

and [omega]6-polyunsaturated fatty acids. The EMBO journal 27 2281-2292.

Sutherland HG, Newton K, Brownstein DG, Holmes MC, Kress C, Semple CA & Bickmore WA 2006 Disruption of Ledgf/Psip1 Results in Perinatal Mortality and Homeotic Skeletal Transformations.

Molecular and cellular biology 26 7201-7210.

Page 182: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

171

Takahashi Y, Coppola D, Matsushita N, Cualing HD, Sun M, Sato Y, Liang C, Jung JU, Cheng JQ,

Mul JJ, Pledger WJ & Wang H 2007 Bif-1 interacts with Beclin 1 through UVRAG and regulates

autophagy and tumorigenesis. Nature cell biology 9 1142-1151.

Takizawa N, Mizuno Y, Ito Y & Kikuchi K 1994 Tissue Distribution of Isoforms of Type-1 Protein

Phosphatase PP1 in Mouse Tissues and Its Diabetic Alterations. Journal of Biochemistry 116 411-415.

Tegelenbosch RA &de Rooij DG 1993 A quantitative study of spermatogonial multiplication and stem

cell renewal in the C3H/101 F1 hybrid mouse. 290 193-200.

Terrak M, Kerff F, Langsetmo K, Tao T & Dominguez R 2004 Structural basis of protein phosphatase

1 regulation. Nature 429 780-784.

Terry-Lorenzo R, Carmody LC, Voltz JW, Connor JH, Li S, Smith FD, Milgram SL, Colbran RJ

& Shenolikar S 2002 The Neuronal Actin-binding Proteins, Neurabin I and Neurabin II, Recruit Specific

Isoforms of Protein Phosphatase-1 Catalytic Subunits. Journal of Biological Chemistry 277 27716-

27724.

Thacker S, Yadav SP, Sharma RK, Kashou A, Willard B, Zhang D & Agarwal A 2011 Evaluation of

Sperm Proteins in Infertile Men: A Proteomic Approach. Fertility and sterility 95 2745-2748.

Thingholm TE, Jensen ON & Larsen MR 2009 Enrichment and Separation of Mono- and Multiply

Phosphorylated Peptides Using Sequential Elution from IMAC Prior to Mass Spectrometric Analysis. In

Phospho-Proteomics, Methods and Protocols, pp 67-78. Ed. M de Graauw, New York: Humana Press.

Thingholm TE, Jensen ON, Robinson PJ & Larsen MR April 2008 SIMAC (Sequential Elution from

IMAC), a Phosphoproteomics Strategy for the Rapid Separation of Monophosphorylated from Multiply

Phosphorylated Peptides. Molecular & Cellular Proteomics 7 661-671.

Tian Y, Kelly-Spratt KS, Kemp CJ & Zhang H 2010 Mapping Tissue-Specific Expression of

Extracellular Proteins Using Systematic Glycoproteomic Analysis of Different Mouse Tissues. Journal of

Proteome Research 9 5837-5847.

Tokuhiro K, Isotani A, Yokota S, Yano Y, Oshio S, Hirose M, Wada M, Fujita K, Ogawa Y, Okabe

M, Nishimune Y & Tanaka H 2009 OAZ-t/OAZ3 Is Essential for Rigid Connection of Sperm Tails to

Heads in Mouse. PLoS Genet 5 e1000712.

Tournaye H 2012 Male factor infertility and ART. Asian Journal of Andrology 14 103-108.

Trinkle-Mulcahy L, Andersen J, Lam YW, Moorhead G, Mann M & Lamond AI 2006 Repo-Man

recruits PP1γ to chromatin and is essential for cell viability. The Journal of cell biology 172 679-692.

Ulloa L, Dombrádi V, Díaz-Nido J, Szücs K, Gergely P, Friedrich P & Avila J 1993

Dephosphorylation of distinct sites on microtubule-associated protein MAP1B by protein phosphatases

1, 2A and 2B. FEBS letters 330 85-89.

Varmuza S, Jurisicova A, Okano K, Hudson J, Boekelheide K & Shipp EB 1999 Spermiogenesis Is

Impaired in Mice Bearing a Targeted Mutation in the Protein Phosphatase 1cγ Gene. Developmental

biology 205 98-110.

Varmuza S &Ling L 2003 Increased recombination frequency showing evidence of loss of interference

is associated with abnormal testicular histopathology. Molecular reproduction and development 64 499-

506.

Page 183: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

172

Vigodner M, Shrivastava V, Gutstein LE, Schneider J, Nieves E, Goldstein M, Feliciano M &

Callaway M 2012 Localization and identification of sumoylated proteins in human sperm: excessive

sumoylation is a marker of defective spermatozoa. Human Reproduction 28 210-223.

Vijayaraghavan S, Stephens DT, Trautman K, Smith GD, Khatra B, da Cruz e Silva,E F. &

Greengard P 1996 Sperm motility development in the epididymis is associated with decreased glycogen

synthase kinase-3 and protein phosphatase 1 activity. Biology of reproduction 54 709-718.

Wakayama T, Sai Y, Ito A, Kato Y, Kurobo M, Murakami Y, Nakashima E, Tsuji A, Kitamura Y &

Iseki S 2007 Heterophilic Binding of the Adhesion Molecules Poliovirus Receptor and Immunoglobulin

Superfamily 4A in the Interaction Between Mouse Spermatogenic and Sertoli Cells. Biology of

reproduction 76 1081-1090.

Wakula P, Beullens M, Ceulemans H, Stalmans W & Bollen M 2003 Degeneracy and Function of the

Ubiquitous RVXF Motif That Mediates Binding to Protein Phosphatase-1. Journal of Biological

Chemistry 278 18817-18823.

Walker A, Taylor J, Rowe D & Summers D 2008 A method for generating sticky-end PCR products

which facilitates unidirectional cloning and the one-step assembly of complex DNA constructs. Plasmid

59 155-162.

Wang J, Wang J, Zhang H, Shi H, Ma D, Zhao H, Lin B & Li R 2009 Proteomic analysis of seminal

plasma from asthenozoospermia patients reveals proteins that affect oxidative stress responses and semen

quality. Asian Journal of Andrology 11 484-491.

Wang R, Kaul A & Sperry AO 2010 TLRR (lrrc67) interacts with PP1 and is associated with a

cytoskeletal complex in the testis. Biology of the Cell 102 173-189.

Wang R &Sperry AO 2011 PP1 Forms an Active Complex with TLRR (lrrc67), a Putative PP1

Regulatory Subunit, during the Early Stages of Spermiogenesis in Mice. PLoS ONE 6 e21767.

Xu B, Hao Z, Jha KN, Zhang Z, Urekar C, Digilio L, Pulido S, Strauss III JF, Flickinger CJ &

Herr JC 2008 Targeted deletion of Tssk1 and 2 causes male infertility due to haploinsufficiency.

Developmental biology 319 211-222.

Xu W, Hu H, Wang Z, Chen X, Yang F, Zhu Z, Fang P, Dai J, Wang L, Shi H, Li Z & Qiao Z 2012

Proteomic characteristics of spermatozoa in normozoospermic patients with infertility. Journal of

Proteomics 75 5426-5436.

Yamagata T, Tsuru T, Momoi MY, Suwa K, Nozaki Y, Mukasa T, Ohashi H, Fukushima Y &

Momoi T 1997 Genome Organization of Human 48-kDa Oligosaccharyltransferase (DDOST). Genomics

45 535-540.

Yan W 2009 Male infertility caused by spermiogenic defects: Lessons from gene knockouts. Molecular

and cellular endocrinology 306 24-32.

Yang F, De La Fuente R, Leu NA, Baumann C, McLaughlin KJ & Wang PJ 2006 Mouse SYCP2 is

required for synaptonemal complex assembly and chromosomal synapsis during male meiosis. 173 497-

507.

Yang J, Medvedev S, Yu J, Tang LC, Agno JE, Matzuk MM, Schultz RM & Hecht NB 2005

Absence of the DNA-/RNA-binding protein MSY2 results in male and female infertility. Proceedings of

the National Academy of Sciences of the United States of America 102 5755-5760.

Page 184: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

173

Yeo G, Holste D, Kreiman G & Burge C 2004 Variation in alternative splicing across human tissues.

Genome biology 5 R74.

Zhang Z &Lutz B 2002 Cre recombinase mediated inversion using lox66 and lox71: method to

introduce conditional point mutations into the CREB binding protein. Nucleic acids research 30 e90.

Zhao C, Huo R, Wang F, Lin M, Zhou Z & Sha J 2007 Identification of several proteins involved in

regulation of sperm motility by proteomic analysis. Fertility and sterility 87 436-438.

Page 185: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

174

Appendix A.1 Mouse Mutations Causing Male Infertility

Author’s Note: This table contains data compiled from Matzuk & Lamb 2008, Kuzmin et al., 2009 in

addition to data that I personally compiled.

Gene Testis Cells Affected Sex Affected Testis/Sperm Phenotype Male Fertility Status

Ace Spermatozoa Male Impaired fertility Subfertile

Acr Spermatozoa Male Impaired fertility Fertile

Acvr2a Sertoli,

peritubular, Leydig

and/or interstitial

cells

Both Oligozoospermia Subfertile

Adad1 Spermatozoa Male Oligoteratozoospermia Infertile

Adam2 Spermatozoa Male Impaired fertility Infertile

Adam24 Spermatozoa Male Polyspermy Subfertile

Adam3 Spermatozoa Male Impaired fertility Infertile

Adamts2 Spermatogonia; Spermatids;

Spermatozoa

Male Oligozoospermia infertile

Adcy10 Spermatozoa Male Asthenozoospermia Infertile

Adcy3 Spermatids; Spermatozoa Male Impaired fertility;

Asthenozoospermia

Subfertile

Adora1 Spermatozoa Male Impaired Fertility Subfertile

Adra1b Sertoli,

peritubular, Leydig

and/or interstitial

cells;

Spermatocytes

Male hypospermatogenesis- Sertoli cell

only

Variable infertility

Aff4 Spermatids Male Azoospermia Infertile

Agfg1 Spermatocytes; Spermatids Male ÒAT/Globozoospermia Infertile

Agps Spermatocytes; Spermatids Male Azoospermia Infertile

Agtpbp1 Spermatozoa Male OAT Infertile

Ahr Spermatozoa Both Oligozoospermia Subfertile

Aire Both Gonadal Atrophy Variable infertility

Akap4 Spermatozoa Male Asthenozoospermia Infertile

Akt1 Spermatozoa Male Oligoasthenozoospermia Subfertile

Akt2 Spermatozoa Male Oligoasthenozoospremia Fertile

Amh Both Obstructive Azoospermia Infertile

Amhr2 Male Obstructive Azoospermia Secondary Infertility

Apaf1 Spermatogonia Male Azoospermia Variable lethality;

infertile

Page 186: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

175

Apob Spermatozoa Male Oligoasthenozoospermia Infertile

Aqp3 Spermatozoa Male Impaired Fertility Subfertile

Ar Sertoli,

peritubular, Leydig

and/or interstitial

cells

Male Morphological Infertile

Arl4 Spermatozoa Male Oligozoospermia Fertile

Arntl Leydig cells; Spermatozoa Both Oligozoospermia; Impaired

Fertility

Infertile

Aspm PGC Both Oligoasthenozoospermia Subfertile

Asz1 Spermatocytes Male Meiotic Arrest Infertile

Atf4 Male Obstructive Subfertile

Atm Spermatocytes Both Meiotic Arrest Infertile

Atn1 ? Male Oligozoospermia Progressive Infertility

Atp2b4 Spermatozoa Male Asthenozoospermia Infertile

Aurkb Spermatocytes Male Hypospermatogenesis Subfertile

Aurkc Spermatids, Spermatozoa Male Teratozoospermia Subfertile

B4galnt1 Sertoli,

peritubular, Leydig

and/or interstitial

cells; Spermatozoa

Male Azoospermia Infertile

Bat3 Spermatocytes Male Azoospermia Infertile

Bax Spermatogonia Both Azoospermia Infertile

Bbs1 Spermatids Male Asthenozoospermia Infertile

Bbs2 Spermatids Male Asthenozoospermia Infertile

Bbs3 Spermatids Male Asthenozoospermia Infertile

Bbs4 Spermatozoa Male Asthenozoospermia Infertile

Bcl2l1 Spermatocytes Both Sertoli-cell only Infertile

Bcl2l2 Sertoli,

peritubular, Leydig

and/or interstitial

cells; Spermatocytes;

Spermatids

Both Azoospermia Infertile

Bcl6 Spermatocytes Male Oligozoospermia Subfertile

Blimp1 PGC Both PGC depletion Infertile

Page 187: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

176

Bmp4 PGC Both PGC depletion Lethal

Bmp8a Spermatocytes Male Progressive germ cell depletion Progressive infertility

Bmp8b Spermatogonia Both Hypospermatogenesis

Variable infertility;

subfertile

Brca2 Spermatocytes Both Meiotic Arrest Infertile

Brdt Spermatids Male OAT Infertile

Brwd1 Spermatids; Spermatozoa Both OAT Infertile

Bsg Spermatocytes Both Meiotic Arrest Infertile

Btrc Spermatocytes Male Oligozoospermia Subfertile

Bub1 Spermatogonia Male Sertoli-cell only Infertile

Bub1b Spermatocytes Both Oligozoospermia, Impaired fertility Infertile

Cadm1 Spermatids; Spermatozoa Male OAT Infertile

Calr3 Spermatozoa Male Impaired Fertility Infertile

Camk4 Spermatids Male Oligoteratozoospermia Infertile

Capza3 Spermatids Male OAT Infertile

Catsper1 Spermatozoa Male Asthenozoospermia Infertile

Catsper2 Spermatozoa Male Asthenozoospermia Infertile

Catsper3 Spermatozoa Male Asthenozoospermia Infertile

Catsper4 Spermatozoa Male Asthenozoospermia Infertile

Ccna1 Spermatocytes Male Meiotic Arrest Infertile

Ccnb1ip1 Spermatocytes Both Meiotic Arrest Infertile

Ccnd2 Both Gonadal hypoplasia Fertile

Ccne2 Spermatocytes Male Oligozoospermia Subfertile

Cd59b Spermatozoa Male OAT Progressive infertility

Cdk16 Spermatids; Spermatozoa Male Asthenoteratozoospermia Infertile

Cdk2 Spermatocytes Both Meiotic Arrest Infertile

Cdk4 Sertoli,

peritubular, Leydig

and/or interstitial

cells; Spermatocytes

Both Age dependent decline in

spermatogenesis

Infertile

Cdkn1b Both Testicular hyperplasia Fertile

Cdkn1c Both Gonadal hypoplasia Infertile

Cdkn2c Sertoli,

peritubular, Leydig

and/or interstitial

cells

Male OAT Fertile

Page 188: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

177

Cdkn2d Spermatogonia Male Testicular atrophy Fertile

Cecr2 Spermatozoa Male Impaired Fertility Subfertile

Cenpb Spermatozoa Both Oligozoospermia Fertile

Cftr Sertoli cells; Spermatocytes;

Spermatids; Spermatozoa

Male Azoospermia Infertile

Cga Spermatozoa Both Hypogonadism Infertile

Cib1 Spermatozoa Male Azoospermia Infertile

Cks2 Spermatocytes Both Meiotic Arrest Infertile

Cldn11 Sertoli,

peritubular, Leydig

and/or interstitial

cells

Male Azoospermia Infertile

Clgn Spermatozoa Male Impaired fertility Infertile

Clock

Cnot7 Spermatocytes; Spermatids Male OAT Infertile

Cpe Both Hypospermatogenesis Subfertile

Cpeb1 Spermatocytes Both Azoospermia Infertile

Cplx1 Spermatozoa Male Impaired Fertility Subfertile

Crem Spermatids; Spermatozoa Male Azoospermia Infertile

Crisp1 Spermatozoa Male Impaired Fertility Fertile

Crtc1 Both ? Infertile

Crybb2 Both Oligozoospermia Subfertile

Csda Spermatocytes Male Oligozoospermia Variable Infertility

Csf1 Spermatogonia Both Oligozoospermia Subfertile

Csf2 Both None Subfertile

Csnk2a2 Spermatids Male Globozoospermia Infertile

Cstf2t Spermatocytes; Spermatozoa Male OAT Infertile

Ctnnb1 Sertoli Cells; Spermatogonia ? Sertoli-cell Only Infertile

Cugbp1 Spermatids Male Azoospermia (variable) Variable Infertility

Cux1 Male None Subfertile

CylD Spermatids Male Azoospermia Infertile

Cyp11a1 Both Gonadal degeneration Not reported

Cyp17a1 Sertoli,

peritubular, Leydig

and/or interstitial

cells

Male Asthenoteratozoospermia Infertile

Page 189: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

178

Cyp19a1 Sertoli,

peritubular, Leydig

and/or interstitial

cells, Spermatogonia

Both Oligozoospermia Progressive Infertility

D6mm5e Spermatocytes Male Oligozoospermia Infertile

Dazap1 Spermatocytes Both Meiotic Arrest Variable lethality;

infertile

Dazl Spermatogonia Both Sertoli-cell only Infertile

Ddr2 Sertoli cells; Leydig cells;

Spermatogonia

Both Testicular atrophy Infertile

Ddx25 Spermatids Male Azoospermia Infertile

Ddx4 Spermatogonia Male Meiotic Arrest Infertile

Dhcr24 Sertoli cells; Spermatocytes;

Spermatids;

Both Seminiferous tubule degeneration;

OAT

Infertile

Dhh Sertoli,

peritubular, Leydig

and/or interstitial

cells

Male Azoospermia Infertile

Dicer1 Spermatocytes Both Oligoteratozoospermia Infertile

Dmc1 Spermatocytes Both Meiotic Arrest Infertile

Dmrt1 Sertoli,

peritubular, Leydig

and/or interstitial

cells; Spermatocytes

Male Azoospermia Infertile

Dmrtc2 Spermatocytes Male Meiotic Arrest Infertile

Dnahc1 Spermatozoa Male Asthenozoospermia Infertile

Dnaja1 Sertoli,

peritubular, Leydig

and/or interstitial

cells

Male Oligoasthenozoospermia Variable

infertility/subfertility

Dnmt3l Spermatogonia Both Azoospermia Infertile

Drosha Spermatocytes; Spermatids ? Azoospermia/Oligoteratozoospermi

a

Infertile

Dyp19l2 Spermatids; Spermatocytes Male Globozoospermia Infertile

Egr1 Both Hypogonadism Infertile

Egr4 Spermatocytes; Spermatozoa Male Hypospermatogenesis Infertile

Ehd1 Spermatids Male Azoospermia Infertile

Ehmt2 Spermatocytes;

Spermatogonia

Both Sertoli-cell only; Meiotic arrest Infertile

Eif4g3 Spermatocytes Male Meiotic Arrest Infertile

Elavl1 Spermatocytes; Spermatids Male Meiotic Arrest Infertile

Elovl2 Spermatocytes Male Azoospermia Infertile

Page 190: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

179

Emx2 Both Gonadal hypoplasia Not assessed

Epas1 Sertoli Cells ? Azoospermia Infertile

Epb4.1l2 Sertoli cells; Spermatids Male Azoospermia Infertile

Ercc1 Spermatocytes Both Sertoli-cell only Infertile

Espl1 Spermatogonia Male Sertoli-cell only Fertile

Esr1 Sertoli cells; spermatogonia;

spermatocytes; spermatids;

spermatozoa

Both Hypospermataogenesis;

Asthenozoospermia.

Infertile

Esr2 Both None Fertile

Etv4 Male None Infertile

Etv5 Spermatogonia, Sertoli,

peritubular, Leydig

and/or interstitial

cells

Male Sertoli cell only Progressive Infertility

Exo1 Spermatocytes Both Meiotic Arrest Infertile

Faah Spermatozoa Both Impaired Fertility Subfertile

Fabp9 Spermatozoa ?? Teratozoospermia Fertile

Fads2 Spermatids Male Azoospermia Infertile

Fanca Spermatocytes Both Reduced Fertility Subfertile

Fancc PGC Both Hypogonadism Infertile

Fancg Sertoli cells; spermatids Both Hypogonadism Subfertile

Fancl PGC Both Sertoli cell only in younger mice Delayed fertility

Fgf9 Male Sex-reversal; testicular hypoplasia Not assessed

Fhl5 Spermatozoa Male Oligoteratozoospermia Fertile

Fkbp4 Both None Infertile

Fkbp6 Spermatocytes Male Meiotic Arrest Infertile

Fmr1 Male Macroorchidism Fertile

Fndc3a Spermatids; Spermatozoa Male Azoospermia Infertile

Fos Male None Fertile

Foxa3 Leydig cells; Spermatogonia Male Progressive to Sertoli-cell only Progressive subfertility

Foxp3 Male Hypogonadism Infertile

Fshb Both Decreased testis size Fertile

Fshr Sertoli cells; Spermatozoa Both OAT Fertile

Fus Spermatocytes Male Azoospermia Infertile

Page 191: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

180

Gal3st1 Spermatocytes Male Meiotic Arrest Infertile

Gamt Spermatids; Sertoli Cells Male Oligozoospermia Subfertile

Gapdhs Spermatozoa Male Asthenozoospermia Infertile

Gata4 Sertoli Cells ? Oligoasthenozoospermia Age dependant

infertility

Gba2 Sertoli Cells; Spermatozoa Male Globozoospermia Subfertile

Gdf7 Male Infertile

Gdi1 Sertoli,

peritubular, Leydig

and/or interstitial

cells; Spermatozoa

Both Azoospermia Infertile

Gdnf Sertoli,

peritubular, Leydig

and/or interstitial

cells

Male Hypospermatogenesis Fertile

Ggt1 Both Hypogonadism Infertile

Gja1 Sertoli,

peritubular, Leydig

and/or interstitial

cells, Spermatogonia

Both Azoospermia Postnatal Lethal

Glp1 Spermatocytes Both Azoospermia Infertile

Gm101 Spermatids; Spermatozoa Male Asthenozoospermia Infertile

Gmcl1 Spermatocytes; Spermatids;

Spermatozoa

Male OAT Variable

infertility/subfertility

Gnpat Spermatocytes Male Azoospermia Infertile

Gnrh1 Both Hypogonadism Infertile

Gnrhr Both Hypogonadism Infertile

Gopc Spermatids Male Globozoospermia Infertile

Gpr64 Male Subfertile

Gpx4 Spermatocytes; Spermatids Both OAT Infertile

Gtsf1 Spermatocytes Male Meiotic Arrest Infertile

H1fnt Spermatids Male OAT Subfertile

H2afx Spermatocytes Male Meiotic Arrest Infertile

H3f3a Spermatocytes Male Reduced fertility and copulatory

behavior

Subfertile

HexB Spermatozoa Both Impaired Fertility Progressive infertility

Hip1 Spermatids Male Azoospermia (Incomplete

penetrance)

Variable infertiltiy

Hmga1 Sertoli, Male Azoospermia Infertile

Page 192: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

181

peritubular, Leydig

and/or interstitial

cells

Hmgb2 Sertoli,

peritubular, Leydig

and/or interstitial

cells

Male Azoospermia Subfertile

Hnf1a Both Hypogonadism Infertile

Hook1 Spermatid Male Teratozoospermia Subfertile

Hormad2 Spermatocytes Male Meiotic Arrest Infertile

Hoxa10 Both Cryptorchidism Progressive infertility

Hoxa11 Both Cryptorchidism Infertile

Hsf1 Spermatocytes Both Meiotic Arrest Infertile

Hsf2 Spermatocytes, Spermatids Both Oligozoospermia Subfertile

Hsp90aa1 Spermatocytes Male Meiotic Arrest Infertile

Hsp90b1 Spermatozoa Male Asthenoteratozoospermia Infertile

Hspa2 Spermatocytes Male Meiotic Arrest Infertile

Hspa4 Spermatocytes Male Oligoasthenozoospermia Infertile

Hspa4l Spermatids; Spermatocytes;

Spermatozoa

Male Oligoasthenozoospermia Variable infertility

Ifnb1 Sertoli cells; Spermatogonia Male Progressive Azoospermia Progressive Infertility

Igf1 Both Hypogonadism Infertile

Il17rb Spermatocytes; Spermatids Both OAT Infertile

Il2rn Spermatozoa Male Teratozoospermia Subfertile

Immp2l Both Subfertile

Ing2 Spermatocytes Male Meiotic Arrest Infertile

Inha Sertoli,

peritubular, Leydig

and/or interstitial

cells

Both Gonadal tumours Secondary Infertility

Inpp5b Spermatozoa Male Asthenozoospermia Infertile

Insl3 Both Cryptorchidism Subfertile

Insl5 Spermatozoa Both Asthenozoospermia Subfertile

Insl6 Spermatocytes Male Oligoasthenozoospermia Variable Infertility

Page 193: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

182

Ip6k1 Spermatocytes; Spermatids Male Azoospermia Infertile

Jam3 Spermatids Male OAT Infertile

Jund Spermatozoa Male Asthenozoospermia; OAT Variable

Katnal1 Sertoli cells; Spermatids Male Oligozoospermia Infertile

Katnb1 Spermatocytes; Spermatids Male OAT Infertile

Kdm3a Spermatocytes/Spermatids Male Oligozoospermia Infertile

Kiss1 Both Hypogonadism Infertile

Kiss1r Both Hypogonadism Infertile

Kit Spermatogonia Both Sertoli-cell only Infertile

Kitl Sertoli,

peritubular, Leydig

and/or interstitial

cells

Both Sertoli cell only Infertile

Klc3 Spermatid ? Oligoasthenozoospermia Reduced Fertility

Klhl10 Spermatid Male OAT Infertile

Knuc1 Spermatozoa Male Asthenozoospermia Infertile

Krt9 Spermatids Male Teratozoospermia Fertile

Ldhc Spermatozoa Male Asthenozoospermia Variable infertility;

subfertility

Lep Both Hypogonadism Infertile

Lepr Both Hypogonadism Infertile

Lfng Both None Subfertile

Lgr4 Male Seminiferous epithelium rupture Infertile

Lhb Leydig cells; Spermatids Both Azoospermia Infertile

Lhcgr Sertoli,

peritubular, Leydig

and/or interstitial

cells

Both Azoospermia Infertile

Limk2 Spermatogonia,

Spermatocytes

Male Azoospermia Fertile

Lipe Spermatids Male Oligozoospermia Infertile

Lmna Spermatocytes Male Azoospermia Infertile

Lmtk2 Spermatids Male Azoospermia Infertile

Lrp8 Spermatozoa Male Asthenoteratozoospermia Infertile

Man2a2 Sertoli,

peritubular, Leydig

and/or interstitial

Male Azoospermia Mostly Infertile

Page 194: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

183

cells

Mark2 Both Gonadal hypotrophy Variable infertility

Mas1 Sertoli,peritubular, Leydig

and/or interstitial

cells; Spermatocytes

Male Hypospermatogenesis Fertile

Mc4r Male None Fertile

Mcph1 Spermatocytes Both Meiotic Arrest Infertile

Mei1 Spermatocytes Both Meiotic Arrest Infertile

Meig1 Spermatocytes, Spermatids Males Azoospermia Infertile

Mfge8 Spermatozoa Male Impaired Fertility Subfertile

Mlh1 Spermatocytes Both Meiotic Arrest Infertile

Mlh3 Spermatocytes Both Meiotic Arrest Infertile

Mll5 Spermatids Male Azoospermia Infertile

Mmel1 Spermatozoa Male Impaired Fertility Subfertile

Morc1 Spermatocytes Male Meiotic Arrest Infertile

Mov10l1 Spermatocytes Male Meiotic Arrest Infertile

Msh4 Spermatocytes Both Meiotic Arrest Infertile

Msh5 Spermatocytes Both Meiotic Arrest Infertile

Mtap7 Sertoli,

peritubular, Leydig

and/or interstitial

cells; Spermatids

Male Azoospermia Infertile

Mthfr Spermatogonia Male Sertoli cell only Infertile

Mtmr2 Spermatocytes; Spermatids Male Azoospermia Infertile

Mybl1 Spermatocytes Male Meiotic Arrest Infertile

Myh10 Spermatocytes Male Azoospermia Infertile

Nanos2 Spermatogonia Male Azoospermia Infertile

Nanos3 PGC Both Sertoli cell only Infertile

Ncoa1 Both Decreased testis size Fertile

Ncoa6 Both Not examined Subfertile

Nek1 Spermatogonia;

Spermatocytes

Both Testicular hypoplasia; Azoospermia Infertile

Nfe2l2 Spermatogonia,

Spermatocytes, Spermatids,

Spermatozoa

? Progressive to Sertoli Cell Only Progressive Infertility

Nhlh2 Both Hypogonadism Infertile

Page 195: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

184

Nkd1 Spermatids Male Oligozoospermia Subfertile

Nmp4 Spermatogonia Male Sertoli-cell only/Focal

spermatogenesis

Variable infertility

Nos1 Both Hypogonadism Infertile

Npc1 Spermatocytes; Spermatozoa Both OAT Infertile

Nphp1 Spermatids Male OAT Infertile

Nphp4 Spermatids; Spermatozoa Male Oligoasthenozoospermia Infertile

Nr0b1 Sertoli,

peritubular, Leydig

and/or interstitial

cells

Male Abnormal Spermatogenesis Infertile

Nr2c2 Spermatocytes Both Oligozoospermia Subfertile

Nr2f2 Sertoli,

peritubular, Leydig

and/or interstitial

cells

Both Azoospermia Variable fertility

Nr5a1 PGC Both Gonadal hypoplasia Infertile

Nsun7 Spermatozoa Male Asthenozoospermia Infertile

Nxf2 Spermatogonia;

Spermatocytes; Spermatozoa

Male Variable by strain Variable infertility

Oaz3 Spermatozoa Male Teratozoospermia Infertile

Odf1 Spermatozoa Male Teratozoospermia Infertile

Odf2 Spermatids, Spermatozoa Male Asthenozoospermia Variable fertility

Otx1 Both Hypogonadism Delayed fertility

Otx2 Both Hypogonadism Infertile

Ovol1 Spermatocytes Both Meiotic Arrest Subfertile

P2rx6 Spermatogonia; Spermatozoa Male Oligozoospermia Infertile

Pacrg Spermatids Male Oligozoospermia Infertile

Pafah1b1 Spermatids Male Azoospermia Infertile

Pank2 Spermatids Both Azoospermia Infertile

Parp2 Spermatids Male Azoospermia Subfertile

Pax8 Both Secondary seminiferous epithelium

degeneration

Infertile

Paxip1 Spermatocytes ? Meiotic Arrest Infertile

Pcsk4 Spermatozoa Male Impaired fertility Infertile

Pcyt1b Both Seminiferous tubule degeneration

(progressive)

Subfertile

Page 196: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

185

Pdilt Spermatozoa Male Impaired Fertility Infertile

Pdrm9 Spermatocytes Both Meiotic Arrest Infertile

Pebp1 Spermatozoa Male Impaired fertility Subfertile

Pfdn5 Spermatocytes; Spermatids Both Oligozoospermia Infertile

Pgap1 Spermatozoa Male Impaired Fertility Postnatal lethal;

infertile

Pgs1 Spermatozoa; Spermatids Male Asthenozoospermia Infertile

Pick1 Spermatids; Spermatocytes Male Globozoospermia Infertile

Piga Both Oligozoospermia Infertile

Pin1 Spermatogonia Male Progressive to Sertoli Cell Only Progressive infertility

Piwil1 Spermatids; Spermatozoa Male Azoospermia Infertile

Piwil2 Spermatocytes Male Meiotic Arrest Infertile

Piwil4 Spermatocytes Male Meiotic Arrest Infertile

Pla2g4c Spermatozoa Male Asthenozoospermia Subfertile

Plcb1 Spermatozoa Both Impaired Fertility Variable lethality;

infertile

Plcd4 Spermatozoa Male Impaired Fertility Variable Infertility/

Subfertile

Plk4 Spermatogonia Male Patchy germ cell loss Fertile

Pltp Spermatozoa Male Asthenozoospermia Subfertile

Pmis2 Spermatozoa Male Impaired fertility Infertile

Pms2 Spermatocytes Both Teratozoospermia Infertile

Pold4 Spermatozoa Male Asthenozoospermia Infertile

Polg Spermatozoa Both Azoospermia Reduced feriltiy

Pou1f1 Both Hypogonadism Infertile

Ppm1d Spermatids Male Testicular atrophy Subfertile

PPP1cc Spermatids; Spermatocytes;

Sertoli Cells

Male Azoospermia Infertile

Prkaa1 Spermatozoa Both Asthenoteratozoospermia Subfertile

Prkaca Spermatozoa Male Asthenozoospermia Infertile

Prkar1a Spermatids; Spermatozoa Male OAT Subfertile

Prlr Spermatozoa Both Impaired Fertility Variable infertility

Prm1 Spermatids; Spermatozoa Male Asthenoteratozoospermia Infertile

Prm2 Spermatids; Spermatozoa Male Asthenoteratozoospermia Infertile

Page 197: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

186

Prnd Spermatids; Spermatozoa Male OAT Infertile

Prop1 Both Hypogonadism Infertile

Psmc3ip Spermatocytes Both Meiotic Arrest Infertile

Psme4 Spermatocytes; Spermatids Male OAT Infertile

Pum1 Spermatocytes ? ` Subfertile

Pvrl2 Spermatids Male Teratozoospermia Infertile

Pxt1 Spermatocytes Male Azoospermia Infertile

Pygo2 Spermatids Male Azoospermia Infertile

Rabl2 Spermatozoa Male Oligoasthenozoospermia Infertile

Rad18 Spermatogonia Male Reduced fertility Progressive subfertility

Rad21l Spermatocytes Both Meiotic Arrest Infertile

Rad23b PGC; spermatogonia Male Sertoli-cell only Infertile

Rad51c Spermatocytes Both Meiotic Arrest Variable infertility

Ranbp1 Spermatids Male Azoospermia Infertile

Rara Spermatocytes Male Azoospermia Infertile

Rarg Male None Infertile

Rasip1 Spermatozoa Male Impaired Fertility Infertile

Rbmxl2 Spermatozoa Male Azoospermia Infertile

Rbp4 Sertoli,

peritubular, Leydig

and/or interstitial

cells, Spermatogonia;

Spermatids

Male Variable Infertile

Rec8 Spermatocytes Both Meiotic Arrest Infertile

Rhox5 Spermatozoa Male OAT Subfertile

Rimbp3 Spermatids Male Teratozoospermia Infertile

Rnf17 Spermatids Male Azoospermia Infertile

Rnf8 Spermatids Male OAT Infertile

Ros1 Spermatozoa Male Asthenozoospermia Infertile

Rxfp1 Spermatocytes Male Azoospermia (variable) Variable infertility

Rxfp2 Male Cryptorchidism Infertile

Rxrb Spermatids; Sertoli Cells Male OAT Infertile

Safb Sertoli,

peritubular, Leydig

and/or interstitial

cells, Spermatogonia;

Spermatids

Both Azoospermia Subfertile

Page 198: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

187

Sbf1 Sertoli,

peritubular, Leydig

and/or interstitial

cells

Male Azoospermia Infertile

Sepp1 Spermatozoa Male Asthenoteratozoospermia Infertile

Sept12 Spermatids; Spermatocytes Male Hypospermatogenesis;

Azoospermia; OAT

Variable infertility

Sept4 Spermatids; Spermatocytes Male Asthenozoospermia Infertile

Serpina5 Sertoli,

peritubular, Leydig

and/or interstitial

cells

Male Asthenoteratozoospermia Infertile

Serpine2 Male None Subfertile

Sgol2 Spermatocytes Both Meiotic Arrest Infertile

Sh2b1 Spermatozoa;

Sertoli,

peritubular, Leydig

and/or interstitial

cells

Both Oligozoospermia Subfertile

Siah1a Spermatocytes Both Meiotic Arrest Infertile

Sin3a Spermatogonia ? Sertoli-cell only Infertile

Sirt1 Spermatocytes; Spermatids;

Spermatozoa

Male OAT Infertile

Six5 Spermatids Male Azoospermia/oligozoospermia Infertile

Slc12a2 Sertoli,

peritubular, Leydig

and/or interstitial

cells; Spermatids;

Spermatozoa

Male Azoospermia Infertile

Slc19a2 Spermatogonia Male Meiotic block Infertile

Slc25a4 Spermatocytes Male Meiotic Arrest Infertile

Slc4a2 Spermatids Male Azoospermia Infertile

Slc9a10 Spermatozoa Male Asthenozoospermia Infertile

Sly1 Spermatids; Spermatozoa Male Teratozoospermia Subfertile

Smad1 PGC Both Gonadal Hypoplasia Not assessed

Smad5 PGC Both Gonadal Hypoplasia Not assessed

Smc1b Spermatocytes Both Meiotic Arrest Infertile

Smcp Spermatozoa Male Athenozoospermia, Impaired

fertility

Infertile

Smpd1 Sertoli cells; Spermatozoa Male Asthenoteratozoospermia Subfertile

Page 199: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

188

Sod1 Spermatozoa Both Impaired Fertility Subfertile

Sohlh1 Spermatogonia Both Azoospermia Infertile

Sohlh2 Spermatogonia Both Sertoli-cell only Infertile

Sox3 Sertoli cells Both Hypogonadism; Variable

Oligozoospermia to Sertoli-cell

only

Subfertile

Sox8 Sertoli,

peritubular, Leydig

and/or interstitial

cells

Male Abnormal Spermatogenesis Variable Subfertility

Spaca1 Spermatids; Spermatozoa Male Globozoospermia Infertile

Spag16 Spermatid Male OAT Infertile

Spag6 Spermatozoa Male Asthenoteratozoospermia Infertile

Spag9 Spermatozoa Male Oligoasthenozoospermia Subfertile

Spam1 Spermatozoa Male Impaired Fertility Subfertile

Spata16

Spata22 Spermatocytes Both Meiotic Arrest Infertile

Spef2 Spermatids Male OAT Infertile

Spem1 Spermatids; Spermatozoa Male Teratozoospermia Infertile

Spo11 Spermatocytes Both Meiotic Arrest Infertile

Sprm1 Spermatozoa Male Impaired Fertility Subfertile

Spz1 Spermatogonia;

Spermatocytes; Spermatids

Male Oligoteratozoospermia Progressive infertility

Star Leydig

and/or interstitial

cells

Both Lipid deposits in interstitial regions Not assessed

Stat3 Both Hypogonadism Infertile

Stk11 Spermatids; Spermatozoa Male OAT Infertile

Stra8 Spermatogonia Both Progressive germ cell depletion Infertile

Strbp Spermatids Male OAT Subfertile

Stx2 Spermatocytes Male Meiotic Arrest Infertile

Styx Spermatids Male OAT Infertile

Sult1e1 Sertoli,

peritubular, Leydig

and/or interstitial

cells; Spermatozoa

Both Oligoasthenozoospermia Progressive Infertility

Sun1 Spermatocytes Both Meiotic Arrest Infertile

Syce3 Spermatocytes Both Meiotic Arrest Infertile

Page 200: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

189

Sycp1 Spermatocytes Both Meiotic Arrest Infertile

Sycp2 Spermatogonia,

spermatocytes

Both Meiotic Arrest Infertile

Sycp3 Spermatocytes Both Meiotic Arrest Infertile

Taf4b Spermatogonia Both Sertoli-cell only Progressive infertility

Taf7l Spermatozoa Male OAT Infertile

Taldo1 Spermatozoa Male Asthenozoospermia Infertile

Tas2r105 Spermatids Male Azoospermia Infertile

Tbpl1 Spermatids Male Azoospermia Infertile

Tcf21 (Developmental) Both Gonadal Hypoplasia Infertile

Tcte3 Spermatozoa Male Asthenozoospermia Infertile

Tdrd1 Spermatids Male Azoospermia Infertile

Tdrd6 Spermatids Male Azoospermia Infertile

Tdrd7 Spermatids Male Azoospermia Infertile

Tdrd9 Spermatocytes Male Meiotic Arrest Infertile

Tekt2 Spermatozoa Male Asthenoteratozoospermia Infertile

Tekt3 Spermatozoa Male Asthenozoospermia fertile

Tekt4 Spermatozoa Male Asthenozoospermia Subfertile

Tert Spermatocytes Both Progressive germ cell loss Progressive infertility

Tex11 Spermatocytes Male Azoospermia Infertile

Tex14 Spermatocytes Male Meiotic Arrest Infertile

Tex15 Spermatocytes Male Meiotic Arrest Infertile

Tgfb1 Both Infertile

Theg Spermatids Male Asthenozoospermia Infertile

Thoc1 Spermatocytes Both Oligoasthenozoospermia Infertile

Tial1 PGC Both Sertoli-cell only Infertile

Tmf1 Spermatids; Spermatozoa Male OAT Infertile

Tnp1 Spermatids; Spermatozoa Male Asthenozoospermia Subfertile

Tnp2 Spermatids; Spermatozoa Male Asthenozoospermia Subfertile

Top3b Spermatocytes Both Sperm aneuploidy Progressive infertility

Tpst2 Spermatozoa Male Asthenozoospermia; Impaired

fertility

Infertile

Trip13 Spermatocytes Both Meiotic Arrest Infertile

Trpv6 Male Impaired Fertility Infertile

Page 201: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

190

Tsc22d3 Spermatogonia; Sertoli cells Male Sertoli-cell only Infertile

Tsn Spermatozoa Male Progressive Oligozoospermia Fertile

Tssk1/2 Spermatids Male OAT Infertile

Tssk6 Spermatids; Spermatozoa Male Asthenoteratozoospermia Infertile

Ttll1 Spermatids; Spermatozoa Male Asthenoteratozoospermai Infertile

Tyst2 Spermatozoa Male Impaired Fertility Infertile

Ubb Spermatocytes Both Meiotic Arrest Infertile

Ube2b Spermatocytes; Spermatids Male Azoospermia Infertile

Ube3a Spermatozoa Both Hypospermatogenesis; Impaired

Fertility

Subfertile

Ubr2 Spermatocytes Male Meiosis Variable lethality;

infertile

Uchl1 Spermatocytes Both Meiotic Arrest Infertile

Usp2 Spermatids; Spermatozoa Male Impaired Fertility Subfertile

Utp14b Spermatogonia Male Progressive germ cell depletion Progressive infertility

Vdac3 Spermatozoa Male Asthenozoospermia Infertile

Vdr Both Oligoasthenozoospermia Infertile

Vhl Sertoli cells; Spermatogonia;

Spermatocytes; Spermatids

? Oligozoospermia Infertile

Vipr2 Spermatids Male Oligozoospermia Progressive Infertility

Vps54 Sertoli cells; Spermatids;

Spermatozoa

Male Globozoospermia Infertile

Vrk1 Spermatogonia Both Sertoli-Cell only Infertile

Wipf3 Spermatozoa Male Impaired Fertility Infertile

Wnt7a Both Obstructive Azoospermia Infertile

Wt1 Spermatogonia Both Sertoli cell only Infertile

Ybx2 Spermatids Both Azoospermia Infertile

Zbtb16 Spermatogonia Male Progressive germ cell depletion Subfertile

Zfx PGC Both Oligozoospermia Fertile

Zmynd15 Spermatids Male Azoospermia Infertile

Zpbp1 Spermatids; Spermatozoa Male Globozoospermia Infertile

Zpbp2 Spermatids; Spermatozoa Male Globozoospermia Subfertile

Page 202: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

191

Appendix A.2: Yeast 3-hybrid screen for substrates of the SH3GLB1T-PPP1CC2 holoenzyme

Introduction

The Yeast 2-hybrid (Y2H) is a commonly applied technique for identifying binary protein

interactions on a large scale(Fields &Song. 1989). Due to its ease of use, and ability to screen large

libraries in a relatively short amount of time the Y2H system has been utilized extensively by many

research groups and has allowed for the identification of thousands of protein interaction pairs. In the

Y2H system, the protein of interest or “bait” is expressed in the yeast S. cerevisiae as a fusion protein

with the GAL4 DNA binding domain (GAL4-BD). Yeast cultures expressing this bait-GAL4-BD fusion

protein are transformed with DNA libraries encoding protein coding sequences fused to the GAL4

trascriptional activation domain (GAL4-AD), otherwise known as “prey”. When the bait protein

interacts with a prey protein from the library, the two GAL4 domains are brought into close contact, and

subsequently are able to stimulate the transcription of a reporter gene. Thus positive interaction clones

can be identified via a selectable marker and the sequence of the prey protein can be determined,

identifying the protein that forms a binary interaction with the bait protein.

Previous studies from the Varmuza lab have utilized the Y2H system to identify two putative

isoform-specific regulatory subunits of PPP1CC2: Spz1 (Hrabchak & Varmuza 2004) and SH3GLB1T

(Endophilin B1t) (Hrabchak et al. 2007). Both of these proteins bind only to the PPP1CC2 isoform, and

the interaction is abolished when the unique 18 amino acid C-terminus of PPP1CC2 is deleted. Spz1 and

SH3GLB1T both contain the “RVxF” motif that is commonly found in PP1 regulatory subunits, and

decrease the activity of PPP1CC2 towards the substrate phosphorylase a in vitro.

SH3GLB1T is a testis specific splice isoform of the Sh3glb1 gene, encoding a protein that lacks

the 10 most C-terminal amino acids of the full-length SH3GLB1 protein (Hrabchak et al. 2007).

SH3GLB1 has been shown to play a role in multiple cellular processes including autophagy, endocytosis,

and apoptosis (reviewed in Takahashi et al., 2009 (Hrabchak et al. 2007, Takahashi et al. 2009)). It has

previously been proposed that premature release of the germ cells from the seminiferous tubules may be

Page 203: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

192

a cause of the infertility phenotype observed in Ppp1cc mutant mice (Varmuza &Ling. 2003). Premature

germ cell release could arise from improper recycling of cell-cell junctional complexes via endocytosis

in the seminiferous tubules. Thus it has been hypothesized that disregulation of SH3GLB1T in the

Ppp1cc mutant testis could play a role in this process (Hrabchak et al. 2007)

After identifying PPP1CC2 specific regulatory subunits, the focus shifted to identifying

substrates of PPP1CC2 holoenzymes. In an attempt to identify substrates of the SH3GLB1T-PPP1CC2

holoenzyme, I conducted a yeast 3-hybrid (Y3H) screen of a mouse testis cDNA library. As the Y2H is

only capable of recognizing binary interactions, attempts to identify ternary interactions (regulatory

subunit, catalytic subunit, substrate) necessitated a more advanced technique. The Y3H system is based

on the Y2H system, but includes the addition of an inducible third protein(Fields & Song 1989, Tirode et

al. 1997). This inducible third protein functions to “bridge” interactions between the bait and prey fusion

proteins. To conduct a Y3H screen, a library is first screened with the inducible protein expressed. After

selecting positive colonies the inducible bridge protein is switched off. Colonies that are no longer

positive without the bridge protein are dependent upon all 3 proteins being present, and thus represent a

ternary protein complex. My Y3H screen featured the regulatory subunit SH3GLB1T-GAL4BD, with an

inducible PPP1CC2, and a mouse testis cDNA library was screened for third-party interactors which we

hypothesized would be putative substrates of the SH3GLB1T-PPP1CC2 holoenzyme.

Materials and Methods

Cloning of Yeast 3-hybrid Plasmid

The pBridge™ plasmid (Clontech) was obtained as a gift from Prof. Darrell Desveuax

(University of Toronto, Department of Cell and Systems Biology). pBridge features two multiple

cloning sites (MCS): MCSI, which is immediately downstream of a GAL4-BD sequence, and MCSII

which is downstream of a methionine responsive promoter. First, SH3GLB1T was cloned into MCSI.

The full-length SH3GLB1T CDS obtained via a SmaI and XhoI digestion of the SH3GLB1T-pGADT7

Page 204: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

193

plasmid (Hrabchak et al. 2007), followed by gel purification. The SH3GLB1T sequence was then ligated

into the pBridge plasmid that had been digested with SmaI and SalI. Next, PPP1CC2 was cloned into

MCSII of pBridge. The PPP1CC2 CDS was amplified from the pGEM7Zf plasmid carrying the full

length cDNA sequence (Mann et al. 1995, Hrabchak et al. 2007) using the following primers: forward –

TCGGCGGCGCGGCCGCGATG; reverse – GTCACCGCAGGATCCAGAATGTAGCCAAAG. The

resultant PCR product was digested with NotI and BglII and ligated into the SH3GLB1T containing

pBridge plasmid that dad been digested with Eco52I and BamHI. The presence and integrity of the

cloned DNA sequences, as well as the frame of insertion was verified by DNA sequencing.

Yeast 3-Hybrid Screening

The Y3H bait vector containing the SH3GLB1T-GAL4BD and inducible PP1cγ2 were

transformed into the S. cerevisiae strain AH109 via the LiAc protocol described in the Clontech Yeast

Protocols Handbook (2001) and maintained on synthetic dropout media lacking tryptophan (SD-W

). Y3H

screening was conducted using the Mouse Testis Matchmaker cDNA Library (clontech), that had been

previously amplified ((Hrabchak et al. 2007)). Library plasmid DNA was purified using Purelink™

HIPure Plasmid Maxiprep Kit (Invitrogen) and transformed into the bait expressing strain according the

protocol outlined the the Clontech Yeast Protocols Handbook. Transformed yeast were plated on

synthetic dropout media lacking tryptophan, leucine, histidine, and adenine (SD-W-L-H-Ade

) supplemented

with 25 µM 3-Amino-1,2,4-triazole (3-AT). Transformation plates were incubated at 30° C for 5 days,

whereupon positive colonies were replicaplated onto both SD-W-L-H-Ade

supplemented with 2mM 3-AT and

SD-W-L-H-Ade

supplemented with 2mM 3-AT and 2mM Methione (SD-W-L-H-Ade+M

). Replicaplates were then

grown at 30°C for a further 3-5 days.

Western blotting

Page 205: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

194

Protein extraction, and SDS-PAGE were conducted using standard protocols. Western blotting

was performed with primary antibodies for SH3GLB1 (anti-Bif-1 Imgenex) and PPP1CC (anti-PPP1CC2

N-19, Santa Cruz Biotechnology) at dilution of 1:1000, and HRP coupled secondary antibodies.

Results and discussion

In an attempt to identify proteins that interact with the SH3GLB1T-PPP1CC2 holoenzyme in the

mouse testis, a Y3H screen was conducted using a mouse testis cDNA library. The bait construct for this

Y3H strain utilized the pBridge backbone and contained both a SH3GLB1T-GAL4BD fusion protein as

mouse PPP1CC2 under the control of the S. cerevisiae MET25 promoter, which is active at a low

methionine concentration and inactive at elevated methionine concentrations. After LiAc mediated

transformation of the bait plasmid into the S. cerevisiae strain AH109, western blotting was used to

verify the expression of both the SH3GLB1T-GAL4BD fusion protein and PPP1CC2 (Figure A.2.1). In

order to optimize the methionine concentration needed to abolish PPP1CC2 expression bait expressing

yeast strains were grown in SD-W

supplemented with methionine concentrations ranging from 0.134 – 1

mM methionine. Western blots for PPP1CC2 from these yeast cultures demonstrated prominent

PPP1CC2 expression until 1 mM methionine where PPP1CC2 expression was not evident (Figure A.2.2).

Three rounds of Y3H screening were conducted, using the SH3GLB1T-GAL4BD + PPP1CC2

bait construct, amounting to an estimated 7 x 106 clones, or 2-fold coverage of the cDNA library. A total

of 71 positive colonies were observed. Upon replicaplating on medium supplemented with 2 mM

methionine to completely abolish PPP1CC2 expression from the bait construct, all 71 colonies survived.

This indicates that none of the interactions were dependent upon the presence of PPP1CC2.

The Y3H method allows for the rapid screening of millions of clones for ternary protein

interactions. We sought to use this approach to identify substrates of the SH3GLB1T-PPP1CC2

holoenzyme. Although 71 positive clones were initially identified, not one was dependent upon the

presence of PPP1CC2. This indicates that the 71 positive clones were likely direct interactors with the

Page 206: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

195

Figure A.2.1: Expression of Yeast 3-hybrid bait proteins in yeast. Western blots depicting expression

of SH3GLB1T-GAL4BD (A) and PPP1CC2 (B).

Page 207: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

196

Page 208: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

197

Figure A.2.2: Methionine regulation of PPP1CC2 expression from pBridge vector in yeast. Western

blot depicting expression of PPP1CC2 in yeast grown in differing methionine concentration. Numerical

values indicated are concentrations in mM.

Page 209: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

198

Page 210: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

199

SH3GLB1T protein, as well as false positives. Therefore, the Y3H approach described in this chapter

was ineffective in identifying any PPP1CC2 substrates.

There are a number of possible explanations for the failure of this approach. First, yeast based

screening methods are a highly artificial system, despite being regarded as an in vivo approach.

Although we screened a mouse testis cDNA library, and thus were looking at testis proteins, there is no

guarantee that testis proteins expressed in yeast behave the same as testis proteins in their native context.

Post-translational modifications, such as phosphorylation, that occur in the testis may not be present

when a testis protein is expressed in yeast. Thus if a potential PPP1CC2 substrate, was never

phosphorylated to begin with, it is unlikely to interact with a protein phosphatase such as PPP1CC2.

Likewise, PPP1CC2 has been demonstrated to be subject to phosphorylation, which affects its activity

and possibly its binding to other proteins (Huang & Vijayaraghavan 2004). Another limitation of the

Y3H approach at identifying PP1 substrates is due to the transient nature of many PP1-substrate

interactions. While some substrates have been shown to form stable complexes with PP1 catalytic

subunits, others form only weak interactions (Bollen et al. 2010). Finally, while the Y3H system allows

for the expression of one additional protein than the traditional Y2H system, it still falls short of

reproducing larger multi-protein complexes that exist in vivo. It remains a possibility that a PPP1CC2-

SH3GLB1T holoenzyme is a part of a larger complex, and additional proteins are required for substrate

binding. Situations such as this would be missed in our Y3H screen.

In order to address the short comings of the Y3H system in identifying PPP1CC2 substrates, a

more powerful approach is needed. As a result we chose to perform the tandem affinity purification

experiments detailed in chapter 2. Utilizing a TAP-tagged PPP1CC2 to purify interacting proteins direct

from the mouse testis would not suffer from the same shortcomings as Y3H. Because prey proteins

would be expressed in their native tissue, at endogenous levels, they would have the appropriate

expression pattern and post-translational modifications necessary to interact with PPP1CC2. In addition

TAP is not limited to binary, or ternary protein complexes like the Y2H and Y3H systesms. During the

Page 211: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

200

course of this research, Takahasi et al., produced a SH3GLB1 knockout mouse (Takahashi et al. 2007).

Loss of SH3GLB1 was found to suppress autophagosome formation, and prolong cell survival under

nutrient starvation conditions, but no impact on male fertility was reported although the testes were not

specifically examined. This likely indicates that a SH3GLB1T-PPP1CC2 holoenzyme does not play a

role in regulation of spermatogenesis. Conversely there remains a possibility that SH3GLB1T-PPP1CC2

plays a minor but not essential role in spermatogenesis, or that functionally redundant proteins exist.

Regardless, future experiments will shift away from examining only SH3GLB1T-PPP1CC2 interacting

proteins, to a more global approach that will seek to identify all PPP1CC2 interacting proteins.

Page 212: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

201

Appendix A.3: The effect of Ppp1cc deletion on the localization of candidate substrates HSPA2 and

β-tubulin in mouse spermatogenic cell nuclei.

Author’s Note: A modified version of the experiments described in this section was published as a part

of the following paper: H. Henderson, G. MacLeod, C. Hrabchak and S. Varmuza (2011). New candidate

targets of protein phosphatase-1c-gamma-2 in mouse testis revealed by a differential phosphoproteome

analysis. Int J Androl. 34(4):339-51.

Included in this section are only those experiments that I personally performed. Text for this section was

written by myself, with editing by H. Henderson and S. Varmuza

Introduction

In an attempt to identify candidate substrates of PPP1CC2 in the testis a comparative

phosphoproteome analysis was conducted according to the rationale that substrates will be

hyperphosphorylated in Ppp1cc knockout testes. Ten hyperphosphorylated proteins were identified via

2-dimensional electrophoresis followed by ProQ phosphoprotein staining. Two of these proteins, have

previously been shown to play a role in spermatogenesis: HSPA2 and β-tubulin. HSPA2 is a testis-

specific 70kDa related heat-shock protein, whose deletion leads to spermatogenic arrest at the pachytene

spermatocyte stage, followed by spermatocyte apoptosis (Dix et al. 1996). β-tubulin is the primary

component of the manchette a structure which envelopes the spermatid nucleus and is thought to aid in

nuclear shaping and condensation (Kierszenbaum. 2002). Upon further investigation, both HSPA2 and

β-tubulin were found to co-immunoprecipitate with PPP1CC2 in mouse testis protein lysate.

To further examine the interaction between PPP1CC2 and HSPA2 in the mouse testis I have

examined the effect of deletion of Ppp1cc on HSPA2 and β-tubulin localization in spermatogenic cell

nuclear suspensions.

Materials and Methods

Preparation of Surface-spread nuclear suspensions and Immunohistochemistry

Page 213: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

202

Dissected testes were placed in MEM after removal of the tunica albuginea and homogenized

using a razor blade to release germ cells from the seminiferous tubules. The resultant homogenate was

mixed and allowed to settle for several minutes. The developing germ cells were then pelleted by

centrifugation at 2,000 rpm for 2 minutes. Following removal of the supernatant the germ cells were re-

suspended in 25 μL of MEM. 4 μL of germ cell suspension was then surface spread and prepared for

immunohistochemistry as previously (Moens et al. 1987, Moens & Earnshaw 1989, Dobson et al. 1994).

The cell suspension was dropped onto a 0.5% NaCl solution and cells were picked up on glass slides.

Cells were fixed for 3 minutes in a 2% paraformaldehyde solution containing 0.36% SDS followed by 3

additional minutes in 2% paraformaldehyde and 3 one minutes washes in 0.4% Photoflo solution. Slides

were then air-dried. Prior to application of primary antibodies, the slides were incubated for 10 minutes

in PBS + 0.4% Photoflo solution followed by 10 minutes in the same solution with 0.05% Triton-X-100

and 10 minutes in 10% antibody dilution buffer. Incubation with anti-β-Tubulin (1:500) or anti-HSPA2

(1:1000) was carried out overnight at room temperature. Select slides were also incubated with CREST

anti-serum (1:500) to visualize kinetchores and/or anti-COR (1:500) to visualize synaptonemal

complexes. Slides were then washed using the same solutions as prior to primary incubation. Secondary

antibody incubations were all carried out at 1:500 using appropriate combinations of goat and rabbit Cy3

(Jackson), goat anti mouse Cy2, goat anti mouse Cy3, goat anti-human Alexa 488, and goat anti-human

rhodamine at 37 °C for 1 hour. Following secondary antibody incubation slides were washed for 10

minutes each in 0.4% Photoflo in PBS, 0.4% Photoflo plus 0.05% Triton-X-100 in PBS and again in

0.4% Photoflo, followed by 2 one minute washes in 0.4% Photoflo in water. Slides were then

counterstained with DAPI (300nM) for 10 minutes, followed by 4 one minute washes in PBS and

mounted as before. Images were captured and processed as previously described using the appropriate

filters at 100X magnification.

Quantification of β-tubulin abnormalities was performed by scoring 50 randomly chosen

manchettes on slides made from wild-type and Ppp1cc mutant mice as abnormal or normal, based on

Page 214: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

203

existing literature on manchette structure (Cherry & Hsu 1984, Russell et al. 1990). Assessment of

abnormalities was done conservatively with any ambiguities being scored as normal, to allow for any

disruptions in manchette structure brought about in slide preparation. Assessment of statistical

significance was performed by a Chi-squared test using a 2x2 contingency table.

Results and discussion

Abnormalities in HSPA2 distribution are present in Ppp1cc mutant germ cell nuclei

To determine if the loss of PPP1CC2 in the testis impaired HSPA2 localization during

spermatogenesis, we performed Immunolocalization studies on surface spread mouse germ cell nuclei.

CREST antibody was included in some experiments as it localizes to the kinetochores, which aids in

staging developing spermatocytes. In wild-type pachytene and diplotene spermatocytes, anti-HSPA2

signal localizes predominantly to the synaptonemal complex, but is also visible in a diffuse cloud

throughout the nucleus (Figure A.3.1A). This localization pattern has been observed previously by

others (Dix et al. 1996). In Ppp1cc mutants, the same cell types display a typically weak HSPA2 signal.

This observation is demonstrated in Figure A.3.1A in which the light intensity in the Ppp1cc mutant

specimen was enhanced to produce a visible signal (note the relative intensities of the CREST signal). In

Ppp1cc mutant spermatocytes HSPA2 was still able to localize to the synaptonemal complex, but

generally lacked or displayed a reduction of the punctate cloud throughout the nucleus (Figure A.3.1A,

arrows). As mentioned previously, deletion of HSPA2 results in a complete meiotic arrest in the

pachytene spermatocyte stage(Dix et al. 1996). Conversely, when Ppp1cc is deleted, germ cells can pass

this stage of development but do display phenotypic effects consistent with defects in meiosis(Varmuza

et al. 1999).

In wild-type elongating spermatid nuclei, HSPA2 displayed a cap-like distribution in the

subacrosomal region that has been previously described (Govin et al. 2006) (Figure A.3.1Bi). This

HSPA2 localization pattern was conserved in some Ppp1cc mutant elongating spermatid nuclei (Figure

A.3.1Bii), while others displayed abnormal localization patterns such as HSPA2 distributed throughout

Page 215: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

204

Figure A.3.1: The localization of HSPA2 in developing germ cells. (A) Surface-spread wild-type and

Ppp1cc null spermatocyte nuclei stained for CREST (green) and HSPA2 (red). Arrows indicate the

presence or absence of punctateHSPA2 staining. (B) Surface-spread wild type(i) and Ppp1cc null (ii, iii)

spermatid nuclei stained for DAPI (blue) and HSPA2 (red).

Page 216: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

205

Page 217: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

206

the nucleus (Figure A.3.1Biii). Generally morphologically normal Ppp1cc mutant elongating spermatid

nuclei displayed wild-type HSPA2 localization, while morphologically abnormal nuclei showed aberrant

localization. As was seen in spermatocytes, HSPA2 localization does not appear to be dependent on the

Ppp1cc gene. However both genes due appear to play a role in re-organization of chromatin in

elongating spermatids(Varmuza et al. 1999, Govin et al. 2006, Forgione et al. 2010)

While HSPA2 localization is impaired in Ppp1cc mutant spermatocytes and spermatids, it’s

localization does not appear to be totally dependent on this gene. This could potentially be due to the

presence of other PP1 isoforms PPP1CA and PPP1CB in the testis. Conversely the localization of

HSPA2 may not be dependent upon its phosphorylation status. While we have demonstrated that HSPA2

is hyperphosphorylated in Ppp1cc mutant testes, direct dephosphorylation of HSPA2 by PPP1CC2 has

not been demonstrated so hyperphosphorylation due to a third-party interaction also remains a

possibility.

The β-tubulin manchette structures are abnormal in Ppp1cc mutant spermatids

To determine if the loss of PPP1CC2 in the testis disrupted β-Tubulin localization in developing

spermatids, surface-spread spermatid nuclei were incubated with anti-β-Tubulin along with DAPI

counterstaining. Analysis of the β-Tubulin containing manchettes from Ppp1cc mutant spermatids

revealed prominent abnormalities when compared to wild-type spermatids and examples from the

existing literature (Table A.3.1). A conservative assessment of manchette abnormalities revealed that

54% of the manchettes examined from Ppp1cc mutant spermatids were abnormal, which is significantly

higher (p < 0.001) than 10% in wild-type spermatids (Figure A.3.2). The β-Tubulin staining pattern in

Ppp1cc mutant spermatid manchettes generally showed an abnormal morphology, and no examples of a

completely developed manchette were observed whereas they were common in wild-type spermatids

(Figure A.3.2iii). Typical abnormalities in Ppp1cc mutant manchettes included incomplete envelopment

of the spermatid nucleus in early elongating spermatids (Figure A.3.2iv), a reduced amount of β-Tubulin

staining (Figure A.3.2v), and mislocalization of the manchette around the spermatid nucleus (Figure

Page 218: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

207

A.3.2vi). Abnormalities in Ppp1cc mutant manchettes were accompanied by defects in nuclear

condensation in the developing spermatids (Figure A.3.2iv-vi).

The results of this experiment demonstrate prominent abnormalities in manchette morphogenesis

in Ppp1cc mutant spermatids. This suggests the possibility that misregulation of β-tubulin due to

hyperphosphorylation may be at least partly responsible for the nuclear shaping/condensation defects

found upon deletion of Ppp1cc. However to date, direct dephosphorylation of β-tubulin by PPP1CC2

has yet to be demonstrated, and will need to be confirmed in future experiments. Interestingly, PPP1CC2

has been shown to interact with another protein PPP1R42 that is associated with the manchette in

developing spermatids (Wang & Sperry 2011). The biological role of this interaction remains unknown,

but its existence provides further evidence towards a role of PPP1CC2 in manchette formation.

Page 219: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

208

Figure A.3.2: The loss of PPP1CC disrupts manchette formation in the mutant testis.

Immunohistochemistry performed on wild-type (i–iii) and Ppp1cc null (iv–vi) testis cell suspensions

using a b-tubulin antibody (red) and DAPI (blue). Wild-type manchettes initially envelop the nuclei of

elongating spermatids (i,ii) and later form normally developed manchette (iii). Ppp1cc null manchettes

do not properly envelop the nucleus (iv, v) and are poorly formed (vi).

Page 220: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

209

Page 221: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

210

Appendix A.4 Supplemental Material

Supplemental File A.4.1 Proteins identified via tandem affinity purification

Gene

symbol

Acession

Number

Unique/non

overlap

Spectra

above

threshold

matched

spectra

matched

peptides

#

matched

spectra

>95

Best

MASCOT

protein

score

%

Coverage

PPP1R7 Q3UM45 17 126 37 104 681 61.5

PPP1R8 Q8R3G1 8 20 11 10 110 37.9

PPP1R2 Q9DCL8 5 35 10 21 151 24.3

Wdr82 Q8BFQ4 4 8 5 4 68 16.3

DDOSt O54734 3 3 3 3 56 6.3

PPP1R10 Q80W00 3 5 3 3 43 6.1

PPP1R11 Q8K1L5 2 11 3 6 75 39.7

RRP1B Q91YK2 3 8 4 4 29 6.9

Atp2a2 O55143 2 3 3 2 85 3.8

AtP5C1 Q91VR2 2 2 2 2 41 7.4

LMtK2 Q3TYD6 2 13 5 6 48 4.6

ADAMtS12 Q811B3 1 1 1 1 28 0.6

APOPt1 Q9CQW7 1 1 1 1 17 5.8

ARID5A Q3U108 1 2 2 1 25 3.6

AtRX Q61687 1 1 1 1 23 0.5

BCL11B Q99PV8 1 3 2 2 23 3.6

BRWD1 Q8BPZ1 1 1 1 1 22 0.5

CCDC25 Q78PG9 1 42 2 8 34 10.6

CgN P59242 1 6 3 2 24 2.5

CLCF1 Q9QZM3 1 2 1 2 26 3.6

Page 222: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

211

CLIP1 Q922J3 1 1 1 1 21 0.6

CYC1 Q9D0M3 1 1 1 1 24 4.9

DENND5A Q6PAL8 1 2 2 1 20 1.8

SKIDA1 Q80YR3 1 1 1 1 23 1.3

DOCK7 Q7TPP0 1 2 1 2 24 5.1

EgLN1 Q91YE3 1 1 1 1 18 2.5

ELK4 P41158 1 1 1 1 20 2.1

FAM91A1 Q3UVG3 1 8 2 1 20 2.7

FANCL Q9CR14 1 1 1 1 25 2.7

FCHSD2 Q3USJ8 1 1 1 1 24 1.5

FtSJD1 Q8BWQ4 1 3 2 2 19 3.5

gAPDHS Q64467 1 3 2 1 77 5.2

gNB5 P62881 1 1 1 1 20 2.8

HADHA Q8BMS1 1 7 2 1 27 2.8

HES3 Q61657 1 1 1 1 23 5.1

HISt1H1A P43275 1 1 1 1 20 3.3

HISt1H2AF Q8CGP5 1 1 1 1 34 6.9

HKDC1 Q91W97 1 1 1 1 22 0.9

HMBS P22907 1 19 1 4 19 1.9

HOXD1 Q01822 1 2 1 1 25 4.9

HSD3B3 P26150 1 2 2 1 23 4.0

HSPB1 P14602 1 3 3 1 28 20.1

INtS7 Q7TQK1 1 1 1 1 17 1.5

KALRN A2CG49 1 2 2 1 19 0.7

KCNB2 A6H8H5 1 1 1 1 30 1.3

LBR Q3U9G9 1 1 1 1 30 1.8

Page 223: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

212

LCP2 Q60787 1 1 1 1 20 1.7

LRP1B Q9JI18 1 3 1 2 30 0.2

MAB21L1 O70299 1 2 2 1 22 7.2

MAgt1 Q9CQY5 1 1 1 1 26 2.4

MOSC2 Q922Q1 1 1 1 1 21 7.1

NDUFA4 Q62425 1 1 1 1 48 12.2

NEFH P19246 1 6 3 2 34 1.6

NES Q6P5H2 1 1 1 1 17 0.6

NOtCH2 O35516 1 1 1 1 17 0.7

PIK3CA P42337 1 4 2 3 45 1.9

PLOD3 Q9R0E1 1 1 1 1 20 0.9

PPP1r42 Q8R1Z4 1 1 1 1 25 2.0

PRR11 Q8BHE0 1 1 1 1 21 3.5

RAB10 P61027 1 3 3 2 53 16.5

RAB12 P35283 1 2 2 1 51 7.8

RAB14 Q91V41 1 2 2 1 51 8.8

RAB35 Q6PHN9 1 6 2 5 80 10.9

RAB8B P61028 1 2 2 1 49 12.1

RAD54B Q6PFE3 1 2 1 1 30 0.9

RPH3AL Q768S4 1 12 1 1 25 2.3

RPS29 P62274 1 1 1 1 23 14.3

SEMA3B Q62177 1 6 2 1 24 2.9

SLC29A1 Q9JIM1 1 1 1 1 43 2.6

Slit3 Q9WVB4 1 2 1 2 24 0.6

Sltm Q8CH25 1 5 1 1 20 1.1

Thsd7a Q69ZU6 1 1 1 1 21 0.4

Page 224: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

213

Tial1 P70318 1 2 2 1 21 5.4

Tnn Q80Z71 1 5 2 1 23 2.0

ttC39B Q8BYY4 1 1 1 1 26 1.9

WDR76 A6PWY4 1 3 2 2 17 3.1

Page 225: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

214

Supplemental file A.4.2 Peptides identified in LC-MS/MS of ~45KDa gel slice

Gene

Symbol Acession Bait Peptide Sequence

MASCOT

Ion

SCORE

Uqcrc2 Q9DB77

GST-

PPP1CC2 (K)AVAFQNSQTR(I) 43.51

(K)AVAQGNLSSADVQAAK(N) 80.66

(R)GIEAVGGK(L) 33.67

(R)GNNTTSLLSQSVAK(G) 74.08

(R)IIENLHDVAYK(N) 88.11

(K)ITSEELHYFVqNHFTSAR(M) 52.71

(R)LASSLTTK(G) 20

(K)NALANPLYcPDYR(M) 53.5

(R)RWEVAALR(S) 38.63

(K)TSAAPGGVPLQPQDLEFTK(L) 52.79

GST-

PPP1CC1 (K)AVAFQNSQTR(I) 37.96

(K)AVAFQNSQTR(I) 52.51

(K)AVAFQNSqTR(I) 28.87

(K)AVAQGNLSSADVQAAK(N) 80.03

(R)ENMAYTVEGIR(S) 62.92

(R)GNNTTSLLSQSVAK(G) 51.68

(R)IIENLHDVAYK(N) 35.2

(K)ITSEELHYFVqNHFTSAR(M) 51.33

(R)LASSLTTK(G) 35

(K)NALANPLYcPDYR(M) 51.7

(K)QVAEQFLNmR(G) 46.76

Page 226: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

215

(R)RWEVAALR(S) 29.13

(K)TSAAPGGVPLQPQDLEFTK(L) 64.68

(R)YEDSNNLGTSHLLR(L) 55.68

Sccpdh Q8R127

GST-

PPP1CC2 (R)EQIASEQSSR(L) 51.72

(K)GGGVFTPGAAFSR(T) 14.62

(K)GGGVFTPGAAFSR(T) 23.36

(K)LQQVLEK(A) 35.34

(K)LVLNcVGPYR(F) 61.15

(R)SVScLKPVPIVGTK(L) 15.51

(R)SVScLKPVPIVGTK(L) 64.18

GST-

PPP1CC1 (R)EQIASEQSSR(L) 28.82

(K)GGGVFTPGAAFSR(T) 23.23

(R)LPWAVAGR(S) 19.68

(K)LQQVLEK(A) 35.04

(K)LVLNcVGPYR(F) 51.33

(R)SVScLKPVPIVGTK(L) 57.47

(R)WPVSYcR(E) 14.71

Tssk1 Q61241

GST-

PPP1CC2 (K)HLTGEcK(D) 17.72

(K)HLTGEcK(D) 31.54

(K)HLTGEcK(D) 26.19

(R)SKHLTGEcK(D) 24.89

GST-

PPP1CC1 (K)APSDFLEK(F) 39.59

(R)DTEEGHPQQPSETHT(-) 53.41

(R)GLSSGAINK(E) 37.62

Page 227: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

216

(K)HLTGEcK(D) 46.45

(K)KAPSDFLEK(F) 45.33

(R)QSENVGLSSELNR(D) 71.08

(R)SESKPQEDTLQVVR(Q) 40.16

(R)SKHLTGEcK(D) 28.63

(K)TYEIFETSDGK(V) 40.56

(K)YcHDLDVVHR(D) 35.38

Fads2 Q9Z0R9

GST-

PPP1CC2 (K)HGIEYQEKPLLR(A) 22.43

GST-

PPP1CC2 (K)SIWNHVVHK(F) 33.45

Page 228: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

217

Supplemental file A.4.3 TSKS testis phosphopeptide site assignment data

TSKS testis phosphopeptides. ScaffoldPTM Protein Report

#20111006_Graham.sptm, Protein Report created on 12/06/2012

Peptide Sequence Variable Modifications

Localization

Probability Ascore Peptide Score Xtandem:Expect

Mascot:Identity

Threshold

AVsFHGVEPR S3 Phosphorylation 100% 1,000.00 61.98 2.523 38.22

HGLSPATPIQGcSGPPGsPEEPPR S18 Phosphorylation 100% 94.9 141.73 10.08 43.28

HGLSPATPIQGcSGPPGsPEEPPR S18 Phosphorylation 90% 18.8 78.41 2.921 43.22

Page 229: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

218

Supplemental file A.4.4 TSKS testis phosphopeptide site assignment charge tables/annotated

spectra

TSKS pS281 Peptide 1

Page 230: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

219

TSKS pS281 Peptide 2

Page 231: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

220

TSKS pS54

Page 232: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

221

Supplemental File A.4.5 3 week testis phosphoproteins of interest

Known and predicted PP1

interacting proteins

Proteins encoded by

genes essential for

male fertility

Proteins with GO Biological Process

"Reproduction" annotation

Proteins identified

in SBP-3XFLAG-

PPP1CC2 tandem

affinity purification

TNS1 ESPL1 AGFG1 DENND5A

MAP1B ACE UBAP2L PPP1R7

HDAC6 DDX25 NIPBL SLTM

SIRT2 RBMXL2 SYCP2 LBR

RB1 SIN3A MAP3K4 PPP1R10

PPP1R7 PRKAR1A AHSG HSPB1

GSK3A SH2B1 EIF2S2 CGN

MKI67 DDX4 MAPK8IP2 DOCK7

MAX CLGN ACSBG1 CCDC25

DDX10 SIRT1 CTNNA1 PPP1R2

PPP1R10 CSTF2T EIF2B5 HIST1H1A

SRSF10 EIF4G3

AMHR

STT3B (VIA

ASSOCIATION

WITH DDOST)

RBM26 SYCP3 SPAG1

WNK1 MYBL1 DDX25

EIF2S2 DMRT1 CLGN

TPRN HMGA1 CSDA

POLR2A CLDN11 H2AFX

PPP1R2 YBX2 XRN2

GNL3 TMF1 DDX4

TRA2B CSDA TDRD1

CD2BP2 RAD18 HORMAD1

Page 233: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

222

SLC9A1 HSPA4 HIST1H1T

PHACTR4 AMHR2 BAG6

MRE11A MTMR2 DNMT3A

POLE PANK2 ACE

PHRF1 MYH10 CLDN11

PPP1R3D ADAM2 HIST1H1A

TRIM28 HSP90AA1 SPE39

HSPA4 H2AFX BOLL

MARK2 DMRT1

TDRD1 TYRO3

NEK1 MTL5

AGFG1 NEDD4

CCNE2

ATP2B4

GJA1

TEX14

HSPA4L

UBE3A

SYCP2

Page 234: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

223

Supplemental File A.4.6 DAVID GO enrichment analysis of 3 week testis Phosphoproteins

Term Count PValue

Fold

Enrichment

GO:0003723~RNA binding 99 1.4E-29 3.6

GO:0006397~mRNA processing 60 1.9E-27 5.5

GO:0016071~mRNA metabolic process 64 2.8E-27 5.1

GO:0043228~non-membrane-bounded organelle 153 6.3E-24 2.2

GO:0043232~intracellular non-membrane-bounded organelle 153 6.3E-24 2.2

GO:0006396~RNA processing 71 4.5E-23 3.9

GO:0031981~nuclear lumen 95 7.4E-23 3.0

GO:0005694~chromosome 60 1.9E-22 4.5

GO:0008380~RNA splicing 46 4.8E-21 5.5

GO:0030529~ribonucleoprotein complex 62 2.0E-19 3.8

GO:0044427~chromosomal part 51 2.8E-19 4.5

GO:0000166~nucleotide binding 172 3.4E-19 1.9

GO:0043233~organelle lumen 99 2.5E-17 2.4

GO:0031974~membrane-enclosed lumen 101 2.6E-17 2.4

GO:0070013~intracellular organelle lumen 98 6.2E-17 2.4

GO:0005681~spliceosome 29 2.6E-15 6.6

GO:0051276~chromosome organization 56 5.0E-15 3.3

GO:0000785~chromatin 32 1.8E-14 5.4

GO:0006325~chromatin organization 47 7.4E-14 3.6

GO:0005654~nucleoplasm 61 2.3E-13 2.9

GO:0016607~nuclear speck 23 4.6E-13 7.2

GO:0003677~DNA binding 132 3.9E-12 1.8

GO:0016604~nuclear body 27 4.5E-12 5.3

Page 235: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

224

GO:0007049~cell cycle 63 5.3E-11 2.5

GO:0000792~heterochromatin 16 1.4E-10 8.8

GO:0032559~adenyl ribonucleotide binding 110 1.8E-10 1.8

GO:0005730~nucleolus 37 3.6E-10 3.3

GO:0005524~ATP binding 107 8.7E-10 1.8

GO:0030554~adenyl nucleotide binding 111 1.6E-09 1.8

GO:0022402~cell cycle process 45 2.0E-09 2.7

GO:0001883~purine nucleoside binding 111 2.6E-09 1.8

GO:0016568~chromatin modification 33 3.4E-09 3.4

GO:0001882~nucleoside binding 111 3.8E-09 1.7

GO:0044451~nucleoplasm part 47 6.6E-09 2.6

GO:0000279~M phase 36 7.3E-09 3.1

GO:0000228~nuclear chromosome 21 8.9E-09 4.9

GO:0006350~transcription 122 1.2E-08 1.7

GO:0000398~nuclear mRNA splicing, via spliceosome 13 1.7E-08 8.4

GO:0000375~RNA splicing, via transesterification reactions 13 1.7E-08 8.4

GO:0000377~RNA splicing, via transesterification reactions with

bulged adenosine as nucleophile 13 1.7E-08 8.4

GO:0003682~chromatin binding 25 1.7E-08 3.9

GO:0032553~ribonucleotide binding 120 3.1E-08 1.6

GO:0032555~purine ribonucleotide binding 120 3.1E-08 1.6

GO:0022403~cell cycle phase 38 3.3E-08 2.8

GO:0045449~regulation of transcription 142 6.7E-08 1.5

GO:0006333~chromatin assembly or disassembly 20 1.0E-07 4.4

GO:0044454~nuclear chromosome part 18 1.2E-07 4.9

GO:0017076~purine nucleotide binding 121 1.6E-07 1.6

Page 236: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

225

GO:0000793~condensed chromosome 18 2.1E-07 4.7

GO:0051301~cell division 33 2.3E-07 2.8

GO:0008134~transcription factor binding 33 2.7E-07 2.8

GO:0016569~covalent chromatin modification 17 2.0E-06 4.3

GO:0006259~DNA metabolic process 40 2.4E-06 2.3

GO:0051321~meiotic cell cycle 16 4.1E-06 4.3

GO:0004386~helicase activity 19 4.9E-06 3.6

GO:0004674~protein serine/threonine kinase activity 38 9.8E-06 2.2

GO:0051327~M phase of meiotic cell cycle 15 1.5E-05 4.1

GO:0007126~meiosis 15 1.5E-05 4.1

GO:0016567~protein ubiquitination 13 1.5E-05 4.7

GO:0006323~DNA packaging 16 1.7E-05 3.8

GO:0000790~nuclear chromatin 11 2.0E-05 5.6

GO:0032446~protein modification by small protein conjugation 14 2.0E-05 4.3

GO:0006511~ubiquitin-dependent protein catabolic process 19 2.2E-05 3.2

GO:0005720~nuclear heterochromatin 9 2.4E-05 7.2

GO:0016570~histone modification 15 2.5E-05 3.9

GO:0006468~protein amino acid phosphorylation 50 2.6E-05 1.9

GO:0040029~regulation of gene expression, epigenetic 13 2.8E-05 4.5

GO:0000278~mitotic cell cycle 26 3.0E-05 2.6

GO:0034621~cellular macromolecular complex subunit

organization 26 3.2E-05 2.5

GO:0000280~nuclear division 22 4.3E-05 2.8

GO:0007067~mitosis 22 4.3E-05 2.8

GO:0016585~chromatin remodeling complex 10 5.8E-05 5.6

GO:0000087~M phase of mitotic cell cycle 22 5.8E-05 2.7

Page 237: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

226

GO:0001739~sex chromatin 7 6.8E-05 9.3

GO:0048285~organelle fission 22 7.3E-05 2.7

GO:0010605~negative regulation of macromolecule metabolic

process 41 7.3E-05 1.9

GO:0006476~protein amino acid deacetylation 7 8.6E-05 8.8

GO:0008092~cytoskeletal protein binding 35 8.7E-05 2.1

GO:0000803~sex chromosome 7 9.0E-05 8.9

GO:0005856~cytoskeleton 65 9.7E-05 1.6

GO:0070647~protein modification by small protein conjugation or

removal 15 9.9E-05 3.5

GO:0016458~gene silencing 11 1.1E-04 4.6

GO:0051603~proteolysis involved in cellular protein catabolic

process 42 1.1E-04 1.9

GO:0004672~protein kinase activity 44 1.3E-04 1.8

GO:0044257~cellular protein catabolic process 42 1.3E-04 1.9

GO:0044265~cellular macromolecule catabolic process 46 1.3E-04 1.8

GO:0003779~actin binding 27 1.3E-04 2.3

GO:0010608~posttranscriptional regulation of gene expression 18 1.4E-04 2.9

GO:0019941~modification-dependent protein catabolic process 40 1.7E-04 1.9

GO:0043632~modification-dependent macromolecule catabolic

process 40 1.7E-04 1.9

GO:0033554~cellular response to stress 34 1.7E-04 2.0

GO:0008135~translation factor activity, nucleic acid binding 14 1.7E-04 3.5

GO:0030695~GTPase regulator activity 31 1.8E-04 2.1

GO:0016310~phosphorylation 51 2.3E-04 1.7

GO:0060589~nucleoside-triphosphatase regulator activity 31 2.4E-04 2.1

GO:0006417~regulation of translation 14 2.5E-04 3.4

Page 238: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

227

GO:0031497~chromatin assembly 12 2.5E-04 3.8

GO:0030163~protein catabolic process 42 2.6E-04 1.8

GO:0034622~cellular macromolecular complex assembly 22 2.8E-04 2.4

GO:0034728~nucleosome organization 12 2.9E-04 3.8

GO:0065004~protein-DNA complex assembly 12 2.9E-04 3.8

GO:0034399~nuclear periphery 9 3.0E-04 5.1

GO:0003729~mRNA binding 10 3.1E-04 4.5

GO:0005096~GTPase activator activity 20 3.3E-04 2.5

GO:0006974~response to DNA damage stimulus 26 4.0E-04 2.2

GO:0003712~transcription cofactor activity 19 4.2E-04 2.6

GO:0001741~XY body 5 4.3E-04 12.7

GO:0006270~DNA replication initiation 5 5.1E-04 12.0

GO:0016887~ATPase activity 25 5.3E-04 2.2

GO:0000775~chromosome, centromeric region 13 5.8E-04 3.3

GO:0004221~ubiquitin thiolesterase activity 11 5.8E-04 3.8

GO:0009057~macromolecule catabolic process 46 6.1E-04 1.7

GO:0031252~cell leading edge 13 6.3E-04 3.3

GO:0000794~condensed nuclear chromosome 8 6.3E-04 5.3

GO:0016363~nuclear matrix 8 6.3E-04 5.3

GO:0043933~macromolecular complex subunit organization 30 7.1E-04 2.0

GO:0042162~telomeric DNA binding 5 7.2E-04 11.1

GO:0015630~microtubule cytoskeleton 31 7.2E-04 1.9

GO:0070035~purine NTP-dependent helicase activity 12 7.3E-04 3.4

GO:0008026~ATP-dependent helicase activity 12 7.3E-04 3.4

GO:0051015~actin filament binding 9 7.6E-04 4.5

Page 239: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

228

GO:0016575~histone deacetylation 5 7.7E-04 10.9

GO:0051493~regulation of cytoskeleton organization 13 8.1E-04 3.2

GO:0006334~nucleosome assembly 11 8.4E-04 3.6

GO:0003743~translation initiation factor activity 10 8.7E-04 3.9

GO:0006281~DNA repair 21 9.7E-04 2.3

GO:0048232~male gamete generation 23 9.9E-04 2.2

GO:0007283~spermatogenesis 23 9.9E-04 2.2

GO:0006793~phosphorus metabolic process 56 1.1E-03 1.6

GO:0006796~phosphate metabolic process 56 1.1E-03 1.6

GO:0006342~chromatin silencing 6 1.1E-03 7.2

GO:0048471~perinuclear region of cytoplasm 17 1.2E-03 2.5

GO:0045934~negative regulation of nucleobase, nucleoside,

nucleotide and nucleic acid metabolic process 31 1.2E-03 1.9

GO:0000151~ubiquitin ligase complex 9 1.2E-03 4.2

GO:0031202~RNA splicing factor activity, transesterification

mechanism 4 1.2E-03 16.3

GO:0005911~cell-cell junction 16 1.3E-03 2.6

GO:0010558~negative regulation of macromolecule biosynthetic

process 32 1.3E-03 1.8

GO:0008047~enzyme activator activity 22 1.4E-03 2.2

GO:0051172~negative regulation of nitrogen compound metabolic

process 31 1.4E-03 1.9

GO:0032259~methylation 11 1.6E-03 3.3

GO:0030027~lamellipodium 9 1.7E-03 4.0

GO:0022613~ribonucleoprotein complex biogenesis 15 1.7E-03 2.6

GO:0005938~cell cortex 13 1.8E-03 2.9

GO:0045814~negative regulation of gene expression, epigenetic 6 1.8E-03 6.5

Page 240: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

229

GO:0043244~regulation of protein complex disassembly 8 1.8E-03 4.5

GO:0065003~macromolecular complex assembly 27 1.9E-03 1.9

GO:0010629~negative regulation of gene expression 31 1.9E-03 1.8

GO:0031327~negative regulation of cellular biosynthetic process 32 2.1E-03 1.8

GO:0003730~mRNA 3'-UTR binding 4 2.1E-03 14.0

GO:0046907~intracellular transport 32 2.1E-03 1.8

GO:0006306~DNA methylation 6 2.2E-03 6.3

GO:0006305~DNA alkylation 6 2.2E-03 6.3

GO:0019904~protein domain specific binding 18 2.3E-03 2.3

GO:0043021~ribonucleoprotein binding 7 2.3E-03 5.0

GO:0009890~negative regulation of biosynthetic process 32 2.4E-03 1.8

GO:0005516~calmodulin binding 13 2.4E-03 2.8

GO:0007010~cytoskeleton organization 26 2.4E-03 1.9

GO:0022618~ribonucleoprotein complex assembly 7 2.5E-03 4.9

GO:0003713~transcription coactivator activity 12 2.5E-03 2.9

GO:0016564~transcription repressor activity 19 2.6E-03 2.2

GO:0043414~biopolymer methylation 10 2.6E-03 3.4

GO:0019887~protein kinase regulator activity 9 2.6E-03 3.7

GO:0006304~DNA modification 6 2.7E-03 6.0

GO:0019899~enzyme binding 20 2.7E-03 2.1

GO:0030029~actin filament-based process 17 2.8E-03 2.3

GO:0000245~spliceosome assembly 5 2.8E-03 8.0

GO:0033558~protein deacetylase activity 5 3.4E-03 7.6

GO:0004407~histone deacetylase activity 5 3.4E-03 7.6

GO:0051494~negative regulation of cytoskeleton organization 8 3.5E-03 4.0

Page 241: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

230

GO:0031111~negative regulation of microtubule polymerization or

depolymerization 5 3.6E-03 7.5

GO:0030036~actin cytoskeleton organization 16 3.7E-03 2.3

GO:0001701~in utero embryonic development 22 3.9E-03 2.0

GO:0009628~response to abiotic stimulus 21 4.1E-03 2.0

GO:0070161~anchoring junction 12 4.4E-03 2.7

GO:0005913~cell-cell adherens junction 6 4.4E-03 5.4

GO:0005912~adherens junction 11 4.5E-03 2.9

GO:0007059~chromosome segregation 9 4.8E-03 3.4

GO:0030911~TPR domain binding 3 4.9E-03 24.4

GO:0000795~synaptonemal complex 5 4.9E-03 7.0

GO:0033043~regulation of organelle organization 15 4.9E-03 2.3

GO:0016574~histone ubiquitination 4 5.0E-03 10.7

GO:0050684~regulation of mRNA processing 4 5.0E-03 10.7

GO:0016584~nucleosome positioning 3 5.0E-03 24.0

GO:0019787~small conjugating protein ligase activity 13 5.1E-03 2.5

GO:0051253~negative regulation of RNA metabolic process 24 5.4E-03 1.9

GO:0030054~cell junction 29 5.4E-03 1.7

GO:0032318~regulation of Ras GTPase activity 10 5.8E-03 3.0

GO:0000776~kinetochore 8 6.3E-03 3.6

GO:0000786~nucleosome 8 6.3E-03 3.6

GO:0006913~nucleocytoplasmic transport 11 6.5E-03 2.8

GO:0019213~deacetylase activity 5 6.6E-03 6.4

GO:0016481~negative regulation of transcription 27 6.9E-03 1.7

GO:0043484~regulation of RNA splicing 4 6.9E-03 9.6

GO:0043073~germ cell nucleus 5 7.0E-03 6.4

Page 242: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

231

GO:0009898~internal side of plasma membrane 20 7.0E-03 2.0

GO:0051169~nuclear transport 11 7.5E-03 2.7

GO:0051082~unfolded protein binding 9 7.5E-03 3.1

GO:0005852~eukaryotic translation initiation factor 3 complex 4 7.7E-03 9.3

GO:0016881~acid-amino acid ligase activity 15 8.2E-03 2.2

GO:0005768~endosome 18 8.8E-03 2.0

GO:0006730~one-carbon metabolic process 12 9.4E-03 2.5

GO:0019207~kinase regulator activity 9 9.6E-03 3.0

GO:0043009~chordate embryonic development 29 9.8E-03 1.7

GO:0004842~ubiquitin-protein ligase activity 11 1.0E-02 2.6

GO:0003697~single-stranded DNA binding 6 1.0E-02 4.4

GO:0006260~DNA replication 14 1.1E-02 2.2

GO:0009792~embryonic development ending in birth or egg

hatching 29 1.1E-02 1.6

GO:0003774~motor activity 13 1.1E-02 2.3

GO:0044430~cytoskeletal part 41 1.1E-02 1.5

GO:0003725~double-stranded RNA binding 6 1.2E-02 4.3

GO:0000791~euchromatin 3 1.2E-02 16.8

GO:0031532~actin cytoskeleton reorganization 4 1.2E-02 8.0

GO:0017148~negative regulation of translation 5 1.2E-02 5.5

GO:0016323~basolateral plasma membrane 12 1.2E-02 2.4

GO:0005874~microtubule 17 1.2E-02 2.0

GO:0043566~structure-specific DNA binding 9 1.2E-02 2.9

GO:0051656~establishment of organelle localization 6 1.3E-02 4.2

GO:0016790~thiolester hydrolase activity 11 1.3E-02 2.5

GO:0016879~ligase activity, forming carbon-nitrogen bonds 16 1.3E-02 2.0

Page 243: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

232

GO:0032268~regulation of cellular protein metabolic process 21 1.3E-02 1.8

GO:0010639~negative regulation of organelle organization 8 1.4E-02 3.1

GO:0043242~negative regulation of protein complex disassembly 6 1.4E-02 4.1

GO:0015629~actin cytoskeleton 15 1.5E-02 2.1

GO:0043087~regulation of GTPase activity 10 1.5E-02 2.6

GO:0051129~negative regulation of cellular component

organization 10 1.5E-02 2.6

GO:0042393~histone binding 5 1.5E-02 5.1

GO:0070507~regulation of microtubule cytoskeleton organization 6 1.6E-02 4.0

GO:0005099~Ras GTPase activator activity 8 1.6E-02 3.0

GO:0032993~protein-DNA complex 8 1.7E-02 3.0

GO:0001939~female pronucleus 3 1.7E-02 14.0

GO:0030528~transcription regulator activity 65 1.8E-02 1.3

GO:0019898~extrinsic to membrane 27 1.8E-02 1.6

GO:0045892~negative regulation of transcription, DNA-

dependent 22 1.8E-02 1.7

GO:0031114~regulation of microtubule depolymerization 4 1.8E-02 6.9

GO:0007026~negative regulation of microtubule depolymerization 4 1.8E-02 6.9

GO:0019953~sexual reproduction 26 1.9E-02 1.6

GO:0050657~nucleic acid transport 8 1.9E-02 2.9

GO:0051236~establishment of RNA localization 8 1.9E-02 2.9

GO:0050658~RNA transport 8 1.9E-02 2.9

GO:0007548~sex differentiation 12 2.0E-02 2.2

GO:0016563~transcription activator activity 19 2.0E-02 1.8

GO:0007276~gamete generation 23 2.1E-02 1.7

GO:0042623~ATPase activity, coupled 17 2.1E-02 1.9

Page 244: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

233

GO:0005083~small GTPase regulator activity 17 2.1E-02 1.9

GO:0006403~RNA localization 8 2.1E-02 2.9

GO:0030898~actin-dependent ATPase activity 3 2.2E-02 12.2

GO:0070567~cytidylyltransferase activity 3 2.2E-02 12.2

GO:0003724~RNA helicase activity 3 2.2E-02 12.2

GO:0048024~regulation of nuclear mRNA splicing, via

spliceosome 3 2.3E-02 12.0

GO:0005721~centromeric heterochromatin 3 2.4E-02 12.0

GO:0000800~lateral element 3 2.4E-02 12.0

GO:0051640~organelle localization 7 2.4E-02 3.1

GO:0051168~nuclear export 5 2.4E-02 4.4

GO:0045120~pronucleus 4 2.5E-02 6.2

GO:0001673~male germ cell nucleus 4 2.5E-02 6.2

GO:0050839~cell adhesion molecule binding 4 2.6E-02 6.1

GO:0000118~histone deacetylase complex 5 2.6E-02 4.4

GO:0003006~reproductive developmental process 19 2.7E-02 1.7

GO:0031110~regulation of microtubule polymerization or

depolymerization 5 2.7E-02 4.3

GO:0007017~microtubule-based process 16 3.0E-02 1.8

GO:0005952~cAMP-dependent protein kinase complex 3 3.1E-02 10.5

GO:0043624~cellular protein complex disassembly 4 3.1E-02 5.6

GO:0043241~protein complex disassembly 4 3.1E-02 5.6

GO:0000209~protein polyubiquitination 4 3.1E-02 5.6

GO:0016079~synaptic vesicle exocytosis 4 3.1E-02 5.6

GO:0045947~negative regulation of translational initiation 3 3.2E-02 10.3

GO:0015931~nucleobase, nucleoside, nucleotide and nucleic acid

transport 8 3.4E-02 2.6

Page 245: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

234

GO:0048609~reproductive process in a multicellular organism 26 3.6E-02 1.5

GO:0032504~multicellular organism reproduction 26 3.6E-02 1.5

GO:0009314~response to radiation 12 3.6E-02 2.0

GO:0005829~cytosol 29 3.6E-02 1.5

GO:0042995~cell projection 30 3.7E-02 1.5

GO:0032886~regulation of microtubule-based process 6 3.8E-02 3.2

GO:0007163~establishment or maintenance of cell polarity 5 3.8E-02 3.9

GO:0000777~condensed chromosome kinetochore 6 4.0E-02 3.2

GO:0031369~translation initiation factor binding 3 4.0E-02 9.2

GO:0001839~neural plate morphogenesis 3 4.1E-02 9.0

GO:0008360~regulation of cell shape 6 4.5E-02 3.1

GO:0031988~membrane-bounded vesicle 23 4.6E-02 1.5

GO:0016208~AMP binding 4 4.6E-02 4.9

GO:0001725~stress fiber 4 4.7E-02 4.9

GO:0016581~NuRD complex 3 4.7E-02 8.4

GO:0031982~vesicle 27 4.9E-02 1.5

Page 246: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

235

Supplemental File A.4.7 Peptides identified via preliminary semi-quantitative analysis as more

abundant in Ppp1cc knockout testes.

Gene Name Method Gene Name Method

Abcf1 Spectral Counting Fip1l1 Spectral Counting

Acin1 MS/MS XIC, Spectral Counting Fn1 Spectral Counting

Acss2 Spectral Counting Fxr2 Spectral Counting

Adam2 Spectral Counting Fyttd1 Spectral Counting

Add1 MS/MS XIC Gapvd1 MS/MS XIC

Aebp2 Spectral Counting Gatad2a Spectral Counting

Agfg1 Spectral Counting Gbf1 Spectral Counting

Ahsg Spectral Counting Gm14569 MS/MS XIC

Akap12 MS/MS XIC Gtpbp1 Spectral Counting

Api5 Spectral Counting Hdac1 Spectral Counting

Arhgap12 Spectral Counting Hdac2 Spectral Counting

Arhgef11 Spectral Counting Hdgf MS/MS XIC

Basp1 MS/MS XIC Hectd1 MS/MS XIC

Baz1b Spectral Counting Hist1h1b Spectral Counting

Bclaf1 Spectral Counting Hmga1 Spectral Counting

Bysl Spectral Counting Hmgn1 MS/MS XIC

Canx Spectral Counting Hnrnpu MS/MS XIC, Spectral Counting

Ccdc6 Spectral Counting Hsf5 Spectral Counting

Ccne2 Spectral Counting Hspa4 Spectral Counting

Ccnk Spectral Counting Htatsf1 Spectral Counting

Cdk12 Spectral Counting Igfn1 Spectral Counting

Cenpv Spectral Counting Ivns1abp Spectral Counting

Clgn Spectral Counting Jcad Spectral Counting

Cnot2 Spectral Counting Kat7 Spectral Counting

Csda Spectral Counting Kdm1a Spectral Counting

Cttn Spectral Counting Kri1 Spectral Counting

D11Wsu99e Spectral Counting Larp1 MS/MS XIC, Spectral Counting

Dcaf6 Spectral Counting Larp7 Spectral Counting

Dgkq Spectral Counting Lsm14a MS/MS XIC

Dhx9 Spectral Counting Luc7l3 Spectral Counting

Dido1 Spectral Counting Mageb10-ps Spectral Counting

Dstn Spectral Counting Mageb4 Spectral Counting

Eif3b Spectral Counting Map1b Spectral Counting

Eif4b Spectral Counting Map4k4 Spectral Counting

Eif4ebp2 Spectral Counting Max Spectral Counting

Eif4g3 MS/MS XIC Mdc1 Spectral Counting

Ensa Spectral Counting Mdn1 Spectral Counting

Enthd1 MS/MS XIC, Spectral Counting Mettl16 Spectral Counting

Esf1 Spectral Counting Mfap1 Spectral Counting

Espl1 MS/MS XIC, Spectral Counting Myh11 Spectral Counting

Espn Spectral Counting Naa30 Spectral Counting

Evl MS/MS XIC Ncl Spectral Counting

Fam122a Spectral Counting Nfatc2ip Spectral Counting

Fam134a Spectral Counting Nipbl Spectral Counting

Page 247: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

236

Fam54b Spectral Counting Nkapl Spectral Counting

Fcho2 MS/MS XIC Nmd3 Spectral Counting

Page 248: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

237

Gene Name Method Gene Name Method

Nol8 Spectral Counting Srsf11 Spectral Counting

Nop56 MS/MS XIC Ssr1 Spectral Counting

Nop58 Spectral Counting Supt6h Spectral Counting

Npm1 MS/MS XIC Sycp2 MS/MS XIC, Spectral Counting

Nvl Spectral Counting Sycp3 Spectral Counting

Pcm1 Spectral Counting Taok3 Spectral Counting

Pcyt1a Spectral Counting Tbc1d4 MS/MS XIC

Pdap1 Spectral Counting Tcp11l2 Spectral Counting

Phf8 MS/MS XIC Tdrd1 Spectral Counting

Ppil4 MS/MS XIC Thrap3 Spectral Counting

Prpf31 Spectral Counting Thumpd1 Spectral Counting

Prpf38a MS/MS XIC Tjp1 Spectral Counting

Psip1 Spectral Counting Tnik Spectral Counting

Ptges3 Spectral Counting Top2b Spectral Counting

Ptpdc1 MS/MS XIC Tp53bp1 Spectral Counting

Pvr Spectral Counting Trappc12 Spectral Counting

Rab11fip1 Spectral Counting Trip12 Spectral Counting

Rad18 Spectral Counting Trmt10a MS/MS XIC

Ralgps2 Spectral Counting Trp53bp1 Spectral Counting

Raly MS/MS XIC, Spectral Counting Ttc15 Spectral Counting

Ranbp2 Spectral Counting Uba2 MS/MS XIC

Ranbp3 MS/MS XIC Ubap2l Spectral Counting

Rbm14 MS/MS XIC Unc119 Spectral Counting

Rcor3 Spectral Counting Usp9x Spectral Counting

Rhox8 Spectral Counting Vcp MS/MS XIC

Rnf20 MS/MS XIC Xrn2 Spectral Counting

Saal1 Spectral Counting Zc3h13 Spectral Counting

Scaf1 Spectral Counting Zc3hc1 Spectral Counting

Scaf8 Spectral Counting Zfp91 Spectral Counting

Scrib Spectral Counting Znf830 Spectral Counting

Sec31a Spectral Counting Setx Spectral Counting Sf1 Spectral Counting Sf3b1 Spectral Counting Sfr1 Spectral Counting Sgk3 Spectral Counting Sltm Spectral Counting Slx4 MS/MS XIC Smarca5 Spectral Counting Smarcc1 Spectral Counting Smarcc2 Spectral Counting Spice1 Spectral Counting Srp72 MS/MS XIC

Page 249: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

238

Supplemental File A.4.8- Quantitative comparison of XIC peak areas of Ppp1cc

hyperphosphorylated peptides assigned to indicated proteins

Page 250: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

239

Appendix Reference List

Bollen M, Peti W, Ragusa MJ & Beullens M 2010 The extended PP1 toolkit: designed to create

specificity. Trends in biochemical sciences 35 450-458.

Cherry L &Hsu TC 1984 Antitubulin immunofluorescence studies of spermatogenesis in the mouse.

Chromosoma 90 265-274.

Dix DJ, Allen JW, Collins BW, Mori C, Nakamura N, Poorman-Allen P, Goulding EH & Eddy EM

1996 Targeted gene disruption of Hsp70-2 results in failed meiosis, germ cell apoptosis, and male

infertility. Proceedings of the National Academy of Sciences 93 3264-3268.

Dobson MJ, Pearlman RE, Karaiskakis A, Spyropoulos B & Moens PB 1994 Synaptonemal complex

proteins: occurrence, epitope mapping and chromosome disjunction. Journal of cell science 107 2749-

2760.

Fields S &Song O 1989 A novel genetic system to detect protein–protein interactions. Nature 340 245-

246.

Forgione N, Vogl AW & Varmuza S 2010 Loss of protein phosphatase 1c{gamma} (PPP1CC) leads to

impaired spermatogenesis associated with defects in chromatin condensation and acrosome development:

an ultrastructural analysis. Reproduction 139 1021-1029.

Govin J, Caron C, Escoffier E, Ferro M, Kuhn L, Rousseaux S, Eddy EM, Garin J & Khochbin S

2006 Post-meiotic Shifts in HSPA2/HSP70.2 Chaperone Activity during Mouse Spermatogenesis.

Journal of Biological Chemistry 281 37888-37892.

Hrabchak C &Varmuza S 2004 Identification of the spermatogenic zip protein Spz1 as a putative

protein phosphatase-1 (PP1) regulatory protein that specifically binds the PP1cgamma2 splice variant in

the mouse testis. Journal of Biological Chemistry 279 37079-37086.

Hrabchak C, Henderson H & Varmuza S 2007 A Testis Specific Isoform of Endophilin B1,

Endophilin B1t, Interacts Specifically with Protein Phosphatase-1cgamma2 in Mouse Testis and Is

Abnormally Expressed in PP1cgamma Null Mice. Biochemistry 46 4635-4644.

Huang Z &Vijayaraghavan S 2004 Increased Phosphorylation of a Distinct Subcellular Pool of Protein

Phosphatase, PP1γ2, During Epididymal Sperm Maturation. Biology of reproduction 70 439-447.

Kierszenbaum AL 2002 Intramanchette transport (IMT): Managing the making of the spermatid head,

centrosome, and tail. Molecular reproduction and development 63 1-4.

Kuzmin A, Jarvi K, Lo K, Spencer L, Chow GYC, Macleod G, Wang Q & Varmuza S 2009

Identification of Potentially Damaging Amino Acid Substitutions Leading to Human Male Infertility.

Biology of reproduction 81 319-326.

Page 251: Identification of PPP1CC2 Interacting Proteins in the ... · Deletion of the PP1 serine/threonine phosphatase gene Ppp1cc in mice results in male infertility due to a severe impairment

240

Mann M, Latham K, E. & Varmuza S 1995 Identification of Genes Showing Altered Expression in

Preimplantation and Early Postimplantation Parthenogenetic Embryos. Developmental Genetics 17 223-

232.

Matzuk MM &Lamb DJ 2008 The biology of infertility: research advances and clinical challenges.

Nature medicine 14 1197-1213.

Moens PB, Heyting C, Dietrich AJ, van Raamsdonk W & Chen Q 1987 Synaptonemal complex

antigen location and conservation. The Journal of cell biology 105 93-103.

Moens P &Earnshaw W 1989 Anti-topoisomerase II recognizes meiotic chromosome cores.

Chromosoma 98 317-322.

Russell LD, Ettlin RA, Sinha Hikim AP & Clegg EG 1990 Histological and Histopathological

Evaluation of the Testis. 286.

Takahashi Y, Meyerkord CL & Wang H 2009 Bif-1/Endophilin B1: a candidate for crescent driving

force in autophagy. Cell death and differentiation 16 947-955.

Takahashi Y, Coppola D, Matsushita N, Cualing HD, Sun M, Sato Y, Liang C, Jung JU, Cheng JQ,

Mul JJ, Pledger WJ & Wang H 2007 Bif-1 interacts with Beclin 1 through UVRAG and regulates

autophagy and tumorigenesis. Nature cell biology 9 1142-1151.

Tirode F, Malaguti C, Romero F, Attar R, Camonis J & Egly JM 1997 A Conditionally Expressed

Third Partner Stabilizes or Prevents the Formation of a Transcriptional Activator in a Three-hybrid

System. Journal of Biological Chemistry 272 22995-22999.

Varmuza S, Jurisicova A, Okano K, Hudson J, Boekelheide K & Shipp EB 1999 Spermiogenesis Is

Impaired in Mice Bearing a Targeted Mutation in the Protein Phosphatase 1cγ Gene. Developmental

biology 205 98-110.

Varmuza S &Ling L 2003 Increased recombination frequency showing evidence of loss of interference

is associated with abnormal testicular histopathology. Molecular reproduction and development 64 499-

506.

Wang R &Sperry AO 2011 PP1 Forms an Active Complex with TLRR (lrrc67), a Putative PP1

Regulatory Subunit, during the Early Stages of Spermiogenesis in Mice. PLoS ONE 6 e21767.