225
CHARACTERIZATION OF SLP-2 FUNCTION IN T LYMPHOCYTES (Spine title: Characterization of SLP-2 function in T lymphocytes) (Thesis format: Integrated Article) by Darah A. Christie Graduate Program in Microbiology and Immunology A thesis submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy The School of Graduate and Postdoctoral Studies The University of Western Ontario London, Ontario, Canada © Darah Christie 2012

Characterization of SLP-2 function in T lymphocytes

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

CHARACTERIZATION OF SLP-2 FUNCTION IN T LYMPHOCYTES

(Spine title: Characterization of SLP-2 function in T lymphocytes)

(Thesis format: Integrated Article)

by

Darah A. Christie

Graduate Program in Microbiology and Immunology

A thesis submitted in partial fulfillment of the requirements for the degree of

Doctor of Philosophy

The School of Graduate and Postdoctoral Studies The University of Western Ontario

London, Ontario, Canada

© Darah Christie 2012

ii

THE UNIVERSITY OF WESTERN ONTARIO School of Graduate and Postdoctoral Studies

CERTIFICATE OF EXAMINATION

Supervisor ______________________________ Dr. Joaquín Madrenas Supervisory Committee ______________________________ Dr. Ewa Cairns ______________________________ Dr. Sean Cregan

Examiners ______________________________ Dr. John McCormick ______________________________ Dr. Lakshman Gunaratnam ______________________________ Dr. Peter Chidiac ______________________________ Dr. Nathalie Labrecque

The thesis by

Darah A. Christie

entitled:

Characterization of SLP-2 function in T lymphocytes

is accepted in partial fulfillment of the requirements for the degree of

Doctor of Philosophy

______________________ _______________________________ Date Chair of the Thesis Examination Board

 

iii

Abstract

Stomatin-like protein 2 (SLP-2) is a widely expressed mitochondrial-resident protein and a

member of the highly conserved stomatin and SPFH families, which includes Stomatins,

Prohibitins, Flotillins and bacterial HflC/K proteins. Studies on SPFH domain-containing

family members have pointed to a role in the organization of membranes into functional

domains as the mechanism underlying the control of a wide variety of cellular functions. The

Madrenas laboratory originally identified SLP-2 in the detergent-insoluble fraction of human

T cells activated through the T cell receptor and found that over-expression of SLP-2

increased T cell activation. Based on these results and on the conservation of function across

other SPFH family members, we hypothesized that SLP-2 acts to facilitate mitochondrial and

plasma membrane organization in response to T cell activation.

To investigate the role of SLP-2 during T cell activation, we generated stably transfected

Jurkat T cells expressing GFP-tagged SLP-2 and also produced recombinant human SLP-2

protein. These tools allowed us to demonstrate a direct interaction of SLP-2 with the

mitochondrial lipid cardiolipin and to show an association of SLP-2 with the prohibitin

complex. Furthermore, an increased recruitment of prohibitin to mitochondrial membranes

upon SLP-2 over-expression was also observed. At the cellular level, cells over-expressing

SLP-2 were found to have an increase in mitochondrial biogenesis, increased respiratory

chain activity and decreased susceptibility to apoptotic induction. Next, T cell-specific SLP-

2 conditional knockout mice were generated and found to have altered T cell mitochondrial

membrane organization in the absence of SLP-2. SLP-2-deficient T cells showed

significantly lower levels of cardiolipin in detergent-resistant mitochondrial fractions, along

with decreased complex I levels and activity. The absence of SLP-2 also led to a decrease in

T cell activation, which translated into a decreased rejection rate for non-matched tissue

transplants. Finally, for the first time it has been shown that SLP-2 is found in two

intracellular pools, one in mitochondria and the other associated with plasma membrane, with

the plasma membrane pool surrounding T cell receptors upon T cell activation. This implies

a fundamental role for SLP-2 in membrane organization.

iv

Taken together, the results presented here demonstrate a role for SLP-2 in the

compartmentalization of mitochondrial and plasma membranes into functional domains.

This, in turn, affects multi-domain receptor and multi-protein complex assembly and thus

affects such functions as mitochondrial biogenesis, respiratory chain activity, apoptotic

induction and T cell activation.

Keywords

Stomatin-like protein 2, mitochondria, membrane organization, cardiolipin, immunology, T

cell activation

v

Statement of Co-Authorship

In regards to the work presented in Chapter 2:

Christie, D., Lemke, C.D., Elias, I.M., Chau, L.A., Kirchhof, M.G., Li, B., Ball, E.H., Dunn, S.D., Hatch, G.M. and Madrenas, J. Stomatin-like protein 2 binds cardiolipin and regulates mitochondrial biogenesis and function. Mol. Cell. Biol 31 (18) 3845-56 (2011).

D. Christie contributed to experimental design, performed experiments for figures 2.1, 2.2,

2.3, 2.4, 2.5, 2.7, and 2.8 and wrote the manuscript. C.D. Lemke contributed to experimental

design and performed experiments for figure 2.1, 2.2, 2.7 and 2.8. I.M. Elias produced

recombinant human SLP-2, used in the vesicle precipitation assay in figure 2.6. L.A. Chau

performed the vesicle assay in figure 2.6. M.G. Kirchhof generated the SLP-2-GFP

expressing Jurkat cell line. B. Li performed apoptosis induction experiments for figure 2.8a

and b. E.H. Ball provided experimental design for the vesicle precipitation assay. S.D. Dunn

provided experimental design for recombinant human SLP-2 production. G.M. Hatch

provided experimental design and performed experiments for cardiolipin analysis and

enzyme activities for figures 2.3 and 2.7. J. Madrenas is the principle investigator of the lab

and contributed to experimental design and manuscript preparation.

In regards to the work presented in Chapter 3: Christie, D., Lian, D., Wang, H., Hatch, G. and Madrenas, J. SLP-2 deficiency in T cells is associated with abnormal mitochondrial compartmentalization of cardiolipin, decreased mitochondrial respiration and impaired CD4+ T cell responses. Manuscript submitted.

D. Christie contributed to experimental design and performed all experiments with the

exception of the heart transplant experiment and wrote the manuscript. D. Lian and H. Wang

performed the heart transplant surgery. G. Hatch performed cardiolipin measurements for

figure 3.2c and complex II+III complex activity for figure 3.2g. J. Madrenas is the principle

investigator of the lab and contributed to experimental design and manuscript preparation.

vi

In regards to the work presented in Chapter 4:

Christie, D., Kirchhof, M.G., Vardhana, S., Dustin, M.L., Madrenas, J. Plasma membrane and mitochondrial stomatin-like protein 2 pools coalesce at the immunological synapse during T cell activation. Manuscript in preparation.

D. Christie contributed to experimental design, performed experiments for figure 4.1, 4.2,

4.3, and 4.5 and wrote the manuscript. M.G. Kirchhof generated the SLP-2-GFP expressing

Jurkat cell line and performed the experiments for figure 4.4. S. Vardhana and M.L. Dustin

performed experiments for figure 4.6. J. Madrenas is the principle investigator of the lab and

contributed to experimental design and manuscript preparation.

vii

Acknowledgments

I would like to thank my supervisor, Dr. Joaquín Madrenas for his guidance and support

throughout my PhD studies, as well as my supervisory committee members Dr. Ewa Cairns

and Dr. Sean Cregan for helpful discussions and suggestions. I would also like to thank Dr.

Greg Dekeban for his role as acting co-supervisor during the last stages of my thesis studies.

I have had the pleasure to work with many excellent colleagues during my studies and I

thank Wendy Teft, Todd Fairhead, Cait Lemke, Mark Kirchhof, Michelle McCully, Samar

Sayedyahossein, Isaac Elias, Peter Mitsopoulos, Teresa Fernández, Holly Lemmon and

Cynthia Tang for many helpful discussions and insights. I would also like to thank Luan

Chau and Thu Chau for their endless technical expertise. As I have relied very heavily on

mouse work, I thank Melissa Pickering and Nikki Vannieuwenhuyze for taking excellent

care of my mouse colony. I would also like to thank Dr. Grant Hatch from the University of

Manitoba for sharing his cardiolipin expertise as well as Dr. Stan Dunn, Dr. Eric Ball and Dr.

Fred Possmayer from the Department of Biochemistry for help with experimental design.

Finally, I thank the department of Microbiology and Immunology. This has been an

excellent environment to complete my PhD studies and I thank all of the undergraduates,

graduate students, post-doctoral fellows, faculty members and staff for creating a supportive,

enthusiastic and creative environment.

viii

Table of Contents

CERTIFICATE OF EXAMINATION ................................................................................. ii  Abstract .................................................................................................................................... iii  Statement of Co-Authorship ..................................................................................................... v  Table of Contents ................................................................................................................... viii  List of Tables .......................................................................................................................... xii  List of Figures ........................................................................................................................ xiii  List of Appendices .................................................................................................................. xv  List of Abbreviations ............................................................................................................. xvi  Chapter 1 ................................................................................................................................... 1  1   Introduction ......................................................................................................................... 1  

1.1   The SPFH family of proteins and membrane compartmentalization ........................... 4  1.1.1   The stomatin family ............................................................................................ 10  1.1.2   Stomatin-like Protein 2 ....................................................................................... 11  1.1.3   Prohibitins ........................................................................................................... 15  

1.2   Mitochondrial functions ............................................................................................. 17  1.2.1   Bioenergetics and metabolism ............................................................................ 17  1.2.2   The metabolic regulation of T cells .................................................................... 21  1.2.3   Calcium signalling .............................................................................................. 23  1.2.4   Apoptosis ............................................................................................................ 25  

1.3   Rationale and Hypothesis .......................................................................................... 30  1.3.1   Rationale ............................................................................................................. 30  1.3.2   Hypothesis .......................................................................................................... 31  1.3.3   Specific Aim 1: To characterize the function of the mitochondrial pool of SLP-2 ............................................................................................................................ 32  1.3.4   Specific Aim 2: To generate and characterize a T cell-specific SLP-2 conditional

knockout mouse .................................................................................................. 33  1.3.5   Specific Aim 3: To characterize the plasma membrane-associated pool of SLP-2 ............................................................................................................................ 35  

1.4   References .................................................................................................................. 37  Chapter 2 ................................................................................................................................. 50  2   Stomatin-Like Protein 2 Binds Cardiolipin and Regulates Mitochondrial Biogenesis and

Function ........................................................................................................................... 50  2.1   Introduction ................................................................................................................ 50  2.2   Materials and Methods ............................................................................................... 51  

2.2.1   Cells .................................................................................................................... 51  2.2.2   Plasmids, siRNA and T cell transfectants ........................................................... 52  2.2.3   Upregulation of SLP-2 expression ...................................................................... 52  2.2.4   Antibodies ........................................................................................................... 52  2.2.5   Reagents .............................................................................................................. 53  2.2.6   Confocal microscopy .......................................................................................... 53  2.2.7   Transmission electron microscopy ..................................................................... 53  2.2.8   Measurement of mitochondrial mass .................................................................. 54  2.2.9   Measurement of cardiolipin ................................................................................ 54  2.2.10   Measurement of cardiolipin mass in stable doxycycline-inducible SLP-2-GFP

transfected Jurkat cells ...................................................................................... 54  

ix

2.2.11   Radiotracer studies of de novo cardiolipin synthesis ........................................ 55  2.2.12   Real time-PCR .................................................................................................. 55  2.2.13   Mitochondrial DNA quantification ................................................................... 55  2.2.14   Cell lysate preparation ...................................................................................... 56  2.2.15   Mitochondria isolation ...................................................................................... 56  2.2.16   Prohibitin and SLP-2 membrane association .................................................... 56  2.2.17   NADH dehydrogenase activity ......................................................................... 56  2.2.18   ATP quantitation ............................................................................................... 56  2.2.19   T cell stimulation .............................................................................................. 57  2.2.20   Oxygen consumption ........................................................................................ 57  2.2.21   Induction and detection of apoptosis ................................................................ 57  2.2.22   Generation of human recombinant SLP-2 (hrSLP-2). ...................................... 57  2.2.23   Phospholipid vesicle co-precipitation assay ..................................................... 59  

2.3   Results ........................................................................................................................ 59  2.3.1   SLP-2 is localized mostly in mitochondria ......................................................... 59  2.3.2   Upregulation of SLP-2 expression increases mitochondrial biogenesis ............. 62  2.3.3   SLP-2 interacts with prohibitins and binds cardiolipin-enriched microdomains 71  2.3.4   Upregulation of SLP-2 expression is associated with increased mitochondrial

functions .............................................................................................................. 76  2.3.5   Upregulation of SLP-2 expression increases T cell responses and resistance to

apoptosis ............................................................................................................. 80  2.4   Discussion .................................................................................................................. 83  2.5   References .................................................................................................................. 87  

Chapter 3 ................................................................................................................................. 91  3   SLP-2 deficiency in T cells is associated with abnormal mitochondrial membrane

compartmentalization of cardiolipin, decreased mitochondrial respiration and impaired CD4+ T cell responses ....................................................................................................... 91  

3.1   Introduction ................................................................................................................ 91  3.2   Materials and Methods ............................................................................................... 93  

3.2.1   Mice .................................................................................................................... 93  3.2.2   Confirmation of SLP-2 deletion ......................................................................... 93  3.2.3   Isolation of detergent-insoluble fraction of mitochondrial membranes .............. 94  3.2.4   Measurement of cardiolipin ................................................................................ 94  3.2.5   Enzyme activity assays ....................................................................................... 95  3.2.6   Thymocyte and T cell population analysis ......................................................... 95  3.2.7   Flow cytometry ................................................................................................... 95  3.2.8   Stimulation of T lymphocytes ............................................................................. 95  3.2.9   In vivo immunization and ex vivo recall response ............................................. 96  3.2.10   Intracellular staining cytokine analysis ............................................................. 96  3.2.11   Real time PCR analysis ..................................................................................... 96  3.2.12   Heart transplants ............................................................................................... 97  3.2.13   Activation-induced cell death assay ................................................................. 97  3.2.14   Statistical analysis ............................................................................................. 97  

3.3   Results ........................................................................................................................ 97  3.3.1   Generation of T cell-specific SLP-2 knockout mice .......................................... 97  3.3.2   SLP-2 deficiency is associated with abnormal cardiolipin compartmentalization

in mitochondrial membranes and reduced mitochondrial respiration ................ 98  3.3.3   Impaired ex vivo responses of SLP-2-deficient T cells ..................................... 104  

x

3.3.4   SLP-2 deficiency impairs T cell responses in vivo ........................................... 109  3.3.5   The IL-2 defect of SLP-2 deficient T cells occurs at the post-transcriptional level .......................................................................................................................... 112  3.3.6   Altered metabolic capacity of SLP-2-deficient T cells ..................................... 116  

3.4   Discussion ................................................................................................................ 119  3.5   References ................................................................................................................ 123  

Chapter 4 ............................................................................................................................... 129  4   Plasma membrane and mitochondrial Stomatin-like Protein 2 pools coalesce at the

immunological synapse during T cell activation ............................................................ 129  4.1   Introduction .............................................................................................................. 130  4.2   Material and Methods .............................................................................................. 131  

4.2.1   Cells .................................................................................................................. 131  4.2.2   Plasmids, siRNA and T cell transfectants ......................................................... 131  4.2.3   Mice .................................................................................................................. 132  4.2.4   Antibodies ......................................................................................................... 132  4.2.5   Mitochondria isolation ...................................................................................... 133  4.2.6   Immunoprecipitations ....................................................................................... 133  4.2.7   Cell surface biotinylation .................................................................................. 133  4.2.8   Confocal microscopy ........................................................................................ 133  

4.3   Results ...................................................................................................................... 135  4.3.1   The largest cellular pool of SLP-2 is associated with mitochondria ................ 135  4.3.2   Detection of a small pool of SLP-2 associated with the plasma membrane ..... 138  4.3.3   Homo-oligomerization of SLP-2 ...................................................................... 141  4.3.4   SLP-2 redistributes to the immunological synapse during T cell activation .... 141  4.3.5   The SLP-2 pools predominantly partition in the pSMAC of the immunological

synapse .............................................................................................................. 146  4.3.6   Mitochondrial recruitment to the IS does not require SLP-2 ............................ 152  

4.4   Discussion ................................................................................................................ 155  4.5   References ................................................................................................................ 159  

Chapter 5 ............................................................................................................................... 164  5   Discussion ....................................................................................................................... 164  

5.1   Summary of results .................................................................................................. 164  5.1.1   Stomatin-like protein 2 binds cardiolipin and regulates mitochondrial biogenesis

and function ...................................................................................................... 164  5.1.2   SLP-2 deficiency in T cells is associated with abnormal mitochondrial

membrane compartmentalization of cardiolipin, decreased mitochondrial respiration and impaired CD4+ T cell responses ............................................... 166  

5.1.3   Plasma membrane and mitochondrial stomatin-like protein 2 coalesce at the immunological synapse during T cell activation .............................................. 167  

5.2   Model of function: SLP-2 organizes membrane domains ....................................... 169  5.2.1   Assembly of multi-domain receptors, including respiratory chain supercomplex

formation ........................................................................................................... 176  5.2.2   Calcium buffering ............................................................................................. 177  5.2.3   Mitochondria membrane associated protease regulation .................................. 179  5.2.4   Specialized membrane microdomain formation ............................................... 180  5.2.5   T cell activation ................................................................................................ 181  

5.3   Future directions ...................................................................................................... 183  5.3.1   Characterization of the structure of SLP-2 ....................................................... 183  

xi

5.3.2   Characterization of cardiolipin-enriched microdomains .................................. 184  5.3.3   Intracellular trafficking of SLP-2 ..................................................................... 186  5.3.4   Functional role of SLP-2 in cell physiology ..................................................... 187  

5.4   SLP-2 and disease .................................................................................................... 188  5.4.1   SLP-2 and mitochondrial disorders .................................................................. 188  5.4.2   SLP-2 in cancer prognosis and metastasis ........................................................ 189  

5.5   Conclusions .............................................................................................................. 190  5.6   References ................................................................................................................ 192  

Appendix A: Ethics Approval ............................................................................................... 199  Appendix B: Copyright Permission ...................................................................................... 201  Curriculum Vitae .................................................................................................................. 202  

xii

List of Tables

Table 3.1: T cell subsets in lymph nodes of SLP-2 T-K/O mice upon OVA immunization 113  Table 4.1: Segregation of mitochondrial SLP-2-GFP from the cSMAC in the mature IS, by

wide field fluorescence microscopy ................................................................... 150  Table 4.2: Percentage of Jurkat T cells on supported planar bilayers organizing SLP-2-GFP

close to pSMAC at the contact interface by wide field fluorescence microscopy ............................................................................................................................ 151  

xiii

List of Figures

Figure 1.1: Alignment of the predicted SPFH domain of SPFH family members in human, mouse and E. coli proteins. .................................................................................... 6  

Figure 1.2: Phylogenetic analysis of the SPFH domain of human, mouse and E. coli family members. ................................................................................................................ 9  

Figure 1.3: Features of the SLP-2 amino acid sequence. ........................................................ 13  

Figure 1.4: Schematic overview of the respiratory chain. ...................................................... 19  Figure 2.1: The major pool of SLP-2 in human T cells is located in mitochondria ............... 61  

Figure 2.2: Induction of SLP-2 expression triggers mitochondrial biogenesis in human T cells. ..................................................................................................................... 64  

Figure 2.3: Induction of SLP-2 expression increases de novo cardiolipin biosynthesis, nuclear transcription programs, and mitochondrial DNA replication. ............................. 67  

Figure 2.4: SLP-2 upregulation precedes PGC-1α upregulation and mitochondrial biogenesis during T cell activation. ....................................................................................... 70  

Figure 2.5: SLP-2 interacts with prohibitins. .......................................................................... 73  Figure 2.6: SLP-2 binds cardiolipin. ....................................................................................... 75  

Figure 2.7: Induction of SLP-2 expression is associated with higher mitochondrial activity. 78  Figure 2.8: SLP-2 upregulation increases T cell responses and protects against apoptosis. .. 82  

Figure 3.1: Generation of T cell-specific SLP-2 knockout mice. ......................................... 100  Figure 3.2: T cells deficient in SLP-2 have abnormal cardiolipin compartmentalization and

decreased activity of respiratory complexes I and II+III. .................................. 103  Figure 3.3: Grossly normal thymic T cell development and peripheral T cell pool in SLP-2 T-

K/O mice. ........................................................................................................... 106  Figure 3.4: T cells deficient in SLP-2 produce less IL-2 in response to T cell activation. ... 108  

Figure 3.5: T cells deficient in SLP-2 have decreased responses in vivo. ............................ 111  

Figure 3.6: SLP-2-deficiency in T cells induces a post-transcriptional defect in IL-2 production. ......................................................................................................... 115  

Figure 3.7: Altered metabolic capacity of SLP-2-deficient T cells during activation. ......... 118  

Figure 4.1: SLP-2 is a mitochondrial protein. ....................................................................... 137  

Figure 4.2: A small pool of SLP-2 is associated with the plasma membrane. ..................... 140  Figure 4.3: Homo-oligomerization of SLP-2. ....................................................................... 143  

Figure 4.4: SLP-2 polarizes to the immunological synapse during T cell activation. .......... 145  

xiv

Figure 4.5: Redistribution of plasma membrane-associated SLP-2 and mitochondria-associated SLP-2 during immunological synapse formation. ............................ 148  

Figure 4.6: SLP-2-deficient T cells show normal mitochondrial recruitment upon T cell stimulation. ......................................................................................................... 154  

Figure 5.1: A step-wise model of the formation of cardiolipin-enriched microdomains in the mitochondrial membrane as facilitated by SLP-2. ............................................. 173  

Figure 5.2: Model of supercomplex formation upon SLP-2 upregulation in response to T cell activation. ........................................................................................................... 175  

xv

List of Appendices

Appendix A: Ethics Approval ............................................................................................... 199  Appendix B: Copyright Permission ...................................................................................... 201  

xvi

List of Abbreviations

AAA ATPases associated with diverse cellular activities

ActD Actinomycin D

ADP Adenosine diphosphate

AMPK AMP-activated protein kinase

ANOVA Analysis of variance

AP-1 Activator protein 1

Apaf-1 Apoptotic protease activating factor 1

APC Antigen presenting cell

ASIC Acid sensing ion channel

ATP Adenosine triphosphate

BAK Bcl-2 homologous antagonist/killer

BAX Bcl-2 associated X protein

BCL-2 B cell lymphoma-2

BLAST Basic local alignment search tool

CCO Cytochrome c oxidase I

CD Cluster of differentiation

Cdk Cyclin dependent kinase

CL Cardiolipin

CoA Coenzyme A

CRAC Calcium release activated calcium

CS Citrate synthase

cSMAC Central supramolecular activation cluster

DAG Diacylglycerol

xvii

DEG/EnaC Degenerin/epithelial sodium channel

DI-GEM Detergent-insoluble glycolipid-enriched microdomain

DIABLO Direct IAP binding protein with low pI

DN Double negative

DP Double positive

Drp-1 Dynamin-related protein-1

ELISA Enzyme linked immunosorbent assay

ER Endoplasmic reticulum

ERK Extracellular signal-regulated kinase

EST Expressed sequence tag

FAD Flavin adenine dinucleotide

Gads GRB2-related adapter downstream of Shc

GAPDH Glyceraldehyde 3-phosphate dehydrogenase

GFP Green fluorescent protein

GRB2 Growth factor receptor-bound protein 2

hrSLP-2 Human recombinant SLP-2

IAP Inhibitor of apoptosis

ICAM-1 Intercellular adhesion molecule 1

IFN Interferon

IL Interleukin

IP Immunoprecipitation

IP3 Inositol triphosphate

IS Immunological synapse

ITAM Immunoreceptor tyrosine-based activation motif

Itk IL-2 inducible T cell kinase

xviii

LAT Linker of activated T cells

LCMV Lymphocytic choriomeningitis virus

LFA-1 Lymphocyte function-associated antigen 1

MAP Mitogen activated protein

MEF Mouse embryonic fibroblast

Mff Mitochondrial fission factor

Mfn Mitofusin

MHC Major histocompatibility complex

mTOR Mammalian target of rapamycin

mtRFP Mitochondrial red fluorescence protein

NAD Nicotinamide adenine dinucleotide

NAO Nonyl acridine orange

NFAT Nuclear factor of activated T cells

NFκB Nuclear factor κB

OPA-1 Optic atrophy 1

OVA Ovalbumin

OXPHOS Oxidative phosphorylation

P Pellet

PBMC Peripheral blood mononuclear cells

PC Phosphatidylcholine

PCC Pigeon cytochrome c

PE Phosphatidylethanolamine

PG Phosphatidylglycerol

PGC PPAR (peroxisome proliferator-activated receptor) - γ coactivator

PHB Prohibitin

xix

PI Phosphatidylinositol

PI3K Phosphoinositide 3-kinase

PKC Protein kinase C

PLC-γ1 Phospholipase C-γ1

PMA Phorbol myristate acetate

PS Phosphatidylserine

pSMAC Peripheral supramolecular activation cluster

RT-PCR Reverse transcriptase polymerase chain reaction

S Supernatant

SDH Succinate dehydrogenase

SDS-PAGE Sodium dodecyl polyacrylamide gel electrophoresis

SEE Staphylococcal enterotoxin E

SHP Src homology phosphatase

siRNA Small interfering RNA

SLP Stomatin-like protein

SLP-76 Src homology 2 domain containing leukocyte protein of 76kDa

SMAC Supramolecular activation cluster

SMAC Second mitochondria-derived activator of caspases

SOS Son of sevenless

SP Single positive

SPFH Stomatin, Prohibitin, Flotillin, HflC/K

STIM1 Stromal interaction molecule 1

T-K/O T cell knockout

tBID Truncated BID

TCA Tricarboxylic acid

xx

TCR T cell receptor

TH T helper

TIRFM Total internal reflection fluorescence microscopy

TMHMM Transmembrane hidden Markov model

TRAF TNF-receptor associated factor

TRAIL TNF-related apoptosis-inducing ligand

Treg T regulatory

UQ Ubiquinone

VDAC Voltage dependent anion channel

ZAP-70 Zeta-chain associated protein kinase 70

ΔΨm Mitochondrial transmembrane potential

1

Chapter 1

1 Introduction

The immune system has evolved to provide a defense against invading pathogens,

including bacteria, viruses and fungi that threaten the health and survival of the animal.

T cells function to directly lyse infected cells or to secrete cytokines, which activate other

immune cells to eliminate pathogens. The function of T cells requires the translation of

antigen recognition into gene expression to facilitate pathogen elimination. This occurs

via intracellular signalling cascades that translate T cell receptor (TCR) binding into the

activation of transcriptional programs. T cell signalling events are complex and require

the interaction of a large number of signalling and adapter proteins at both the

extracellular and cytoplasmic faces of the plasma membrane. The organization of the

plasma membrane into specialized micro-domains to sequester signalling components

away from other plasma membrane proteins could strengthen the association of signalling

components to promote T cell activation 1, 2.

The TCR is a dimer composed of two transmembrane proteins with large extracellular

domains forming a ligand recognition site. The TCR ligand is a specific antigenic

peptide bound to a major histocompatibility complex (MHC) molecule. The TCR is

found at the cell surface in association with CD3 complex composed of molecules of γε,

δε or ζζ pairs 3, 4. The intracellular domains of the CD3 proteins contain regions known

as immunoreceptor tyrosine-based activation motifs (ITAM), which consist of two

tyrosine residues separated by a defined amino acid sequence (YXXL/I-X6-8-YXXL/I) 5, 6.

These tyrosine residues can be phosphorylated and act as a docking site for proteins

involved in the T cell signalling cascade. Upon antigen recognition, CD4 or CD8

molecules cluster together with the TCR, by virtue of their binding to MHC of class II or

class I respectively, on antigen presenting cells. Both CD4 and CD8 molecules are

associated with the Src family kinase Lck, which phosphorylates the ζ chain ITAMs 7.

The Syk family kinase ZAP-70 then binds the phosphorylated ITAMs and, in turn,

2

phosphorylates the adapters LAT (linker of activated T cells) and SLP-76 (Src homology

2 domain containing leukocyte protein of 76 kDa). LAT is a transmembrane adapter

protein which upon phosphorylation by ZAP-70, provides docking sites for a number of

signalling and adapter molecules, including Gads (GRB2-related adapter downstream of

Shc), which bind to and recruit other adapters, such as SLP-76 8. The complex of

signalling and scaffold proteins recruited to the TCR upon antigen binding is known as

the TCR signalosome.

A major signalling event triggered at the LAT/SLP-76 scaffold is the recruitment and

activation of phospholipase C γ1 (PLCγ1). This enzyme is recruited upon

phosphorylation of LAT and is activated by another kinase recruited to the LAT/SLP-76

scaffold, the Tec family kinase Itk (IL-2 inducible T cell kinase). Activated PLCγ

cleaves the lipid phosphatidylinositol-4,5-bisphosphate into diacylglycerol (DAG) and

inositol-1,4,5-triphosphate (IP3), each of which initiate signalling pathways 5. DAG

activates the mitogen activated protein (MAP) kinase cascade, leading to the activation of

Elk1, a subunit of the dimeric AP-1 transcription factor complex. The MAP kinase

cascade is also activated by guanine nucleotide exchange factor son of sevenless (SOS)

which is recruited to the LAT/SLP-76 scaffold by the adapter GRB2. In addition to the

MAP kinase cascade, DAG also activates protein kinase Cθ (PKCθ), which leads to the

activation of the nuclear factor κB (NFκB) family of transcription factors. In parallel, IP3

triggers the release of intracellular calcium stores, which triggers the opening of cell

surface calcium channels. The surge of intracellular calcium then activates calcineurin,

which in turn, activates the nuclear factor of activated T cells (NFAT) family of

transcription factors 9. The transcription factors activated from these various signalling

cascades upregulate the expression of effector molecules needed for T cell responses.

TCR ligation and signalling also triggers the formation of the immunological synapse, a

unique organization of the T cell membrane into specialized domains involved in the

maintenance and regulation of T cell signalling 10, 11. The peripheral supramolecular

activation cluster (pSMAC) is a ring of high avidity lymphocyte function associated

antigen 1 (LFA-1) and intracellular adhesion molecule 1 (ICAM-1), which facilitate the

tight association with the antigen presenting cell. On the other hand, as the TCR:MHC

3

complexes form in the pSMAC, they migrate into and concentrate at the center of the

SMAC (cSMAC), driven by actin polymerization 12. The pSMAC excludes large

inhibitory molecules, including phosphatases such as CD45, which is segregated to the

distal SMAC, to prevent inactivation of the T cell signalosome. Although it was

originally thought that T cell signalling occurred at the cSMAC, blocking the formation

of new TCR:MHC complexes showed that as TCR:MHC complexes migrate into the

cSMAC they dissociate from ZAP-70, indicating a loss of T cell signalling 13. Thus, the

organization of the immune synapse allows for the termination of TCR signalling

followed by TCR degradation upon entering the cSMAC 14.

In addition to the membrane organization following antigen recognition, T cell signalling

is thought to involve membrane compartmentalization into lipid microdomains enriched

in cholesterol and glycolipids known as detergent-insoluble glycolipid-enriched

microdomains (DI-GEMs) or lipid rafts. These are highly structured assemblies of

tightly packed lipids and proteins found in the plasma membrane that are thought to exist

in nano-units that coalesce in response to cell stimulation 1, 2. Studies have shown that T

cell stimulation leads to the aggregation of lipid rafts at the T cell signalosome, which

may be important for T cell activation 15-17. In addition, a number of T cell signalosome

components are constitutively associated with lipid raft domains, including Lck and LAT 18. This localization appears to be essential for T cell activation as a mutant LAT unable

to associate with lipid rafts leads to the inhibition of T cell responses 19. The formation of

lipid raft domains from the nanoassemblies is likely facilitated by lipid-lipid, lipid-protein

and protein-protein interactions and as such, actin rearrangement during T cell activation

is thought to help raft organization 20. A number of studies have also shown that

cholesterol and sphingomyelin disruption leads to altered T cell responses, likely due to

disruption in raft formation 21, 22. This membrane organization is likely important for

sequestering signalling components while also excluding inhibitors such as CD45 or

SHP-1, which are normally found outside DI-GEMs 23, 24.

Within this framework and with the aim to identify novel proteins involved in the

regulation of T cell activation and membrane organization, our lab identified stomatin-

like protein 2 (SLP-2), a member of the highly conserved stomatin family. The

4

investigation of the functional role played by SLP-2 in T cell activation will be the focus

of this thesis.

1.1 The SPFH family of proteins and membrane compartmentalization

The organization of membrane components into defined functional domains plays an

important role in many cellular processes, including cell signalling, membrane

trafficking, ion channel activity and viral budding 1, 25. The SPFH family of proteins,

consisting of Stomatin, Prohibitin, Flotillin and the bacterial HflC/K proteins, has been

proposed to play a role in the organization of membrane domains by binding directly to

lipids, as well as to other proteins, to facilitate membrane compartmentalization 26, 27.

Members of this family are anchored to various cellular membranes, including the plasma

membrane, early endosomes, Golgi apparatus, mitochondria and endoplasmic reticulum.

SPFH family members are defined by the highly conserved, shared SPFH domain (Fig.

1.1, 1.2), which is proposed to facilitate lipid association as the SPFH family members

podocin and Mec-2 have been shown to bind directly to the lipid raft component

cholesterol and many SPFH family members are enriched in DI-GEMs, where they carry

out many diverse cellular functions 26, 27. Prohibitins are mitochondrial proteins proposed

to be involved in mitochondrial chaperone activity 28, 29, organization of mitochondrial

DNA nucleoids 30, regulation of lifespan 31, 32, proliferation and apoptosis 33,

compartmentalization of mitochondrial membranes 34, 35 and mitochondrial biogenesis 29,

36, 37. Flotillins are widely expressed and found at the plasma membrane as well as

intracellular membranes and have been proposed to organize lipid rafts, which may

regulate a number of functions, including insulin-stimulated glucose uptake, neuronal

regeneration, phagocytosis, cell proliferation, signal transduction and cytoskeletal

remodeling (reviewed in 38, 39). Finally, the bacterial proteins HflC/K are hypothesized to

be involved in the regulation of FtsH, a zinc metalloprotease with high similarity to the

5

Figure 1.1: Alignment of the predicted SPFH domain of SPFH family members in

human, mouse and E. coli proteins.

The predicted SPFH domains from human (h) and mouse (m) SPFH family members

stomatin, SLP-1, SLP-2, SLP-3, podocin, flotillin 1, flotillin 2, PHB1 and PHB2 along

with E. coli HflC and HflK proteins were aligned using Clustal W and conserved residues

and the SPFH domain consensus sequence were predicted using Boxshade software.

6

Figure 1.1

hStomatin 52 MCIKIIKEYERAIIFRLGRILQGG-----AKGPGLFFILPCTDSFIKVD-MRTISFDIPP mStomatin 52 ICIKIVKEYERVIIFRLGRILQGG-----AKGPGLFFILPCTDSLIKVD-MRTISFDIPP hSLP-3 49 ---------ERAVVFRLGRIQADK-----AKGPGLILVLPCIDVFVKVD-LRTVTCNIPP mSLP-3 45 MCLKIIKEYERAVVFRLGRIQADK-----AKGPGLILVLPCIDVFVKVD-LRTVTCNIPP hPodocin 123 FCVKVVQEYERVIIFRLGHLLPGR-----AKGPGLFFFLPCLDTYHKVD-LRLQTLEIPF mPodocin 125 FCIKVVQEYERVIIFRLGHLLPGR-----AKGPGLFFFLPCLDTYHKVD-LRLQTLEIPF hSLP-1 77 FALKIVPTYERMIVFRLGRIRT-------PQGPGMVLLLPFIDSFQRVD-LRTRAFNVPP mSLP-1 77 FALKIVPTYERMIVFRLGRIRN-------PQGPGMVLLLPFIDSFQRVD-LRTRAFNVPP hSLP-2 36 TVVLFVPQQEAWVVERMGRFHR-------ILEPGLNILIPVLDRIRYVQSLKEIVINVPE mSLP-2 36 TVILFVPQQEAWVVERMGRFHR-------ILEPGLNVLIPVLDRIRYVQSLKEIVINVPE hFlotillin1 4 ---TCGPNEAMVVSG-FCRSP-PV-----MVAGGRVFVLPCIQQIQRIS-LNTLTLNVKS mFlotillin1 4 ---TCGPNEAMVVSG-FCRSP-PV-----MVAGGRVFVLPCIQQIQRIS-LNTLTLNVKS hFlotillin2 6 ---TVGPNEALVVSGGCCGSDYKQ-----YVFGGWAWAWWCISDTQRIS-LEIMTLQPRC mFlotillin2 6 ---TVGPNEALVVSGGCCGSDYKQ-----YVFGGWAWAWWCISDTQRIS-LEIMTLQPRC hPHB1 26 ALYNVDAGHRAVIFDRFRGVQ-DI-----VVGEGTHFLIPWVQKPIIFD-CRSRPRNVP- mPHB1 26 ALYNVDAGHRAVIFDRFRGVQ-DI-----VVGEGTHFLIPWVQKPIIFD-CRSRPRNVP- hPHB2 39 SVFTVEGGHRAIFFNRIGGVQQDT-----ILAEGLHFRIPWFQYPIIYD-IRARPRKIS- mPHB2 39 SVFTVEGGHRAIFFNRIGGVQQDT-----ILAEGLHFRIPWFQYPIIYD-IRARPRKIS- E.coliHflC 18 MSVFVVKEGERGITLRFGKVLRDDDNKPLVYEPGLHFKIPFIETVKMLD-ARIQTMDNQA E.coliHflK 95 SGFYTIKEAERGVVTRFGKFSH-------LVEPGLNWKPTFIDEVKPVN-VEAVRELAAS consensus 1 i ii ervii rlgri ilgpGl filpcid kvd lrtis nip hStomatin 106 QEILTKDSVTISVDGVVYYRVQN----------ATLAVANITNADSATRLLAQTTLRNVL mStomatin 106 QEVLTKDSVTISVDGVVYYRVQN----------ATLAVANITNADSATRLLAQTTLRNAL hSLP-3 94 QEILTRDSVTTQVDGVVYYRIYS----------AVSAVANVNDVHQATFLLAQTTLRNVL mSLP-3 99 QEILTRDSVTTQVDGVVYYRIYS----------AVSAVANVNDVHQATFLLAQTTLRNVL hPodocin 177 HEIVTKDMFIMEIDAICYYRMEN----------ASLLLSSLAHVSKAVQFLVQTTMKRLL mPodocin 179 HEVVTKDMFIMEIDAVCYYRMEN----------ASLLLSSLAHVSKAIQFLVQTTMKRLL hSLP-1 129 CKLASKDGAVLSVGADVQFRIWD----------PVLSVMTVKDLNTATRMTAQNAMTKAL mSLP-1 129 CKLASKDGAVLSVGADVQFRIWD----------PVLSVMAVKDLNTATRMTAHNAMTKAL hSLP-2 89 QSAVTLDNVTLQIDGVLYLRIMD----------PYKASYGVEDPEYAVTQLAQTTMRSEL mSLP-2 89 QSAVTLDNVTLQIDGVLYLRIMD----------PYKASYGVEDPEYAVTQLAQTTMRSEL hFlotillin1 53 EKVYTRHGVPISVTGIAQVKIQGQNKEMLAAACQMFLGKTEAEIAHIALETLEGHQRAIM mFlotillin1 53 EKVYTRHGVPISVTGIAQVKIQGQNKEMLAAACQMFLGKTEAEIAHIALETLEGHQRAIM hFlotillin2 57 EDVETAEGVALTVTGVAQVKIMT-EKELLAVACEQFLGKNVQDIKNVVLQTLEGHLRSIL mFlotillin2 57 EDVETAEGVALTVTGVAQVKIMT-EKELLAVACEQFLGKNVQDIKNVVLQTLEGHLRSIL hPHB1 78 VITGSKDLQNVNITLRILFRPVASQLP------RIFTSIGEDYDERVLPSITTEILKSVV mPHB1 78 VITGSKDLQNVNITLRILFRPVASQLP------RIYTSIGEDYDERVLPSITTEILKSVV hPHB2 92 SPTGSKDLQMVNISLRVLSRPNAQELP------SMYQRLGLDYEERVLPSIVNEVLKSVV mPHB2 92 SPTGSKDLQMVNISLRVLSRPNAQELP------SMYQRLGLDYEERVLPSIVNEVLKSVV E.coliHflC 77 DRFVTKEKKDLIVDSYIKWRISDFSR-----YYLATGGGDISQAEVLLKRKFSDRLRSEI E.coliHflK 147 GVMLTSDENVVRVEMNVQYRVTN----------PEKYLYSVTSPDDSLRQATDSALRGVI consensus 61 iltkd v i vdgvv yri v i dvd av l q lr vl

7

hStomatin 156 GTKNLSQILSD-REEIAHNMQSTLD----------------------------------- mStomatin 156 GTKNLSQILSD-REEIAHHMQSTLD----------------------------------- hSLP-3 144 GTQTLSQILAG-REEIAHSIQTLLD----------------------------------- mSLP-3 149 GTQTLSQILSG-REEIAHSIQTLLD----------------------------------- hPodocin 227 AHRSLTEILLE-RKSIAQDAKVALD----------------------------------- mPodocin 229 AHRSLTEILLE-RKSIAQDVKVALD----------------------------------- hSLP-1 179 LKRPLREIQME-KLKISDQLLLEIN----------------------------------- mSLP-1 179 LRRPLQEIQME-KLKIGDQLLLEIN----------------------------------- hSLP-2 139 GKLSLDKVFRE-RESLNASIVDAIN----------------------------------- mSLP-2 139 GKLSLDKVFRE-RESLNANIVDAIN----------------------------------- hFlotillin1 113 AHMTVEEIYKD-RQKFSEQVFKVAS----------------------------------- mFlotillin1 113 AHMTVEEIYKD-RQKFSEQVFKVAS----------------------------------- hFlotillin2 116 GTLTVEQIYQD-RDQFAKLVREVAA----------------------------------- mFlotillin2 116 GTLTVEQIYQD-RDQFAKLVREVAA----------------------------------- hPHB1 132 ARFDAGELITQ-RELVSRQVSDDLT----------------------------------- mPHB1 132 ARFDAGELITQ-RELVSRQVSDDLT----------------------------------- hPHB2 146 AKFNASQLITQ-RAQVSLLIRRELT----------------------------------- mPHB2 146 AKFNASQLITQ-RAQVSLLIRRELT----------------------------------- E.coliHflC 132 GRLDVKDIVTDSRGRLTLEVRDALNSGSAGTEDEVTTPAADNAIAEAAERVTAETKGKVP E.coliHflK 197 GKYTMDRILTEGRTVIRSDTQRELEET--------------------------------- consensus 121 gh tl il d re i m l hStomatin 180 ----DATDAWGIKVERVEIKDVKLPVQLQRAMAAEAEASREARAKVIAAEGE-------- mStomatin 180 ----DATDDWGIKVERVEIKDVKLPVQLQRAMAAEAEAAREARAKVIAAEGE-------- hSLP-3 168 ----DATELWGIRVARVEIKDVRIPVQLQRSMAAEAEATREARAKVLAAEGEMNASKSLK mSLP-3 173 ----DATELWGIRVARVEIKDVRIPVQLQRSMAAEAEATREARAKVLAAEGE-------- hPodocin 251 ----SVTCIWGIKVERIEIKDVRLPAGLQHSLAVEAEAQRQAKVRMIAAEAEK------- mPodocin 253 ----AVTCIWGIKVERTEIKDVRLPAGLQHSLAVEAEAQRQAKVRVIAAEGEK------- hSLP-1 203 ----DVTRAWGLEVDRVELAVEAVLQPPQDSPAG-------------------------- mSLP-1 203 ----DVTRAWGLEVDRVELAVEAVLQPPQDSLTVPSLDS--------------------- hSLP-2 163 ----QAADCWGIRCLRYEIKDIHVPPRVKESMQMQVEAERRKRATVLESEG--------- mSLP-2 163 ----QAADCWGIRCLRYEIKDIHVPPRVKESMQMQVEAERRKRATVLESEG--------- hFlotillin1 137 ----SDLVNMGISVVSYTLKDIHDDQDYLHSLGKARTAQVQKDARIGEAEAKRDAGIREA mFlotillin1 137 ----SDLVNMGISVVSYTLKDIHDDQDYLHSLGKARTAQVQKDARIGEAEAKRDAGIREA hFlotillin2 140 ----PDVGRMGIEILSFTIKDVYDKVDYLSSLGKTQTAVVQRDADIGVAEAERDAGIREA mFlotillin2 140 ----PDVGRMGIEILSFTIKDVYDKVDYLSSLGKTQTAVVQRDADIGVAEAERDAGIREA hPHB1 156 ----ERAATFGLILDDVSLTHLTFGKEFTEAVEAKQVAQQEAERARFVVEKAEQQ----- mPHB1 156 ----ERAATFGLILDDVSLTHLTFGKEFTEAVEAKQVAQQEAERARFVVEKAEQQ----- hPHB2 170 ----ERAKDFSLILDDVAITELSFSREYTAAVEAKQVAQQEAQRAQFLVEKAKQE----- mPHB2 170 ----ERAKDFSLILDDVAITELSFSREYTAAVEAKQVAQQEAQRAQFLVEKAKQE----- E.coliHflC 192 VINPNSMAALGIEVVDVRIKQINLPTEVSEAIYNRMRAEREAVARRHRSQGQ-------- E.coliHflK 224 ----IRPYDMGITLLDVNFQAARPPEEVKAAFDDAIAARENEQQYIREAEAY-------- consensus 181 d wgi verveikdvklp dl sma a a a v aeg

8

Figure 1.2: Phylogenetic analysis of the SPFH domain of human, mouse and E. coli

family members.

The predicted SPFH domains from human (h) and mouse (m) SPFH family members

stomatin, SLP-1, SLP-2, SLP-3, podocin, flotillin 1, flotillin 2, PHB1 and PHB2 along

with E. coli HflC and HflK proteins were aligned using Clustal W and a phylogenetic tree

was generated.

9

Figure 1.2

10

AAA (ATPases associated with diverse cellular activities) family of ATPases 40, a

function that may be conserved in prohibitin 37. While each member has unique

functions, many of which are currently unknown, all seem to be involved in regulating

the formation or organization of DI-GEMs to affect various cell processes, a function that

is likely conserved in the stomatin family 27, 38, 39.

1.1.1 The stomatin family

The stomatin family consists of 5 members, stomatin, stomatin-like protein 1 (SLP-1),

SLP-2, SLP-3 and podocin 41. This is a highly conserved family of proteins with

homologues found in primates, rodents, birds, amphibians, teleosts, insects, nematodes,

fungi, plants, prokaryotes and Archaea 41. Stomatin was originally identified in patients

with stomatocytosis, a hemolytic anemia resulting from increased membrane

permeability to sodium and potassium 42. Erythrocytes from these patients were found to

be deficient in stomatin 43, indicating a possible role in cation channel regulation.

Surprisingly, although there is high similarity between human and mouse stomatin, a

stomatin knockout mouse failed to show a stomatocytosis phenotype, indicating that

while stomatin may be involved in regulating cation transport, it is not responsible for the

phenotype of erythrocytes of patients with stomatocytosis 44. Further evidence of

stomatin-mediated ion channel regulation has been shown for the degenerin / epithelial

sodium channel (DEG/ENaC) family of ion channels, including the acid-sensing ion

channels (ASIC) 45-49. Initial studies into DEG/ENaC regulation was carried out in

Caenorhabditis elegans, which expresses three stomatin homologues, mec-2, unc-1 and

unc-24 50-52. Mec-2 was originally identified in touch-insensitive mutants and is thought

to regulate DEG/ENaC mechanosensory transduction channels as Mec-2 localizes to the

touch-receptor neurons in C. elegans and co-expression in oocytes with the DEG/ENaC

ion channel components Mec-4 and Mec-10 leads to increased current in cells 47. This

finding was corroborated with human and mouse stomatin, both of which associate with

ASIC when co-expressed in COS-7 cells and co-expression with ASIC3 leads to reduced

current in cells, indicating a regulatory role for stomatin 46. Furthermore, SLP-3 is also

expressed in mouse dorsal root ganglions and SLP-3 mutant mice have decreased

11

mechanosensitivity 53, similar to Mec-2 mutants and suggesting SLP-2 and its

homologues may be important in complex receptor assemblies.

Unc-1 was originally identified in C. elegans mutants with altered sensitivity to volatile

anesthetics, including an increase in sensitivity to the inhaled anesthetic diethyl ether 51.

Interestingly, this phenotype is conserved in the stomatin knockout mouse, as these mice

also show an increased sensitivity to diethyl ether 54. Taken together, studies of stomatin

family members in both C. elegans and mouse models have shown a role for this family

in ion channel regulation, yet the exact mechanisms remain elusive.

Stomatin family members have also been shown to localize to DI-GEMs and have been

proposed to organize these membranes into functional domains through associations with

actin microfilaments 55-57. Podocin is found in detergent-resistant membranes in

glomeruli 58, 59 and is thought to control intracellular distribution of proteins involved in

kidney ultrafiltration as HEK 293 cells co-expressing podocin and nephrin show co-

localization in DI-GEMs while cells lacking podocin fail to recruit nephrin to these

domains 60. Similarly, SLP-1 has also been shown to control the localization of stomatin 61, as cells over-expressing SLP-1 show an increase in recruitment of stomatin to the late

endosomal compartment, which is the subcellular localization of SLP-1. The localization

of these proteins to lipid domains is likely important for proper cellular function.

1.1.2 Stomatin-like Protein 2

SLP-2 was originally identified in a BLAST search of the EST database using the human

stomatin cDNA sequence 62. The 1071 nucleotide SLP-2 gene encodes a 357 amino acid

protein and is located on human chromosome 9p13. Sequence analysis indicates that

SLP-2 lacks a putative transmembrane domain and palmitoylation sites found in other

stomatin family members. However, SLP-2 does share sequence homology with the

other stomatin family members in the predicted C-terminal α-helical region and proximal

β-sheet domains. Similar to other stomatin family members, SLP-2 is also found in

12

Figure 1.3: Features of the SLP-2 amino acid sequence.

Alignment of human and mouse SLP-2 amino acid sequence, showing conservation of

sequence as well as putative post-translational modifications. The mitochondrial

targeting sequence, predicted by the Mitoprot program, is shown in the yellow shaded

sequences. The SPFH domain is shown in the underlined sequence, as predicted by

InterPro scan. Putative post-translational modifications of the SLP-2 protein, as

predicted by Prosite, are indicated by bold text and arrows above the sequence to denote

the type of modification. The stomatin family consensus sequence is indicated by *

above the sequence alignment, where the consensus sequence is defined as

RX2(L/I/V)(S/A/N)X6(L/I/V)DX2TX2WG(L/I/V)X(K/R)(L/I/V)E(L/I/V)(K/R) 62. The

conservation of sequence between mouse and human SLP-2 is indicated below the

sequence alignment, where * indicates a conservation, : indicates highly similar residues

and . indicates weakly similar residues. Abbreviations: myr: myristoylation site,

PKC: protein kinase C phosphorylation site, cAMP: cAMP and cGMP – dependent

protein kinase phosphorylation site, CKII: casein kinase II phosphorylation site, N-gly:

N-linked glycosylation site, h – human modification only, m – mouse modification only.

13

Figure 1.3

cAMP myr PKC myr hSLP-2 MLARAARGTGALLLRGSLLASGRAPRRASSGLPRNTVVLFVPQQEAWVVERMGRFHRILE 60 mSLP-2 MLARAARGTGALLLRGSVQASGRVPRRASSGLPRNTVILFVPQQEAWVVERMGRFHRILE 60

*****************: ****.*************:********************** PKC/CKII N-Gly hSLP-2 PGLNILIPVLDRIRYVQSLKEIVINVPEQSAVTLDNVTLQIDGVLYLRIMDPYKASYGVE 120 mSLP-2 PGLNVLIPVLDRIRYVQSLKEIVINVPEQSAVTLDNVTLQIDGVLYLRIMDPYKASYGVE 120 ****:******************************************************* N-Gly(h) PKC CKII(h)

* ** ** * ***** ***** hSLP-2 DPEYAVTQLAQTTMRSELGKLSLDKVFRERESLNASIVDAINQAADCWGIRCLRYEIKDI 180 mSLP-2 DPEYAVTQLAQTTMRSELGKLSLDKVFRERESLNANIVDAINQAADCWGIRCLRYEIKDI 180

***********************************.************************ cAMP/CKII myr amidation CKII hSLP-2 HVPPRVKESMQMQVEAERRKRATVLESEGTRESAINVAEGKKQAQILASEAEKAEQINQA 240 mSLP-2 HVPPRVKESMQMQVEAERRKRATVLESEGTRESAINVAEGKKQAQILASEAEKAEQINQA 240

************************************************************ CKII hSLP-2 AGEASAVLAKAKAKAEAIRILAAALTQHNGDAAASLTVAEQYVSAFSKLAKDSNTILLPS 300 mSLP-2 AGEASAVLAKAKAKAEAIRILAGALTQHNGDAAASLTVAEQYVSAFSKLAKDSNTVLLPS 300

**********************.********************************:**** myr myr PKC/CKII CKII hSLP-2 NPGDVTSMVAQAMGVYGALTKAPVPGTPDSLSSGSSRDVQGTDASLDEELDRVKMS 356 mSLP-2 NPSDVTSMVAQAMGVYGALTKAPVPGAQN--SSQSRRDVQATDTSI-EELGRVKLS 353

**.***********************: : ** * ****.**:*: ***.***:* N-Gly(m) CKII (m)

14

association with DI-GEM 63, which may be mediated by protein myristoylation or may

occur through the SPFH domain, in a similar manner to Mec-2 and podocin, both of

which have been shown to associate with cholesterol through the SPFH domain 26. As

shown in Fig. 1.3 human and mouse SLP-2 are highly conserved and share a number of

putative post-translational modifications, including possible myristoylation sites to

mediate membrane association. Furthermore, identification of multiple putative

phosphorylation sites indicates a possible role in cell signalling cascades and a

mitochondrial targeting sequence at the amino terminus of both SLP-2 sequences

indicates mitochondrial localization.

Northern blot analysis demonstrated that SLP-2 was widely expressed, including heart,

brain, lung, liver, placenta, pancreas, skeletal muscle and kidney, with the highest

expression levels found in heart, liver and pancreas. Intracellularly, SLP-2 is found

predominantly in mitochondria 64, 65. This subcellular localization is dictated by a

mitochondrial targeting sequence at the amino terminus of SLP-2, which is cleaved upon

transport into the mitochondria 64, 65. The mitochondrial localization of SLP-2 has been

confirmed in several studies investigating the mitochondrial proteome in a variety of

tissues, including mouse liver, brain, heart and kidney, as well as human heart and T

cells66-69. Furthermore, confocal imaging studies have shown full length SLP-2 localized

within mitochondria and fusion of SLP-2 residues 1-50 to a green fluorescent protein

(GFP) tag resulted in mitochondrial localization of GFP, whereas deletion of SLP-2

residues 1-50 eliminated the mitochondrial localization64, 65. SLP-2 is tightly associated

with the inner mitochondrial membrane, facing the intermembrane space 64, 65.

In the initial report of SLP-2 identification, SLP-2 was found in a high molecular weight

complex 62. Subsequent studies have shown SLP-2 to complex with mitochondrial

resident proteins mitofusin 2 and prohibitins 1 and 2, although the function of these

associations is currently unknown 64, 65. Knockdown of SLP-2 leads to a decrease in

mitochondrial transmembrane potential as well as a decrease in protein levels of complex

I and IV of the respiratory chain and decreased prohibitin 1 and 2 64, 65. The function of

SLP-2 is currently unknown.

15

1.1.3 Prohibitins

The SPFH-domain-containing proteins prohibitin (PHB)-1 and -2 are mitochondrial

proteins associated with the inner mitochondrial membrane 36, 70, 71. Both proteins contain

a hydrophobic region at the amino-terminus, which may facilitate the association with the

inner membrane 37. PHB-1 and PHB-2 hetero-oligomerize to form a large ring-like

complex and are dependent on co-expression for stability, as knockdown of PHB-1

results in the loss of PHB-2 and vice versa 29, 37, 70, 72. As such, PHB-1 and PHB-2 have

similar expression patterns and are widely expressed, although the expression levels vary

between tissues. Although studies have pointed to many roles for prohibitins, the

function of these proteins is currently unknown. While prohibitins have been implicated

in cellular proliferation, their expression levels do not correlate with the proliferative

state of the tissue 33,70. Prohibitins have also been linked to aging, as some senescent

cells show decreased PHB levels compared to young cells 70. Furthermore, prohibitin

knockdown during embryogenesis results in embryonic arrest in C. elegans 73. Prohibitin

knockdown after embryogenesis results in shortened life span in wild type animals, but

mutants unable to undergo diapause were protected by prohibitin deletion 32. Although

this result seems contradictory, it has been proposed that the loss of prohibitins initiates

signalling cascades that change cellular metabolism for fat utilization. In this way, wild

type animals that fail to undergo diapause cannot utilize fat stores for energy production,

resulting in decreased survival, while prohibitin loss in diapause mutants lead to fat store

utilization and increased survival.

Prohibitins associate with the mitochondrial AAA (m-AAA) protease and knockdown of

prohibitin leads to increased protein degradation by the protease, leading to the

suggestion that the prohibitin complex acts as a chaperone to prevent degradation of

functional proteins 29, 37. Moreover, prohibitins have also been shown to play a role in the

cleavage of the mitochondrial protein optic atrophy 1 (OPA-1), which is involved in

maintaining cristae structure 74. Deletion of PHB-2 in mouse embryonic fibroblasts led to

massive mitochondrial fragmentation along with loss of cristae or the formation of very

large cristae 33. These cells were found to have altered processing of OPA-1 with a loss

16

of the long forms. Over-expression of a long-form OPA-1 mutant that is not cleavable

led to recovery of cristae structure and less fragmentation of the mitochondrial network.

The prohibitin knockdown cells also showed increased susceptibility to apoptotic

induction, which was rescued by the mutant OPA-1. This is likely due to the role for

OPA-1 and cristae remodeling as an early step in cytochrome c release leading to the

induction of the intrinsic pathway of apoptosis 75. In this way, the prohibitin complex

plays a role in the sensitivity to apoptotic induction.

The mechanism of action for the prohibitin complex is still not understood. However, it

has recently been proposed that prohibitins may facilitate membrane

compartmentalization at the inner mitochondrial membrane 34. As this membrane has

extremely high protein content, proper functioning of these proteins may benefit from

segregation, in a way reminiscent of that described for T cell activation. Yeast cells

lacking prohibitins remain viable, but to further understand the function of these proteins,

synthetic lethal mutations were created to identify the genomic interactome of prohibitins 35. The loss of a number of non-essential genes were shown to be synthetic lethal in

combination with the loss of prohibitins, including genes involved in the assembly of

respiratory chain components and in the control of mitochondrial morphology. Two new

genes were identified in this screen, one required for phosphatidylethanolamine

biosynthesis and the other for cardiolipin biosynthesis, two lipids found at the inner

mitochondrial membrane. Cardiolipin is unique to mitochondrial membranes, and as

such, has been shown to be important for various mitochondrial functions 76. Cardiolipin

interacts with many mitochondrial proteins, including components of the electron

transport chain and mitochondrial translocases, and this interaction is required for optimal

activity of these complexes 76-78. In addition, there is evidence that cardiolipin is required

for the maintenance of mitochondrial membrane structure, which is essential for

oxidative phosphorylation 79, 80. The ring-like prohibitin complex has been proposed to

organize cardiolipin, and perhaps other proteins, into domains to facilitate various

mitochondrial functions. In the absence of prohibitins, these domains would not be

formed and increased cardiolipin biosynthesis may be required to maintain proper

mitochondrial functions 34, 35. The importance of mitochondrial membrane organization

will be discussed in the context of the major mitochondrial functions, including

17

bioenergetics, calcium signalling and apoptosis and in the importance of these functions

for T cell activation and function.

1.2 Mitochondrial functions

1.2.1 Bioenergetics and metabolism

Energy production is the predominant function associated with mitochondria as the

electron transport chain resides at the inner membrane and is responsible for producing

the majority of cellular ATP. Mitochondria have separate inner and outer membranes,

each with a unique lipid and protein composition. The inner membrane is enriched in the

lipid cardiolipin, which has been linked to the organization of the membrane into folded

cristae. This, in turn, gives a much larger membrane surface area in the small organelle,

which allows for a higher concentration of the protein complexes of the electron transport

chain. Glycolysis and the tricarboxylic acid (TCA) cycle break down glucose into the

substrates oxidized by the electron transport chain. Glucose is first metabolized to two

pyruvate molecules, two ATP and two NADH (nicotinamide dinucleotide) molecules

during glycolysis. Acetyl groups from pyruvate are then transferred to coenyzme A

(CoA), with the generation of one NADH molecule and this marks the entry into the

TCA. The oxidation of fatty acids by β-oxidation also generates acetyl-CoA, which can

enter the TCA cycle to generate ATP from the respiratory chain, which occurs under

conditions of limiting glucose. The enzymes of the TCA cycle are located within the

mitochondrial matrix and this cycle gives rise to three NADH molecules, one FADH2

(flavin adenine dinucleotide) molecule and one ATP. The NADH and FADH2 molecules

carry high-energy electrons, which are used to drive ATP production through the electron

transport chain. The principle behind this relies on the generation of energy as electrons

are transferred from carriers with low reduction potential to donors with high reduction

potential 81. The electron transport chain transfers electrons between a series of electron

acceptors with successively higher reduction potential. The energy released by these

transfers is harnessed to shuttle hydrogen ions across the inner membrane, resulting in a

net negative charge in the matrix. This gives rise to a transmembrane potential and

18

Figure 1.4: Schematic overview of the respiratory chain.

The components of the respiratory chain in association with the inner mitochondrial

membrane are shown along with the flow of electrons and hydrogen ions through the

complexes. NADH dehydrogenase and succinate dehydrogenase represent the entry

point for electrons into the respiratory chain at complex I and complex II, respectively.

Electrons are transferred through a series of electron acceptors associated with complex

III and IV and are ultimately transferred to oxygen to form water as an end product. At

complex I, III and IV, hydrogen ions are transported across the inner membrane into the

intermembrane space, resulting in a net negative charge in the mitochondrial matrix.

ATP synthase uses the energy released by hydrogen ions moving back into the matrix to

synthesis ATP from ADP. This figure is adapted from Rich and Marechal, 2010 81.

19

Figure 1.4

Intermembrane Space (+)

Inner Mitochondrial Membrane

Matrix (-)

NADH +H+

NAD+ Succinate Fumarate

UQ UQ

UQH2 UQH2

2H+ 2H+

Cytochrome c

½ O2 + 2H+

H2O ADP + Pi

ATP

4H+ 4H+ 2H+ nH+

Complex I NADH

Dehydrogenase

Complex II Succinate

Dehydrogenase

Complex III Cytochrome bc1

Complex IV Cytochrome c

Oxidase

Complex V ATP

Synthase

20

pH gradient across the inner membrane. The double membrane structure allows for two

distinct regions within the mitochondria, which allows this gradient to exist separately

from the cytosol. The transmembrane potential is exploited for ATP synthase, which

uses the energy released by transport of hydrogen ions along the potential gradient to

drive the phosphorylation of ADP to ATP.

The respiratory chain is composed of four molecular complexes located within the inner

mitochondrial membrane (Fig. 1.4). Complex I, NADH dehydrogenase, is a complex

formed by as many as 45 subunits and transfers electrons from NADH to a series of

cofactors resulting in the reduction of ubiquinone (UQ). Complex I is the largest of the

respiratory complexes and seven of the core hydrophobic subunits are encoded by the

mitochondrial DNA 81. Complex II, succinate dehydrogenase, is an enzyme of the TCA

cycle and converts succinate to fumarate with the transfer of two electrons to FAD to

form FADH2. FADH2 is then oxidized by the transfer of these electrons through a

number of cofactors to UQ. Complex I and II represent the entry point of electrons into

the respiratory chain. UQH2 is a mobile electron carrier that can freely diffuse in the

membrane to Complex III, cytochrome bc1, where the electrons are transferred from

UQH2 to a number of cofactors and then to cytochrome c. Cytochrome c is another

mobile electron carrier that diffuses through the inner membrane to Complex IV,

Cytochrome c oxidase, which again transfers electrons through a series of cofactors to the

final electron acceptor, O2 forming H2O, the final product of the respiratory chain. ATP

synthase forms a pore in the inner membrane through which H+ is transported back into

the matrix. The energy released drives the phosphorylation of ADP to ATP.

Complex I, III and IV span the inner membrane and use the energy released from electron

transfer to transport hydrogen ions across the inner membrane into the inter-membrane

space. Although these complexes are often described as separate complexes, recent work

has indicated that the complexes associate with one another, forming super complexes 82.

These complexes include I/III and II/III/IV and are quite prevalent as most complex I and

complex III in mouse liver mitochondria are found in super complexes. Some of these

complexes also contain the mobile electron carriers UQ and cytochrome c and together

this organization is thought to increase the efficiency of electron transfer between the

21

complexes, increasing the ATP production. To facilitate super complex formation and

efficient electron transfer, the components of the respiratory chain may be sequestered

into defined domains within the mitochondrial inner membrane. At the cell surface,

cholesterol is a major component involved in micro-domain formation but the cholesterol

content in the mitochondrial membrane is low. However, the mitochondrial lipid

cardiolipin may perform a similar role in membrane organization, as this lipid is

important for the proper function of the components of the electron transport chain and

super complexes are not stable in the absence of cardiolipin 77, 78, 83, 84. Furthermore,

cardiolipin has a unique structure that can alter membrane structure and curvature, which

may facilitate micro-domain formation and has been shown to form clusters in the

mitochondrial membrane 85, 86. Through the organization of the inner mitochondrial

membrane into cardiolipin-enriched domains, super complex formation may be favored,

leading to more efficient electron transport, giving rise to increased ATP production.

Studies altering the expression levels of SLP-2 indicate a role for SLP-2 in regulating the

respiratory chain. In cells treated with siRNA to knock down SLP-2 expression levels,

there is a slight decrease in the mitochondrial transmembrane potential and lower ATP

production 65, 87. Furthermore, SLP-2 expression is increased upon cellular stress, as is

ATP production, whereas knocking down SLP-2 expression before inducing cellular

stress inhibits this increase in ATP production 88. Thus, SLP-2 appears to play a role in

regulating the respiratory chain and ATP production. As SLP-2 has been shown to

interact with the prohibitin complex 34, 64, this regulation may be related to the role

proposed for prohibitins in the mitochondrial membrane compartmentalization and

respiratory chain function.

1.2.2 The metabolic regulation of T cells

The regulation of ATP production plays an important role in T cell activation as this is an

anabolic process requiring energy for cellular growth and function 89. As such, T cell

activation is tightly linked to metabolism, as co-stimulation of T cells through the CD28

receptor increases glycolysis while decreasing the requirement for ATP production

22

through the respiratory chain 90. Although there is much more ATP produced per glucose

molecule if the products of glycolysis enter the respiratory chain, these products can also

provide the building blocks for amino acid, nucleotide and lipid biosynthesis, all of which

are needed for cellular proliferation and function 91. Thus, under conditions of unlimited

glucose supply, T cells fulfill energy requirements as well as biosynthetic requirements

by relying on high throughput glycolysis.

The importance of metabolic regulation during T cell activation has recently been shown

in both CD4+ and CD8+ T cells. Upon CD28 stimulation, the PI3K/Akt pathway is

activated and leads to the activation of the mammalian target of rapamycin (mTOR), a

serine/threonine kinase involved in activating protein synthesis. The generation of T cell-

specific mTOR-deficient mice showed a failure to differentiate into TH1, TH2 or TH17

with increased differentiation into Tregs 92. Subsequently, an investigation into the

metabolic profiles of these cell types demonstrated that TH1, TH2 and TH17 cells all had

significantly higher glycolytic rates than Treg cells, while Treg cells had significantly

higher rates of β-oxidation, which relies on the respiratory chain for ATP production 93.

Treating cells with an inhibitor of β-oxidation led to a decrease in Treg cell counts, while

treatment with fatty acids to promote β-oxidation decreased TH1/2/17 cell numbers and

function and increased Treg function. Finally, treatment with the AMP-activated kinase

(AMPK) activator metformin led to a decrease in total CD4+ T cell populations while

increasing the Treg population. AMPK is activated in response to an increase in the ratio

of AMP to ATP and acts to inhibit protein synthesis through the inhibition of mTOR. It

also activates β-oxidation with the overall effect of decreasing ATP consumption 94.

The control of the mTOR pathway and β-oxidation has also been shown to play an

essential role in the development of CD8+ memory T cells 95, 96. Treatment of mice with

rapamycin, an inhibitor of mTOR, results in an increase in CD8+ T cell memory

responses to LCMV infection 96. Finally, TNF-receptor associated factor 6 (TRAF6)-

deficient mice with a defect in the expression of genes involved in β-oxidation were

found to have severely decreased CD8+ memory T cell formation 95. These mice also

showed low rates of β-oxidation and treatment with metformin or rapamycin to induce β-

oxidation led to a recovery of CD8+ memory T cells. Taken together, these results

23

demonstrate the importance of metabolic regulation in the generation of both CD4+ and

CD8+ T cell subsets, where glycolysis and β-oxidation play opposing roles in the T cell

subsets.

1.2.3 Calcium signalling

In addition to ATP synthesis, mitochondrial membrane potential also drives the uptake of

cytosolic calcium 97, 98. The mitochondrial outer membrane is relatively permeable to

ions, while the inner membrane is impermeable, an essential feature for the respiratory

chain. The inner membrane has a number of ion transporters, including a calcium

uniporter that transports calcium into the matrix 99. Calcium is exported from the matrix

by the sodium/calcium antiporter. The rate of Ca2+ uptake by the Ca2+ uniporter is

controlled by cytosolic calcium levels and transport is increased in response to high

cytosolic calcium, while the Na+/Ca2+ antiporter is regulated by free matrix Ca2+ levels.

In the matrix, calcium binds to phosphate and prevents the activation of the antiporter and

allows the mitochondria to sequester calcium from the cytosol in response to cytosolic

calcium spikes. Mitochondrial calcium activates a number of enzymes and leads to

increased ATP production, but prolonged elevated mitochondrial calcium levels lead to

apoptosis 100.

The ability of mitochondria to buffer calcium signals is of special importance in T cells

where stimulation triggers cytosolic Ca2+ elevations required for NFAT activation 101-103.

TCR signalling through the PLCγ pathway leads to the generation of IP3, which binds to

the IP3 receptor on the endoplasmic reticulum membrane to trigger the release of

intracellular Ca2+ stores. Upon depletion of the intracellular Ca2+ stores, the ER resident

protein stromal interaction molecule 1 (STIM1) then triggers the activation of the calcium

release-activated calcium (CRAC) channels at the cell surface, further increasing the

cytosolic Ca2+ levels 104, 105. Once activated, the CRAC channels are sensitive to

cytosolic calcium levels with elevated levels leading to channel inactivation 97,106. Thus,

in order to achieve elevated cytosolic Ca2+ levels, local Ca2+ levels must remain low.

Recent studies have demonstrated that mitochondrial calcium uptake plays an important

24

role in maintaining CRAC-dependent calcium uptake 107. Mitochondria were shown to

migrate to the plasma membrane upon thapsigargin-triggered ER calcium release and the

inhibition of this migration led to a rapid inactivation of CRAC channels. In addition, T

cell activation through the TCR triggers mitochondrial translocation to the

immunological synapse and the inhibition of this translocation led to decreased

intracellular Ca2+ levels 108. Thus, mitochondria are thought to act as a localized calcium

sink necessary to shuttle calcium away from CRAC channels to allow a sustained

calcium influx required for T cell activation. In this way, the organization of

mitochondrial membranes to facilitate supercomplex formation may also lead to

increased calcium buffering capacity as the increased transmembrane potential would

drive more calcium into the mitochondria, away from the CRAC channels, resulting in

prolonged calcium signalling.

To investigate the role of SLP-2 in mitochondrial calcium buffering Da Cruz et al treated

HeLa cells with shRNA to knock down SLP-2 levels 109. Cells expressing lower levels of

SLP-2 took up less calcium in response to histamine treatment and had a higher rate of

calcium efflux after stimulation. Treatment of these cells with an inhibitor of the

mitochondrial sodium/calcium exchanger normalized the calcium efflux rate between

control and knockdown cells, whereas the initial peak of calcium response remained

lower in the knockdown cells. This difference in calcium uptake was not due to a

difference in cellular calcium concentration as permeabilized cells also showed a

decreased mitochondrial calcium uptake and increased efflux rate in SLP-2 knockdown

cells. Likewise, in cells over-expressing SLP-2, the initial calcium uptake was slightly

higher compared to wild type cells and efflux rate was reduced, resulting in a prolonged

calcium signal. This role for SLP-2 in regulating mitochondrial calcium buffering

capacity is likely involved in the regulation of T cell responses, as Jurkat T cell activation

level is directly proportional to SLP-2 expression level 63.

25

1.2.4 Apoptosis

Mitochondria are not only involved in T cell activation but also in the regulation of T cell

responses by inducing cell death. Apoptosis is a form of controlled cell death induced in

response to extracellular signals or cellular stress, such as DNA damage or growth factor

withdrawal 110. The activation of the apoptotic pathway leads to chromatin condensation

and DNA fragmentation, cell shrinkage and membrane blebbing, ultimately reducing

cells to small apoptotic bodies that can be engulfed by macrophages to prevent

inflammatory responses. Apoptosis is essential for multi-cellular organisms to eliminate

damaged or unneeded cells. In T cell development, the failure of cells to succeed through

pre-TCR β selection, positive or negative selection leads to apoptosis and elimination of

TCR-defective or auto-reactive T cells 111. Moreover, apoptosis is also required after a

pathogen has been cleared by the immune response. In this case, T cells specific for the

pathogen have undergone massive proliferation and activation. While some will survive

as memory cells, most cells will undergo activation-induced cell death. In this way, the

body returns to homeostasis and reduces possible immunopathological effects of

activated T cells.

The main effectors of apoptosis are the caspases, a family of asparagine-proteases that

cleave a wide variety of structural and regulatory proteins 112. Capsases are composed of

two main subfamilies, in which capase 1 subfamily members are involved in pro-

inflammatory responses through the cleavage and activation of IL-1 and IL-18, while

caspase 3 subfamily members, including caspase 3 and 6-10 are involved in apoptosis 113,

114. Caspases exist as zymogens in healthy cells, but apoptotic stimuli induce signalling

cascades that result in procaspase homo-oligomerization, auto-cleavage and activation.

The activated caspases then cleave and activate additional caspases, leading to wide

spread protein degradation including complex I subunit NDUFS1, which in turn, leads to

the loss of the transmembrane potential. Caspases also activate a number of additional

apoptotic effectors, including DNA fragmentation factor 45 and pro-apoptotic kinases

involved in membrane blebbing and apoptotic body formation 110.

Mitochondria are important regulators of apoptosis and represent the convergence of both

extrinsic and intrinsic apoptosis pathways 110. The extrinsic pathway is activated by

26

extracellular signals, in response to ligation of cell surface death receptors, such as Fas or

TNF related apoptosis-inducing ligand (TRAIL), which induce a signal cascade resulting

in the activation of caspase 8 115. The intrinsic pathway is activated by intracellular

signals that result in the release of cytochrome c and direct IAP binding protein with low

pI (DIABLO) from the mitochondrial inner membrane space, allowing the formation of

the apoptosome, composed of Apaf-1, cytochrome c and procaspase 9 115.

Oligomerization of these components result in the activation of caspase 9, another

activator that initiates the caspase cascade 110. DIABLO inhibits the inhibitor of

apoptosis proteins (IAPs), which can bind to and inactivate various caspases 116.

The B cell lymphoma 2 (Bcl-2) family of proteins is responsible for sensing cellular

stress signals to induce the activation of the intrinsic pathway 116. This family consists of

both pro-apoptotic and anti-apoptotic family members, and cell signalling in normal

conditions or under cell stress tips the balance in favor of pro- or anti-apoptotic

responses. Under stress conditions, the pro-apoptotic members Bak and Bax oligomerize

and insert into the mitochondrial outer membrane, forming a pore that causes cytochrome

c release. There are currently two models explaining the control of Bax and Bak in the

transition from healthy to apoptotic conditions. In the activation model, Bax and Bak are

thought to be inactive in healthy cells, but apoptotic signals lead to the activation of pro-

apoptotic family members BIM, tBID and PUMA, which in turn, activate Bax and Bak

and lead to membrane insertion and cytochrome c release. In the neutralizing model, Bax

and Bak are thought to be constitutively active, but prevented from pore formation by the

association with the anti-apoptotic family members, including Bcl-2. Apoptotic signals

upregulate or release multiple pro-apoptotic family members from confined cellular

locations, and allow these proteins to bind the anti-apoptotic members, releasing Bax and

Bak from Bcl-2, to permit pore formation. Mitochondria link the extrinsic pathway to the

induction of the intrinsic pathway via caspase 8, as one target of caspase 8 is the pro-

apoptotic protein Bid, which upon cleavage, associates with the mitochondrial membrane

to activate Bax/Bak pore formation to trigger cytochrome c release 110, 117. In this way,

mitochondria play a role in both intrinsic and extrinsic apoptosis.

27

Apoptosis is also tightly coupled to changes in mitochondrial morphology. Although

mitochondria have traditionally been thought of as isolated organelles, they have been

shown to be a highly dynamic population undergoing fusion and fission events, which

affect a wide variety of cellular functions 118-120. Mitochondrial fusion is mediated by the

GTPases mitofusin (Mfn) 1 and 2 and OPA-1. Mfn1 and 2 facilitate outer membrane

fusion by undergoing homo- or hetero-oligomerizion between adjacent mitochondria.

The intermembrane space resident OPA-1 facilitates inner membrane fusion, although the

mechanism is currently unclear 121. Fission is carried out by the GTPase Drp1, which

oligomerizes into a ring-like structure around the mitochondrial outer membrane at the

site of the fission event. Drp1 is a cytosolic protein with a small pool localized to the

mitochondrial membrane in a punctate pattern. The recruitment of Drp1 is thought to be

facilitated by the outer membrane protein Fis1, although the outer membrane protein

mitochondrial fission factor (Mff) has also been proposed to recruit Drp1 to fission sites 122-124. The importance of mitochondrial dynamics has been shown in development as

Drp1, Mfn1, Mfn2 and OPA-1 deletions are all embryonic lethal 125-128. Furthermore,

fusion is important for content sharing of the mitochondrial network, including the

replacement of damaged mitochondrial DNA (mtDNA). Indeed, in fusion-deficient cells,

mtDNA is lost 129. Finally, fusion and fission events are also regulated during the cell

cycle, with hyperfusion occurring as cells transition from G1 to S phase, while the mitotic

kinase Cdk1/cyclin B phosphorylates Drp1 to promote fission 130, 131. In this way,

contents of the mitochondrial network are shared to replace damaged components before

splitting and dividing into the daughter cells.

Changes in the expression levels of the fusion or fission components have been shown to

play a role in apoptosis. Mitochondrial fission, facilitated by Drp1 recruitment, is an

early apoptotic event 132, 133. Inhibition of Fis1 or Drp1 or over-expression of OPA-1

inhibits fission, which is protective against apoptosis 133, 134. In addition, the loss of

transmembrane potential that accompanies apoptosis also results in elevated OPA-1

cleavage, which then inactivates OPA-1 and decreases mitochondrial fusion, leading to

an imbalance of mitochondrial dynamics in favor of mitochondrial fission 74. The

components of fusion and fission machinery also interact with members of the Bcl-2

family of apoptotic proteins to regulate apoptosis. The pro-apoptotic Bax co-localizes

28

with Drp1 and Mfn2 upon apoptotic induction and the knockdown of Fis1 inhibits the

recruitment, whereas knockdown of both Drp1 and Fis1 inhibit mitochondrial fission to

protect against apoptosis 135, 136.

SLP-2 may play a role in regulating mitochondrial fusion upon cellular stress to protect

against apoptotic induction 88. Mitochondrial stress induced by chloramphenicol

treatment led to the specific upregulation of SLP-2 64. Interestingly, mouse embryonic

fibroblasts (MEF) subjected to stress stimuli such as UV irradiation, actinomycin D or

cycloheximide show mitochondrial hyperfusion, resulting in elongated mitochondria 88.

Deletion of OPA-1 and Mfn1, both required for fusion, abrogated this response, as did

SLP-2 knockdown. This loss of hyperfusion appears to be due to altered OPA-1

processing as SLP-2 deficient cells lack the longer isoforms of OPA-1. Transfecting cells

with a non-cleavable mutant OPA-1 protein restored mitochondrial hyperfusion.

Treatment of SLP-2 knockdown cells with a metalloprotease inhibitor prevented the loss

of long form OPA-1, indicating a possible role for SLP-2 in protease regulation. This is

supported by an earlier study in which metalloprotease inhibition restored prohibitin and

respiratory chain component loss upon SLP-2 knockdown 64, 88. Although prohibitins

control OPA-1 processing 33 and are known to associate with SLP-2, cells lacking

prohibitin 1 and 2 show similar hyperfusion to wild type cells upon cycloheximide

treatment 88 indicating these hyperfusion events are independent of the prohibitin-SLP-2

complex. Upon hyperfusion, cells have increased ATP production compared to untreated

cells, while mutant cells unable to undergo hyperfusion (OPA-1-/- or shSLP-2) do not

show increased ATP production. Hyperfusion appears to be protective against apoptosis

as cells lacking Mfn1 or SLP-2 had increased apoptosis in response to actinomycin D

treatment or UV irradiation.

Membrane organization in the mitochondria may also play a role in apoptosis. As

discussed above, there are many players involved in the induction of apoptosis, including

the pro- and anti-apoptotic members of the Bcl family, the members of the caspase

cascade as well as the proteins involved in mitochondrial membrane dynamics 110, 116, 133.

The organization of the mitochondrial membrane into specialized microdomains would

facilitate the interaction of these components to induce apoptosis, leading to membrane

29

permeabilization, cytochrome c release and caspase activation in response to apoptotic

signals. Apoptotic induction via anti-Fas treatment led to a re-organization of pro-

apoptotic Bax and truncated Bid (tBid) from detergent-soluble to detergent-insoluble

domains isolated from whole cells 137. These results demonstrate the re-organization of

apoptotic components into membrane microdomains during apoptotic responses. Further

characterization of these mitochondrial detergent-insoluble membranes demonstrated an

enrichment in cardiolipin whereby all cardiolipin was found in the detergent-insoluble

domains and none in the detergent-soluble domains 138.

The enrichment of cardiolipin in these detergent-insoluble domains likely plays an

important functional role in the induction of apoptosis as cardiolipin can bind to many

components of the apoptotic pathway, including cytochrome c, tBid and caspase 8 139-142.

The pro-apoptotic protein tBid may also play a role in the organization of membranes

into cardiolipin-enriched detergent-insoluble domains as tBid binds to cardiolipin

monolayers and induces re-organization of the cardiolipin into regions of very tightly

packed lipid 143. This binding of tBid to cardiolipin is an essential step in apoptotic

induction as incubation of recombinant tBid with isolated mitochondria leads to

cytochrome c release, yet the addition of excess free cardiolipin prevents this binding and

leads to a decrease in cytochrome c release 141. Furthermore, apoptotic induction results

in the accumulation of many of the proteins known to associate with cardiolipin into

these cardiolipin-enriched domains, including activated caspase 8, Bak, tBid and DLP1 138, 144. Interestingly, the fission protein hFis1 and the fusion protein OPA-1 are

constitutively localized to the cardiolipin-enriched membrane domains, but DLP1 is

recruited upon apoptotic induction, indicating the coordination of apoptosis and

mitochondrial fission 144. The importance of these domains is demonstrated in cells

lacking cardiolipin, either by knockdown of cardiolipin synthase or tafazzin, the

remodeling enzyme involved in the final steps of cardiolipin synthesis, as these cells

exhibit an increased resistance to apoptotic induction 142, 145.

30

1.3 Rationale and Hypothesis

1.3.1 Rationale

In light of the important role played by cell membrane organization at both the plasma

and mitochondrial membranes, initial experiments were performed to identify novel

players involved in membrane organization during T cell activation. To this end, Jurkat

T cells were activated through their T cell receptor and DI-GEMs were isolated. Proteins

were extracted from these fractions and subjected to mass spectrometry 63. In addition to

proteins known to be involved in T cell activation, previous work in the Madrenas

laboratory also identified a novel protein, stomatin-like protein 2 (SLP-2), which is a

member of the highly conserved stomatin family. As discussed above, members of this

family have been proposed to facilitate membrane microdomain formation and

organization, 55-57 although the function of SLP-2 remains unclear.

Preliminary experiments were performed to investigate the role of SLP-2 in the immune

response in general and in T cell activation in particular. Immunohistochemistry and

immunoblot analysis demonstrated SLP-2 expression in a number of human lymphoid

tissues, including lymph nodes, thymus, and tonsils, as well as in peripheral blood

mononuclear cells, including T cells, B cells and monocytes 63. SLP-2 expression was

also detected in areas of lymphocyte activation, such as germinal centers. Although the

expression of SLP-2 was low in naïve T cells, stimulation of these cells with

Staphylococcal enterotoxin E (SEE) increased the expression of SLP-2, pointing to a role

during T cell activation. Furthermore, SLP-2 was also most highly expressed in activated

and memory B cells, with much lower expression in naïve B cells, demonstrating a

widespread role for SLP-2 during adaptive immune responses.

To investigate the role of SLP-2 in membrane organization, DI-GEM fractions were

collected from both Jurkat T cells and primed peripheral blood mononuclear cells at

various time points during T cell stimulation with SEE and analyzed for SLP-2 levels.

Under resting conditions, a small pool of SLP-2 was associated with the detergent-

insoluble fraction and SEE stimulation increased the recruitment of SLP-2 to these

domains in a time-dependent manner, peaking at five minutes after activation. These

31

microdomains are likely important for T cell activation as membrane organization has

long been hypothesized to play an important role in T cell signalling 1, 2 and as such, the

association of SLP-2 with T cell signalling components was investigated. SLP-2 was

found to be associated with a number of T cell signalosome components, including

CD3ε, Lck, ZAP-70, LAT and PLC-γ1. SLP-2 was also shown to associate with actin,

another important player in membrane domain organization 20, and the inhibition of actin

polymerization decreased T cell activation upon SEE stimulation. The functional

importance of these associations was demonstrated by SLP-2 siRNA knockdown studies,

in which SLP-2 depletion resulted in decreased duration of ERK and PLC-γ1

phosphorylation, indicative of a shortened duration of T cell signalling. This decrease in

T cell signalling upon SLP-2 depletion led to decreased T cell activation, shown by lower

IL-2 production in response to SEE stimulation. The loss of SLP-2 had a greater impact

on primed T cells, whereas knockdown of SLP-2 in naïve T cells had a negligible effect

on activation, as these cells have very low SLP-2 expression. However, in primed cells,

which have upregulated SLP-2, the loss of SLP-2 led to a significant decrease in T cell

activation. Furthermore, increasing SLP-2 expression by transfection of Jurkat T cells

with a doxycycline-inducible SLP-2-GFP construct resulted in increased T cell activation

in response to SEE stimulation. Taken together, these results point to an important role

for SLP-2 in T cell activation, potentially through the organization of membrane domains

into specialized regions for T cell signalling.

1.3.2 Hypothesis

These preliminary data document a role for SLP-2 in T cell signalling and activation as

both over-expression and knockdown of SLP-2 affects T cell responses. The importance

of SLP-2 during T cell activation is further supported by the upregulation of SLP-2

expression in response to T cell stimulation. However, this work focused primarily on

events at the plasma membrane, yet SLP-2 is found predominantly at the mitochondrial

inner membrane 64, 65. In line with the proposed roles of other stomatin and SPFH family

members in membrane organization, and given the mitochondrial localization of SLP-2, I

hypothesize that SLP-2 acts to facilitate mitochondrial and plasma membrane

32

organization in response to T cell activation. In order to address this hypothesis, I

investigated the function of the mitochondrial pool of SLP-2 by examining mitochondrial

membrane composition as well as mitochondrial biogenesis and function in response to

SLP-2 over-expression. I also generated T cell-specific SLP-2 conditional knockout mice

and examined the activation and effector functions of SLP-2-deficient T cells to further

characterize the role of SLP-2 in mitochondrial membrane organization and function as

well as T cell activation. Finally, although the majority of SLP-2 is localized to the

mitochondria, there appears to be a small pool at the plasma membrane, as indicated by

the association of SLP-2 with the T cell signalosome. This plasma membrane pool of

SLP-2 thus was further investigated in resting T cells and in response to T cell

stimulation and the re-organization of both surface and mitochondrial SLP-2 pools in

response to T cell stimulation was be examined.

1.3.3 Specific Aim 1: To characterize the function of the mitochondrial pool of SLP-2

To further investigate the function of the mitochondrial pool of SLP-2, Jurkat T cells

were stably transfected with a doxycycline-inducible SLP-2 construct, tagged with GFP

at the C-terminus. When SLP-2 expression was induced in these cells, there was an

increase in mitochondrial populations, as shown by increased staining with the

mitochondrial-specific dye MitoTracker Red measured by both flow cytometry and

confocal microscopy. Furthermore, these cells also had increased mitochondrial counts

per cell slice as shown by transmission electron microscopy. To further investigate this

increase, the pathways required for mitochondrial biogenesis, including nuclear

transcription of mitochondrial-targeted genes, replication of mitochondrial DNA and lipid

biosynthesis were examined. SLP-2 over-expressing cells were found to have increased

PGC-1α transcript levels, indicative of activated nuclear transcription programs. In

addition, mitochondrial DNA levels were also elevated in the SLP-2 over-expressing

cells, as was cardiolipin biosynthesis. Taken together, these results point to an increase

in mitochondrial biogenesis in response to SLP-2 over-expression.

33

The mechanism of SLP-2-induced mitochondrial biogenesis is currently unclear.

However, it was hypothesized that SLP-2 organizes the mitochondrial membrane into

specialized domains to control mitochondrial function and biogenesis. As SLP-2 was

shown to interact with both the prohibitin complex and with cardiolipin in a dose-

dependent manner, we propose that SLP-2 may act as a bridge to facilitate the association

of the prohibitin complex with the mitochondrial inner membrane. In support of this

model, over-expression of SLP-2 increased the recruitment of prohibitins to

mitochondrial membranes. SLP-2 over-expressing cells also demonstrated increased

enzymatic activities of components of the citric acid cycle and the electron transport

chain, including citrate synthase, NADH dehydrogenase and succinate dehydrogenase.

As a result, SLP-2 over-expressing cells had increased ATP production and oxygen

consumption, indicative of enhanced activity of the respiratory chain. Furthermore, SLP-

2 over-expression was protective against induction of the intrinsic apoptotic pathway.

Taken together, we propose that SLP-2 facilitates mitochondrial membrane organization

through association with the prohibitin complex, which in turn, increases mitochondrial

biogenesis and function.

1.3.4 Specific Aim 2: To generate and characterize a T cell-specific SLP-2 conditional knockout mouse

To further investigate the function of SLP-2 in vivo, conventional genome-wide SLP-2

knockout mice were generated (C. Lemke and J. Madrenas, unpublished results).

However, the loss of SLP-2 expression resulted in embryonic lethality at the pre-

implantation stage, indicating an essential role for SLP-2 in early embryonic

development. Therefore, a conditional SLP-2 knockout was created using the Cre-lox

system. As preliminary work focused on the role of SLP-2 in T cells, mice expressing

Cre-recombinase under the control of the CD4 promoter were utilized to initiate genomic

deletion at the double-positive stage of T cell development. This resulted in mice with

genomic SLP-2 deletion in all double positive αβ T cells. Thus, all mature peripheral

CD4+ and CD8+ T cells lack expression of SLP-2 and the mice are termed SLP-2 T-K/O.

34

SLP-2 T-K/O mice showed grossly normal T cell development compared to control mice.

The total number of thymocytes, splenocytes and lymph node cells were similar between

control and SLP-2 T-K/O mice, as was double negative, double positive and single

positive thymic populations, as well as non T cells and both CD4+ and CD8+ T cells in

the spleen and lymph nodes. Confirming earlier results with human T cells, murine SLP-

2 T-K/O cells also showed decreased T cell activation ex vivo in response to a number of

different T cell stimuli, with the defect being more pronounced in primed SLP-2 T-K/O

cells. This defect in T cell activation was shown to be a post-transcriptional block in IL-2

production. However, analysis of the hallmark cytokines showed normal differentiation

into TH1, TH2, and TH17 subsets upon SEE stimulation in both naïve and primed SLP-2

T-K/O cells. Analysis of FoxP3+ staining also showed similar Treg populations in both

control and SLP-2 T-K/O mice.

In demonstrating the biological significance of these ex vivo studies, SLP-2 T-K/O mice

exhibited impaired responses to in vivo T cell challenge. SLP-2 T-K/O mice showed

decreased recall responses to OVA immunization and were also slower to reject an

allograft from a non-matched donor. In agreement with the decrease in T cell responses,

the SLP-2 T-K/O mice also showed a small but significant reduction in CD4+ T cell

populations, which did not appear to be due to apoptosis. The decreased T cell activation

may lead to less T cell proliferation in vivo, resulting in lower T cell populations and

decreased T cell responses.

As previous work with SLP-2 over-expression in the Jurkat system indicated a role for

SLP-2 in membrane organization, detergent-resistant glycolipid-enriched microdomains

were investigated in SLP-2 T-K/O mice. Although there was no significant difference in

total cardiolipin levels in these mice, there was significantly less cardiolipin in the

detergent-insoluble mitochondrial membrane fractions, accompanied by a decrease in

associated prohibitin 1, although total prohibitin levels were similar to control cells. In

addition, a number of subunits of complex I of the respiratory chain were also found to be

decreased in SLP-2 T-K/O cells, including the NDU FA9, NDU FS3 and NDU FB8

subunits, while there was no change in the levels of the other complexes of the

respiratory chain. The SLP-2 T-K/O cells also had a significant reduction in the activity

35

of complex I, suggesting a functional outcome of the loss of complex I subunits. The

defect in respiratory chain function was extended to complex II + III, which also showed

decreased activity in SLP-2 T-K/O cells. Together, these results point to an altered

mitochondrial membrane organization, which may lead to decreased complex I assembly

and increased degradation of complex I subunits, ultimately resulting in decreased

complex I activity and decreased T cell responses in SLP-2-deficient T cells.

1.3.5 Specific Aim 3: To characterize the plasma membrane-associated pool of SLP-2

Although SLP-2 has been shown to localize predominantly to the mitochondria, it has

also been detected in detergent-insoluble glycosphingolipid-enriched membrane fractions

isolated from whole T cells, which include many surface proteins. To confirm the

existence of a second pool of SLP-2 at the plasma membrane and to further investigate

the function of this pool, Jurkat T cells stably transfected with doxycycline-inducible

SLP-2-GFP were utilized for SLP-2 over-expression and confocal imaging studies. In

addition to full length SLP-2-GFP, a deletion construct lacking the amino-terminal

region, which includes the mitochondrial targeting sequence and the putative

myristoylation sites, was also examined.

In agreement with the results found in specific aim 1, both endogenous SLP-2 and SLP-

2-GFP localized to the mitochondria. As expected, the truncation mutant lacking the

mitochondrial-targeting domain failed to localize to mitochondria and instead showed a

diffuse cytosolic localization. Analysis of the plasma membrane-associated pool

demonstrated the presence of both full length and the truncation mutant of SLP-2,

although higher expression levels of the truncation mutant were required for detection of

the plasma membrane-associated pool. This may indicate a role for the amino terminal

region in membrane association. In addition to human T cells, both the mitochondrial

and surface pool of SLP-2 were also found in primary mouse T cells, indicating a

conservation of two distinct localizations for SLP-2 across species.

36

In agreement with earlier results showing recruitment of SLP-2 to detergent-insoluble

membranes upon T cell activation, confocal microscopy demonstrated the recruitment of

SLP-2 to the immunological synapse upon T cell activation. Furthermore, this pool of

SLP-2 was actively maintained at the synapse, indicating a role for SLP-2 in T cell

activation. To better visualize the cell surface pool of SLP-2 during T cell activation,

total internal reflection fluorescence microscopy was used to limit the detection to a

depth of 200nm from the cell surface. These images showed the surface pool of SLP-2

surrounding TCR clusters at the immunological synapse, with SLP-2 being weakly

excluded from the TCR clusters. The ability of SLP-2 to self-associate may allow SLP-2

to organize the T cell membrane to facilitate TCR microcluster formation and to prevent

signal complex dissociation.

Taken together, the investigation of SLP-2 function using both human and mouse

systems, in both over-expression and knockout studies, indicates a role for SLP-2 in

membrane organization, predominantly of the mitochondrial membranes, but also for the

plasma membrane.

37

1.4 References

1. Simons, K. & Gerl, M.J. Revitalizing membrane rafts: new tools and insights. Nat Rev Mol Cell Biol 11, 688-699 (2010).

2. Lingwood, D. & Simons, K. Lipid rafts as a membrane-organizing principle. Science 327, 46-50 (2010).

3. Pitcher, L.A., Young, J.A., Mathis, M.A., Wrage, P.C., Bartok, B. & van Oers, N.S. The formation and functions of the 21- and 23-kDa tyrosine-phosphorylated TCR zeta subunits. Immunol Rev 191, 47-61 (2003).

4. Alarcon, B., Gil, D., Delgado, P. & Schamel, W.W. Initiation of TCR signaling: regulation within CD3 dimers. Immunol Rev 191, 38-46 (2003).

5. Smith-Garvin, J.E., Koretzky, G.A. & Jordan, M.S. T cell activation. Annu Rev Immunol 27, 591-619 (2009).

6. Love, P.E. & Hayes, S.M. ITAM-mediated signaling by the T-cell antigen receptor. Cold Spring Harb Perspect Biol 2, a002485 (2010).

7. Salmond, R.J., Filby, A., Qureshi, I., Caserta, S. & Zamoyska, R. T-cell receptor proximal signaling via the Src-family kinases, Lck and Fyn, influences T-cell activation, differentiation, and tolerance. Immunol Rev 228, 9-22 (2009).

8. Wange, R.L. LAT, the linker for activation of T cells: a bridge between T cell-specific and general signaling pathways. Sci STKE 2000, re1 (2000).

9. Hogan, P.G., Chen, L., Nardone, J. & Rao, A. Transcriptional regulation by calcium, calcineurin, and NFAT. Genes Dev 17, 2205-2232 (2003).

10. Lin, J., Miller, M.J. & Shaw, A.S. The c-SMAC: sorting it all out (or in). J Cell Biol 170, 177-182 (2005).

11. Dustin, M.L., Chakraborty, A.K. & Shaw, A.S. Understanding the structure and function of the immunological synapse. Cold Spring Harb Perspect Biol 2, a002311 (2010).

12. Monks, C.R., Freiberg, B.A., Kupfer, H., Sciaky, N. & Kupfer, A. Three-dimensional segregation of supramolecular activation clusters in T cells. Nature 395, 82-86 (1998).

13. Varma, R., Campi, G., Yokosuka, T., Saito, T. & Dustin, M.L. T cell receptor-proximal signals are sustained in peripheral microclusters and terminated in the central supramolecular activation cluster. Immunity 25, 117-127 (2006).

38

14. Fooksman, D.R., Vardhana, S., Vasiliver-Shamis, G., Liese, J., Blair, D.A., Waite, J., Sacristan, C., Victora, G.D., Zanin-Zhorov, A. & Dustin, M.L. Functional anatomy of T cell activation and synapse formation. Annu Rev Immunol 28, 79-105 (2010).

15. Xavier, R., Brennan, T., Li, Q., McCormack, C. & Seed, B. Membrane compartmentation is required for efficient T cell activation. Immunity 8, 723-732 (1998).

16. Janes, P.W., Ley, S.C. & Magee, A.I. Aggregation of lipid rafts accompanies signaling via the T cell antigen receptor. J Cell Biol 147, 447-461 (1999).

17. Viola, A., Schroeder, S., Sakakibara, Y. & Lanzavecchia, A. T lymphocyte costimulation mediated by reorganization of membrane microdomains. Science 283, 680-682 (1999).

18. Webb, Y., Hermida-Matsumoto, L. & Resh, M.D. Inhibition of protein palmitoylation, raft localization, and T cell signaling by 2-bromopalmitate and polyunsaturated fatty acids. J Biol Chem 275, 261-270 (2000).

19. Zhang, W., Irvin, B.J., Trible, R.P., Abraham, R.T. & Samelson, L.E. Functional analysis of LAT in TCR-mediated signaling pathways using a LAT-deficient Jurkat cell line. Int Immunol 11, 943-950 (1999).

20. Chichili, G.R. & Rodgers, W. Clustering of membrane raft proteins by the actin cytoskeleton. J Biol Chem 282, 36682-36691 (2007).

21. Kabouridis, P.S., Janzen, J., Magee, A.L. & Ley, S.C. Cholesterol depletion disrupts lipid rafts and modulates the activity of multiple signaling pathways in T lymphocytes. Eur J Immunol 30, 954-963 (2000).

22. Jin, Z.X., Huang, C.R., Dong, L., Goda, S., Kawanami, T., Sawaki, T., Sakai, T., Tong, X.P., Masaki, Y., Fukushima, T., Tanaka, M., Mimori, T., Tojo, H., Bloom, E.T., Okazaki, T. & Umehara, H. Impaired TCR signaling through dysfunction of lipid rafts in sphingomyelin synthase 1 (SMS1)-knockdown T cells. Int Immunol 20, 1427-1437 (2008).

23. Zhang, M., Moran, M., Round, J., Low, T.A., Patel, V.P., Tomassian, T., Hernandez, J.D. & Miceli, M.C. CD45 signals outside of lipid rafts to promote ERK activation, synaptic raft clustering, and IL-2 production. J Immunol 174, 1479-1490 (2005).

24. Su, M.W., Yu, C.L., Burakoff, S.J. & Jin, Y.J. Targeting Src homology 2 domain-containing tyrosine phosphatase (SHP-1) into lipid rafts inhibits CD3-induced T cell activation. J Immunol 166, 3975-3982 (2001).

25. Dart, C. Lipid microdomains and the regulation of ion channel function. J Physiol 588, 3169-3178 (2010).

39

26. Huber, T.B., Schermer, B., Muller, R.U., Hohne, M., Bartram, M., Calixto, A., Hagmann, H., Reinhardt, C., Koos, F., Kunzelmann, K., Shirokova, E., Krautwurst, D., Harteneck, C., Simons, M., Pavenstadt, H., Kerjaschki, D., Thiele, C., Walz, G., Chalfie, M. & Benzing, T. Podocin and MEC-2 bind cholesterol to regulate the activity of associated ion channels. Proc Natl Acad Sci U S A 103, 17079-17086 (2006).

27. Browman, D.T., Hoegg, M.B. & Robbins, S.M. The SPFH domain-containing proteins: more than lipid raft markers. Trends Cell Biol 17, 394-402 (2007).

28. Langer, T., Kaser, M., Klanner, C. & Leonhard, K. AAA proteases of mitochondria: quality control of membrane proteins and regulatory functions during mitochondrial biogenesis. Biochem Soc Trans 29, 431-436 (2001).

29. Nijtmans, L.G., de Jong, L., Artal Sanz, M., Coates, P.J., Berden, J.A., Back, J.W., Muijsers, A.O., van der Spek, H. & Grivell, L.A. Prohibitins act as a membrane-bound chaperone for the stabilization of mitochondrial proteins. EMBO J 19, 2444-2451 (2000).

30. Kasashima, K., Sumitani, M., Satoh, M. & Endo, H. Human prohibitin 1 maintains the organization and stability of the mitochondrial nucleoids. Exp Cell Res 314, 988-996 (2008).

31. Coates, P.J., Jamieson, D.J., Smart, K., Prescott, A.R. & Hall, P.A. The prohibitin family of mitochondrial proteins regulate replicative lifespan. Curr Biol 7, 607-610 (1997).

32. Artal-Sanz, M. & Tavernarakis, N. Prohibitin couples diapause signalling to mitochondrial metabolism during ageing in C. elegans. Nature 461, 793-797 (2009).

33. Merkwirth, C., Dargazanli, S., Tatsuta, T., Geimer, S., Lower, B., Wunderlich, F.T., von Kleist-Retzow, J.C., Waisman, A., Westermann, B. & Langer, T. Prohibitins control cell proliferation and apoptosis by regulating OPA1-dependent cristae morphogenesis in mitochondria. Genes Dev 22, 476-488 (2008).

34. Osman, C., Merkwirth, C. & Langer, T. Prohibitins and the functional compartmentalization of mitochondrial membranes. J Cell Sci 122, 3823-3830 (2009).

35. Osman, C., Haag, M., Potting, C., Rodenfels, J., Dip, P.V., Wieland, F.T., Brugger, B., Westermann, B. & Langer, T. The genetic interactome of prohibitins: coordinated control of cardiolipin and phosphatidylethanolamine by conserved regulators in mitochondria. J Cell Biol 184, 583-596 (2009).

36. Berger, K.H. & Yaffe, M.P. Prohibitin family members interact genetically with mitochondrial inheritance components in Saccharomyces cerevisiae. Mol Cell Biol 18, 4043-4052 (1998).

40

37. Steglich, G., Neupert, W. & Langer, T. Prohibitins regulate membrane protein degradation by the m-AAA protease in mitochondria. Mol Cell Biol 19, 3435-3442 (1999).

38. Morrow, I.C. & Parton, R.G. Flotillins and the PHB domain protein family: rafts, worms and anaesthetics. Traffic 6, 725-740 (2005).

39. Langhorst, M.F., Reuter, A. & Stuermer, C.A. Scaffolding microdomains and beyond: the function of reggie/flotillin proteins. Cell Mol Life Sci 62, 2228-2240 (2005).

40. Kihara, A., Akiyama, Y. & Ito, K. A protease complex in the Escherichia coli plasma membrane: HflKC (HflA) forms a complex with FtsH (HflB), regulating its proteolytic activity against SecY. EMBO J 15, 6122-6131 (1996).

41. Green, J.B. & Young, J.P. Slipins: ancient origin, duplication and diversification of the stomatin protein family. BMC Evol Biol 8, 44 (2008).

42. Lande, W.M. & Mentzer, W.C. Haemolytic anaemia associated with increased cation permeability. Clin Haematol 14, 89-103 (1985).

43. Stewart, G.W., Hepworth-Jones, B.E., Keen, J.N., Dash, B.C., Argent, A.C. & Casimir, C.M. Isolation of cDNA coding for an ubiquitous membrane protein deficient in high Na+, low K+ stomatocytic erythrocytes. Blood 79, 1593-1601 (1992).

44. Zhu, Y., Paszty, C., Turetsky, T., Tsai, S., Kuypers, F.A., Lee, G., Cooper, P., Gallagher, P.G., Stevens, M.E., Rubin, E., Mohandas, N. & Mentzer, W.C. Stomatocytosis is absent in "stomatin"-deficient murine red blood cells. Blood 93, 2404-2410 (1999).

45. Rajaram, S., Spangler, T.L., Sedensky, M.M. & Morgan, P.G. A stomatin and a degenerin interact to control anesthetic sensitivity in Caenorhabditis elegans. Genetics 153, 1673-1682 (1999).

46. Price, M.P., Thompson, R.J., Eshcol, J.O., Wemmie, J.A. & Benson, C.J. Stomatin modulates gating of acid-sensing ion channels. J Biol Chem 279, 53886-53891 (2004).

47. Goodman, M.B., Ernstrom, G.G., Chelur, D.S., O'Hagan, R., Yao, C.A. & Chalfie, M. MEC-2 regulates C. elegans DEG/ENaC channels needed for mechanosensation. Nature 415, 1039-1042 (2002).

48. Sedensky, M.M., Siefker, J.M., Koh, J.Y., Miller, D.M., 3rd & Morgan, P.G. A stomatin and a degenerin interact in lipid rafts of the nervous system of Caenorhabditis elegans. Am J Physiol Cell Physiol 287, C468-474 (2004).

41

49. Tavernarakis, N., Shreffler, W., Wang, S. & Driscoll, M. unc-8, a DEG/ENaC family member, encodes a subunit of a candidate mechanically gated channel that modulates C. elegans locomotion. Neuron 18, 107-119 (1997).

50. Huang, M., Gu, G., Ferguson, E.L. & Chalfie, M. A stomatin-like protein necessary for mechanosensation in C. elegans. Nature 378, 292-295 (1995).

51. Rajaram, S., Sedensky, M.M. & Morgan, P.G. Unc-1: a stomatin homologue controls sensitivity to volatile anesthetics in Caenorhabditis elegans. Proc Natl Acad Sci U S A 95, 8761-8766 (1998).

52. Barnes, T.M., Jin, Y., Horvitz, H.R., Ruvkun, G. & Hekimi, S. The Caenorhabditis elegans behavioral gene unc-24 encodes a novel bipartite protein similar to both erythrocyte band 7.2 (stomatin) and nonspecific lipid transfer protein. J Neurochem 67, 46-57 (1996).

53. Wetzel, C., Hu, J., Riethmacher, D., Benckendorff, A., Harder, L., Eilers, A., Moshourab, R., Kozlenkov, A., Labuz, D., Caspani, O., Erdmann, B., Machelska, H., Heppenstall, P.A. & Lewin, G.R. A stomatin-domain protein essential for touch sensation in the mouse. Nature 445, 206-209 (2007).

54. Sedensky, M.M., Pujazon, M.A. & Morgan, P.G. Tail clamp responses in stomatin knockout mice compared with mobility assays in Caenorhabditis elegans during exposure to diethyl ether, halothane, and isoflurane. Anesthesiology 105, 498-502 (2006).

55. Snyers, L., Umlauf, E. & Prohaska, R. Association of stomatin with lipid-protein complexes in the plasma membrane and the endocytic compartment. Eur J Cell Biol 78, 802-812 (1999).

56. Salzer, U. & Prohaska, R. Stomatin, flotillin-1, and flotillin-2 are major integral proteins of erythrocyte lipid rafts. Blood 97, 1141-1143 (2001).

57. Snyers, L., Thines-Sempoux, D. & Prohaska, R. Colocalization of stomatin (band 7.2b) and actin microfilaments in UAC epithelial cells. Eur J Cell Biol 73, 281-285 (1997).

58. Boute, N., Gribouval, O., Roselli, S., Benessy, F., Lee, H., Fuchshuber, A., Dahan, K., Gubler, M.C., Niaudet, P. & Antignac, C. NPHS2, encoding the glomerular protein podocin, is mutated in autosomal recessive steroid-resistant nephrotic syndrome. Nat Genet 24, 349-354 (2000).

59. Schwarz, K., Simons, M., Reiser, J., Saleem, M.A., Faul, C., Kriz, W., Shaw, A.S., Holzman, L.B. & Mundel, P. Podocin, a raft-associated component of the glomerular slit diaphragm, interacts with CD2AP and nephrin. J Clin Invest 108, 1621-1629 (2001).

42

60. Huber, T.B., Simons, M., Hartleben, B., Sernetz, L., Schmidts, M., Gundlach, E., Saleem, M.A., Walz, G. & Benzing, T. Molecular basis of the functional podocin-nephrin complex: mutations in the NPHS2 gene disrupt nephrin targeting to lipid raft microdomains. Hum Mol Genet 12, 3397-3405 (2003).

61. Mairhofer, M., Steiner, M., Salzer, U. & Prohaska, R. Stomatin-like protein-1 interacts with stomatin and is targeted to late endosomes. J Biol Chem 284, 29218-29229 (2009).

62. Wang, Y. & Morrow, J.S. Identification and characterization of human SLP-2, a novel homologue of stomatin (band 7.2b) present in erythrocytes and other tissues. J Biol Chem 275, 8062-8071 (2000).

63. Kirchhof, M.G., Chau, L.A., Lemke, C.D., Vardhana, S., Darlington, P.J., Marquez, M.E., Taylor, R., Rizkalla, K., Blanca, I., Dustin, M.L. & Madrenas, J. Modulation of T cell activation by stomatin-like protein 2. J Immunol 181, 1927-1936 (2008).

64. Da Cruz, S., Parone, P.A., Gonzalo, P., Bienvenut, W.V., Tondera, D., Jourdain, A., Quadroni, M. & Martinou, J.C. SLP-2 interacts with prohibitins in the mitochondrial inner membrane and contributes to their stability. Biochim Biophys Acta 1783, 904-911 (2008).

65. Hajek, P., Chomyn, A. & Attardi, G. Identification of a novel mitochondrial complex containing mitofusin 2 and stomatin-like protein 2. J Biol Chem 282, 5670-5681 (2007).

66. Da Cruz, S., Xenarios, I., Langridge, J., Vilbois, F., Parone, P.A. & Martinou, J.C. Proteomic analysis of the mouse liver mitochondrial inner membrane. J Biol Chem 278, 41566-41571 (2003).

67. Mootha, V.K., Bunkenborg, J., Olsen, J.V., Hjerrild, M., Wisniewski, J.R., Stahl, E., Bolouri, M.S., Ray, H.N., Sihag, S., Kamal, M., Patterson, N., Lander, E.S. & Mann, M. Integrated analysis of protein composition, tissue diversity, and gene regulation in mouse mitochondria. Cell 115, 629-640 (2003).

68. Rezaul, K., Wu, L., Mayya, V., Hwang, S.I. & Han, D. A systematic characterization of mitochondrial proteome from human T leukemia cells. Mol Cell Proteomics 4, 169-181 (2005).

69. Taylor, S.W., Fahy, E., Zhang, B., Glenn, G.M., Warnock, D.E., Wiley, S., Murphy, A.N., Gaucher, S.P., Capaldi, R.A., Gibson, B.W. & Ghosh, S.S. Characterization of the human heart mitochondrial proteome. Nat Biotechnol 21, 281-286 (2003).

70. Coates, P.J., Nenutil, R., McGregor, A., Picksley, S.M., Crouch, D.H., Hall, P.A. & Wright, E.G. Mammalian prohibitin proteins respond to mitochondrial stress and decrease during cellular senescence. Exp Cell Res 265, 262-273 (2001).

43

71. Ikonen, E., Fiedler, K., Parton, R.G. & Simons, K. Prohibitin, an antiproliferative protein, is localized to mitochondria. FEBS Lett 358, 273-277 (1995).

72. Tatsuta, T., Model, K. & Langer, T. Formation of membrane-bound ring complexes by prohibitins in mitochondria. Mol Biol Cell 16, 248-259 (2005).

73. Artal-Sanz, M., Tsang, W.Y., Willems, E.M., Grivell, L.A., Lemire, B.D., van der Spek, H. & Nijtmans, L.G. The mitochondrial prohibitin complex is essential for embryonic viability and germline function in Caenorhabditis elegans. J Biol Chem 278, 32091-32099 (2003).

74. Ishihara, N., Fujita, Y., Oka, T. & Mihara, K. Regulation of mitochondrial morphology through proteolytic cleavage of OPA1. EMBO J 25, 2966-2977 (2006).

75. Arnoult, D., Grodet, A., Lee, Y.J., Estaquier, J. & Blackstone, C. Release of OPA1 during apoptosis participates in the rapid and complete release of cytochrome c and subsequent mitochondrial fragmentation. J Biol Chem 280, 35742-35750 (2005).

76. Schlame, M., Rua, D. & Greenberg, M.L. The biosynthesis and functional role of cardiolipin. Prog Lipid Res 39, 257-288 (2000).

77. Fry, M. & Green, D.E. Cardiolipin requirement for electron transfer in complex I and III of the mitochondrial respiratory chain. J Biol Chem 256, 1874-1880 (1981).

78. Joshi, A.S., Zhou, J., Gohil, V.M., Chen, S. & Greenberg, M.L. Cellular functions of cardiolipin in yeast. Biochim Biophys Acta 1793, 212-218 (2008).

79. Choi, S.Y., Gonzalvez, F., Jenkins, G.M., Slomianny, C., Chretien, D., Arnoult, D., Petit, P.X. & Frohman, M.A. Cardiolipin deficiency releases cytochrome c from the inner mitochondrial membrane and accelerates stimuli-elicited apoptosis. Cell Death Differ 14, 597-606 (2007).

80. Benard, G., Bellance, N., James, D., Parrone, P., Fernandez, H., Letellier, T. & Rossignol, R. Mitochondrial bioenergetics and structural network organization. J Cell Sci 120, 838-848 (2007).

81. Rich, P.R. & Marechal, A. The mitochondrial respiratory chain. Essays Biochem 47, 1-23 (2010).

82. Acin-Perez, R., Fernandez-Silva, P., Peleato, M.L., Perez-Martos, A. & Enriquez, J.A. Respiratory active mitochondrial supercomplexes. Mol Cell 32, 529-539 (2008).

44

83. Zhang, M., Mileykovskaya, E. & Dowhan, W. Cardiolipin is essential for organization of complexes III and IV into a supercomplex in intact yeast mitochondria. J Biol Chem 280, 29403-29408 (2005).

84. Wenz, T., Hielscher, R., Hellwig, P., Schagger, H., Richers, S. & Hunte, C. Role of phospholipids in respiratory cytochrome bc(1) complex catalysis and supercomplex formation. Biochim Biophys Acta 1787, 609-616 (2009).

85. Epand, R.F., Tokarska-Schlattner, M., Schlattner, U., Wallimann, T. & Epand, R.M. Cardiolipin clusters and membrane domain formation induced by mitochondrial proteins. J Mol Biol 365, 968-980 (2007).

86. Maniti, O., Lecompte, M.F., Marcillat, O., Desbat, B., Buchet, R., Vial, C. & Granjon, T. Mitochondrial creatine kinase binding to phospholipid monolayers induces cardiolipin segregation. Biophys J 96, 2428-2438 (2009).

87. Wang, Y., Cao, W., Yu, Z. & Liu, Z. Downregulation of a mitochondria associated protein SLP-2 inhibits tumor cell motility, proliferation and enhances cell sensitivity to chemotherapeutic reagents. Cancer Biol Ther 8, 1651-1658 (2009).

88. Tondera, D., Grandemange, S., Jourdain, A., Karbowski, M., Mattenberger, Y., Herzig, S., Da Cruz, S., Clerc, P., Raschke, I., Merkwirth, C., Ehses, S., Krause, F., Chan, D.C., Alexander, C., Bauer, C., Youle, R., Langer, T. & Martinou, J.C. SLP-2 is required for stress-induced mitochondrial hyperfusion. EMBO J 28, 1589-1600 (2009).

89. Frauwirth, K.A. & Thompson, C.B. Regulation of T lymphocyte metabolism. J Immunol 172, 4661-4665 (2004).

90. Frauwirth, K.A., Riley, J.L., Harris, M.H., Parry, R.V., Rathmell, J.C., Plas, D.R., Elstrom, R.L., June, C.H. & Thompson, C.B. The CD28 signaling pathway regulates glucose metabolism. Immunity 16, 769-777 (2002).

91. Vander Heiden, M.G., Cantley, L.C. & Thompson, C.B. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science 324, 1029-1033 (2009).

92. Delgoffe, G.M., Kole, T.P., Zheng, Y., Zarek, P.E., Matthews, K.L., Xiao, B., Worley, P.F., Kozma, S.C. & Powell, J.D. The mTOR kinase differentially regulates effector and regulatory T cell lineage commitment. Immunity 30, 832-844 (2009).

93. Michalek, R.D., Gerriets, V.A., Jacobs, S.R., Macintyre, A.N., MacIver, N.J., Mason, E.F., Sullivan, S.A., Nichols, A.G. & Rathmell, J.C. Cutting edge: distinct glycolytic and lipid oxidative metabolic programs are essential for effector and regulatory CD4+ T cell subsets. J Immunol 186, 3299-3303 (2011).

45

94. Hue, L. & Rider, M.H. The AMP-activated protein kinase: more than an energy sensor. Essays Biochem 43, 121-137 (2007).

95. Pearce, E.L., Walsh, M.C., Cejas, P.J., Harms, G.M., Shen, H., Wang, L.S., Jones, R.G. & Choi, Y. Enhancing CD8 T-cell memory by modulating fatty acid metabolism. Nature 460, 103-107 (2009).

96. Araki, K., Turner, A.P., Shaffer, V.O., Gangappa, S., Keller, S.A., Bachmann, M.F., Larsen, C.P. & Ahmed, R. mTOR regulates memory CD8 T-cell differentiation. Nature 460, 108-112 (2009).

97. Gunter, T.E., Yule, D.I., Gunter, K.K., Eliseev, R.A. & Salter, J.D. Calcium and mitochondria. FEBS Lett 567, 96-102 (2004).

98. Rimessi, A., Giorgi, C., Pinton, P. & Rizzuto, R. The versatility of mitochondrial calcium signals: from stimulation of cell metabolism to induction of cell death. Biochim Biophys Acta 1777, 808-816 (2008).

99. Nicholls, D.G. Mitochondrial ion circuits. Essays Biochem 47, 25-35 (2010).

100. Santo-Domingo, J. & Demaurex, N. Calcium uptake mechanisms of mitochondria. Biochim Biophys Acta 1797, 907-912 (2010).

101. Razzaq, T.M., Ozegbe, P., Jury, E.C., Sembi, P., Blackwell, N.M. & Kabouridis, P.S. Regulation of T-cell receptor signalling by membrane microdomains. Immunology 113, 413-426 (2004).

102. Goodridge, H.S. & Harnett, M.M. Introduction to immune cell signalling. Parasitology 130 Suppl, S3-9 (2005).

103. Oh-hora, M. & Rao, A. Calcium signaling in lymphocytes. Curr Opin Immunol 20, 250-258 (2008).

104. Gallo, E.M., Cante-Barrett, K. & Crabtree, G.R. Lymphocyte calcium signaling from membrane to nucleus. Nat Immunol 7, 25-32 (2006).

105. Baba, Y. & Kurosaki, T. Physiological function and molecular basis of STIM1-mediated calcium entry in immune cells. Immunol Rev 231, 174-188 (2009).

106. Hoth, M., Button, D.C. & Lewis, R.S. Mitochondrial control of calcium-channel gating: a mechanism for sustained signaling and transcriptional activation in T lymphocytes. Proc Natl Acad Sci U S A 97, 10607-10612 (2000).

107. Quintana, A., Schwarz, E.C., Schwindling, C., Lipp, P., Kaestner, L. & Hoth, M. Sustained activity of calcium release-activated calcium channels requires translocation of mitochondria to the plasma membrane. J Biol Chem 281, 40302-40309 (2006).

46

108. Quintana, A., Schwindling, C., Wenning, A.S., Becherer, U., Rettig, J., Schwarz, E.C. & Hoth, M. T cell activation requires mitochondrial translocation to the immunological synapse. Proc Natl Acad Sci U S A 104, 14418-14423 (2007).

109. Da Cruz, S., De Marchi, U., Frieden, M., Parone, P.A., Martinou, J.C. & Demaurex, N. SLP-2 negatively modulates mitochondrial sodium-calcium exchange. Cell Calcium 47, 11-18 (2009).

110. Jin, Z. & El-Deiry, W.S. Overview of cell death signaling pathways. Cancer Biol Ther 4, 139-163 (2005).

111. Bouillet, P. & O'Reilly, L.A. CD95, BIM and T cell homeostasis. Nat Rev Immunol 9, 514-519 (2009).

112. Parsons, M.J. & Green, D.R. Mitochondria in cell death. Essays Biochem 47, 99-114 (2010).

113. Earnshaw, W.C., Martins, L.M. & Kaufmann, S.H. Mammalian caspases: structure, activation, substrates, and functions during apoptosis. Annual review of biochemistry 68, 383-424 (1999).

114. Green, D.R., Galluzzi, L. & Kroemer, G. Mitochondria and the autophagy-inflammation-cell death axis in organismal aging. Science 333, 1109-1112 (2011).

115. Lawen, A. Apoptosis-an introduction. Bioessays 25, 888-896 (2003).

116. Youle, R.J. & Strasser, A. The BCL-2 protein family: opposing activities that mediate cell death. Nat Rev Mol Cell Biol 9, 47-59 (2008).

117. Luo, X., Budihardjo, I., Zou, H., Slaughter, C. & Wang, X. Bid, a Bcl2 interacting protein, mediates cytochrome c release from mitochondria in response to activation of cell surface death receptors. Cell 94, 481-490 (1998).

118. McBride, H.M., Neuspiel, M. & Wasiak, S. Mitochondria: more than just a powerhouse. Curr Biol 16, R551-560 (2006).

119. Scott, I. & Youle, R.J. Mitochondrial fission and fusion. Essays Biochem 47, 85-98 (2010).

120. Chan, D.C. Mitochondrial fusion and fission in mammals. Annu Rev Cell Dev Biol 22, 79-99 (2006).

121. Song, Z., Ghochani, M., McCaffery, J.M., Frey, T.G. & Chan, D.C. Mitofusins and OPA1 mediate sequential steps in mitochondrial membrane fusion. Mol Biol Cell 20, 3525-3532 (2009).

122. Yoon, Y., Krueger, E.W., Oswald, B.J. & McNiven, M.A. The mitochondrial protein hFis1 regulates mitochondrial fission in mammalian cells through an

47

interaction with the dynamin-like protein DLP1. Mol Cell Biol 23, 5409-5420 (2003).

123. Mozdy, A.D., McCaffery, J.M. & Shaw, J.M. Dnm1p GTPase-mediated mitochondrial fission is a multi-step process requiring the novel integral membrane component Fis1p. J Cell Biol 151, 367-380 (2000).

124. Otera, H., Wang, C., Cleland, M.M., Setoguchi, K., Yokota, S., Youle, R.J. & Mihara, K. Mff is an essential factor for mitochondrial recruitment of Drp1 during mitochondrial fission in mammalian cells. J Cell Biol 191, 1141-1158 (2010).

125. Chen, H., Detmer, S.A., Ewald, A.J., Griffin, E.E., Fraser, S.E. & Chan, D.C. Mitofusins Mfn1 and Mfn2 coordinately regulate mitochondrial fusion and are essential for embryonic development. J Cell Biol 160, 189-200 (2003).

126. Davies, V.J., Hollins, A.J., Piechota, M.J., Yip, W., Davies, J.R., White, K.E., Nicols, P.P., Boulton, M.E. & Votruba, M. Opa1 deficiency in a mouse model of autosomal dominant optic atrophy impairs mitochondrial morphology, optic nerve structure and visual function. Hum Mol Genet 16, 1307-1318 (2007).

127. Ishihara, N., Nomura, M., Jofuku, A., Kato, H., Suzuki, S.O., Masuda, K., Otera, H., Nakanishi, Y., Nonaka, I., Goto, Y., Taguchi, N., Morinaga, H., Maeda, M., Takayanagi, R., Yokota, S. & Mihara, K. Mitochondrial fission factor Drp1 is essential for embryonic development and synapse formation in mice. Nat Cell Biol 11, 958-966 (2009).

128. Wakabayashi, J., Zhang, Z., Wakabayashi, N., Tamura, Y., Fukaya, M., Kensler, T.W., Iijima, M. & Sesaki, H. The dynamin-related GTPase Drp1 is required for embryonic and brain development in mice. J Cell Biol 186, 805-816 (2009).

129. Chen, H., McCaffery, J.M. & Chan, D.C. Mitochondrial fusion protects against neurodegeneration in the cerebellum. Cell 130, 548-562 (2007).

130. Mitra, K., Wunder, C., Roysam, B., Lin, G. & Lippincott-Schwartz, J. A hyperfused mitochondrial state achieved at G1-S regulates cyclin E buildup and entry into S phase. Proc Natl Acad Sci U S A 106, 11960-11965 (2009).

131. Taguchi, N., Ishihara, N., Jofuku, A., Oka, T. & Mihara, K. Mitotic phosphorylation of dynamin-related GTPase Drp1 participates in mitochondrial fission. J Biol Chem 282, 11521-11529 (2007).

132. Jeong, S.Y. & Seol, D.W. The role of mitochondria in apoptosis. BMB Rep 41, 11-22 (2008).

133. Suen, D.F., Norris, K.L. & Youle, R.J. Mitochondrial dynamics and apoptosis. Genes Dev 22, 1577-1590 (2008).

48

134. Frezza, C., Cipolat, S., Martins de Brito, O., Micaroni, M., Beznoussenko, G.V., Rudka, T., Bartoli, D., Polishuck, R.S., Danial, N.N., De Strooper, B. & Scorrano, L. OPA1 controls apoptotic cristae remodeling independently from mitochondrial fusion. Cell 126, 177-189 (2006).

135. Karbowski, M., Lee, Y.J., Gaume, B., Jeong, S.Y., Frank, S., Nechushtan, A., Santel, A., Fuller, M., Smith, C.L. & Youle, R.J. Spatial and temporal association of Bax with mitochondrial fission sites, Drp1, and Mfn2 during apoptosis. J Cell Biol 159, 931-938 (2002).

136. Lee, Y.J., Jeong, S.Y., Karbowski, M., Smith, C.L. & Youle, R.J. Roles of the mammalian mitochondrial fission and fusion mediators Fis1, Drp1, and Opa1 in apoptosis. Mol Biol Cell 15, 5001-5011 (2004).

137. Garofalo, T., Giammarioli, A.M., Misasi, R., Tinari, A., Manganelli, V., Gambardella, L., Pavan, A., Malorni, W. & Sorice, M. Lipid microdomains contribute to apoptosis-associated modifications of mitochondria in T cells. Cell Death Differ 12, 1378-1389 (2005).

138. Sorice, M., Manganelli, V., Matarrese, P., Tinari, A., Misasi, R., Malorni, W. & Garofalo, T. Cardiolipin-enriched raft-like microdomains are essential activating platforms for apoptotic signals on mitochondria. FEBS Lett 583, 2447-2450 (2009).

139. Kagan, V.E., Tyurin, V.A., Jiang, J., Tyurina, Y.Y., Ritov, V.B., Amoscato, A.A., Osipov, A.N., Belikova, N.A., Kapralov, A.A., Kini, V., Vlasova, II, Zhao, Q., Zou, M., Di, P., Svistunenko, D.A., Kurnikov, I.V. & Borisenko, G.G. Cytochrome c acts as a cardiolipin oxygenase required for release of proapoptotic factors. Nat Chem Biol 1, 223-232 (2005).

140. Kim, T.H., Zhao, Y., Ding, W.X., Shin, J.N., He, X., Seo, Y.W., Chen, J., Rabinowich, H., Amoscato, A.A. & Yin, X.M. Bid-cardiolipin interaction at mitochondrial contact site contributes to mitochondrial cristae reorganization and cytochrome C release. Mol Biol Cell 15, 3061-3072 (2004).

141. Liu, J., Weiss, A., Durrant, D., Chi, N.W. & Lee, R.M. The cardiolipin-binding domain of Bid affects mitochondrial respiration and enhances cytochrome c release. Apoptosis 9, 533-541 (2004).

142. Gonzalvez, F., Schug, Z.T., Houtkooper, R.H., Mackenzie, E.D., Brooks, D.G., Wanders, R.J., Petit, P.X., Vaz, F.M. & Gottlieb, E. Cardiolipin provides an essential activating platform for caspase-8 on mitochondria. J Cell Biol 183, 681-696 (2008).

143. Gonzalvez, F., Pariselli, F., Dupaigne, P., Budihardjo, I., Lutter, M., Antonsson, B., Diolez, P., Manon, S., Martinou, J.C., Goubern, M., Wang, X., Bernard, S. & Petit, P.X. tBid interaction with cardiolipin primarily orchestrates mitochondrial

49

dysfunctions and subsequently activates Bax and Bak. Cell Death Differ 12, 614-626 (2005).

144. Ciarlo, L., Manganelli, V., Garofalo, T., Matarrese, P., Tinari, A., Misasi, R., Malorni, W. & Sorice, M. Association of fission proteins with mitochondrial raft-like domains. Cell Death Differ 17, 1047-1058 (2010).

145. Huang, Z., Jiang, J., Tyurin, V.A., Zhao, Q., Mnuskin, A., Ren, J., Belikova, N.A., Feng, W., Kurnikov, I.V. & Kagan, V.E. Cardiolipin deficiency leads to decreased cardiolipin peroxidation and increased resistance of cells to apoptosis. Free Radic Biol Med 44, 1935-1944 (2008).

50

Chapter 2

2 Stomatin-Like Protein 2 Binds Cardiolipin and Regulates Mitochondrial Biogenesis and Function 1

Stomatin-like protein 2 (SLP-2) is a widely expressed mitochondrial inner membrane

protein of unknown function. Here we show that human SLP-2 interacts with prohibitin-

1 and -2 and binds to the mitochondrial membrane phospholipid cardiolipin.

Upregulation of SLP-2 expression increases cardiolipin content and the formation of

metabolically active mitochondrial membranes and induces mitochondrial biogenesis. In

human T lymphocytes, these events correlate with increased complex I and II activities,

increased intracellular ATP stores, and increased resistance to apoptosis through the

intrinsic pathway, ultimately enhancing cellular responses. We propose that the function

of SLP-2 is to recruit prohibitins to cardiolipin to form cardiolipin-enriched

microdomains in which electron transport complexes are optimally assembled. Likely

through the prohibitin functional interactome, SLP-2 then regulates mitochondrial

biogenesis and function.

2.1 Introduction

The demands for energy-generating substrates in a eukaryotic cell rise as the cell

transitions from a resting state to an activated state. This process is well documented in

lymphocytes undergoing mitogenic responses. In these cells, the increasing energy

requirements are met by increased production of ATP either by glycolysis or by oxidative

1 The material contained in this chapter was published in: Christie, D., Lemke, C.D., Elias, I.M.,

Chau, L.A., Kirchhof, M.G., Li, B., Ball, E.H., Dunn, S.D., Hatch, G.M. and Madrenas, J. Stomatin-like protein 2 binds cardiolipin and regulates mitochondrial biogenesis and function. Mol. Cell. Biol. 31 (18): 3845-3856 (2011).

Copyright 2011. American Society for Microbiology

51

phosphorylation (OXPHOS); both processes being regulated by signalling from antigen

receptors and costimulatory molecules 1-3. Scattered evidence suggests that these

demands on cellular bioenergetics correlate with augmentation of mitochondrial

membrane surface and of mitochondrial numbers 4-6, a finding consistent with the key

role played by these organelles in supplying most cellular ATP 3. However, the

molecules that regulate mitochondrial biogenesis in response to cell activation remain

mostly unknown.

Recent evidence suggests that the biogenesis of mitochondrial membranes may be

regulated by the functional interactome of prohibitins (PHB) -1 and -2 7, but it is not

known how prohibitins sense the need to increase mitochondrial membrane biogenesis.

We have previously reported that SLP-2, an evolutionarily conserved mitochondrial

protein belonging to the stomatin family 8-10 and enriched in detergent-insoluble

microdomains of T lymphocytes, is upregulated during activation of these cells and

enhances T cell responses in vivo and in vitro 11. The function of SLP-2 is unknown.

Since SLP-2 is mostly located in mitochondrial membranes and has been shown to

interact with PHB-1 and PHB-2 12 we hypothesized that upregulation of SLP-2

expression may act as a linker between PHBs and mitochondrial biogenesis. Here, we

show that SLP-2 binds cardiolipin (CL) and facilitates the assembly of respiratory chain

components. Upregulation of SLP-2 expression then translates into enhanced

mitochondrial biogenesis and function.

2.2 Materials and Methods

2.2.1 Cells

Jurkat T cells were obtained from American Type Culture Collection (Manassas, VA).

The LG2 B lymphoblastoid cell line used as antigen-presenting cells was provided by Dr.

Eric Long (NIAID, NIH, Rockville, MD). Peripheral blood mononuclear cells (PBMCs)

were isolated from normal donors using Ficoll-Hypaque (Amersham Pharmacia Biotech,

Uppsala, Sweden). Cells were washed and resuspended at 1 x 106 cells/ml. PBMC blasts

52

were generated with Phorbol Myristate Acetate (PMA; 1 ng/ml) and Ionomycin (100

ng/ml) at 37oC for 72 h.

2.2.2 Plasmids, siRNA and T cell transfectants

Full length human SLP-2 cDNA was subcloned into the pEGFPN1 expression vector

(Clontech Inc., Palo Alto, CA) to create an in-frame translational fusion of SLP-2 with

GFP at the 3’ end (SLP-2-GFP) as described 11. The full length SLP-2-GFP was placed

into the doxycycline-inducible pBig2i vector 13. A doxycycline-inducible mutant of

human SLP-2 lacking the amino-terminal domain tagged with GFP was also constructed.

Stable transfectants were generated by nucleofection.

2.2.3 Upregulation of SLP-2 expression

For induction of high levels of SLP-2 expression, doxycycline (Sigma, St. Louis, MO)

was added in culture at 1 µg/mL for 18-24 h. Parental cells, which express low levels of

SLP-2, are referred to as low expressors (SLP-2lo) whereas SLP-2-GFP-transfected T

cells cultured in the presence of doxycycline are referred as high expressors (SLP-2hi).

Transfection of pBig2iGFP, pBig2iSLP2GFP, pmaxGFP® (Amaxa, Gaithersburg, MD)

or mock transfection into human PBMC or isolated primary CD4+ T helper cells were

conducted using Human T Cell Nucleofector Kit (Amaxa). After transfection, cells were

cultured for 24 h before stimulation.

2.2.4 Antibodies

The following monoclonal antibodies (MAb) were used in these studies: OPA-1 and

DLP1(Drp1) (BD Bisosciences, Mississauga, ON); cytochrome c , complex I subunit

NDUFS3, the 70kDa subunit of Complex II, Complex III core II, subunit II of Complex

IV, the alpha subunit of Complex V (MitoSciences, Eugene, OR), β-actin (Santa Cruz

Biotechnology, Santa Cruz, CA), and glyceraldehyde-3-phosphate dehydrogenase

(GAPDH) (Chemicon International, Temecula, CA). Rabbit polyclonal antibodies

against Mfn2 (Sigma, St. Louis, MO), PGC-1α (Santa Cruz Biotechnology, Santa Cruz,

CA), and SLP-2 (ProteinTech Group, Inc., Chicago, IL) were also used.

53

2.2.5 Reagents

The following working concentrations were used: 4 µM oligomycin, 1 µg/mL ionomycin

(both from Sigma). [3H]Acetate and [1-14C]linoleic acid were obtained from Amersham

(Oakville, Ontario, Canada). Lipid standards were obtained from Serdary Research

Laboratories (Englewood Cliffs, NJ). Thin layer chromatographic plates (silica gel G;

0.25 mm thickness) were obtained from Fisher Scientific (Winnipeg, Manitoba, Canada).

Ecolite scintillant was obtained from ICN Biochemicals (Montreal, Quebec, Canada).

Primers for real time-PCR were obtained from Invitrogen. All other chemicals were

certified ACS grade or better and obtained from Sigma Chemical Company or Fisher

Scientific.

2.2.6 Confocal microscopy

Confocal microscopy was performed with a Zeiss LSM 510 microscope (Carl Zeiss, Inc.) 14. Photobleaching experiments to test for mitochondrial fusion were carried out by

targeting an area of MitoTracker Red CMXRos labeled mitochondria within a cell for

repeated exposure to high power HeNe1 laser. Loss of fluorescence at the targeted point

versus an unrelated point within the same cell or another cell were then compared.

Similar loss in fluorescent intensity from the unrelated point within the same cell

indicated mitochondrial fusion between the two points.

2.2.7 Transmission electron microscopy

Cells were fixed in 2.5% gluteraldehyde in 0.1M sodium cacodylate buffer for 2 h,

washed in the buffer, fixed in 1% osmium tetroxide in 0.1M sodium cacodylate buffer for

1 h, washed in the buffer again and enrobed in Noble agar. Following a wash in distilled

water, cells were stained with 2% uranyl acetate for 2 h, then dehydrated through a

graded series of ethanol, cleared in propylene oxide, infiltrated with Epon resin and

embedded. Thin sections were stained with 2% uranyl acetate and lead citrate and were

viewed with a Philips 410 transmission electron microscope.

54

2.2.8 Measurement of mitochondrial mass

Human PBMCs were labeled with 100 nM MitoTracker Red® CMXRos (InvitrogenTM)

for 20 min at 37°C in complete RPMI 1640. Fluorescence was detected by flow

cytometry and analyzed with FlowJo flow cytometry analysis software (Treestar, Inc.,

Ashland, OR). To inhibit cardiolipin synthase during in vitro activation, human PBMCs

were stimulated with 1 ng/mL PMA and 100 ng/mL ionomycin in the presence of 4µM

triacsin C at 37°C for 24 hours 15.

2.2.9 Measurement of cardiolipin

Human PBMCs, resting or blasted were labeled with 10nM nonyl-acridine orange

(Sigma) for 15 min at 37°C. Fluorescence was detected by flow cytometry.

2.2.10 Measurement of cardiolipin mass in stable doxycycline-inducible SLP-2-GFP transfected Jurkat cells

Jurkat cells stably transfected with doxycycline-inducible SLP-2-GFP were incubated

without (SLP-2lo) or with (SLP-2hi) 1 µg/ml doxycycline for 72 h. After incubation, the

medium was aspirated, and the dishes were washed twice with 2 ml of ice-cold

phosphate-buffered saline. Cells were harvested in 2 ml of methanol:water (1:1, by

volume), and 25 µL aliquots were taken for protein determination 16. Subsequently, 0.5

ml of water and 2 ml chloroform were added to initiate phase separation. Samples were

centrifuged at 2000 rpm for 10 min, and the upper phase was aspirated. Two milliliters of

theoretical upper phase (methanol-0.9% NaCl-chloroform) (48:47:3, by volume) was

added, and centrifugation was repeated for 5 min. The organic phase was removed, dried

under nitrogen gas, and resuspended in chloroform:methanol (2:1, by volume). A 50-µl

aliquot was spotted onto thin-layer plates for separation of cardiolipin from other

phospholipids as described elsewhere 35. Phospholipids were removed from the plate, and

the phospholipid phosphorus content of cardiolipin was determined as described

previously 17.

55

2.2.11 Radiotracer studies of de novo cardiolipin synthesis

SLP-2lo or SLP-2hi cells were grown in Dulbecco’s modified Eagle medium (DMEM)

with 10% fetal bovine serum (FBS) and 100 U Penicillin, and 100 µg Streptomycin in 60

mm-diameter dishes and were incubated at 37°C under a humidified atmosphere of 5%

CO2. Cells were then incubated immediately after doxycycline treatment with 0.1 µM

[3H]acetate (10 µCi/dish) for 16 h or 0.1 mM [1-14C]linoleic acid (3 µCi/dish) bound to

albumin (molar ratio, 1:1) for 8 h. Cells were harvested, and the amount of radioactivity

incorporated into cardiolipin determined as described previously 18.

2.2.12 Real time-PCR

The cycler protocol for real-time PCR consisted of a 30-min reverse transcription (RT)

step at 50oC, followed by a 15 min Taq activation step at 95oC, followed by a 1-min

separation at 95oC and a melting curve that increased in temperature incrementally from

60oC to 95oC over the course of 20 min. The primers used for real time-PCR were

Invitrogen’s D-LUX 6-carboxyfluorescein (FAM)-labeled primer sets: for cardiolipin

synthase, reverse primer CGAACCGTGGTGTTGGAAGAGTT-FAM-G, forward primer

CGAGAGATGTAATGTTGATTGCTG; for phosphatidylglycerol phosphate synthase,

reverse primer CGGTGAGTCACTCAGGTTTGCACC-FAM-G, forward primer

TCGGCCTCCAGCACATTAAG; for PGC-1α, reverse primer TGC TTC GTC GTC

AAA AAC AG, forward primer TCA GTC CTC ACT GGT GGA CA; and for 18srRNA

reverse primer CGGGTGCTCTTAGCTGAGTGTCC-FAM-G, forward primer

CTCGGGCCTGCTTTGAACAC. Changes in mRNAs were analyzed on an Eppendorf

Mastercycler ep Realplex, software version 1.5.474, and data are presented as the mean

fold change (2-ΔΔCT) in mRNA expression relative to 18s rRNA expression 19.

2.2.13 Mitochondrial DNA quantification

Mitochondrial cytochrome c oxidase I (CCO) and nuclear polymerase γ accessory subunit

were amplified from total cellular DNA 20. Relative levels of mitochondrial DNA were

determined by comparing the threshold cycle (CT) detection values of CCO in SLP-2hi

and SLP-2lo cells and were normalized to nuclear DNA levels by the ΔΔCT method 19.

56

2.2.14 Cell lysate preparation

Cells were lysed in a 1% Triton X-100-containing buffer (1% Triton X-100, 150mM

NaCl, 10mM Tris [pH7.6], 5mM EDTA, 1mM sodium orthovanadate, 10µg/mL

leupeptin, 10µg/mL aprotinin, 25µM p-nitrophenyl-p’-guanidinobenzoate) at 4ºC for 30

min. Lysates were cleared by centrifugation (10,700 x g, 4ºC, 10 min), mixed with

sample buffer containing β-mercaptoethanol, boiled and blotted 21.

2.2.15 Mitochondria isolation

Intact mitochondria were isolated from 5 x 106 to 10 x 106 Jurkat T cells using the

Qproteome Mitochondria Isolation Kit (Qiagen). Mitochondrial preparations were mixed

with sample buffer containing β-mercaptoethanol, boiled, and analyzed by sodium

dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE).

2.2.16 Prohibitin and SLP-2 membrane association

Mitochondria were isolated and fractionated with 0.1M Na2CO3 as described previously 22. Fractions treated with H2O were used as an input control. Soluble and membrane

fractions were analyzed by immunoblotting for PHB-1 and SLP-2.

2.2.17 NADH dehydrogenase activity

NADH dehydrogenase activity from whole cell lysates was measured using the complex I

enzyme activity microplate assay kit (Mitosciences).

2.2.18 ATP quantitation

Mitochondrial and total-cell-lysate ATP levels were measured using the ATP

Determination Kit (Molecular ProbesTM, Eugene, OR). Briefly, 10 µL of ATP standards

(ranging from 1.95 to 1000 nM), mitochondrial fractions or cell lysates were added in

triplicate to 90 µL of a standard reaction solution in a 96-well Chromalux luminescent

assay microplate (Dynex Technologies, Chantilly, VA). Sample luminescence was

measured using an MLX microtiter plate luminometer (Dynex Technologies). Sample

ATP concentrations were calculated from the standard curve and were normalized to cell

equivalents.

57

2.2.19 T cell stimulation

Jurkat T cells were stimulated as described previously 14. A CD4+ T cell isolation kit and

MACS (magnetic cell sorting) columns (Miltenyi Biotec Inc., Auburn, CA) were used to

isolate primary CD4+ T helper cells from PBMCs. pBig2iGFP, pBig2iSLP2GFP,

pmaxGFP® or mock transfected PBMCs (0.2x106 cells/group) in the presence or absence

of 1µg/mL doxycycline were plated in triplicate on 96 well plates with various

concentrations of staphylococcal enterotoxin E (SEE) at 37°C and 5% CO2 for 24 h.

Supernatants were collected, and interleukin-2 (IL-2) was measured by an enzyme-linked

immunosorbent assay (ELISA) 23.

2.2.20 Oxygen consumption

Cells were incubated in a 96-well oxygen biosensor system (Becton Dickinson [BD], San

Jose, CA). Background fluorescence was measured before plating cells, and specific

fluorescence due to oxygen consumption was measured after 24 h of incubation at 37°C.

The fluorescent signal was measured on a Safire fluorescent plate reader (Tecan,

Switzerland). To calculate [O2] from fluorescence intensity, we followed the protocol

supplied by BD: [O2] = [(DR/NRF)-1]/KSV, where NRF is normalized relative

fluorescence, calculated as the fluorescence intensity of cells (I) divided by the

fluorescence intensity of a blank well (IB); DR is calculated as the fluorescence intensity

of 100mM sodium sulfite (IS) divided by IB; and KSV is calculated as (DR-1)/[O2]A,

where [O2]A equals 195µM at 37°C with 5% CO2.

2.2.21 Induction and detection of apoptosis

T cells were treated with 5 µg/mL of actinomycin D (Sigma) for 3 h to induce apoptosis,

which was measured by flow cytometry as ΔΨm (mitochondrial transmembrane potential)

loss using the cell-permeate probe MitoTracker Red CMXRos (InvitrogenTM), or as an

increase in Annexin V staining.

2.2.22 Generation of human recombinant SLP-2 (hrSLP-2).

We generated a hrSLP-22 cDNA coding for amino acids 36 to 356 of human SLP-2

downstream of a sequence coding for 6xHIS tag, a thrombin cleavage site, Thioredoxin,

58

and a tobacco etch virus (TEV) cleavage site in a pET28 plasmid. Rosetta strain E. coli

containing plasmids were streaked and single colonies were picked and grown to a A600

of 0.5 to 0.6. The culture was cooled to 15°C and induced with 0.1 mM isopropyl-β-D-

thiogalactopyranoside (IPTG) overnight. Bacteria were pelleted and scraped into pre-

weighed 50 ml centrifuge tubes, resuspended in 25 ml 50/10 buffer (50 mM Tris pH 8.0,

10 mM MgCl2), and centrifuged at 10,000 rpm for 10 min. The supernatant was decanted,

and the pellet was weighed and resuspended in 10x (wet weight) 50/10 buffer. The buffer

was fortified with 1 mM PMSF (Sigma), 5 mM p-aminobenzamidine (Sigma) and

Complete protease inhibitor cocktail (Roche). The bacteria were lysed by two passes at

10,000 PSI in a French pressure cell, and the lysates were centrifuged for 10 min at

10,000 rpm at 4°C. The supernatant was decanted into 60 ml ultracentrifuge tubes, spun

at 38,000 rpm at 4°C for 90 min, decanted again, and stored at -80°C.

Chromatography was run using Chelating Sepharose Fast Flow resin (GE BioScience).

Two column volumes of NiSO4 were applied to the column in order to bind the nickel.

The column was equilibrated with 10 column volumes of Buffer A (20 mM Tris-HCl [pH

8.0], 100 mM NaCl, 5 mM 2-mercaptoethanol, 10% glycerol, 20 mM imidazole). The

sample was added to the column and was allowed to flow through the resin at a rate of

approximately 0.5 ml/min. The column was washed with 10 column volumes of Buffer

A, followed by 2 column volumes of Buffer B (20 mM Tris-HCl [pH 8.0], 1 M NaCl, 5

mM 2-mercaptoethanol, 10% glycerol) and a further 2 volumes of Buffer A. Bound

proteins were eluted by competitive binding with Buffer C (20 mM Tris-HCl [pH 8.0],

100 mM NaCl, 5 mM 2-mercaptoethanol, 10% glycerol, 0-500 mM imidazole) and were

collected in 1.5 ml or 5 ml fractions. Fractions were stained with Coomassie Brilliant

Blue and were analysed on SDS-PAGE.

Protein samples were dialysed into 10 mM Tris (pH 8.0) overnight at 4°C. The protein

solution was then brought up to 2 mM EDTA and 1 mM dithiothreitol (DTT) and was

quantitated by the bicinchoninic acid (BCA) method (BCA protein assay kit; Pierce). The

SLP-2 construct was then incubated with TEV protease (ratio, 1:10) at room temperature

for 2 h. After cleavage, the sample was dialyzed into 10 mM Tris (pH 8.0) overnight at

4°C to remove EDTA and DTT.

59

2.2.23 Phospholipid vesicle co-precipitation assay

The phospholipid vesicle co-precipitation assay was performed as described previously 24. The required amounts of phospholipids were transferred into glass vials and were

dried under a gentle stream of Nitrogen gas. The required amount of binding buffer

(20mM Tris [pH7.5], 150mM NaCl, 1mM EDTA) was added, and samples incubated for

3 h at 52ºC. After this incubation, hrSLP-2 protein was added to the lipid vesicles

solution at a 5:1 ratio of lipid to protein and was incubated for 30 min at 37ºC. Samples

were centrifuged at 40,000 rpm for 10 min at room temperature; the supernatant was

removed; and pellets were resuspended in binding buffer. Laemmli sample buffer was

added to supernatant and pellet fractions, boiled for 7 min, and run on an SDS-PAGE.

Bovine heart cardiolipin was purchased from Sigma; chicken egg L-α-

phosphatidylcholine (PC), L-α-phosphadidylethanolamine (PE) and L-α-

phosphadidylglycerol, porcine brain L-α-phosphatidylserine, and soy L-α-

phosphadidylinositol were purchased from Avanti Polar Lipids, Inc. (Alabaster AB).

2.3 Results

2.3.1 SLP-2 is localized mostly in mitochondria

Studies by different groups, including our own, have shown that SLP-2 in human cells is

expressed in two pools 11, 12, 22. The major pool of SLP-2 is associated with the

mitochondrial inner membrane, whereas a quantitatively minor pool is associated with

the plasma membrane. This compartmentalization was corroborated biochemically by

subcellular fractionation of human T cells (Fig. 2.1a). SLP-2 was detected in membrane

and organelle/cytoskeleton fractions but not in nuclear or cytosolic compartments. Porin

(voltage-dependent anion channel [VDAC]), a protein detected in mitochondrial and

plasma membranes, showed partitioning pattern similar to that of SLP-2.

To study the role of SLP-2, we developed stable transfected cells in which SLP-2

expression can be tightly regulated using a doxycycline-inducible promoter coding for a

C-terminally GFP-tagged SLP-2 protein (SLP-2-GFP). This system allowed us to control

60

Figure 2.1: The major pool of SLP-2 in human T cells is located in mitochondria

(a) Subcellular fractions of Jurkat T cells were isolated by differential centrifugation and

were immunoblotted for SLP-2. Serial blotting for GAPDH, Histone H1, CD45, and

talin was performed to control for the quality of the fractions. Blotting of porin (VDAC),

another protein with a major subcellular pool in mitochondrial and a smaller pool in the

plasma membrane, is also shown. (b) Mitochondrial and cytosolic fractions of stable T

cell SLP-2-GFP transfectants expressing low (SLP-2lo) and high (SLP-2hi) levels of SLP-

2-GFP were isolated by differential centrifugation and immunoblotted for SLP-2,

electron transport complex III, and β-actin. Note that in addition to endogenous SLP-2

and SLP-2-GFP forms, we detected bands likely reflecting intermediate SLP-2

degradation fragments. (c) Stably expressed SLP-2-GFP colocalizes with MitoTracker

Red CMXRos by confocal microscopy. Bars, 5 µm (d) Three-dimensional reconstitution

image analysis. A lateral view of a T cell attached to a glass slide is shown. The yellow

units represent mitochondria and the mitochondrial network. Images are representative

of more than 100 cells from more than 5 independent experiments.

61

Figure 2.1

62

the levels of SLP-2 in human cells (T cells for these experiments) from very low (basal

levels of expression by Jurkat T cells stably transfected with this cDNA in the absence of

doxycycline; referred to below as SLP-2lo) to high (10-100 times higher expression upon

addition of doxycycline; referred to below as SLP-2hi). With these cells, we corroborated

the predominant localization of SLP-2 in mitochondria both by biochemical fractionation

(Fig. 2.1b) and by confocal microscopy, in which SLP-2-GFP colocalized with the

mitochondrial dye MitoTracker Red (Fig. 2.1c and 2.1d). Such an intracellular

distribution was determined by an N-terminal mitochondrial targeting sequence 12, 22, as

evidenced by the fact that SLP-2-GFP deletion mutants lacking the N-terminus abrogated

the mitochondrial localization, whereas expression of the SLP-2 N-terminal domain fused

to GFP was sufficient to determine mitochondrial localization of this protein (data not

shown 12).

2.3.2 Upregulation of SLP-2 expression increases mitochondrial biogenesis

To learn about the function of SLP-2, given its mitochondrial localization, we first

examined the effect of induction of SLP-2 expression on mitochondrial mass by using the

system mentioned above. We found that upregulation of SLP-2 expression, but not of a

control protein, induced a significant increase in the mass of metabolically-active

mitochondria (P < 0.01), as determined with the intact mitochondria label MitoTracker

Red® (Fig. 2.2a and b), and a significant increase in the number of mitochondria per T

cell (P < 0.001) (Fig. 2.2c and 2d). Such mitochondrial biogenesis required appropriate

targeting of SLP-2 to mitochondria, as evidenced by the fact that overexpression of a

SLP-2-GFP mutant lacking the N terminal mitochondrial targeting sequence (ΔNSLP-2)

did not induce a significant increase in the number of mitochondria (Fig. 2.2c and 2.2d),

although a slight increase over baseline was observed, likely due to limited transport of

the mutant as a dimer with endogenous full length SLP-2. Furthermore, the increased

mitochondrial mass observed after upregulation of SLP-2 expression was corroborated by

flow cytometry for primary human leukocytes from peripheral blood in the transition

63

Figure 2.2: Induction of SLP-2 expression triggers mitochondrial biogenesis in

human T cells.

(a and b) T cells expressing high levels of SLP-2 (SLP-2hi) and GFP-expressing control

cells, which contain lower levels of SLP-2 (SLP-2lo), were stained with MitoTracker Red,

and the signal was quantified in each cell. Bars, 5 µm. (c) Mitochondria visualized by

transmission electron microscopy were counted for 30 cell slices of SLP-2hi T cells, SLP-

2lo T cells, or T cells expressing a SLP-2 mutant in which the N-terminal mitochondrial

targeting sequence was deleted (ΔNSLP-2). Asterisks indicate significant differences

(***, P<0.001; **, P<0.01) as determined by Student’s t test. (d) Representative

transmission electron microscopy images of SLP-2lo, SLP-2hi and ΔNSLP-2 T cells.

Mitochondria are indicated by white stars. Bars, 2 µm. (e) Mitochondrial mass was

measured by flow cytometry in resting (shaded histogram) and activated (filled

histogram) human peripheral blood mononuclear cells after labeling with MitoTracker

Red. The result for the unstained control is shown by a dotted line. (f) Immunoblotting

of PBMC whole-cell lysates for SLP-2 demonstrated that SLP-2 levels were upregulated

after blasting. β−Actin blotting was used as a loading control. Densitometric units of

SLP-2 for each group normalized to actin levels are shown.

64

Figure 2.2

65

from resting state (in which they express low levels of SLP-2) to the primed blasted state

(in which they express high levels of SLP-2) (Fig. 2.2e and 2f).

Mitochondrial biogenesis requires de novo synthesis of cardiolipin, a phospholipid in the

mitochondrial membrane. Since upregulation of SLP-2 expression was associated with

increased mitochondrial biogenesis, we next tested whether SLP-2 overexpression

increased cardiolipin synthesis. A 37% increase in cardiolipin content (as a percentage of

total phospholipid phosphorus mass) was detected in SLP-2hi expressing cells (Fig. 2.3a).

This finding was corroborated in human PBMCs transitioning from resting state (i.e.,

expressing low levels of SLP-2) to the activated state (i.e., expressing high levels of SLP-

2) by flow cytometry with the cardiolipin dye nonyl-acridine orange (Fig. 2.3b). Since

the loading and retention of this dye are sensitive to changes in mitochondrial potential 25,

we verified that the increase in cardiolipin content was due to de novo synthesis of this

phospholipid by using radiolabeling assays incubating SLP-2lo and SLP-2hi cells with 3H-

acetate and 14C-linoleic acid, both of which are incorporated into newly synthesized

cardiolipin. SLP-2hi cells showed higher levels of radiolabeled cardiolipin under both

incubation conditions than did SLP-2lo cells (P < 0.05) (Fig. 2.3c and 2.3d). Finally, we

used real time RT-PCR to look at the levels of phosphatidylglycerol phosphate synthase

mRNA and cardiolipin synthase mRNA, since these resulting enzymes are involved in

the final steps of cardiolipin biosynthesis. As expected, the SLP-2hi cells had consistently

higher transcript levels of both enzymes (Fig. 2.3e and 2.3f), consistent with a state of

activation of lipid biosynthesis at the transcriptional level. Together, these data led us to

conclude that SLP-2 regulates cardiolipin synthesis, which is required for the formation

of mitochondrial membranes. To examine the requirement of cardiolipin synthase

upregulation for mitochondrial biogenesis in SLP-2hi cells, PBMCs were activated with 1

ng/mL PMA and 100 ng/mL ionomycin in the presence of 4 µM triacsin C, an inhibitor

of cardiolipin synthase. Under these conditions, the increase in mitochondrial mass upon

T cell activation was blocked (Fig. 2.3g), indicating a requirement for cardiolipin

synthesis in mitochondrial biogenesis induced by SLP-2 upregulation.

66

Figure 2.3: Induction of SLP-2 expression increases de novo cardiolipin

biosynthesis, nuclear transcription programs, and mitochondrial DNA replication.

(a) Upregulation of SLP-2 expression in Jurkat T cells induces cardiolipin synthesis.

Upregulation of SLP-2 expression in T cells (SLP-2hi) (open bar) was induced with

doxycycline and was compared with baseline expression of SLP-2 (SLP-2lo) (filled bar).

The pool size of cardiolipin was determined and was expressed as a percent of the total

phospholipid phosphorus mass. (b) Cardiolipin levels in naïve (shaded histogram) and in

vitro-activated (filled histogram) PBMCs were measured by flow cytometry after staining

with nonyl-acridine orange. The result for the unstained control is shown by a dotted

line. (c and d) Incorporation of [3H]-acetate (c) and [14C]-linoleic acid (d) into

cardiolipin in SLP-2lo and SLP-2hi T cells. Data are shown as dpm/mg total cellular

protein. (e and f) Real-time PCR analysis of the transcript levels of phosphatidylglycerol

phosphate synthase (e) and cardiolipin synthase (f) in SLP-2lo and SLP-2hi T cells. (g)

The mitochondrial mass in naïve (shaded histograms) and in vitro-activated PBMC in the

absence (thin line) or presence (thick line) of triacsin C was measured by flow cytometry

after staining with MitoTracker Red. The result for the unstained control is shown by a

dotted line. (h and i) Real-time PCR analysis of the transcript levels of PGC-1α (h) and

mitochondrial DNA content (i) in SLP-2lo and SLP-2hi T cells. The results shown here

are representative of at least 3 independent experiments. *: P < 0.05.

67

Figure 2.3

68

In addition to membrane formation, mitochondrial biogenesis also requires activation of a

nuclear transcription program that encodes the majority of mitochondrial proteins. This

nuclear transcriptional program is dependent on PGC-1α, a transcriptional co-activator

and master regulator of mitochondrial biogenesis 26. Thus, we performed real-time RT-

PCR to measure the levels of PGC-1α mRNA and found that these were increased in

SLP-2hi cells (Fig. 2.3h), supporting the idea that upregulation of SLP-2 expression is

associated with an increase in the expression of mitochondrially targeted genes. Of

interest, induction of SLP-2 expression also resulted in significant increases in the levels

of OPA-1 and mitofusin-2 (P < 0.05), both integral mitochondrial membrane proteins

associated with mitochondrial fusion, but not in the levels of Drp1 (involved in

mitochondrial fission), GAPDH, or cytochrome c (data not shown). However, the

increase in OPA-1 and mitofusin-2 did not translate functionally into enhanced

mitochondrial fusion.

Mitochondrial biogenesis requires replication of mitochondrial DNA. As expected from

the data presented above, we found that upregulation of SLP-2 correlated with a 70%

increase in the level of mitochondrial DNA (Fig. 2.3i). Taken together, these findings

lead us to conclude that higher levels of SLP-2 stimulate cardiolipin synthesis and

mitochondrial biogenesis.

To determine the kinetics of mitochondrial biogenesis in response to SLP-2 upregulation,

MitoTracker Red was used to measure mitochondrial mass in PBMCs stimulated with

PMA and ionomycin for 48 h. During the first 18 h of stimulation, the cells had a slight

decrease in mitochondrial mass, which was recovered by 24 h, and mitochondrial mass

was elevated by 48 h of stimulation (Fig. 2.4a). During this time, SLP-2 and PHB-1 were

both found to be upregulated as early as 1 h after stimulation, while PGC-1α was elevated

by 18 h poststimulation (Fig. 2.4b). This preceded the increase in mitochondrial

biogenesis detected by MitoTracker Red at 24 h poststimulation, implying that SLP-2

upregulation in response to T cell activation precedes PGC-1α upregulation and the

subsequent increase in mitochondrial biogenesis.

69

Figure 2.4: SLP-2 upregulation precedes PGC-1α upregulation and mitochondrial

biogenesis during T cell activation.

(a) PBMCs were stimulated with 1ng/mL PMA and 100ng/mL ionomycin for as long as

48 h. At the indicated time points, cells were stained with Mitotracker Red and were

analyzed for mitochondrial mass by flow cytometry. Shaded histograms represent

unstimulated cells while filled histograms represent stimulated cells. (b) Whole-cell

lysates were made at each indicated time point during stimulation with PMA and

ionomycin. The lysates were separated by SDS-PAGE and were serially blotted for

PGC-1α, PHB-1, SLP-2, and actin.

70

Figure 2.4

71

2.3.3 SLP-2 interacts with prohibitins and binds cardiolipin-enriched microdomains

Next, we examined how upregulation of SLP-2 expression may induce mitochondrial

biogenesis. Emerging data suggest that mitochondrial biogenesis is coordinated by a

mechanism involving PHB-1 and -2 7, 27. These proteins localize within phospholipid-

enriched, detergent-insoluble microdomains in mitochondrial membranes, and their

functional interactome includes genes such as Ups1 and Gep1 that are linked to the

synthesis of mitochondrial membrane phospholipids cardiolipin and

phosphatidylethanolamine, respectively 7, 27. We noticed that Saccharomyces cerevisiae

PHB-2 has a putative transmembrane domain (as predicted by the TMHMM

[transmembrane hidden Markov model] algorithm 28), but neither mouse nor human

PHBs have such a domain. Of interest, S. cerevisiae does not have a homolog of SLP-2,

the closest being PHB-1, with a clustal W score of 15 29. This suggested that, in

mammals, SLP-2 may act to link PHBs and phospholipid-enriched detergent-insoluble

microdomains in mitochondrial membranes. Such a possibility is consistent with the

observations that PHBs are upregulated in parallel with SLP-2 during activation 30 (Fig.

2.4b) and that SLP-2 interacts with PHBs 12.

We first confirmed that human SLP-2 interacted with PHB-1 and PHB-2 in T cells using

standard coprecipitation experiments (Fig. 2.5a). We found that immunoprecipitates of

SLP-2 contained both PHB-1 and PHB-2, as previously reported 12. Furthermore,

coprecipitation of PHB-1, PHB-2 and SLP-2 increased as the expression of SLP-2

increased with increasing levels of doxycycline in the culture (Fig. 2.5b). To demonstrate

that SLP-2 plays a role in linking prohibitins to phospholipid-enriched detergent-

insoluble microdomains, we isolated mitochondria from SLP-2lo and SLP-2hi cells and

extracted mitochondrial membranes with Na2CO3. We found a higher association of

PHB-1 with the mitochondrial membrane fraction in the SLP-2hi cells, although the total

level of PHB-1 was similar in the SLP-2lo and SLP-2hi cells (Fig. 2.5c).

Next, we generated human recombinant SLP-2 (hrSLP-2) and examined the profile of

72

Figure 2.5: SLP-2 interacts with prohibitins.

(a) Immunoprecipitates with an antiserum against human SLP-2 or with a pre-immune

serum from SLP-2-expressing Jurkat T cells were serially blotted for prohibitin 1 (PHB-

1), PHB-2 or SLP-2. WCL, whole cell lysate; IP, immunoprecipitating antiserum; Beads,

no cell lysate control; Pre-Imm, control immunoprecipitation with pre-immune serum.

(b) Immunoprecipitates and whole-cell lysates from SLP-2-GFP cells induced to express

increasing amounts of SLP-2 by increasing concentration of doxycycline were collected

and blotted as for Fig. 2.4b. (c) Mitochondria were isolated from SLP-2hi and SLP-2lo

cells and treated with Na2CO3 in order to examine the membrane association of

prohibitins in the presence of elevated SLP-2 expression. The fractions were serially

blotted for PHB-1 and SLP-2.

73

Figure 2.5

74

Figure 2.6: SLP-2 binds cardiolipin.

(a) Coprecipitation of hrSLP-2 with cardiolipin (CL) vesicles but not with other

phopholipid vesicles as shown by Western blotting of pellet (P) and supernatant (S)

fractions. PC, phosphatidylcholine; PE, phosphatidylethanolamine; PI,

phosphatidylinositol; PS, phosphatidylserine; PG, phosphatidylglycerol; Total protein,

hrSLP-2 aliquot as used for coprecipitation. (b) Titration of hrSLP-2 coprecipitation with

CL in mixed CL-PC-PE vesicles at the indicated ratios. An additional group containing

twice the amount of hrSLP-2 (cardiolipin + 2xSLP-2) was added.

75

Figure 2.6

76

interaction of this protein with different phospholipids using a phospholipid vesicle co-

precipitation assay 24. In this assay, molecules that interact with phospholipids

cosediment with the vesicles whereas non-interacting proteins remain in the supernatant.

We found that hrSLP-2 coprecipitated with cardiolipin vesicles (Fig. 2.6a, P [pellet

fraction]), whereas most of the hrSLP-2 remained in solution when vesicles made of

other phospholipids found in mitochondria (phosphatidylcholine and

phosphatidylethanolamine) or vesicles made of mostly non-mitochondrial phospholipids

(phosphatidylinositol and phosphatidylserine) were used (Fig. 2.6a; S [supernatant

fraction]). Of interest, hrSLP-2 bound to phosphatidylglycerol vesicles in lower amounts,

a finding likely due to the fact that cardiolipin is a composite of two phosphatidylglycerol

molecules. To further investigate the association of hrSLP-2 with cardiolipin, we

prepared phospholipid vesicles containing cardiolipin along with the two other major

phospholipids in mitochondria, phosphatidylcholine and phosphatidylethanolamine at

ratios of 1:1:1 (33% CL, 33% PC, 33% PE), 2.5:0.25:0.25 (83% CL, 8% PC, 8% PE) and

2.75:0.12:0.12 (92% CL, 4% PC, 4% PE). The interaction between hrSLP-2 and

cardiolipin was selective and titrated with the enrichment of this phospholipid in vesicles

also containing phosphatidylcholine and phosphatidylethanolamine (Fig. 2.6b). These

data demonstrate that SLP-2 binds to the prohibitin complex as well as to cardiolipin-

enriched membrane microdomains, potentially mediating the association between

prohibitins and mitochondrial membranes.

2.3.4 Upregulation of SLP-2 expression is associated with increased mitochondrial functions

To examine the functional effects of upregulation of SLP-2 expression, we first

determined the activity of citrate synthase, an enzyme of the citric acid cycle, which is

localized to the mitochondrial matrix. We found that SLP-2hi cells showed significantly

increased citrate synthase activity (P < 0.01), (Fig. 2.7a). Next, we measured the activity

of the electron transport complexes in the respiratory chain. We found that SLP-2hi cells

had significantly higher activities of NADH dehydrogenase and succinate dehydrogenase

(P < 0.05), complex I and II of the electron transport chain (Fig. 2.7b and 2.7c). In

77

Figure 2.7: Induction of SLP-2 expression is associated with higher mitochondrial

activity.

(a) Citrate synthase (CS) activity was measured in SLP-2lo (filled bar) and SLP-2hi (open

bar) Jurkat T cells. Data are means and standard deviations (SD) for triplicate samples;

**P < 0.01. Data are expressed as nmol/min/mg mitochondrial protein. (b) NADH

dehydrogenase was measured in SLP-2lo (filled bar) and SLP-2hi (open bar) Jurkat T

cells, plotted as in panel a, *P < 0.05. (c) Succinate dehydrogenase (SDH) was measured

in SLP-2lo and SLP-2hi Jurkat T cells, plotted as in panel a; *P < 0.05. Data are expressed

as nmol/min/mg mitochondrial protein. (d) Mitochondrial and total cellular ATP levels

were measured in SLP-2lo and SLP-2hi Jurkat T cells. Means and standard deviations

(SDs) for triplicate samples are plotted. *P < 0.05, **P < 0.01. (e and f) IL-2 levels in

culture supernatants of SLP-2lo (e) and SLP-2hi (f) Jurkat T cells pre-treated or not with

oligomycin (4 µM) and stimulated with antigen-presenting cells and SEE for 24 h.

Means and standard deviations for triplicate samples are plotted. Asterisks indicate

significant differences (*P < 0.05; ***P < 0.001) as determined by analysis of variance

(ANOVA) with a Bonferroni posttest. (g) Oxygen consumption was measured in SLP-2lo

and SLP-2hi T cells in media containing high (11mM) or low (0.4mM) glucose. Means

and standard deviations for triplicate samples are plotted. Asterisks indicate significant

differences (*P < 0.05; ***P < 0.001) as determined by analysis of variance (ANOVA)

with a Bonferroni posttest. (h) Mitochondria were isolated from SLP-2lo and SLP-2hi

cells, separated by SDS-PAGE, and serially blotted for subunits of each complex of the

respiratory chain as well as SLP-2.

78

Figure 2.7

79

addition, mitochondrial and whole-cell ATP levels in resting SLP-2hi T cells were

significantly higher than those in SLP-2lo T cells (P < 0.05) (Fig. 2.7d). Such an increase

in ATP levels in SLP-2hi T cells correlated with increased resistance to depletion of

mitochondrial ATP with oligomycin, as evidenced by increased T cell responses,

indicated by IL-2 production. In these experiments, oligomycin-treated SLP-2lo T cells

and SLP-2hi T cells were stimulated with antigen-presenting cells and staphylococcal

enterotoxin E (SEE), and the production of interleukin-2 (IL-2) was determined after 24 h

of stimulation. Oligomycin treatment significantly reduced the IL-2 response to SEE of

SLP-2lo T cells (P < 0.001) (Fig. 2.7e) but did not affect the response of SLP-2hi T cells

(Fig. 2.7f). Consistent with our previous findings, SLP-2hi T cells had a higher IL-2

response than SLP-2lo T cells 11.

Given that the generation of ATP by OXPHOS is linked to the respiratory chain in the

mitochondrial inner membrane, we examined the effect of modulating SLP-2 expression

on oxygen consumption by T cells. We found that SLP-2hi T cells had slightly but

significantly higher oxygen consumption than SLP-2lo T cells (P < 0.05). In a low-

glucose medium, SLP-2hi T cells had much higher oxygen consumption than SLP-2lo T

cells (P < 0.01) (Fig. 2.7g). This is expected, since cells rely more heavily on OXPHOS

under low-glucose conditions. Therefore, we concluded that upregulation of SLP-2

expression is associated with increased oxygen consumption and OXPHOS, leading to

increased ATP stores in the cell.

Cardiolipin plays an essential role in the mitochondrial protein import machinery 31, and

as such, the increase in cardiolipin seen in response to SLP-2 upregulation could lead to

an increased rate of protein transport into the mitochondria. If so, the increased

enzymatic rates we detected in the SLP-2hi cells could be due to elevated protein levels in

the mitochondria. To test this, mitochondria were isolated from SLP-2lo and SLP-2hi

cells, and lysates from these mitochondria were blotted for representative subunits of

each complex of the respiratory chain. SLP-2lo and SLP-2hi cells had similar protein

levels for each complex of the respiratory chain (Fig. 2.7h), indicating that the increase in

80

enzymatic activity seen in the SLP-2-overexpressing cells was not due to increased levels

or increased translocation of electron transport complexes into mitochondria.

2.3.5 Upregulation of SLP-2 expression increases T cell responses and resistance to apoptosis

In light of the significantly enhanced mitochondrial biogenesis and function associated

with SLP-2 upregulation, we examined the physiological effects of these changes by

looking at the functional response of primary human T cells in which SLP-2 expression

was upregulated. As shown in Fig. 2.8a, peripheral blood T cells from normal volunteers

transiently nucleofected with doxycycline-inducible SLP-2-GFP cDNA and expressing

high levels of SLP-2 produced significantly higher levels of IL-2 (P < 0.05) upon

stimulation with SEE and antigen-presenting cells. In addition, these cells showed

increased resistance to apoptosis through the intrinsic pathway (Fig. 2.8b). Such

resistance to apoptosis was also observed in response to actinomycin D in Jurkat SLP-2hi

T cells (Fig. 2.8c). As an additional marker of apoptosis, actinomycin D-induced cell

death was also measured by an increase in annexin V labeling. In agreement with the

loss of transmembrane potential as measured by MitoTracker Red, SLP-2hi cells also

showed lower annexin V staining, which is indicative of a protection against the

induction of the intrinsic apoptosis pathway (Fig. 2.8d and e). Thus, consistent with the

increased mitochondrial biogenesis and function, upregulation of SLP-2 expression

enhances cell function.

81

Figure 2.8: SLP-2 upregulation increases T cell responses and protects against

apoptosis.

(a) Human peripheral blood T cells nucleofected with doxycycline-inducible SLP-2-GFP

and expressing high levels of SLP-2 (SLP-2hi) upon doxycycline-induction produce

significantly more IL-2 in response to antigen-presenting cells and SEE stimulation than

control (noninduced) SLP-2-GFP transfected T cells. **P < 0.01; * P < 0.05. (b) SLP-

2hi primary T cells are less susceptible to actinomycin D-induced apoptosis. Human

PBMCs were transiently transfected with doxycycline-inducible SLP-2-GFP and cultured

with (SLP-2hi) (shaded histogram) or without (SLP-2lo) (open histogram) doxycycline

and with actinomycin D (5µg/mL) for 3 h at 37°C. Cell death was measured by

MitoTracker Red staining, which was analyzed by flow cytometry. (c) Upregulation of

SLP-2 expression in doxycycline-inducible SLP-2-GFP transfected Jurkat T cells results

in increased resistance to actinomycin D-induced apoptosis. SLP-2hi (shaded histogram)

and SLP-2lo (open histogram) T cells were treated with 5µg/mL actinomycin D for 3 h at

37°C. (d) SLP-2hi (open bars) and SLP-2lo (filled bars) T cells were treated with

actinomycin D and stained for annexin V, an alternative marker of apoptosis.

***P<0.001. (e) One representative flow cytometry plot of annexin V staining of SLP-

2hi (shaded histogram) and SLP-2lo (open histogram) cells after induction of apoptosis

with actinomcyin D. The result for the unstained control is shown by a dotted line. The

data are representative of three independent experiments.

82

Figure 2.8

83

2.4 Discussion The functions of stomatins and other proteins sharing the

stomatin/prohibitin/flotillin/Hflk (SPFH) domain remain enigmatic. It has been proposed

that through this domain, stomatins, PHBs and flotillins interact with different cell

membranes and facilitate signalling. We report here that SLP-2, a mitochondrial member

of the stomatin family, binds cardiolipin and may help to recruit PHBs to cardiolipin-

enriched microdomains. Furthermore, upregulation of SLP-2 expression is associated

with increased mitochondrial biogenesis and function, ultimately resulting in enhanced

effector cell function.

Our data show that SLP-2 binds cardiolipin-enriched microdomains in a way that titrates

with the content of cardiolipin in phospholipid membranes. The mitochondrial

membrane is composed of 40% phosphatidylcholine, 30% phosphatidylethanolamine and

10-15% phosphatidylinositol and cardiolipin 32. But the cardiolipin content in the

mitochondrial membrane is not homogenously distributed. Our data suggest that SLP-2

can bind to clusters of cardiolipin, i.e., microdomains with a much higher localized

cardiolipin concentration. This selective binding of SLP-2 to cardiolipin is reminiscent

of the observation that two other members of the stomatin/PHB/flotillin/Hflk superfamily

(MEC-2 and podocin) bind cholesterol 33 and leads us to propose that SPFH domain-

containing proteins bind to selective phospholipid-enriched membrane microdomains and

may act as markers that recruit other proteins to the lipid microdomains. How these

proteins bind lipids is still unknown. The binding of SLP-2 to cardiolipin in the

mitochondrial inner membrane is likely independent of transmembrane insertion, since

SLP-2 lacks a putative transmembrane domain 8.

Upregulation of SLP-2 expression increases cardiolipin synthesis and mitochondrial

biogenesis. Recent evidence indicates that these two processes are tightly coordinated 27,

likely through the PHB functional interactome, which includes genes involved in

phospholipid synthesis, such as Ups1, whose deletion, in yeast, translates into a marked

decreased in cardiolipin content 7. Although we have not shown a direct interaction

among PHBs, SLP-2 and cardiolipin, our data suggest that SLP-2 may bring PHBs to

84

cardiolipin and may contribute to the organization of cardiolipin-enriched microdomains

in the mitochondrial inner membrane 34. Data from our laboratory using T cell-specific

SLP-2 knockout mice seem to corroborate this model (Chapter 3). This possible

mechanism accommodates the upregulation of both SLP-2 and PHBs during T cell

activation 30 and the detection of both proteins in detergent-insoluble microdomains of

cell membranes (i.e., cardiolipin-enriched microdomains in the mitochondrial inner

membrane and cholesterol-enriched microdomains in the plasma membrane) 11, 35-37. In

this context, an increase in SLP-2 levels as a result of cell activation would increase the

interaction of PHBs with cardiolipin in the mitochondrial inner membrane and activate

the PHB functional interactome, leading to de novo cardiolipin synthesis.

Using human T cell activation as a model, we have shown that upregulation of SLP-2

expression is a molecular step between mitochondrial biogenesis and cell activation and

function. SLP-2 does not seem to be critical under resting conditions, as suggested by the

absence or very low level of SLP-2 expression in resting naïve T cells. However, during

T cell activation, SLP-2 expression is dramatically upregulated, and this increases T cell

function, leading to increased IL-2 production and increased resistance to apoptosis

through the intrinsic pathway. PHBs are also upregulated during T cell activation 30,

whereas PHB-2 deletion decreases the ability of cells to proliferate and increases their

sensitivity to apoptosis through the intrinsic pathway 38. These observations corroborate

the model proposed here by which SLP-2 - PHB complexes would act in concert to

regulate mitochondrial biogenesis and function during T cell activation. Of interest,

modulation of PHB-2 levels also alters OPA-1 levels (e.g., cells lacking PHB-2 have

impaired processing of OPA-1 34), and in our experiments, T cells overexpressing SLP-2

have increased OPA-1 levels.

The increased mitochondrial function observed in SLP-2 overexpressing cells is in line

with the observation that SLP-2 controls the stability and the assembly of electron

transport complexes 39, 40. This is further corroborated by emerging data from our

laboratory using T cell-specific SLP-2-deficient mice, showing that a lack of SLP-2 is

associated with decreased recruitment of PHB-1 to cardiolipin-enriched mitochondrial

membranes, as well as a loss of complex I subunits (Chapter 3). Optimal assembly of

85

these complexes may require the formation of specialized membrane microdomains, and

this would correlate with increased respiratory chain function. Of interest, SLP-2 is

required for mitochondrial hyperfusion 41, a phenomenon linked with increased metabolic

demands and cellular stress. Levels of both OPA-1 and mitofusin-2, proteins required

for this process, were increased upon SLP-2 upregulation, but cytochrome c levels were

not. It is possible that SLP-2 expression specifically increases the expression of proteins

involved in mitochondrial fusion, or prevents these proteins from degradation, without

altering expression of other mitochondrial proteins.

Taken together, the evidence presented here leads us to propose that binding of SLP-2 to

cardiolipin recruits PHBs, helping to form cardiolipin-enriched membrane microdomains

in which the respiratory chain components are optimally assembled. The formation of

these microdomains is likely the result of the self-oligomerizing properties of SLP-2 8.

The increase in respiratory chain function may then induce further cardiolipin synthesis

through its effect on the pH gradient across the mitochondrial inner membrane 42,

enhancing mitochondrial biogenesis. The result of this proposed mechanism is the

provision of the enhanced mitochondrial function required for an optimal T cell response.

How the increased mitochondrial membrane formation increases nuclear transcription

and mitochondrial DNA replication, both required to increase mitochondrial numbers,

remains to be determined. A retrograde nuclear signalling pathway in yeast is starting to

emerge, but little is known about this pathway in mammalian cells. Specific signals may

involve mitochondrial stress, since it has been shown that mitochondrial dysfunction

increases nuclear transcription 26. In this context, SLP-2 may play a role by regulating

mitochondrial membrane biogenesis and its subsequent effect of calcium-dependent

signalling, which has been linked to the activation of retrograde signalling 43.

In summary, we have shown that increased levels of SLP-2, seen, for example, during

lymphocyte activation, stimulate mitochondrial biogenesis and function, providing for the

energetic requirements of activation. Such a function involves the binding of SLP-2 and

its interacting partners (PHBs) to cardiolipin to form membrane microdomains in which

86

respiratory complexes are optimally assembled. The ultimate result of this function is the

net enhancement of effector cell functions.

87

2.5 References

1. Frauwirth, K.A., Riley, J.L., Harris, M.H., Parry, R.V., Rathmell, J.C., Plas, D.R., Elstrom, R.L., June, C.H. & Thompson, C.B. The CD28 signaling pathway regulates glucose metabolism. Immunity 16, 769-777 (2002).

2. Frauwirth, K.A. & Thompson, C.B. Regulation of T lymphocyte metabolism. J Immunol 172, 4661-4665 (2004).

3. Fox, C.J., Hammerman, P.S. & Thompson, C.B. Fuel feeds function: energy metabolism and the T-cell response. Nat Rev Immunol 5, 844-852 (2005).

4. Carpentieri, U. & Sordahl, L.A. Respiratory and calcium transport functions of mitochondria isolated from normal and transformed human lymphocytes. Cancer Res 40, 221-224 (1980).

5. Nagy, G., Barcza, M., Gonchoroff, N., Phillips, P.E. & Perl, A. Nitric oxide-dependent mitochondrial biogenesis generates Ca2+ signaling profile of lupus T cells. J Immunol 173, 3676-3683 (2004).

6. D'Souza, A.D., Parikh, N., Kaech, S.M. & Shadel, G.S. Convergence of multiple signaling pathways is required to coordinately up-regulate mtDNA and mitochondrial biogenesis during T cell activation. Mitochondrion 7, 374-385 (2007).

7. Osman, C., Haag, M., Potting, C., Rodenfels, J., Dip, P.V., Wieland, F.T., Brugger, B., Westermann, B. & Langer, T. The genetic interactome of prohibitins: coordinated control of cardiolipin and phosphatidylethanolamine by conserved regulators in mitochondria. J Cell Biol 184, 583-596 (2009).

8. Wang, Y. & Morrow, J.S. Identification and characterization of human SLP-2, a novel homologue of stomatin (band 7.2b) present in erythrocytes and other tissues. J Biol Chem 275, 8062-8071 (2000).

9. Owczarek, C.M., Treutlein, H.R., Portbury, K.J., Gulluyan, L.M., Kola, I. & Hertzog, P.J. A novel member of the STOMATIN/EPB72/mec-2 family, stomatin-like 2 (STOML2), is ubiquitously expressed and localizes to HSA chromosome 9p13.1. Cytogenet Cell Genet 92, 196-203 (2001).

10. Green, J.B. & Young, J.P. Slipins: ancient origin, duplication and diversification of the stomatin protein family. BMC evolutionary biology 8, 44 (2008).

11. Kirchhof, M.G., Chau, L.A., Lemke, C.D., Vardhana, S., Darlington, P.J., Marquez, M.E., Taylor, R., Rizkalla, K., Blanca, I., Dustin, M.L. & Madrenas, J. Modulation of T cell activation by stomatin-like protein 2. J Immunol 181, 1927-1936 (2008).

88

12. Da Cruz, S., Parone, P.A., Gonzalo, P., Bienvenut, W.V., Tondera, D., Jourdain, A., Quadroni, M. & Martinou, J.C. SLP-2 interacts with prohibitins in the mitochondrial inner membrane and contributes to their stability. Biochim Biophys Acta 1783, 904-911 (2008).

13. Baroja, M.L., Luxenberg, D., Chau, T., Ling, V., Strathdee, C.A., Carreno, B.M. & Madrenas, J. The inhibitory function of CTLA-4 does not require its tyrosine phosphorylation. J Immunol 164, 49-55 (2000).

14. Arp, J., Kirchhof, M.G., Baroja, M.L., Nazarian, S.H., Chau, T.A., Strathdee, C.A., Ball, E.H. & Madrenas, J. Regulation of T-cell activation by phosphodiesterase 4B2 requires its dynamic redistribution during immunological synapse formation. Mol Cell Biol 23, 8042-8057 (2003).

15. Hauff, K., Linda, D. & Hatch, G.M. Mechanism of the elevation in cardiolipin during HeLa cell entry into the S-phase of the human cell cycle. Biochem J 417, 573-582 (2009).

16. Lowry, O.H., Rosebrough, N.J., Farr, A.L. & Randall, R.J. Protein measurement with the Folin phenol reagent. J Biol Chem 193, 265-275 (1951).

17. Rouser, G., Fkeischer, S. & Yamamoto, A. Two dimensional thin layer chromatographic separation of polar lipids and determination of phospholipids by phosphorus analysis of spots. Lipids 5, 494-496 (1970).

18. Hatch, G.M. & McClarty, G. Regulation of cardiolipin biosynthesis in H9c2 cardiac myoblasts by cytidine 5'-triphosphate. J Biol Chem 271, 25810-25816 (1996).

19. Livak, K.J. & Schmittgen, T.D. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods 25, 402-408 (2001).

20. Cote, H.C., Brumme, Z.L., Craib, K.J., Alexander, C.S., Wynhoven, B., Ting, L., Wong, H., Harris, M., Harrigan, P.R., O'Shaughnessy, M.V. & Montaner, J.S. Changes in mitochondrial DNA as a marker of nucleoside toxicity in HIV-infected patients. N Engl J Med 346, 811-820 (2002).

21. Bueno, C., Lemke, C.D., Criado, G., Baroja, M.L., Ferguson, S.S., Rahman, A.K., Tsoukas, C.D., McCormick, J.K. & Madrenas, J. Bacterial superantigens bypass Lck-dependent T cell receptor signaling by activating a Galpha11-dependent, PLC-beta-mediated pathway. Immunity 25, 67-78 (2006).

22. Hajek, P., Chomyn, A. & Attardi, G. Identification of a novel mitochondrial complex containing mitofusin 2 and stomatin-like protein 2. J Biol Chem 282, 5670-5681 (2007).

89

23. Chau, T.A., McCully, M.L., Brintnell, W., An, G., Kasper, K.J., Vines, E.D., Kubes, P., Haeryfar, S.M., McCormick, J.K., Cairns, E., Heinrichs, D.E. & Madrenas, J. Toll-like receptor 2 ligands on the staphylococcal cell wall downregulate superantigen-induced T cell activation and prevent toxic shock syndrome. Nat Med 15, 641-648 (2009).

24. Bakolitsa, C., de Pereda, J.M., Bagshaw, C.R., Critchley, D.R. & Liddington, R.C. Crystal structure of the vinculin tail suggests a pathway for activation. Cell 99, 603-613 (1999).

25. Jacobson, J., Duchen, M.R. & Heales, S.J. Intracellular distribution of the fluorescent dye nonyl acridine orange responds to the mitochondrial membrane potential: implications for assays of cardiolipin and mitochondrial mass. J Neurochem 82, 224-233 (2002).

26. Scarpulla, R.C. Transcriptional paradigms in mammalian mitochondrial biogenesis and function. Physiol Rev 88, 611-638 (2008).

27. Gohil, V.M. & Greenberg, M.L. Mitochondrial membrane biogenesis: phospholipids and proteins go hand in hand. J Cell Biol 184, 469-472 (2009).

28. Krogh, A., Larsson, B., von Heijne, G. & Sonnhammer, E.L. Predicting transmembrane protein topology with a hidden Markov model: application to complete genomes. J Mol Biol 305, 567-580 (2001).

29. Thompson, J.D., Higgins, D.G. & Gibson, T.J. CLUSTAL W: improving the sensitivity of progressive multiple sequence alignment through sequence weighting, position-specific gap penalties and weight matrix choice. Nucleic Acids Res 22, 4673-4680 (1994).

30. Ross, J.A., Nagy, Z.S. & Kirken, R.A. The PHB1/2 phosphocomplex is required for mitochondrial homeostasis and survival of human T cells. J Biol Chem 283, 4699-4713 (2008).

31. Joshi, A.S., Zhou, J., Gohil, V.M., Chen, S. & Greenberg, M.L. Cellular functions of cardiolipin in yeast. Biochim Biophys Acta 1793, 212-218 (2008).

32. Osman, C., Voelker, D.R. & Langer, T. Making heads or tails of phospholipids in mitochondria. J Cell Biol 192, 7-16 (2011).

33. Huber, T.B., Schermer, B., Muller, R.U., Hohne, M., Bartram, M., Calixto, A., Hagmann, H., Reinhardt, C., Koos, F., Kunzelmann, K., Shirokova, E., Krautwurst, D., Harteneck, C., Simons, M., Pavenstadt, H., Kerjaschki, D., Thiele, C., Walz, G., Chalfie, M. & Benzing, T. Podocin and MEC-2 bind cholesterol to regulate the activity of associated ion channels. Proc Natl Acad Sci U S A 103, 17079-17086 (2006).

90

34. Artal-Sanz, M. & Tavernarakis, N. Prohibitin and mitochondrial biology. Trends Endocrinol Metab 20, 394-401 (2009).

35. Mielenz, D., Vettermann, C., Hampel, M., Lang, C., Avramidou, A., Karas, M. & Jack, H.M. Lipid rafts associate with intracellular B cell receptors and exhibit a B cell stage-specific protein composition. J Immunol 174, 3508-3517 (2005).

36. Kim, K.B., Lee, J.W., Lee, C.S., Kim, B.W., Choo, H.J., Jung, S.Y., Chi, S.G., Yoon, Y.S., Yoon, G. & Ko, Y.G. Oxidation-reduction respiratory chains and ATP synthase complex are localized in detergent-resistant lipid rafts. Proteomics 6, 2444-2453 (2006).

37. Foster, L.J., De Hoog, C.L. & Mann, M. Unbiased quantitative proteomics of lipid rafts reveals high specificity for signaling factors. Proc Natl Acad Sci U S A 100, 5813-5818 (2003).

38. Merkwirth, C., Dargazanli, S., Tatsuta, T., Geimer, S., Lower, B., Wunderlich, F.T., von Kleist-Retzow, J.C., Waisman, A., Westermann, B. & Langer, T. Prohibitins control cell proliferation and apoptosis by regulating OPA1-dependent cristae morphogenesis in mitochondria. Genes Dev 22, 476-488 (2008).

39. Houtkooper, R.H. & Vaz, F.M. Cardiolipin, the heart of mitochondrial metabolism. Cell Mol Life Sci 65, 2493-2506 (2008).

40. Acin-Perez, R., Fernandez-Silva, P., Peleato, M.L., Perez-Martos, A. & Enriquez, J.A. Respiratory active mitochondrial supercomplexes. Mol Cell 32, 529-539 (2008).

41. Tondera, D., Grandemange, S., Jourdain, A., Karbowski, M., Mattenberger, Y., Herzig, S., Da Cruz, S., Clerc, P., Raschke, I., Merkwirth, C., Ehses, S., Krause, F., Chan, D.C., Alexander, C., Bauer, C., Youle, R., Langer, T. & Martinou, J.C. SLP-2 is required for stress-induced mitochondrial hyperfusion. EMBO J 28, 1589-1600 (2009).

42. Gohil, V.M., Hayes, P., Matsuyama, S., Schagger, H., Schlame, M. & Greenberg, M.L. Cardiolipin biosynthesis and mitochondrial respiratory chain function are interdependent. J Biol Chem 279, 42612-42618 (2004).

43. Biswas, G., Adebanjo, O.A., Freedman, B.D., Anandatheerthavarada, H.K., Vijayasarathy, C., Zaidi, M., Kotlikoff, M. & Avadhani, N.G. Retrograde Ca2+ signaling in C2C12 skeletal myocytes in response to mitochondrial genetic and metabolic stress: a novel mode of inter-organelle crosstalk. EMBO J 18, 522-533 (1999).

91

Chapter 3 2

3 SLP-2 deficiency in T cells is associated with abnormal mitochondrial membrane compartmentalization of cardiolipin, decreased mitochondrial respiration and impaired CD4+ T cell responses

Stomatin-like protein 2 (SLP-2) is a mostly mitochondrial protein that regulates

mitochondrial biogenesis and function and modulates T cell activation. To determine the

mechanism of action of SLP-2, we generated T cell-specific SLP-2-deficient mice. These

mice had normal numbers of thymocytes and T cells in the periphery. However,

conventional SLP-2-deficient T cells had a post-transcriptional defect in IL-2 production

in response to TCR ligation, even under conditions of non-limiting glycolysis, and this

translated into reduced CD4+ T cell responses. SLP-2 deficiency was associated with

impaired cardiolipin compartmentalization in mitochondrial membranes, decreased levels

of the NDUFS3, NDUFB8 and NDUFA9 subunits of complex I of the respiratory chain

and subsequent decreased activity of this complex as well as of complex II + III of this

chain. Based on these results, we propose that SLP-2 organizes mitochondrial membrane

compartmentalization of cardiolipin, which is required for optimal assembly and function

of respiratory chain complexes. This function, in T cells, helps to ensure proper

metabolic response during activation.

3.1 Introduction

Stomatin-like protein 2 (SLP-2) is an abundant protein in the proteome of detergent-

insoluble microdomains of mitochondrial membranes 1. It is a member of the highly

conserved stomatin family, which is composed of stomatin, SLP-1, SLP-2 and SLP-3 2-7,

2 The material contained in this chapter is based on a submitted manuscript: Christie, D., Lian, D., Wang,

H., Hatch, G. and Madrenas, J. SLP-2 deficiency in T cells is associated with abnormal mitochondrial compartmentalization of cardiolipin, decreased mitochondrial respiration and impaired CD4+ T cell responses. 2012

92

and as such, is part of the SPFH superfamily that also includes prohibitins (PHB),

flotillins and the bacterial proteins HflC and HflK 8, 9. The function of these proteins has

been linked to the regulation of assembly and function of complex multi-chain receptors 2, 10-12. In line with this proposed function, we have recently shown that SLP-2 binds

cardiolipin and promotes the formation of specialized cardiolipin-enriched microdomains

that could facilitate the assembly of protein complexes within mitochondrial membranes 12.

SLP-2 can be detected in many tissues including lymph nodes, thymus and tonsils. In

addition, it is expressed at low levels in human peripheral blood mononuclear cells

(PBMC) including T cells, B cells and monocytes and its expression is upregulated upon

activation in vitro 1. Consistent with this distribution, it has been shown that SLP-2 plays

a role in T cell activation 1: upregulation of SLP-2 correlates with enhanced activation

and functional responses, whereas downregulation of SLP-2 expression is associated with

decreased duration of TCR signaling as well as a decrease in T cell activation.

To study the mechanism of action of SLP-2 in vivo, we initially generated a SLP-2

knockout mouse. However, this mouse was embryonic lethal at the pre-implantation

stage (C.D. Lemke and J. Madrenas, unpublished observation). To circumvent this

limitation, we generated a T cell-specific SLP-2 knockout mouse model. Using this

model, we report here that SLP-2-deficient T cells show abnormal cardiolipin

compartmentalization in mitochondrial membranes, decreased levels of the NDUFS3,

NDUFB8 and NDUFA9 subunits of complex I of the electron transport chain and

decreased activity of this complex, as well as decreased activity of complexes II + III of

this chain. This defect is associated with a selective post-transcriptional reduction in IL-2

production and decreased responses to stimulation through the TCR in vivo and ex vivo.

We propose that SLP-2 contributes to T cell activation primarily by regulating the

compartmentalization of cardiolipin in mitochondrial membranes and ensuring proper

assembly and function of the respiratory chain components in these membranes. This, in

turn, ensures optimal metabolic response during T cell activation.

93

3.2 Materials and Methods

3.2.1 Mice

A full length genomic fragment containing the mouse SLP-2 gene was cloned within the

lox sites of the 3LoxP3NwCD vector. Upon sequence confirmation, the plasmid was

electroporated into C57BL/6 embryonic stem cells and clones were selected for

neomycin resistance. The resistant clones were screened by Southern blot and PCR

analysis to confirm homologous recombination of the SLP-2-floxed DNA sequence.

Clones containing the SLP-2-floxed sequence were injected into B6/Try blastocysts and

implanted into a pseudo-pregnant mouse. The offspring was selected by chimeric coat

colour and backcrossed with C57BL/6 mice to produce SLP-2lox/wt mice in the C57BL/6

background. SLP-2 floxed mice were crossed with C57BL/6 mice transgenic for Cre

recombinase under the control of the CD4 promoter, purchased from Taconic (Hudson,

NY) 13, to generate a T cell-specific knockout of SLP-2 strain (SLP-2 T-K/O). SLP-2

floxed mice lacking Cre were used as control mice. Breeding colonies were derived from

the same original SLP-2 floxed breeders and kept in parallel. C3H/HeJ mice were

obtained from Jackson Labs. Mice were maintained in the animal facility at the

University of Western Ontario with approval from the Animal Use Subcommittee in

accordance with the Canadian Council on Animal Care Guidelines.

3.2.2 Confirmation of SLP-2 deletion

T cells were isolated by magnetic separation using the MACS pan T cell isolation kit

(Miltenyi Biotec, Auburn, CA). To confirm deletion at the genetic level, total genomic

DNA was extracted using the Qiagen DNeasy blood and tissue kit (Qiagen, Mississauga,

ON) and full length SLP-2 PCR was performed using the following primers: forward

primer 5’ ACTTCCACCCTTCAGTCCAGGTCG 3’, reverse primer 5’

ACTTGGATTCTGTGAAAGCAGACAC 3’. Samples were separated by DNA

electrophoresis in a 1% agarose gel and visualized by ethidium bromide staining. To

confirm deletion at the protein level, thymocytes and T cells were pelleted in PBS

containing sodium orthovanadate (400 μM) and EDTA (400 μM) and resuspended in

lysis buffer (1% Triton X-100, 150 mM NaCl, 10 mM Tris (pH7.6), 5 mM EDTA, 1 mM

94

sodium orthovanadate, 10 μg/mL leupeptin, 10 μg/mL aprotinin, 25 μM p-nitrophenyl-p’

guanidinobenzoate) at 4ºC for 30 min. These lysis conditions are sufficient to disrupt all

cellular membranes, excluding the nuclear membrane. Lysates were then cleared of

debris by centrifugation (10,700 x g, 4ºC, 10 min), mixed with sample buffer containing

β-mercaptoethanol and boiled. Cell lysates were resolved by standard SDS-PAGE and

western blotting with rabbit polyclonal anti-SLP-2 antisera (Proteintech Group, Chicago,

IL) and mouse monoclonal anti-GAPDH antibody (Chemicon International, Temecula,

CA). Detection of proteins after western blotting was performed using the BM

Chemiluminescence Substrate POD system (Roche, Laval, QC, Canada).

3.2.3 Isolation of detergent-insoluble fraction of mitochondrial membranes

Intact mitochondria were isolated from primed T cells using the QproteomeTM

Mitochondria Isolation Kit (Qiagen). Detergent-insoluble fractions were isolated by

incubation on ice in 25 mM HEPES buffer with 0.1% triton-X, as previously described 14.

Fractions were analyzed by western blotting or thin layer chromatography.

3.2.4 Measurement of cardiolipin

Soluble and insoluble mitochondrial fractions were washed in 2 ml of methanol:water

(1:1, by vol) and 25 µL aliquots were taken for protein determination 15. Subsequently,

0.5 ml of water and 2 ml chloroform were added to initiate phase separation. Samples

were centrifuged at 2000 rpm for 10 min and the upper phase was aspirated. Two ml of

theoretical upper phase (methanol:0.9% NaCl:chloroform) (48:47:3, by vol) was added

and centrifugation was repeated for 5 min. The organic phase was removed and dried

under nitrogen gas and resuspended in chloroform:methanol (2:1, by vol). A 50 µl aliquot

was spotted onto thin layer plates for separation of cardiolipin from other phospholipids

as described 16. Phospholipids were removed from the plate and phospholipid phosphorus

content of cardiolipin was determined as described 17.

95

3.2.5 Enzyme activity assays

NADH dehydrogenase activity was measured from whole cell lysates using the complex I

enzyme activity microplate assay kit (Mitosciences). The activity of Complex II + III

(succinate cytochrome c reductase), measured as reduction of cytochrome c after addition

of succinate, was determined as described previously 18.

3.2.6 Thymocyte and T cell population analysis

Thymocytes, splenocytes and lymph node cells were isolated from control and SLP-2 T-

knockout mice and stained with the following antibodies: PE-conjugated or APC-

conjugated rat anti-CD4 antibody, FITC-conjugated or PerCP-Cy5.5-conjugated rat anti-

CD8 antibody, APC-conjugated Armenian hamster anti-CD3 antibody, PE-conjugated rat

anti-CD25 antibody, FITC-conjugated rat anti-CD44 antibody or with isotype control

antibodies (all from Becton Dickinson, San Jose, CA).

3.2.7 Flow cytometry

Fluorescence was detected by flow cytometry, using a BD FACSCaliburTM flow

cytometer and CellQuest computer software (Becton Dickinson, San Jose, CA) and

analyzed with FlowJo flow cytometry analysis software (Treestar, Inc., Ashland, OR).

3.2.8 Stimulation of T lymphocytes

Splenocytes isolated from control and SLP-2 T-K/O mice were stimulated with

staphylococcal enterotoxin E (SEE) superantigen (Toxin Technologies, Sarasota, FL) or

0.01µg/mL to 1µg/mL of anti-CD3 antibody (Cedarlane, Burlington, ON) and 0.2 µg/mL

anti-CD28 antibody (eBioscience, San Diego, CA) in 96 well round bottom or flat bottom

plates for 24h at 37°C 19. After 24 h stimulation, supernatants were collected for IL-2

and IFNγ measurement by ELISA 20. Cultures were then supplemented with 3H-

thymidine and incubated for 24 h to measure cell proliferation. For glucose experiments,

splenocytes were stimulated with SEE for 24 h in 10% FBS supplemented RPMI media

containing either high (11 mM) or low (4 µM) glucose concentrations.

96

3.2.9 In vivo immunization and ex vivo recall response

Mice were immunized subcutaneously with 100µg ovalbumin in emulsion with Freund’s

complete adjuvant (Difco, Becton Dickinson, San Jose, CA). Eight to ten days later mice

were sacrificed, lymph nodes collected and single cell suspensions prepared for in vitro

stimulation with ovalbumin or pigeon cytochrome c. Culture supernatants were collected

after 48 h for cytokine assays, and cultures supplemented with 3H-thymidine for

proliferation assay 24 h later. Antigen presenting cells were treated with 50µg/mL

mitomycin C for 30 min at 37°C followed by washing, and used at a 1:4 ratio with CD4+

T cells.

3.2.10 Intracellular staining cytokine analysis

Splenocytes were stimulated with 1µg/mL anti-CD3 antibody and 0.2µg/mL anti-CD28

antibody in flat bottom plates for 24 to 36 h at 37°C, in the presence of brefeldin A

(eBioscience, San Diego, CA). Cells were surface stained for CD4-APC and CD8-

PerCP-Cy5.5 followed by intracellular staining for IL-2-PE and IFNγ-FITC using

Invitrogen fix and permeabilization reagents (Life Technologies, Grand Island, NY).

3.2.11 Real time PCR analysis

Splenocytes were stimulated with 1µg/mL anti-CD3 antibody and 0.2µg/mL anti-CD28

antibody in flat bottom plates for 6 h at 37°C. Total RNA was isolated from stimulated

cells or an equal number of un-stimulated cells using the Qiagen RNeasy kit and RNA

was quantified using a NanoDrop (Thermo Scientific, Wilmington, DE). cDNA was

synthesized from 500 ng RNA using the iScript cDNA synthesis kit (BioRad,

Mississauga, Ont) and transcript levels were quantified on the BioRad CFX96 real time

PCR detection system, using iTaq SYBR green supermix with ROX (BioRad). IL-2

primer sequences: Fwd CCTGAGCAGGATGGAGAATTACA, Rev

TCCAGAACATGCCGCAGAG 21, mouse GAPDH real time primers were purchased

from SABiosciences (Qiagen). Relative levels of IL-2 transcript were determined by

comparing the threshold cycle (CT) detection values of IL-2 in WT and SLP-2 T-K/O

cells in stimulated and un-stimulated cells and were normalized to GAPDH levels by the

ΔΔCT method 22.

97

3.2.12 Heart transplants

Abdominal heterotopic transplants were performed as described previously 23. After

transplant, graft viability was assessed daily by abdominal palpation of the transplant

heartbeat. Graft rejection was determined by lack of heartbeat.

3.2.13 Activation-induced cell death assay

Splenocytes were stimulated with 250 ng/mL ionomycin and 1ng/mL PMA (Sigma, St

Louis, MO) for 12, 24, 36, 48 and 60 h in complete RPMI media. At each time point

cells were collected and stained with FITC-conjugated annexin V and propidium iodide

(Clontech Inc, Palo Alto, CA). Fluorescence was detected by flow cytometry.

3.2.14 Statistical analysis

Data analysis was performed with GraphPad Prism software (GraphPad Software, La

Jolla, CA). Data are presented at mean ± SEM. Two tailed Student’s t test or ANOVA

with Bonferroni post-hoc testing were used to determine p values. P values lower than

0.05 were considered significant.

3.3 Results

3.3.1 Generation of T cell-specific SLP-2 knockout mice

To study the effect of SLP-2 in vivo, we first tried to generate conventional SLP-2

knockout mice but no viable embryos were obtained after more than 300 term

pregnancies (C. D. Lemke and J Madrenas, unpublished observations), suggesting that

SLP-2 deficiency is embryonic lethal at the pre-implantation stage. To circumvent this

problem, conditional SLP-2 knockout mice were generated in which the whole SLP-2

gene was replaced by a lox flanked genomic SLP-2 gene. The resulting SLP-2lox mouse

was bred together with a transgenic mouse containing the Cre-recombinase gene under

the control of the CD4 promoter 13 to generate mice in which T cells lacked SLP-2

expression. These resulting knockout mice are called SLP-2 T-K/O throughout the paper.

98

SLP-2 knockout in the T cell lineage was confirmed at the genomic level by PCR

analysis and at the protein level by western blot analysis. Using genomic PCR with

primers flanking the entire SLP-2 sequence to give a 4kb fragment for full length SLP-2

and a 500bp fragment for SLP-2 deletion, we detected the full length SLP-2 fragment in

CD4+ thymocytes, naïve and primed T cells and splenic non-T cells from control mice.

In contrast, we failed to detect the full length SLP-2 in SLP-2 T-K/O CD4+ thymocytes

and naïve and blast T cells. In these mice, only the truncated product, indicative of

genomic deletion of SLP-2, was observed (Fig. 3.1a). In the non-T cell population from

spleen, we still detected the truncated SLP-2 fragment in addition to the full length SLP-2

fragment, likely due to contamination of the prep by T cells (up to 2% of total cell

numbers in the prep).

The deletion of genomic SLP-2 translated into significantly lower SLP-2 protein

expression in SLP-2 T-K/O CD4+ thymocytes, and complete loss of SLP-2 expression in

peripheral T cells but not in spleen CD4-CD8- cells (Fig. 3.1b), confirming the restriction

of SLP-2 deletion to conventional T cells. Moreover, and as reported with primary

human T cells 1, these experiments showed the upregulation of SLP-2 protein levels upon

activation of wild type T cells (see blast vs. naïve, in Fig. 3.1b).

3.3.2 SLP-2 deficiency is associated with abnormal cardiolipin compartmentalization in mitochondrial membranes and reduced mitochondrial respiration

Proteins of the SPFH superfamily have been linked to the organization of membrane

microdomains 24-29. Based on this assumption, and since SLP-2 is mostly a mitochondrial

protein localized in the intermembrane space 30, 31, we examined if SLP-2 deficiency

would alter mitochondrial membrane compartmentalization. We first looked at the effect

of SLP-2 deficiency on PHBs since these proteins interact with each other and contribute

to the formation of detergent-insoluble microdomains in human mitochondria 14, 32.

Triton-X insoluble domains from T cell mitochondria from control and SLP-2 T-K/O

mice were prepared and blotted for PHB1 and SLP-2. Although total cellular levels of

99

Figure 3.1: Generation of T cell-specific SLP-2 knockout mice.

(a) Genomic DNA was extracted from control and SLP-2 T-K/O CD4+ thymocytes, naïve

and blast T cells and non T cells and genomic SLP-2 deletion was analyzed by PCR using

primers flanking the entire SLP-2 gene. (b) Whole cell lysates from CD4+ thymocytes,

naïve and blast T cells and non T cells were sequentially immunoblotted for SLP-2 and

for GAPDH (as a loading control). CD4+ thymocytes were isolated from single cell

suspension of control (WT) and SLP-2 T-K/O thymocytes; splenocyte single cell

suspensions were separated into T cell and non T cell fractions. Untreated T cells are

referred to as naïve T cells while T cells stimulated for 72 hours with ionomycin and

PMA are referred to as blast T cells. Data are representative of 3 independent

experiments using multiple mice with material from a representative mouse per group ran

per lane.

100

Figure 3.1

101

PHB1 were not affected in SLP-2-deficient T cells (see below), the level of PHB1 in the

mitochondrial detergent-insoluble microdomain fraction was reduced, although the levels

in the detergent-soluble fraction were similar between control and SLP-2 T-K/O cells

(Fig. 3.2a and e). As the amount of PHB1 in detergent-soluble fractions of control and

SLP-2 T-K/O cells was similar, the loss of PHB1 from the insoluble fraction suggested

an abnormal compartmentalization in the SLP-2 T-K/O mitochondrial membrane.

We have recently shown that SLP-2 not only interacts with PHBs but also binds

cardiolipin, the signature phospholipid linked to detergent-insoluble microdomains of

mitochondrial membranes 12. Therefore, the defect in mitochondrial membrane

compartmentalization of PHB1 associated with SLP-2 deficiency prompted us to test if

the content or distribution of cardiolipin was also impaired. Total cardiolipin content in

T cells from control and SLP-2 T-K/O mice as detected with nonyl-acridine orange

staining was not significantly different (Fig. 3.2b). However, the cardiolipin content in

mitochondrial detergent-insoluble microdomains isolated from primed SLP-2 T-K/O T

cells was significantly (p<0.05) lower than that of control samples (Fig. 3.2c). Altogether,

these results imply that SLP-2 deficiency in T cells is associated with impaired

mitochondrial membrane compartmentalization of cardiolipin.

Proper compartmentalization of mitochondrial membranes into cardiolipin-enriched

microdomains is required for optimal assembly and function of the components of the

respiratory chain 33, 34. Therefore, we examined the effects of SLP-2 deficiency in

assembly and function of the complexes of the respiratory chain. First, we analyzed the

levels of mitochondrial respiratory chain components in SLP-2-deleted T cells using a

cocktail of antibodies against different subunits of complexes I to V of the respiratory

chain. SLP-2 T-K/O cells had lower levels of the NDUFB8 subunit of complex I and

slight decrease of the 30 kDa subunit of complex II compared to control cells (Fig. 3.2d).

We corroborated this finding with two other subunits of the 45 described subunits of

complex I, including the NDUFS3 and NDUFA9 subunits (Fig. 3.2e). Note that the total

cellular levels of PHB1 were not affected by SLP-2 deficiency. SLP-2 deficiency

102

Figure 3.2: T cells deficient in SLP-2 have abnormal cardiolipin

compartmentalization and decreased activity of respiratory complexes I and II+III.

(a) Mitochondrial detergent-soluble and insoluble fractions from WT and T-K/O cells

were separated by SDS-PAGE and analyzed by serial blotting for SLP-2 and PHB1. Blot

is representative of 3 independent experiments. (b) Control and SLP-2 T-K/O T cells

were stained with 10 nM nonyl-acridine orange to assess total cellular cardiolipin content

by flow cytometry. The plot represents one control (heavy line) and one knockout mouse

(fine line), representing 3 independent experiments with 3 mice per group. (c)

Cardiolipin was measured in mitochondrial detergent-insoluble fractions collected from

control (black bars) and SLP-2 T-K/O (white bars) T cells. (d) Whole cell lysates from

control (WT) and SLP-2 T-K/O T cells were sequentially immunoblotted for components

of each complex of the respiratory chain and SLP-2, or (e) blotted for individual subunits

of complex I, PHB1 and actin (as a loading control). Blots are representative of 3

independent experiments. (f) Complex I activity was measured in control (black bars)

and SLP-2 T-K/O T cells (white bars). (g) Complex II+III activity was measured in

control (black bars) and SLP-2 T-K/O T cells (white bars). *: p<0.05, **: p<0.01.

103

Figure 3.2

104

did not grossly affect the levels of complexes III to V, including ATP synthase, which

were similar in control and SLP-2 T K/O T cells.

The observed decrease in levels of complex I subunits translated into a significant

(p<0.01) decrease in the functional activity of complex I NADH dehydrogenase in SLP-2

T-K/O T cells (Fig. 3.2f). In addition, an assay of the combined activity of succinate

dehydrogenase and cytochrome c oxidoreductase (complex II + III activity) also showed

a significant decrease in SLP-2-deficient T cells (p<0.05 Fig. 3.2g).

3.3.3 Impaired ex vivo responses of SLP-2-deficient T cells

Next we examined the effect of SLP-2 deletion and the altered mitochondrial membrane

compartmentalization of cardiolipin on T cell function. First, we looked at the cellularity

of thymus, spleen and lymph nodes. Organ sizes were similar between control and SLP-2

T-K/O mice. The total thymocyte and splenocyte cell counts (Fig. 3.3a) as well as the

number of double negative, double positive, CD4+ single positive and CD8+ single

positive thymocytes and splenic cells (Fig. 3.3b and c) were similar in unmanipulated

SLP-2 T-K/O and control mice, implying a grossly normal T cell development in these

mice. Although the total cell count in lymph nodes was similar in control and SLP-2 T-

K/O mice (Fig. 3.3a), there was a slight but consistent decrease in CD4+ T cells in the

lymph nodes of SLP-2 T-K/O mice (p = 0.09, Fig. 3.3d).

Next, we tested the functional responses of SLP-2-deficient T cells. Naïve splenocytes

from control and SLP-2 T-K/O mice were stimulated ex vivo with increasing

concentrations of staphylococcal enterotoxin (SEE) superantigen. We found that SLP-2

T-K/O mice produced significantly less IL-2 upon superantigen stimulation compared to

control mice (p<0.01) (Fig. 3.4a). The decrease in IL-2 production, however, did not

translate into significantly less T cell proliferation (Fig. 3.4b), and SLP-2 T-K/O

splenocytes produced similar levels of IFNγ and IL-17 in response to superantigen

stimulation (data not shown).

105

Figure 3.3: Grossly normal thymic T cell development and peripheral T cell pool in

SLP-2 T-K/O mice.

(a) Eight-week-old wild type (black bars) and SLP-2 T-K/O (white bars) mice were

sacrificed and single cell suspensions were made from whole thymus, spleens and lymph

nodes and live cells were counted. To analyze CD4+ and CD8+ T cell populations, single

cell suspension of (b) thymocytes, (c) splenocytes and (d) lymph nodes were stained with

anti-CD4-PE and anti-CD8-FITC, or isotype-matched control antibodies and surface

expression detected by FACS. T cell populations were calculated by multiplying the total

thymocyte count or total splenocyte count by the population percentage obtained by

FACS. Splenocytes and lymph nodes were also stained with anti-CD3-APC antibodies

and total CD3+ T cell populations were calculated as described above. These plots

represent an average of 6 individual mice for each genotype, performed in 4 separate

experiments. DN: double negative thymocytes; DP: double positive thymocytes; SP:

single positive.

106

Figure 3.3

107

Figure 3.4: T cells deficient in SLP-2 produce less IL-2 in response to T cell activation.

Naive or ex vivo primed splenocytes from WT (black bars) or SLP-2 T-K/O (white bars)

mice were analyzed for IL-2 production and proliferation in response to superantigen

stimulation. Naïve splenocytes were incubated with increasing concentrations of SEE

superantigen at 37°C. After 24 hours, supernatants were collected and IL-2 secretion was

measured by ELISA (a). Cultures were supplemented with additional media and 3H-

thymidine and incubated for an additional 24 hours after which time the cells were

harvested and 3H-thymidine incorporation was measured (b). In C and D, splenocytes

from WT and SLP-2 T-K/O mice were stimulated with 250 ng/mL ionomycin and 1

ng/mL PMA for 72 hours and rested for 48 hours. The primed cells were then analyzed

for IL-2 secretion (c) and proliferation (d) in response to superantigen stimulation in the

same manner as described for naïve cells. Plots in this figure represent an average of 4

mice per group and are representative of at least 3 independent experiments, *: p<0.05,

**: p<0.01, ***: p<0.001.

108

Figure 3.4

109

We have previously reported that SLP-2 levels increase upon priming 1 implying that the

role of this protein is greater upon T cell priming. To test the secondary response of SLP-

2-deficient T cells, control and SLP-2 T-K/O splenocytes were stimulated in vitro with

PMA and ionomycin for 72 hours followed by 48 hours resting, before being stimulated

with SEE superantigen. Again, primed SLP-2 T-K/O cells produced significantly less IL-

2 compared to control cells (p<0.001, Fig. 3.4c). We observed a trend towards less

proliferation in SLP-2 T-K/O cells but it did not reach statistical significance (Fig. 3.4d).

The significant decrease in IL-2 responses seen in SLP-2 T-K/O T cells was also

observed when naïve and in vitro primed cells were stimulated with anti-CD3 and anti-

CD28 antibodies (data not shown). Together, these results demonstrate that SLP-2-

deficient T cells have decreased activation leading to decreased IL-2 responses.

3.3.4 SLP-2 deficiency impairs T cell responses in vivo

To assess how the impairment in T cell responses observed ex vivo translated into T cell

responses in vivo, we first assessed the capacity of SLP-2-deficient T cells to respond to

immunization. Mice were immunized with ovalbumin (OVA) and ex vivo recall

responses tested 10 days after immunization. The response to pigeon cytochrome c

(PCC) was used as a control. In these experiments, SLP-2-deficient cells had

significantly decreased responses (p<0.05), measured by both IL-2 production and

proliferation (Fig. 3.5a and b).

As an additional model for in vivo T cell activation, we examined the response to

vascularized allografts in SLP-2 T-K/O mice compared to that in SLP-2 competent mice.

Heterotopic heart transplants from C3H mice (H-2k) into C57BL/6 (H-2b) wild type and

SLP-2 T-K/O mice were performed and graft survival monitored. In these experiments,

we observed a slight but consistent and significant (p<0.05) delay in the rejection of

allografts in SLP-2 T-K/O mice (Fig. 3.5c). Together, these results corroborated that

110

Figure 3.5: T cells deficient in SLP-2 have decreased responses in vivo.

Mice were immunized subcutaneously with 100 µg ovalbumin in emulsion with Freund’s

complete adjuvant. Ten days post-immunization lymph nodes were isolated and single

cell suspension from WT (black bars) or SLP-2 T-K/O (white bars) mice were stimulated

in vitro with various concentrations of ovalbumin as well as the control peptide pigeon

cytochrome c (PCC). After 48 hours, supernatants were collected and IL-2 levels were

measured by ELISA (a). Cultures were supplemented with additional media and 3H-

thymidine and incubated for an additional 24 hours, after which time the cells were

harvested and the 3H-thymidine incorporation was measured (b). These plots are

representative of 4 mice for each group and are representative of 3 independent

experiments. (c) Wild type and SLP-2 knockout mice were grafted with hearts isolated

from C3H mice. Rejection of the transplant tissue was measured by the beating of the

transplant heart, with the endpoint of rejection being the loss of heartbeat. These plots

report graft survival for 6 mice per group, *: p<0.05, **: p<0.01, ***: p<0.001.

111

Figure 3.5

112

SLP-2 deficiency in T cells is associated with impaired responses to TCR-dependent

stimulation in vivo.

The lymph nodes from control and SLP-2 T-K/O mice had comparable numbers of

CD19+ B cells and CD3+ T cells both before and after immunization (data not shown).

Although there was no difference in the total number of T cells, SLP-2 T-K/O mice had a

consistent and significant decrease in the percentage of CD4+ T cells but not in the

percentage of CD8+ T cells after immunization (Table 3.1). The decrease in the

percentage of CD4+ T cells was primarily due to decreased proportion of activated T

cells, as demonstrated by the decrease in CD4+CD25+ and CD4+CD44hi T cell

populations. The percentage and function of regulatory CD4+CD25+FoxP3+ T cells were

comparable between control and SLP-2 T-K/O mice (Table 1 and data not shown).

Furthermore, the decrease in activated CD4+ T cells was not due to an increased

susceptibility to activation-induced cell death (data not shown).

3.3.5 The IL-2 defect of SLP-2 deficient T cells occurs at the post-transcriptional level

To further investigate the low IL-2 responses in SLP-2 T-K/O mice, we examined IL-2

production at the single cell level by performing intracellular staining and analyzing cell

populations by flow cytometry. Naïve splenocytes were isolated from WT and SLP-2 T-

K/O mice and stimulated for 24 hours in the presence of anti-CD3 and anti-CD28

antibodies. In agreement with the IL-2 data presented above, significantly fewer CD4+

and CD8+ SLP-2 T-K/O cells produced IL-2 in response to stimulation, compared to WT

T cells (p<0.001 and p<0.01 respectively, Fig. 3.6a and b). In addition, the geometric

mean of the IL-2 fluorescence staining was significantly lower in the SLP-2 T-K/O

CD4+IL-2+ and CD8+IL-2+ cell population compared to WT populations (p<0.001 and

p<0.01 respectively, Fig. 3.6c and d). Together, these data indicate that fewer SLP-2-

deficient T cells became activated to express IL-2, and those T cells that were activated

produced less IL-2 compared to WT T cells. Surprisingly, when we investigated the

transcript levels of IL-2 in stimulated T cells, we found no significant difference in IL-2

113

Table 3.1

Table 3.1: T cell subsets in lymph nodes of SLP-2 T-K/O mice upon OVA

immunization

WT T-K/O

CD4+ 29.0% ± 0.9% 23.0% ± 0.9% 1

CD8+ 22.5% ± 0.7% 24.1% ± 1.0%

CD4+CD25+ 3.0% ± 0.2% 2.5% ± 0.1% 2

CD8+CD25+ 0.08% ± 0.01% 0.09% ± 0.01%

CD4+CD44lo 5.9% ± 0.4% 4.5% ± 0.7% 2

CD8+CD44lo 10.9% ± 0.6% 12.1% ± 0.6%

CD4+CD44hi 16.3% ± 0.4% 13.4% ± 0.9% 2

CD8+CD44hi 7.4% ± 0.3% 7.4% ± 0.3%

CD4+CD25+FoxP3+ 8.3% ± 0.2% 8.2% ± 0.3%

1 p < 0.001 2 p < 0.01

Control and SLP-2 T-K/O mice were immunized subcutaneously with 100 µg OVA in

emulsion with Freund’s complete adjuvant. Nine days post-immunization lymph nodes

were isolated and single cell suspensions were stained for CD4, CD8, CD25, CD44 and

FoxP3 and cells were analyzed by FACS. Values are reported as a percentage of total

lymph node population and represent an average of 8 mice per group ± SEM. These

results are representative of 3 independent experiments.

114

Figure 3.6: SLP-2-deficiency in T cells induces a post-transcriptional defect in IL-2 production.

(a-d) Naive splenocytes from WT (black bars) or SLP-2 T-K/O (white bars) mice were

analyzed for intracellular IL-2 staining in response to anti-CD3 and anti-CD28

stimulation using flow cytometry for CD4, CD8 and IL-2. Plots show the percentage of

CD4 or CD8 cells expressing IL-2 (a and b respectively) or the geometric mean of the

IL-2-PE stain in CD4+IL-2+ or CD8+IL-2+ populations (c and d respectively). (e) Real-

time PCR analysis of IL-2 mRNA in WT (black bars) and SLP-2 T-K/O (white bars)

cells from same samples as in a to d. (f-i) Naive splenocytes from WT (black bars) or

SLP-2 T-K/O (white bars) mice were analyzed for intracellular IFNγ staining in response

to anti-CD3 and anti-CD28 stimulation using flow cytometry for CD4, CD8 and IFNγ.

Plots show the percentage of CD4+ or CD8+ cells expressing IFNγ (f and g respectively)

or the geometric mean of the IFNγ-FITC stain in CD4+IFNγ+ or CD8+IFNγ+ populations

(h and i respectively). All plots represent an average of 4 mice and are representative of 3

independent experiments. *:p<0.05, **: p<0.01, ***: p<0.001.

115

Figure 3.6

116

transcript levels between SLP-2 T-K/O and WT cells (Fig. 3.6e), which may point to a

possible role for SLP-2 in the processing of IL-2 mRNA.

Interestingly, the defect in cytokine production seen in SLP-2-deficient T cells seemed to

be limited to IL-2. In addition to normal IFNγ and IL-17 secretion from stimulated SLP-

2 T-K/O cells, we also found that a similar proportion of WT and SLP-2 T-K/O cells

become IFNγ expressing cells in response to T cell stimulation (Fig. 3.6f and g).

Furthermore, quantification of fluorescent signal from IFNγ+ populations indicate a

similar expression level in both WT and SLP-2 T-K/O cells (Fig. 3.6h and i). These

findings support the idea of a unique role for SLP-2 in the regulation of IL-2 expression.

3.3.6 Altered metabolic capacity of SLP-2-deficient T cells

The metabolic requirements of conventional T cell activation are mostly dependent on

glycolysis if glucose is not limiting 35, whereas the contribution of mitochondrial

oxidative phosphorylation is relatively secondary. Given the involvement of SLP-2 in

mitochondrial function, we next tested the effect of limiting glucose supply on the

impairment of T cell activation due to SLP-2 deficiency. Compared to WT T cells, SLP-

2 T-K/O cells showed significantly less IL-2 production in response to stimulation with

SEE superantigen under both high glucose (11 mM) and low glucose (4 µM) conditions

(Fig. 3.7). Under non-limiting glucose, significantly less IL-2 was produced by SLP-2 T-

K/O cells in response to 10 ng/mL SEE stimulation (p<0.001) compared to WT cells,

whereas limiting glucose (to low levels in media and fetal bovine serum) led to a broader

reduction of IL-2 production, i.e. seen in response to 1ng/mL SEE (p<0.05) and to 10

ng/mL SEE (p<0.001). However, limiting glucose supply did not decrease further the

already lower IL-2 response of SLP-2 T-K/O cells, implying an altered metabolic

capacity of SLP-2-deficient T cells that is independent of and insensitive to glycolysis.

117

Figure 3.7: Altered metabolic capacity of SLP-2-deficient T cells during activation.

Splenocytes from WT (black bars) and SLP-2 T-K/O (white bars) mice were analyzed for

IL-2 production in response to indicated concentrations of SEE superantigen stimulation

under conditions of high glucose (11 mM) or low glucose (4 µM), both in the presence of

10% fetal bovine serum in the culture medium. Results represent mean and standard

deviation of triplicate samples. **: p<0.01, ***: p<0.001.

118

Figure 3.7

119

3.4 Discussion

We report here the characterization of SLP-2 deficiency in T cells in vivo. To do so, and

given that systemic SLP-2-deficiency is embryonically lethal, we had to generate a

conditional knockout mouse strain utilizing the Cre-Lox system with Cre recombinase

under the control of the CD4 promoter. This led to the deletion of the SLP-2 gene during

the double positive stage of thymocyte development. The SLP-2 T-K/O mice were viable

and bred well, giving sufficient numbers for biochemical and functional studies of the

effects of SLP-2-deficiency in T lymphocytes.

We found that SLP-2 deficiency in T cells is associated with abnormal cardiolipin

compartmentalization in mitochondrial membranes. SLP-2 is tightly associated with the

mitochondrial inner membrane 30, 31, and other members of the SPFH family have been

linked to the organization of various membrane microdomains 24-29. Therefore, it is

plausible to suggest that SLP-2 promotes the compartmentalization of the mitochondrial

inner membrane into cardiolipin-enriched microdomains. This proposed role is

consistent with the effects of SLP-2 deficiency on mitochondrial function. Formation of

cardiolipin microdomains is required for optimal assembly of the electron transport

complexes in the mitochondrial inner membrane, likely into supercomplexes 36, to allow

for rapid and efficient electron transport through the respiratory chain. Thus, in the

absence of SLP-2, mitochondrial inner membrane compartmentalization is altered and

this interferes with the optimal assembly of the respiratory chain, ultimately resulting in

defective respiration.

How does SLP-2 contribute to the formation of cardiolipin-enriched microdomains? We

have recently shown that SLP-2 binds cardiolipin and interacts with the mitochondrial

resident proteins PHB 1 and 2 12, 31. Thus, SLP-2 may couple PHBs with cardiolipin-

enriched microdomains. This model supports the proposed involvement of PHBs in the

compartmentalization of the mitochondrial inner membrane 37. Also, by facilitating

mitochondrial membrane organization, SLP-2 enhances mitochondrial function

increasing respiration and resistance to apoptosis as we have recently shown under

120

conditions of SLP-2 upregulation in vitro 12. In addition, this model helps to explain the

observations that cardiolipin is required for proper function of the respiratory chain

complexes 38 and that cardiolipin-enriched microdomains play a role in the induction of

apoptosis 14, 32, 39.

Deficiency in SLP-2 is associated with a decrease in mitochondrial respiration as

measured by the activities of complexes I and II+III. The physiological relevance of a

reduction of complex I activity is likely greater than what is measured since the assay

commonly used to determine the activity of complex I only detects partial reaction of

complex I 40-42. Defective complex I activity is considered the most common defect of

the respiratory chain in humans 43 and causes a diverse set of diseases including Leigh

syndrome and renal tubular acidosis. Complex I represents the major entry point into the

respiratory chain through the oxidation of NADH to NAD+ with the transfer of two

electrons into the respiratory chain 44. We show that, in the absence of SLP-2, there is

inefficient assembly of complex I subunits and confirm the suggestion of a previous in

vitro study that downregulation of SLP-2 levels may cause a reduction in the level of

components of the respiratory chain 31. Cardiolipin-enriched microdomains may be

especially required for optimal assembly of complex I because of the large, multichain

nature of this complex, which includes at least 45 subunits 44. Such an inefficient

assembly of this multichain complex may result in increased degradation of the subunits

by mitochondrial proteases, explaining the decrease in complex I activity and protein

levels in SLP-2 T-K/O T cells 31, 45. The defect in mitochondrial membrane

compartmentalization of cardiolipin in the absence of SLP-2 not only affects the function

of complex I but also of complex II+III and likely of other protein supercomplexes in the

mitochondrial membrane as suggested by the data presented here and by preliminary

proteome characterization of detergent-insoluble microdomains (S. D. Dunn and J.

Madrenas, unpublished observations).

In T cells, the impairment in mitochondrial membrane compartmentalization and

complexes I and II+III activities associated with SLP-2 deficiency results in a minimal

defect under resting conditions. Such a paucity of immune manifestations is also

observed in humans with complex I deficiency. Although these patients have a reduction

121

of complex I activity in peripheral blood leukocytes, they do not show overt immune

deficiency 46, 47. This may be due to a compensatory reliance of T cells on glycolysis for

ATP production inasmuch as access to glucose is not limited 35.

However, the phenotype of SLP-2 deficiency in T cells becomes apparent upon TCR-

mediated activation and results in lower IL-2 production and decreased T cell responses.

It is likely that this reflects the increased metabolic demands on T cells during activation.

The impairment in electron uptake by complex I and decreased complex II+III activity

will result in less protons shuttled across the inner membrane, leading to lower ATP

production and decreased mitochondrial function. The activation-dependent phenotype is

consistent with the recent observation that complex I plays an important role in the

activation of T cells 48, 49. Also, the decrease in CD4+ T cell expansion is expected

because low NAD+ levels, seen under conditions of decreased complex I activity, are

associated with mild lymphopenia 46, 50-52.

A surprising finding in SLP-2-deficient T cells is the selective post-transcriptional defect

in IL-2 production in response to TCR stimulation, documented by the decrease in both

intracellular and secreted levels of IL-2 protein with abundant IL-2 transcripts. A similar

phenotype has been reported in anergic T cells in which IL-2 transcript levels were

similar in T cells stimulated under both activating and anergic conditions, but IL-2

protein was only detected in activated T cells 53, 54. This post-transcriptional block in

cytokine production affected other cytokines, such as IFNγ, IL-4 and IL-17 and was

dependent on AU-rich regions in the 3’ UTR of cytokine mRNA 54, 55. In contrast to

anergic conditions, the post-transcriptional block in SLP-2-deficient T cells appears to be

limited to IL-2 expression as IFNγ and IL-17 were expressed normally. The precise

nature of the SLP-2-dependent metabolic checkpoint for the processing of IL-2 mRNA is

currently unknown.

Why the metabolic demands upon activation would be higher in CD4+ T cells than in

CD8+ T cells as implied by the selective effect of SLP-2 deficiency on CD4+ T cell

expansion remains to be determined. Recent work has started to unravel the role of

metabolic pathways in the differentiation of various T cell subsets 49, 56, 57. Memory CD8

122

T cells seem to rely on fatty acid oxidation, wherein mice with a block in fatty acid

metabolism showed significantly decreased memory CD8 T cells, and upregulation of

fatty acid oxidation with metformin, an AMPK activator, increased the generation of

CD8+ T cells 57. Furthermore, rapamycin treatment increased CD8 memory generation,

likely through the upregulation of fatty acid oxidation 56, 57. Treg cells also require high

levels of fatty acid oxidation 49. Indeed, precursor cells can be driven into the Treg

lineage by the addition of excess fatty acid, metformin or rapamycin. In contrast, effector

CD4+ T cells (TH1, TH2 and TH17 cells) require high levels of glycolysis 35. Thus, the

difference in the numbers CD4+ and CD8+ T cells in SLP-2 T-K/O mice may be a

reflection of differences in the metabolic requirements of these cells such as CD4+ T cells

are more sensitive to mitochondrial defects than CD8+ T cells.

In summary, we show that the loss of SLP-2 in T cells alters cardiolipin

compartmentalization in mitochondrial membranes and prevents proper function of

complexes I and II+III of the respiratory chain. Such a decreased respiratory activity

provides a straightforward explanation for the impaired activation and defective

downstream T cell responses in SLP-2-deficient mice. Availability of the SLP-2 T-K/O

mouse strain may thus be helpful to dissect different aspects of metabolic regulation of T

cell responses.

123

3.5 References

1. Kirchhof, M.G., Chau, L.A., Lemke, C.D., Vardhana, S., Darlington, P.J., Marquez, M.E., Taylor, R., Rizkalla, K., Blanca, I., Dustin, M.L. & Madrenas, J. Modulation of T cell activation by stomatin-like protein 2. J Immunol 181, 1927-1936 (2008).

2. Stewart, G.W., Hepworth-Jones, B.E., Keen, J.N., Dash, B.C., Argent, A.C. & Casimir, C.M. Isolation of cDNA coding for an ubiquitous membrane protein deficient in high Na+, low K+ stomatocytic erythrocytes. Blood 79, 1593-1601 (1992).

3. Seidel, G. & Prohaska, R. Molecular cloning of hSLP-1, a novel human brain-specific member of the band 7/MEC-2 family similar to Caenorhabditis elegans UNC-24. Gene 225, 23-29 (1998).

4. Wang, Y. & Morrow, J.S. Identification and characterization of human SLP-2, a novel homologue of stomatin (band 7.2b) present in erythrocytes and other tissues. J Biol Chem 275, 8062-8071 (2000).

5. Green, J.B., Fricke, B., Chetty, M.C., von During, M., Preston, G.F. & Stewart, G.W. Eukaryotic and prokaryotic stomatins: the proteolytic link. Blood Cells Mol Dis 32, 411-422 (2004).

6. Green, J.B. & Young, J.P. Slipins: ancient origin, duplication and diversification of the stomatin protein family. BMC Evol Biol 8, 44 (2008).

7. Goldstein, B.J., Kulaga, H.M. & Reed, R.R. Cloning and characterization of SLP3: a novel member of the stomatin family expressed by olfactory receptor neurons. J Assoc Res Otolaryngol 4, 74-82 (2003).

8. Browman, D.T., Hoegg, M.B. & Robbins, S.M. The SPFH domain-containing proteins: more than lipid raft markers. Trends Cell Biol 17, 394-402 (2007).

9. Tavernarakis, N., Driscoll, M. & Kyrpides, N.C. The SPFH domain: implicated in regulating targeted protein turnover in stomatins and other membrane-associated proteins. Trends Biochem Sci 24, 425-427 (1999).

10. Huang, M., Gu, G., Ferguson, E.L. & Chalfie, M. A stomatin-like protein necessary for mechanosensation in C. elegans. Nature 378, 292-295 (1995).

11. Wetzel, C., Hu, J., Riethmacher, D., Benckendorff, A., Harder, L., Eilers, A., Moshourab, R., Kozlenkov, A., Labuz, D., Caspani, O., Erdmann, B., Machelska, H., Heppenstall, P.A. & Lewin, G.R. A stomatin-domain protein essential for touch sensation in the mouse. Nature 445, 206-209 (2007).

124

12. Christie, D.A., Lemke, C.D., Elias, I.M., Chau, L.A., Kirchhof, M.G., Li, B., Ball, E.H., Dunn, S.D., Hatch, G.M. & Madrenas, J. Stomatin-like protein 2 binds cardiolipin and regulates mitochondrial biogenesis and function. Mol Cell Biol 31, 3845-3856 (2011).

13. Lee, P.P., Fitzpatrick, D.R., Beard, C., Jessup, H.K., Lehar, S., Makar, K.W., Perez-Melgosa, M., Sweetser, M.T., Schlissel, M.S., Nguyen, S., Cherry, S.R., Tsai, J.H., Tucker, S.M., Weaver, W.M., Kelso, A., Jaenisch, R. & Wilson, C.B. A critical role for Dnmt1 and DNA methylation in T cell development, function, and survival. Immunity 15, 763-774 (2001).

14. Ciarlo, L., Manganelli, V., Garofalo, T., Matarrese, P., Tinari, A., Misasi, R., Malorni, W. & Sorice, M. Association of fission proteins with mitochondrial raft-like domains. Cell Death Differ 17, 1047-1058 (2010).

15. Lowry, O.H., Rosebrough, N.J., Farr, A.L. & Randall, R.J. Protein measurement with the Folin phenol reagent. J Biol Chem 193, 265-275 (1951).

16. Hatch, G.M. Regulation of cardiolipin biosynthesis in the heart. Mol Cell Biochem 159, 139-148 (1996).

17. Rouser, G., Fkeischer, S. & Yamamoto, A. Two dimensional thin layer chromatographic separation of polar lipids and determination of phospholipids by phosphorus analysis of spots. Lipids 5, 494-496 (1970).

18. Sangle, G.V., Chowdhury, S.K., Xie, X., Stelmack, G.L., Halayko, A.J. & Shen, G.X. Impairment of mitochondrial respiratory chain activity in aortic endothelial cells induced by glycated low-density lipoprotein. Free Radic Biol Med 48, 781-790 (2010).

19. Chau, T.A., McCully, M.L., Brintnell, W., An, G., Kasper, K.J., Vines, E.D., Kubes, P., Haeryfar, S.M., McCormick, J.K., Cairns, E., Heinrichs, D.E. & Madrenas, J. Toll-like receptor 2 ligands on the staphylococcal cell wall downregulate superantigen-induced T cell activation and prevent toxic shock syndrome. Nat Med 15, 641-648 (2009).

20. Madrenas, J., Wange, R.L., Wang, J.L., Isakov, N., Samelson, L.E. & Germain, R.N. Zeta phosphorylation without ZAP-70 activation induced by TCR antagonists or partial agonists. Science 267, 515-518 (1995).

21. Fairhead, T., Lian, D., McCully, M.L., Garcia, B., Zhong, R. & Madrenas, J. RIP2 is required for NOD signaling but not for Th1 cell differentiation and cellular allograft rejection. Am J Transplant 8, 1143-1150 (2008).

22. Livak, K.J. & Schmittgen, T.D. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods 25, 402-408 (2001).

125

23. Zhang, Z., Zhu, L., Quan, D., Garcia, B., Ozcay, N., Duff, J., Stiller, C., Lazarovits, A., Grant, D. & Zhong, R. Pattern of liver, kidney, heart, and intestine allograft rejection in different mouse strain combinations. Transplantation 62, 1267-1272 (1996).

24. Snyers, L., Umlauf, E. & Prohaska, R. Association of stomatin with lipid-protein complexes in the plasma membrane and the endocytic compartment. Eur J Cell Biol 78, 802-812 (1999).

25. Snyers, L., Thines-Sempoux, D. & Prohaska, R. Colocalization of stomatin (band 7.2b) and actin microfilaments in UAC epithelial cells. Eur J Cell Biol 73, 281-285 (1997).

26. Salzer, U. & Prohaska, R. Stomatin, flotillin-1, and flotillin-2 are major integral proteins of erythrocyte lipid rafts. Blood 97, 1141-1143 (2001).

27. Mairhofer, M., Steiner, M., Salzer, U. & Prohaska, R. Stomatin-like protein-1 interacts with stomatin and is targeted to late endosomes. J Biol Chem 284, 29218-29229 (2009).

28. Morrow, I.C. & Parton, R.G. Flotillins and the PHB domain protein family: rafts, worms and anaesthetics. Traffic 6, 725-740 (2005).

29. Langhorst, M.F., Reuter, A. & Stuermer, C.A. Scaffolding microdomains and beyond: the function of reggie/flotillin proteins. Cell Mol Life Sci 62, 2228-2240 (2005).

30. Hajek, P., Chomyn, A. & Attardi, G. Identification of a novel mitochondrial complex containing mitofusin 2 and stomatin-like protein 2. J Biol Chem 282, 5670-5681 (2007).

31. Da Cruz, S., Parone, P.A., Gonzalo, P., Bienvenut, W.V., Tondera, D., Jourdain, A., Quadroni, M. & Martinou, J.C. SLP-2 interacts with prohibitins in the mitochondrial inner membrane and contributes to their stability. Biochim Biophys Acta 1783, 904-911 (2008).

32. Sorice, M., Manganelli, V., Matarrese, P., Tinari, A., Misasi, R., Malorni, W. & Garofalo, T. Cardiolipin-enriched raft-like microdomains are essential activating platforms for apoptotic signals on mitochondria. FEBS Lett 583, 2447-2450 (2009).

33. Osman, C., Haag, M., Potting, C., Rodenfels, J., Dip, P.V., Wieland, F.T., Brugger, B., Westermann, B. & Langer, T. The genetic interactome of prohibitins: coordinated control of cardiolipin and phosphatidylethanolamine by conserved regulators in mitochondria. J Cell Biol 184, 583-596 (2009).

34. Osman, C., Voelker, D.R. & Langer, T. Making heads or tails of phospholipids in mitochondria. J Cell Biol 192, 7-16 (2011).

126

35. Frauwirth, K.A., Riley, J.L., Harris, M.H., Parry, R.V., Rathmell, J.C., Plas, D.R., Elstrom, R.L., June, C.H. & Thompson, C.B. The CD28 signaling pathway regulates glucose metabolism. Immunity 16, 769-777 (2002).

36. Acin-Perez, R., Fernandez-Silva, P., Peleato, M.L., Perez-Martos, A. & Enriquez, J.A. Respiratory active mitochondrial supercomplexes. Mol Cell 32, 529-539 (2008).

37. Osman, C., Merkwirth, C. & Langer, T. Prohibitins and the functional compartmentalization of mitochondrial membranes. J Cell Sci 122, 3823-3830 (2009).

38. Fry, M. & Green, D.E. Cardiolipin requirement for electron transfer in complex I and III of the mitochondrial respiratory chain. J Biol Chem 256, 1874-1880 (1981).

39. Garofalo, T., Giammarioli, A.M., Misasi, R., Tinari, A., Manganelli, V., Gambardella, L., Pavan, A., Malorni, W. & Sorice, M. Lipid microdomains contribute to apoptosis-associated modifications of mitochondria in T cells. Cell Death Differ 12, 1378-1389 (2005).

40. Li, Q., Vande Velde, C., Israelson, A., Xie, J., Bailey, A.O., Dong, M.Q., Chun, S.J., Roy, T., Winer, L., Yates, J.R., Capaldi, R.A., Cleveland, D.W. & Miller, T.M. ALS-linked mutant superoxide dismutase 1 (SOD1) alters mitochondrial protein composition and decreases protein import. Proc Natl Acad Sci U S A 107, 21146-21151 (2010).

41. Favaro, E., Ramachandran, A., McCormick, R., Gee, H., Blancher, C., Crosby, M., Devlin, C., Blick, C., Buffa, F., Li, J.L., Vojnovic, B., Pires das Neves, R., Glazer, P., Iborra, F., Ivan, M., Ragoussis, J. & Harris, A.L. MicroRNA-210 regulates mitochondrial free radical response to hypoxia and krebs cycle in cancer cells by targeting iron sulfur cluster protein ISCU. PLoS One 5, e10345 (2010).

42. McIntosh, R., Lee, S., Ghio, A.J., Xi, J., Zhu, M., Shen, X., Chanoit, G., Zvara, D.A. & Xu, Z. The critical role of intracellular zinc in adenosine A(2) receptor activation induced cardioprotection against reperfusion injury. J Mol Cell Cardiol 49, 41-47 (2010).

43. Finsterer, J. Leigh and Leigh-like syndrome in children and adults. Pediatr Neurol 39, 223-235 (2008).

44. Rich, P.R. & Marechal, A. The mitochondrial respiratory chain. Essays Biochem 47, 1-23 (2010).

45. Steglich, G., Neupert, W. & Langer, T. Prohibitins regulate membrane protein degradation by the m-AAA protease in mitochondria. Mol Cell Biol 19, 3435-3442 (1999).

127

46. Distelmaier, F., Koopman, W.J., van den Heuvel, L.P., Rodenburg, R.J., Mayatepek, E., Willems, P.H. & Smeitink, J.A. Mitochondrial complex I deficiency: from organelle dysfunction to clinical disease. Brain 132, 833-842 (2009).

47. Ma, Y.Y., Zhang, X.L., Wu, T.F., Liu, Y.P., Wang, Q., Zhang, Y., Song, J.Q., Wang, Y.J. & Yang, Y.L. Analysis of the Mitochondrial Complex I-V Enzyme Activities of Peripheral Leukocytes in Oxidative Phosphorylation Disorders. J Child Neurol Epub ahead of print (2011).

48. Kaminski, M.M., Sauer, S.W., Klemke, C.D., Suss, D., Okun, J.G., Krammer, P.H. & Gulow, K. Mitochondrial reactive oxygen species control T cell activation by regulating IL-2 and IL-4 expression: mechanism of ciprofloxacin-mediated immunosuppression. J Immunol 184, 4827-4841 (2010).

49. Michalek, R.D., Gerriets, V.A., Jacobs, S.R., Macintyre, A.N., MacIver, N.J., Mason, E.F., Sullivan, S.A., Nichols, A.G. & Rathmell, J.C. Cutting edge: distinct glycolytic and lipid oxidative metabolic programs are essential for effector and regulatory CD4+ T cell subsets. J Immunol 186, 3299-3303 (2011).

50. Bruzzone, S., Fruscione, F., Morando, S., Ferrando, T., Poggi, A., Garuti, A., D'Urso, A., Selmo, M., Benvenuto, F., Cea, M., Zoppoli, G., Moran, E., Soncini, D., Ballestrero, A., Sordat, B., Patrone, F., Mostoslavsky, R., Uccelli, A. & Nencioni, A. Catastrophic NAD+ depletion in activated T lymphocytes through Nampt inhibition reduces demyelination and disability in EAE. PLoS One 4, e7897 (2009).

51. Triepels, R.H., Van Den Heuvel, L.P., Trijbels, J.M. & Smeitink, J.A. Respiratory chain complex I deficiency. Am J Med Genet 106, 37-45 (2001).

52. Holen, K., Saltz, L.B., Hollywood, E., Burk, K. & Hanauske, A.R. The pharmacokinetics, toxicities, and biologic effects of FK866, a nicotinamide adenine dinucleotide biosynthesis inhibitor. Invest New Drugs 26, 45-51 (2008).

53. Garcia-Sanz, J.A. & Lenig, D. Translational control of interleukin 2 messenger RNA as a molecular mechanism of T cell anergy. The Journal of experimental medicine 184, 159-164 (1996).

54. Villarino, A.V., Katzman, S.D., Gallo, E., Miller, O., Jiang, S., McManus, M.T. & Abbas, A.K. Posttranscriptional silencing of effector cytokine mRNA underlies the anergic phenotype of self-reactive T cells. Immunity 34, 50-60 (2011).

55. Shaw, G. & Kamen, R. A conserved AU sequence from the 3' untranslated region of GM-CSF mRNA mediates selective mRNA degradation. Cell 46, 659-667 (1986).

128

56. Araki, K., Turner, A.P., Shaffer, V.O., Gangappa, S., Keller, S.A., Bachmann, M.F., Larsen, C.P. & Ahmed, R. mTOR regulates memory CD8 T-cell differentiation. Nature 460, 108-112 (2009).

57. Pearce, E.L., Walsh, M.C., Cejas, P.J., Harms, G.M., Shen, H., Wang, L.S., Jones, R.G. & Choi, Y. Enhancing CD8 T-cell memory by modulating fatty acid metabolism. Nature 460, 103-107 (2009).

129

Chapter 4 3

4 Plasma membrane and mitochondrial Stomatin-like Protein 2 pools coalesce at the immunological synapse during T cell activation

Stomatin-like protein 2 (SLP-2) is a member of the stomatin – prohibitin – flotillin –

HflC/K (SPFH) superfamily. Recent evidence indicates that SLP-2 is involved in the

organization of cardiolipin-enriched microdomains in mitochondrial membranes and the

regulation of mitochondrial biogenesis and function. In T cells, this role translates into

enhanced T cell activation. Although the major pool of SLP-2 is associated with

mitochondria, we show here that there is an additional pool of SLP-2 associated with the

plasma membrane of T cells. Both plasma membrane-associated and mitochondria-

associated pools of SLP-2 coalesce at the immunological synapse (IS) upon T cell

activation. SLP-2 is not required for formation of IS nor for the re-localization of

mitochondria to the IS because SLP-2-deficient T cells showed normal re-localization of

these organelles in response to T cell activation. Interestingly, upon T cell activation, we

found the surface pool of SLP-2 mostly excluded from the central supramolecular

activation complex, and enriched in the peripheral area of the IS where signalling TCR

microclusters are located. Based on these results, we propose that SLP-2 facilitates the

compartmentalization not only of mitochondrial membranes but also of the plasma

membrane into functional microdomains. In this latter location, SLP-2 may facilitate the

optimal assembly of TCR signalosome components. Our data also suggest that there may

be a net exchange of membrane material between mitochondria and plasma membrane,

explaining the presence of some mitochondrial proteins in the plasma membrane.

3 The material contained in this chapter is based on a submitted manuscript: Christie, D., Kirchhof, M.G.,

Vardhana, S., Dustin, M.L., and Madrenas, J. Mitochondrial and plasma membrane pools of Stomatin-like protein 2 coalesce at the immunological synapse during T cell activation. 2012

130

4.1 Introduction

Stomatin-like protein 2 (SLP-2), a widely expressed mitochondrial protein identified in

proteome analyses from several tissues and species, is a member of the stomatin family

as well as the stomatin – prohibitin – flotillin – HflC/K (SPFH) superfamily 1-4. In

mitochondria, SLP-2 associates tightly with the mitochondrial inner membrane. How

this association occurs is unclear because, unlike other stomatin family members, SLP-2

lacks a putative transmembrane domain. It contains though a SPFH domain, a highly

conserved region found in SPFH superfamily members 5-8, and this domain has been

linked to protein : lipid membrane interactions 9-11.

Recent studies have begun to shed light on the function of SLP-2. Similar to other SPFH

family members, SLP-2 has been found in large protein complexes and has been shown

to interact with mitochondrial proteins, including prohibitin (PHB) 1 and 2 as well as

mitofusin 2 5, 6, 8. We also have shown that SLP-2 binds directly to cardiolipin and can

regulate the localization of the prohibitin complex to cardiolipin-enriched domains in the

mitochondrial membrane 12. Furthermore, over-expression of SLP-2 leads to increased

mitochondrial biogenesis and function 5, 6, 8 whereas SLP-2 depletion results in decreased

mitochondrial transmembrane potential, reduced mitochondrial calcium uptake in

response to cell stimulation, enhanced apoptotic responses to cell stress and increased

degradation of mitochondrial proteins 5, 8, 12-14 (Christie et al, manuscript submitted). All

this evidence led us to propose that SLP-2 regulates all these mitochondrial functions (i.e.

energy production, calcium buffering and apoptosis) by organizing mitochondrial

membranes into defined cardiolipin-enriched microdomains, which then facilitate the

optimal assembly of membrane-associated molecular complexes.

Although most recent work on SLP-2 has focused on the function in mitochondria, we

originally identified SLP-2 in the glycolipid-enriched, detergent-insoluble microdomains

of human T cells activated through the T cell receptor 12. In these cells, we demonstrated

that SLP-2 was recruited to glycolipid-enriched detergent-insoluble microdomains and

interacted with numerous components of the TCR signalosome upon T cell stimulation,

including CD3, ZAP-70, LAT, lck and PLCγ. Furthermore, the level of SLP-2

expression correlated with the response of T cells to stimulation. Under conditions of

131

SLP-2 over-expression, T cells had significantly increased responses while knockdown of

SLP-2 resulted in significantly decreased T cell signalling. Together, these results

suggested that SLP-2 plays an important role in T cell activation, either by increasing

mitochondrial biogenesis 6 or by regulating T cell signalling at the plasma membrane 12.

To investigate the role of SLP-2 during T cell activation, we utilized a Jurkat T cell line

stably transfected with an inducible construct encoding either full-length SLP-2 tagged

with gfp or a gfp-tagged mutant construct lacking the amino-terminal region. As

expected, we found that most SLP-2 was associated with mitochondria. This primary

location was determined by an amino-terminal mitochondrial-targeting sequence of SLP-

2. Interestingly, a small pool of SLP-2 was also detected in association with the plasma

membrane. Upon T cell stimulation both pools of SLP-2 coalesced at the immunological

synapse (IS), and localized mostly in the peripheral supramolecular activation complex

(pSMAC). SLP-2 was not required for mitochondrial trafficking to the IS. Our data

indicate that there is a small pool of SLP-2 in the plasma membrane that, by analogy with

the mitochondrial pool of this protein, may play a role in the compartmentalization of the

plasma membrane, ensuring optimal assembly of multimolecular complexes.

4.2 Material and Methods

4.2.1 Cells

Jurkat T cells (E6.1) were obtained from American Type Culture Collection (Manassas,

VA) and cultured in supplemented RPMI 1640 medium. The B lymphoblastoid cell line,

LG2, was kindly provided by Dr. Eric Long (NIAID, NIH, Rockville, MD) and cultured

in standard supplemented RPMI 1640 media.

4.2.2 Plasmids, siRNA and T cell transfectants

Human SLP-2 cDNA was subcloned into the pEGFPN1 expression vector (Clontech Inc.

Palo Alto, CA) to create an in-frame translational fusion of the 3’ end of SLP-2 to gfp, as

previously described 15, 16. Subsequently, the SLP-2-gfp fusion was cloned into the

doxycycline inducible pBIG2i vector 15. A mutant construct lacking the amino-terminal

132

region (deletion of amino acid residues 1-77) was generated and cloned into pBIG2i.

Stable transfectants were generated by electroporating linearized plasmid into Jurkat E6.1

T cells and screening for stable expression of SLP-2-gfp. Doxycycline (Sigma, St. Louis,

MO) was added to overnight cultures at 1 µg/mL to induce SLP-2-gfp expression, which

was monitored by direct flow cytometry (Becton Dickinson [BD], San Jose, CA).

4.2.3 Mice

A full-length genomic fragment containing the mouse SLP-2 gene was cloned within the

lox sites of the 3LoxP3NwCD vector. Upon sequence confirmation, the plasmid was

electroporated into C57BL/6 embryonic stem cells and clones were selected for

neomycin resistance. The resistant clones were screened by Southern blot and PCR

analysis to confirm homologous recombination of the SLP-2-floxed DNA sequence.

Clones containing the SLP-2-floxed sequence were injected into B6/Try blastocysts and

implanted into a pseudo-pregnant mouse. The offspring was selected by chimeric coat

colour and backcrossed with C57BL/6 mice to produce SLP-2lox/wt mice in the C57BL/6

background. SLP-2 floxed mice were crossed with C57BL/6 mice transgenic for Cre

recombinase under the control of the CD4 promoter, purchased from Taconic (Hudson,

NY) 17, to generate a T cell specific SLP-2 knockout strain (SLP-2 T-K/O, Christie et al,

manuscript submitted). SLP-2 floxed mice lacking Cre were used as control mice.

Breeding colonies were derived from the same original SLP-2 floxed breeders and kept in

parallel. Mice were maintained in the animal facility at the University of Western

Ontario with approval from the Animal Use Subcommittee in accordance with the

Canadian Council on Animal Care Guidelines.

4.2.4 Antibodies

A rabbit polyclonal antiserum against human SLP-2 was generated by immunization with

a peptide spanning amino acids 343 to 356 (ProSci Inc., Poway, CA). Polyclonal

antibodies against SLP-2 (Protein Tech Group, Chicago, IL) and gfp (BD Bioscience,

San Jose, CA) were used in these studies. Pre-immune rabbit serum was used as an

isotype control (ProSci Inc., Poway, CA). Monoclonal antibodies against β-actin (Santa

Cruz Biotechnology, Santa Cruz, CA), the α-subunit of Complex V (MitoSciences,

133

Eugene, OR), biotin (Jackson ImmunoResearch, West Grove, PA), Caspase 3 (Cell

Signaling, Danvers, MA), GAPDH (Chemicon International, Temecula, CA) and PE-

labelled anti-CD3 (UCHT-I) were used (BD Bioscience, San Jose, CA) were used.

4.2.5 Mitochondria isolation

Intact mitochondria were isolated from 20 x 106 Jurkat T cells, SLP-2 T-K/O or wild type

control cells using the Qproteome Mitochondria Isolation Kit (Qiagen). Cytosolic

fractions were concentrated in Amicon Ultra-0.5 centrifugal filter units with Ultracel-30

membrane (Millipore, Billerica, MA). Mitochondrial and cytosolic preparations were

mixed with sample buffer containing β-mercaptoethanol, boiled, and analyzed by sodium

dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE).

4.2.6 Immunoprecipitations

Protein A or protein G agarose beads were coated with either SLP-2 antisera, pre-immune

sera, anti-gfp or anti-biotin antibodies overnight at 4oC 16. Beads were washed to remove

excess antibody and cell lysates were incubated with the antibody-coated beads overnight

at 4oC. Following immunoprecipitation, unbound proteins were removed and antibody-

bound protein complexes were eluted in sample buffer with β-mercaptoethanol.

Immunoprecipitated samples were separated by SDS-PAGE and immunoblotted with the

appropriate antibodies.

4.2.7 Cell surface biotinylation

Jurkat or mouse T cells were incubated with EZ-link sulfo-NHS-biotin (Pierce, Rockford,

IL) in PBS pH 8.0, for 30 minutes at room temperature. Cells were then washed three

times with cold PBS, lysed and immunoprecipitated for biotin 15, 18. Lysates and

immunoprecipitates were separated by SDS-PAGE and blotted with antibodies against

SLP-2.

4.2.8 Confocal microscopy

Confocal microscopy was performed with a Zeiss LSM 510 microscope. Jurkat T cells or

murine T cells (1x106/ml) were incubated on poly-L-lysine-coated (0.01%, Sigma) glass

134

bottom microwell dishes (MatTek Corp., MA) for 10 minutes to promote cell adherence

at 37ºC. SLP-2-gfp localization and mitochondrial movement was monitored by staining

cells with 100 nM MitoTracker Red CMXRos (Invitrogen, Burlington, ON, Canada) for

15 min at 37°C in complete RPMI 1640. SLP-2-gfp distribution during IS formation was

assessed by culturing doxycycline-induced SLP-2-gfp stably transfected T cells with LG2

antigen-presenting cells (APC) pre-incubated with 1 µg/ml staphylococcal enterotoxin E

(SEE) for either 10 or 30 minutes. Following the allotted time of co-incubation, the T

cell-APC conjugates were rapidly fixed with 4% paraformaldehyde, washed with

PBS+1% FCS and stained with PE conjugated anti-CD3 for 30 minutes on ice. For

experiments imaging mitochondrial localization in primary mouse cells, T cells were

isolated by magnetic separation using the MACS pan T cell isolation kit (Miltenyi Biotec,

Auburn, CA). Cells were stained with MitoTracker Red, stimulated with dynabeads

mouse T activator CD3/CD28 (Invitrogen, Burlington, ON, Canada) for 30 minutes and

cells were fixed and imaged 19. For experiments using planar membranes, glass-

supported dioleoylphosphatidylcholine bilayers incorporating Cy5-ICAM-1 (300

molecules/µm2) and 0.1% cap-biotin were prepared in a Bioptechs flow cell. Unlabelled

Streptavidin (8 µg/mL) and Cy3-conjugated anti-human CD3 (OKT3 clone, 10 µg/mL),

were loaded sequentially in HBS/HSA buffer (Hepes buffered saline supplemented with

5 mM glucose, 2 mM MgCl2, 1 mM CaCl2, and 1% human serum albumin). Jurkat T

cells were also suspended in HBS/HAS buffer. All microscopy was performed on an

automated microscope with a Hamamatsu USA Orca-ER cooled CCD camera. The

hardware on the microscope was controlled using IPLAB software (Scanalytics) on a

PowerMac G4 Macintosh computer. Images were exposed in wide-field for 1-2 s at a

resolution of 0.11 µm per pixel using the 60x 1.45 N.A. objective. Interference reflection

microscopy (IRM) is based on destructive interference in green light reflected from the

bilayer-cell interface leading to a dark area where cells are in close contact with the

bilayer. Images of cell interaction with the planar membrane were collected by IRM and

fluorescent images to examine re-localization of T cell proteins were collected by total

internal reflection fluorescence microscopy (TIRFM). Images were inspected using

Metamorph (Molecular Devices).

135

4.3 Results

4.3.1 The largest cellular pool of SLP-2 is associated with mitochondria

In order to assess the subcellular localization and movement of SLP-2 in T cells, we

constructed stably transfected Jurkat T cells with a doxycycline inducible construct

encoding SLP-2 tagged with gfp at the C-terminus. In addition, we also constructed a

truncated version of SLP-2 lacking the N-terminus of the protein (ΔN-SLP-2-gfp), as this

region contains a predicted mitochondrial targeting sequence 8. Transfected T cells were

visualized using confocal microscopy (Fig. 4.1a). As expected, we found that most SLP-

2-gfp was distributed in aggregates within the cytosol, which were shown to be

mitochondria by staining with the mitochondrial dye MitoTracker Red. In the absence of

the predicted mitochondrial targeting sequence (ΔN-SLP-2-gfp), the protein failed to

partition selectively in mitochondria and was instead found in a diffuse cytosolic pattern,

confirming the functionality of the mitochondrial-targeting domain in the N-terminus of

SLP-2.

The mitochondrial localization of SLP-2 was further supported by subcellular

fractionation and western blotting. As shown in Fig. 4.1b, both endogenous SLP-2 and

SLP-2-gfp were detected in the mitochondrial but not the cytosolic fractions of

transfected Jurkat T cells. The SLP-2-gfp blots showed intermediate degradation

products from the full length chimeric construct. The ΔN-SLP-2-gfp was absent in the

mitochondrial fraction and detected only in the cytosolic fraction, further confirming the

presence of a mitochondrial targeting domain at the amino terminus of SLP-2.

The previous results were corroborated using T cells from wildtype and T-cell-specific

SLP-2 knockout mice (Fig. 4.1c). Similar to human SLP-2, murine SLP-2 also partitioned

in mitochondria. As expected, the SLP-2 T-K/O cells had no detectable SLP-2, verifying

the identity of the mitochondrial SLP-2 band in the wild type mice. Altogether, these

results confirmed the predominant mitochondrial location of SLP-2 and validated the use

of the inducible SLP-2-gfp transfected T cells.

136

Figure 4.1: SLP-2 is a mitochondrial protein.

(a) Full-length SLP-2-gfp and ΔN-SLP-2-gfp were subcloned into a doxycycline-

inducible vector and transfected into Jurkat T cells. These stable transfectants were

imaged by confocal microscopy for SLP-2-gfp (first column of micrographs – green), or

after staining with MitoTracker Red (second column of micrographs – red). The third

colum of photomicrographs show overlapping of red and green signals as yellow signal.

(b) Mitochondrial and cytosolic fractions of parental, SLP-2-gfp and ΔN-SLP-2-gfp T

cell transfectants were isolated by differential centrifugation and immunoblotted for SLP-

2, the α-subunit of ATP synthase and actin. (c) Mitochondrial and cytosolic fractions

were isolated from T cells of wild type mice and T-cell specific SLP-2 conditional

knockout mice and blotted as in B. These results are representative of at least 3

independent experiments, and of more than 100 imaged cells.

137

Figure 4.1

138

4.3.2 Detection of a small pool of SLP-2 associated with the plasma membrane

Although initial studies of SLP-2 have focused on the mitochondrial localization 5, 6, 8, we

originally identified SLP-2 in total glycolipid-enriched, detergent-insoluble

microdomains from activated T cells 12. This suggested to us that there may be SLP-2

associated with other membranes, potentially plasma membranes. In order to test the

presence of SLP-2 associated with the plasma membrane, T cells were biotinylated and a

a biotin immunoprecipitation performed. In these experiments, untransfected Jurkat T

cells as well as SLP-2-gfp and ΔN-SLP-2-gfp stably transfected Jurkat T cells were used

to determine the association of full length SLP-2 and SLP-2 lacking the mitochondrial

targeting domain with T cell surface molecules (Fig. 4.2a). Both the endogenous and

SLP-2-gfp were found in association with cell surface molecules. In addition, the ΔN-

SLP-2-gfp was also found in association with the plasma membrane, indicating that the

mitochondrial targeting sequence is not required for association with surface molecules.

However, the interaction of ΔN-SLP-2-gfp with membrane proteins was much less

efficient because when expression of full length SLP-2-gfp and ΔN-SLP-2-gfp were

titrated the full-length SLP-2 associated with the plasma membrane at lower levels of

expression compared to ΔN-SLP-2-gfp (Fig. 4.2b). Even the very low level of leaky

expression of SLP-2-gfp in uninduced cells was sufficient for some SLP-2-gfp to interact

with plasma membrane proteins while ΔN-SLP-2-gfp surface pool was only detected

when cells expressed high levels of this mutant (by induction with 0.2 µg/mL of

doxycycline or higher).

The small surface-associated pool of SLP-2 was also found in mouse T cells. Biotin

immunoprecipitation was performed with wild type and SLP-2 T-K/O T cells and SLP-2

was found to co-precipitate with surface proteins in the wild type T cells but not in cells

lacking SLP-2 (Fig. 4.2c). This result confirms the existence of a plasma membrane-

associated pool of SLP-2 in association with surface proteins with an extracellular

domain (i.e. biotinylated).

139

Figure 4.2: A small pool of SLP-2 is associated with the plasma membrane.

(a) Intact parental Jurkat T cells, as well as Jurkat T cells stably transfected with full

length SLP-2-gfp or ΔN-SLP-2-gfp were biotinylated. Next, cells were lysed and surface

proteins were immunoprecipitated with an antibody against biotin. The

immunoprecipitate samples (Biotin ip) were immunoblotted for SLP-2 to detect the pool

of SLP-2 associated with surface molecules. Whole cell lysates (WCL) from the

transfectants were blotted to show expression levels of the transgenes. Samples were also

blotted for caspase 3 as a negative control for surface protein pull-down. (b) Increasing

levels of SLP-2-gfp or ΔN-SLP-2-gfp were induced with increasing concentrations of

doxycycline and biotin immunoprecipitation was performed. Whole cell lysates (WCL)

and biotin immunoprecipitates (biotin ip) were blotted as in a. (c) T cells isolated from

wild type and SLP-2 T-K/O mice were analyzed by biotin immunoprecipitation and

blotted as in a. Samples were also blotted for actin as a loading control for SLP-2 T-K/O

samples and also for GAPDH as a negative control for surface protein pull-down. These

results are representative of at least 3 independent experiments.

140

Figure 4.2

141

4.3.3 Homo-oligomerization of SLP-2

We have previously shown that SLP-2 is capable of interacting with numerous proteins,

including multiple components of the TCR signalosome and prohibitins 6, 12. However, it

is currently unclear if SLP-2 is capable of homo-oligomerization in vivo, without

exogenous cross-linkers. This may be an important feature of SLP-2 required to organize

specialized membrane microdomains. To test for homo-oligomerization of SLP-2, we

utilized the gfp-tagged constructs to examine co-precipitation of endogenous SLP-2 with

gfp-tagged SLP-2. In these experiments, parental Jurkat cells, SLP-2-gfp or ΔN-SLP-2-

gfp expressing Jurkat cells were immunoprecipitated with anti-gfp antibodies and

samples were blotted sequentially for SLP-2 and gfp (Fig. 4.3). As expected, gfp

immunoprecipitation of the parental cells failed to bring down any proteins,

demonstrating the specificity of this system. GFP-immunoprecipitation of SLP-2-gfp co-

precipitated endogenous SLP-2, documenting the ability of full-length SLP-2 to homo-

oligomerize. Of interest, ΔN-SLP-2-gfp did not co-precipitate with endogenous SLP-2 in

significant amounts. This suggests that the N-terminal region of SLP-2, by itself or

through its effect on mitochondrial targeting, is important for effective homo-

oligomerization of SLP-2.

4.3.4 SLP-2 redistributes to the immunological synapse during T cell activation

Next, we investigated the intracellular movement of SLP-2 during T cell activation.

Upon activation of SLP-2-gfp stably transfected T cells with APC and SEE, both the

plasma membrane-associated pool and the intracellular pool of SLP-2 polarized towards

putative immunological synapses, coalescing into major clusters close to the plasma

membrane, in the periphery of the IS and underneath the T cell-APC interface (Fig. 4.4a).

At the cell population level, such polarization was apparent in 60% of T cells forming

putative immunological synapses (Fig. 4.4b). Video capture of the redistribution of SLP-

2-gfp during T cell activation showed that as activation proceeded, greater than 80% of

the polarized SLP-2-gfp partitioned in the periphery of the synapse and underneath

142

Figure 4.3: Homo-oligomerization of SLP-2.

Parental Jurkat T cells and Jurkat T cells stably transfected with SLP-2-gfp or ΔN-SLP-2-

gfp were lysed. Lysates were used for immunoprecipitation with anti-GFP antibodies

(GFP ip), and immunoblotted serially for SLP-2 and gfp. Whole cell lysates (WCL) were

also blotted to show expression levels of endogenous SLP-2 and the SLP-2 transgenes.

This result is representative of at least 3 independent experiments.

143

Figure 4.3

144

Figure 4.4: SLP-2 polarizes to the immunological synapse during T cell activation.

(a) Stable SLP-2-gfp-transfected E6.1 Jurkat T cells were cultured with APC in the

presence (+SEE) or absence (-SEE) of SEE and examined by confocal microscopy for the

formation of putative synapses (identified as CD3-positive red clusters at the interface

between T cell and APC) and for the location of SLP-2-gfp signal (green). Images are

representative of at least 50 putative IS. Concomitant studies done with control

transfected T cells demonstrated that expression of SLP-2-gfp did not interfere with IS

formation. (b) Confocal images collected from 50 putative IS images (in A) were

analyzed for SLP-2-gfp intracellular localization. SLP-2-gfp signal in un-stimulated (0

min) or stimulated cells (30 min) was classified as predominantly proximal to the IS

(white bars), predominantly distal to the IS (black bars), or diffuse throughout the cell

(grey bars). (c) Redistribution of SLP-2 during T cell activation is shown by a series of

videomicroscopy capture images during IS formation. The dotted circle outlines the APC

interacting with the Jurkat T cell and the arrow indicates the mature IS with the SLP-2

localization. (d) SLP-2-gfp-transfected Jurkat T cells were stimulated with antibodies

against CD3 and examined by confocal microscopy. Photobleaching was induced at the

arrow-indicated site and regaining of the signal at that site was monitored for 6 minutes at

20 second intervals. Non-specific distribution is documented by progressive regaining of

signal with non-bleached SLP-2, while active distribution correlates with lack of

regaining of signal.

145

Figure 4.4

146

the putative IS at late time points (Fig. 4.4c). Only a small fraction of SLP-2 was

detectable in the centre of the synapse. Accumulation of SLP-2 in the periphery of the IS

mirrored the distribution of pSMACs, known to be enriched in LFA-1. The SLP-2

clusters during T cell activation were actively retained and stable as shown by

photobleaching experiments (Fig. 4.4d). In these experiments, the SLP-2-gfp signal was

restored after 400 seconds of bleaching the plasma membrane of non-stimulated T cells

but was not restored when SLP-2-gfp clusters in OKT3-stimulated T cells were bleached

within the same timeline, indicating that T cell activation was associated with stable SLP-

2-gfp clusters unable to freely move within the cell.

4.3.5 The SLP-2 pools predominantly partition in the pSMAC of the immunological synapse

To further refine our data concerning SLP-2-gfp redistribution to the IS, we used

supported planar bilayers containing anti-CD3 and ICAM-1 to which T cells form IS-like

structures. This system has excellent optics for resolution of components in the IS. With

this system, we visualized the formation of IS by Jurkat cells expressing SLP-2-gfp (Fig.

4.5). Under conditions of doxycycline-induced SLP-2-gfp overexpression, we confirmed

the presence of two pools of this molecule. The plasma membrane-associated pool of

SLP-2 was visualized by total internal reflection fluorescence microscopy (TIRFM) as

clusters located at less than 200nm from the glass plane (i.e., sites of TCR and LFA-1

engagement) (Fig. 4.5a). The small plasma membrane pool of SLP-2 distributed

uniformly in a granular appearance interspersed with TCR microclusters in the nascent IS

(Fig. 4.5a). As activation proceeded, the TCR microclusters were surrounded by densely

packed SLP-2 clusters. These TCR microclusters weakly excluded the SLP-2 clusters

resulting in up to 50% reduction of SLP-2 fluorescence in the cSMACs.

147

Figure 4.5: Redistribution of plasma membrane-associated SLP-2 and

mitochondria-associated SLP-2 during immunological synapse formation.

Jurkat T cells stably expressing SLP-2-gfp were incubated on planar membranes

containing ICAM-1 and anti-CD3 to induce immunosynapse formation. Cells were

imaged at early (<5 min) and late (15-30 min) stages of immunosynapse formation to

demonstrate SLP-2 redistribution upon TCR ligation. (a) Plasma membrane-associated

SLP-2 redistribution during immunosynapse formation was imaged by TIRFM, to

eliminate signal from the mitochondrial pool of SLP-2. Cell contact with the planar

membrane was imaged by IRM, TCR images were obtained by wide-field fluorescence

microscopy and the image overlay represents SLP-2-gfp as green and TCR as red. (b)

Total SLP-2-gfp, including both plasma membrane-associated and mitochondrial pools

was imaged at early and late stages of immunosynapse formation. The bright field image

shows cells being imaged, IRM images show contact with the bilayer as dark areas, and

the TCR, ICAM-1 and SLP-2-gfp fluorescence channels are shown in gray scale and two

red-green merges (green is always SLP-2). The dotted lines in the ICAM-1 picture

represent, from the periphery to the centre of the picture, the outer boundaries of the

distal SMAC, of the pSMAC, and of the cSMAC. Images are representative 3 separate

experiments. (c) SLP-2-gfp expressing Jurkat cells were transfected with mitochondria-

targeted RFP to verify mitochondrial localization of intracellular SLP-2-gfp. Images were

obtained by wide-field fluorescence microscopy and are representative of three separate

experiments. The dotted lines in the ICAM-1 image are as described in b. The

percentage of cells showing segregation of the mitochondrial pool of SLP-2 away from

the cSMAC is shown in Table 4.1 and this organization of SLP-2 into the pSMAC

requires anti-CD3 ligation (Table 4.2).

148

Figure 4.5

149

In addition, the major mitochondria-associated pool of SLP-2-gfp located at more than

200 nm from the plasma membrane, and also polarized to the IS and segregated from the

cSMACs to distribute close to the pSMACs as activation proceeded (Fig. 4.5b and Table

4.1). The mitochondrial origin of this pool of SLP-2 was confirmed by co-localization of

the gfp signal with a mitochondria-targeted red fluorescence protein (mtRFP) (Fig. 4.5c).

The polarization and partitioning of SLP-2 to the synapse required TCR engagement. In

experiments using planar membranes containing only ICAM-1 without anti-CD3, ICAM-

1 ligated LFA-1 on the Jurkat T cell to induce immunosynapse formation. Under these

conditions, SLP-2 failed to redistribute to the pSMAC and was instead distributed

throughout the membrane (Table 4.2). However, in the presence of both ICAM-1 and

anti-CD3 in the planar membrane, immunosynapse formation was accompanied by

redistribution of SLP-2-gfp to the pSMAC, with 86% of cells showing exclusion of SLP-

2 from the cSMAC (Table 4.1), indicating the requirement of TCR ligation for SLP-2

redistribution. Together, the results presented here confirm the presence of both a

mitochondrial and plasma membrane pool of SLP-2 and demonstrates the redistribution

of both SLP-2 pools to surround TCR microclusters in the pSMAC during T cell

activation. These data support a general model of SLP-2 function involving the

organization of membranes into functional microdomains to support multiple cell

functions and specifically T cell activation, as shown here.

150

Table 4.1

Table 4.1: Segregation of mitochondrial SLP-2-gfp from the cSMAC in the mature

IS

Segregation No Segregation

25/29 (86.2%) 4/29 (13.8%)

Only cells forming IRM contacts for 3 or more data points by wide field fluorescence

microscopy were considered. Images of each field were taken at 2-3 minute intervals.

Data is representative of 3 experiments.

151

Table 4.2

Table 4.2: Percentage of Jurkat T cells on supported planar bilayers organizing

SLP-2-gfp close to pSMAC at the contact interface

Peripheral ring No peripheral ring

10µg/mL OKT3 + 300 mol/µm2 ICAM-1 291 8

300 mol/µm2 ICAM-1 only 0 29

1P<0.0001 vs. ICAM-1 only

152

4.3.6 Mitochondrial recruitment to the IS does not require SLP-2

It has been shown previously that mitochondria relocate to the IS upon T cell stimulation

and that this migration is important for T cell activation 20. We have previously shown

that SLP-2 interacts with the actin cytoskeleton and treatment of Jurkat cells with

cytochalasin D inhibits T cell responses 12. Interestingly, the migration of mitochondria

to the IS during T cell stimulation is dependent on microtubules as well as the actin

cytoskeleton, and inhibition of actin polymerization inhibits mitochondrial movement and

T cell activation 20. To determine if SLP-2 was involved in mitochondrial translocation

to the IS, we imaged mitochondrial movement in response to TCR stimulation in wild

type and SLP-2-deficient primary T cells using confocal microscopy (Fig. 4.6a). For

these experiments, we used cells from conditional SLP-2 knockout mice we have recently

generated (Chapter 3), and stimulated these T cells with anti-CD3 + anti-CD28 antibody

coated beads for 30 minutes at 37°C 15. We found that, in unstimulated cells, the

mitochondrial population was located primarily around the large nucleus (representative

confocal images shown in Fig. 4.6a, quantification of mitochondrial localization shown in

Fig. 4.6b). Upon T cell stimulation, the mitochondrial population re-distributed and was

generally found proximal to the stimulating bead, under the IS (Fig. 4.5c). In addition,

many cells also showed mitochondrial movement to the uropod, classified here as distal

localization (Fig. 4.6a and 4.6c), which is characteristic of migrating lymphocytes 21.

Importantly, there was no apparent defect in mitochondrial migration in T cells lacking

SLP-2, indicating that mitochondrial movement in T cells is independent of SLP-2.

153

Figure 4.6: SLP-2-deficient T cells show normal mitochondrial recruitment upon T

cell stimulation.

T cells were isolated from wild type and SLP-2 T-K/O (T-K/O) mice and stained with

MitoTracker Red. Stained cells were plated on poly-L-lysine coated confocal dishes and

incubated for 10 minutes to promote adherence. Cells were stimulated for 30 minutes

with anti-CD3 and anti-CD28 antibody coated beads or left unstimulated. Cells were

then fixed and imaged by confocal microscopy. These images are representative of at

least 100 individual cells, imaged in three independent experiments. (a) Representative

confocal images are shown for wild type and SLP-2-deficient T cells in the absence (0

min) and presence (30 min) of T cell stimulation (2 different cells for each).

Mitochondria are shown in red and stimulating beads can be seen in the light image

overlay. (b) The location of the mitochondria was quantified in un-stimulated wild type

(black bars) and SLP-2-deficient (white bars) T cells was quantified as either a polar

distribution with mitochondria being clustered together at one end of the cell or a uniform

distribution, with mitochondria located throughout the entire cell. (c) The location of the

mitochondria in stimulated wild type and SLP-2-deficient T cells was quantified as

proximal to the stimulating bead, distal to the bead or uniformly distributed throughout

the cytoplasm, in a manner similar to that in figure 4.4. These plots represent an average

of 3 independent experiments, in which 30 cells for each group were counted.

154

Figure 4.6

155

4.4 Discussion

SLP-2 is a member of the highly conserved stomatin family as well as the SPFH

superfamily, consisting of stomatins, prohibitins, flotillins and HflC/K proteins 9.

Although the function of the stomatin family is currently unknown, these proteins are

found in glycolipid-enriched, detergent-insoluble microdomains and may be involved in

the interaction between protein and lipids in cellular membranes 22-24. Specifically, SLP-

2 is a mostly mitochondrial protein that has been linked to the formation of cardiolipin-

enriched microdomains, thus regulating mitochondrial functions, including energy

production and apoptotic responses 6. SLP-2 has also been identified in detergent-

resistant, glycolipid-enriched microdomains of activated T cells and shown to regulate T

cell activation through the TCR. The mechanism of action by which SLP-2 does it is

unknown 12. The work presented here investigates the link between T cell activation and

the role of SLP-2 in membrane organization. We show that there are two intracellular

pools of SLP-2, one major pool associated to mitochondria and another much less

abundant pool associated to the plasma membrane. Both pools coalesce at the IS upon

TCR ligation, and partition in the peripheral areas of the synapse and are actively

maintained in this location during activation. Together, these results suggest that SLP-2

regulates T cell activation by organizing functional microdomains in mitochondrial and

plasma membranes. These microdomains can then facilitate the optimal assembly and

function of multi-chain complexes or receptors.

The compartmentalization of cellular membranes into specialized microdomains is

thought to facilitate the assembly of multi-molecular complexes. This role has been

shown in the mitochondrial inner membrane for individual complexes of the respiratory

chain and supercomplexes of these individual units, together leading to increased electron

transport efficiency and increased ATP production. This role correlates with SLP-2

directly binding to cardiolipin and interacting with prohibitins 6, 12. Other SPFH family

members, such as prohibitin and flotillin are also proposed to organize membrane

domains 10, 11. Alternatively, SLP-2-deficient T cells have less cardiolipin-enriched

domains and decreased mitochondrial respiration (Chapter 3), and loss of transmembrane

156

potential 5, 25. Altogether, the net effect of the increase in SLP-2 levels upon T cell

activation would be an increase in cardiolipin-enriched microdomains and enhanced

mitochondrial function resulting in increased T cell function.

The data presented here extends the role of SLP-2 as an organizer of membrane

compartmentalization to the plasma membrane. Upon TCR ligation, there is recruitment

of a large number of adapter and signalling molecules to form the TCR signalosome 26.

Assembly of this multi-molecular complex is also associated with re-organization of

plasma membranes, through the aggregation of detergent-resistant, glycolipid-enriched

microdomains previously referred to as lipid rafts 27-30, and the formation of an IS 31.

Within the IS, stable signalling TCR microclusters containing kinases and adapters are

detected in the outer ring of the synapse together with LFA-1 and other adhesion

molecules forming the pSMAC 32. As activation proceeds, TCR microclusters move

from the periphery to the centre of the synapse (cSMACs), destined for internalization

and degradation 33-35, a process that ensures signal down-regulation 36. The results

presented here are compatible with the idea that SLP-2 facilitates the assembly of the

TCR signalosome during T cell activation. In a manner similar to Mec-2 and podocin,

members of the SPFH superfamily, SLP-2 may associate with cholesterol at the plasma

membrane and with components of the TCR signalosome the stability of this

multimolecular complex 7, 12. The ability of SLP-2 to homo-oligomerize may allow SLP-

2 to form a ring-like structure around the signalosome to separate components from other

proteins at the plasma membrane and to prevent dissociation of the complex, in a manner

similar to what SLP-2:prohibitin complexes may do in mitochondria 6, 11.

How SLP-2 specifically interacts with cardiolipin in the mitochondrial membrane and

other lipids in the plasma membrane is an issue for further investigation. SLP-2 lacks a

putative transmembrane domain and palmitoylation sites found in other stomatin family

members 5. We have previously indicated that there may be putative myristoylation sites

at the amino-terminus of SLP-2, which may facilitate membrane association 12.

Alternatively, the SPFH domain may be directly involved in this association as it has

been shown for the superfamily members Mec-2 and podocin and its binding to

cholesterol and for SLP-2 and its binding to cardiolipin 6, 7.

157

Using video capture microscopy we have shown that the mitochondrial-associated pool

of SLP-2 redistributed to the IS upon T cell activation. This result corroborates the

observation of mitochondrial relocation to the IS upon T cell stimulation 20. In this work,

the authors suggest a model in which mitochondria act as a localized calcium sink to

maintain Calcium Release Activated Calcium (CRAC) channel activation, strengthening

T cell signalling and activation. Studies looking at knockdown of SLP-2 with siRNA

have shown that the loss of SLP-2 leads to decreased calcium uptake by the mitochondria 13. Similarly, we have found that mitochondrial calcium uptake is increased in cells over-

expressing full-length SLP-2 (C. Lemke and J. Madrenas, unpublished observations).

However, it is currently unclear how SLP-2 modulates calcium uptake by the

mitochondria. While an inhibitor of mitochondrial sodium/calcium exchange channel

equalized the rate of calcium extrusion from the mitochondria in control and SLP-2-

depleted cells, it did not restore full calcium uptake into the mitochondria 13, indicating

that calcium exchange rates alone cannot account for the role of SLP-2 in mitochondrial

calcium uptake.

Since we had previously shown that SLP-2 interacts with actin and it has been shown that

the mitochondrial translocation to the IS requires actin polymerization 12, 20, it was

plausible that SLP-2 may link the mitochondria to the actin cytoskeleton and facilitate the

movement of mitochondria to the IS. However, our data indicate that this is not the case

because we found that mitochondrial translocation to the IS proceeded normally in SLP-

2-deficient T cells. Thus, SLP-2 is not required for mitochondrial translocation to the IS

during T cell activation.

The identification of SLP-2 and other mitochondrial proteins at the plasma membrane

may reflect membrane exchange between these organelles and other cellular

compartments and organelles, and point to a novel pathway of intracellular trafficking.

We have confirmed the presence of a mitochondrial targeting sequence in the amino-

terminus of SLP-2. This raised the question of the intracellular trafficking pathway

leading to the presence of SLP-2 in the cell surface. It is possible that the plasma

membrane pool of SLP-2 arises by default when a small proportion of newly synthesized

SLP-2 fails to be transported to the mitochondria. However, it is also possible that a

158

small amount of membrane exchange occurs between the mitochondrial and plasma

membranes, allowing for the exchange of membrane-associated proteins, which is

supported by the existence of mitochondria-derived vesicles and its fusion with other

organelles such as lysosomes 37-40. In addition to SLP-2, a small surface pool has also

been shown for other mitochondrial proteins such as prohibitin, porin, NADH

dehydrogenase, ubiquinol-cytochrome c reducatase and ATP synthase, indicating that the

surface association of mitochondrial proteins is not unique to SLP-2 41-44. The purpose of

mitochondrial proteins at the plasma membrane is unclear, although the association of

SLP-2 with components of the T cell signalosome suggest that both pools of SLP-2 are

likely functional.

In summary, the work presented here provides the first evidence of two pools of SLP-2 in

human and mouse T cells and it suggests to a conservation of function at both

membranes. In the mitochondria, SLP-2 organizes the membrane into cardiolipin-

enriched domains to facilitate respiratory chain function. At the plasma membrane, SLP-

2 may facilitate immunosynapse organization, leading to increased T cell signalling and

activation. It is likely that both of these functions have the same mechanistic basis, i.e.

organization of specialized microdomains that facilitate the assembly of multi-molecular

complexes and ultimately ensure their optimal function.

159

4.5 References

1. Da Cruz, S., Xenarios, I., Langridge, J., Vilbois, F., Parone, P.A. & Martinou, J.C. Proteomic analysis of the mouse liver mitochondrial inner membrane. J Biol Chem 278, 41566-41571 (2003).

2. Chevallet, M., Lescuyer, P., Diemer, H., van Dorsselaer, A., Leize-Wagner, E. & Rabilloud, T. Alterations of the mitochondrial proteome caused by the absence of mitochondrial DNA: A proteomic view. Electrophoresis 27, 1574-1583 (2006).

3. Rezaul, K., Wu, L., Mayya, V., Hwang, S.I. & Han, D. A systematic characterization of mitochondrial proteome from human T leukemia cells. Mol Cell Proteomics 4, 169-181 (2005).

4. Taylor, S.W., Fahy, E., Zhang, B., Glenn, G.M., Warnock, D.E., Wiley, S., Murphy, A.N., Gaucher, S.P., Capaldi, R.A., Gibson, B.W. & Ghosh, S.S. Characterization of the human heart mitochondrial proteome. Nat Biotechnol 21, 281-286 (2003).

5. Hajek, P., Chomyn, A. & Attardi, G. Identification of a novel mitochondrial complex containing mitofusin 2 and stomatin-like protein 2. J Biol Chem 282, 5670-5681 (2007).

6. Christie, D.A., Lemke, C.D., Elias, I.M., Chau, L.A., Kirchhof, M.G., Li, B., Ball, E.H., Dunn, S.D., Hatch, G.M. & Madrenas, J. Stomatin-like protein 2 binds cardiolipin and regulates mitochondrial biogenesis and function. Mol Cell Biol 31, 3845-3856 (2011).

7. Huber, T.B., Schermer, B., Muller, R.U., Hohne, M., Bartram, M., Calixto, A., Hagmann, H., Reinhardt, C., Koos, F., Kunzelmann, K., Shirokova, E., Krautwurst, D., Harteneck, C., Simons, M., Pavenstadt, H., Kerjaschki, D., Thiele, C., Walz, G., Chalfie, M. & Benzing, T. Podocin and MEC-2 bind cholesterol to regulate the activity of associated ion channels. Proc Natl Acad Sci U S A 103, 17079-17086 (2006).

8. B, W.A., G, I.H., L, M.I., M, L.J., V, P.M., H, I.N., V, A.M., P, J.S., H, M.T. & A, I.Z. [Part VI. Antiparasitic treatment for Chagas disease]. Rev Chilena Infectol 25, 384-389, discussion 387 (2008).

9. Browman, D.T., Hoegg, M.B. & Robbins, S.M. The SPFH domain-containing proteins: more than lipid raft markers. Trends Cell Biol 17, 394-402 (2007).

10. Morrow, I.C. & Parton, R.G. Flotillins and the PHB domain protein family: rafts, worms and anaesthetics. Traffic 6, 725-740 (2005).

160

11. Osman, C., Merkwirth, C. & Langer, T. Prohibitins and the functional compartmentalization of mitochondrial membranes. J Cell Sci 122, 3823-3830 (2009).

12. Kirchhof, M.G., Chau, L.A., Lemke, C.D., Vardhana, S., Darlington, P.J., Marquez, M.E., Taylor, R., Rizkalla, K., Blanca, I., Dustin, M.L. & Madrenas, J. Modulation of T cell activation by stomatin-like protein 2. J Immunol 181, 1927-1936 (2008).

13. Da Cruz, S., De Marchi, U., Frieden, M., Parone, P.A., Martinou, J.C. & Demaurex, N. SLP-2 negatively modulates mitochondrial sodium-calcium exchange. Cell Calcium 47, 11-18 (2009).

14. Tondera, D., Grandemange, S., Jourdain, A., Karbowski, M., Mattenberger, Y., Herzig, S., Da Cruz, S., Clerc, P., Raschke, I., Merkwirth, C., Ehses, S., Krause, F., Chan, D.C., Alexander, C., Bauer, C., Youle, R., Langer, T. & Martinou, J.C. SLP-2 is required for stress-induced mitochondrial hyperfusion. EMBO J 28, 1589-1600 (2009).

15. Arp, J., Kirchhof, M.G., Baroja, M.L., Nazarian, S.H., Chau, T.A., Strathdee, C.A., Ball, E.H. & Madrenas, J. Regulation of T-cell activation by phosphodiesterase 4B2 requires its dynamic redistribution during immunological synapse formation. Mol Cell Biol 23, 8042-8057 (2003).

16. Baroja, M.L., Cieslinski, L.B., Torphy, T.J., Wange, R.L. & Madrenas, J. Specific CD3 epsilon association of a phosphodiesterase 4B isoform determines its selective tyrosine phosphorylation after CD3 ligation. J Immunol 162, 2016-2023 (1999).

17. Lee, P.P., Fitzpatrick, D.R., Beard, C., Jessup, H.K., Lehar, S., Makar, K.W., Perez-Melgosa, M., Sweetser, M.T., Schlissel, M.S., Nguyen, S., Cherry, S.R., Tsai, J.H., Tucker, S.M., Weaver, W.M., Kelso, A., Jaenisch, R. & Wilson, C.B. A critical role for Dnmt1 and DNA methylation in T cell development, function, and survival. Immunity 15, 763-774 (2001).

18. Darlington, P.J., Baroja, M.L., Chau, T.A., Siu, E., Ling, V., Carreno, B.M. & Madrenas, J. Surface cytotoxic T lymphocyte-associated antigen 4 partitions within lipid rafts and relocates to the immunological synapse under conditions of inhibition of T cell activation. The Journal of experimental medicine 195, 1337-1347 (2002).

19. Tomas, E.M., Chau, T.A. & Madrenas, J. Clustering of a lipid-raft associated pool of ERM proteins at the immunological synapse upon T cell receptor or CD28 ligation. Immunol Lett 83, 143-147 (2002).

20. Quintana, A., Schwindling, C., Wenning, A.S., Becherer, U., Rettig, J., Schwarz, E.C. & Hoth, M. T cell activation requires mitochondrial translocation to the immunological synapse. Proc Natl Acad Sci U S A 104, 14418-14423 (2007).

161

21. Campello, S., Lacalle, R.A., Bettella, M., Manes, S., Scorrano, L. & Viola, A. Orchestration of lymphocyte chemotaxis by mitochondrial dynamics. J Exp Med 203, 2879-2886 (2006).

22. Snyers, L., Umlauf, E. & Prohaska, R. Association of stomatin with lipid-protein complexes in the plasma membrane and the endocytic compartment. Eur J Cell Biol 78, 802-812 (1999).

23. Snyers, L., Thines-Sempoux, D. & Prohaska, R. Colocalization of stomatin (band 7.2b) and actin microfilaments in UAC epithelial cells. Eur J Cell Biol 73, 281-285 (1997).

24. Mairhofer, M., Steiner, M., Salzer, U. & Prohaska, R. Stomatin-like protein-1 interacts with stomatin and is targeted to late endosomes. J Biol Chem 284, 29218-29229 (2009).

25. Wang, Y., Cao, W., Yu, Z. & Liu, Z. Downregulation of a mitochondria associated protein SLP-2 inhibits tumor cell motility, proliferation and enhances cell sensitivity to chemotherapeutic reagents. Cancer Biol Ther 8, 1651-1658 (2009).

26. Smith-Garvin, J.E., Koretzky, G.A. & Jordan, M.S. T cell activation. Annu Rev Immunol 27, 591-619 (2009).

27. Xavier, R., Brennan, T., Li, Q., McCormack, C. & Seed, B. Membrane compartmentation is required for efficient T cell activation. Immunity 8, 723-732 (1998).

28. Janes, P.W., Ley, S.C. & Magee, A.I. Aggregation of lipid rafts accompanies signaling via the T cell antigen receptor. J Cell Biol 147, 447-461 (1999).

29. Janes, P.W., Ley, S.C., Magee, A.I. & Kabouridis, P.S. The role of lipid rafts in T cell antigen receptor (TCR) signalling. Semin Immunol 12, 23-34 (2000).

30. Viola, A., Schroeder, S., Sakakibara, Y. & Lanzavecchia, A. T lymphocyte costimulation mediated by reorganization of membrane microdomains. Science 283, 680-682 (1999).

31. Fooksman, D.R., Vardhana, S., Vasiliver-Shamis, G., Liese, J., Blair, D.A., Waite, J., Sacristan, C., Victora, G.D., Zanin-Zhorov, A. & Dustin, M.L. Functional anatomy of T cell activation and synapse formation. Annu Rev Immunol 28, 79-105 (2010).

32. Gascoigne, N.R. & Zal, T. Molecular interactions at the T cell-antigen-presenting cell interface. Curr Opin Immunol 16, 114-119 (2004).

33. Lee, K.H., Dinner, A.R., Tu, C., Campi, G., Raychaudhuri, S., Varma, R., Sims, T.N., Burack, W.R., Wu, H., Wang, J., Kanagawa, O., Markiewicz, M., Allen,

162

P.M., Dustin, M.L., Chakraborty, A.K. & Shaw, A.S. The immunological synapse balances T cell receptor signaling and degradation. Science 302, 1218-1222 (2003).

34. Lee, K.H., Holdorf, A.D., Dustin, M.L., Chan, A.C., Allen, P.M. & Shaw, A.S. T cell receptor signaling precedes immunological synapse formation. Science 295, 1539-1542 (2002).

35. Varma, R., Campi, G., Yokosuka, T., Saito, T. & Dustin, M.L. T cell receptor-proximal signals are sustained in peripheral microclusters and terminated in the central supramolecular activation cluster. Immunity 25, 117-127 (2006).

36. Mossman, K.D., Campi, G., Groves, J.T. & Dustin, M.L. Altered TCR signaling from geometrically repatterned immunological synapses. Science 310, 1191-1193 (2005).

37. Braschi, E., Goyon, V., Zunino, R., Mohanty, A., Xu, L. & McBride, H.M. Vps35 mediates vesicle transport between the mitochondria and peroxisomes. Curr Biol 20, 1310-1315 (2010).

38. Neuspiel, M., Schauss, A.C., Braschi, E., Zunino, R., Rippstein, P., Rachubinski, R.A., Andrade-Navarro, M.A. & McBride, H.M. Cargo-selected transport from the mitochondria to peroxisomes is mediated by vesicular carriers. Curr Biol 18, 102-108 (2008).

39. Soubannier, V., McLelland, G.L., Zunino, R., Braschi, E., Rippstein, P., Fon, E.A. & McBride, H.M. A Vesicular Transport Pathway Shuttles Cargo from Mitochondria to Lysosomes. Curr Biol (2012).

40. Andrade-Navarro, M.A., Sanchez-Pulido, L. & McBride, H.M. Mitochondrial vesicles: an ancient process providing new links to peroxisomes. Curr Opin Cell Biol 21, 560-567 (2009).

41. Bathori, G., Parolini, I., Tombola, F., Szabo, I., Messina, A., Oliva, M., De Pinto, V., Lisanti, M., Sargiacomo, M. & Zoratti, M. Porin is present in the plasma membrane where it is concentrated in caveolae and caveolae-related domains. The Journal of biological chemistry 274, 29607-29612 (1999).

42. Bae, T.J., Kim, M.S., Kim, J.W., Kim, B.W., Choo, H.J., Lee, J.W., Kim, K.B., Lee, C.S., Kim, J.H., Chang, S.Y., Kang, C.Y., Lee, S.W. & Ko, Y.G. Lipid raft proteome reveals ATP synthase complex in the cell surface. Proteomics 4, 3536-3548 (2004).

43. Kim, K.B., Lee, J.W., Lee, C.S., Kim, B.W., Choo, H.J., Jung, S.Y., Chi, S.G., Yoon, Y.S., Yoon, G. & Ko, Y.G. Oxidation-reduction respiratory chains and ATP synthase complex are localized in detergent-resistant lipid rafts. Proteomics 6, 2444-2453 (2006).

163

44. Kim, B.W., Lee, C.S., Yi, J.S., Lee, J.H., Lee, J.W., Choo, H.J., Jung, S.Y., Kim, M.S., Lee, S.W., Lee, M.S., Yoon, G. & Ko, Y.G. Lipid raft proteome reveals that oxidative phosphorylation system is associated with the plasma membrane. Expert Rev Proteomics 7, 849-866 (2010).

164

Chapter 5

5 Discussion

5.1 Summary of results

5.1.1 Stomatin-like protein 2 binds cardiolipin and regulates mitochondrial biogenesis and function

To investigate the role of SLP-2 in T cells, a doxycycline-inducible SLP-2 construct

tagged with GFP at the C-terminus was stably transfected into Jurkat T cells, a human T

cell line. Sub-cellular fractionation demonstrated that SLP-2 was localized to the

mitochondria and this was confirmed by confocal imaging showing co-localization of

SLP-2-GFP with the mitochondrial-specific dye MitoTracker Red. SLP-2-GFP over-

expressing cells had an increase in mitochondrial mass and an increase in the number of

mitochondria per cell slice in transmission electron microscopy images. This was

confirmed in primary human peripheral blood mononuclear cells (PBMCs), in which

ionomycin and PMA treatment led to increased SLP-2 expression and increased

mitochondrial mass.

To investigate the mechanism of increased mitochondrial populations upon SLP-2 over-

expression, the cellular programs required for mitochondrial biogenesis, including lipid

synthesis, nuclear transcription and mitochondrial DNA replication, were examined. In

both SLP-2 over-expressing Jurkat T cells and stimulated PBMCs, the level of the

mitochondrial-specific lipid cardiolipin was increased. An increase in radiolabelled

precursor incorporation demonstrated that the increase in cardiolipin was due to increased

biosynthesis. Supporting this finding, expression of enzymes required for the final two

steps of cardiolipin biosynthesis were also elevated in SLP-2 over-expressing cells. This

increased cardiolipin biosynthesis was required for mitochondrial biogenesis as inhibition

of cardiolipin synthase blocked mitochondrial biogenesis in response to T cell

stimulation. In addition, SLP-2 over-expressing cells also had increased expression

levels of PGC-1α, a transcriptional co-activator and master regulator of nuclear

165

transcription programs for mitochondrial targeted genes. Finally, SLP-2 over-expressing

cells showed increased levels of mitochondrial DNA, which is essential for functional

mitochondrial populations. The timeline of mitochondrial biogenesis upon T cell

stimulation demonstrated that expression of SLP-2 and prohibitins increased at early time

points, followed by an increase in PGC-1α expression. These events preceded increased

mitochondrial mass, which was seen by 24 hours post-stimulation.

As a possible mechanism for the increase in mitochondrial biogenesis upon SLP-2 over-

expression, the interaction of SLP-2 with the prohibitin complex was examined, as the

functional interactome of prohibitins has been shown to contain components of the

cardiolipin biosynthetic machinery 1. SLP-2 was found to associate with both PHB-1 and

PHB-2 and it was hypothesized that SLP-2 may recruit the prohibitin complex to the

mitochondrial membrane. Indeed, upon SLP-2 over-expression, there was a specific

increase in the association of PHB-1 with the mitochondrial membrane, while total

cellular expression levels of PHB-1 remained unchanged. Furthermore, it was found that

recombinant human SLP-2 bound to cardiolipin-enriched vesicles in a titratable fashion,

in which vesicles with higher cardiolipin content bound more SLP-2. Based on these

findings, it is proposed that SLP-2 may act as a bridge between cardiolipin in the

mitochondrial membrane and the prohibitin complex, recruiting prohibitins to facilitate

mitochondrial membrane compartmentalization.

The major function associated with the mitochondria is the energy production capacity of

the respiratory chain. As such, the effect of increased mitochondrial biogenesis upon

SLP-2 over-expression on respiratory chain function was examined. As expected, these

cells had higher enzymatic activity for both complex I and complex II as well as

increased ATP levels and oxygen consumption. This increased respiratory chain function

was due to an increase in activity but not expression levels of the respiratory chain

subunits. Finally, SLP-2 over-expressing cells were also less susceptible to the induction

of the intrinsic apoptotic pathway. Together, these results demonstrate that over-

expression of SLP-2 increases mitochondrial biogenesis, energy production and apoptosis

resistance, likely through the formation of cardiolipin-enriched microdomains.

166

5.1.2 SLP-2 deficiency in T cells is associated with abnormal mitochondrial membrane compartmentalization of cardiolipin, decreased mitochondrial respiration and impaired CD4+ T cell responses

To further investigate the role of SLP-2 in T cell activation, SLP-2 knockout mice were

generated and found to be embryonic lethal at the pre-implantation stage. To circumvent

this problem, conditional knockout mice were generated using the Cre-lox system

wherein SLP-2 was flanked by lox sites and bred with mice expressing Cre recombinase

under the control of the CD4 promoter. This resulted in T cell-specific conditional SLP-2

deletion at the double positive stage of T cell development with total loss of SLP-2

expression in mature T cells.

T cell-specific SLP-2 conditional knockout mice (SLP-2 T-K/O) showed normal T cell

development, with cell numbers in thymus, spleen and lymph nodes comparable to those

found in control mice. In addition, thymic subsets were also similar between control and

SLP-2 T-K/O mice, including double negative, double positive and CD4+ or CD8+ single

positive populations. As well, CD3+, CD4+, CD8+ and non-T cell populations were

similar between control and SLP-2 T-K/O mice in both the spleen and lymph nodes.

These results indicate that T cell development proceeds independently of SLP-2

expression.

It has been shown previously that SLP-2 expression levels correlate with T cell activation 2. Similarly, T cells from SLP-2 T-K/O mice had decreased activation in response to ex

vivo stimulation. To demonstrate the importance of this result in vivo, T cell recall

responses to primary immunization were examined in SLP-2 T-K/O mice. As expected

from ex vivo data, SLP-2 T-K/O mice showed significantly lower activation during

secondary exposure to antigen. As an additional in vivo model of SLP-2 function, a

transplant model was examined and control mice were found to reject non-matched

allografts an average of two days earlier than SLP-2 T-K/O mice, indicating that

decreased T cell activation upon SLP-2 deletion delays allograft rejection. This decrease

167

in T cell activation in SLP-2 T-K/O mice was shown to be a post-transcriptional block in

IL-2 production.

Examination of the effect of SLP-2 deletion on mitochondrial function showed that SLP-

2 T-K/O T cells had decreased protein levels of multiple subunits of complex I of the

respiratory chain while the remaining complexes were found in normal levels.

Furthermore, SLP-2 T-K/O T cells also had decreased activity of complex I as well as

complex II + III. Investigation of cardiolipin indicated that while SLP-2 T-K/O T cells

had normal total cellular levels of cardiolipin, there were decreased levels of cardiolipin

in detergent-insoluble microdomains and less recruitment of prohibitins to these domains.

To examine the effect of glucose starvation on SLP-2 T-K/O cells, T cells were

stimulated under conditions of excess or limiting glucose and responses were measured.

Under both excess and limiting glucose, SLP-2 T-K/O T cells had significantly lower

activation than control cells. Interestingly, under conditions of limiting glucose, control

cells had partial inhibition of T cell activation while SLP-2 T-K/O T cells were activated

to a similar extent under both glucose conditions. These results further demonstrate the

altered metabolic capacity of T cells lacking SLP-2 and point to the importance of

membrane organization for proper cellular functions.

5.1.3 Plasma membrane and mitochondrial stomatin-like protein 2 coalesce at the immunological synapse during T cell activation

To further investigate the subcellular localization and movement of SLP-2 in T cells,

SLP-2-GFP expressing Jurkat T cells were utilized. In addition, a Jurkat T cell line

stably expressing a doxycycline-inducible truncation mutant of SLP-2, lacking the amino

terminus and tagged at the carboxy terminus with GFP (ΔN-SLP-2-GFP) was also

generated. In comparison with full length SLP-2-GFP, which showed co-localization

with MitoTracker Red, ΔN-SLP-2-GFP showed diffuse cytosolic staining and no longer

co-localized with Mitotracker Red, confirming the presence of a mitochondrial targeting

domain at the amino-terminus of SLP-2 3. This divergent localization of full length and

168

truncated SLP-2 was confirmed by subcellular fractionation, in which SLP-2-GFP was

found in the mitochondrial fraction while ΔN-SLP-2-GFP was found only in the cytosol.

Surprisingly, a small pool of SLP-2 was also found in association with the plasma

membrane. While both SLP-2-GFP and ΔN-SLP-2-GFP were shown to associate with

the plasma membrane, full length SLP-2-GFP retained plasma membrane association at

lower expression levels compared to ΔN-SLP-2-GFP. This result indicates that while the

amino terminal region may play a role in membrane association, it is not essential.

It has been shown that mitochondria translocate to the immunological synapse upon T

cell activation and this translocation is required for optimal activation 4. In light of this

and given that SLP-2 is predominantly mitochondrial, the location of SLP-2-GFP during

T cell activation by superantigen stimulation was examined. In resting Jurkat T cells,

SLP-2-GFP distributed uniformly throughout the cell, surrounding the large nucleus.

Upon T cell activation, SLP-2-GFP re-localized to the immunological synapse and this

localization was actively maintained during T cell activation. Since the actin

cytoskeleton is required for mitochondrial translocation and SLP-2 has been shown

previously to associate with actin 2, 4, mitochondrial translocation in cells lacking SLP-2

was examined. The mitochondria in SLP-2 T-K/O T cells were found to redistribute to

the immunological synapse in a manner similar to that in wild type T cells, indicating that

mitochondrial trafficking during T cell activation proceeds independently of SLP-2.

To further investigate the surface pool of SLP-2, total internal reflection fluorescence

microscopy (TIRFM) was used to image 200 nm from the support plain, eliminating in

this way the mitochondrial pool of SLP-2. With this technique, the surface pool of SLP-2

was shown to be preferentially excluded from the cSMAC during T cell activation and

was found to surround TCR microclusters in the pSMAC. SLP-2 has been shown to

homo-oligomerize and it is possible that SLP-2 may form a large complex surrounding

the T cell signalosome, leading to increased association and activation of T cell signal

complexes. Furthermore, the presence of a small pool of the mitochondrial resident

protein SLP-2 at the plasma membrane may be indicative of membrane exchange

between the mitochondria and the cell surface, representing a novel mechanism for

protein trafficking in the cell.

169

5.2 Model of function: SLP-2 organizes membrane domains

SLP-2 is a member of the highly conserved stomatin family and therefore of the SPFH

superfamily, consisting of stomatins, prohibitins, flotillins and bacterial HflC/K proteins 5-7. This family of proteins is defined by the shared SPFH domain, a globular domain

composed of both a hydrophobic region and a hydrophilic region containing conserved

charged residues on the surface of the domain 8. These proteins are found in association

with either the plasma membrane or various intracellular membranes, either as integral or

peripheral membrane proteins, many of which are proposed to associate through

palmitoylation modifications 5, 9. The function of the SPFH domain is currently unclear,

although it has been linked to protein-lipid interactions such as those seen in association

with the plasma membrane 10. It is likely that a highly conserved domain such as the

SPFH domain would have a conservation of function. Indeed, studies of SPFH family

members have pointed to a general mechanism of action involving the organization of

membranes into defined domains to affect a wide variety of cellular functions depending

on the subcellular localization of these proteins 11-15. Plasma membrane-associated

flotillins have been shown to play a role in insulin signalling, phagocytosis, axon growth

and lymphocyte activation, while mitochondrial-associated prohibitins are involved in

cellular lifespan, mitochondrial morphology and assembly of the respiratory chain and

plasma membrane-associated stomatin has been proposed to be involved in ion channel

regulation 13, 16, 17. Based on the conserved mechanism of action across SPFH family

members and on the results presented here, we propose that SLP-2 organizes membranes

into specialized microdomains, thus affecting many mitochondrial and cellular functions.

The organization of the plasma membrane into specialized domains has long been

proposed to play an important role in various cellular processes, including signal

transduction and membrane trafficking 18. These domains, previously termed lipid rafts

but referred to here as detergent-insoluble glycolipid-enriched microdomains (DI-GEMs),

are highly ordered membrane domains enriched in sphingolipids, cholesterol and GPI-

anchored proteins 18-20. DI-GEMs are thought to exist in resting cells as nanoscale

assemblies, which coalesce in response to stimulation 18, 19. The coalescence of the

170

smaller assemblies into larger domains has been proposed to occur through lipid-lipid,

lipid-protein and protein-protein interactions. Although most studies of DI-GEMs

involve the plasma membrane, SLP-2 is associated predominantly with the mitochondrial

membrane, where there is a very limited level of cholesterol, the major component of

plasma membrane DI-GEMs. However, recent studies have indicated the possibility of

DI-GEM regions in the mitochondrial membrane 21-25. These studies have pointed to a

role for the mitochondrial-specific lipid cardiolipin in the formation of specialized

cardiolipin-enriched microdomains involved in induction of apoptosis, mitochondrial

fission and cristae formation 23, 25, 26. Our finding that SLP-2 binds cardiolipin may reveal

a key step in the formation of these microdomains in the mitochondria. Cardiolipin is a

lipid of unique structure that contains four acyl chains and has been found to bind directly

to a number of proteins, and this association is required for proper function of

components of the respiratory chain as well as the protein import machinery 27-29. As

such, the formation of cardiolipin-enriched microdomains not only segregates physically

mitochondrial proteins but also plays an important role in proper protein function.

Although previous work has pointed to the existence of cardiolipin-enriched

microdomains in the mitochondrial membrane, it is unclear how these regions arise. We

propose a model in which SLP-2 contributes to the formation of cardiolipin-enriched

microdomains through a direct association with cardiolipin and through interactions with

various proteins. We have presented here a number of findings to support a role for SLP-

2 in membrane organization. Similar to other SPFH family members, SLP-2 has been

found in DI-GEM fractions, specifically in T cells undergoing activation and as T cell

activation proceeds, SLP-2 is increasingly recruited to DI-GEM fractions, supporting a

role in membrane organization 2. In addition, we have found that recombinant SLP-2

binds directly to cardiolipin, in such a way that increased concentrations of cardiolipin

leads to increased SLP-2 binding. This is reminiscent of work showing the plasma

membrane-associated SPFH domain containing proteins Mec-2 and podocin binding

directly to cholesterol 10. Furthermore, we have found that SLP-2 alters the recruitment

of proteins to DI-GEM regions, as the over-expression of SLP-2 leads to an increased

recruitment of prohibitins to the mitochondrial membrane. This is similar to results

looking at stomatin-like protein 1 and podocin, both of which have been shown to control

171

localization of other proteins 15, 30, 31. Finally, the formation of DI-GEMs is altered in the

absence of SLP-2, as these fractions have significantly less cardiolipin content in mice

lacking SLP-2.

We also propose that the oligomerization of SLP-2 may play a role in membrane

organization. Similar to other SFPH family members, we have shown here that SLP-2 is

capable of undergoing homo-oligomerization 10, 32, 33. Oligomerization of SPFH proteins

has been proposed to play a role in protein stabilization, as loss of either prohibitin 1 or

prohibitin 2 prevents the formation of the large heteroligomeric prohibitin complex and

results in the loss of both prohibitin proteins 5, 34. However, we found that while the

truncation mutant of SLP-2 lacking the amino terminal region failed to oligomerize with

full length endogenous SLP-2, its expression was not lost. This indicates that

oligomerization of SLP-2 is not required for protein stability and instead we propose that

the oligomerization of SLP-2 into large complexes may facilitate the concentration of

cardiolipin into cardiolipin-enriched microdomains. Although cardiolipin only

constitutes approximately 10-15% of the total lipid content of the mitochondrial

membrane 14, the oligomerization of cardiolipin-bound SLP-2 proteins could alter the

cardiolipin distribution in the mitochondrial membrane and concentrate it into regions in

which the localized cardiolipin content is much higher than 15%. Furthermore, those

proteins bound to cardiolipin or with SLP-2, either directly or indirectly, would also be

concentrated into these regions, leading to the formation of large protein complexes (Fig.

5.1). The proteins recruited to these regions would in turn determine the function of the

domain. In this way, treatment with the inducer of apoptosis, anti-Fas could trigger the

formation of domains enriched in pro-apoptotic factors and fission proteins 23, 35.

172

Figure 5.1: A step-wise model of the formation of cardiolipin-enriched

microdomains in the mitochondrial membrane as facilitated by SLP-2.

(a) The mitochondrial membrane is composed of a mixture of lipids, including the

mitochondrial-specific lipid cardiolipin, which compromises approximately 10-15% of

the total mitochondrial lipids 36. (b) SLP-2 binds directly to cardiolipin and undergoes

homo-oligomerization (c) to concentrate cardiolipin into cardiolipin-enriched

microdomains. (d) Additional proteins are recruited to these domains through direct or

indirect association with either cardiolipin (respiratory chains components) or SLP-2

(prohibitin complex). Abreviations: CL – cardiolipin, SLP-2 – Stomatin-like protein 2,

PHB – prohibitin, C – complex of the respiratory chain, CI – NADH dehydrogenase, CII

– succinate dehydrogenase, CIII – cytochrome bc1 complex, CIV – cytochrome c

oxidase, CV – ATP synthase.

173

Figure 5.1

CL Other lipids

SLP-2

Mitochondrial membrane

Homo-oligomerization of SLP-2

PHB1 + PHB2

CI CII CII

I

CIV CV

CI CII CII

I

CIV CV

CI CII

CII

I CIV CV

CI CII

CII

I CIV CV

a

b

c

d

174

Figure 5.2: Model of supercomplex formation upon SLP-2 upregulation in response

to T cell activation.

SLP-2 is expressed at low levels in naive T cells but both SLP-2 and prohibitin 1 and 2

are upregulated upon T cell activation. Through self-oligomerization, prohibitin

association and the interaction with cardiolipin, SLP-2 organizes the mitochondrial

membrane into cardiolipin-enriched domains, which facilitates supercomplex formation,

leading to more efficient respiratory chain function and increased mitochondrial

biogenesis. Abreviations: CL – cardiolipin, SLP-2 – Stomatin-like protein 2, PHB –

prohibitin, C – complex of the respiratory chain, CI – NADH dehydrogenase, CII –

succinate dehydrogenase, CIII – cytochrome bc1 complex, CIV – cytochrome c oxidase,

CV – ATP synthase.

175

Figure 5.2

Top View of Mitochondrial Inner Membrane

Inner mitochondrial membrane

T cell activation

Inner mitochondrial membrane

CI

CIII

CII

CIV

CV

CI CIII

CII

CIV

CV

PHB1 PHB2

SLP-2 CL

Respiratory chain complex

176

5.2.1 Assembly of multi-domain receptors, including respiratory chain supercomplex formation

A major function of the mitochondria is the energy production capacity of the respiratory

chain. Given that the respiratory chain is composed of a large number of subunits

organized into five separate complexes, it is attractive to speculate a role for cardiolipin-

enriched domains to enhance the efficiency of the respiratory chain. This is supported by

the detection of respiratory chain components in the cardiolipin-enriched DI-GEMs of

mouse T cells. As electrons are transported through the respiratory chain, closer packing

of the complexes in the mitochondrial membrane would facilitate more efficient transfer

of electrons and reduce the possibility of energy loss. Indeed, it has been shown that

multiple components of the respiratory chain, including complexes I, III and IV are found

in association with each other, leading to the formation of supercomplexes 37. Given that

cardiolipin is known to be required for proper functioning of various components of the

respiratory chain and that supercomplex formation is inhibited in cells lacking cardiolipin 27, 28, 38-40, we propose that supercomplexes may be formed in cardiolipin-enriched

domains organized by SLP-2 and cardiolipin interactions (Fig. 5.2). In the absence of

SLP-2, cardiolipin-enriched DI-GEM domains may be less stable, causing supercomplex

dissociation and decreased respiratory chain efficiency. Supporting this model, studies

have shown a small decrease in transmembrane potential upon SLP-2 knockdown and we

have found a decrease in cardiolipin content in DI-GEMs along with decreased complex I

activity in SLP-2-deficient T cells 41, 42. As complex I is the largest of the respiratory

chain complexes, consisting of 45 subunits, and cardiolipin is known to be required for

complex I activity, it is likely that cardiolipin-enriched DI-GEMs play a role in the

assembly and/or the activity of complex I 29, 43. In addition, we and others have shown an

association between SLP-2 and the prohibitin complex and we have found increased

recruitment of prohibitins upon SLP-2 over-expression 3. This recruitment of the

prohibitin complex to cardiolipin-enriched microdomains may also play a role in the

assembly of complex I, as suggested by studies looking at the assembly of complex I 44,

45. We have also shown that cells over-expressing SLP-2 have increased ATP production

and oxygen consumption, although these cells do not express higher levels of the

respiratory chain components. Together, these results support a model in which

177

increased SLP-2 expression would lead to increased supercomplex formation, giving rise

to a better functioning respiratory chain.

In addition to increased ATP production, we propose that increased supercomplex

formation in response to SLP-2 over-expression also triggers an increase in mitochondrial

biogenesis. This response appears to rely on increased cardiolipin biosynthesis as

inhibition of cardiolipin synthase blocks mitochondrial biogenesis. It has been shown

that increases in matrix pH leads to increased cardiolipin biosynthesis 46. We propose

that enhanced cardiolipin-enriched domain formation upon SLP-2 over-expression

increases supercomplex formation, leading to more efficient respiratory chain function,

thus increasing the matrix pH and inducing cardiolipin biosynthesis. This, in turn, may

trigger nuclear transcription programs by an undefined mitochondria to nucleus signalling

pathway 47.

5.2.2 Calcium buffering

In addition to energy production, the mitochondria also play a major role in calcium

signalling as the transmembrane potential is a critical driving force to take calcium up

from the cytosol. SLP-2 has been shown to play a role in the calcium buffering capacity

of the mitochondria as cells treated with siRNA to deplete SLP-2 were found to have

decreased calcium uptake into the mitochondria and also to have an increased rate of

calcium efflux through the sodium/calcium exchanger 48. Treatment of these cells with

an inhibitor of the sodium/calcium exchanger equalized the rate of calcium efflux to that

of control cells, but SLP-2-depleted cells still had lower calcium uptake. This result

indicates that the loss of SLP-2 affects both the net calcium uptake as well as the loss of

calcium through the ion exchanger.

Our model of SLP-2 as an organizer of mitochondrial membranes could explain both the

ion channel-dependent and independent effects of SLP-2 depletion. As discussed above,

cardiolipin-enriched microdomains formed by association with SLP-2 would lead to more

efficient electron transport and a higher transmembrane potential, thus allowing an

178

increase in calcium uptake, which we have seen upon SLP-2 over-expression in Jurkat T

cells (C. Lemke and J. Madrenas, unpublished results). Furthermore, work by Da Cruz et

al has found a decrease in mitochondrial calcium efflux when SLP-2 is over-expressed in

HeLa cells, but no difference in the initial uptake 48. In the absence of SLP-2, the

formation of cardiolipin-enriched domains would be inhibited, leading to decreased

complex I activity, which could lead to a decrease in transmembrane potential. This, in

turn, would inhibit the net uptake of calcium into the mitochondria. Surprisingly, our

SLP-2-defecient T cells did not show a deficiency in mitochondrial calcium uptake, as

was expected from the work of Da Cruz et al, demonstrating decreased calcium uptake in

SLP-2-depleted HeLa cells 48. The discrepancy concerning the effect of SLP-2 on

mitochondrial calcium uptake could be due to differences in SLP-2 expression and over-

expression levels in mouse T cells, Jurkat T cells and HeLa cells. Both HeLa cells and

Jurkat T cells express high levels of endogenous SLP-2 and since the level of SLP-2

over-expression in the HeLa system was not reported, it is difficult to compare this

system to our Jurkat system. Perhaps our Jurkat T cells were over-expressing much

higher levels of SLP-2, leading to increased membrane compartmentalization and

respiratory chain activity, resulting in a higher transmembrane potential to drive in higher

levels of calcium. Primary mouse T cells express very low levels of SLP-2 and may rely

less heavily on SLP-2 membrane compartmentalization for calcium signalling compared

to HeLa cells. In this way, the loss of SLP-2 from the high expressing HeLa cells may

have a more prominent and detectable effect on mitochondrial calcium uptake compared

to the low SLP-2 expressing primary mouse T cells. These discrepancies also point to the

importance of examining the role of SLP-2 across different cell types, as it is likely that

different expression levels across tissues may alter the sensitivity to the loss of SLP-2 on

cell function.

Altered membrane organization could also explain the increased activity of the

sodium/calcium exchanger in Hela cells as DI-GEMs have been shown to play a role in

the regulation of a large number of ion channels 49-52. In this light, cardiolipin-enriched

microdomains formed by SLP-2 may play a role in the inhibition of calcium efflux from

the mitochondria, possibly through a direct inhibition of the ion channel by SLP-2 or

179

through an indirect mechanism in which an inhibitor of the channel is also recruited to

the cardiolipin-enriched microdomain.

5.2.3 Mitochondria membrane associated protease regulation

Early studies of the SPFH family members pointed to a role in the regulation of

proteases, as the loss of prohibitins was found to increase proteolysis of mitochondrial

proteins, reminiscent of the role of the bacterial protein HflC/K in the regulation of the

FtsH protease 7, 53. Subsequent work demonstrated that prohibitins bind directly to the

mitochondrial AAA-protease to negatively regulate its function and prevent proteolysis

of target proteins 53. Supporting this model of protease regulation, cells deficient in SLP-

2 due to treatment with targeted siRNA have decreased protein levels of prohibitins,

subunits of complex I and IV as well as altered processing of OPA-1 3, 54. Pointing to a

role for proteolysis over altered expression, treatment of SLP-2-deficient cells with a

metalloprotease inhibitor led to a partial recovery of protein levels, although the levels

did not return to those found in control cells 3. This result could indicate that both altered

expression and proteolysis may contribute to the decrease in protein levels or it could

point to wider variety of proteases acting to degrade these mitochondrial proteins in the

absence of SLP-2. As an alternative explanation for this degradation pattern, it is likely

that the organization of the mitochondrial membrane into defined domains could protect

proteins from proteolysis by excluding proteases, similar to the exclusion of the CD45

phosphatase from the immunological synapse to prevent premature termination of T cell

signalling 55. In this model, and as we have shown here, the loss of SLP-2 would lead to

altered distribution of cardiolipin into DI-GEMs. This in turn could prevent the physical

separation of the proteases from the target proteins, leading to the increase in protein

turnover. Furthermore, the partial recovery of protein levels upon metalloprotease

inhibition points to a role for a wider variety of proteases in protein turnover and the

physical segregation due to cardiolipin-enriched microdomains could protect proteins

from multiple proteases.

180

This regulation of protein turnover by physical segregation of target proteins from

proteases has implications for multiple mitochondrial functions, including energy

production as well as mitochondrial dynamics. Indeed, recently, SLP-2-deficient cells

have been shown to have an altered pattern of OPA-1 degradation, which in turn, inhibits

mitochondrial fusion in response to stress induction 54. In this study, cells were found to

undergo mitochondrial hyperfusion in response to stress induction by the treatment with

actinomycin D, cyclohexamide or UV and this response was protective against apoptotic

cell death. Knockdown of OPA-1, mitofusin1 or SLP-2 prevented hyperfusion and cells

showed increased cell death in response to stress induction. The induction of hyperfusion

also led to an increase in ATP production, indicating increased respiratory chain activity,

which may be partially explained by an increased formation of cardiolipin-enriched

domains, leading to enhanced supercomplex formation, as cells lacking SLP-2 failed to

increase ATP production. In agreement, we have found that SLP-2 over-expressing cells

are protected against apoptotic cell death, possibly through enhanced hyperfusion events

and increased ATP production.

5.2.4 Specialized membrane microdomain formation

In addition to the protection of proteins from proteolysis, individual cardiolipin-enriched

domains may recruit specific sets of proteins to alter various mitochondrial functions. As

discussed above, formation of domains enriched in respiratory chain components would

facilitate supercomplex assembly and lead to increased ATP production. Mitochondrial

fusion and hyperfusion may be facilitated by the formation of domains enriched in OPA-

1 and the recruitment of the mitofusins, as OPA-1 is found to constitutively associate

with cardiolipin-enriched domains and mitofusin 2 could be recruited through association

with SLP-2 23, 41. Similarly, these domains could also regulate mitochondrial fission as

hFis is constitutively associated with cardiolipin-enriched domains and DLP1 is recruited

in response to various treatments 23. In this case, the balance between fusion and fission

may be regulated by recruitment of factors to cardiolipin-enriched domains in response to

cellular signals. This has been demonstrated during induction of apoptosis, which

induces recruitment of pro-apoptotic factors tBid and Bax to cardiolipin-enriched

181

domains, indicating a role for cellular signalling in recruitment of proteins to these

domains 25.

5.2.5 T cell activation

Although the main pool of SLP-2 is found in association with mitochondrial membranes,

a small pool is also detected at the cell surface in T cells. While we initially hypothesized

that SLP-2 may be involved in mitochondrial translocation and maintenance at the

immunological synapse in response to stimulation, we found normal localization in SLP-

2-deficient cells. Based on this result, we propose that while the mechanism of the

surface pool and mitochondrial pool is likely conserved, it is probable that they act

independently to control unique cellular functions.

Immunosynapse formation plays an important role in regulating T cell activation upon

ligand binding. At early time points, up to 15 minutes after ligand binding, LFA-1 is

concentrated at the center of the contact site after which time TCR microclusters migrate

into the center of the synapse, surrounded by LFA-1, forming the cSMAC and pSMAC

regions respectively. By 60 minutes after binding, the TCR is no longer detected in the

cSMAC, which is likely required for down-regulation of T cell responsiveness 56. The

importance of the immunosynapse structure for T cell signalling has been show in mice

lacking CD2AP, which fail to form pSMAC and cSMAC structures 57. These cells show

altered responses to stimulation, with delayed ZAP-70 and ζ chain phosphorylation. This

delay is thought to occur because the formation of the immunosynapse is required to

optimize interactions between the TCR and MHC, which essential for proper TCR

signalling. For optimal TCR triggering, the TCR must be serially engaged by MHC

complexes. If ligand-binding affinity is too high, dissociation is prevented and there is no

serial triggering of TCR complexes, resulting in decreased TCR signalling. Likewise,

weak ligand binding also prevents TCR triggering. The immunological synapse is

proposed to promote TCR concentration at the interface with the APC, allowing

intermediate affinity ligands to activate T cells through rapid rebinding upon MHC

dissociation to strengthen T cell signalling responses.

182

Given the localization of the surface pool of SLP-2 surrounding TCR microclusters at the

interface with LFA-1, we propose that SLP-2 may strengthen the formation of the

immunological synapse by stabilizing the assembly of the TCR signalosome. As DI-

GEM domains coalesce during T cell activation, the plasma membrane-associated pool of

SLP-2 may be involved in facilitating and stabilizing these interactions, by self-

oligomerization as well as interactions with components of the T cell signalosome and

actin 2. This may result in the assembly of larger signalling complexes or may stabilize

the signalling complexes to allow stronger signalling responses. In addition, although not

shown experimentally, SLP-2 may interact with cholesterol to facilitate DI-GEM

clustering, as stomatin family members MEC-2 and podocin have both been shown to

interact with cholesterol through the shared SPFH domain 10. As SLP-2 levels increase,

the strength and duration of T cell signalosome association would also increase, leading

to increased T cell signalling and activation. Likewise, a loss of SLP-2 would result in

decreased stability of the DI-GEM domains in activated T cells, causing a dissociation of

signalling platforms, ultimately leading to impaired T cell responses. This model is

supported by results showing a decrease in T cell signalling upon SLP-2 depletion in

human T cells 2. Surprisingly, we did not see a loss of T cell signalling in SLP-2-

deficient murine T cells in response to a variety of stimulation conditions. As we have

not yet shown the plasma membrane-associated pool of SLP-2 to localize with TCR

microclusters in murine T cells, it is possible that surface-associated SLP-2 is not

involved in TCR signalosome formation in mice. Alternatively, this result could also

indicate that there is a redundancy in the function of SLP-2 in the formation or

stabilization of TCR microclusters in mice. We performed microarray analysis of both

naïve and stimulated SLP-2-deficient T cell but found no upregulation of any SLP-2-

related genes. However, it is possible that endogenous levels of other SPFH family

members could rescue TCR signalosome formation/stability in the absence of SLP-2.

Indeed, flotillins have been detected at the plasma membrane in T cells and previous

work in our lab has shown both stomatin and SLP-1 expression in Jurkat T cells (M.

Kirchhof and J. Madrenas, unpublished results) 11. It is possible that these family

members could facilitate TCR signalosome formation/stability in the absence of SLP-2

183

but since none are targeted to the mitochondria, these proteins are unable to rescue the

mitochondrial phenotype.

Although it is currently unclear how SLP-2 is localized to two cellular membranes or

why a small pool of a mitochondrial protein would be localized to the plasma membrane,

the localization of SLP-2 to the pSMAC of the immunological synapse indicates a role

for SLP-2 in T cell signalling and demonstrates that both pools of SLP-2 are likely

functional. Thus, at both the mitochondrial and plasma membrane, SLP-2 facilitates

membrane organization to regulate various cellular functions.

5.3 Future directions

5.3.1 Characterization of the structure of SLP-2

We have shown in this work that the deletion of the amino terminal region of SLP-2

prevents association with full length endogenous SLP-2. However, deletion mutants of

stomatin indicate that a short region at the C-terminus is required for homo-

oligomerization 32. Given the high degree of similarity between SLP-2 and the other

stomatin family members, it is likely that the region involved in oligomerization would

be conserved. An explanation for this discrepancy may involve the subcellular

localization of the SLP-2 truncation mutant. Since the mitochondrial target sequence is

located at the amino terminus and we have shown that loss of this region prevents

mitochondrial localization, it is likely that the loss of oligomerization between the

truncation mutant and endogenous full length SLP-2 is due to different subcellular

localization. Since endogenous SLP-2 is localized to the mitochondria and the truncation

mutant is cytosolic, it is unlikely that these proteins would form oligomers. In this way,

the work presented here has only touched on the role of the amino terminal region in

mitochondrial localization and the requirement for this localization to control

mitochondrial functions.

If our model of SLP-2 function is correct, proper functioning of SLP-2 will require

homo-oligomerization, as well as association with cardiolipin and with various

184

mitochondrial proteins. Each of these associations likely require distinct regions of the

SLP-2 protein, allowing SLP-2 to form homo-oligomers while also binding to cardiolipin

and other proteins, which would be required for the formation of cardiolipin-enriched

microdomains. A series of SLP-2 deletion constructs retaining mitochondrial targeting,

expressed in Jurkat T cells, could identify the regions required for homo-oligomerization

and for the association with known binding partners, such as the prohibitin complex.

Furthermore, since we know that SLP-2 over-expression increases mitochondrial

biogenesis and function, the link between loss of physical associations and SLP-2

function could be measured. The association with cardiolipin could be investigated by

generating recombinant proteins with point mutations to identify residues or regions

required for cardiolipin binding.

Information regarding the structure of the SPFH domain is essential to better understand

its functional role in membrane compartmentalization across all family members.

Currently, there is no crystal structure available for any member of the SPFH family.

However, we have generated recombinant human SLP-2, which could be used to generate

the crystal structure of SLP-2. This would provide a three-dimentional model of SLP-2

and could provide insight into the physical interaction with its binding partners as well as

information concerning the formation of microdomains in mitochondrial membranes.

This would represent the first crystal structure for a full length SFPH family member, as

only the SPFH domain has been crystalized thus far 8, 58.

5.3.2 Characterization of cardiolipin-enriched microdomains

Cardiolipin-enriched microdomains in the mitochondrial membrane have been isolated as

detergent-insoluble fractions and are thought to play a role in apoptosis induction 23, 25.

We have proposed here, that similar to other SPFH family members, SLP-2 plays a role

in membrane organization by facilitating the formation of these cardiolipin-enriched

microdomains 11, 14. However, while we have shown an interaction between SLP-2 and

cardiolipin, we have not directly shown the formation of cardiolipin domains and have

instead relied on detergent-insolubility to define these regions. As such, more work is

required to directly visualize cardiolipin-enriched regions within mitochondrial

185

membranes and to visualize the distribution of cardiolipin in the presence and absence of

SLP-2. This has proved to be difficult thus far due to the limitations of confocal

microscopy as it is difficult to image regions within the mitochondrial membrane.

Electron microscopy would provide excellent resolution of the mitochondrial membrane

structure and immunogold labeling could provide information on the distribution of SLP-

2 within the mitochondrial membrane, as well as the localization of SLP-2 binding

partners in the presence and absence of SLP-2. However, using this method to image

cardiolipin would be more complex and would require the use of a purified, specific

antibody against cardiolipin, which is not commercially available at this time.

Alternatively, cryo-electron microscopy and tomography could be used to obtain high-

resolution structures of the mitochondrial membrane 26, 59, 60. In this way, mitochondrial

membranes could be isolated and imaged to identify cardiolipin-enriched and protein-

enriched domains.

Although we have found prohibitins and components of the respiratory chain in

association with cardiolipin-enriched microdomains and others have reported the

association of pro-apoptotic proteins and proteins involved in mitochondrial dynamics 22,

25, there has not been a large-scale analysis of proteins localized to cardiolipin-enriched

microdomains. To this end, these regions could be isolated on the basis of detergent-

insolubility and subjected to mass spectrometry analysis. The proteins identified could

be organized into functional groups, such as respiratory chain components, components

of the protein import machinery, mitochondrial dynamics, etc. In this way, we could

identify those mitochondrial functions that might rely on cardiolipin-enriched

microdomain formation. This could also provide more insight into the function of SLP-2

in the formation of cardiolipin-enriched microdomains, as it is possible that some

mitochondrial functions may rely on SLP-2 for microdomain formation while others may

rely on microdomains formed independently of SLP-2. To investigate the requirement of

SLP-2, we could look at the targeting of the various proteins to cardiolipin-enriched

microdomains in the presence and absence of SLP-2.

186

5.3.3 Intracellular trafficking of SLP-2

While we have shown here that SLP-2 is found in two intracellular pools, at both the

mitochondrial and plasma membranes, it remains to be shown how this pattern of

localization arises. As demonstrated here, SLP-2 contains an amino-terminal

mitochondrial targeting sequence, which is required for mitochondrial localization 3, 41.

How SLP-2 traffics to the plasma membrane is currently unclear. It is possible that a

small portion of newly synthesized SLP-2 escapes mitochondrial trafficking and is

diverted to the plasma membrane by the default pathway of protein segregation 61.

However, an alternative explanation arises from the discovery of mitochondrial vesicles,

which may play a role in membrane exchange between the mitochondrial membrane and

other cellular membranes 62. In this way, it is possible that all newly synthesized SLP-2

is transported into the mitochondria, but the association with the mitochondrial

membrane allows SLP-2 to be transported to the plasma membrane in mitochondrial

vesicles, either by the passive loading of a mitochondrial protein or by an unidentified

pathway to target mitochondrial proteins for exchange. Given that membrane

organization is involved in vesicular transport, it is plausible that the organization of the

mitochondrial membrane into cardiolipin-enriched microdomains could facilitate

mitochondrial vesicle formation 18. Since work has shown that newly synthesized SLP-2

is cleaved upon transport into mitochondria 41, the amino acid sequence of plasma

membrane SLP-2 could provide insight into the cellular trafficking pathway taken by

SLP-2. To perform this analysis, SLP-2 could be isolated from both the mitochondria

and plasma membrane and subjected to mass spectrometry analysis to determine protein

sequence. If the plasma membrane pool is found to contain the full SLP-2 sequence,

while the mitochondrial pool is lacking the amino-terminal region, it is likely that the

plasma membrane pool traffics directly. However, if both mitochondrial- and plasma

membrane-associated SLP-2 are lacking the amino-terminal region, it is likely that the

plasma membrane pool arises from membrane exchange with the mitochondria. If SLP-2

does traffic through the mitochondria, this could point to a new paradigm in protein

trafficking, one that does not necessarily depend on a single targeting domain resulting in

a single intracellular localization. Instead, membrane exchange between various

187

organelles and the plasma membrane could result in protein trafficking independent of

targeting signalling domains encoded in the protein.

5.3.4 Functional role of SLP-2 in cell physiology

Although we have focused primarily on the role played by SLP-2 in T cells, expression of

SLP-2 is not restricted to T cells, but is widely expressed, being detected in such tissues

as brain, muscle and heart, among others 63. Indeed, as SLP-2 is a mitochondrial protein,

it is expressed across many tissues, especially those with high energy demand and the

loss of SLP-2 could lead to a wide variety of potential phenotypes, depending on the

affected tissue and its dependence on the mitochondria, as discussed below. In addition,

although mitochondria play an important role in T cell differentiation, activation of these

cells relies primarily on glycolysis 64-66. As such, it is not surprising that the deletion of

SLP-2 in T cells leads to a very subtle phenotypic effect. However, it is likely that

tissues relying more heavily on the energy production capacity of the mitochondria would

show a more prominent phenotype upon deletion of SLP-2. To this end, tamoxifen-

inducible SLP-2 knockout mice have been generated, in which treatment of mice with

tamoxifen results in widespread deletion of SLP-2 across all tissues. This model will

allow for the study of the effects of SLP-2 deletion on multiple tissues, including high

energy demanding tissues such as muscle and brain. It is likely that the loss of SLP-2 and

the subsequent depression of respiratory chain activity will impact the function of these

tissues, leading to a more prominent phenotype than that seen with T cell-specific

deletion. This model could point out tissues where SLP-2 plays a more essential

functional role, allowing more thorough investigation of SLP-2 function in these tissues.

A tamoxifen-inducible SLP-2 knockout model will also allow for the deletion of SLP-2 at

various time points during development, for further investigation of the role of SLP-2

during embryogenesis. As SLP-2 has been proposed to play a role in mitochondrial

dynamics and given that the deletion of multiple components of the fusion and fission

machinery are also embryonic lethal, the tamoxifen-inducible model could provide a

system to further study the role of mitochondrial dynamics during embryogenesis 54, 67.

188

5.4 SLP-2 and disease

5.4.1 SLP-2 and mitochondrial disorders

Mitochondria carry out many essential functions, primarily the generation of energy

required to power cellular functions. Given the requirement for mitochondria across all

tissues, it follows that dysfunctions of the mitochondria would have wide reaching

effects. Indeed, mitochondrial dysfunction has been implicated in a wide variety of

diseases, including cardiovascular disease, diabetes, gastrointestinal disease, neurological

diseases such as Alzheimer’s disease and Parkinson’s disease as well as cancer 68, 69.

Mitochondrial dysfunction is also the leading cause of inborn metabolic disorders 70.

This group of disorders is widely variable across patients, with differing symptoms and

levels of severity 71. This variability arises due to the complexity of the respiratory chain

and the large number of genes required for proper function. A large number of studies

have been performed on patients with a variety of mitochondrial disorders and many

inherited mutations have been identified, in both nuclear and mitochondrial DNA 71, 72.

The most common form of mitochondrial disorder in infants is Leigh syndrome, in which

patients tend to have normal pre-natal development, but develop symptoms during the

first year, with most dying before 3 years of age 73, 74. These patients generally exhibit

central and peripheral nervous system abnormalities, including optic atrophy,

opthalmoparesis, hypotonia, ataxia and dystonia 71, 74. While causative mutations have

been identified in all respiratory chain complexes, the largest number of mutations have

been identified within complex I genes, which is to be expected, as this complex requires

the correct expression and assembly of 45 subunits 73.

Complex I disorders also present as a spectrum of disorders with varying severity, in

some cases affecting isolated tissues while in other cases affecting multiple systems 70.

As expected, a number of genetic mutations have been identified as causative for this

group of disorders, including mutations in all of the subunits composing the core of

complex I 74. However, the essential role of complex I assembly has also been found to

play a role in mitochondrial disorders, as mutations in accessory subunits have also been

189

shown to be causative for complex I disorders 70, 73. Given the proposed role for SLP-2 in

the assembly of complex I, further study of SLP-2 and cardiolipin-enriched domains may

provide insight into regulation of complex I assembly, which would in turn provide

insight into the role of complex I assembly and mitochondrial disorders.

Another group of mitochondrial disorders involves mutations in proteins involved in

mitochondrial dynamics. The mutation of mitofusin 2 and OPA-1 both cause neural

disorders, characterized by impairment of motor and sensory neurons or the optic nerve,

respectively 75. Furthermore, altered regulation of mitochondrial fusion and fission have

been found in a number of neurological disorders, including Alzheimer’s disease,

Parkinson’s disease and Huntington’s disease 76. Given that SLP-2 has been shown to

play a role in mitochondrial hyperfusion in response to cellular stress, possibly by

regulating the cleavage of OPA-1, a further understanding of the role for SLP-2 in

mitochondrial dynamics could provide insight into these neurological diseases.

5.4.2 SLP-2 in cancer prognosis and metastasis

The role of SLP-2 in membrane organization has effects on multiple cellular functions,

which when deregulated may have health implications. Indeed, a number of studies have

found an upregulation of SLP-2 expression in multiple types of cancer, including

esophageal squamous cell carcinoma, lung cancer, laryngeal cancer, endometrial

adenocarcinoma, breast cancer, laryngeal squamous cell carcinoma, osteosarcoma and

pulmonary squamous cell carcinoma 77-81. Furthermore, SLP-2 expression has been

shown to positively correlate with tumor size, lymph node metastasis, clinical stage,

distant metastasis and decreased overall survival and disease-free survival 78, 79, 81. To

investigate possible mechanisms of SLP-2 in cancer progression, SLP-2 was knocked

down in esophageal squamous cell carcinoma cells, which led to decreased cell growth

due to S-phase arrest with no change in apoptosis 77. When these cells were injected into

nude mice, they showed slower tumor progression and smaller tumors compared to mice

injected with the parental cells. Upon treatment with chemotherapeutic agents, the SLP-

2-depleted cells showed increased apoptosis compared to parental cells 42. Finally, SLP-

190

2-depleted cells were also reported to have slightly decreased transmembrane potential

and ATP levels. The reported changes were very small and likely did not influence the

energy requirements of the cells as cancer cells are known to rely more heavily on

glycolysis for ATP production 82. However, these changes may reflect the ability of SLP-

2 to induce hyperfusion upon stress induction, which may decrease the apoptotic

response of the SLP-2 expressing cancer cells in response to chemotherapeutics. The loss

of SLP-2 expression by siRNA depletion also led to decreased cell motility, both in the

migration of cells in a wound healing model and in cell migration across a transwell 42.

Given that the expression of SLP-2 is positively correlated with lymph node and distant

metastasis in multiple types of cancer 78, 79, 81, it is likely that SLP-2 may play a direct role

in metastasis by regulating the migration of cancer cells. Indeed, we have found SLP-2

in association with the adhesion factor LFA-1, which may point to a role in the regulation

of both cell adhesion and migration. Together, these results indicate that proper

regulation of SLP-2 expression may have an important role in cancer progression and

treatment.

5.5 Conclusions

Although SLP-2 is a widely expressed mitochondrial protein belonging to a highly

conserved protein family, very little work has been done to study this protein and the

mechanism of action has remained unclear. The work presented in this thesis provides

novel insight into the role of SLP-2 in T cells and points to a mechanism for SLP-2 in the

organization of membranes into functional domains.

We initially confirmed the mitochondrial localization of SLP-2 in Chapter 2 by

constructing stably transfected Jurkat T cell expressing SLP-2-GFP. The co-localization

of SLP-2-GFP with MitoTracker Red demonstrated the mitochondrial localization of

SLP-2. In addition, the over-expression of this SLP-2-GFP construct led to the finding

that increased expression of SLP-2 caused an increase in mitochondrial biogenesis, along

with an increase in activity of the respiratory chain and protection against apoptosis.

Providing evidence for the mechanism of action, we also showed that SLP-2 can bind

191

directly to cardiolipin as well as to the prohibitin complex and over-expression of SLP-2

increased the recruitment of the prohibitin complex to the mitochondrial membrane. We

next constructed T cell-specific SLP-2 conditional knockout mice, as described in

Chapter 3, and found that while SLP-2 was not required for T cell development, it was

required for optimal T cell activation. Importantly, we have shown that the decreased T

cell activation upon in vitro stimulation is biologically relevant, as our SLP-2 T-K/O

mice show a decreased rate of non-matched transplant rejection. Furthermore, this model

also provided additional evidence for the mechanism of action for SLP-2 in that T cells

lacking SLP-2 showed decreased levels of complex I subunits as well as lower activity of

complex I. Additionally, these cells also had lower levels of cardiolipin recruited to

detergent-insoluble microdomains within the mitochondrial membrane. Finally, in

Chapter 4 we have identified a second subcellular localization of SLP-2. In addition to

the mitochondrial localization, a small pool of SLP-2 was found at the plasma membrane.

We have shown that this pool was recruited to the pSMAC of the immunological synapse

in response to T cell stimulation, pointing to a role for the surface pool in T cell

activation. Furthermore, we have also shown the ability of SLP-2 to homo-oligomerize,

providing further evidence for our proposed model of SLP-2 function.

Based on these results, we propose that SLP-2 exerts effects on mitochondrial and

cellular functions by facilitating the organization of membranes into functional

microdomains. At the mitochondrial membrane, this is likely carried out in the following

steps: i) SLP-2 binds directly to cardiolipin, ii) SLP-2 undergoes homo-oligomerization

to concentrate cardiolipin into cardiolipin-enriched region, iii) SLP-2 and cardiolipin bind

to other proteins, recruiting them to cardiolipin-enriched domains to define domain

function. This model may be extended to the plasma membrane pool of SLP-2, if SLP-2

is capable of binding to cholesterol in a manner similar to its binding to cardiolipin. In

this way, SLP-2 facilitates membrane microdomain formation to effect such

mitochondrial and cellular functions as energy production, apoptotic induction and T cell

activation.

192

5.6 References

1. Osman, C., Haag, M., Potting, C., Rodenfels, J., Dip, P.V., Wieland, F.T., Brugger, B., Westermann, B. & Langer, T. The genetic interactome of prohibitins: coordinated control of cardiolipin and phosphatidylethanolamine by conserved regulators in mitochondria. J Cell Biol 184, 583-596 (2009).

2. Kirchhof, M.G., Chau, L.A., Lemke, C.D., Vardhana, S., Darlington, P.J., Marquez, M.E., Taylor, R., Rizkalla, K., Blanca, I., Dustin, M.L. & Madrenas, J. Modulation of T cell activation by stomatin-like protein 2. J Immunol 181, 1927-1936 (2008).

3. Da Cruz, S., Parone, P.A., Gonzalo, P., Bienvenut, W.V., Tondera, D., Jourdain, A., Quadroni, M. & Martinou, J.C. SLP-2 interacts with prohibitins in the mitochondrial inner membrane and contributes to their stability. Biochim Biophys Acta 1783, 904-911 (2008).

4. Quintana, A., Schwindling, C., Wenning, A.S., Becherer, U., Rettig, J., Schwarz, E.C. & Hoth, M. T cell activation requires mitochondrial translocation to the immunological synapse. Proc Natl Acad Sci U S A 104, 14418-14423 (2007).

5. Browman, D.T., Hoegg, M.B. & Robbins, S.M. The SPFH domain-containing proteins: more than lipid raft markers. Trends Cell Biol 17, 394-402 (2007).

6. Green, J.B. & Young, J.P. Slipins: ancient origin, duplication and diversification of the stomatin protein family. BMC Evol Biol 8, 44 (2008).

7. Tavernarakis, N., Driscoll, M. & Kyrpides, N.C. The SPFH domain: implicated in regulating targeted protein turnover in stomatins and other membrane-associated proteins. Trends Biochem Sci 24, 425-427 (1999).

8. Kuwahara, Y., Unzai, S., Nagata, T., Hiroaki, Y., Yokoyama, H., Matsui, I., Ikegami, T., Fujiyoshi, Y. & Hiroaki, H. Unusual thermal disassembly of the SPFH domain oligomer from Pyrococcus horikoshii. Biophys J 97, 2034-2043 (2009).

9. Snyers, L., Umlauf, E. & Prohaska, R. Cysteine 29 is the major palmitoylation site on stomatin. FEBS Lett 449, 101-104 (1999).

10. Huber, T.B., Schermer, B., Muller, R.U., Hohne, M., Bartram, M., Calixto, A., Hagmann, H., Reinhardt, C., Koos, F., Kunzelmann, K., Shirokova, E., Krautwurst, D., Harteneck, C., Simons, M., Pavenstadt, H., Kerjaschki, D., Thiele, C., Walz, G., Chalfie, M. & Benzing, T. Podocin and MEC-2 bind cholesterol to regulate the activity of associated ion channels. Proc Natl Acad Sci U S A 103, 17079-17086 (2006).

193

11. Langhorst, M.F., Reuter, A. & Stuermer, C.A. Scaffolding microdomains and beyond: the function of reggie/flotillin proteins. Cell Mol Life Sci 62, 2228-2240 (2005).

12. Morrow, I.C. & Parton, R.G. Flotillins and the PHB domain protein family: rafts, worms and anaesthetics. Traffic 6, 725-740 (2005).

13. Stuermer, C.A. Microdomain-forming proteins and the role of the reggies/flotillins during axon regeneration in zebrafish. Biochim Biophys Acta 1812, 415-422 (2011).

14. Osman, C., Merkwirth, C. & Langer, T. Prohibitins and the functional compartmentalization of mitochondrial membranes. J Cell Sci 122, 3823-3830 (2009).

15. Huber, T.B., Simons, M., Hartleben, B., Sernetz, L., Schmidts, M., Gundlach, E., Saleem, M.A., Walz, G. & Benzing, T. Molecular basis of the functional podocin-nephrin complex: mutations in the NPHS2 gene disrupt nephrin targeting to lipid raft microdomains. Hum Mol Genet 12, 3397-3405 (2003).

16. Artal-Sanz, M. & Tavernarakis, N. Prohibitin and mitochondrial biology. Trends Endocrinol Metab 20, 394-401 (2009).

17. Goodman, M.B., Ernstrom, G.G., Chelur, D.S., O'Hagan, R., Yao, C.A. & Chalfie, M. MEC-2 regulates C. elegans DEG/ENaC channels needed for mechanosensation. Nature 415, 1039-1042 (2002).

18. Simons, K. & Gerl, M.J. Revitalizing membrane rafts: new tools and insights. Nat Rev Mol Cell Biol 11, 688-699 (2010).

19. Lingwood, D. & Simons, K. Lipid rafts as a membrane-organizing principle. Science 327, 46-50 (2010).

20. Leslie, M. Mysteries of the cell. Do lipid rafts exist? Science 334, 1046-1047 (2011).

21. Epand, R.F., Tokarska-Schlattner, M., Schlattner, U., Wallimann, T. & Epand, R.M. Cardiolipin clusters and membrane domain formation induced by mitochondrial proteins. J Mol Biol 365, 968-980 (2007).

22. Sorice, M., Manganelli, V., Matarrese, P., Tinari, A., Misasi, R., Malorni, W. & Garofalo, T. Cardiolipin-enriched raft-like microdomains are essential activating platforms for apoptotic signals on mitochondria. FEBS Lett 583, 2447-2450 (2009).

23. Ciarlo, L., Manganelli, V., Garofalo, T., Matarrese, P., Tinari, A., Misasi, R., Malorni, W. & Sorice, M. Association of fission proteins with mitochondrial raft-like domains. Cell Death Differ 17, 1047-1058 (2010).

194

24. Garofalo, T., Tinari, A., Matarrese, P., Giammarioli, A.M., Manganelli, V., Ciarlo, L., Misasi, R., Sorice, M. & Malorni, W. Do mitochondria act as "cargo boats" in the journey of GD3 to the nucleus during apoptosis? FEBS Lett 581, 3899-3903 (2007).

25. Garofalo, T., Giammarioli, A.M., Misasi, R., Tinari, A., Manganelli, V., Gambardella, L., Pavan, A., Malorni, W. & Sorice, M. Lipid microdomains contribute to apoptosis-associated modifications of mitochondria in T cells. Cell Death Differ 12, 1378-1389 (2005).

26. Acehan, D., Malhotra, A., Xu, Y., Ren, M., Stokes, D.L. & Schlame, M. Cardiolipin affects the supramolecular organization of ATP synthase in mitochondria. Biophys J 100, 2184-2192 (2011).

27. Schlame, M., Rua, D. & Greenberg, M.L. The biosynthesis and functional role of cardiolipin. Prog Lipid Res 39, 257-288 (2000).

28. Joshi, A.S., Zhou, J., Gohil, V.M., Chen, S. & Greenberg, M.L. Cellular functions of cardiolipin in yeast. Biochim Biophys Acta 1793, 212-218 (2008).

29. Fry, M. & Green, D.E. Cardiolipin requirement for electron transfer in complex I and III of the mitochondrial respiratory chain. J Biol Chem 256, 1874-1880 (1981).

30. Mairhofer, M., Steiner, M., Salzer, U. & Prohaska, R. Stomatin-like protein-1 interacts with stomatin and is targeted to late endosomes. J Biol Chem 284, 29218-29229 (2009).

31. Sedensky, M.M., Siefker, J.M., Koh, J.Y., Miller, D.M., 3rd & Morgan, P.G. A stomatin and a degenerin interact in lipid rafts of the nervous system of Caenorhabditis elegans. Am J Physiol Cell Physiol 287, C468-474 (2004).

32. Umlauf, E., Mairhofer, M. & Prohaska, R. Characterization of the stomatin domain involved in homo-oligomerization and lipid raft association. J Biol Chem 281, 23349-23356 (2006).

33. Tatsuta, T., Model, K. & Langer, T. Formation of membrane-bound ring complexes by prohibitins in mitochondria. Mol Biol Cell 16, 248-259 (2005).

34. Berger, K.H. & Yaffe, M.P. Prohibitin family members interact genetically with mitochondrial inheritance components in Saccharomyces cerevisiae. Mol Cell Biol 18, 4043-4052 (1998).

35. Sorice, M., Matarrese, P., Tinari, A., Giammarioli, A.M., Garofalo, T., Manganelli, V., Ciarlo, L., Gambardella, L., Maccari, G., Botta, M., Misasi, R. & Malorni, W. Raft component GD3 associates with tubulin following CD95/Fas ligation. FASEB J 23, 3298-3308 (2009).

195

36. Osman, C., Voelker, D.R. & Langer, T. Making heads or tails of phospholipids in mitochondria. J Cell Biol 192, 7-16 (2011).

37. Acin-Perez, R., Fernandez-Silva, P., Peleato, M.L., Perez-Martos, A. & Enriquez, J.A. Respiratory active mitochondrial supercomplexes. Mol Cell 32, 529-539 (2008).

38. Choi, S.Y., Gonzalvez, F., Jenkins, G.M., Slomianny, C., Chretien, D., Arnoult, D., Petit, P.X. & Frohman, M.A. Cardiolipin deficiency releases cytochrome c from the inner mitochondrial membrane and accelerates stimuli-elicited apoptosis. Cell Death Differ 14, 597-606 (2007).

39. Zhang, M., Mileykovskaya, E. & Dowhan, W. Gluing the respiratory chain together. Cardiolipin is required for supercomplex formation in the inner mitochondrial membrane. J Biol Chem 277, 43553-43556 (2002).

40. Zhang, M., Mileykovskaya, E. & Dowhan, W. Cardiolipin is essential for organization of complexes III and IV into a supercomplex in intact yeast mitochondria. J Biol Chem 280, 29403-29408 (2005).

41. Hajek, P., Chomyn, A. & Attardi, G. Identification of a novel mitochondrial complex containing mitofusin 2 and stomatin-like protein 2. J Biol Chem 282, 5670-5681 (2007).

42. Wang, Y., Cao, W., Yu, Z. & Liu, Z. Downregulation of a mitochondria associated protein SLP-2 inhibits tumor cell motility, proliferation and enhances cell sensitivity to chemotherapeutic reagents. Cancer Biol Ther 8, 1651-1658 (2009).

43. McKenzie, M. & Ryan, M.T. Assembly factors of human mitochondrial complex I and their defects in disease. IUBMB Life 62, 497-502 (2010).

44. Marques, I., Dencher, N.A., Videira, A. & Krause, F. Supramolecular organization of the respiratory chain in Neurospora crassa mitochondria. Eukaryot Cell 6, 2391-2405 (2007).

45. Bourges, I., Ramus, C., Mousson de Camaret, B., Beugnot, R., Remacle, C., Cardol, P., Hofhaus, G. & Issartel, J.P. Structural organization of mitochondrial human complex I: role of the ND4 and ND5 mitochondria-encoded subunits and interaction with prohibitin. Biochem J 383, 491-499 (2004).

46. Gohil, V.M., Hayes, P., Matsuyama, S., Schagger, H., Schlame, M. & Greenberg, M.L. Cardiolipin biosynthesis and mitochondrial respiratory chain function are interdependent. J Biol Chem 279, 42612-42618 (2004).

47. Scarpulla, R.C. Transcriptional paradigms in mammalian mitochondrial biogenesis and function. Physiol Rev 88, 611-638 (2008).

196

48. Da Cruz, S., De Marchi, U., Frieden, M., Parone, P.A., Martinou, J.C. & Demaurex, N. SLP-2 negatively modulates mitochondrial sodium-calcium exchange. Cell Calcium 47, 11-18 (2010).

49. Dart, C. Lipid microdomains and the regulation of ion channel function. J Physiol 588, 3169-3178 (2010).

50. Levitan, I., Fang, Y., Rosenhouse-Dantsker, A. & Romanenko, V. Cholesterol and ion channels. Subcell Biochem 51, 509-549 (2010).

51. Pani, B. & Singh, B.B. Lipid rafts/caveolae as microdomains of calcium signaling. Cell Calcium 45, 625-633 (2009).

52. Pristera, A. & Okuse, K. Building Excitable Membranes: Lipid Rafts and Multiple Controls on Trafficking of Electrogenic Molecules. Neuroscientist Epub ahead of print (2011).

53. Steglich, G., Neupert, W. & Langer, T. Prohibitins regulate membrane protein degradation by the m-AAA protease in mitochondria. Mol Cell Biol 19, 3435-3442 (1999).

54. Tondera, D., Grandemange, S., Jourdain, A., Karbowski, M., Mattenberger, Y., Herzig, S., Da Cruz, S., Clerc, P., Raschke, I., Merkwirth, C., Ehses, S., Krause, F., Chan, D.C., Alexander, C., Bauer, C., Youle, R., Langer, T. & Martinou, J.C. SLP-2 is required for stress-induced mitochondrial hyperfusion. EMBO J 28, 1589-1600 (2009).

55. Fooksman, D.R., Vardhana, S., Vasiliver-Shamis, G., Liese, J., Blair, D.A., Waite, J., Sacristan, C., Victora, G.D., Zanin-Zhorov, A. & Dustin, M.L. Functional anatomy of T cell activation and synapse formation. Annu Rev Immunol 28, 79-105 (2010).

56. Lee, K.H., Holdorf, A.D., Dustin, M.L., Chan, A.C., Allen, P.M. & Shaw, A.S. T cell receptor signaling precedes immunological synapse formation. Science 295, 1539-1542 (2002).

57. Lee, K.H., Dinner, A.R., Tu, C., Campi, G., Raychaudhuri, S., Varma, R., Sims, T.N., Burack, W.R., Wu, H., Wang, J., Kanagawa, O., Markiewicz, M., Allen, P.M., Dustin, M.L., Chakraborty, A.K. & Shaw, A.S. The immunological synapse balances T cell receptor signaling and degradation. Science 302, 1218-1222 (2003).

58. Yokoyama, H., Fujii, S. & Matsui, I. Crystal structure of a core domain of stomatin from Pyrococcus horikoshii Illustrates a novel trimeric and coiled-coil fold. J Mol Biol 376, 868-878 (2008).

197

59. Dudkina, N.V., Kouril, R., Bultema, J.B. & Boekema, E.J. Imaging of organelles by electron microscopy reveals protein-protein interactions in mitochondria and chloroplasts. FEBS letters 584, 2510-2515 (2010).

60. Fujiyoshi, Y. & Unwin, N. Electron crystallography of proteins in membranes. Curr Opin Struct Biol 18, 587-592 (2008).

61. Burgess, T.L. & Kelly, R.B. Constitutive and regulated secretion of proteins. Annu Rev Cell Biol 3, 243-293 (1987).

62. Andrade-Navarro, M.A., Sanchez-Pulido, L. & McBride, H.M. Mitochondrial vesicles: an ancient process providing new links to peroxisomes. Curr Opin Cell Biol 21, 560-567 (2009).

63. Wang, Y. & Morrow, J.S. Identification and characterization of human SLP-2, a novel homologue of stomatin (band 7.2b) present in erythrocytes and other tissues. J Biol Chem 275, 8062-8071 (2000).

64. Pearce, E.L., Walsh, M.C., Cejas, P.J., Harms, G.M., Shen, H., Wang, L.S., Jones, R.G. & Choi, Y. Enhancing CD8 T-cell memory by modulating fatty acid metabolism. Nature 460, 103-107 (2009).

65. Michalek, R.D., Gerriets, V.A., Jacobs, S.R., Macintyre, A.N., MacIver, N.J., Mason, E.F., Sullivan, S.A., Nichols, A.G. & Rathmell, J.C. Cutting edge: distinct glycolytic and lipid oxidative metabolic programs are essential for effector and regulatory CD4+ T cell subsets. J Immunol 186, 3299-3303 (2011).

66. Frauwirth, K.A., Riley, J.L., Harris, M.H., Parry, R.V., Rathmell, J.C., Plas, D.R., Elstrom, R.L., June, C.H. & Thompson, C.B. The CD28 signaling pathway regulates glucose metabolism. Immunity 16, 769-777 (2002).

67. Chen, H. & Chan, D.C. Emerging functions of mammalian mitochondrial fusion and fission. Hum Mol Genet 14 Spec No. 2, R283-289 (2005).

68. Fosslien, E. Mitochondrial medicine--molecular pathology of defective oxidative phosphorylation. Ann Clin Lab Sci 31, 25-67 (2001).

69. Brandon, M., Baldi, P. & Wallace, D.C. Mitochondrial mutations in cancer. Oncogene 25, 4647-4662 (2006).

70. Lazarou, M., Thorburn, D.R., Ryan, M.T. & McKenzie, M. Assembly of mitochondrial complex I and defects in disease. Biochim Biophys Acta 1793, 78-88 (2009).

71. Smeitink, J., van den Heuvel, L. & DiMauro, S. The genetics and pathology of oxidative phosphorylation. Nat Rev Genet 2, 342-352 (2001).

198

72. Calvo, S., Jain, M., Xie, X., Sheth, S.A., Chang, B., Goldberger, O.A., Spinazzola, A., Zeviani, M., Carr, S.A. & Mootha, V.K. Systematic identification of human mitochondrial disease genes through integrative genomics. Nat Genet 38, 576-582 (2006).

73. Koene, S., Willems, P.H., Roestenberg, P., Koopman, W.J. & Smeitink, J.A. Mouse models for nuclear DNA-encoded mitochondrial complex I deficiency. J Inherit Metab Dis 34, 293-307 (2011).

74. Finsterer, J. Leigh and Leigh-like syndrome in children and adults. Pediatr Neurol 39, 223-235 (2008).

75. Chen, H. & Chan, D.C. Physiological functions of mitochondrial fusion. Ann N Y Acad Sci 1201, 21-25 (2010).

76. Cho, D.H., Nakamura, T. & Lipton, S.A. Mitochondrial dynamics in cell death and neurodegeneration. Cellular and molecular life sciences : CMLS 67, 3435-3447 (2010).

77. Zhang, L., Ding, F., Cao, W., Liu, Z., Liu, W., Yu, Z., Wu, Y., Li, W. & Li, Y. Stomatin-like protein 2 is overexpressed in cancer and involved in regulating cell growth and cell adhesion in human esophageal squamous cell carcinoma. Clin Cancer Res 12, 1639-1646 (2006).

78. Cao, W., Zhang, B., Liu, Y., Li, H., Zhang, S., Fu, L., Niu, Y., Ning, L., Cao, X., Liu, Z. & Sun, B. High-level SLP-2 expression and HER-2/neu protein expression are associated with decreased breast cancer patient survival. Am J Clin Pathol 128, 430-436 (2007).

79. Cao, W.F., Zhang, L.Y., Liu, M.B., Tang, P.Z., Liu, Z.H. & Sun, B.C. Prognostic significance of stomatin-like protein 2 overexpression in laryngeal squamous cell carcinoma: clinical, histologic, and immunohistochemistry analyses with tissue microarray. Hum Pathol 38, 747-752 (2007).

80. Guo, Q.C., Shen, J.N., Jin, S., Wang, J., Huang, G., Zhang, L.J., Yin, J.Q., Zou, C.Y. & Li, M.T. Comparative proteomic analysis of human osteosarcoma and SV40-immortalized normal osteoblastic cell lines. Acta Pharmacol Sin 28, 850-858 (2007).

81. Chang, D., Ma, K., Gong, M., Cui, Y., Liu, Z.H., Zhou, X.G., Zhou, C.N. & Wang, T.Y. SLP-2 overexpression is associated with tumour distant metastasis and poor prognosis in pulmonary squamous cell carcinoma. Biomarkers 15, 104-110 (2009).

82. Warburg, O. On respiratory impairment in cancer cells. Science 124, 269-270 (1956).

199

Appendix A: Ethics Approval

200

201

Appendix B: Copyright Permission

AMERICAN SOCIETY FOR MICROBIOLOGY LICENSE TERMS AND CONDITIONS

Sep 19, 2011 This is a License Agreement between Darah A Christie ("You") and American Society for Microbiology ("American Society for Microbiology") provided by Copyright Clearance Center ("CCC"). The license consists of your order details, the terms and conditions provided by American Society for Microbiology, and the payment terms and conditions. License Number 2752590906268 License date Sep 19, 2011 Licensed content publisher American Society for Microbiology Licensed content publication Molecular and Cellular Biology Licensed content title Stomatin-Like Protein 2 Binds Cardiolipin and

Regulates Mitochondrial Biogenesis and Function

Licensed content author Darah A. Christie, Joaquin Madrenas Licensed content date Sep 15, 2011 Volume 31 Issue 18 Start page 3845 End page 3856 Type of Use Dissertation/Thesis Format Electronic Portion Full article Order reference number Title of your thesis / dissertation

Characterization of SLP-2 function in T lymphocytes

Expected completion date Jan 2012 Estimated size(pages) 200 Billing Type Invoice Billing address Robarts Research Institute Room 2278, 100 Perth Drive London, ON N6A 5K8 Canada Customer reference info Total 0.00 USD

202

Curriculum Vitae

Name           Darah  Christie    

Education

2004 B.Sc. University of Waterloo, Waterloo, Ontario, Canada Honours Biochemistry

2007 M.Sc. University of Waterloo, Waterloo, Ontario, Canada

Department of Biology Supervisor: Dr. B. Dixon

2008-12 Ph.D. University of Western Ontario, London, Ontario Canada (Expected) Department of Microbiology and Immunology Supervisor: Dr. J. Madrenas

Honours and Awards

2004 NSERC – Undergraduate Student Research Award Entrance Scholarship – University of Waterloo

2005 CIHR Canada Graduate Scholarship – Master’s Award (Declined) Ontario Graduate Scholarship President’s Graduate Scholarship – University of Waterloo Entrance Scholarship – University of Waterloo 2007 Graduate Studies Research Travel Assistantship – University of Waterloo 2008 CIHR Doctoral Research Award (2008-2011)

Microbiology and Immunology Graduate Entrance Award Western Graduate Research Scholarship – Microbiology Schulich Scholarship for Medical Research

2009 eBioscience Award for Excellence in Graduate Poster Presentation

Western Graduate Research Scholarship – Microbiology Schulich Scholarship for Medical Research

2011 Microbiology and Immunology Graduate Travel Award Ontario Graduate Scholarship

203

Research Publications

1. Christie, D., Wei, G., Fujiki, K. and Dixon, B. Cloning and characterization of a cDNA encoding walleye (Sander vitreum) beta-2 microglobulin. Fish Shellfish Immun. 22:727-733 (2007).

2. Yue, D., Brintnell, W., Mannik, L.A., Christie, D., Haeryfar, S. M., Madrenas, J., Chakrabarti, S., Bell, D.A. and Cairns, E. CTLA4-Ig blocks the development and progression of citrullinated human fibrinogen-induced arthritis in DR4 tg mice. Arthritis Rheum 62(10) 2941-52 (2010).

3. Christie, D., Lemke, C.D., Elias, I.M., Chau, L.A., Kirchhof, M.G., Li, B., Ball, E.H., Dunn, S.D., Hatch, G.M. and Madrenas, J. Stomatin-like protein 2 binds cardiolipin and regulates mitochondrial biogenesis and function. Mol. Cell. Biol 31 (18) 3845-56 (2011).

4. Christie, D., Lian, D., Wang, H., Hatch, G. and Madrenas, J. SLP-2 deficiency in T cells is associated with abnormal mitochondrial compartmentalization of cardiolipin, decreased mitochondrial respiration and impaired CD4+ T cell responses. Manuscript in revision.

5. Christie, D., Kirchhof, M.G., Vardhana, S., Dustin, M.L., Madrenas, J. Mitochondrial and plasma membrane pools of stomatin-like protein 2 coalesce at the immunological synapse during T cell activation. Manuscript submitted.

Abstracts and Presentations

1. Graduate Student Research Conference. Waterloo, Ont. May 2007. Abstract and

oral presentation. “Regulation of Major Histocompatibility Class II Associated Invariant Chain Genes in Rainbow Trout”

2. Canadian Society of Zoology Annual Meeting. Montreal, Que. May 2007. Abstract and poster presentation. “Regulation of Major Histocompatibility Class II Associated Invariant Chain Genes in Rainbow Trout”

3. Robarts Research Day. London, Ont. March 2008. Abstract and poster

presentation. “Generation of a T Cell-Specific Knockout Mouse for Stomatin-like Protein 2 (SLP-2)”

204

4. Margaret Moffat Research Day. London, Ont. March 2008. Abstract and poster presentation. “Generation of a T Cell-Specific Knockout Mouse for Stomatin-like Protein 2 (SLP-2)”

5. Infection and Immunity Research Forum. London, Ont. November 2008.

Abstract and poster presentation. “Generation of a T Cell-Specific Knockout Mouse for Stomatin-like Protein 2 (SLP-2)”

6. American Association of Immunology Annual Meeting. Seattle, WA. May 2009. Abstract, oral and poster presentation. “Stomatin-like protein 2: a link between T cell activation and mitochondrial function”

7. Infection and Immunity Research Forum. London, Ont. November 2009.

Abstract and poster presentation. “The role of stomatin-like protein 2 in mitochondrial biogenesis”

8. Cold Spring Harbor, Gene Expression & Signaling in the Immune System. Cold

Spring Harbor, NY. April 2010. Abstract and poster presentation. “Stomatin-like protein 2: a link between T cell activation and mitochondrial function”

9. American Association of Immunology Annual Meeting. San Francisco, CA.

May 2011. Abstract and poster presentation. “SLP-2 deficiency in T cells is associated with decreased respiratory complex I levels and impaired TCR-dependent responses”

Teaching Experience

2004 Fundamentals of Microbiology - Laboratory Techniques

2005 Techniques in Molecular Biology Virology - Laboratory Techniques

2006 Techniques in Molecular Biology

Cellular and Molecular Immunology - Laboratory Techniques

2007 Cellular and Molecular Immunology - Laboratory Techniques Introduction to Applied Microbiology

2008 - 11 Laboratory Techniques in Microbiology and Immunology

Relevant Technical Experience

Bacterial expression of recombinant protein Polyclonal antibody production and purification

205

Southern, northern and western blotting One-dimensional and two-dimensional SDS-PAGE Reverse Transcriptase (RT) PCR Real Time PCR Mitochondrial isolation Chemical cell transformation Enzyme-Linked Immunosorbent Assay (ELISA) In vitro cell culture – primary culture and established cell lines Flow cytometry Confocal microscopy Animal handling

Training and Workshops

2008 Mitochondrial Molecular Biology and Pathology, May

National Institutes of Health, Bethesda, Maryland

Voluntary Services

2005-07 Grand River Regional Cancer Center – Present the New Patient

Orientation Session weekly

2006 TA Workshop – help train new teaching assistants 2009 Fall Preview Day – Discuss Microbiology and Immunology undergraduate

program with prospective students 2010 Fall Graduate Student Recruitment Fair – Discuss graduate work in the

Schulich School of Medicine and Dentistry 2010-11 Immunity and Infection Research Forum Committee Member – V.P. of

Finance and V.P. of Travel