237
Using Saccharomyces cerevisiae for the Biosynthesis of Tetracycline Antibiotics Ehud Herbst Submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy in the Graduate School of Arts and Sciences COLUMBIA UNIVERSITY 2019

Saccharomyces cerevisiae for the Biosynthesis of

  • Upload
    others

  • View
    0

  • Download
    0

Embed Size (px)

Citation preview

Using Saccharomyces cerevisiae for the Biosynthesis of Tetracycline Antibiotics

Ehud Herbst

Submitted in partial fulfillment of the

requirements for the degree of

Doctor of Philosophy

in the Graduate School of Arts and Sciences

COLUMBIA UNIVERSITY

2019

© 2019

Ehud Herbst

All rights reserved

ABSTRACT

Using Saccharomyces cerevisiae for the biosynthesis of tetracycline antibiotics

Ehud Herbst

Developing treatments for antibiotic resistant bacterial infections is among the most urgent public

health challenges worldwide. Tetracyclines are one of the most important classes of antibiotics,

but like other antibiotics classes, have fallen prey to antibiotic resistance. Key small changes in

the tetracycline structure can lead to major and distinct pharmaceutically essential improvements.

Thus, the development of new synthetic capabilities has repeatedly been the enabling tool for

powerful new tetracyclines that combatted tetracycline-resistance. Traditionally, tetracycline

antibiotics were accessed through bacterial natural products or semisynthetic analogs derived from

these products or their intermediates. More recently, total synthesis provided an additional route

as well. Importantly however, key promising antibiotic candidates remained inaccessible through

existing synthetic approaches.

Heterologous biosynthesis is tackling the production of medicinally important and structurally

intriguing natural products and their unnatural analogs in tractable hosts such as Saccharomyces

cerevisiae. Recently, the heterologous biosynthesis of several tetracyclines was achieved in

Streptomyces lividans through the expression of their respective biosynthetic pathways. In

addition, the heterologous biosynthesis of fungal anhydrotetracyclines was shown in S. cerevisiae.

This dissertation describes the use of Saccharomyces cerevisiae towards the biosynthesis of target

tetracyclines that have promising prospects as antibiotics based on the established structure-

activity relationship of tetracyclines but have been previously synthetically inaccessible.

Chapter 1 provides an introduction to the pursuit of tetracycline antibiotics using S. cerevisiae.

Following an overview of tetracycline drugs, the chapter describes the methods for making

tetracyclines and their limitations in accessing the tetracycline analogs targeted in this study. The

desirability of making these target analogs as well as key desired properties are then exemplified

by natural products, totally synthetic and semisynthetic derivatives. The target tetracycline analogs

pursued in this study are then outlined and the considerations in choosing their desired properties

are discussed, as well as the reasons for employing S. cerevisiae in their synthesis.

Chapter 2 describes the use of Saccharomyces cerevisiae for the final steps of tetracycline

biosynthesis, setting the stage for total biosynthesis of tetracyclines in Saccharomyces cerevisiae.

Chapter 3 describes the work towards biosynthesis of the target tetracycline analogs using

Saccharomyces cerevisiae, utilizing successful expression optimization and gene biomining

approaches. Chapter 4 describes the work towards the target tetracycline analogs from fungal

anhydrotetracyclines in Saccharomyces cerevisiae.

The challenge of enzyme evolution towards unnatural substrates and the complex environment of

cells require metabolic engineering efforts to be performed in libraries, as it is currently impossible

to predetermine which modifications will prove beneficial. Traditional methods in DNA

mutagenesis and increasingly, advances in DNA synthesis, DNA assembly and genome

engineering are enabling high throughput strain construction. Thus, there is a need for a general,

high-throughput, versatile and readily implemented assay for the detection of target molecule

biosynthesis. The development of such an assay is described in Chapter 5. The assay is

demonstrated to detect tetracycline derivatives, and differentiate a producer and a nonproducer

strain of the fungal anhydrotetracycline TAN-1612. The yeast three hybrid assay for metabolic

engineering of tetracycline derivatives described in this chapter could be used in the next steps

towards the heterologous biosynthesis of the target tetracycline analogs in S. cerevisiae and

beyond.

i

Table of Contents

List of Figures……………………………………………………………………………………vii

List of Schemes……………………………………………………………………………………x

List of Tables……………………………………………………………………………………...xi

List of Abbreviations……………………………………………………………………………xiii

1 An introduction to tetracycline biosynthesis using Saccharomyces cerevisiae ...................... 1

1.1 Chapter outline ................................................................................................................. 2

1.2 An antibiotic crisis ........................................................................................................... 2

1.3 Tetracycline antibiotics .................................................................................................... 3

1.3.1 General historical context for tetracycline antibiotics and tetracycline-resistance ....... 3

1.3.2 Tetracycline analogs – small structural changes can lead to major pharmaceutical

improvements .......................................................................................................................... 3

1.3.3 A structural overview of the FDA-approved tetracyclines ........................................... 4

1.4 Making tetracycline antibiotics ........................................................................................ 8

1.4.1 Biosynthesis and semisynthesis of tetracyclines using the original microbial hosts .... 9

1.4.2 Total synthesis of tetracycline antibiotics ................................................................... 12

1.4.3 Heterologous biosynthesis and semisynthesis of tetracyclines ................................... 14

1.5 Tetracycline analogs in the 6-position ........................................................................... 17

1.5.1 Dactylocyclines – natural product 6-epiglycotetracyclines ........................................ 17

1.5.2 6-demethyl-6-deoxy-6α-aryltetracyclines – totally synthetic tetracyclines ................ 18

ii

1.5.3 6-demethyl-6-deoxy-6α-methylenemercaptantetracyclines – semisynthetic

tetracyclines ........................................................................................................................... 19

1.6 Design of the biosynthesis of 6-demethyl-6-epiglycotetracyclines using S. cerevisiae 21

1.6.1 Targeting 6-demethyl derivatives of 6-epiglycotetracyclines for acid stability ......... 21

1.6.2 Targeting glycoside libraries on 6-demethyl-6-epiglycotetracyclines for broad

spectrum activity.................................................................................................................... 23

1.6.3 Targeting the 6-epi forms of 6-demethylglycotetracyclines for antibiotic efficacy ... 25

1.6.4 S. cerevisiae as the heterologous host of choice for 6-demethyl-6-epiglycotetracycline

biosynthesis ........................................................................................................................... 26

1.7 Research progress and future goals in the biosynthesis of 6-demethyl-6-

epiglycotetracyclines using S. cerevisiae .................................................................................. 29

1.7.1 Chapter 2 – Using Saccharomyces cerevisiae for the final steps of tetracycline

biosynthesis ........................................................................................................................... 29

1.7.2 Chapter 3 – Towards the biosynthesis of 6-demethyl-6-epitetracyclines using S.

cerevisiae ............................................................................................................................... 30

1.7.3 Chapter 4 – Towards the biosynthesis of 6-demethyl-6-epitracyclines from TAN-1612

in Saccharomyces cerevisiae ................................................................................................. 30

1.7.4 Chapter 5 – a yeast three hybrid system for the metabolic engineering of tetracycline

derivatives .............................................................................................................................. 31

1.8 References ...................................................................................................................... 32

2 Using Saccharomyces cerevisiae for the final steps of tetracycline biosynthesis ................. 41

iii

2.1 Chapter outline ............................................................................................................... 42

2.2 Introduction .................................................................................................................... 42

2.3 Materials and methods ................................................................................................... 43

2.3.1 General protocol for hydroxylation/reduction assay in cell lysate ............................. 43

2.3.2 Protocol for hydroxylation/reduction assay in whole cells ......................................... 44

2.4 Results ............................................................................................................................ 45

2.4.1 6β-hydroxylation of anhydrotetracycline in S. cerevisiae .......................................... 45

2.4.2 5a(11a)-dehydrotetracycline reduction in S. cerevisiae .............................................. 50

2.5 Discussion ...................................................................................................................... 53

2.5.1 6β-hydroxylation of anhydrotetracycline in S. cerevisiae .......................................... 53

2.5.2 5a(11a)-dehydrotetracycline reduction in S. cerevisiae .............................................. 55

2.6 Conclusion ...................................................................................................................... 56

2.7 Appendix ........................................................................................................................ 58

2.8 References ...................................................................................................................... 64

3 Towards the biosynthesis of 6-demethyl-6-epitetracyclines using S. cerevisiae ................. 70

3.1 Chapter outline ............................................................................................................... 71

3.2 Introduction .................................................................................................................... 72

3.3 Materials and methods ................................................................................................... 72

3.3.1 General protocol for high throughput assay of hydroxylation/reduction in whole cells

in microtiter plates ................................................................................................................. 72

iv

3.3.2 General protocol for western blots .............................................................................. 73

3.4 Results ............................................................................................................................ 74

3.4.1 Optimizing DacO1 expression in S. cerevisiae – rational design ............................... 74

3.4.2 Searching the bacterial hydroxylase space for 6α-hydroxylation of

anhydrotetracyclines .............................................................................................................. 77

3.4.3 Developing a microtiter plate assay for anhydrotetracycline hydroxylation .............. 79

3.4.4 Evolving a 6α-hydroxylase of anhydrotetracyclines – DacO1 random mutagenesis . 80

3.4.5 Testing DacO1 expression optimization constructs and other bacterial hydroxylases 82

3.5 Discussion ...................................................................................................................... 97

3.5.1 Optimizing DacO1 expression in S. cerevisiae – rational design ............................... 97

3.5.2 Searching the bacterial hydroxylase space for 6α-hydroxylation of

anhydrotetracyclines .............................................................................................................. 98

3.5.3 Developing a microtiter plate assay for anhydrotetracycline hydroxylation .............. 98

3.5.4 Evolving a 6α-hydroxylase of anhydrotetracyclines – DacO1 random mutagenesis . 99

3.5.5 Testing DacO1 expression optimization constructs and other hydroxylases............ 100

3.6 Conclusion .................................................................................................................... 102

3.7 Appendix ...................................................................................................................... 105

3.8 References .................................................................................................................... 127

4 Towards the biosynthesis of 6-demethyl-6-epitracyclines from TAN-1612 in Saccharomyces

cerevisiae .................................................................................................................................... 131

v

4.1 Chapter outline ............................................................................................................. 132

4.2 Introduction .................................................................................................................. 132

4.3 Materials and methods ................................................................................................. 133

4.3.1 General protocol for high throughput assay of TAN-1612 hydroxylation in whole cells

in microtiter plates ............................................................................................................... 133

4.3.2 Protocol for selecting amino acids for mutagenesis in hypothesized binding pockets of

monooxygenases .................................................................................................................. 134

4.4 Results .......................................................................................................................... 135

4.4.1 Testing PgaE and other bacterial hydroxylases for TAN-1612 hydroxylation......... 135

4.4.2 Searching the fungal hydroxylase space for 6α-hydroxylation of TAN-1612 .......... 145

4.4.3 Evolving monooxygenases for 6α-hydroxylation of anhydrotetracyclines – OxyS

example ................................................................................................................................ 149

4.4.4 Library screening for TAN-1612 hydroxylation in S. cerevisiae ............................. 152

4.5 Discussion .................................................................................................................... 154

4.5.1 Testing PgaE and other bacterial hydroxylases for TAN-1612 hydroxylation......... 154

4.5.2 Searching the fungal hydroxylase space for 6α-hydroxylation of TAN-1612 .......... 159

4.5.3 Evolving monooxygenases for 6α-hydroxylation of anhydrotetracyclines .............. 159

4.5.4 Library screening for TAN-1612 hydroxylation in S. cerevisiae ............................. 160

4.6 Conclusion .................................................................................................................... 161

4.7 Appendix ...................................................................................................................... 163

vi

4.8 References .................................................................................................................... 179

5 A yeast three hybrid assay for metabolic engineering of tetracycline derivatives .............. 181

5.1 Chapter outline ............................................................................................................. 182

5.2 Introduction .................................................................................................................. 183

5.3 Materials and methods ................................................................................................. 185

5.4 Results .......................................................................................................................... 192

5.4.1 Y3H system design and CID design and synthesis ................................................... 192

5.4.2 Initial characterization of the Y3H assay for tetracycline derivatives ...................... 194

5.4.3 Applying the Y3H assay to the detection of target molecule biosynthesis ............... 198

5.5 Discussion .................................................................................................................... 200

5.6 Conclusion .................................................................................................................... 203

5.7 Appendix ...................................................................................................................... 204

5.7.1 Supplementary figures .............................................................................................. 204

5.7.2 Supplementary methods ............................................................................................ 205

5.7.3 Supplementary tables ................................................................................................ 206

5.8 References .................................................................................................................... 211

vii

List of Figures

Figure 1-1 Structural comparison of FDA-approved tetracyclines................................................. 5

Figure 1-2 Comparison of the tetracycline lipophilic and zwitterionic forms ................................ 7

Figure 1-3 Key steps in the Myers convergent total synthesis of tetracyclines ............................ 14

Figure 1-4 Structures of the dactylocycline family of natural products ....................................... 18

Figure 1-5 6-demethyl-6-deoxy-6α-methylenemercaptantetracyclines and their antibiotic activities

....................................................................................................................................................... 20

Figure 1-6 Two possible C-O bond cleavage pathways in glycoside hydrolysis ......................... 22

Figure 1-7 Retrosynthetic analysis of 6-demethyl-6-epiglycotetracyclines ................................. 27

Figure 1-8 Structural Comparison between anhydrotetracycline, chloranhydrotetracycline, TAN-

1612 and anhydrotdactylocyclinone ............................................................................................. 28

Figure 2-1 Mass spectrometry analysis of anhydrotetracycline hydroxylation in cell lysate

expressing OxyS ........................................................................................................................... 47

Figure 2-2 UV/Vis spectroscopy analysis of the reaction of anhydrotetracycline in whole cells

expressing OxyS ........................................................................................................................... 49

Figure 2-3 Mass spectrometry of anhydrotetracycline hydroxylation and reduction in cell lysate

expressing OxyS in the presence of G6P ...................................................................................... 52

Figure 3-1 Western Blot of DacO1 and OxyS .............................................................................. 75

Figure 3-2 A simplified outline of the microtiter plate assay for anhydrotetracycline hydroxylation.

....................................................................................................................................................... 79

Figure 3-3 DacO1 error-prone mutagenesis screen excerpt ......................................................... 81

Figure 3-4 ΔExcitation spectrum for DacO1 and DacO1 error-prone PCR mutant ..................... 82

viii

Figure 3-5 Western blot analysis of DacO1 fusion proteins and other bacterial hydroxylases in

FY251 ........................................................................................................................................... 86

Figure 3-6 Western blot analysis of bacterial hydroxylases in BJ-5464-NpgA ........................... 87

Figure 3-7 Western blot analysis of DacO1-DacO4 and DacO4-DacO1 fusion proteins ............ 88

Figure 3-8 Western blot analysis of DacO1 and its fusion proteins in BJ5464-NpgA ................. 90

Figure 3-9 Mass spectrometry analysis of anhydrotetracycline hydroxylation and reduction by cell

lysates of strains expressing DacO1, its fusion proteins and other bacterial hydroxylases .......... 93

Figure 3-10 Mass spectrometry analysis of anhydrotetracycline hydroxylation and reduction in cell

lysates of strains expressing DacO1, its fusion proteins and other bacterial hydroxylases .......... 96

Figure 3-11 Mass spectrometry analysis of anhydrotetracycline hydroxylation and reduction in cell

lysates of strains expressing OxyS and PgaE ............................................................................... 97

Figure 4-1 UV/Vis analysis of hydroxylation attempt of TAN-1612 by bacterial hydroxylases 137

Figure 4-2 UV/Vis chromatogram of the HPLC separation of extracts from +/- PgaE strains

encoding the TAN-1612 pathway. .............................................................................................. 139

Figure 4-3 Mass spectrometry analysis of isolate from +/-PgaE strain encoding the TAN-1612

pathway. ...................................................................................................................................... 142

Figure 4-4 MSMS analysis of isolate from +PgaE strain encoding the TAN-1612 pathway..... 142

Figure 4-5 1H-NMR spectra of isolates from +/-PgaE strains encoding the TAN-1612 pathway.

..................................................................................................................................................... 143

Figure 4-6 COSY NMR spectrum of isolate from +PgaE strain encoding the TAN-1612 pathway.

..................................................................................................................................................... 144

Figure 4-7 Western blot analysis of fungal hydroxylase expression in BJ5464-NpgA .............. 148

ix

Figure 4-8 PyMOL illustrations of amino acids in proximity to the tetracycline substrate in

aklavinone-11-Hydroxylase and in OxyS ................................................................................... 151

Figure 4-9 Library screening for TAN-1612 hydroxylation in S. cerevisiae ............................. 153

Figure 4-10 Possible moieties in the product isolated from +PgaE strain coexpressing the TAN-

1612 bioysnthetic pathway ......................................................................................................... 155

Figure 4-11 Possible intermediates in the biosynthesis of TAN-1612 ....................................... 156

Figure 4-12 xanthurenic acid and its derivative that could correspond to experimental mass and

NMR spectra ............................................................................................................................... 158

Figure 5-1 Detection of a target molecule biosynthesis by the yeast three hybrid system. ........ 184

Figure 5-2 Characterization of the dynamic range of the Y3H assay for tetracyclines .............. 195

Figure 5-3 The modularity of the yeast three hybrid assay enables both screening and selecting for

biosynthesis of target tetracyclines ............................................................................................. 197

Figure 5-4 Differentiation of a producer and a non-producer strain of a target molecule by the Y3H

assay for tetracyclines, demonstrating the applicability of the Y3H assay to metabolic engineering.

..................................................................................................................................................... 199

Figure 5-5 LacZ readout of strain harboring plasmid encoding for LexA-TetR (PBA-8) and strain

without the plasmid (PBA-5). ..................................................................................................... 204

Figure 5-6 standard curve for TAN-1612 quantification in cultures. ......................................... 205

x

List of Schemes

Scheme 2-1 Two-step enzymatic conversion of anhydrotetracycline to tetracycline ................... 46

Scheme 3-1 Bacterial monooxygenases and their hypothesized substrates and products ............ 78

Scheme 3-2 Two-step enzymatic conversion of (6-demethyl-)anhydrotetracycline to (6-demethyl-

)6-epitetracycline .......................................................................................................................... 91

Scheme 4-1 Biosynthetic plan for TAN-1612 6α-hydroxylation ............................................... 135

Scheme 4-2 Bacterial monooxygenases and their hypothesized substrates and products .......... 136

Scheme 5-1 Synthesis of the chemical inducer of dimerization (CID) Min-Mtxa ...................... 193

xi

List of Tables

Table 1-1 FDA approved tetracycline derivatives and their differentiating functional groups ...... 6

Table 1-2 Rates of hydrolysis of β-D-glycopyranosides in 0.5 M aqueous sulfuric acid ............. 23

Table 2-1 Strains used in this study .............................................................................................. 58

Table 2-2 Plasmids used in this study ........................................................................................... 58

Table 2-3 Sequences used in this study ........................................................................................ 59

Table 3-1 Potential hits in the screen of DacO1 expression optimization constructs and other

bacterial hydroxylases ................................................................................................................... 85

Table 3-2 UV/Vis spectroscopy results for hydroxylase strains cell lysates incubated with

anhydrotetracycline and glucose-6-phosphate .............................................................................. 94

Table 3-3 Assay conditions for anhydrotetracycline hydroxylation and reduction in cell lysates of

strains expressing DacO1, its fusion proteins and other bacterial hydroxylases .......................... 95

Table 3-4 Strains used in this study ............................................................................................ 105

Table 3-5 Plasmids used in this study ......................................................................................... 106

Table 3-6 Sequences used in this study ...................................................................................... 107

Table 4-1 Fungal monooxygenases biomined for TAN-1612 6α-hydroxylation in S. cerevisiae

..................................................................................................................................................... 146

Table 4-2 Residues chosen for mutagenesis in OxyS ................................................................. 149

Table 4-3 Rank of PgaE positive control in screen for TAN-1612 hydroxylase ........................ 152

Table 4-4 Strains used in this study ............................................................................................ 163

Table 4-5 Plasmids used in this study ......................................................................................... 164

Table 4-6 Plasmid Library A – Strain EH-5-217-1 .................................................................... 165

Table 4-7 Plasmid Library B – Strain EH-5-217-3 ..................................................................... 165

xii

Table 4-8 Plasmid Library C – Strain EH-5-217-1 and EH-5-217-3.......................................... 165

Table 4-9 Plasmid Library D – Strains EH-5-217-2 and EH-217-4 ........................................... 166

Table 4-10 Sequences used in this study .................................................................................... 166

Table 4-11 Primers used for OxyS saturation mutagenesis in this study ................................... 177

Table 5-1 Strains used in this study ............................................................................................ 206

Table 5-2 Plasmids used in this study ......................................................................................... 207

Table 5-3 Sequences of the various TetR classes in the within the pMW103 backbone. .......... 207

xiii

List of Abbreviations

5-FOA 5-fluoroorotic acid

AD activation domain

DBD DNA binding domain

DCM dichloromethane

DHFR dihydrofolate reductase

DMF dimethylformamide

EtOAc ethyl acetate

FAD flavin adenine dinucleotide

G6P glucose-6-phosphate

HPLC high-pressure liquid chromatography

HRMS high resolution mass spectroscopy

kb kilobase pair

LCMS liquid chromatography mass spectrometry

MeCN Acetonitrile

MeOH methanol

MIC minimal inhibitory concentration

MS-MS tandem mass spectrometry

NADPH dihydronicotinamide adenine dinucleotide phosphate

NMR nuclear magnetic resonance

ONPG o-nitrophenyl β-d-galactopyranoside

PAGE polyacrylamide gel electrophoresis

xiv

PR protein receptor

PKS polyketide synthase

r.t. room temperature

SDS sodium dodecyl sulfate

TetR tetracycline repressor

TIC total ion count

TFA trifluoroacetic acid

Tris Tris(hydroxymethyl)aminomethane

w.t. wild type

X-Gal 5-bromo-4-chloro-3-indolyl-β-d-galactopyranoside

xv

Acknowledgments

I would like to thank my advisor Prof. Virginia Cornish for a wide cast mentorship, for providing

a very fertile environment for sciencemaking and for the pushes to dare scientifically.

I would like to thank my thesis committee members, Prof. Scott Snyder, Dr. George Ellestad, Prof.

Ruben Gonzalez and Prof. Anne-Catrin Uhlemann as well as my advisory committee member

Prof. Tristan Lambert. The valuable discussions with you throughout the years have been

tremendously helpful to shape this research direction. I consider myself extremely lucky to have

benefited from your highly relevant expertise to this research and your will to mentor.

I have profited immensely in various ways from interacting with the highly talented and committed

members, past and present, of the Cornish and neighboring labs. Specifically, I would like to thank

Arden Lee, Pedro Baldera-Aguayo and Hyunwook Lee for their large contribution to this research

and for being, each in their distinctive style, a pleasure to work with. I would like to thank Dr.

Andrew Anzalone, Dr. Nili Ostrov, Dr. Casey Brown, Dr. Sonja Billerbeck, Dr. Zhixing Chen, Dr.

Caroline Patenode, Dr. Marie Harton, Dr. Bertrand Adanve, Dr. Miguel Jimenez, Dr. Andy (Yao

Zong) Ng, Dr. Mia Shandell, Sean Bannier, Dr. Anna Kaplan and Dr. Andreas Hartel for

collaborations and/or mentorship and/or technical assistance.

I would like to thank Prof. Yi Tang for sharing his time and wide-ranging knowledge, as well as

plasmids pYR291 and pYR342 and yeast strain BJ5464-NpgA, Prof. Alan R. Davidson for sharing

pET21d plasmid encoding TetR(B), Prof. Frank Foss for sharing synthetic FO and Dr. Bertrand T.

Adanve for the synthesis of Methotrexate-α-OtBu.

I would like to thank my family for making the immense effort it takes to be close while an ocean

apart. I would like to thank my friends in Israel and the US for their support and company. Finally,

I would like to thank Jess for the love and support throughout the years. It’s been quite a journey.

xvi

To my Savtas. For your perseverance.

1

1 An introduction to tetracycline biosynthesis using

Saccharomyces cerevisiae

2

1.1 Chapter outline

This chapter provides an introduction to the pursuit of tetracycline antibiotics using S. cerevisiae.

First, the significance of the production of new antibiotic analogs from a human health perspective

is mentioned (Section 1.2). Then, the general historical context for developing tetracycline

antibiotics is given (Section 1.3.1), the antibiotic promise in making tetracycline analogs is

discussed (Section 1.3.2) and a structural overview of FDA-approved tetracyclines follows

(Section 1.3.3).

Three modes of synthesis of tetracycline antibiotic are then introduced in the historical order in

which they appeared, biosynthesis and semisynthesis using the original microbial hosts (Section

1.4.1), total synthesis (Section 1.4.2) and heterologous biosynthesis and semisynthesis (Section

1.4.3). The desirability of making 6-position analogs of tetracyclines as well as key desired

properties of such analogs is then exemplified by natural products (Section 1.5.1), totally synthetic

(Section 1.5.2) and semisynthetic derivatives (Section 1.5.3).

The target tetracycline analogs pursued in this study are then outlined and the considerations in

choosing their desired properties are then discussed (Sections 1.6.1, 1.6.2 and 1.6.3) as well as the

reasons for employing S. cerevisiae in their synthesis (Section 1.6.4). Finally, an overview of the

research plan and its execution throughout this dissertation is given (Section 1.7).

1.2 An antibiotic crisis

Developing treatments for antibiotic resistant bacterial infections is among the most urgent public

health challenges world-wide.(1) The US Center for Disease Control reports over 2 million

illnesses and 20,000 deaths annually in the United States as a result of antibiotic resistance.(1)

Novel antibiotic drug candidates have traditionally been accessed through natural product

3

screening, total synthesis, semisynthesis and combinatorial chemistry. However, the number of

new antibiotics approved by the FDA has dropped significantly from previous decades, from 29

approvals in the 1980s to 9 in the 2000s.(2, 3) To reverse this trend, transformative new

technologies for discovery of new antibiotics are needed.

1.3 Tetracycline antibiotics

1.3.1 General historical context for tetracycline antibiotics and tetracycline-

resistance

Tetracyclines are one of the most important classes of antibiotics,(4) but like other classes of

antibiotics, have fallen prey to resistance.(5) From their discovery in the 1940’s, tetracyclines were

used as broad spectrum antibiotics, as well as for other types of disease, such as periodontitis.(6)

The tetracyclines inhibit bacterial protein synthesis by binding reversibly to the 30S ribosomal

subunit and sterically hindering aminoacyl-tRNA binding to the ribosomal A-site.(7, 8) However,

bacteria have evolved a growing number of resistance mechanisms – including efflux pumps,(9)

ribosomal protection proteins,(10) rRNA mutations,(11) and enzymatic degradation.(12)

1.3.2 Tetracycline analogs – small structural changes can lead to major

pharmaceutical improvements

Small changes in the tetracycline structure can lead to major and distinct pharmaceutically

essential improvements. Examples include tetracycline analogs differing from tetracycline in only

up to three positions and showing improved pharmacokinetic properties, binding affinity to the

ribosome, activity against resistant strains and non-antimicrobial properties.(13-16)

Thus, for example, a difference in the position of one hydroxy group between tetracycline and

doxycycline improves the half-life, tissue penetration and matrix metalloproteinase (MMP)

4

inhibition properties of the latter (Figure 1-1 and Table 1-1).(13) Due to such advantages,

doxycycline is not only one of the most commonly clinically indicated tetracycline antibiotics, it

is the only clinically available MMP inhibitor as of 2010 and is indicated for the treatment of

periodontitis.(14)

Minocycline differs from tetracycline in only three functional groups, a hydroxy, a methyl and an

N,N-dimethylamino (Figure 1-1 and Table 1-1). Impressively, these differences are enough to

improve minocycline relative to tetracycline in its pharmacokinetic profile, radical oxygen species

scavenging and other anti-inflammatory properties.(13, 14) While tetracycline is unable to cross

the blood-brain barrier (BBB), minocycline is known for its neuroprotective properties that have

been demonstrated pre-clinically and clinically in the treatment of various neurological disease.

Examples include ischemia, traumatic brain injury and neuropathic pain, Parkinson's disease,

Huntington's disease, amyotrophic lateral sclerosis, Alzheimer's disease, multiple sclerosis and

spinal cord injury.(15)

Lastly, tigecycline, being identical to minocycline except for the 9-t-butyl-glycylamido group has

a better binding affinity to the A-site of the ribosome and an improved antimicrobial activity

against tetracycline-resistant strains (Figure 1-1 and Table 1-1).(16) On the other hand, these

changes also led to tigecycline being orally unavailable and furthermore, tigecycline use is

associated with an increased risk of death.(17)

1.3.3 A structural overview of the FDA-approved tetracyclines

As of 2018, all nine FDA approved tetracycline antibiotics were either bacterial natural products

or semisynthetic analogs derived from bacterial natural products or their intermediates, and all

nine are produced commercially partly or wholly by fermentation (Figure 1-1).(6, 18-20) Three of

these nine tetracycline antibiotics, chlortetracycline, meclocycline and metacycline, were

5

discontinued.(20) In 2018, three additional tetracycline antibiotics have received their FDA

approval, omadacycline, sarecycline and eravacycline.(20-22) Importantly, FDA approved

tetracycline derivatives are identical in all positions but for the 5α-, 6α-, 6β-, 7- and 9-positions

(Table 1-1).

Figure 1-1 Structural comparison of FDA-approved tetracyclines

The common name, a full name outlining the structural relationship between the different

derivatives and the year of FDA approval, are mentioned below the structures.(6, 20, 23)

6

Table 1-1 FDA approved tetracycline derivatives and their differentiating functional groups

Namea 5α 6α 6β 7 9

Chlortetracycline Me OH Cl

Oxytetracycline OH Me OH

Tetracycline Me OH

Demeclocycline OH Cl

Doxycycline OH Me

Minocycline dimethylamino

Meclocycline OH CH2 CH2 Cl

Metacycline OH CH2 CH2

Tigecycline dimethylamino tBu-glycylamido

Sarecycline (methoxy(methyl)amino)

methyl

Omadacycline dimethylamino neopentylaminomethyl

Eravacycline F pyrrolidinoacetamido aFDA approved tetracycline derivatives are identical in all positions but the 5α-, 6α-, 6β-, 7- and

9-positions. Empty cells indicate implicit Hs.

The identity of the FDA approved tetracyclines at all positions but for 5α, 6α, 7, 8 and 9 could be

explained as stemming from two major reasons. One is the antibacterial ineffectiveness of

tetracycline analogs generated so far in positions 10, 11, 11a, 12, 12a, 1, 2, 3 and 4. The other is

limited semisynthetic and total synthetic access to modifications in positions such as 4a and 6α

that could potentially yield successful tetracycline antibiotics but are awaiting efficient synthetic

methods to access them. Both of these topics will be expanded on below.

Given that positions 10, 12a, 1, 2 and 3, as well as the β-diketone of 11,11a and 12 of tetracyclines

(Figure 1-1) are considered to interact with the ribosome and thus to be directly responsible for

tetracyclines antibiotic activity,(8, 24) it is not surprising that modifications in these positions do

not yield effective antibiotics. The 2-position is somewhat of an exception as modification at this

position can yield tetracycline antibiotic prodrugs that hydrolyze to the unmodified 2-carboxamido

functional group (Section 1.4.1). Modifications at the 4-position have also not yielded

antibiotically active tetracyclines even though this position is not considered to be important for

7

ribosome binding. The prevalent hypothesis to explain the requirement for the 4α-dimethylamino

group in tetracycline antibiotics is the importance of maintaining an equilibrium between a

zwitterionic and a nonionic (lipophilic) structures (Figure 1-2, Section 1.4.1).(25) This equilibrium

is considered crucial for tetracycline antibiotic activity as the zwitterionic form is the form of

tetracycline that binds to the ribosome,(8) and the nonionic form is the form that can cross the

cytoplasmic membrane.(26) It is therefore clear that the promising positions for modifications that

could yield tetracycline antibiotics are likely to be limited to positions 4a, 5, 5a, 6, 7, 8 and 9.

Figure 1-2 Comparison of the tetracycline lipophilic and zwitterionic forms

Adapted from (26).

In order to explain which modifications in which positions are of promise to obtain tetracycline

analogs with desired antibiotic activity but have not yet been accessed, the following two

subsections of the introduction are given. The first (Section 1.4) introduces three different fronts

of tetracycline synthesis to access tetracycline antibiotic analogs, biosynthesis and semisynthesis

using the original microbial hosts (Section 1.4.1), total synthesis (Section 1.4.2) and heterologous

biosynthesis and semisynthesis (Section 1.4.3). Specifically, Section 1.4 discusses which

modifications were so far inaccessible by all existing methods. The following section introduces

desirable tetracyclines that are inaccessible by biosynthesis and semisynthesis using the original

microbical hosts and by total synthesis and could be conceived to made by heterologous

biosynthesis (Section 1.5). Finally, the last two sections of the introduction describe the plan to

8

access these target antibiotics using S. cerevisiae (Section 1.6) and the overview of the

experimental progress and future plans towards these analogs (Section 1.7).

1.4 Making tetracycline antibiotics

In this section an overview of the three main methods for the production of tetracycline antibiotics

is given, biosynthesis and semisynthesis using the original microbial hosts (Section 1.4.1), total

synthesis (Section 1.4.2) and heterologous biosynthesis and semisynthesis (Section 1.4.3). This

overview emphasizes two main points. For one, it is shown here that synthetic advancements in

the tetracycline field have time and again led to the development of FDA approved tetracyclines.

The second point emphasized in this overview is the inaccessibility of key tetracycline antibiotic

analogs through both of the established methods of tetracycline analog production, biosynthesis

and semisynthesis using the original microbial hosts (Section 1.4.1) and total synthesis (Section

1.4.2).

Some example for the first main point include the FDA approval of minocycline on 1971 following

the mutgenesis studies that enabled the fermentation of 6-demethyltetracyclines (e.g.

demeclocycline) and the followup synthetic studies that enabled aromatic substitutions in the 7-

position. This synthetic achievement was in turn followed up with studies on aromatic substitions

of minocycline in the 9-position that gave birth to the tigecycline that received its FDA approval

on 2005 (Section 1.4.1). Similarly, a breakthrough in the younger field of tetracycline total

synthesis, enabling unprecedented modifications in the tetracycline D-ring such as 7-

fluorotetracyclines, gave birth to eravacycline that was FDA approved on 2018 (Section 1.4.2).

Some examples for the second main point include promising tetracycline analogs at the 6- and 4a-

positions that are further discussed in Section 1.5. It remains to be seen whether unique

9

transformations enabled by the much younger field of heterologous biosynthesis and semisynthesis

of tetracyclines would yield FDA approved antibiotics (Section 1.4.3).

1.4.1 Biosynthesis and semisynthesis of tetracyclines using the original

microbial hosts

Tetracycline biosynthesis and semisynthesis using the original microbial hosts products can yield

modifications in positions 1, 2, 3, 4, 5, 5a, 6, 7, 9, 11, 11a, 12 and 12a.(25) The original microbial

hosts referred to here include Streptomyces aureofaciens and Streptomyces rimosus, the original

producers of chlortetracycline and oxytetracycline, as well as the S. aureofaciens mutants that

produce tetracycline and demeclocycline.(27) Despite the limitations on the positions that can be

modified and more importantly despite the limitations on the functional group that could be

introduced in these positions, this strategy yielded all FDA-approved tetracyclines that are not

natural products prior to 2018.(19)

Considering that positions 12a, 1, 2 and 3 and the β-diketone of 11,11a and 12 are known to be

important for the interaction of tetracyclines with the ribosome,(8, 24, 25) it is not surprising that

modifications in these positions have never led to a new tetracycline antibiotic drug. I will therefore

provide a short overview for biosynthetic and semisynthetic modifications using the original host

that led to tetracyclines modified in positions 4, 5, 5a, 6, 7, and 9. For a broader review of the

semisynthesis of tetracyclines using the original microbial hosts the reader is directed to the

excellent book chapter by Rogalski.(25) For more recent reviews in the field, covering tigecycline

and related derivatives as well, the reader is referred to references (6, 22, 28-31).

Modifications at the 4-position have so far not presented tetracycline analogs with improved

antibiotic activity and most commonly have worsened antibiotic outcomes in comparison to the

unmodified counterparts. For example, 4-epitetracycline, 4-dedimethylaminotetracycline and 4-

10

hydroxy-4-dedimethylaminotetracycline all present much higher MIC values relative to

tetracycline.(25) Increasing bulk on the 4-position in 6-demethyltetracycline derivatives leads to

reduced antibiotic activity as well. 6-demethyltetracycline derivatives containing

methylethylamino, methylpropylamino, diethylamino and methyl(2-hydroxyethyl)-amino at the

4α-position instead of the 4α-dimethylamino group of 6-demethyltetracycline show 75%, 50%,

25% and 12%, respectively, of the antibiotic activity of tetracycline against Staphylococcus aureus

while 6-demethyltetracycline maintains 96% of the activity.(32)

The study the effect of reducing bulk on the 4-amino group was attempted as well by synthesizing

4-monoalkylamines, but only the synthesis of 4β-methylamines has succeeded. Considering that

4-epitetracycline has much reduced antibiotic activity it is not surprising that these derivatives

were much less effective antibiotics than tetracycline as well. The one monoalkylamino

successfully generated, 6-demethyl-4-dedimethylamino-4-methylaminotetracycline has shown

only 7% of the antibiotic activity of tetracycline.(32)

While 6-methylene tetracycline has effective antibiotic activity (e.g., MIC value of 0.2 against

tetracycline-sensitive S. aureus), its 4-trimethylammonium derivative 6-methylenetetracycline

methiodide does not (e.g., MIC value of 12 against tetracycline-sensitive S. aureus).(23) While

some of the difference could stem from the partial epimerization at the 4-position for the

methiodide, the partial epimerization alone cannot explain the difference. The lack of the proton

on the 4-dimethylammonium required to form the characteristic H-bond of the zwitterionic form

(Figure 1-2) is hypothesized to explain why 4-quaternary ammonium derivatives cannot form the

zwitterionic tetracycline form,(33) and could be a reason for their higher MIC values. 4-

epitetracyclines would be incapable of forming the zwitterionic form (Figure 1-2) as well and

similar problems could arise with increasing bulk on the 4-amino group.

11

The 4a proton was unmodified by biosynthesis and semisynthesis of tetracycline derivatives using

the original microbial hosts, as is to be expected given that C-H activation of the tertiary 4a-proton

is challenging in a highly functionalized molecule such as tetracycline and its natural and

semisynthetic analogs.(25, 34) A 4a(12a)-anhydrotetracycline was made and found to have less

than 1% the activity of tetracycline.(25)

Modifications at the 5-position enabled by biosynthesis and semisynthesis using the original

microbial hosts were focused on modifications of oxytetracycline and doxycycline (Figure 1-1),

such as esterification, and oxidation. In general, these modifications have either yielded

tetracycline derivatives with similar or worse antibiotic activity.(25) Modifications at the 5a-

position were mostly limited to dehydration, isomerization and 5a-methyl installation but have not

produced improvements on biological activity.

Modifications at the 6-position have yielded many of the currently FDA-approved tetracyclines

(Figure 1-1, Table 1-1). Some of the 6-position analogs made had the following functional groups

in the α and β positions, respectively: Me,OH; Me,H; H,Me; Me,-; H,OH; H,H; CH2,-; RSCH2,H;

OH,-; OH, HOCH2; PhCH2,H; RSOCH2,H; CH3COCH2,H; HOCH2CH2CH2,H; PhCH2CH2,H;

F,H; H,F; F,-; F,Me; H, OAc; with R indicating and alkyl, phenyl or a derivatized alkyl or phenyl

and a hyphen indicating an SP2 carbon at the 6-position.(25) Thus, it is clear that alkyl

derivatizations of the 6α-position were explored, as well as some heteroatoms such as fluorine.

However, the scope was still limited and has not included 6-epi-6-demethytetracyclines, a point

that will be revisited in section 1.6. This is to be expected considering that natural tetracyclines

such as tetracycline, oxytetracycline and chlortetracycline give ready access to a 6β-

hydroxytetracyclines. Notably, 6-demethyltetracyclines such as demeclocycline (Figure 1-1) were

accessed by strain mutagenesis.(25)

12

Experimental success with aromatic substitutions enabled initially the semisynthetic derivatization

of the 7-position generating minocycline that later received FDA approval (Figure 1-1).(6) 9-

position substation followed and thus the road was paved for the newer generation of semisynthetic

tetracyclines, tigecycline, sarecycline and omadacycline (Figure 1-1).(21, 22, 25, 28, 35, 36)

However, the scope of possible substitutions in the D-ring has been significantly expanded with

the Myers convergent approach to tetracycline total synthesis, covered in Section 1.4.2.

Modifications at the 2-position have generated tetracycline antibiotics that were approved outside

the United States. Rolitetracycline, lymecycline (N-lysinomethyltetracycline) and clomocycline

are such examples. However, the modifications in general made for tetracycline prodrugs with

improved water solubility that hydrolyze to tetracyclines unmodified in the 2-position, which is

their active form.(25)

1.4.2 Total synthesis of tetracycline antibiotics

Early efforts at the total synthesis of tetracyclines include the first total synthesis of a biologically

active tetracycline, 6-demethyl-6-deoxytetracycline by the Woodward laboratory in the 1960s,

albeit as a racemic mixture.(37) Later synthetic efforts of interest to antibiotically active analog

generation included generating heteroatoms at the 6-position in derivatives such as 6-

thiatetracycline.(25) However, the latter was found to be acting as an antibiotic by an alternative

mechanism that does not involve the ribosome and was labeled, along with anhydrotetracycline,

anhydrochlortetracycline, chelocardin and 4-epianhydrochlortetracycline, as an atypical

tetracycline. Importantly, adverse side effects precluded the latter group from being perused as

antibiotic agents.(38)

While other total syntheses of tetracyclines appeared over the years, (39, 40) the Myers’ group

convergent enantioselective total synthesis of tetracyclines,(41) was the first one to produce a

13

totally synthetic FDA approved tetracycline.(20, 42) This convergent approach to tetracycline total

synthesis permitted tetracycline analog modifications previously impossible, particularly in the 7-

, 8- and 9-positions. These modifications enabled the synthesis of TP-271, a clinical candidate and

the FDA approved eravacycline, both 7-dedimethylamino-7-fluoro-9-amidominocyclines (Figure

1-1).(18, 43, 44) Additional tetracycline analogs that became accessible by later developments in

this total synthesis approach are 5-oxo analogs with an oxygen instead of the C5 carbon and 5a-

quaternary carbon analogs.(45, 46) Importantly, the Myers convergent synthesis of tetracyclines

does involve a fermentation process in its first step, the enantioselective dihydroxylation of

benzoic acid.(41, 47) However, this synthesis is clearly distinct from the semisynthetic efforts

towards tetracyclines that modify fully formed tetracycline fermentation products (Section 1.4.1).

On the other hand, the synthetic strategy for forming the AB-ring precursor and then coupling it

with the D-ring precursor across the C-ring limits modifications to the AB-ring junction and C-

ring.(41) Specifically, the Michael-Claisen cyclization that forms the ABCD tetracycline core from

the AB- and D-ring precursors demands a D-ring precursor that can undergo anionization at the

carbon precursor to the C6 carbon and then serve as a nucleophile for 1,4-addition reaction to an

α,β-unsaturated carbonyl (Figure 1-3-a). This demand limits the functional diversity that can be

convergently generated at the 6α-position (Figure 1-1). 6α-substituents that were thus published in

the Myers group include alkyl, aryl, heteroaryl, and proton substituents.(41, 42) Thus, some 6-

deoxytetracyclines such as doxycycline and some 6-deoxy-6-demethyl tetracyclines such as

eravacycline can be convergently synthesized using this total synthesis strategy.(41, 42, 44)

However, other simple tetracyclines, such as tetracycline itself, require an alternative cyclization

strategy and further oxygenation following the cyclization, and thus while the overall yield for

doxycycline was 8.3%, the overall yield for tetracycline was 1.1%.(41, 48)

14

Substitutions at the 4a-positions also appear limited by the convergent approach to tetracycline

total synthesis, as epoxide formation and opening on this position are critical to the formation of

the AB-ring intermediate (Figure 1-3-b).(41) Finally, it is also unclear at this point whether this

method of tetracycline total synthesis can compete economically with fermentation, or with

semisynthesis from a fermentation intermediate, for commercial process chemistry of a wide range

of tetracyclines indicated for a wide range of infectious diseases.

Figure 1-3 Key steps in the Myers convergent total synthesis of tetracyclines

The key step to form the (a) ABCD precursor and (b) AB-ring precursor in the total synthesis of

doxycycline. Adapted from (41)

1.4.3 Heterologous biosynthesis and semisynthesis of tetracyclines

Heterologous biosynthesis of natural products and their analogs is a rapidly developing field and

is importing important biosynthetic pathways into tractable hosts, such as Escherichia coli and

Saccharomyces cerevisiae. This field relies heavily on recent breakthrough in the fields of DNA

synthesis, DNA sequencing, methods in genetic manipulation of model organisms and information

15

regarding natural product pathways in a variety of organisms.(49) Heterologous biosynthesis is

tackling the production of medicinally important and structurally complex natural products such

as taxol,(50) erythromycin,(51) and lovastatin.(52) Notably, heterologous biosynthesis offers a

greener or more economically feasible approach to the production of known compounds.(53, 54)

While much of the effort in heterologous biosynthesis is still focused on the production of natural

products themselves, an ultimate goal is to obtain their unnatural derivatives as well.(55)

Heterologous biosynthesis in the tetracycline field was spearheaded by the Tang laboratory who

produced in Streptomyces lividans K4 by herelogous expression oxytetracycline (Figure 1-1),

dactylocyclinone (Figure 1-4) and SF2575 through the expression of their respective biosynthetic

pathways.(56) Analog production of the antitumor agent SF2575 was also shown in this study by

the elimination of biosynthetic enzymes from the SF2575 pathway. As is the common case for the

heterologous production of other glycosylated natural products,(57) the aglycone,

dactylocyclinone and not the glycosylated natural products dactylocyclines has been

produced.(56)

The power of genetic modifications to the biosynthetic pathway in yielding tetracycline derivatives

was historically used in the original hosts. For example, in the production of tetracycline and

demeclocycline by mutants of Streptomyces aureofaciens that naturally produces

chlortetracycline.(58) Demeclocycline is an intermediate in the semisynthesis of the important

drug minocycline and a tetracycline drug on its own right (Figure 1-1).(59) While impressive for

having taken place before full deciphering of the tetracycline biosynthetic pathway and prior to

the development of efficient tools for genomic engineering, these two analogs were obtained by

elimination of function rather than by introduction of function, which is more challenging. The

16

genetic revolution that intervened and the use of genetically tractable heterologous organisms such

as S. cerevisiae are a potential avenue towards such challenging transformations.(49)

Importantly the Tang group has also heterologously expressed in S. cerevisiae the biosynthetic

pathway of the fungal anhydrotetracycline derivative, TAN-1612.(60) Fungal

anhydrotetracyclines such as TAN-1612 and viridicatumtoxin serve as excellent scaffolds for

generating tetracycline analogs in S. cerevisiae (Section 1.6.4 and Chapter 4). While not pursued

thus far in tetracyclines, heterologous semisynthesis combining the power of heterologous

biosynthesis with the often complimentary advantages of synthetic chemistry is exciting. Such

combination has led to the production of artemisinin from artemisinic acid fermented in yeast,(61)

and could prove especially beneficial for generating tetracycline analogs (Section 1.6.4).

In conclusion, for each of the fronts of tetracycline synthesis, biosynthesis and semisynthesis using

the original microbial hosts, total synthesis and heterologous biosynthesis and semisynthesis, the

emergence of new capabilities is the enabling tool for powerful new antibiotics. Thus, the

development of semisynthetic capabilities in the 6-, 7- and 9-positions enabled the synthesis of

novel more powerful tetracycline antibiotics such as doxycycline, minocycline and tigecycline

(Section 1.4.1). The expansion of the range of possible modifications at the 7- and 9-positions by

total synthesis enabled the synthesis of 7-fluoro-9-amidotetracycline antibiotics such as

eravacycline (Section 1.4.2). Importantly however, key tetracycline analogs such as 6-demethyl-

6-epitetracyclines and 4a-hydroxytetracyclines, as well as their derivatives remained inaccessible

through the existing synthetic approaches (Sections 1.4.1 and 1.4.2) It is now to be tested whether

the new efforts enabling the heterologous expression of tetracycline biosynthetic pathways will

give rise to new tetracycline antibiotic drugs (Section 1.4.3). Section 1.5 describes exciting

potential structures for such tetracyclines.

17

1.5 Tetracycline analogs in the 6-position

The 6-position of tetracycline has been particularly useful for generating tetracycline analogs.

Indeed, no less than five different functional group combinations in the 6-position exist for FDA-

approved tetracyclines (Figure 1-1 and Table 1-1). As noted by Rogalski in 1985, “Modifications

at the C6 atom have produced by far the greatest success in evolving highly active

tetracyclines.”(25) Three different classes of 6-position tetracycline analogs will be outlined here

as inspiration for future tetracycline antibiotics analogs in the 6-position, the natural product

dactylocyclines (Section 1.5.1) the totally synthetic 6α-aryltetracyclines (Section 1.5.2) and the

semisynthetic 6α-methylenemercaptantetracyclines (Section 1.5.3).

1.5.1 Dactylocyclines – natural product 6-epiglycotetracyclines

Dactylocyclines are tetracycline antibiotics isolated in the early 1990’s from Dactylosporangium

sp. (ATCC 53693) in a screen by Bristol-Myers Squibb for antibiotics active against tetracycline-

resistant strains.(62) Dactylocyclines are 4a-hydroxy-6-epi-6-glyco-7-chloro-8-

methoxytetracyclines (Figure 1-4). While the 7-chloro substitution is known for existing

tetracycline antibiotics on the market, the 4a, 6, and 8 substitutions are unique (Figure 1-1).

Dactylocyclines were found to be antibiotically active, although not of an exceptional potency,

with MIC values of 3.1-6.3 μg/mL against gram-positive tetracycline-resistant strains of

Straphylococcus aureus, Staphylococcus epidermis and Streptococcus faecalis. However,

dactylocyclines exhibited high MIC values (>100 μg/mL) against gram-negative tetracycline-

resistant strains.(63) Importantly, the antibiotic activity of dactylocyclines against tetracycline-

resistant strains is dependent on the glycoside group, with dactylocyclinone, the aglycone of

dactylocycline, not presenting similar antibiotic properties.(63)

18

Despite their promising antibacterial activity outlook, two main disadvantages prevented

dactylocyclines’ further development as antibiotics. For one, dactylocyclines are extremely acid

labile. Specifically, the glycoside installed on the tertiary hydroxyl group of the 6-epitetracycline

aglycone dactylocyclinone is acid labile in mildly acidic conditions.(56, 62) Secondly, the fields

of synthetic biology and metabolic engineering were not yet ripe to generate dactylocycline

analogs at the early 1990’s when dactylocyclines were discovered. Twenty years later, with higher

maturity of these fields, the pathway towards the dactylocycline aglycone, dactylocyclinone, was

successfully heterologously expressed by the Tang group.(56) Thus, today there is an outlook for

generating dacylocycline analogs that build on the advantages of the natural dactylocyclines and

overcome their disadvantages (Section 1.6).

Figure 1-4 Structures of the dactylocycline family of natural products

Adapted from (62).

1.5.2 6-demethyl-6-deoxy-6α-aryltetracyclines – totally synthetic tetracyclines

6-demethyl-6-deoxy-6α-aryltetracyclines and 6-demethyl-6-deoxy-6α-heteroaryltetracyclines

have been accessed through the Myers convergent total synthesis of tetracyclines and described in

2008.(42) Among the 6α-aryls synthesized are included, phenyl, 4-toluyl and 3,5-eifluorophenyl.

While these derivatives showed good MIC values of <1 μg/mL against gram-positive tetracycline-

19

resistant strains, they showed values of ≥8 μg/mL against gram-negative tetracycline-resistant

strains.(42) However, despite promising MIC values, when one of these derivatives, 6-demethyl-

6-deoxy-6α-phenyltetracycline was tested in vivo in mice infected with tetracycline-sensitive

gram-positive S. aureus, it was found to be much less effective than tetracycline, requiring a

concentration at least 10 times higher to achieve the same rescuing ability.(42) A possible reason

for the ineffectiveness of 6α-aryltetracyclines against gram-negative tetracycline-resistant bacteria

in vitro and against tetracycline-sensitive infections in vivo could be their high expected

lipophilicity, a topic explored in Section 1.5.3.

1.5.3 6-demethyl-6-deoxy-6α-methylenemercaptantetracyclines –

semisynthetic tetracyclines

6-demethyl-6-deoxy-6α-methylenemercaptantetracyclines are an example for the potential

importance of the polarity of 6α-substituents on the antibacterial activity of 6α-substituted 6-

demethyltetracycline analogs. 6-demethyl-6-deoxy-6α-methylenemercaptantetracyclines were

synthesized from the semisynthetic 6-methylenetetracycline and 6-methyleneoxytetracycline

(metacycline, Figure 1-1).(23)

Mercaptan derivatives of 6-methylenetetracycline with lipophilic substituents such as Ph and Bn

showed a much reduced activity relative to tetracycline against the gram-negative Klebsiella

pneumoniae (Figure 1-5-a). However, in the case of the phenyl and benzyl derivatives of 6-

methyleneoxytetracycline the reduction in activity was lower, presumably due to the mediating

hydrophilicity of the 5α-hydroxy (Figure 1-5-b). Similarly, in the case of the more polar benzyl

and phenyl sulfoxide derivatives of 6-methylenetetracycline the reduction in activity relative to

tetracycline was lower as well (Figure 1-5-c). The smaller and more polar acetyl derivatives

20

showed better activity in both mercaptan derivatives of 6-methylenetetracycline and 6-

methyleneoxytetracycline (Figure 1-5-a and Figure 1-5-b).(23)

Figure 1-5 6-demethyl-6-deoxy-6α-methylenemercaptantetracyclines and their antibiotic

activities

(a) 6-demethyl-6-deoxy-6α-methylenemercaptantetracyclines derived from 6-

methylenetetracycline, (b) 6-demethyl-6-deoxy-6α-methylenemercaptanoxytetracyclines derived

from 6-methyleneoxytetracycline (metacycline) and (c) 6-demethyl-6-deoxy-6α-

methylenemercaptantetracycline oxides derived from 6-methylenetetracycline. The numeric value

stated for each of the derivative is its normalized turbidimetric bioassay value (relative to

tetracycline) against the gram-negative Klebsiella pneumoniae.(23, 64)

Based on these results it was hypothesized that bulky and lipophilic substituents on the 6-position

reduce antibacterial activity, especially against gram-negative organisms. Importantly, however,

MIC values for the derivatives of Figure 1-5 did not follow the above mentioned trend and were

generally high, with the acetyl derivatives of 6-methyleneoxytetracycline performing better than

the others.(23) It is of interest to see whether this strategy of improving gram-negative efficacy by

reducing lipophilicity of the 6α-substituent, namely the glycoside, could be effective in the case of

6-demethyl-6-epiglycotetracyclines (Section 1.6).

21

1.6 Design of the biosynthesis of 6-demethyl-6-

epiglycotetracyclines using S. cerevisiae

Given the promise in tetracycline analogs in the 6-position to make effective antibiotics (Section

1.5) and given the limitations in existing synthetic methods to access key analogs in this position

(Section 1.4), I decided to focus my efforts on 6-position analogs. Specifically, inspired by the

natural products dactylocyclines (Section 1.5.1) I focused my efforts towards making acid stable

broad spectrum antibiotic derivatives of dactylocyclines effective against tetracycline-resistant

strains. The sections below outline the specific structural elements of the target tetracycline

analogs. Namely, making 6-demethyl derivatives for acid stability (Section 1.6.1), aiming for

broad spectrum activity using glycoside libraries (Section 1.6.2) and ensuring antibiotic efficacy

using 6-demethyl-6-epitetracyclines as opposed to 6-demethyltetracyclines (Section 1.6.3).

Finally, I describe why S. cerevisiae was chosen as the heterologous host for making these target

tetracycline analogs (Section 1.6.4).

1.6.1 Targeting 6-demethyl derivatives of 6-epiglycotetracyclines for acid

stability

To improve on the extreme acid lability of dactylocyclines, I propose making 6-demethyl-6-

epiglycotetracyclines, instead of 6-epiglycotetracyclines such as dactylocyclines. Dactylocyclines

show promising antibacterial activity against tetracycline-resistant strains. However,

dactylocyclines’ extreme acid lability prevents their development as antibiotics (Section 1.5.1). In

weak acidic conditions dactylocyclines are known to convert to their aglycone, dactylocyclinone

and thereby lose their antibiotic activity against tetracycline-resistant strains.(62, 63)

22

The extreme acid lability of dactylocyclines is likely originating in the tertiary 6α-hydroxy to

which the glycoside is bound in dactylocyclines (Figure 1-4). As Figure 1-6 shows, the acidic

hydrolysis bond splitting mechanism of the glycosidic bond is hypothesized to shift based on the

aglycone ability to form a stable carbonium ion. Thus, in aglycones that cannot form stable

carbonium ion, the oxonium ion would form on the glycoside and in aglycones that can form a

stable carbonium ion the carbonium ion would form on the aglycone. Examples for aglycones that

can form a stable carbonium ion are aglycones that are bound to the glycoside through a tertiary

or benzylic hydroxyl.(65)

As expected according to this hypothesis, the acid hydrolysis reaction rate is increased in the case

of a tert-butyl aglycone that can form a stable tertiary carbocation relative to the case of a methyl

aglycone (Table 1-2). However, the adamantyl carbocation is not similarly stabilized and

nevertheless when adamantyl is the aglycone the reaction rate is higher than in the case of other

substituents such as Me, Et and Pr. This incongruence led to the hypothesis that F-strain produced

by bulky aglycones enhances the hydrolysis rate as well.(65) Thus, in the case of the aglycone

dactylocyclinone, where the aglycone is both bulky tertiary and benzylic, it is indeed expected to

undergo facile hydrolysis in low acid concentrations.

Figure 1-6 Two possible C-O bond cleavage pathways in glycoside hydrolysis

Adapted from (65).

23

Table 1-2 Rates of hydrolysis of β-D-glycopyranosides in 0.5 M aqueous sulfuric acid

Aglycone Methyl Ethyl isopropyl tButyl Benzyl Adamantyl

Hydrolysis ratea 1 1.1 1.9 555.8 1.1 48.6

ahydroylsis rate is normalized to a methyl aglycone. The rate was calculated at 60 oC at 0.5 M

aqueous sulfuric acid or extrapolated or interpolated from data at different temperatures.(66, 67)

To produce 6-epiglycotetracycline analogs inspired by dactylocyclines but with a much increased

acid stability, I therefore propose to produce 6-demethyl-6-epiglycotetracyclines rather than 6-

epiglycotetracyclines. An example for such tetracycline derivatives would be 6-

demethyldactylocyclines. However, additional modifications are likely to be required to lead to

improved activity of 6-demethyl-6-epiglycotetracyclines against tetracycline-resistant gram-

positive and especially gram-negative strains. Such modifications are the subject of Section 1.6.2.

1.6.2 Targeting glycoside libraries on 6-demethyl-6-epiglycotetracyclines for

broad spectrum activity

For improving broad spectrum antibiotic activity against tetracycline-resistant strains I propose

installing a library of glycoside moieties on the 6-demethyl-6-epitetracycline aglycone. The

extremely limited tools in synthetic biology around the time of discovery of dactylocyclines

restricted exploring unnatural derivatives in this manner. However, throughout the intervening

years much progress has been achieved in producing diversified glycosylated natural products by

employing glycoside libraries for natural product aglycones.(68-70) Progress in this field is

fostered by the natural glycoside promiscuity of many glycosyltransferase.(68)

It was recently shown that installing sterically accessible amines on antibiotics that are only

effective against gram-positive strains can assist in transforming them into broad spectrum

antibiotics.(71) Thus, installing a library of glycosamines on 6-demethyl-6-epitetracyclines could

24

be a promising avenue to test whether tetracyclines active against gram-positive strains alone could

be transformed into broad-spectrum tetracyclines in this manner as well. Many glycosamines with

a primary amine have been previously reported in the literature and could be employed in this

glycodiversification effort.(69)

More specifically for tetracyclines, it was shown that reducing lipophilicity of 6α-substituents or

overall lipophilicity of 6-demethyl-6-deoxy-6α-methylmarcaptantetracyclines can increase their

gram-negative activity (Section 1.5.3). Importantly, dactylocyclines were noted for being

unusually more lipophilic than commercially available tetracyclines.(62) Their lipophilicity could

theoretically be stemming from any of the differentiating elements between dactylocyclines and

commercially available tetracyclines (Figure 1-1). These include a 4a-hydroxy, a 6-epi

configuration, a 7-chloro and an 8-methoxy (Section 1.5.1, Figure 1-4). Given that the 4a-hydroxy

and the 8-methoxy are unlikely to make dactylocyclines more lipophilic and given the presence of

the 7-chloro in commercially available tetracyclines (Table 1-1) it is likely the uniqueness in the

6-position that makes dactylocyclines more lipophilic. The 6-epi configuration of dactylocyclines

is unlikely to contribute to their unusually high lipophilicity relative to commercially available

tetracyclines because 6α-methyltetracyclines are generally known to be more lipophilic than 6β-

methyltetracyclines (Section 1.6.3). Thus, the 6-glycoside is the likely main contributor to

dactylocyclines lipophilicity, especially considering that the glycoside of dactylocyclines is rather

lipophilic with a 2,6-dideoxy and a 4-methoxy, leaving it with only one or zero hydroxyls (Figure

1-4). It would therefore be of interest to test whether 6-demethyl-6-epiglycotetracyclines with a

variety of less lipophilic glycosides have broad-spectrum activity against tetracycline-resistant

strains.

25

1.6.3 Targeting the 6-epi forms of 6-demethylglycotetracyclines for antibiotic

efficacy

C6 monosubstituted tetracyclines are generally more antibiotically active when substituted at the

α-position than when substituted at the β-position (Figure 1-1).(25) For example, doxycycline (6-

deoxyoxytetracycline) had lower MIC values than 6-epidoxycycline (6-epi-6-

deoxyoxytetracycline) in 7 / 7 gram-positive and gram-negative strains tested in one set of

experiments and its in vivo protective dose was lower as well.(23) Similar results, although less

pronounced, were reported for 6-deoxytetracycline versus 6-epi-6-deoxytetracycline,(23) and 6α-

fluoro-6-demethyl-6-deoxytetracycline was also shown to be more effective than its 6-epimer, 6β-

fluoro-6-demethyl-6-deoxytetracycline.

A possible explanation to the advantage 6α-tetracycline analogs have over their 6β counterparts is

that 6α-analogs more efficiently maintain the important balance between the zwitterionic and the

lipophilic forms of tetracyclines (Section 1.3.3 and Section 1.4.1). This explanation is supported

by the following known trend in lipophilicity, with lipophilicity levels ranking oxytetraycline < 6-

epi-6-deoxyoxytetracycline < 6-deoxyoxytetracycline, and with 6-epi-6-deoxyoxytetracycline

showing only a modest increase in lipophilicity relative to oxytetracycline. While the increase in

lipophilcity between oxytetracycline and its two 6-deoxy derivatives is expected based on a loss

of a hydrophilic hydroxy group, the increase in lipophilicity between the two deoxyoxytetracycline

epimers is less immediately reasoned. The commonly proposed reasoning is that a 6β-functional

group leads to steric clashes with the 4α-dimethylamino and thus destabilizes the lipophilic

conformation of the tetracycline, making the tetracycline derivative less lipophilic as a whole. In

contrast, an 6α-substiuent does not disrupt the important balance between the lipophilic and

zwitterionic forms of tetracycline derivatives.(72)

26

While these difference in lipophilicity and antibiotic activity are noted for 6α- and 6β-methyl

groups, they are all the more relevant when a much bulkier glycoside is installed on the 6-hydroxyl.

It is thus perhaps not surprising that dactylocyclines, the only known 6-glycotetrayclines natural

products, are 6-epitetracyclines while other tetracycline natural products such as oxytetracycline

and chlortetracyclines are not. Thus, 6-demethyl-6-epiglycotetracyclines are proposed in this study

and not 6-demethyl-6-glycotetracyclines. This point is of special importance given the higher

accessibility of 6β-hydroxytetracyclines over their 6α-hydroxytetracycline counterparts in all

methods of tetracycline preparation (Section 1.4.1 and Section 1.4.2 and Chapter 3).

1.6.4 S. cerevisiae as the heterologous host of choice for 6-demethyl-6-

epiglycotetracycline biosynthesis

Given that 6-demethyl-6-epiglycotetracyclines were marked here as highly desirable tetracycline

targets to synthesize and test as antibiotics (Sections 1.6.1, 1.6.2 and 1.6.3), the question arises

what would be the appropriate synthetic tool. Section 1.4.1 mentions the limitations in accessing

6-demethyl-6-epiglycotetracyclines by semisynthesis from the mainstream tetracycline

fermentation products, such as tetracycline, oxytetracycline, chlortetracycline and demeclocycline,

that are all 6β-hydroxy. Similarly, Section 1.4.2 mentions the difficulty in accessing 6-demethyl-

6-epitetracyclines by total synthesis due to the inherent limitations in the synthetic approach that

requires constructing the C-ring from AB- and D-ring precursors. Recent advances in metabolic

engineering and synthetic biology render it especially exciting to test heterologous biosynthesis

for generating 6-demethyl-6-epitetracyclines, the precursors to the desired 6-demethyl-6-

epiglycotetracyclines. Importantly, the following glycodiversification step can take place either in

vivo or by isolating 6-demethyl-6-epitetracyclines and appending activated glycosides on them

using glycosyltransferases in vitro or through chemical synthesis (Figure 1-7).

27

Figure 1-7 Retrosynthetic analysis of 6-demethyl-6-epiglycotetracyclines

Saccharomyces cerevisiae is uniquely positioned to the synthesis of 6-demethyl-6-

epiglycotetracyclines. First, as a highly genetically tractable model organism that is easy to culture

and quick to reproduce, S. cerevisiae has been demonstrated a useful heterologous host for the

production of a variety of natural products of importance to human health.(61, 73-75) The

advantages, sometimes unexpected, of performing chemistry in S. cerevisiae are also exemplified

in this study (e.g., Section 2.4.2).

Nevertheless, 6-demethyl-6-epiglycotetracyclines or 6-demethyl-6-epitetracyclines could also

theoretically be accessed from the native host Dactylosporangium sp. (ATCC 53693) or from an

alternative heterologous host, such as S. lividans K4 previously used for dactylocyclinone

biosynthesis.(56, 63) However, the native host Dactylosporangium sp. (ATCC 53693) is reported

to grow at an exceedingly slow rate and to be completely inaccessible for genetic

modifications.(56) It is yet to be determined whether an alternative heterologous host such as S.

lividans K4 has better or worse outcomes than S. cerevisiae for this biosynthesis goal.

Importantly, the Tang laboratory has reported the heterologous biosynthesis of the fungal

anhydrotetracycline TAN-1612 in S. cerevisiae (Section 1.4.3) and TAN-1612 is well positioned

to serve as a scaffold for the production of 6-demethyl-6-epiglycotetracyclines in S. cerevisiae.

TAN-1612 holds important structural similarities to anhydrodactylocyclinone, such as the 4a-

hydroxy and the 8-methoxy functional groups, that can render it a good starting point for further

derivatization. These substitutions are not observed in the anhydrotetracycline precursors of the

28

commonly fermented tetracyclines, such as chloranhydrotetracycline and anhydrotetracycline.

Also favorably to the target molecules outlined (Figure 1-7), TAN-1612 lacks a 6-methyl group

(Figure 1-8).

Figure 1-8 Structural Comparison between anhydrotetracycline, chloranhydrotetracycline,

TAN-1612 and anhydrotdactylocyclinone

In grey are marked potentially desirable functional groups TAN-1612 has relative to the

commercially available anhydrotetracycline as a scaffold for 6-demethyl-6-epiglycotetracycline

biosynthesis. The stereochemistry of TAN-1612 at positions 4a and 12a was not yet confirmed by

X-ray crystallography. Empty cells indicate implicit Hs.

However, TAN-1612 is also lacking structural features that are important for antibiotic activity in

tetracyclines in the A-ring such as the 4α-dimethylamino and the 2-carboxamido (Figure 1-8). The

furnishing of a 2-carboxamido derivative of TAN-1612 or a related molecule in S. cerevisiae could

be envisioned by heterologously expressing the relevant genes from the oxytetracycline

biosynthetic pathway of S. rimosus. An alternative pathway to harvest the relevant genes is the

biosynthetic pathway towards the fungal polyketide viridicatumtoxin, which is homologous to that

of TAN-1612. This effort would likely require evolving the TAN-1612 biosynthetic enzymes to

the unnatural substrate.(76, 77)

The Yeast three hybrid (Y3H) system for metabolic engineering of tetracycline analogs could be

crucial to assay mutants of the TAN-1612 pathway enzymes for relaxed substrate specificity

towards the new 2-carboxamido intermediates (Section 1.7.4 and Chapter 5). Likewise,

hydroxylation and oxidation of the 4-position of TAN-1612 can be envisioned by OxyE and OxyL

29

from the oxytetracycline pathway (78) evolved for TAN-1612 specificity by employing the Y3H

as well. These steps could be followed by reductive amination to furnish the 4α-dimethylamino or

a related functional group on TAN-1612 or a related derivative. Finally, fermentation of 6-

demethyl-6-epiglycotetracyclines or 6-demethyl-6-epitetracyclines in S. cerevisiae could readily

open up possibilities in the longer term for commercial production of new tetracycline

therapeutics.

1.7 Research progress and future goals in the biosynthesis of

6-demethyl-6-epiglycotetracyclines using S. cerevisiae

1.7.1 Chapter 2 – Using Saccharomyces cerevisiae for the final steps of

tetracycline biosynthesis

This chapter describes the use of Saccharomyces cerevisiae for the final steps of tetracycline

biosynthesis, setting the stage for total biosynthesis of tetracycline in S. cerevisiae. Specifically,

data presented in this chapter supports the conversion of anydrotetracycline to tetracycline or an

epimer in S. cerevisiae by heterologous expression of the 6β-hydroxylase enzyme form the

oxytetracycline pathway, OxyS. This result is surprising as it was expected that the expression of

a reductase enzyme that uses a cofactor not native to yeast would be required as well. It was also

expected that OxyS would lead to an oxytetracycline intermediate and not to tetracycline by

performing two hydroxylation steps as reported previously and not one as shown in this study. The

ability to perform the required steps for tetracycline biosynthesis from anhydrotetracycline

supports the ongoing efforts to produce 6-demethyl-6-epitetracyclines from anhydrotetracyclines

(Figure 1-7, Chapter 3 and Chapter 4).

30

1.7.2 Chapter 3 – Towards the biosynthesis of 6-demethyl-6-epitetracyclines

using S. cerevisiae

This chapter describes the work towards 6-demethyl-6-epitetracyclines biosynthesis in S.

cerevisiae. 6α-hydroxylation of a 6-demethylanhydrotetracycline by an enzyme such as DacO1 is

required for the synthesis of 6-demethyl-6-epitetracyclines as opposed to 6β-hydroxylation by

enzymes such as OxyS leading to 6-demethyltetracyclines. However, previous attempts using

multiple heterologous hosts to heterologously express DacO1 without the rest of the

dactylocyclinone pathway have not yielded stably expressed protein. The chapter describes the

successful development of a stably expressing form of DacO1 in S. cerevisiae by employing

rational design approaches for optimizing DacO1 expression. This chapter also describes the

successful hydroxylation of anhydrotetracycline by another 6α-hydroxylase candidate, PgaE. The

next step towards 6-demethyl-6-epitetracyclines is to test the stably expressed DacO1 and PgaE in

larger scale for 6α-hydroxylation of anhydrotetracycline and/or anhydrodactylocyclinone and/or

6-demethylanhydrodactylocyclinone.

1.7.3 Chapter 4 – Towards the biosynthesis of 6-demethyl-6-epitracyclines

from TAN-1612 in Saccharomyces cerevisiae

This chapter describes the work towards 6-demethyl-6-epitracycline derivatives of the fungal

anhydrotetracycline TAN-1612 in Saccharomyces cerevisiae. TAN-1612 is indicated as a unique

anhydrotetracycline scaffold for the production of 6-demethyl-6-epitetracyclines (Section 1.6.4),

the precursors to the target molecules 6-demethyl-6-epiglycotetracyclines (Figure 1-7). The

progress and future steps towards the directed evolution and biomining of a TAN-1612 6α-

hydroxylase are outlined and the isolation and characterization of a new major product in a strain

coexpressing PgaE and the TAN-1612 pathway is described.

31

1.7.4 Chapter 5 – a yeast three hybrid system for the metabolic engineering of

tetracycline derivatives

The transformation of anhydrotetracyline to tetracycline could be readily detected by a UV/Vis

based assay due to the blue-shift in the excitation and emission of the molecule (Chapters 2-4).

However, most transformations of tetracyclines and other molecules that do not involve such a

major UV/Vis shift, such as the proposed glycosylation and A-ring modifications (Section 1.6.4)

require the development of a general assay for their detection. This chapter describes the

development of a yeast three hybrid (Y3H) assay as such a general, high-throughput, versatile and

readily implemented approach for the detection of target molecule biosynthesis. The Y3H assay is

presented here as a tool for the reactions in the next steps towards the heterologous biosynthesis

of 6-demethyl-6-epiglycotetracyclines in S. cerevisiae and beyond.

32

1.8 References

1. Antibiotic Resistance Threats in the United States. US Department of Health and Human

Services, Center for Disease Control and Prevention, 2013.

2. Mullard A. Momentum builds around new antibiotic business models. Nature Reviews

Drug Discovery. 2014;13(10):711-713. doi: 10.1038/nrd4455.

3. Henssler EM, Scholz O, Lochner S, Gmeiner P, Hillen W. Structure-based design of Tet

repressor to optimize a new inducer specificity. Biochemistry. 2004;43(29):9512-9518. doi:

10.1021/bi049682j.

4. Tatsuta K. Total synthesis of the big four antibiotics and related antibiotics. The Journal of

Antibiotics. 2013;66(3):107-129. doi: 10.1038/ja.2012.126.

5. Eisen DP. Doxycycline. In: M. Lindsay Grayson SEC, Suzanne Crowe, William Hope,

James S. McCarthy, John Mills, Johan W. Mouton, David L. Paterson, editor. Kucers' The Use of

Antibiotics: A Clinical Review of Antibacterial, Antifungal, Antiparasitic, and Antiviral Drugs. 6

ed. Boca Raton, FL: CRC Press; 2010. p. 851-869.

6. Nelson ML, Levy SB. The history of the tetracyclines. Ann NY Acad Sci. 2011;1241:17-

32. doi: 10.1111/j.1749-6632.2011.06354.x.

7. Chopra I, Roberts M. Tetracycline antibiotics: mode of action, applications, molecular

biology, and epidemiology of bacterial resistance. Microbiol Mol Biol Rev. 2001;65(2):232-260.

doi: 10.1128/MMBR.65.2.232-260.2001.

8. Brodersen DE, Clemons WM, Jr., Carter AP, Morgan-Warren RJ, Wimberly BT,

Ramakrishnan V. The structural basis for the action of the antibiotics tetracycline, pactamycin,

and hygromycin B on the 30S ribosomal subunit. Cell. 2000;103(7):1143-1154. doi:

10.1016/S0092-8674(00)00216-6.

9. Speer BS, Shoemaker NB, Salyers AA. Bacterial resistance to tetracycline: mechanisms,

transfer, and clinical significance. Clin Microbiol Rev. 1992;5(4):387-399. doi:

10.1128/CMR.5.4.387.

33

10. Connell SR, Tracz DM, Nierhaus KH, Taylor DE. Ribosomal protection proteins and their

mechanism of tetracycline resistance. Antimicrob Agents Chemother. 2003;47(12):3675-3681.

doi: 10.1128/AAC.47.12.3675-3681.2003.

11. Gerrits MM, de Zoete MR, Arents NL, Kuipers EJ, Kusters JG. 16S rRNA mutation-

mediated tetracycline resistance in Helicobacter pylori. Antimicrob Agents Chemother.

2002;46(9):2996-3000. doi: 10.1128/AAC.46.9.2996-3000.2002.

12. Speer BS, Salyers AA. Novel aerobic tetracycline resistance gene that chemically modifies

tetracycline. J Bacteriol. 1989;171(1):148-153.

13. Toon S, Rowland M. Quantitative Structure Pharmacokinetic Activity Relationships with

Some Tetracyclines. J Pharm Pharmacol. 1979;31(S1):43P-43P. doi: 10.1111/j.2042-

7158.1979.tb11591.x.

14. Griffin MO, Fricovsky E, Ceballos G, Villarreal F. Tetracyclines: a pleitropic family of

compounds with promising therapeutic properties. Review of the literature. AJP: Cell Physiology.

2010;299(3):C539-C548. doi: 10.1152/ajpcell.00047.2010.

15. Garrido-Mesa N, Zarzuelo A, Gálvez J. Minocycline: far beyond an antibiotic. Br J

Pharmacol. 2013;169(2):337-352. doi: 10.1111/bph.12139.

16. Jenner L, Starosta AL, Terry DS, Mikolajka A, Filonava L, Yusupov M, Blanchard SC,

Wilson DN, Yusupova G. Structural basis for potent inhibitory activity of the antibiotic tigecycline

during protein synthesis. Proceedings of the National Academy of Sciences. 2013;110(10):3812-

3816. doi: 10.1073/pnas.1216691110.

17. Prasad P, Sun J, Danner RL, Natanson C. Excess Deaths Associated With Tigecycline

After Approval Based on Noninferiority Trials. Clin Infect Dis. 2012;54(12):1699-1709. doi:

10.1093/cid/cis270.

18. Liu F, Myers AG. Development of a platform for the discovery and practical synthesis of

new tetracycline antibiotics. Curr Opin Chem Biol. 2016;32:48-57. doi:

10.1016/j.cbpa.2016.03.011.

19. Nelson ML, Levy SB. 3.25 - Tetracyclines and Tetracycline Derivatives. In: Moo-Young

M, editor. Comprehensive Biotechnology (Second Edition). Burlington: Academic Press; 2011. p.

269-283.

34

20. Approved drug products with therapeutic equivalence evaluations (Orange book). U.S.

Department of Health and Human Services, 2019.

21. Butler MS, Blaskovich MA, Cooper MA. Antibiotics in the clinical pipeline in 2013. J

Antibiot. 2013;66(10):571-591. doi: 10.1038/ja.2013.86.

22. Honeyman L, Ismail M, Nelson ML, Bhatia B, Bowser TE, Chen J, Mechiche R, Ohemeng

K, Verma AK, Cannon EP, Macone A, Tanaka SK, Levy S. Structure-Activity Relationship of the

Aminomethylcyclines and the Discovery of Omadacycline. Antimicrob Agents Chemother.

2015;59(11):7044-7053. doi: 10.1128/aac.01536-15.

23. Blackwood RK, English AR. Structure–Activity Relationships in the Tetracycline Series.

In: Perlman D, editor. Adv Appl Microbiol: Academic Press; 1970. p. 237-266.

24. Khosla C, Tang Y. A new route to designer antibiotics. Science. 2005;308(5720):367-368.

doi: 10.1126/science.1111415.

25. Rogalski W. Chemical Modification of the Tetracyclines. In: Hlavka JJ, Boothe JH, editors.

The Tetracyclines. Berlin, Heidelberg: Springer Berlin Heidelberg; 1985. p. 179-316.

26. Rasmussen B, Noller HF, Daubresse G, Oliva B, Misulovin Z, Rothstein DM, Ellestad GA,

Gluzman Y, Tally FP, Chopra I. Molecular basis of tetracycline action: identification of analogs

whose primary target is not the bacterial ribosome. Antimicrob Agents Chemother.

1991;35(11):2306-2311. doi: 10.1128/AAC.35.11.2306.

27. Hošťálek Z, Vaněk Z. Biosynthesis of the Tetracyclines. In: Hlavka JJ, Boothe JH, editors.

The Tetracyclines. Berlin, Heidelberg: Springer Berlin Heidelberg; 1985. p. 137-178.

28. Sum PE, Petersen P. Synthesis and structure-activity relationship of novel glycylcycline

derivatives leading to the discovery of GAR-936. Bioorg Med Chem Lett. 1999;9(10):1459-1462.

doi: 10.1016/S0960-894X(99)00216-4.

29. Chopra I. New developments in tetracycline antibiotics: glycylcyclines and tetracycline

efflux pump inhibitors. Drug Resistance Updates. 2002;5(3-4):119-125. doi: 10.1016/s1368-

7646(02)00051-1.

30. Zhanel GG, Homenuik K, Nichol K, Noreddin A, Vercaigne L, Embil J, Gin A, Karlowsky

JA, Hoban DJ. The Glycylcyclines. Drugs. 2004;64(1):63-88. doi: 10.2165/00003495-200464010-

00005.

35

31. Wright PM, Seiple IB, Myers AG. The Evolving Role of Chemical Synthesis in

Antibacterial Drug Discovery. Angew Chem Int Ed. 2014;53(34):8840-8869. doi:

10.1002/anie.201310843.

32. Esse RC, Lowery JA, Tamorria CR, Sieger GM. Tetracycloxides. II. Transformations at

the C-4 Position. J Am Chem Soc. 1964;86(18):3875-3877. doi: 10.1021/ja01072a057.

33. Prewo R, Stezowski JJ. Chemical-structural properties of tetracycline derivatives. 3. The

integrity of the conformation of the nonionized free base. J Am Chem Soc. 1977;99(4):1117-1121.

doi: 10.1021/ja00446a024.

34. Mitscher LA. Degradation and Structure Proofs. The Chemistry of the Tetracycline

Antiobitcs. New York: Marcel Dekker, Inc.; 1978. p. 122-164.

35. Sum PE, Lee VJ, Testa RT, Hlavka JJ, Ellestad GA, Bloom JD, Gluzman Y, Tally FP.

Glycylcyclines .1. A New-Generation of Potent Antibacterial Agents through Modification of 9-

Aminotetracyclines. J Med Chem. 1994;37(1):184-188. doi: 10.1021/jm00027a023.

36. Tanaka SK, Steenbergen J, Villano S. Discovery, pharmacology, and clinical profile of

omadacycline, a novel aminomethylcycline antibiotic. Biorg Med Chem. 2016;24(24):6409-6419.

doi: 10.1016/j.bmc.2016.07.029.

37. Conover LH, Butler K, Johnston JD, Korst JJ, Woodward RB. The Total Synthesis of 6-

Demethyl-6-Deoxytetracycline. J Am Chem Soc. 1962;84(16):3222-3224. doi:

10.1021/ja00875a063.

38. Chopra I. Tetracycline analogs whose primary target is not the bacterial ribosome.

Antimicrob Agents Chemother. 1994;38(4):637-640. doi: 10.1128/aac.38.4.637.

39. Wasserman HH, Lu TJ, Scott IA. The total synthesis of tetracycline. J Am Chem Soc.

1986;108(14):4237-4238. doi: 10.1021/ja00274a072.

40. Stork G, La Clair JJ, Spargo P, Nargund RP, Totah N. Stereocontrolled Synthesis of (±)-

12a-Deoxytetracycline. J Am Chem Soc. 1996;118(22):5304-5305. doi: 10.1021/ja960434n.

41. Charest MG, Lerner CD, Brubaker JD, Siegel DR, Myers AG. A convergent

enantioselective route to structurally diverse 6-deoxytetracycline antibiotics. Science.

2005;308(5720):395-398. doi: 10.1126/science.1109755.

36

42. Sun C, Wang Q, Brubaker JD, Wright PM, Lerner CD, Noson K, Charest M, Siegel DR,

Wang Y-M, Myers AG. A Robust Platform for the Synthesis of New Tetracycline Antibiotics. J

Am Chem Soc. 2008;130(52):17913-17927. doi: 10.1021/ja806629e.

43. Clark RB, Hunt DK, He M, Achorn C, Chen C-L, Deng Y, Fyfe C, Grossman TH, Hogan

PC, O’Brien WJ, Plamondon L, Rönn M, Sutcliffe JA, Zhu Z, Xiao X-Y. Fluorocyclines. 2.

Optimization of the C-9 Side-Chain for Antibacterial Activity and Oral Efficacy. J Med Chem.

2012;55(2):606-622. doi: 10.1021/jm201467r.

44. Xiao X-Y, Hunt DK, Zhou J, Clark RB, Dunwoody N, Fyfe C, Grossman TH, O’Brien WJ,

Plamondon L, Rönn M, Sun C, Zhang W-Y, Sutcliffe JA. Fluorocyclines. 1. 7-Fluoro-9-

pyrrolidinoacetamido-6-demethyl-6-deoxytetracycline: A Potent, Broad Spectrum Antibacterial

Agent. J Med Chem. 2012;55(2):597-605. doi: 10.1021/jm201465w.

45. Liu F, Wright PM, Myers AG. Diastereoselective Michael–Claisen Cyclizations of γ-Oxa-

α,β-unsaturated Ketones en Route to 5-Oxatetracyclines. Org Lett. 2016;19(1):206-209. doi:

10.1021/acs.orglett.6b03491.

46. Wright PM, Myers AG. Methodological advances permit the stereocontrolled construction

of diverse fully synthetic tetracyclines containing an all-carbon quaternary center at position C5a.

Tetrahedron. 2011;67(51):9853-9869. doi: 10.1016/j.tet.2011.09.143.

47. Myers AG, Siegel DR, Buzard DJ, Charest MG. Synthesis of a Broad Array of Highly

Functionalized, Enantiomerically Pure Cyclohexanecarboxylic Acid Derivatives by Microbial

Dihydroxylation of Benzoic Acid and Subsequent Oxidative and Rearrangement Reactions. Org

Lett. 2001;3(18):2923-2926. doi: 10.1021/ol010151m.

48. Charest MG, Siegel DR, Myers AG. Synthesis of (−)-Tetracycline. J Am Chem Soc.

2005;127(23):8292-8293. doi: 10.1021/ja052151d.

49. Woolston BM, Edgar S, Stephanopoulos G. Metabolic engineering: past and future. Annu

Rev Chem Biomol Eng. 2013;4:259-288. doi: 10.1146/annurev-chembioeng-061312-103312.

50. Jiang M, Stephanopoulos G, Pfeifer BA. Downstream reactions and engineering in the

microbially reconstituted pathway for Taxol. Appl Microbiol Biotechnol. 2012;94(4):841-849.

doi: 10.1007/s00253-012-4016-1.

51. Pfeifer BA. Biosynthesis of Complex Polyketides in a Metabolically Engineered Strain of

E. coli. Science. 2001;291(5509):1790-1792. doi: 10.1126/science.1058092.

37

52. Ma SM, Li JWH, Choi JW, Zhou H, Lee KKM, Moorthie VA, Xie X, Kealey JT, Da Silva

NA, Vederas JC, Tang Y. Complete Reconstitution of a Highly Reducing Iterative Polyketide

Synthase. Science. 2009;326(5952):589-592. doi: 10.1126/science.1175602.

53. Fasciotti M. Perspectives for the use of biotechnology in green chemistry applied to

biopolymers, fuels and organic synthesis: from concepts to a critical point of view. Sustainable

Chem Pharm. 2017;6:82-89. doi: 10.1016/j.scp.2017.09.002.

54. Keasling JD. Manufacturing Molecules Through Metabolic Engineering. Science.

2010;330(6009):1355-1358. doi: 10.1126/science.1193990.

55. Tsoi CJ, Khosla C. Combinatorial biosynthesis of 'unnatural' natural products: the

polyketide example. Chem Biol. 1995;2(6):355-362. doi: 10.1016/1074-5521(95)90214-7.

56. Wang P, Kim W, Pickens LB, Gao X, Tang Y. Heterologous Expression and Manipulation

of Three Tetracycline Biosynthetic Pathways. Angewandte Chemie-International Edition.

2012;51(44):11136-11140. doi: 10.1002/anie.201205426.

57. Feng Z, Kallifidas D, Brady SF. Functional analysis of environmental DNA-derived type

II polyketide synthases reveals structurally diverse secondary metabolites. Proc Natl Acad Sci U

S A. 2011;108(31):12629-12634. doi: 10.1073/pnas.1103921108.

58. Goodman JJ. Fermentation and Mutational Development of the Tetracyclines. In: Hlavka

JJ, Boothe JH, editors. The Tetracyclines. Berlin, Heidelberg: Springer Berlin Heidelberg; 1985.

p. 5-57.

59. Mccormick JRD, Sjolander NO, Hirsch U, Jensen ER, Doerschuk AP. A New Family of

Antibiotics - the Demethyl-Tetracyclines. J Am Chem Soc. 1957;79(16):4561-4563. doi:

10.1021/ja01573a089.

60. Li YR, Chooi YH, Sheng YW, Valentine JS, Tang Y. Comparative Characterization of

Fungal Anthracenone and Naphthacenedione Biosynthetic Pathways Reveals an alpha-

Hydroxylation-Dependent Claisen-like Cyclization Catalyzed by a Dimanganese Thioesterase. J

Am Chem Soc. 2011;133(39):15773-15785. doi: 10.1021/ja206906d.

61. Ro DK, Paradise EM, Ouellet M, Fisher KJ, Newman KL, Ndungu JM, Ho KA, Eachus

RA, Ham TS, Kirby J, Chang MC, Withers ST, Shiba Y, Sarpong R, Keasling JD. Production of

the antimalarial drug precursor artemisinic acid in engineered yeast. Nature. 2006;440(7086):940-

943. doi: 10.1038/nature04640.

38

62. Tymiak AA, Aklonis C, Bolgar MS, Kahle AD, Kirsch DR, O'Sullivan J, Porubcan MA,

Principe P, Trejo WH. Dactylocyclines: novel tetracycline glycosides active against tetracycline-

resistant bacteria. J Org Chem. 1993;58(3):535-537. doi: 10.1021/jo00055a002.

63. Wells JS, O'Sullivan J, Aklonis C, Ax HA, Tymiak AA, Kirsch DR, Trejo WH, Principe

P. Dactylocyclines, novel tetracycline derivatives produced by a Dactylosporangium sp. I.

Taxonomy, production, isolation and biological activity. J Antibiot. 1992;45(12):1892-1898. doi:

10.7164/antibiotics.45.1892.

64. Kersey RC. A Turbidimetric Assay for Terramycin. Journal of the American

Pharmaceutical Association (Scientific ed). 1950;39(5):252-253. doi: 10.1002/jps.3030390503.

65. Bochkov AF, Zaikov GE. Cleavage of O-glycosidic Bonds. Chemistry of the O-glycosidic

bond: Pergamon Press; 1979. p. 177-201.

66. Timell TE. The Acid Hydrolysis of Glycosides: I. General Conditions and the Effect of the

Nature of the Aglycone. Can J Chem. 1964;42(6):1456-1472. doi: 10.1139/v64-221.

67. Cocker D, Jukes LE, Sinnott ML. Alkyl–oxygen versus glycosyl–oxygen fission in the

acid-catalysed hydrolyses of some alkyl β-D-glycopyranosides. J Chem Soc, Perkin Trans 2.

1973(2):190-194. doi: 10.1039/p29730000190.

68. Thibodeaux CJ, Melançon CE, Liu H-w. Natural-Product Sugar Biosynthesis and

Enzymatic Glycodiversification. Angew Chem Int Ed. 2008;47(51):9814-9859. doi:

10.1002/anie.200801204.

69. Elshahawi SI, Shaaban KA, Kharel MK, Thorson JS. A comprehensive review of

glycosylated bacterial natural products. Chem Soc Rev. 2015;44(21):7591-7697. doi:

10.1039/c4cs00426d.

70. Jiang M, Zhang H, Park S-H, Li Y, Pfeifer BA. Deoxysugar pathway interchange for

erythromycin analogues heterologously produced through Escherichia coli. Metab Eng.

2013;20:92-100. doi: 10.1016/j.ymben.2013.09.005.

71. Richter MF, Drown BS, Riley AP, Garcia A, Shirai T, Svec RL, Hergenrother PJ.

Predictive compound accumulation rules yield a broad-spectrum antibiotic. Nature. 2017. doi:

10.1038/nature22308.

39

72. Stezowski JJ. Chemical-structural properties of tetracycline derivatives. 1. Molecular

structure and conformation of the free base derivatives. J Am Chem Soc. 1976;98(19):6012-6018.

doi: 10.1021/ja00435a039.

73. Galanie S, Thodey K, Trenchard IJ, Filsinger Interrante M, Smolke CD. Complete

biosynthesis of opioids in yeast. Science. 2015;349(6252):1095-1100. doi:

10.1126/science.aac9373.

74. Awan AR, Blount BA, Bell DJ, Shaw WM, Ho JCH, McKiernan RM, Ellis T. Biosynthesis

of the antibiotic nonribosomal peptide penicillin in baker’s yeast. Nature Communications. 2017;8.

doi: 10.1038/ncomms15202.

75. Luo X, Reiter MA, d’Espaux L, Wong J, Denby CM, Lechner A, Zhang Y, Grzybowski

AT, Harth S, Lin W, Lee H, Yu C, Shin J, Deng K, Benites VT, Wang G, Baidoo EEK, Chen Y,

Dev I, Petzold CJ, Keasling JD. Complete biosynthesis of cannabinoids and their unnatural

analogues in yeast. Nature. 2019;567(7746):123-126. doi: 10.1038/s41586-019-0978-9.

76. Lesnik U, Lukezic T, Podgorsek A, Horvat J, Polak T, Sala M, Jenko B, Harmrolfs K,

Ocampo-Sosa A, Martinez-Martinez L, Herron PR, Fujs S, Kosec G, Hunter IS, Muller R, Petkovic

H. Construction of a New Class of Tetracycline Lead Structures with Potent Antibacterial Activity

through Biosynthetic Engineering. Angewandte Chemie-International Edition. 2015;54(13):3937-

3940. doi: 10.1002/anie.201411028.

77. Chooi YH, Cacho R, Tang Y. Identification of the viridicatumtoxin and griseofulvin gene

clusters from Penicillium aethiopicum. Chem Biol. 2010;17(5):483-494. doi:

10.1016/j.chembiol.2010.03.015.

78. Pickens LB, Tang Y. Oxytetracycline biosynthesis. J Biol Chem. 2010;285(36):27509-

27515. doi: 10.1074/jbc.R110.130419.

40

41

2 Using Saccharomyces cerevisiae for the final steps of

tetracycline biosynthesis

*The contents of this chapter will be published in:

E. Herbst, V. W. Cornish, et al “Using Saccharomyces cerevisiae for the final steps of tetracycline

biosynthesis”, in preparation.

42

2.1 Chapter outline

This chapter describes the use of Saccharomyces cerevisiae for the final steps of tetracycline

biosynthesis, setting the stage for total biosynthesis of tetracycline in S. cerevisiae. Key results

described in this chapter are that OxyS performs in S. cerevisiae just one hydroxylation step as

opposed to two performed in vitro, thus enabling the biosynthesis of tetracycline instead of

oxytetracycline (Section 2.4.1 and 2.5.1). In addition, this chapter describes the reduction of the

hydroxylation product of OxyS in yeast cell lysate, without the need for heterologous expression

of a dedicated reduction enzyme, such as OxyR (Section 2.4.2). This result is important as OxyR

uses a cofactor not native to yeast. Thus, circumventing the need to express OxyR also circumvents

the need to heterologously express the biosynthetic pathway of the cofactor or to extragenously

supply it or its equivalent (Sections 2.4.2 and 2.5.2). Finally, this chapter describes mass

spectrometry and UV/Vis results supporting the production of 5a(11a)-dehydrotetracycline using

S. cerevsiae even though its further hydroxylation and/or degradation hampered previous analysis

when produced by OxyS in vitro and in vivo (Sections 2.4.2 and 2.5.2).

2.2 Introduction

S. cerevisiae has proven itself a useful heterologous host for the production of a variety of natural

products of importance to human health. A key example is the production of artemisinic acid, a

key intermediate in the production of artemisinin,(1) with more recent examples being the

production of hydrocodone, Δ9-tetrahydrocannabinolic acid (THCA) and penicillin.(2-4) One of

the promises in natural product biosynthesis using S. cerevisiae is the access to unnatural analogs

of natural products.(3) Tetracyclines are one of the most important classes of antibiotics and

tetracycline analogs are needed to combat the resistance to current tetracycline antibiotics on the

43

market given the current and impending antibiotic crisis (Chapter 1). Thus, from an applied science

perspective, research on heterologous biosynthesis of tetracyclines is important for its potential to

lead to new and effective tetracycline antibiotics. From a pure science perspective, heterologous

biosynthesis of is often highly instructive for the study of the biosynthetic steps that take place in

the natural hosts, as is exemplified throughout this chapter.

Granted, Heterologous biosynthesis of oxytetracycline was already shown in Streptomyces

lividans K4‐11. This is a more readily achievable feat given the existence in that host of a

biosynthetic pathway to cofactor F420, a required cofactor in the biosynthesis of tetracyclines that

is not native to S. cerevisiae, and the capacity of bacterial heterologous hosts to express whole

gene clusters from other bacteria.(5) However, its eukaryotic family lineage is also a key advantage

for S. cerevisiae as a heterologous host to bacterial natural products such as tetracycline.

Expression in S. cerevisiae allows orthogonal and complementary chemistry to be performed,

giving access to analogs that are otherwise inaccessible in heterologous expression in bacterial

hosts.(6) Examples for the rich, useful, and sometimes unexpected chemical possibilities of S.

cerevisiae in the heterologous biosynthesis of bacterial natural products such as tetracycline are

demonstrated in this chapter (2.4.2 and 2.5.2).

2.3 Materials and methods

See Chapter 5 for general methods.

2.3.1 General protocol for hydroxylation/reduction assay in cell lysate

Fresh patches of strains harboring the plasmid for the hydroxylation and/or reduction enzyme and

control strains were inoculated in 5 mL selective media (U- or HU-) in 15 mL culture tubes

(Corning 352059) and placed in shaker overnight to OD600 2-3. Overnight cultures were used to

inoculate 100 mL selective media (U- or HU-) cultures in 500 mL conical flasks with a starting OD

44

of 0.01-0.05. Cells were grown to final OD of 0.6-0.8 before pelleting in 2 50 mL tubes (Corning

352098) at 4 °C, 4,000 rpm for 20 min. Each pellet was redissolved in 0.5 mL H2O and the

suspension was distributed into two presterilized 1.5 mL Eppendorf tubes and pelleted at 14,000

rpm for 10 min at 4 °C. Pellets were stored at -20 °C prior to further use.

Pellets were weighed and thawn on ice. A 99:1 mixture of Y-PER yeast protein extraction reagent

(ThermoFisher Scientific 78991) and HALT protease inhibitor cocktail (ThermoFisher Scientific

PI87786) was added in a ratio of 3 μL mixture per mg pellet and placed on orbital shaker for 20

min at r.t., followed by 10 min centrifugation at 14,000 rpm at 4 °C and the cell lysate was

transferred to a new 1.5 mL Eppendorf tube, kept on ice and used within 1 h.

The cell lysate (80 μL) was added as the last component to a 4 mL vial (Chemglass CG-4900-01)

containing 280 μL of 143 mM Tris (pH 7.45), 7.7 mM anhydrotetracycline HCl (AdipoGen CDX-

A0197-M500), 4.3 mM NADPH tetrasodium hydrate (Sigma-Aldrich N7505), 26.4 mM

mercaptoehtanol and 14.285 mM glucose-6-phosphate (in experiments labeled +G6P, 0 mM

glucose-6-phophsate in all other experiments) and 40 μL glucose (278 mM) for final

concentrations of 100 mM Tris, 5.4 mM anhydrotetracycline HCl, 3 mM NADPH, 18.5 mM

mercaptoethanol, 0 or 10 mM G6P as indicated and 27.8 mM glucose. A septum was placed on

top of the vial through which a needle was inserted to allow air exchange and the reaction was left

at r.t. overnight. After night, 1 mL of MeOH was added, the contents were mixed and the reaction

was filtered through a PTFE 0.2 μm filter (Acrodisc 4423) prior to analysis by mass and UV/VIS

spectrometry.

2.3.2 Protocol for hydroxylation/reduction assay in whole cells

Fresh patches of strains harboring the plasmid for the hydroxylation and/or reduction enzyme and

control strains were inoculated in 5 mL selective media (U- or HU-) in 15 mL culture tubes

45

(Corning 352059) and placed in shaker overnight to OD600 2-3. Overnight cultures were used to

inoculate 100 mL selective media (U- or HU-) cultures in 500 mL conical flasks with a starting OD

of 0.08. Cells were grown to an OD of 1.3 before placing at 15 °C for an additional 10 h until a

final OD of 1.65. Cells were then pelleted in 2 50 mL tubes (Corning 352098) at 10 °C, 3,500 rpm

for 5 min. Each pellet was redissolved in 0.5 mL H2O and the suspension was distributed into a

presterilized 1.5 mL Eppendorf tube and pelleted at 11,000 rpm for 3 min at 10 °C. Pellets were

placed on ice and used within 1 h.

Pellets from 50 mL culture were redissolved in H2O (1,025 μL) and added as the last component

to 15 mL culture tubes (Corning 352059) containing 1,100 μL of 8 mg/mL anhydrotetracycline

HCl, 125 μL glucose solution in H2O (40%) and 250 μL 1 M Tris buffer pH 7.45 and were placed

in shaker at 350 rpm at 21 °C for 27 h. Cultures were then pelleted and the supernatant was diluted

10X into H2O before being used for UV/VIS spectral measurements.

2.4 Results

2.4.1 6β-hydroxylation of anhydrotetracycline in S. cerevisiae

The first step required to convert anhydrotetracyclines to tetracyclines is the 6-hydroxylation of

the anhydrotetracyclines. This step was studied in the oxytetracycline biosynthetic pathway and it

was shown that anhydrotetracycline 6-hydroxylation is catalyzed by OxyS. A similar step is

hypothesized to be catalyzed on 7-chloroanhydrotetracycline by the homologous enzyme Cts8 of

the chlortetracycline pathway.(7) As tetracycline is biosynthesized in 7-chlorotetracycline

producers with reduced chlorination ability,(8) anhydrotetracycline hydroxylation is likely to be

the first in the final sequence of steps in tetracycline biosynthesis.

To test the capacity of Saccharomyces cerevisiae to hydroxylate an anhydrotetracycline, the model

hydroxylating enzyme OxyS was used together with its native substrate, anhydrotetracycline

46

(Scheme 2-1). This enzyme-substrate pair was chosen as OxyS was shown to functionally express

in Escherichia coli by the Tang laboratory and anhydrotetracycline is commercially available.(7)

The expression plasmid pSP-G1 was chosen in order to facilitate the coexpression of a reductase

enzyme, to append antibody tags to both hydroxylase and reductase and to constitutively express

both enzymes.(9) OxyS was cloned into pSP-G1 under the transcriptional control of the strong

constitutive promoter TEF1 with a FLAG antibody tag at its C-terminus. The resulting plasmid

(AL-1-101) was transformed into FY251 and BJ5464-NpgA to generate strains EH-3-98-6 and

EH-3-248-1, respectively.

Scheme 2-1 Two-step enzymatic conversion of anhydrotetracycline to tetracycline

The OxyS-catalyzed reaction to convert anhydrotetracycline to 5a(11a)-dehydrotetracycline in S.

cerevisiae is supported by mass spectrometry. When a cell lysate expressing OxyS is added to

anhydrotetracycline the molecular ion corresponding to 5a(11a)-dehydrotetracycline ([M+H]+) has

much higher ion counts compared to the molecular ion corresponding to anhydrotetracycline

([M+H]+). However, the opposite is the case when a cell lysate not expressing OxyS is added to

anhydrotetracycline (Figure 2-1).

47

Figure 2-1 Mass spectrometry analysis of anhydrotetracycline hydroxylation in cell lysate

expressing OxyS

Cell lysate of OxyS expressing strain EH-3-248-1 (+OxyS) or a no-hydroxylase control EH-3-

248-8 (-OxyS) was placed overnight in Tris buffer (100 mM, pH 7.45) containing

48

anhydrotetracycline (5.4 mM), glucose (27.8 mM), NADPH (3 mM) and mercaptoethanol (18.5

mM). After night, MeOH was added, the contents were mixed and the reaction was filtered prior

to MS analysis.

Following this encouraging result with a cell lysate, the ability of OxyS to hydroxylate

anhydrotetracycline was tested in whole cells as well. As for the cell lysate, the molecular ion

corresponding to 5a(11a)-dehydrotetracycline had much higher ion counts in the +OxyS sample

relative to the -OxyS sample (data not shown). Given that anhydrotetracycline absorbs and

fluoresces in the visible range, it was logical to test whether the reaction could be monitored using

a simple UV/Vis assay using a spectrophotometer. Indeed, reducing the -OxyS spectrum from the

+OxyS spectrum shows a reduction in the 440 nm absorption and 570 nm emission peaks

corresponding to anhydrotetracycline and a formation of 380 nm absorption and 500 nm emission

peaks (Figure 2-2).

The blue shift in absorption and emission is supportive for the formation of 5a(11a)-

dehydrotetracycline from anhydrotetracycline as anhydrotetracycline’s conjugation in the CD-

rings is expected to be significantly reduced by 6-hydroxylation (Scheme 2-1). Indeed, the

corresponding red shift in the absorption spectrum for the opposite transformation, where

chlortetracycline is converted to anhydrochlortetracycline was previously reported (λmax = 373

nm to 438 nm, respectively).(10)

49

Figure 2-2 UV/Vis spectroscopy analysis of the reaction of anhydrotetracycline in whole cells

expressing OxyS

The spectra shown are Δ spectra, that is, the absorption/emission values for the OxyS expressing

cells EH-98-6 minus the absorption/emission values of the no hydroxylase control EH-3-80-3 are

shown.

50

2.4.2 5a(11a)-dehydrotetracycline reduction in S. cerevisiae

The second step in the synthesis of tetracycline from anhydrotetracycline is to reduce the 5a(11a)

α,β-unsaturated double bond (Scheme 2-1). The reduction at the 5a(11a) bond is known to be key

to tetracyclines’ antibiotic activity, with 7-chlorotetracycline being over 20X more potent than 7-

chloro-5a(11a)-dehydrotetracycline against Staphylococcus aureus.(11) In order to execute the

reduction step OxyR, the reductase of 5a(11a)-dehydrooxytetracycline,(7) was placed under the

control of a PGK1 promoter in the pSP-G1-OxyS plasmid AL-1-101 to generate AL-1-119-A-C7.

To function properly, OxyR requires cofactor F420, a cofactor that is not native to S. cerevisiae.

Cofactor Fo is an intermediate in cofactor F420 biosynthesis and is known to successfully replace

cofactor F420 in several F420 dependent enzymes with similar Km and Kcat values.(12-15) Given that

Fo synthases, employing known yeast metabolites to biosynthesize Fo were previously

characterized, an attempt was made to heterologousy produce Fo in S. cerevisiae. Fo synthase from

Thermobifida fusca and Chlamydomonas reinhardtii, was placed under the control of promoter

pADH1 to generate plasmids AL-1-183-C and AL-1-183-D, respectively.(16, 17) However,

following transformations of the plasmids to S. cerevisiae no Fo biosynthesis was confirmed by

MS (data not shown).

In order to test whether OxyR could be functional in S. cerevisiae with Fo as a cofactor, synthetic

Fo,(18) a generous gift from Frank Foss, was exogenously added to a yeast strain expressing OxyS,

OxyR and an Fo reductase. Fo reductase was placed under the control of pGPD (pTDH3) on PRS413 to

generate AL-215-D-C1, AL-255-C1 and AL-235-C5, encoding F420 reductase from Mycobacterium

tuberculosis, Archeoglobus fulgidus and Streptomyces griseus, respectively.(19-22) The reaction

mixture in Tris buffer (pH 7.45) also contained anhydrotetracycline, glucose, NADPH, and in the

51

case of Fo reductase from M. tuberculosis, glucose-6-phosphate for Fo reduction. It was found that

the levels of the molecular ion peak corresponding to tetracycline, the reduction product of

5a(11a)-dehydrotetracycline, are increased when glucose-6-phosphate is added (Figure 2-3).

Contrary to the expectation that glucose-6-phosphate serves as a substrate for the Fo reductase, it

was found that neither Fo, the Fo reductase or OxyR increased the levels of the molecular ion peak

corresponding to tetracycline either in the presence or absence of glucose-6-phosphate (data not

shown).

52

Figure 2-3 Mass spectrometry of anhydrotetracycline hydroxylation and reduction in cell

lysate expressing OxyS in the presence of G6P

Cell lysate of OxyS expressing strain EH-3-248-1 was placed overnight in Tris buffer (100 mM,

pH 7.45) containing anhydrotetracycline (5.4 mM), glucose (27.8 mM), NADPH (3 mM), 10 mM

glucose-6-phosphate (+G6P) or 0 mM glucose-6-phosphate (-G6P) and mercaptoethanol (18.5

mM).

53

To test whether a native reductase to S. cerevisiae is reducing 5a(11a)-dehydrotetracycline, the

native old yellow enzyme (OYE) reductases, OYE2 and OYE3 from S. cerevisiae were expressed

under the strong promoter pPGK1, along with their fungal homolog OYE1 from Saccharomyces

carlsbergensis.(23-26) OYE proteins are known to reduce the olefinic bond in α,β-unsaturated

carbonyls, such as the reduction of 2-cyclohexenone to cyclohexanone and are known to have a

flexible substrate specificity.(23) However, coexpression of the OYE proteins together with OxyS

did not enhance the reduction compared to the expression of OxyS alone (data not shown).

2.5 Discussion

2.5.1 6β-hydroxylation of anhydrotetracycline in S. cerevisiae

When OxyS and OxyR were used in vitro, oxytetracycline (5-hydroxytetracycline) and not

tetracycline was formed as the major product, indicating that OxyS is catalyzing both

hydroxylation steps at C5 and C6 prior to OxyR reduction of the 5a(11a) double bond (Scheme

2-1).(7) Interestingly however, when expressed in S. cerevisiae, both in cell lysate and whole cells,

OxyS is catalyzing just one hydroxylation step (Figure 2-1 and data not shown). It could be that

the absence of the 5-hydroxyl is the reason for the lack of contribution of OxyR to reduction as

OxyR’s native substrate is the doubly hydroxylated 5a(11a)-dehydrooxytetracycline and not the

singly hydroxylated 5a(11a)-dehydrotetracycline. Given that DacO4 and CtcR are known to

hydroxylate 5a(11a)-dehydrotetracycline,(7) an attempt of the hydroxylase-reductase pairs OxyS-

DacO4 and OxyS-CtcR is warranted in S. cerevisiae where OxyS performs only a single

hydroxylation step.

Furthermore, previously in the absence of OxyR the reaction product of OxyS rapidly degraded

both in vitro and in vivo in ΔoxyR Streptomyces lividans.(7) However, in the S. cerevisiae

54

expression system both in whole cells and in cell lysate the hydroxylation product of OxyS was

analyzable (Figure 2-1, Figure 2-2). This could be explained by the additional degradation

pathways possible due to the presence of the 5-hydroxy. For example, anhydrooxytetracycline (5-

hydroxyanhydrotetracycline) is known to undergo B-ring scission degradation that is not possible

in anhydrotetracycline.(27)

The ability to observe the hydroxylated product of anhydrotetracycline could also be because in

this study prior to mass spectrometry analysis MeOH added and the samples were filtered, while

in the in vitro study of OxyS acidic organic extraction using 1% AcOH in EtOAc was employed.(7)

Anhydrooxytetracycline is known to undergo B-ring scission specifically in the presence of dilute

acid.(27).

Importantly, it could be that some double hydroxylation product is formed in the reactions of the

+OxyS S. cerevisiae strain with anhydrotetracycline (Figures 2-1, 2-2 and 2-3) but degraded prior

to analysis. However, even if that is the case, it is still notable that a significant proportion of a

singly hydroxylated product is formed as well (Figure 2-1), in contrast to the previously reported

OxyS double hydroxylation in vitro and in S. lividans.(7)

The literature reported ability to isolate and analyze 7-chloro-5a(11a)-dehydrotetracycline could

provide further support for the potential role of the second hydroxylation step in destabilizing the

5a(11a)-dehydro product. 7-chloro-5a(11a)-dehydrotetracycline, lacking the 5α-hydroxy present

in 5a(11a)-dehydrooxytetracycline, is produced by mutant strain S-1308 of Streptomyces lividans

and can be isolated and analyzed.(11) The corresponding S. lividans wild type strain normally

produces 7-chlorotetracycline, which, like tetracycline is also lacking the additional 5-hydroxy

group. However, the 7-chloro group could also be responsible for the stability of 7-chloro-5a(11a)-

dehydrotetracycline, previously reported to be the only stable 5a(11a)-dehydrotetetracycline of the

55

ones that have been prepared.(28) Purification of the hydroxylation product of anhydrotetracycline

and NMR characterization will enable verifying whether 5a(11a)-dehydrotetracycline is indeed

produced.

2.5.2 5a(11a)-dehydrotetracycline reduction in S. cerevisiae

Recently, the biosynthetic pathway of cofactor F420 was fully characterized and F420 was

successfully heterologously produced in E. coli by recombinant expression of four genes.(29) The

substrates for the F420 pathway, lactate, tyrosine, and 5-amino-6-ribitylamino-2,4[1H,3H]-

pyrimidinedione, are native to yeast.(30, 31) Thus, a future attempt to natively produce F420 in

yeast to allow functional expression of cofactor F420-dependent enzymes such as OxyR is now

more feasible. Alternatively, cofactor F420 or its precursors might become more readily available

to be exogenously supplied to S. cerevisiae through heterologous production in another organism

such as E. coli. Importantly however, a reduction that takes place in S. cerevisiae in the absence

of OxyR and F420 (Figure 2-3) potentially circumvents the need to heterologously express F420 in

S. cerevisiae, a formidable challenge on its own right.

Another alternative route to tetracycline from 5a(11a)-dehydrotetracycline is chemical reduction.

However, the process is not necessarily stereoselective and can yield an additional stereoisomer to

tetracycline, 5a-epitetracycline, of significantly lesser antibiotic activity (e.g. against S. aureus)

than tetracycline.(11) Stereoselectivity was achieved however in the chemical reduction of the

5a(11a)-bond of 7-chloro-5a(11a)-dehydrotetracycline and of the 6-peroxidated derivative of

5a(11a)-dehydrotetracycline.(32-34) Larger scale culturing, isolation and NMR analysis of the

reduction product of OxyS hydroxylation (Figure 2-3) will be needed to confirm whether the

reduction takes place in the 5a(11a)-bond and if it does, whether it is stereoselective.

56

While the conversion percent for the hydroxylation of anhydrotetracycline by OxyS is high (Figure

2-1), the conversion percent of the reduction reaction is much lower (Figure 2-3), assuming equal

ionization capacity of anhydrotetracycline, its hydroxylated intermediate and its hydroxylated and

reduced product (Scheme 2-1). Thus, optimization of the reduction reaction is likely to be needed.

An obvious method for optimization of the reduction reaction would be to identify the native

reductase enzyme in S. cerevisiae and overexpress it. In order to identify the endogenous reductase

enzyme in S. cerevisiae a screen could be done using a plasmid collection of yeast genes and a

screening method such as described in the next chapter.(35) An alternative assay would be an

overlay of the yeast on bacteria given the much higher antibiotic activity of tetracycline over

5a(11a)-dehydrotetracycline.(11) Instead of a library of yeast genes, a library of yeast deletion

mutants could be screened, using similar assays, to identify yeast mutants that cannot reduce the

hydroxylation products of OxyS.(36, 37)

Importantly however, for these assays to function, the reduction of OxyS’s hydroxylation product

needs to happen in whole cells and not only in cell lysates and whether and in which conditions

this is the case is still unanswered. Thus, a cell lysate assay employing a more specific library of

yeast reductases or ene-reductases or even ene-reductases of α,β-unsat carbonyls is preferable.(38-

40) An alternative method will be to screen additional OYE proteins for the reduction step and

potentially perform directed evolution for improved substrate recognition and stereoselectivity in

similar methods to those described in the next chapters.(41-50) Finally, OxyR homologs with F420

or an intermediate in its biosynthesis are a potential alternative as well (Section 2.5.1).

2.6 Conclusion

The data presented in this chapter supports that the use of OxyS as the sole heterologously

expressed enzyme in S. cerevisiae allows both a single hydroxylation and reduction steps of

57

anhydrotetracycline to take place using S. cerevisiae in the presence of G6P (Figure 2-3). This

result is unexpected for two reasons. One, OxyS is known to perform two hydroxylation steps on

anhydrotetracycline in vitro and in vivo. Two, it was expected that the coexpression of the

dedicated reductase enzyme and the supply / heterologous biosynthesis of its nonnative cofactor

would be required for the reduction step. Work on isolation and NMR analysis of the hydroxylation

and hydroxylation + reduction products of anhydrotetracycline and 6-

demethylanhydrotetracycline to complement the results presented in this chapter is ongoing.

The work presented in this chapter paves the road for total biosynthesis of tetracyclines using S.

cerevisiae by expressing the additional bacterial biosynthetic enzymes from the oxytetracycline or

the chlortetracycline pathways.(51-54) In addition, the work presented in this chapter paves the

road for producing key tetracycline analogs from bacterial and fungal anhydrotetracyclines such

as anhydrotetracycline, anhydrodactylocyclinone and TAN-1612, the subject of the next chapters.

58

2.7 Appendix

Table 2-1 Strains used in this study

Genotype Source/Reference

FY251 MATa leu2-Δ1 trpΔ63 ura3-52 his3-Δ200 ATCC 96098

BJ5464-

NpgA

MATα ura3-52 his3-Δ200 leu2-Δ1

trp1 pep4::HIS3 δ:: pADH2-npgA-tADH2 prb1Δ1.6R can1

GAL

a / (55)

EH-3-80-3 FY251 pSP-G1 This Study

EH-3-98-6 FY-251 AL-1-101-C10 This Study

EH-3-142-A AL-1-141-A-C6 This Study

EH-3-142-B AL-1-151-A-C4 This Study

EH-3-142-C AL-1-151-B-C2 This Study

ΕΗ-5-153-8 FY-251 AL-1-119-A-C7 This Study

EH-3-204-5 FY-251 AL-1-119-A-C7, AL-215-D-C1 This Study

EH-3-204-6 FY-251 AL-1-119-A-C7, AL-255-C1 This Study

EH-3-204-7 FY-251 AL-1-119-A-C7, AL-235-C5 This Study

EH-3-204-8 FY-251 AL-1-119-A-C7, pRS413-pGAL1 This Study

EH-3-204-9 FY-251 AL-1-101-C10, pRS413-pGAL1 This Study

EH-3-248-1 BJ5464-NpgA AL-1-101-C10 This Study

EH-3-248-8 BJ5464-NpgA pSP-G1 This Study a Y. Tang, University of California, Los Angeles, Department of Chemical and Biomolecular

Engineering & Department of Chemistry and Biochemistry, Department of Bioengineering

Table 2-2 Plasmids used in this study

Description Source/Reference

pSP-G1 pTEF1-FLAG-tADH1, pPGK1-MYC-tCYC1, 2 μ, Ura Addgene 64736

pRS413-

pGAL1

Shuttle vector (empty), His marker, CEN ATCC 87326

AL-1-101-

C10

pTEF1-oxyS-tADH1 in pSP-G1 This Study

AL-1-119-A-

C7

pTEF1-oxyS-tADH1, pPGK1-oxyR-tCYC1 in pSP-G1 This Study

AL-1-141-A-

C6

pTEF1-oxyS-tADH1, pPGK1-OYE1-tCYC1 in pSP-G1 This Study

AL-1-151-A-

C4

pTEF1-oxyS-tADH1, pPGK1-OYE2-tCYC1 in pSP-G1 This Study

AL-1-151-B-

C2

pTEF1-oxyS-tADH1, pPGK1-OYE3-tCYC1 in pSP-G1 This Study

AL-1-183-C-

C8

Fo synthase Thermobifida fusca This Study

59

AL-1-183-D-

C7

Fo synthase Chlamydomonas reinhardtii This Study

AL-1-215-D-

C1

FGD1 from Mycobacterium tuberculosis This Study

AL-1-255-C1 F420-dependent NADP+ oxidoreductase from

Archeoglobus fulgidus

This Study

AL-1-235-C5 NADPH-dependent F420 reductase from Streptomyces

griseus

This Study

Table 2-3 Sequences used in this study

The sequence for the hydroxylase OxyS from Streptomyces rimosus (GenBank AAZ78342.1) is

shown capitalized within the context of the pSP-G1 backbone containing pTEF1, the FLAG tag

and tADH1 (partial, decapitalized).

The sequence for the reductase OxyR from Streptomyces rimosus (GenBank: DQ143963.2) is

shown capitalized within the context of the pSP-G1 backbone containing pPGK1, the myc tag and

tCYC1 (partial, decapitalized).

Sequences for the three F420 reductases from M. tuberculosis, A. fulgidus and S. griseus are shown

along with the pGPD (pTDH3) promoter capitalized within the context of the pRS413 backbone

containing the tCYC1 terminator (partial, decapitalized). The NCBI/Genbank reference sequences

used for the Fo reductases from M. tuberculosis, A. fulgidus and S. griseus are CP023708.1,

NC_000917.1 and NC_010572.1 (6172267..6172977). Prior to codon optimization, leucine codon

in M. tuberculosis F420 reductases in subsequence LTGAACAACACCCGGTTT was changed to

methionine so that the total sequence matches the protein sequence used for M. tuberculosis F420

crystallization.(19)

pTEF1-

oxyS-

tADH1-in-

pSP-G1-

AL-1-101-

C10 and

AL-1-119-

A-C7

tcaagtttcagtttcatttttcttgttctattacaactttttttacttcttgctcattagaaagaaagcatagcaatctaatctaa

gttttaattacaagcggccgcATGAGGTACGATGTTGTTATAGCTGGTGCAGGAC

CCACCGGTTTGATGTTAGCATGTGAACTTCGGCTGGCGGGTGCCAGA

ACTTTGGTTTTAGAAAGATTAGCCGAGCCTGTTGACTTCTCGAAAGCT

CTAGGAGTCCACGCTCGCACTGTTGAACTATTAGATATGAGAGGCCT

CGGTGAAGGATTCCAGGCTGAAGCACCAAAGTTAAGAGGTGGTAATT

TTGCCTCATTAGGCGTCCCCCTGGATTTCTCATCATTTGATACTAGAC

ACCCATATGCATTGTTTGTTCCACAAGTACGAACTGAAGAACTGCTA

ACAGGTAGAGCTTTGGAGCTAGGGGCGGAGCTGCGTCGTGGTCATGC

CGTGACCGCCTTGGAACAAGATGCTGATGGTGTTACTGTGAGCGTGA

CAGGCCCTGAAGGCCCATACGAAGTAGAATGTGCTTATTTGGTGGGC

TGCGACGGTGGAGGCAGTACGGTGAGGAAACTATTGGGCATAGATTT

TCCAGGTCAAGACCCACATATGTTTGCTGTCATCGCAGACGCAAGAT

TCAGAGAAGAGCTTCCTCACGGAGAAGGTATGGGTCCTATGCGTCCT

60

TATGGTGTCATGAGACATGACCTTCGTGCATGGTTCGCAGCATTTCCG

CTAGAACCAGACGTCTACAGAGCAACAGTCGCCTTTTTCGATAGACC

GTATGCTGACAGGAGGGCGCCGGTAACGGAGGAGGATGTCAGAGCC

GCGTTAACAGAAGTTGCTGGATCTGACTTTGGAATGCATGATGTAAG

ATGGTTATCTCGTTTGACCGATACAAGTAGACAAGCAGAAAGGTATA

GAGATGGGAGAGTTCTTTTGGCTGGTGATGCATGCCATATTCATTTGC

CCGCTGGTGGCCAGGGACTGAACTTAGGATTTCAAGATGCCGTTAAC

TTGGGTTGGAAGCTTGGTGCTACCATTGCCGGGACCGCTCCACCAGA

GTTATTGGATACGTATGAAGCTGAGAGAAGGCCGATAGCCGCCGGTG

TTTTGAGAAATACAAGGGCTCAAGCTGTTCTAATTGATCCAGATCCTC

GCTACGAGGGTTTAAGGGAATTAATGATTGAATTGTTGCACGTTCCT

GAGACTAATAGATATTTAGCGGGGCTAATCTCCGCATTAGACGTTAG

ATACCCAATGGCTGGGGAACATCCATTGCTGGGAAGAAGAGTACCCG

ACTTACCTCTTGTCACCGAAGATGGAACAAGACAGTTGTCCACTTATT

TCCATGCTGCACGTGGCGTATTATTAACGTTAGGTTGTGATCAACCAC

TAGCAGATGAAGCCGCTGCTTGGAAAGATAGGGTTGATTTAGTTGCA

GCTGAAGGTGTGGCGGACCCTGGTTCTGCAGTAGATGGCTTAACTGC

TTTACTTGTAAGGCCTGATGGTTACATTTGTTGGACAGCTGCCCCAGA

AACTGGTACTGATGGATTGACAGACGCGCTGCGTACTTGGTTCGGCC

CACCTGCAATGgtatcgatggattacaaggatgacgacgataagatctgagctcttaattaacaattcttcg

ccagaggtttggtcaagtctccaatcaaggttgtcggct

FGD1-in-

pRS413-

pGPD-

tCYC1-AL-

1-215-D-

C1

ccctcactaaagggaacaaaagctggagctcAGTTTATCATTATCAATACTGCCATTTC

AAAGAATACGTAAATAATTAATAGTAGTGATTTTCCTAACTTTATTTA

GTCAAAAAATTAGCCTTTTAATTCTGCTGTAACCCGTACATGCCCAAA

ATAGGGGGCGGGTTACACAGAATATATAACATCGTAGGTGTCTGGGT

GAACAGTTTATTCCTGGCATCCACTAAATATAATGGAGCCCGCTTTTT

AAGCTGGCATCCAGAAAAAAAAAGAATCCCAGCACCAAAATATTGT

TTTCTTCACCAACCATCAGTTCATAGGTCCATTCTCTTAGCGCAACTA

CAGAGAACAGGGGCACAAACAGGCAAAAAACGGGCACAACCTCAAT

GGAGTGATGCAACCTGCCTGGAGTAAATGATGACACAAGGCAATTG

ACCCACGCATGTATCTATCTCATTTTCTTACACCTTCTATTACCTTCTG

CTCTCTCTGATTTGGAAAAAGCTGAAAAAAAAGGTTGAAACCAGTTC

CCTGAAATTATTCCCCTACTTGACTAATAAGTATATAAAGACGGTAG

GTATTGATTGTAATTCTGTAAATCTATTTCTTAAACTTCTTAAATTCTA

CTTTTATAGTTAGTCTTTTTTTTAGTTTTAAAACACCAAGAACTTAGTT

TCGACGGATACTAGTAAAATGGCTGAGTTGAAACTTGGCTACAAGGC

ATCAGCTGAACAGTTTGCCCCAAGGGAGTTAGTCGAACTAGCAGTCG

CTGCCGAAGCTCACGGTATGGATTCCGCTACTGTTTCCGACCATTTCC

AACCATGGAGACACCAAGGTGGCCATGCACCATTCTCACTCAGTTGG

ATGACAGCTGTTGGAGAAAGAACTAATAGATTGTTATTGGGGACGTC

GGTACTCACCCCGACGTTTAGATACAACCCTGCGGTAATAGCACAGG

CCTTTGCTACAATGGGATGTCTATATCCAAACCGGGTGTTTTTAGGTG

TTGGTACTGGAGAGGCCTTGAATGAAATCGCCACTGGTTATGAAGGT

GCTTGGCCTGAGTTCAAAGAAAGGTTTGCGCGTCTGCGCGAAAGCGT

GGGGCTGATGAGACAATTATGGTCTGGTGATAGGGTAGATTTTGATG

GAGATTATTATAGATTAAAAGGCGCGTCTATATATGACGTTCCGGAT

61

GGTGGTGTCCCTGTATATATTGCCGCCGGAGGACCAGCAGTTGCTAA

ATATGCTGGCCGAGCTGGTGACGGTTTCATATGCACTTCAGGCAAGG

GTGAAGAACTTTACACAGAAAAGTTGATGCCCGCCGTCAGAGAAGG

GGCGGCAGCCGCTGACAGGTCTGTTGATGGCATAGACAAAATGATTG

AGATCAAGATTTCATACGACCCAGATCCCGAATTAGCAATGAATAAC

ACAAGATTTTGGGCACCTCTTAGTTTGACCGCAGAACAAAAGCATAG

CATCGATGATCCAATTGAAATGGAAAAAGCTGCTGATGCTTTACCCA

TCGAGCAAATTGCAAAAAGATGGATTGTTGCAAGTGATCCTGATGAA

GCAGTGGAGAAAGTAGGACAGTACGTGACCTGGGGTTTAAATCATCT

AGTTTTCCACGCTCCTGGGCATGATCAAAGAAGATTCTTGGAATTGTT

TCAATCTGACCTAGCGCCAAGACTTCGTCGTCTGGGTTAACTCGAGtca

tgtaattagttatgtcacgcttacattcacgccctccccccacatccgctctaaccgaaaaggaaggagttagacaac

ctgaagtctaggtccctatttatttttttatagttatgttagtattaagaacgttatttatatttcaaatttttcttttttttctgtac

agacgcgtgtacgcatgtaacattatactgaaaaccttgcttgagaaggttttgggacgctcgaaggctttaatttgcg

gccggtacccaat

FNO-A-

fulgidus-in-

pRS413-

pGPD-

tCYC1-

AL-1-255-

C1

ccctcactaaagggaacaaaagctggagctcAGTTTATCATTATCAATACTGCCATTTC

AAAGAATACGTAAATAATTAATAGTAGTGATTTTCCTAACTTTATTTA

GTCAAAAAATTAGCCTTTTAATTCTGCTGTAACCCGTACATGCCCAAA

ATAGGGGGCGGGTTACACAGAATATATAACATCGTAGGTGTCTGGGT

GAACAGTTTATTCCTGGCATCCACTAAATATAATGGAGCCCGCTTTTT

AAGCTGGCATCCAGAAAAAAAAAGAATCCCAGCACCAAAATATTGT

TTTCTTCACCAACCATCAGTTCATAGGTCCATTCTCTTAGCGCAACTA

CAGAGAACAGGGGCACAAACAGGCAAAAAACGGGCACAACCTCAAT

GGAGTGATGCAACCTGCCTGGAGTAAATGATGACACAAGGCAATTG

ACCCACGCATGTATCTATCTCATTTTCTTACACCTTCTATTACCTTCTG

CTCTCTCTGATTTGGAAAAAGCTGAAAAAAAAGGTTGAAACCAGTTC

CCTGAAATTATTCCCCTACTTGACTAATAAGTATATAAAGACGGTAG

GTATTGATTGTAATTCTGTAAATCTATTTCTTAAACTTCTTAAATTCTA

CTTTTATAGTTAGTCTTTTTTTTAGTTTTAAAACACCAAGAACTTAGTT

TCGACGGATACTAGTAAAATGAGGGTAGCTTTACTTGGTGGTACAGG

AAATCTTGGAAAAGGCCTTGCCCTACGACTGGCCACGCTGGGCCATG

AAATCGTAGTCGGAAGCAGAAGAGAAGAAAAGGCAGAGGCTAAAGC

AGCAGAATATAGGCGCATTGCAGGTGACGCTTCCATCACTGGTATGA

AAAATGAAGATGCCGCTGAGGCATGCGACATTGCTGTCTTGACCATT

CCTTGGGAACATGCTATTGACACTGCCAGAGATTTGAAGAATATTTT

ACGTGAGAAAATTGTTGTATCACCATTAGTTCCAGTGTCAAGAGGTG

CAAAGGGCTTCACCTACTCGTCCGAAAGATCAGCGGCCGAGATTGTG

GCTGAAGTTCTGGAATCTGAAAAAGTTGTTTCTGCGTTGCACACAAT

ACCTGCTGCAAGATTTGCCAACTTGGATGAAAAATTTGATTGGGATG

TTCCTGTTTGTGGTGATGATGACGAAAGTAAGAAAGTCGTCATGTCTT

TAATAAGTGAAATAGATGGGTTGAGGCCGTTAGATGCTGGTCCCCTC

TCTAACTCCCGTTTAGTGGAGAGCCTAACTCCATTGATATTGAACATC

ATGAGATTCAATGGAATGGGTGAACTAGGGATCAAGTTTTTATAACT

CGAGtcatgtaattagttatgtcacgcttacattcacgccctccccccacatccgctctaaccgaaaaggaagga

gttagacaacctgaagtctaggtccctatttatttttttatagttatgttagtattaagaacgttatttatatttcaaatttttctt

62

ttttttctgtacagacgcgtgtacgcatgtaacattatactgaaaaccttgcttgagaaggttttgggacgctcgaagg

ctttaatttgcggccggtacccaat

FNO-S-

Griseus-in-

pRS413-

pGPD-

tCYC1-

AL-1-235-

C5

ccctcactaaagggaacaaaagctggagctcAGTTTATCATTATCAATACTGCCATTTC

AAAGAATACGTAAATAATTAATAGTAGTGATTTTCCTAACTTTATTTA

GTCAAAAAATTAGCCTTTTAATTCTGCTGTAACCCGTACATGCCCAAA

ATAGGGGGCGGGTTACACAGAATATATAACATCGTAGGTGTCTGGGT

GAACAGTTTATTCCTGGCATCCACTAAATATAATGGAGCCCGCTTTTT

AAGCTGGCATCCAGAAAAAAAAAGAATCCCAGCACCAAAATATTGT

TTTCTTCACCAACCATCAGTTCATAGGTCCATTCTCTTAGCGCAACTA

CAGAGAACAGGGGCACAAACAGGCAAAAAACGGGCACAACCTCAAT

GGAGTGATGCAACCTGCCTGGAGTAAATGATGACACAAGGCAATTG

ACCCACGCATGTATCTATCTCATTTTCTTACACCTTCTATTACCTTCTG

CTCTCTCTGATTTGGAAAAAGCTGAAAAAAAAGGTTGAAACCAGTTC

CCTGAAATTATTCCCCTACTTGACTAATAAGTATATAAAGACGGTAG

GTATTGATTGTAATTCTGTAAATCTATTTCTTAAACTTCTTAAATTCTA

CTTTTATAGTTAGTCTTTTTTTTAGTTTTAAAACACCAAGAACTTAGTT

TCGACGGATACTAGTAAAATGACTACTCAAGACAGTGGGTCTGCACC

GAAGCCTCCCGCCAAAGATCCATGGGATTTGCCAGATGTCAGCGCAC

TGTCTGTTGGTGTCCTAGGTGGTACAGGACCACAGGGCAGGGGATTA

GCCTACAGATTGGCGCGTGCTGGTCAAAAAGTGACCTTGGGCTCAAG

AGATGCTGGACGCGCTGCTGATGCAGCGGCAGAGCTGGGCCATGGTG

TGGAAGGTACTGATAATGCAGAATGCGCAAGAAGATCCGACATCGTT

ATTGTTGCTGTACCTTGGGACGGTCATGCTAAAACTTTGGAAAGTTTA

AGAGAAGAGTTGGCTGGCAAGCTGGTTATCGACTGTGTTAACCCCTT

GGGGTTTGATAAGAAAGGTGCTTATGCTTTAAAACCAGAGGAGGGCT

CGGCCGCTGAACAAGCTGCGGCGCTTCTCCCTGATTCACGAGTAACA

GCAGCTTTCCACCATTTATCTGCTGTGCTTTTACAAGATGAATCCATT

GAAGAAATTGATACCGATGTTTTGGTCTTAGGTGAAGCAAGGGCAGA

CACGGATATTGTCCAGGCACTAGCAGGGCGTATACCAGGTATGAGAG

GAATATTTGCCGGTCGTCTAAGAGGTGCCCACCAAGTTGAATCACTT

GTAGCCAATTTAATATCTGTAAACAGAAGGTATAAGGCTCATGCCGG

ACTAAGGGCCACAGACGTGTAACTCGAGtcatgtaattagttatgtcacgcttacattcac

gccctccccccacatccgctctaaccgaaaaggaaggagttagacaacctgaagtctaggtccctatttatttttttat

agttatgttagtattaagaacgttatttatatttcaaatttttcttttttttctgtacagacgcgtgtacgcatgtaacattatac

tgaaaaccttgcttgagaaggttttgggacgctcgaaggctttaatttgcggccggtacccaat

pPGK1-

oxyR-

tCYC1 in

pSP-G1

AL-1-119-

A-C7 and

AL-1-26-1-

C1

tggaatggcgggaaagggtttagtaccacatgctatgatgcccactgtgatctccagagcaaagttcgttcgatcgt

actgttactctctctctttcaaacagaattgtccgaatcgtgtgacaacaacagcctgttctcacacactcttttcttctaa

ccaagggggtggtttagtttagtagaacctcgtgaaacttacatttacatatatataaacttgcataaattggtcaatgc

aagaaatacatatttggtcttttctaattcgtagtttttcaagttcttagatgctttctttttctcttttttacagatcatcaagga

agtaattatctactttttacaacaaatataaaacaaggatccATGCCATTCACTCAAAAGGAGAT

CACGTATTTAAGGGCTCAAGGCTACGGCCGACTAGCCACCGTCGGTG

CTCACGGTGAACCTCACAACGTGCCGGTATCTTTTGAAATTGATGAA

GAAAGAGGTACCATTGAAATAACAGGAAGGGATATGGGACGTTCCA

GAAAATTCAGAAATGTTGCCAAAAGTGACAGAGTGGCATTTGTTGTT

GATGATGTTCCATGCAGAGACCCAGAAGTTGTCAGAGCTGTTGTAAT

ACATGGTACTGCACAGGCGCTTCCCACAGGCGGAAGAGAAAGGCGT

63

CCTCATTGTGCTGACGAAATGATTAGAATTCATCCAAGAAGAATAGT

AACATGGGGTATCGAAGGTGATTTGTCAACTGGTGTTCATGCAAGAG

ATATTACTGCTGAAGATGGTGGTAGAAGGgtcgacatggaacagaagttgatttccga

agaagacctcgagtaagcttggtaccgcggctagctaagatccgctctaaccgaaaaggaaggagttagacaacc

tgaagtctaggtccctatttatttttttatagttatgttagtattaagaacgttatttatatttcaaatttttcttttttttctgtaca

gacgcgtgtacgcatgtaacattatactgaaaaccttgcttgagaaggttttgggacgctcgaagatccagctgcatt

aatgaatcggccaacgcgcggggagaggcggtttgcgtattgggcgctcttccgcttcctcgctcactgactcgct

gcgctcgg

64

2.8 References

1. Ro DK, Paradise EM, Ouellet M, Fisher KJ, Newman KL, Ndungu JM, Ho KA, Eachus

RA, Ham TS, Kirby J, Chang MC, Withers ST, Shiba Y, Sarpong R, Keasling JD. Production of

the antimalarial drug precursor artemisinic acid in engineered yeast. Nature. 2006;440(7086):940-

943. doi: 10.1038/nature04640.

2. Galanie S, Thodey K, Trenchard IJ, Filsinger Interrante M, Smolke CD. Complete

biosynthesis of opioids in yeast. Science. 2015;349(6252):1095-1100. doi:

10.1126/science.aac9373.

3. Luo X, Reiter MA, d’Espaux L, Wong J, Denby CM, Lechner A, Zhang Y, Grzybowski

AT, Harth S, Lin W, Lee H, Yu C, Shin J, Deng K, Benites VT, Wang G, Baidoo EEK, Chen Y,

Dev I, Petzold CJ, Keasling JD. Complete biosynthesis of cannabinoids and their unnatural

analogues in yeast. Nature. 2019;567(7746):123-126. doi: 10.1038/s41586-019-0978-9.

4. Awan AR, Blount BA, Bell DJ, Shaw WM, Ho JCH, McKiernan RM, Ellis T. Biosynthesis

of the antibiotic nonribosomal peptide penicillin in baker’s yeast. Nature Communications. 2017;8.

doi: 10.1038/ncomms15202.

5. Wang P, Kim W, Pickens LB, Gao X, Tang Y. Heterologous Expression and Manipulation

of Three Tetracycline Biosynthetic Pathways. Angewandte Chemie-International Edition.

2012;51(44):11136-11140. doi: 10.1002/anie.201205426.

6. Zhou K, Qiao K, Edgar S, Stephanopoulos G. Distributing a metabolic pathway among a

microbial consortium enhances production of natural products. Nat Biotechnol. 2015;33(4):377-

383. doi: 10.1038/nbt.3095.

7. Wang P, Bashiri G, Gao X, Sawaya MR, Tang Y. Uncovering the Enzymes that Catalyze

the Final Steps in Oxytetracycline Biosynthesis. J Am Chem Soc. 2013;135(19):7138-7141. doi:

10.1021/ja403516u.

8. Hošťálek Z, Vaněk Z. Biosynthesis of the Tetracyclines. In: Hlavka JJ, Boothe JH, editors.

The Tetracyclines. Berlin, Heidelberg: Springer Berlin Heidelberg; 1985. p. 137-178.

9. Partow S, Siewers V, Bjorn S, Nielsen J, Maury J. Characterization of different promoters

for designing a new expression vector in Saccharomyces cerevisiae. Yeast. 2010;27(11):955-964.

doi: 10.1002/yea.1806.

65

10. Tymiak AA, Aklonis C, Bolgar MS, Kahle AD, Kirsch DR, O'Sullivan J, Porubcan MA,

Principe P, Trejo WH. Dactylocyclines: novel tetracycline glycosides active against tetracycline-

resistant bacteria. J Org Chem. 1993;58(3):535-537. doi: 10.1021/jo00055a002.

11. McCormick JRD, Miller PA, Growich JA, Sjolander NO, Doerschuk AP. Two new

tetracycline-related compounds: 7-chloro-5a(11a)-dehydrotetracycline and 5a-epi-tetracycline. a

new route to tetracycline. J Am Chem Soc. 1958;80(20):5572-5573. doi: 10.1021/ja01553a066.

12. Greening C, Ahmed FH, Mohamed AE, Lee BM, Pandey G, Warden AC, Scott C,

Oakeshott JG, Taylor MC, Jackson CJ. Physiology, Biochemistry, and Applications of F420- and

Fo-Dependent Redox Reactions. Microbiol Mol Biol Rev. 2016;80(2):451-493. doi:

10.1128/mmbr.00070-15.

13. Jacobson FS, Daniels L, Fox JA, Walsh CT, Orme-Johnson WH. Purification and

properties of an 8-hydroxy-5-deazaflavin-reducing hydrogenase from Methanobacterium

thermoautotrophicum. J Biol Chem. 1982;257(7):3385-3388.

14. Yamazaki S, Tsai L, Stadtman TC. Analogs of 8-hydroxy-5-deazaflavin cofactor: relative

activity as substrates for 8-hydroxy-5-deazaflavin-dependent NADP+ reductase from

Methanococcus vannielii. Biochemistry. 1982;21(5):934-939. doi: 10.1021/bi00534a019.

15. Eirich LD, Vogels GD, Wolfe RS. Distribution of coenzyme F420 and properties of its

hydrolytic fragments. J Bacteriol. 1979;140(1):20-27.

16. Decamps L, Philmus B, Benjdia A, White R, Begley TP, Berteau O. Biosynthesis of F0,

Precursor of the F420 Cofactor, Requires a Unique Two Radical-SAM Domain Enzyme and

Tyrosine as Substrate. J Am Chem Soc. 2012;134(44):18173-18176. doi: 10.1021/ja307762b.

17. Petersen JL, Ronan PJ. Critical role of 7,8-didemethyl-8-hydroxy-5-deazariboflavin for

photoreactivation in Chlamydomonas reinhardtii. J Biol Chem. 2010;285(42):32467-32475. doi:

10.1074/jbc.M110.146050.

18. Hossain MS, Le CQ, Joseph E, Nguyen TQ, Johnson-Winters K, Foss FW. Convenient

synthesis of deazaflavin cofactor FO and its activity in F420-dependent NADP reductase. Org

Biomol Chem. 2015;13(18):5082-5085. doi: 10.1039/c5ob00365b.

19. Bashiri G, Squire CJ, Moreland NJ, Baker EN. Crystal Structures of F420-dependent

Glucose-6-phosphate Dehydrogenase FGD1 Involved in the Activation of the Anti-tuberculosis

Drug Candidate PA-824 Reveal the Basis of Coenzyme and Substrate Binding. J Biol Chem.

2008;283(25):17531-17541. doi: 10.1074/jbc.M801854200.

66

20. Le CQ, Joseph E, Nguyen T, Johnson-Winters K. Optimization of Expression and

Purification of Recombinant Archeoglobus fulgidus F420H2:NADP+ Oxidoreductase, an F420

Cofactor Dependent Enzyme. The Protein Journal. 2015;34(6):391-397. doi: 10.1007/s10930-015-

9633-y.

21. Warkentin E. Structures of F420H2:NADP+ oxidoreductase with and without its substrates

bound. The EMBO Journal. 2001;20(23):6561-6569. doi: 10.1093/emboj/20.23.6561.

22. Ohnishi Y, Ishikawa J, Hara H, Suzuki H, Ikenoya M, Ikeda H, Yamashita A, Hattori M,

Horinouchi S. Genome Sequence of the Streptomycin-Producing Microorganism Streptomyces

griseus IFO 13350. J Bacteriol. 2008;190(11):4050-4060. doi: 10.1128/jb.00204-08.

23. Karplus PA, Fox KM, Massey V. Flavoprotein structure and mechanism. 8. Structure-

function relations for old yellow enzyme. The FASEB Journal. 1995;9(15):1518-1526. doi:

10.1096/fasebj.9.15.8529830.

24. Saito K, Thiele DJ, Davio M, Lockridge O, Massey V. The cloning and expression of a

gene encoding Old Yellow Enzyme from Saccharomyces carlsbergensis. J Biol Chem.

1991;266(31):20720-20724.

25. Swiderska MA, Stewart JD. Stereoselective enone reductions by Saccharomyces

carlsbergensis old yellow enzyme. J Mol Catal B: Enzym. 2006;42(1-2):52-54. doi:

10.1016/j.molcatb.2006.06.023.

26. Hall M, Stueckler C, Hauer B, Stuermer R, Friedrich T, Breuer M, Kroutil W, Faber K.

Asymmetric Bioreduction of Activated C=C Bonds Using Zymomonas mobilis NCR Enoate

Reductase and Old Yellow Enzymes OYE 1–3 from Yeasts. Eur J Org Chem. 2008;2008(9):1511-

1516. doi: 10.1002/ejoc.200701208.

27. Mitscher LA. Degradation and Structure Proofs. The Chemistry of the Tetracycline

Antiobitcs. New York: Marcel Dekker, Inc.; 1978. p. 122-164.

28. Miller PA, Hash JH. [47] NADP:Tetracycline 5a(11a)dehydrogenase1975;43:606-607.

doi: 10.1016/0076-6879(75)43123-8.

29. Bashiri G, Antoney J, Jirgis ENM, Shah MV, Ney B, Copp J, Stuteley SM, Sreebhavan S,

Palmer B, Middleditch M, Tokuriki N, Greening C, Scott C, Baker EN, Jackson CJ. A revised

biosynthetic pathway for the cofactor F420 in prokaryotes. Nature Communications. 2019;10(1).

doi: 10.1038/s41467-019-09534-x.

67

30. Fischer M, Bacher A. Biosynthesis of flavocoenzymes. Nat Prod Rep. 2005;22(3). doi:

10.1039/b210142b.

31. Abbas CA, Sibirny AA. Genetic Control of Biosynthesis and Transport of Riboflavin and

Flavin Nucleotides and Construction of Robust Biotechnological Producers. Microbiol Mol Biol

Rev. 2011;75(2):321-360. doi: 10.1128/mmbr.00030-10.

32. Tatsuta K, Yoshimoto T, Gunji H, Okado Y, Takahashi M. The First Total Synthesis of

Natural (−)-Tetracycline. Chem Lett. 2000;29(6):646-647. doi: 10.1246/cl.2000.646.

33. Scott AI, Bedford CT. Simulation of the Biosynthesis of Tetracyclines. A Partial Synthesis

of Tetracycline from Anhydroaureomycin. J Am Chem Soc. 1962;84(11):2271-2272. doi:

10.1021/ja00870a061.

34. Charest MG, Siegel DR, Myers AG. Synthesis of (−)-Tetracycline. J Am Chem Soc.

2005;127(23):8292-8293. doi: 10.1021/ja052151d.

35. Rose MD, Novick P, Thomas JH, Botstein D, Fink GR. A Saccharomyces cerevisiae

genomic plasmid bank based on a centromere-containing shuttle vector. Gene. 1987;60(2-3):237-

243. doi: 10.1016/0378-1119(87)90232-0.

36. Giaever G, Nislow C. The Yeast Deletion Collection: A Decade of Functional Genomics.

Genetics. 2014;197(2):451-465. doi: 10.1534/genetics.114.161620.

37. Scherens B, Goffeau A. The uses of genome-wide yeast mutant collections. Genome Biol.

2004;5(7). doi: 10.1186/gb-2004-5-7-229.

38. Knaus T, Toogood HS, Scrutton NS. Ene-reductases and their Applications. Green

Biocatalysis2016. p. 473-488.

39. Winkler CK, Tasnádi G, Clay D, Hall M, Faber K. Asymmetric bioreduction of activated

alkenes to industrially relevant optically active compounds. J Biotechnol. 2012;162(4):381-389.

doi: 10.1016/j.jbiotec.2012.03.023.

40. Cherry JM, Hong EL, Amundsen C, Balakrishnan R, Binkley G, Chan ET, Christie KR,

Costanzo MC, Dwight SS, Engel SR, Fisk DG, Hirschman JE, Hitz BC, Karra K, Krieger CJ,

Miyasato SR, Nash RS, Park J, Skrzypek MS, Simison M, Weng S, Wong ED. Saccharomyces

Genome Database: the genomics resource of budding yeast. Nucleic Acids Res.

2011;40(D1):D700-D705. doi: 10.1093/nar/gkr1029.

68

41. Hollmann F, Arends IWCE, Holtmann D. Enzymatic reductions for the chemist. Green

Chem. 2011;13(9). doi: 10.1039/c1gc15424a.

42. Hollmann F, Arends IWCE, Buehler K. Biocatalytic Redox Reactions for Organic

Synthesis: Nonconventional Regeneration Methods. ChemCatChem. 2010;2(7):762-782. doi:

10.1002/cctc.201000069.

43. Fryszkowska A, Toogood H, Sakuma M, Gardiner JM, Stephens GM, Scrutton NS.

Asymmetric Reduction of Activated Alkenes by Pentaerythritol Tetranitrate Reductase:

Specificity and Control of Stereochemical Outcome by Reaction Optimisation. Adv Synth Catal.

2009;351(17):2976-2990. doi: 10.1002/adsc.200900574.

44. Cheng JZ, Coyle CM, Panaccione DG, O’Connor SE. A Role for Old Yellow Enzyme in

Ergot Alkaloid Biosynthesis. J Am Chem Soc. 2010;132(6):1776-1777. doi: 10.1021/ja910193p.

45. Hall M, Stueckler C, Kroutil W, Macheroux P, Faber K. Asymmetric Bioreduction of

Activated Alkenes Using Cloned 12-Oxophytodienoate Reductase Isoenzymes OPR-1 and OPR-

3 fromLycopersicon esculentum (Tomato): A Striking Change of Stereoselectivity. Angew Chem

Int Ed. 2007;46(21):3934-3937. doi: 10.1002/anie.200605168.

46. Hall M, Stueckler C, Ehammer H, Pointner E, Oberdorfer G, Gruber K, Hauer B, Stuermer

R, Kroutil W, Macheroux P, Faber K. Asymmetric Bioreduction of C C Bonds using Enoate

Reductases OPR1, OPR3 and YqjM: Enzyme-Based Stereocontrol. Adv Synth Catal.

2008;350(3):411-418. doi: 10.1002/adsc.200700458.

47. Messiha HL, Munro AW, Bruce NC, Barsukov I, Scrutton NS. Reaction of Morphinone

Reductase with 2-Cyclohexen-1-one and 1-Nitrocyclohexene. J Biol Chem. 2005;280(11):10695-

10709. doi: 10.1074/jbc.M410595200.

48. Shimoda K, Kubota N, Hirata T, Kondo Y, Hamada H. Stereoselective reduction of 2-

butenolides to chiral butanolides by reductases from cultured cells of Glycine max. Tetrahedron

Lett. 2007;48(8):1345-1347. doi: 10.1016/j.tetlet.2006.12.126.

49. Shimoda K, Kubota N. Asymmetric reduction of 2-substituted 2-butenolides with

reductase from Marchantia polymorpha. Tetrahedron: Asymmetry. 2004;15(24):3827-3829. doi:

10.1016/j.tetasy.2004.11.006.

50. Wallwey C, Matuschek M, Xie X-L, Li S-M. Ergot alkaloid biosynthesis in Aspergillus

fumigatus: Conversion of chanoclavine-I aldehyde to festuclavine by the festuclavine synthase

69

FgaFS in the presence of the old yellow enzyme FgaOx3. Org Biomol Chem. 2010;8(15). doi:

10.1039/c003823g.

51. Pickens LB, Tang Y. Oxytetracycline biosynthesis. J Biol Chem. 2010;285(36):27509-

27515. doi: 10.1074/jbc.R110.130419.

52. Zhang W, Watanabe K, Cai X, Jung ME, Tang Y, Zhan J. Identifying the minimal enzymes

required for anhydrotetracycline biosynthesis. J Am Chem Soc. 2008;130(19):6068-6069. doi:

10.1021/ja800951e.

53. Zhu T, Cheng X, Liu Y, Deng Z, You D. Deciphering and engineering of the final step

halogenase for improved chlortetracycline biosynthesis in industrial Streptomyces aureofaciens.

Metab Eng. 2013;19:69-78. doi: 10.1016/j.ymben.2013.06.003.

54. Dairi T, Nakano T, Aisaka K, Katsumata R, Hasegawa M. Cloning and Nucleotide

Sequence of the Gene Responsible for Chlorination of Tetracycline. Biosci, Biotechnol, Biochem.

2014;59(6):1099-1106. doi: 10.1271/bbb.59.1099.

55. Ma SM, Li JWH, Choi JW, Zhou H, Lee KKM, Moorthie VA, Xie X, Kealey JT, Da Silva

NA, Vederas JC, Tang Y. Complete Reconstitution of a Highly Reducing Iterative Polyketide

Synthase. Science. 2009;326(5952):589-592. doi: 10.1126/science.1175602.

70

3 Towards the biosynthesis of 6-demethyl-6-

epitetracyclines using S. cerevisiae

*The contents of this chapter will be published in:

E. Herbst, V. W. Cornish, et al “Biosynthesis of 6-demethyl-6-epitetracyclines using

Saccharomyces cerevisiae”, in preparation.

71

3.1 Chapter outline

This chapter describes the work towards 6-demethyl-6-epitetracyclines biosynthesis using S.

cerevisiae. 6α-hydroxylation by an enzyme such as DacO1 is required for the synthesis of 6-

demethyl-6-epitetracyclines as opposed to 6β-hydroxylation by enzymes such as OxyS leading to

6-demethyltetracycline (Scheme 3-1 and Scheme 3-2). However, previous attempts in multiple

heterologous hosts to heterologously express DacO1 without the rest of the dactylocyclinone

pathway have not yielded stably expressed protein. Thus, the chapter first describes rational design

approaches for optimizing DacO1 expression in S. cerevisiae (Section 3.4.1). The rationale behind

the choices for bacterial hydroxylases screened for 6α-hydroxylase activity on anhydrotetracycline

is explained (Section 3.4.2). Screening for 6-hydroxylase activity towards the biosynthesis of 6-

demethyl-6-epitetracyclines required the development of a multititer plate assay for

anhydrotetracycline biosynthesis (Section 3.4.3). This assay was put to the use in screening for

error-prone mutants of DacO1 in an attempt to identify 6α-hydroxylases of anhydrotetracycline

(Section 3.4.4). Finally, the results of testing the rational design DacO1 expression optimization

library as well as the other bacterial candidates for 6α-hydroxylation in S. cerevisiae are described

(Section 3.4.5). Key results in this chapter are the the development of a stably expressing form of

DacO1 in S. cerevisiae (Figure 3-8), the identification of PgaE as an anhydrotetracycline

hydroxylase (Figure 3-9 and Figure 3-10), and the discovery that a larger proportion of PgaE

hydroxylated intermediates undergo reduction in the presence of glucose-6-phosphate than is the

case in OxyS (Figure 3-11). Finally, the next required steps towards 6-demethyl-6-epitetracyclines

biosynthesis in S. cerevsiae are discussed (Section 3.5).

72

3.2 Introduction

As mentioned in Chapter 1, the synthesis of 6-demethyl-6-epiglycotetracyclines for testing is

highly desirable due to their highly promising outlook as antibiotics. The first two steps in the

synthesis of 6-demethyl-6-epiglycotetracyclines are the synthesis of 6-demethyl-6-epitetracyclines

from anhydrotetracyclines. This chapter shows the progress towards developing an S. cerevisiae

platform for 6-demethyl-6-epitetracyclines biosynthesis using bacterial hydroxylase enzymes as a

starting point for directed evolution and rational design as well as anhydrotetracycline as a model

substrate. DacO1, a bacterial flavin-dependent monooxygenase homologous to OxyS was

hypothesized to perform a 6α-hydroxylation on its native substrate that leads to 6-

epiglycotetracyclines in its native host, Dactylosporangium sp. SC 14051 (ATCC 53693).(1, 2)

Thus, DacO1, along with other bacterial hydroxylases, is used here as a template to evolve a 6α-

hydroxylase in S. cerevisiae towards the biosynthesis of 6-demethyl-6-epiglycotetracyclines.

3.3 Materials and methods

See Chapter 5 for general methods. See Chapter 2 for general protocol for hydroxylation/reduction

assay in cell lysate.

3.3.1 General protocol for high throughput assay of hydroxylation/reduction

in whole cells in microtiter plates

Fresh colonies of strains harboring a plasmid encoding a hydroxylase and/or reductase and control

strains (EH-3-98-6 encoding OxyS as positive control and EH-3-80-3 with no hydroxylase as

negative control) were inoculated in 200 μL selective media (U- or HU-) in 96-well plates (Corning

353072) and placed in shaker overnight. Overnight cultures were used to inoculate 1 mL selective

media (U- or HU-) cultures in deep well plates (Corning P-2ML-SQ-C-S) with an average starting

73

OD of 0.01. The plate was covered with two layers of SealMate film (Excel Scientific, SM-KIT-

BS) and placed in shaker overnight. Cells were then pelleted at 4 °C at 4,000 rpm. Each pellet was

redissolved in 0.3 mL containing a 150 μL solution of 5 mg / mL anhydrotetracycline HCl, 30 μL

1 M Tris buffer pH 7.45, 15 μL 40% glucose solution in H2O and 105 μL H2O for final

concentrations of 2.5 mg/mL anhydrotetracycline HCl, 100 mM Tris pH 7.45 and 2% glucose. The

plates were then covered with two layers of SealMate film (Excel Scientific, SM-KIT-BS) and

placed in shaker overnight at 800 rpm. 2 μL of Overnight suspensions were diluted into 198 μL of

H2O in Corning 3603 plates before UV/VIS spectroscopic measurements. UV/VIS spectroscopic

measurements taken were as follows: (i) absorption spectrum 350 nm – 500 nm, (ii) emission

spectrum 450 nm – 550 nm (λexcitation = 400 nm), (iii) excitation spectrum 305 nm – 455 nm (λemission

= 500 nm). Spectral steps in the spectra were 25 nm.

3.3.2 General protocol for western blots

Fresh patches or colonies of strains harboring the plasmid for the hydroxylation and/or reduction

enzyme and control strains were inoculated in 3 mL selective media (U-) in 15 mL culture tubes

(Corning 352059) and placed in shaker overnight. Overnight cultures were used to reinoculate 3

mL selective media (U-) in 15 mL culture tubes (Corning 352059) and placed in shaker overnight

with a starting OD of 0.01-0.05 (all western Blots shown in this study except for western blots

shown in Figure 3-1 which were pelleted and lysed immediately). Cells were grown to final OD

of 0.6-0.8 before pelleting in 2 separate microcentrifuge tubes (1 mL culture each) at 4 °C at 14,000

rpm, removing the supernatant and freezing at -20 °C prior to further use.

100 μL of a 99:1 mixture of Y-PER yeast protein extraction reagent (ThermoFisher Scientific

78991) and HALT protease inhibitor cocktail (ThermoFisher Scientific PI87786) were added to

each microcentrifuge tube. The tubes were placed on orbital shaker for 20 min at r.t., followed by

74

10 min centrifugation at 14,000 rpm at 4 °C and the cell lysate was transferred to a new 1.5 mL

microcentrifuge tube and kept on ice. Following 5 min at 95 °C with SDS loading buffer,(3) an

SDS-page was run with a Kaleidoscope Prestained Standard (Bio-Rad 161-0324, Figure 3-1) or a

Precision Plus Protein Kaleidoscope (Bio-Rad 161-0375, all other western blots shown),(4) the

gels were transferred to PVDF blots (Thermo Fisher Scientific IB24001) and western blots were

performed according to manufacturer guidelines (Thermo Fisher R951-25 and Sigma-Aldrich

A8592 for myc-and FLAG-tag, respectively) with BSA used instead of milk and analyzed with

Thremo Scientific 34000 substrate kit for horseradish peroxidase.

3.4 Results

3.4.1 Optimizing DacO1 expression in S. cerevisiae – rational design

Following the successful hydroxylation of anhydrotetracycline in S. cerevisiae by OxyS described

in Chapter 2, the 6α-hydroxylation of anhydrotetracycline in S. cerevisiae by DacO1 was

attempted. DacO1 is 66% homologous to OxyS and is assumed to perform the 6α-hydroxylation

on its native substrate, anhydrodactylocyclinone, as opposed to the 6β-hydroxylation that OxyS

performs on anhydrotetracycline (Scheme 3-1).(1)

DacO1 heterologous expression, along with the rest of the Dactylocyclinone pathway was

previously attempted in Streptomyces lividans K4‐114 and has led to the successful biosynthesis

of dactylocyclinone. Assuming correct assignment for the role of DacO1 in the dactylocycline

pathway, given its homology to OxyS, this result supports that DacO1 was successfully

heterologously expressed in S. lividans.(2) However, previous attempts to obtain soluble forms of

heterologously expressed DacO1 from Streptomyces and E. coli, when heterologously expressed

without the rest of its pathway, were not successful.(1)

75

My initial attempts to hydroxylate anhydrotetracycline with DacO1 yielded either little or no

hydroxylation when assayed by mass spectrometry (data not shown) but little hydroxylation was

supported by UV/Vis spectroscopy (Figure 3-4). Therefore, the expression levels of DacO1 were

examined by western blotting with FLAG-tag to determine whether a problem in expression leads

to the low/no hydroxylation levels with DacO1. Another likely explanation for low/no

hydroxylation is difference between anhydrotetracycline, the model substrate I used, and

anhydrodactylocyclinone, the native substrate of DacO1 (Scheme 3-1). Indeed, DacO1 expression

levels were found to be significantly lower than its counterpart OxyS (Figure 3-1-a).

Figure 3-1 Western Blot of DacO1 and OxyS

Cultures of DacO1, DacO4 and OxyS, OxyR encoding strains EH-3-153-7 and EH-3-153-8,

respectively were lysed with Y-PER and labeled with Monoclonal ANTI-FLAG HRP antibody.

Two biological replicates were used for each strain indicated as colony 1 and 2 (C1 and C2),

respectively. The expected sizes are 55.1, 55.9, 16.3 and 17.5 for DacO1, OxyS, DacO4 and OxyR,

respectively.

I therefore attempted to optimize the functional expression of DacO1 by a number of approaches.

Namely, fusion to a stable protein, switching to a transcriptional control of an inducible promoter,

expression in a strain background that has protease deletions, lower temperature culturing and

76

coexpression of pathway proteins. The testing of these expression optimization approaches is

described in Section 3.4.5.

Specifically, I attempted N-terminal fusions proteins previously shown to improve expression of

heterologous proteins in yeast, ubiquitin,(5-8) superoxide dismutase,(9-11) Gal1(8) and γ-

IFN.(12) I also attempted a C- and N-terminal fusion to DacO4, the 5a(11a) reductase enzyme of

the dactylocycline pathway that I verified to be stably expressed in yeast (Figure 3-1-b).

Given the homology between OxyS and DacO1, the successful heterologous expression of OxyS

and the potential importance of the N-terminus of the protein to its in vivo stability,(13) I attempted

three fusion proteins composed of an N-terminal portion of OxyS and a C-terminal portion of

DacO1. These fusion proteins include the first 37, 87 and 191 amino acids of OxyS, followed by

the last 461, 411 and 307 amino acids of DacO1, respectively. In order to determine where to fuse

the proteins I considered the structure of OxyS as determined previously by X-ray

crystallography,(1) and made fusions proteins in disordered regions of the structure around

positions 37, 87 and 191. The homology between DacO1 and OxyS made it simple to determine

what are the corresponding amino acids at the DacO1 protein, although there is no guarantee the

disordered regions in OxyS are also disordered in DacO1.

For inducible promoters I tested pGAL1, induced by galactose, and pADH2, a late stage promoter

induced in low glucose / high EtOH.(14) The alternative strain background I chose is BJ5464-

NpgA, as the BJ-5464 strain background was previously shown useful for heterologous protein

production in yeast.(15, 16) For a lower temperature culturing I attempted 25 °C.(17, 18)

The activity of polyketide biosynthetic enzymes is known to be dependent on an oligomeric

complexation of biosynthetic enzymes. This is claimed for instance in the case of JadF, JadG and

JadH of the Jadomycin biosynthetic pathway.(19) Similarly, in the case of an oxytetracycline

77

producing strain, deletion of the gene encoding for OtcC, which is 99% identical to OxyS used in

this study, led to production of a polyketide with a shorter chain length. Expression of a triply

mutated, catalytically disfunctional OtcC, restored production of a polyketide with the same chain

length. Thus, a post-polyketdie synthase (PKS) polyketide biosynthetic enzyme could have a

structural role in the oligomeric protein complex responsible for polyketide biosynthesis.(20)

Potential further support for the potential that coexpression of additional proteins from the

dactylocycline pathway has for improving DacO1 expression could be found in the case of DacO1

itself. As mentioned above, expression of DacO1 together with the rest of the dactylocyclinone

pathway in S. lividans led to the production of dactylocyclinone.(2) However, expression of

DacO1 in the absence of the rest of the dactylocycline pathway in E. coli, Streptomyces,(1) or S.

cerevisiae (Figure 3-1) yielded no/low levels of soluble protein. In order to provide DacO1 with

enzymes potentially required to form a functional oligomeric complex I attempted the

coexpression of DacJ and DacM2. DacJ and DacM2 were chosen since they are the closest

enzymes to DacO1 in the dactylocycline gene cluster and they are proposed to share with DacO1

the functional role of aglycone tailoring.(2)

3.4.2 Searching the bacterial hydroxylase space for 6α-hydroxylation of

anhydrotetracyclines

Given the goal to perform 6α-hydroxylation on anhydrotetracyclines it was clear that there is a

need to go beyond the 6β-hydroxylase OxyS. Expected challenges with the expression of Daco1

(Section 3.4.1) as well as incongruencies between the DacO1 native substrate

anhydrodactylocyclinone and anhydrotetracycline affirmed the need to test other hydroxylases as

well (Scheme 3-1). I therefore looked for homologous enzymes catalyzing hydroxylation in

polyketides that are structurally related to anhydrotetracycline.

78

Scheme 3-1 Bacterial monooxygenases and their hypothesized substrates and products

Bacterial flavin-dependent monooxygenases OxyS, DacO1, SsfO1, CtcN and PgaE and their

native substrates. Marked in red are differences between the native substrate and

anhydrotetracycline.

SsfO1, the hypothesized flavin-dependent monooxygenases of the 6α-position in SF2575 pathway,

with 54% homology to OxyS, was an obvious candidate (Scheme 3-1).(2) Flavin-dependent

monooxygenases of angucyclines, seemed like another promising source for relevant

monooxygenases, given the structural similarity between angucyclines and tetracyclines.

Specifically, enzymes that catalyze monooxygenation of position 12 of the angucyclines,

79

analogous to position 6 of the tetracyclines seemed of particular interest.(21) I chose to test PgaE

(56% homology to OxyS), catalyzing the hydroxylation of UWM6 (Scheme 3-1), in the

biosynthesis of gaudimycin C since it was previously both analyzed by X-ray crystallography and

used in chemoenzymatic synthesis.(22, 23) Finally, CtcN, with 69% homology to OxyS was

chosen since it was shown to be necessary for the 6-hydroxylation step in the chlortetracycline

pathway.(24) The testing of these bacterial hydroxylases is described in Section 3.4.5.

3.4.3 Developing a microtiter plate assay for anhydrotetracycline

hydroxylation

Given the need to screen for a 6α-hydroxylase of anhydrotetracyclines I developed a high

throughput screen for hydroxylation of anhydrotetracyclines based on the ready detection of

anhydrotetracycline hydroxylation by UV/Vis spectroscopy (Figure 2-2). Following colony

picking onto microtiter plates and overnight culturing to even out the OD across the different wells

the cells were reinoculated for a 2nd overnight culture, pelleted and resuspended in a Tris reaction

buffer containing anhydrotetracycline and glucose. The suspension was then diluted in H2O for

UV/VIS measurements (Figure 3-2).

Figure 3-2 A simplified outline of the microtiter plate assay for anhydrotetracycline

hydroxylation.

80

3.4.4 Evolving a 6α-hydroxylase of anhydrotetracyclines – DacO1 random

mutagenesis

Given the low/no hydroxylation activity of DacO1 on anhydrotetracycline, DacO1 optimization

was attempted by directed evolution. An error-prone mutagenesis library from DacO1 encoding

plasmid AL-1-31-C7 was made with an average of 3 mutations per 1 kb and transformed into

FY251. The library was then assayed for anhydrotetracycline hydroxylation by the microtiter plate

assay for anhydrotetracycline hydroxylation (Figure 3-2) with strains expressing OxyS, DacO1

(w.t.) and empty plasmid (EH-3-98-6, EH-3-80-2 and EH-3-80-3, respectively) as controls. Over

500 colonies were screened in the assay using 96-well plates with two potential hits identified. A

typical plate with no hits, showing only the OxyS expressing strain positive control above

background fluorescence as well as a plate displaying a hit are shown in Figure 3-3.

81

Figure 3-3 DacO1 error-prone mutagenesis screen excerpt

DacO1 error-prone mutagenesis screen using the microtiter plate assay for anhydrotetracycline

hydroxylation. (a) a plate with only the OxyS positive control significantly above background

fluorescence. (b) a plate with both the OxyS positive control and a DacO1 mutant hit with

fluorescence significantly above background. Wells 60 and 72 contain the positive control OxyS

encoding strain EH-3-98-6. λex = 400 nm.

A followup on the two potential DacO1 mutant hits in the screen showed that one of them has a

slightly increased fluorescence in the Δexcitation spectrum relative to DacO1 (Figure 3-4). The

82

new excitation peak at 375 nm, in 25 nm resolution, is similar to that found for OxyS hydroxylation

of anhydrotetractycline at 380 nm, in 10 nm resolution, but of a much lower intensity (Figure 2-

2). Considering that the increase in fluorescence is rather minor I decided to focus in the interim

on alternatives such as rational design approaches to optimize hydroxylation by DacO1 and

exploring DacO1 homologs (Sections 3.4.1 and 3.4.2, respectively). The testing of these

candidates is described in Section 3.4.5.

Figure 3-4 ΔExcitation spectrum for DacO1 and DacO1 error-prone PCR mutant

The spectrum shown is a Δ spectrum, that is, the values shown are the emission values for the

hydroxylase expressing cells minus the emission values of the no hydroxylase control EH-3-80-3.

λemission = 500 nm. Each value is the average of six biological replicates and the error bars represent

standard error.

3.4.5 Testing DacO1 expression optimization constructs and other bacterial

hydroxylases

I screened the strains generated for the DacO1 optimization attempts as well as their controls, EH-

5-98-1 through EH-5-98-19, EH-3-248-1 through EH-3-248-8, EH-3-80-2, EH-3-80-3, EH-3-98-

6 and EH-5-115-1 through EH-5-115-5 (Sections 3.4.1 and 3.4.2) using the microtiter plate

83

UV/Vis assay (Section 3.4.3). Each of the 35 strains was assayed in four biological replicates, as

well as in two culturing temperatures, 30 °C and 25 °C, starting from the 2nd inoculation stage

(Figure 3-2). The protocol for the microtiter plate UV/Vis assay (Section 3.3.1) was slightly

modified to accommodate the strains encoding DacO1 controlled by inducible promoters. Thus,

EH-5-98-7 and EH-5-98-8 encoding pADH2-dacO1 were inoculated in YPD in the 2nd inoculation

and EH-5-98-4 and EH-5-98-5, encoding pGAL1-dacO1 were inoculated in U- Raffinose and

supplemented in the next morning with 66.7 μL of 30% galactose in H2O for a total concentration

of 2%. UV/Vis measurements were taken following one night and after three nights of incubation

with anhydrotetracycline for the 30 °C plates and after one night only for the 25 °C plates.

Strains that showed a blue shift in their peak in the Δexcitation spectrum from that of negative

control strain EH-3-80-2 harboring empty pSP-G1 were indicated as potential hits in the assay and

are shown in Table 3-1. Specifically, strains listed in the table have shown in the Δexcitation

spectrum a peak of 405 nm, 380 nm or 355 nm as opposed to the 430 nm peak of negative control

EH-3-80-3, associated with anhydrotetracycline. Results from three measurements are shown, two

measurements after overnight incubation of the cells suspensions in the buffer containing

anhydrotetracycline, one for each of the culturing temperatures (25 °C and 30 °C) and another

final measurement for the 30 °C culturing condition after 3 nights of suspension in the buffer

containing anhydrotetracycline. The rank was calculated per plate measured for one of the

measurements according to the emission at the peak maximum, and should be interpreted only as

a rough indicator as peak emissions at three different wavelengths are ranked on the same scale.

The number 1 or 2 after the dot indicates the plate number from which the measurement was taken.

Reassuringly, the positive control OxyS, whose native substrate is anhydrotetracycline is leading

the rank in highest emission resulting from a blue-shifted excitation. It is followed by the various

84

DacO1 fusion proteins as well as by hydroxylase alternatives to OxyS and DacO1. Surprisingly,

empty pSP-G1 and OxyR in the alternative background strain, BJ-5464-NpgA have also displayed

a blue-shifted peak in the excitation spectrum, although they are the lowest ranking (Table 3-1).

Also surprisingly, EH-3-80-2 encoding unoptimized DacO1 has not displayed a blue shifted

excitation peak in the reduction spectrum in contrast to previous experiments (Figure 3-4). Strains

that exhibited a blue-shifted excitation peak at 25 °C and not in 30 °C are: DacO1-DacO4,

OxySDacO1-191, no-AB-tags and DacO1-JCAT-BJ. I proceeded to analyze by western blot the

hydroxylase expression of the strains that displayed a blue-shifted excitation peak in all three

measurements (Figure 3-5, Figure 3-6 and Figure 3-7).

85

Table 3-1 Potential hits in the screen of DacO1 expression optimization constructs and other

bacterial hydroxylases

Strain

number

Strain description 30 °C

measurement

I

25 °C

measure

ment I

30 °C

measure

ment II

Rank 30 °C

measurement I

EH-3-98-6 OxyS Y Y Y 1.1

EH-3-98-6 OxyS Y Y Y 1.2

EH-5-98-9 Ubiquitin-γ-IFN-

DacO1

Y Y Y 2.1

EH-3-248-2 OxyS-OxyR-BJ Y Y Y 2.2

EH-5-98-1 Ubiquitin-DacO1 Y Y Y 3.1

EH-3-248-1 OxyS BJ Y Y Y 3.2

EH-5-98-10 γ-IFN-DacO1 Y Y Y 4.1

EH-5-115-4 OxyS JCAT Y Y Y 4.2

EH-3-98-2 hSOD-DacO1 Y Y 5.1

EH-3-248-7 PgaE-JCAT-BJ Y Y Y 5.2

EH-5-98-3 Gal1-DacO1 Y Y Y 6.1

EH-3-248-5 CtcN-JCAT-BJ Y Y Y 6.2

EH-5-98-12 DacO4-DacO1 Y Y Y 7.1

EH-5-115-5 DacO1-JCAT-BJ Y Y Y 7.2

EH-5-98-14 OxyS-DacO1-87 Y Y Y 8.1

EH-3-248-6 SsfO1-JCAT-BJ Y Y Y 8.2

EH-5-98-13 OxyS-DacO1-37 Y Y Y 9.1

EH-3-248-4 DacO1-BJ Y Y Y 9.2

EH-3-248-3 OxyR Y 10.2

EH-3-248-8 pSP-G1-empty-BJ Y Y Y 11.2

EH-98-11 DacO1-DacO4 Y

EH-5-98-15 OxyS-DacO1-191 Y

EH-5-98-6 DacO1-no-AB-

tags

Y

EH-5-98-16 DacO1-JCAT-BJ Y

EH-5-98-5 pGAL-DacO1-426 Y Y

86

Figure 3-5 Western blot analysis of DacO1 fusion proteins and other bacterial hydroxylases

in FY251

Strain cultures were lysed with Y-PER and labeled with monoclonal ANTI-FLAG HRP antibody.

Two biological replicates were used for each strain indicated as colony 1 and 2 (C1 and C2),

respectively. Strain EH-5-98-1, EH-5-98-3, EH-5-98-9, EH-5-98-10 and EH-5-98-13 encode

DacO1 fusions with ubiquitin, Gal1, Ubiquitin-γ-IFN, γ-IFN, and OxyS (first 37 amino acids)

respectively. In the latter only the last 461 amino acids of DacO1 are encoded, while in the other

87

DacO1 fusion proteins the complete DacO1 protein is encoded. Strains EH-3-80-2, EH-3-80-3 and

EH-3-98-6 encode DacO1, no hydroxylase and OxyS, respectively. The size (kDa) indicated below

the gels is the expected size of the protein based on the amino acid sequence.

Figure 3-6 Western blot analysis of bacterial hydroxylases in BJ-5464-NpgA

Strain cultures were lysed with Y-PER and labeled with monoclonal ANTI-FLAG HRP antibody.

Two biological replicates were used for each strain indicated as colony 1 and 2 (C1 and C2),

respectively. The size (kDa) indicated below the gels is the expected size of the protein based on

the amino acid sequence.

88

Figure 3-7 Western blot analysis of DacO1-DacO4 and DacO4-DacO1 fusion proteins

Strain cultures were lysed with Y-PER and labeled with monoclonal ANTI-Myc HRP antibody.

Two biological replicates were used for each strain indicated as colony 1 and 2 (C1 and C2),

respectively. The size (kDa) indicated below the gels is the expected size of the protein based on

the amino acid sequence.

For the fusion protein constructs in FY251, ubiquitin-DacO1-C1, Gal1-DacO1-C1 and Ubiquitin-

IFN-DacO1-C1 slight bands are observed in the expected sizes that are not observed in the DacO1-

C1 control (55.0, 86.5 and 72.0 kDa, respectively, Figure 3-5). Bands at the expected sizes were

also observed for OxyS-DacO1-37, SsfO1, OxyS and DacO1-C2, with SsfO1 and OxyS bands

looking more prominent (55.5, 56.2, 55.9 and 55.1 kDa, respectively, Figure 3-5). OxyS-DacO1-

37 and DacO1-C2 show a 50 < band < 75 kDa that is not observed in the empty pSP-G1 negative

89

control. In all bands, proteins of lower mass are noticeable, potentially corresponding to

proteolysis products of the hydroxylases retaining the C-terminal FLAG-tag.

For the protein constructs in BJ-5464-NpgA, clearly less degradation products are observed

(Figure 3-6). OxyS, SsfO1 and PgaE show very prominent bands at the expected sizes (55.9, 56.2

and 53.8 kDa) for both colonies; CtcN-C1 shows a slighter band in the expected size (51.9 kDa)

and DacO1-JCAT-C2 shows an even slighter band of the expected size (55.1 kDa). In addition,

the codon optimization method (COOL vs JCAT) does not seem to significantly alter expression

levels in the case of OxyS, or significantly remediate expression levels in the case of DacO1

(Compare strains EH-3-248-1 and EH-3-248-4 with strains EH-5-115-4 and EH-5-115-5,

respectively, Figure 3-6). Finally, even though BJ-5464-NpgA expression apparently reduced

protein degradation for other hydroxylases (e.g. OxyS and SsfO1) it does not seem to have

promoted significantly expression levels for DacO1 (Figure 3-5 and Figure 3-6).

The DacO4-DacO1 fusion did not yield a stably expressed protein. While DacO4 under pPGK1

and labeled with myc-tag is clearly expressed when unfused to DacO1, the DacO4-DacO1 fusion

is not observed in the gel when under pPGK1 and labeled with myc-tag, in either FY251 or

BJ5464-NpgA background strain (Figure 3-7).

Since less protein degradation was observed for OxyS and SsfO1 in BJ5464-NpgA relative to

FY251 (Compare strain EH-3-248-1 and EH-3-248-6 to strains EH-3-98-6 and EH-5-98-17,

Figure 3-6 and Figure 3-5, respectively) it was logical to test whether the expression of the DacO1

N-terminal fusion proteins would be enhanced in BJ5464-NpgA as well. Strains EH-5-163-1

through EH-5-163-7 were therefore cloned and their DacO1 fusion expression was tested by

western blot (Figure 3-8).

90

Figure 3-8 Western blot analysis of DacO1 and its fusion proteins in BJ5464-NpgA

Strain cultures were lysed with Y-PER and labeled with monoclonal ANTI-FLAG HRP antibody.

Two biological replicates were used for each strain indicated as colony 1 and 2 (C1 and C2),

respectively. Strains EH-5-163-1, through EH-5-163-7 encode DacO1 fusions with ubiquitin,

91

Gal1, Ubiquitin-γ-IFN, γ-IFN, DacO4, OxyS (first 37 amino acids) and OxyS (first 87 amino

acids), respectively. In the last two strains only the last 461 and 411 amino acids of DacO1 are

encoded, respectively, while the other DacO1 fusion proteins the complete DacO1 protein is

encoded. In contrast to DacO1 and its fusion proteins, PgaE expressing strain in this assay has an

FY251 background. The size (kDa) indicated below the gels is the expected size of the protein

based on the amino acid sequence.

In BJ5464-NpgA background, Ubiquitin-DacO1 fusion expression levels seem similar to that of

unfused DacO1; Gal1-DacO1, Ubiquitin-IFN-DacO1-C2 and IFN-DacO1-C2 fusions gave bands

at the expected sizes of 86.5 and 72.0, respectively, with IFN-DacO1-C2 showing the most

prominent band; The OxyS N-terminal fusions to C-terminal DacO1, OxyS-DacO1-37 and OxyS-

DacO1-87 both gave bands at the expected size with OxyS-DacO1-87-C1 showing the more

prominent band (Figure 3-8). Based on these results, the colonies showing better DacO1

expression levels were chosen for analysis of anhydrotetracycline hydroxylation by mass

spectrometry (Scheme 3-2).

Scheme 3-2 Two-step enzymatic conversion of (6-demethyl-)anhydrotetracycline to (6-

demethyl-)6-epitetracycline

Cell lysates of strains expressing hydroxylases that showed stronger bands of the expected sizes

in the Western Blots were incubated in a Tris buffer with anhydrotetracycline and glucose-6-

phosphate. The overnight incubations were then assayed by mass and UV/Vis spectrometry

(Figure 3-9 and Table 3-2). The strains that show hydroxylation levels potentially above

background, as indicated by 443.4 ion counts that are more than double those for the no

92

hydroxylase negative control, are those encoding OxyS, PgaE and Ubiquitin-γ-IFN-DacO1

(labeled as 1, 4 and 9, respectively in Figure 3-9). These three strains, along with three additional

ones encoding γ-IFN-DacO1, OxySDac37 and OxySDac87, also show a maximal emission and/or

excitation in the Δspectra that differs from that of the background strain EH-3-248-8 encoding

empty pSP-G1 (Table 3-2). Therefore, these six strains along with EH-3-248-4 and EH-3-248-8

as controls were assayed again by mass spectrometry in a wider set of conditions prior to larger

scale reactions, isolation and analysis by NMR.

93

Figure 3-9 Mass spectrometry analysis of anhydrotetracycline hydroxylation and reduction

by cell lysates of strains expressing DacO1, its fusion proteins and other bacterial

hydroxylases

The analyzed strains shown are EH-3-248-1, EH-3-248-4, EH-3-248-6, EH-3-248-7, EH-3-248-8,

EH-5-163-1-C1, EH-5-163-2-C1, EH-5-163-2-C2, EH-5-163-3-C2, EH-5-163-4-C2, EH-5-163-

6-C1, EH-5-163-7-C1 encoding hydroxylases OxyS, DacO1, SsfO1, PgaE, pSPG1 (no

hydroxylase negative control), Ubiquitin-DacO1, Gal1-DacO1, Gal1-DacO1, Ubiquitin-γ-IFN-

DacO1, γ-IFN-DacO1, OxyS-DacO1-37 and OxyS-DacO1-87, respectively. Cell lysates were

placed overnight in Tris buffer (100 mM, pH 7.45) containing anhydrotetracycline (5.4 mM),

94

glucose (27.8 mM), NADPH (3 mM), glucose-6-phosphate (10 mM) and mercaptoethanol (18.5

mM).

Table 3-2 UV/Vis spectroscopy results for hydroxylase strains cell lysates incubated with

anhydrotetracycline and glucose-6-phosphate

Strain no. Hydroxylase Max emission

(λexcitation = 280 nm)

Max excitation

(λemission = 500 nm)

1 EH-3-248-1 OxySb 490 390

2 EH-3-248-4 DacO1 590 350

3 EH-3-248-6 SsfO1 580 350

4 EH-3-248-7 PgaEb 580 370

5 EH-3-248-8 pSPG1a 580 350

6 EH-5-163-1-C1 UbiDac 570 350

7 EH-5-163-2-C1 GalDac 570 350

8 EH-5-163-2-C2 GalDac 570 350

9 EH-5-163-3-C2 UbiIFNDacb 690 350

10 EH-5-163-4-C2 IFNDacb 460 350

11 EH-5-163-6-C1 OxySDac37b 420 370

12 EH-5-163-7-C1 OxySDac87b 430 470 a The maximum emission and maximum absorption spectra of EH-3-248-8 encoding no

hydroxylase (empty pSP-G1) are shown in italics. For all other strains, the maximum value shown

are from the Δspectrum of the corresponding strain minus the spectrum of the EH-3-248-8 strain

encoding no hydroxylase (empty pSP-G1).

b Maximum emission and excitation values for the Δspectra that are more than 10 nm different

than the corresponding values obtained for strain EH-3-248-8 encoding no hydroxylase (empty

pSP-G1) are shown in bold.

In order to select strains and conditions for larger scale reactions, strains encoding hydroxylases

that performed favorably according to the Western Blot, mass spectrometry and UV/Vis

spectroscopy results were analyzed in a larger set of conditions using mass spectrometry.

Following cell lysis, lysates were incubated overnight in four different conditions (Table 3-3) and

analyzed by mass spectrometry. The rationale behind trying different conditions was that given

that the conversion rates for all non-OxyS hydroxylases were significantly lower than for OxyS

(Figure 3-9), the reaction should be optimized to allow enough product to be isolated for NMR

95

analysis. The strains that show hydroxylation levels potentially above background, as indicated by

443.4 ion counts that are more than double those for the no hydroxylase negative control encoding

empty pSP-G1, are OxyS and PgaE, with OxyS-DacO1-37 as the closest runner up (Figure 3-10).

These four strains will be used for a larger scale lysate experiment followed by product isolation

and analysis by NMR spectroscopy. Condition D, maximizing the m/z peak associated with

tetracycline ([M + H]+) per amount of cell lysate used will be used to attempt the isolation of

tetracycline or its 6-epimer and condition B maximizing the m/z peak associated with 5a(11a)-

dehydrotetracycline ([M + H]+) will be used to attempt the isolation of the latter or its 6-epimer.

Table 3-3 Assay conditions for anhydrotetracycline hydroxylation and reduction in cell

lysates of strains expressing DacO1, its fusion proteins and other bacterial hydroxylases

Condition no.a Atc (mM) G6P (10 mM) Cell lysate dilution factor into reaction mix

A 5.4 + 5X

B 5.4 - 5X

C 21.6 + 5X

D 21.6 + 10X aCell lysates were placed overnight in Tris buffer (100 mM, pH 7.45) containing in addition to

anhydrotetracycline and glucose-6-phosphate at the concentrations mentioned above, glucose

(27.8 mM), NADPH (3 mM) and mercaptoethanol (18.5 mM).

96

Figure 3-10 Mass spectrometry analysis of anhydrotetracycline hydroxylation and reduction

in cell lysates of strains expressing DacO1, its fusion proteins and other bacterial

hydroxylases

The analyzed strains shown EH-3-248-1, EH-3-248-4, EH-3-248-7, EH-3-248-8, EH-5-163-3-C2,

EH-5-163-4-C2, EH-5-163-6-C1, EH-5-163-7-C1 are encoding hydroxylases OxyS, DacO1,

PgaE, pSPG1 (no hydroxylase negative control), Ubiquitin-γ-IFN-DacO1, γ-IFN-DacO1, OxyS-

DacO1-37 and OxyS-DacO1-87, respectively.

While for both OxyS and PgaE, in the presence of G6P there is an increase in 445 ion counts

associated with the product of both hydroxylation and reduction reactions (Figure 2-3 and Figure

3-10), the PgaE samples are consistently associated with higher 445/443 ion count ratios relative

97

to the OxyS samples in the presence of G6P (Figure 3-11). This supports that in the case of PgaE

hydroxylation a larger fraction of the hydroxylation product ([M-H]+ = 443) gets further reduced

([M-H]+ = 445).

Figure 3-11 Mass spectrometry analysis of anhydrotetracycline hydroxylation and reduction

in cell lysates of strains expressing OxyS and PgaE

The analyzed strains shown are EH-3-248-1 and EH-3-248-7 encoding hydroxylases OxyS and

PgaE, respectively in condition A (Table 3-3). TIC is total ion count. 443 and 445 refer to ion

counts of these m/z values.

3.5 Discussion

3.5.1 Optimizing DacO1 expression in S. cerevisiae – rational design

DacO1 is hypothesized to be the 6-hydroxylase enzyme in the dactylocycline pathway based on

its homology to OxyS, the 6-hydroxylase of the oxytetracycline pathway. Based on this

assignment, DacO1 is performing its 6-hydroxylase function when heterologously expressed

together with the rest of the dactylocyclinone pathway in S. lividans.(2) Heterologous expression

of DacO1 exclusively does not yield a stably expressed protein in either E. coli or Stryptomyces,(1)

and as shown in this study neither does it stably heterolougsly expressed in S. cerevisiae (Figure

3-1). These results taken together with the importance of coexpression of other pathway proteins

to the function of other polyketide enzymes,(19, 20) led to the hypothesis that coexpression of

98

pathway proteins might be beneficial for DacO1 stable expression in S. cerevisiae. For the

particular proteins whose coexpression with DacO1 in S. cerevisiae was attempted, DacJ and

DacM2, this hypothesis was not confirmed (Table 3-1). However, it could be that other or

additional proteins from the dactylocycline pathway are needed for stable heterologous expression

of DacO1. In contrast, the strategy to improve DacO1 expression by N-terminal fusions was

successful (Sections 3.4.5 and 3.5.5).

3.5.2 Searching the bacterial hydroxylase space for 6α-hydroxylation of

anhydrotetracyclines

The successful identification of PgaE as a hydroxylase of anhydrotetracycline is shown in Section

3.4.5 and discussed in Section 3.5.5. PgaE heterologous expression in S. cerevisiae thus

exemplifies the power of natural enzyme diversity harvesting and testing in S. cerevisiae as a

heterologous host.

3.5.3 Developing a microtiter plate assay for anhydrotetracycline

hydroxylation

The microtiter plate assay for hydroxylation of anhydrotetracyclines is functional, as confirmed

by differentiation between the OxyS encoding strain and the negative control strain (Figure 3-3)

and its throughput could be further increased. One approach to increase the assay throughput would

be to employ an automatic liquid dispenser / colony picker as well as to reduce the volume of each

well in the assay.(25, 26) In the current assay format only 2 μL of the 300 μL suspension are

needed for the final UV/Vis analysis. An alternative approach to increase the assay throughput

would be to grow colonies on agar plates supplemented with anhydrotetracycline and monitor the

change in fluorescence by a fluorescence agar plate imager and thus circumventing the need for

99

microtiter plate handling. However, given that anhydrotetracycline is toxic to S. cerevisiae at

elevated concentrations it remains to be determined whether an agar plate UV/Vis assay is

practical.(27, 28) On the other hand, that very toxicity could prove useful to develop a growth-

based assay, as tetracyclines eukaryotic toxicity is significantly lower than that of

anhydrotetracyclines. Thus, conversion of anhydrotetracycline to tetracycline could lead to an

increased growth rate in media containing anhydrotetracycline.(27, 28)

The stereochemical difference between the expected products of anhydrotetracycline

hydroxylation by a 6β-hydroxylase such as OxyS and a 6α-hydroxylase such as DacO1, 5a(11a)-

dehydrotetracycline and 5a(11a)-6-epidehydrotetracycline, respectively, might lead to a difference

in the absorption/emission of the respective products. Thus, an optimized spectrophotometer

measurement protocol for the microtiter plate assay for anhydrotetracycline hydroxylation might

be desired. However, in both cases both absorption and emission are expected to be blue-shifted,

a difference that the current protocol is capable of detecting. DacO1 hydroxylation is supported by

UV/Vis evidence of the current protocol (Figure 3-4) although it is still to be determined whether

5a(11a)-6-epidehydrotetracycline is indeed synthesized from the reaction of DacO1 with

anhydrotetracycline (Section 3.5.5).

3.5.4 Evolving a 6α-hydroxylase of anhydrotetracyclines – DacO1 random

mutagenesis

Importantly, the library-based (Section 3.4.4 and Section 4.4.3) and rational design-based (Section

3.4.1) approaches for optimizing DacO1 for 6α-hydroxylation of anhydrotetracycline in S.

cerevisiae are not mutually exclusive. The winner of the DacO1 error-prone mutagenesis screen

and additional winners of future screens could be sequenced to identify the amino acids at which

mutations occurred and these sites could be chosen for saturation mutagenesis. The resulting

100

library of size ~100 could be then sequenced by the microtiter plate assay for anhydrotetracycline

hydroxylation with a followup on screen hits as shown (Figure 3-4). The sequence of the confirmed

hits can then be fused in DacO1-fusions that were confirmed to be effective in enhancing DacO1

expression levels such as UbiIFN-DacO1, IFN-DacO1, OxyS-DacO1-37 and OxyS-DacO1-87

(Figure 3-8). Alternatively the expression optimized DacO1 fusion porteins could be used for EP-

PCR or saturation mutagenesis as per Section 3.4.4 and Section 4.4.3.

3.5.5 Testing DacO1 expression optimization constructs and other

hydroxylases

The attempt to fuse a stable protein or protein fragment at the N-terminus of DacO1, or a DacO1

fragment in the case of the OxyS fusions, to improve DacO1 expression levels was successful

(Figure 3-8). However, the more stably expressed DacO1 fusions are still poor hydroxylases of

anhydrotetracycline at best compared to OxyS (Figure 3-9 and Figure 3-10). A very likely

hypothesis to explain the poor hydroxylation activity is that the natural substrate of DacO1,

anhydrodactylocyclinone, is different enough from anhydrotetracycline to impede efficient

hydroxylation of the latter. In fact, the hydroxylase SsfO1, whose heterologous expression was

confirmed to be effective in S. cerevisiae (Figure 3-5 and Figure 3-6) did not prove an effective

hydroxylase of anhydrotetracycline at all (Figure 3-9). The native substrate of DacO1 differs from

anhydrotetracycline by having 8-methoxy, 7-chloro and 4a-hydroxy instead of protons in

anhydrotetracycline and it could be that these functional groups serve as recognition elements in

the substrate (Scheme 3-1). The 8-methoxy and 7-chloro substituents can could further have an

electronic contribution to the activation energy of the hydroxylation reaction.

With stably expressed DacO1 fusion proteins at hand such as ubiquitin-γ-IFN-DacO1, γ-IFN-

DacO1, OxyS-DacO1-37 and OxyS-DacO1-87 (Figure 3-8), a few avenues could now be

101

explored: (i) upscale current hydroxylation setup (Table 3-3 and Section 3.4.5) and attempt

isolation of product and analysis by NMR to verify whether the desired 6α-hydroxylation occurred.

(ii) mutagenize stably expressed DacO1 fusion proteins by error-prone PCR (Section 3.4.4) or

saturation mutagenesis (Section 4.4.3) or a combination thereof and then test for

anhydrotetracycline or 6-demethylanhydrotetracycline(29) hydroxylation by the mutants using the

microplate UV/Vis assay (Section 3.4.3); (iii) test DacO1 fusion proteins hydroxylation of the

DacO1 native substrate, anhydrodactylocyclinone (Scheme 3-1) or 6-

demethyanhydrodactylocyclinone; and (iv) test TAN-1612 hydroxylation by coexpressing the

DacO1 fusion proteins and the TAN-1612 pathway (Chapter 4). Given the high biological

variation between the different colonies expressing DacO1-fusion proteins (Figure 3-8), testing

several biological replicates for each strain for these approaches might be crucial.

Specifically for (iii), Anhydrodactylocyclinone and 6-demethylanhydrodactylocyclinone could be

obtained either by deleting the dacO1 and dacO4 or the dacO1, dacO4 and dacM2 genes from

plasmid pDac, respectively, and transforming the resulting plasmid into a K4-114 strain harboring

pDacT1O3E.(2) Importantly, isolating anhydrodactylocyclinone and/or 6-

demethylanhydrodactylocyclinone using such plasmids would support the functional assignment

of DacO1, DacO4 and DacM2. Alternatively the DacO1 native substrate could be pursued by

acidic dehydration of dactylocyclinone,(29) the aglycone of dactylocycline A, which can be

purified either from its native producer Dactylosporangium sp. SC 14051 (ATCC 53693) or from

the heterologous producer K4/pDac-T1O3E.(2, 30, 31) A coculture of a (6-demethyl-

)anhydrodactylocyclinone producing strain and the strain encoding the DacO1 fusion protein is

another potential alternative. Finally, it is also potentially possible to feed the strain encoding the

102

DacO1 fusion protein with crude (6-demethyl-)anhydrodactylocyclinone without prior

purification.

PgaE, a bacterial monooxygenase tested as a potential 6α-hydroxylase candidate (Section 3.4.2)

was found to hydroxylate anhydrotetracycline (Sections 3.4.5 and 3.5.2). Furthermore, in the

presence of PgaE more reduced product was obtained relative to the hydroxylated product, when

compared to OxyS (Figure 3-11). Possible reasons for increased reduction proportion of

hydroxylated anhydrotetracycline in the PgaE sample (Figure 3-11) could be (i) a lower

hydroxylation rate in the case of PgaE results in less overload on reduction capacities (ii) a different

hydroxylation product between OxyS and PgaE, such as different stereochemistry that leads to a

more ready reduction of the PgaE hydroxylation product, (iii) an involvement of OxyS and/or

PgaE in the reduction process either directly by catalyzing it or indirectly by facilitating the process

through product binding. These reasons could be tested by isolating the hydroxylation product

from two different cell lysates expressing OxyS or PgaE and not containing G6P and analyzing

the hydroxylated product by NMR. Studying and comparing the spectra would support or reject

reason (ii). The isolated hydroxylated product can then be added to one of three cell lysates in the

presence of G6P, expressing OxyS or PgaE or no hydroxylase. This will eliminate hydroxylation

levels influence as per reason (i) and will enable testing whether OxyS and/or PgaE are involved

in the reduction as per reason (iii), although it is still perhaps possible that involvement of OxyS

and/or PgaE in the reduction happens only immediately after the hydroxylation before product

release.

3.6 Conclusion

This chapter describes the progress towards 6-demethyl-6-epitetracyclines biosynthesis in S.

cerevisiae (Scheme 3-2). The biosynthesis of 6-demethyl-6-epitetracyclines as opposed to 6-

103

demethyltetracyclines requires a 6α-hydroxylase, such as DacO1, as opposed to a 6β-hydroxylase

such as OxyS (Scheme 3-1). However, past attempts to stably heterologoulsy express DacO1

without the rest of its pathway have been unsuccessful.(1) With the rational design attempt to

stabilize DacO1 expression by N-terminal fusion proteins proving successful (Figure 3-8), the next

step is to test the stably expressed DacO1 fusion proteins in larger scale for 6α-hydroxylation of

anhydrotetracycline and/or anhydrodactylocyclinone and/or 6-demethylanhydrodactylocyclinone

(Section 3.5.5). This will allow product isolation followed by NMR analysis. In addition, given

the differences between DacO1’s native substrate and anhydrotetracycline (Scheme 3-1) and given

that DacO1 fusion proteins are poor hydroxylases of anhydrotetracycline at best (Figure 3-9 and

Figure 3-10), the stably expressed DacO1 fusion proteins could be subjected to mutagenesis

(Section 3.4.4 and Section 4.4.3) to identify mutants that readily hydroxylate the desired

anhydrotetracycline substrate.

In addition to the use of rational design to improve DacO1 expression, this chapter also describes

harvesting natural diversity in bacterial hydroxylases towards the search for a 6α-hydroxylase

(Section 3.4.2). With PgaE, one of these bacterial hydroxylases, proving as a hydroxylase of

anhydrotetracycline (Figure 3-9 and Figure 3-10) the next step is to analyze its product after large

scale preparation and isolation and to verify whether it is performing 6α-hydroxylation or 6-

hydroxylation at all on anhydrotetracycline. It also remains to be determined why a larger

proportion of the hydroxylation product of anhydrotetracycline undergoes reduction when PgaE is

used as the hydroxylase in comparison to when OxyS is used (Figure 3-11) and whether either of

these hydroxylases participates in reduction as well. Participation of OxyS or PgaE in the reduction

reaction could theoretically take place by direct catalysis of the reduction or by facilitating catalysis

through product binding. These open questions could be better understood by adding the isolated

104

hydroxylation product of OxyS and of PgaE, each to three separate cell lysates of strains encoding

OxyS, PgaE and no hydroxylases.

Finally, using a variant of the microtiter plate assay for anhydrotetracycline hydroxylation (Figure

3-2 and Figure 3-3), the promising 6α-hydroxylation candidates identified in this chapter (Table

3-2) were screened for hydroxylation of the fungal anhydrotetracycline TAN-1612 in the next

chapter.

105

3.7 Appendix

Table 3-4 Strains used in this study

Genotype Source/Reference

FY251 MATa leu2Δ1 trpΔ63 ura3-52 his3-200 ATCC 96098

BJ5464-

NpgA

MATα ura3-52 his3-Δ200 leu2-Δ1

trp1 pep4::HIS3 δ:: pADH2-npgA-tADH2 prb1Δ1.6R can1

GAL

a / (15)

EH-3-80-2 FY251 AL-1-31-C7 This Study

EH-3-80-3 FY251 pSP-G1 Chapter 2

EH-3-98-6 FY-251 AL-1-101-C10 Chapter 2

ΕΗ-5-153-7 FY-251 AL-1-173-B-C4 This Study

ΕΗ-5-153-8 FY-251 AL-1-119-A-C7 Chapter 2

EH-3-248-1 BJ5464-NpgA AL-1-101-C10 Chapter 2

EH-3-248-2 BJ5464-NpgA AL-1-119-A-C7 This Study

EH-3-248-3 BJ5464-NpgA AL-1-26-1-C1 This Study

EH-3-248-4 BJ5464-NpgA AL-1-31-C7 This Study

EH-3-248-5 BJ5464-NpgA AL-1-234-B-C2 This Study

EH-3-248-6 BJ5464-NpgA AL-1-239-B-C3 This Study

EH-3-248-7 BJ5464-NpgA AL-1-234-C-C1 This Study

EH-3-248-8 BJ5464-NpgA pSP-G1 Chapter 2

EH-3-270-9 FY-251 AL-2-8-D This Study

EH-5-19-2 FY-251 AL-2-8-D This Study

EH-5-98-1 FY251 EH-5-49-A This Study

EH-5-98-2 FY251 EH-5-49-B This Study

EH-5-98-3 FY251 EH-5-49-C This Study

EH-5-98-4 FY251 EH-5-80-1 This Study

EH-5-98-5 FY251 EH-5-80-2 This Study

EH-5-98-6 FY251 EH-5-80-3 This Study

EH-5-98-7 FY251 EH-5-80-4 This Study

EH-5-98-8 FY251 EH-5-80-5 This Study

EH-5-98-9 FY251 EH-5-80-6 This Study

EH-5-98-10 FY251 EH-5-80-7 This Study

EH-5-98-11 FY251 EH-5-80-8 This Study

EH-5-98-12 FY251 EH-5-80-9 This Study

EH-5-98-13 FY251 EH-5-80-10 This Study

EH-5-98-14 FY251 EH-5-80-11 This Study

EH-5-98-15 FY251 EH-5-80-12 This Study

EH-5-98-16 FY251 AL-1-239-A This Study

EH-5-98-17 FY251 AL-1-239-B This Study

EH-5-98-18 FY251 EH-5-90-A This Study

EH-5-98-19 FY251 EH-5-90-A This Study

EH-5-115-1 FY251 EH-5-90-A EH-5-105-Α This Study

EH-5-115-2 FY251 AL-1-31-C7 EH-5-105-A This Study

106

EH-5-115-3 FY251 pSP-G1 EH-5-105-A This Study

EH-5-115-4 BJ5464-NpgA AL-1-226-A This Study

EH-5-115-5 BJ5464-NpgA AL-1-239-A This Study

EH-5-163-1 BJ5464-NpgA EH-5-49-A This Study

EH-5-163-2 BJ5464-NpgA EH-5-49-C This Study

EH-5-163-3 BJ5464-NpgA EH-5-80-6 This Study

EH-5-163-4 BJ5464-NpgA EH-5-80-7 This Study

EH-5-163-5 BJ5464-NpgA EH-5-80-9 This Study

EH-5-163-6 BJ5464-NpgA EH-5-80-10 This Study

EH-5-163-7 BJ5464-NpgA EH-5-80-11 This Study

EH-5-163-8 FY251 AL-1-234-C-C1 This Study a Y. Tang, University of California, Los Angeles, Department of Chemical and Biomolecular

Engineering & Department of Chemistry and Biochemistry, Department of Bioengineering

Table 3-5 Plasmids used in this study

Description Source/Reference

pSP-G1 pTEF1-FLAG-tADH1, pPGK1-MYC-tCYC1, 2 μ, Ura Addgene 64736

pRS413-pGAL1 Shuttle vector (empty), His marker, CEN ATCC 87326

pRS416- pGAL1 Shuttle vector (empty), Ura marker, CEN ATCC 87332

pRS426- pGAL1 Shuttle vector (empty), Ura marker, 2 μ ATCC 87361

AL-1-26-1-C1 pPGK1-oxyR-tCYC1 in pSP-G1 This Studya

AL-1-31-C7 pTEF1-dacO1-tADH1 in pSP-G1 This Study

AL-1-101-C10 pTEF1-oxyS-tADH1 in pSP-G1 Chapter 2

AL-1-119-A-C7 pTEF1-oxyS-tADH1, pPGK1-oxyR-tCYC1 in pSP-G1 Chapter 2

AL-1-173-B-C4 pTEF1-dacO1-tADH1, pPGK1-dacO4-tCYC1 in pSP-

G1

This Study

AL-1-226-A-C2 pTEF1-oxyS-tADH1 in pSP-G1 JCAT codon opt. This Study

AL-1-234-B-C2 pTEF1-ctcN-tADH1 in pSP-G1 JCAT codon opt. This Study

AL-1-234-C-C1 pTEF1-pgaE-tADH1 in pSP-G1 JCAT codon opt. This Study

AL-1-239-A-C2 pTEF1-dacO1-tADH1 in pSP-G1 JCAT codon opt. This Study

AL-1-239-B-C3 pTEF1-ssfO1-tADH1 in pSP-G1 JCAT codon opt. This Study

AL-2-8-D Error-prone mutagenesis library of AL-1-31-C7 This Study

EH-5-49-A pTEF1-UBI4-dacO1-tADH1 in pSP-G1 This Study

EH-5-49-B pTEF1-SOD1-dacO1-tADH1 in pSP-G1 This Study

EH-5-49-C pTEF1-GAL1-dacO1-tADH1 in pSP-G1 This Study

EH-5-80-1 pGAL1-dacO1-tCYC1 in pRS416 This Study

EH-5-80-2 pGAL1-dacO1-tCYC1 in pRS426 This Study

EH-5-80-3 pTEF1-dacO1-tADH1 in pSP-G1 (no antibody tag) This Study

EH-5-80-4 pADH2-dacO1-tCYC1 in pRS416 This Study

EH-5-80-5 pADH2-dacO1-tCYC1 in pRS426 This Study

EH-5-80-6 pTEF1-UBI4-IFNG-dacO1-tADH1 in pSP-G1b This Study

EH-5-80-7 pTEF1-IFNG-dacO1-tADH1 in pSP-G1b This Study

EH-5-80-8 pPGK1-dacO4-dacO1-tCYC1 in pSP-G1 This Study

107

EH-5-80-9 pPGK1-dacO1-dacO4-tCYC1 in pSP-G1 This Study

EH-5-80-10 pTEF1-oxyS-dacO1-37-tADH1 in pSP-G1 This Study

EH-5-80-11 pTEF1-oxyS-dacO1-87-tADH1 in pSP-G1 This Study

EH-5-80-12 pTEF1-oxyS-dacO1-191-tADH1 in pSP-G1 This Study

EH-5-90-A pTEF1-dacO1-tADH1, pPGK1-dacJ-tCYC1 in pSP-G1 This Study

EH-5-105-Α pGPD-dacM2-tCYC1 in pRS413 This Study a See Chapter 2 for sequence.

b the sequence of UBI4 and of IFNG used in the gene contrasts here is partial. See Table 3-6

Table 3-6 Sequences used in this study

Sequences for the hydroxylases DacO1 (GenBank: AFU65892.1), PgaE (PDB: 2QA1_A), SsfO1

(GenBank: GQ409537.1), CtcN (GenBank: D38214.1) and the DacO1 fusion proteins with UBI4

(GenBank: AF134781.1), SOD1 (GenBank: KJ892183.1), GAL1 (NM_001178368.1), IFNG

(GenBank: X62468.1) and OxyS (AAZ78342.1) are shown capitalized within the context of the

pSP-G1 backbone containing pTEF1, the FLAG tag and tADH1 (partial, decapitalized).

Sequences for DacO4 (GenBank: JX262387.1), the DacO1-DacO4 and DacO4-DacO1 fusions and

DacJ (GenBank: JX262387.1) are shown capitalized within the context of the pSP-G1 backbone

containing pPGK1, the myc tag and tCYC1 (partial, decapitalized).

The sequence for DacM2 (GenBank: JX262387.1) is shown along with the pGPD (pTDH3)

promoter capitalized within the context of the pRS413 backbone containing the tCYC1 terminator

(partial, decapitalized).

The sequences for DacO1 in pRS backbone under the control of pGAL1 or pADH2 is shown

capitalized along with the promoter (decapitalized in the case of pGAL1 and capitalized in the case

of pADH2) within the context of the pRS backbone containing the tCYC1 terminator (partial,

decapitalized).

pTEF1-

dacO1-

tADH1-in-

pSP-G1-

AL-1-31-

C7 and AL-

1-173-B-

C4

tcaagtttcagtttcatttttcttgttctattacaactttttttacttcttgctcattagaaagaaagcatagcaatctaatctaa

gttttaattacaagcggccgcATGTTAGTAGCCGGTGGTGGACCAACAGGGTTAT

GGTTGGCTGGTGAACTAAGGTTGGCCGGGGTTAGACCTCTTGTTTTA

GAGCGTAGGACAGAACCCTTTCCACATAGCAAAGCACTGGGTATCCA

TCCAAGAACTATTGAAATGTTGGAGTTGAGAGGCCTTGCTGGTAGAT

TTTTAGATGGCGCCCCGAAGCTGCCGAAAGGTCACTTTGCATCGCTTC

CTGTGCCCTTAGACTATGGTGCAATGGATACTCGACATCCGTACGGT

GTCTACCGTAAACAAGTGGAAACGGAAGCTTTATTAGCCGGCCACGC

AACCAGATTAGGAGTTCCAGTCCGCAGAGGTCATGAACTTGTAGGCC

TAAGACAGCACGATGATGCTGTAGTCGGTACTGTTCAAGGTCCTCAA

GGTAGATACGAAGTCAGAGCCGCTTATCTTGTTGGTTGTGATGGAGG

108

AGGTTCTACCACAAGACGCCTGGCTGGGATAGATTTCCCTGGACAAG

ATCCTGGTGTTGCCGTTTTGATGGCTGACGCAAGATTCCGGGACCCAT

TACCCTCTGATCCAGCTATGGGTCCATTCAGACGCTATGGAGTCCTGA

GACCTGATTTAAGGGCATGGTTTAGTGCTATTCCTTTACCAGAAGAA

CAGGGTTTATGCAGAGTTATGGTTTTTTGGTATGGACGACAATTTGAA

GATAGACGTGCTCCAGTAACTGAGGATGAAATGAGAGCAGCACTTGT

TGATCTAGCGGGAACTGACTTTGGTATGTACGATGTTCAATGGTTGA

CCAGATTCACAGACGCTTCCAGGCAAGCTGCAAGGTATAGATCTGGA

AGGGTTTTCTTAGCTGGAGATGCTGCTCATACTGTCTTCCCCACTGGT

GGCCCAGGTTTGAATGCAGGTTTACAAGATGCCATGAATTTAGGTTG

GAAGTTAGCTGGAGCTGTTCATGGCTGGGGTGGTGCTTTACTAGATT

CATATCATGATGAGCGTTACCCTGTAGCTGAACAAGTTTTGCGTGAC

ACAAGAACACAATGTCTATTGTTAGATCCAGACCCTCGCTTTGTTCCA

TTAAGAGAGACGTTAGCAGAACTATTGAGACTAGAACCCGTAAACA

GATATTTCACCGGACACAACACGGCTCTAGCAGTTAGATACGATTTG

GGTGGTGCCCATCCTGCAACGGGAGGCAGAATGCCAGATGTCTTGGT

CGCAACTGGTGCTGGTCAAAGAAGGTTTTCAGATTGTTGTGTTACCG

GGAGAGGGGTTCTTTTAGTCACTTCTGGTGCAGATAGGGGGGAACTG

TTACGAGGATGGAAAGATAGAGTGGATGTGGTAACAGGTACCGTGG

CAGGACCTCTTGATGCAGCTGATCATTTAGTTAGACCTGATGGTTATG

TTGCATGGGCCGGTGGAGCTGGACACGATGAACCTGGTTTAGAAACT

GCTCTTAGATTATGGTTTGGTGAACCAGTTAGTgtatcgatggattacaaggatgac

gacgataaAatctgagctcttaattaacaattcttcgccagaggtttggtcaagtctccaatcaaggttgtcggct

pPGK1-

dacO4-

tCYC1-AL-

1-173-B-

C4

tggaatggcgggaaagggtttagtaccacatgctatgatgcccactgtgatctccagagcaaagttcgttcgatcgt

actgttactctctctctttcaaacagaattgtccgaatcgtgtgacaacaacagcctgttctcacacactcttttcttctaa

ccaagggggtggtttagtttagtagaacctcgtgaaacttacatttacatatatataaacttgcataaattggtcaatgc

aagaaatacatatttggtcttttctaattcgtagtttttcaagttcttagatgctttctttttctcttttttacagatcatcaagga

agtaattatctactttttacaacaaatataaaacaaggatccATGTCATTCACTGCCAAGGAAGT

TCAATATTTGCGTTCTCAGCAATTAGGAAGATTAGCCACCGTTGGTGC

TGATGGTACACCACATAATGTTCCAGTAGGCTACAGATACAACGCTG

ACTTGGGGACCATTGACATCACAGGAAGGGACTTAAGAAGAAGTAG

GAAATATAGAGATGTTCAGGCTGGCTCGCGGGTGGCATTTATTGTTG

ATGATCTACCGAGCACAGCACCTATAGTCGCCCGCGGGCTGGAGGTG

AGGGGAACAGCTGAGGCGCTTTCTGGTGAAGAAGATTTGATACGTGT

ACGACCTGCAAGAATCGTCACTTGGGGTATTGAAGCAGATTGGCAAG

CTGGTCCTACTGGTAGAACGGTAAGGCCCACCACTCCAAGATCCGCG

ACGgtcgacatggaacagaagttgatttccgaagaagacctcgagtaagcttggtaccgcggctagctaagatc

cgctctaaccgaaaaggaaggagttagacaacctgaagtctaggtccctatttatttttttatagttatgttagtattaag

aacgttatttatatttcaaatttttcttttttttctgtacagacgcgtgtacgcatgtaacattatactgaaaaccttgcttgag

aaggttttgggacgctcgaagatccagctgcattaatgaatcggccaacgcgcggggagaggcggtttgcgtattg

ggcgctcttccgcttcctcgctcactgactcgctgcgctcgg

pTEF1-

oxyS-

tADH1 in

pSP-G1

tcaagtttcagtttcatttttcttgttctattacaactttttttacttcttgctcattagaaagaaagcatagcaatctaatctaa

gttttaattacaagcggccgcATGAGATACGACGTTGTTATCGCTGGTGCTGGTCC

AACTGGTTTGATGTTGGCTTGTGAATTGAGATTGGCTGGTGCTAGAA

CTTTGGTTTTGGAAAGATTGGCTGAACCAGTTGACTTCTCTAAGGCTT

TGGGTGTTCACGCTAGAACTGTTGAATTGTTGGACATGAGAGGTTTG

109

JCAT

codon opt.

(AL-1-226-

A)

GGTGAAGGTTTCCAAGCTGAAGCTCCAAAGTTGAGAGGTGGTAACTT

CGCTTCTTTGGGTGTTCCATTGGACTTCTCTTCTTTCGACACTAGACA

CCCATACGCTTTGTTCGTTCCACAAGTTAGAACTGAAGAATTGTTGAC

TGGTAGAGCTTTGGAATTGGGTGCTGAATTGAGAAGAGGTCACGCTG

TTACTGCTTTGGAACAAGACGCTGACGGTGTTACTGTTTCTGTTACTG

GTCCAGAAGGTCCATACGAAGTTGAATGTGCTTACTTGGTTGGTTGT

GACGGTGGTGGTTCTACTGTTAGAAAGTTGTTGGGTATCGACTTCCCA

GGTCAAGACCCACACATGTTCGCTGTTATCGCTGACGCTAGATTCAG

AGAAGAATTGCCACACGGTGAAGGTATGGGTCCAATGAGGCCATAC

GGTGTTATGAGACACGACTTGAGAGCTTGGTTCGCTGCTTTCCCATTG

GAACCAGACGTTTACAGAGCTACTGTTGCTTTCTTCGACAGACCATA

CGCTGACAGAAGAGCGCCAGTTACTGAAGAAGATGTTAGAGCTGCTT

TGACTGAAGTTGCTGGTTCTGACTTCGGTATGCACGACGTTAGATGGT

TGTCTCGTTTGACTGACACTTCTCGTCAAGCTGAAAGATACAGAGAT

GGTAGAGTTTTGTTGGCTGGTGACGCTTGTCACATCCACTTGCCAGCT

GGTGGTCAAGGTTTGAACTTGGGTTTCCAAGACGCTGTTAACTTGGG

TTGGAAGTTGGGTGCTACTATCGCTGGTACTGCTCCACCAGAATTGTT

GGACACTTACGAAGCTGAAAGAAGGCCAATCGCTGCTGGTGTTTTGA

GAAACACTAGAGCGCAAGCTGTTTTGATCGACCCAGACCCAAGATAC

GAAGGTTTGAGAGAATTGATGATCGAATTGTTGCACGTTCCAGAAAC

TAACAGATACTTGGCTGGTTTGATCTCTGCTTTGGACGTTAGATACCC

AATGGCTGGTGAACACCCATTGTTGGGTAGAAGAGTTCCAGACTTGC

CATTGGTTACTGAAGATGGTACTAGACAATTGTCTACTTACTTCCACG

CTGCTAGAGGTGTTTTGTTGACTTTGGGTTGTGACCAACCATTGGCTG

ACGAAGCTGCTGCTTGGAAGGACAGAGTTGACTTGGTTGCTGCTGAA

GGTGTTGCTGACCCAGGTTCTGCTGTTGACGGATTAACTGCTTTGTTG

GTTAGACCAGACGGTTACATCTGTTGGACTGCTGCTCCAGAAACTGG

TACTGACGGTTTGACTGACGCTTTGAGAACTTGGTTCGGTCCACCTGC

TATGgtatcgatggattacaaggatgacgacgataagatctgagctcttaattaacaattcttcgccagaggtttg

gtcaagtctccaatcaaggttgtcggct

pTEF1-

dacO1-

tADH1 in

pSP-G1

JCAT

codon opt. (

AL-1-239-

A)

tcaagtttcagtttcatttttcttgttctattacaactttttttacttcttgctcattagaaagaaagcatagcaatctaatctaa

gttttaattacaagcggccgcATGTTGGTTGCTGGTGGTGGTCCAACTGGTTTGTG

GTTGGCTGGTGAATTGAGATTGGCTGGTGTTAGACCATTGGTTTTGGA

AAGAAGAACTGAACCATTCCCACACTCTAAGGCTTTGGGTATCCACC

CAAGAACTATCGAAATGTTGGAATTGAGAGGTTTGGCTGGTAGATTC

TTGGACGGTGCTCCAAAGTTGCCAAAGGGTCACTTCGCTTCTTTGCCA

GTTCCATTGGACTACGGTGCTATGGACACTAGACACCCATACGGTGT

TTACAGAAAGCAAGTTGAAACTGAAGCTTTGTTGGCTGGTCACGCTA

CTAGATTGGGTGTTCCAGTTAGAAGAGGTCACGAATTGGTTGGTTTG

AGACAACACGACGACGCTGTTGTTGGTACTGTTCAAGGTCCACAAGG

TAGATACGAAGTTAGAGCTGCTTACTTGGTTGGTTGTGACGGTGGTG

GTTCTACTACTAGAAGATTGGCTGGTATCGACTTCCCAGGTCAAGAC

CCAGGTGTTGCTGTTTTGATGGCTGACGCTAGATTCAGAGATCCATTG

CCATCTGACCCAGCTATGGGTCCATTCAGAAGATACGGTGTTTTAAG

ACCAGACTTGAGAGCTTGGTTCTCTGCTATCCCATTGCCAGAAGAAC

AAGGTTTGTGTAGAGTTATGGTTTTCTGGTACGGTAGACAATTCGAA

110

GATAGAAGAGCGCCAGTTACTGAAGATGAAATGAGAGCTGCTTTGGT

TGACTTGGCTGGTACTGACTTCGGTATGTACGACGTTCAATGGTTGAC

TAGATTCACTGACGCTTCTCGTCAAGCTGCTAGATACAGATCGGGTA

GAGTTTTCTTGGCTGGTGACGCTGCTCACACTGTTTTCCCAACTGGTG

GTCCAGGTTTGAACGCTGGTTTGCAAGACGCTATGAACTTGGGTTGG

AAGTTGGCTGGTGCTGTTCACGGTTGGGGTGGTGCTTTGTTGGACTCT

TACCACGACGAAAGATACCCAGTTGCTGAACAAGTTTTGAGAGACAC

TAGAACTCAATGTTTGTTGTTGGACCCAGACCCAAGATTCGTTCCATT

GAGAGAAACTTTGGCTGAATTGTTGAGATTGGAACCAGTTAACAGAT

ACTTCACTGGTCACAACACTGCTTTGGCTGTTAGATACGACTTGGGTG

GTGCTCACCCAGCTACTGGTGGTAGAATGCCAGACGTTTTGGTTGCT

ACTGGTGCTGGTCAAAGAAGATTCTCTGACTGTTGTGTTACTGGTAG

AGGTGTTTTGTTGGTTACTTCTGGTGCTGACAGAGGTGAATTGTTGAG

AGGTTGGAAGGACAGAGTTGACGTTGTTACTGGTACTGTTGCTGGTC

CATTGGACGCTGCTGACCACTTGGTTAGACCAGACGGTTACGTTGCTT

GGGCTGGTGGTGCTGGTCACGACGAACCAGGTTTGGAAACTGCTTTG

AGATTGTGGTTCGGTGAACCAGTTTCTgtatcgatggattacaaggatgacgacgataag

atctgagctcttaattaacaattcttcgccagaggtttggtcaagtctccaatcaaggttgtcggct

pTEF1-

ctcN-

tADH1 in

pSP-G1

JCAT

codon opt.

(AL-1-234-

B-C2)

tcaagtttcagtttcatttttcttgttctattacaactttttttacttcttgctcattagaaagaaagcatagcaatctaatctaa

gttttaattacaagcggccgcATGGTTGTTGCTGGTGCTGGTCCAACTGGTTTGAT

GTTGGCTTGTGAATTGGCTTTGGGTGGTGCTAGAGCTGTTGTTGTTGA

AAGAAGAAGAGAACCAGAAAAGCACTCTAAGGCTATGGGTATGCAA

GGTAGAACTGTTGAATTGTTGGAATTGAGAGGTTTGTTGGACAGATT

CAAGGAAGGTGCTGGTGTTTTGCAAGGTGGTAACTTCGCTTCTTTGG

GTGTTCCAATGAGATTCGAAGAATTTGACACTCAACCAACTCAATAC

GTTTTGTTGGTTCCACAATTGAGAACTGAAGAATTGTTGGCTGAAAG

AGCTAGAGAATTGGGTGTTAGAATCGTTAGAGGTTCTGGTGTTACTG

GTTTCGCTCAAGACGCTGACGGTGTTACTGTTGAAACTGACACTGGTT

TGTTGAGAGCTAGATACTTGGTTGGTTGTGACGGTGGTTCTACTGTTA

GAAAGGCTGCTGGTATCGGTTTCACTGGTCAAGACCCACACATGTAC

GCTTTGATCGGTGACATGAGATTCTCTGGTGACTTGCCAAGAGGTGA

AGGTTTGGGTCCAATGAGGCCAGTTGGTTTGGTTTACAGAGCTACTG

TTGCTTGGTTCGACAGACCATTCGCTGACAGAAGAGCGCCAGTTACT

GAAGAAGAAATGAGAGCTGCTTTGGTTGAACACACTGGTTCTGACCA

CGGTATGCACGACGTTACTTGGTTGTCTCGTTTGACTGACGTTTCTCG

TTTGGCTGACTCTTACAGATTGGGTAGAGTTTTGTTGGCTGGTGACGC

TGCTCACATCCACTTGCCAGCTGGTGGTCAAGGTTTGAACTTGGGTTT

CCAAGACGCTGTTAACTTGGGTTGGAAGTTGGCTGCTGTTGTTAGAG

GTCACGGTACTGAAGAATTGTTGGACTCTTACGGTAGAGAAAGAAGG

CCAATCGCTGACGGTGTTGTTAGAAACACTAGAACTCAAGCTGTTTT

GATCGACCCAGACCCAAGATACGAAGCTCTCAGACAAACTTTGCCAC

ACTTGATGGCTTTGCCAGACACTAACAGACACATGGCTGGTATGTTG

TCTGGTTTCGACGTTGCTTACGGTGGTGGTGACCACCCATTGGTTGGT

AGAAGAATGCCAGACGCTGAATTGATCACTGCTGACGGTCCAAGAA

GAATCTCTGACTGTTTCGCTGGTGCTAGAGGTTTGTTGTTGTTGCCAG

AACAAGGTCCAACTGCTTCTCCATTGGCTGCTTGGGCTGACAGAGTT

111

GACACTTTGACTGTTAAGTCTGGTGGTCCAGACCCAGACACTGCTCA

CTTGGTTAGACCAGACGGTTACGTTGCTTGGGCTGGTGAACCAGCTA

GAACTGAAGAATTGCACCACGCTGCTACTACTTGGTTCGGTGCTGCT

GCTgtatcgatggattacaaggatgacgacgataagatctgagctcttaattaacaattcttcgccagaggtttggt

caagtctccaatcaaggttgtcggct

pTEF1-

pgaE-

tADH1 in

pSP-G1

JCAT

codon opt.

(AL-1-234-

C-C1)

tcaagtttcagtttcatttttcttgttctattacaactttttttacttcttgctcattagaaagaaagcatagcaatctaatctaa

gttttaattacaagcggccgcATGGACGCTGCTGTTATCGTTGTTGGTGCTGGTCC

AGCTGGTATGATGTTGGCTGGTGAATTGAGATTGGCTGGTGTTGAAG

TTGTTGTTTTGGAAAGATTGGTTGAAAGAACTGGTGAATCTCGTGGTT

TGGGTTTCACTGCTAGAACTATGGAAGTTTTCGACCAAAGAGGTATC

TTGCCAAGATTCGGTGAAGTTGAAACTTCTACTCAAGGTCACTTCGGT

GGTTTGCCAATCGACTTCGGTGTTTTGGAAGGTGCTTGGCAAGCTGCT

AAGACTGTTCCACAATCTGTTACTGAAACTCACTTGGAACAATGGGC

TACTGGTTTGGGTGCTGACATCAGAAGAGGTCACGAAGTTTTGTCTTT

GACTGACGACGGTGCTGGTGTTACTGTTGAAGTTAGAGGTCCAGAAG

GTAAGCACACTTTGAGAGCTGCTTACTTGGTTGGTTGTGACGGTGGT

AGATCGTCTGTTAGAAAGGCTGCTGGTTTCGACTTCCCAGGTACTGCT

GCTACTATGGAAATGTACTTGGCTGACATCAAGGGTGTTGAATTGCA

ACCAAGAATGATCGGTGAAACTTTGCCAGGTGGTATGGTTATGGTTG

GTCCATTGCCAGGTGGTATCACTAGAATCATCGTTTGTGAAAGAGGT

ACTCCACCACAAAGAAGAGAAACTCCACCATCTTGGCACGAAGTTGC

TGACGCTTGGAAGAGATTGACTGGTGACGACATCGCTCACGCTGAAC

CAGTTTGGGTTTCTGCTTTCGGTAACGCTACTAGACAAGTTACTGAAT

ACAGAAGAGGTAGAGTTATCTTGGCTGGTGACTCTGCTCACATCCAC

TTGCCAGCTGGTGGTCAAGGTATGAACACTTCTATCCAAGACGCTGT

TAACTTGGGTTGGAAGTTGGGTGCTGTTGTTAACGGTACTGCTACTGA

AGAATTGTTGGACTCTTACCACTCTGAAAGACACGCTGTTGGTAAGA

GATTGTTGATGAACACTCAAGCTCAAGGTTTGTTGTTCTTGTCTGGTC

CAGAAGTTCAACCATTGAGAGATGTTTTGACTGAATTGATCCAATAC

GGTGAAGTTGCTAGACACTTGGCTGGTATGGTTTCTGGTTTGGAAATC

ACTTACGACGTTGGTACTGGTTCTCACCCATTGTTGGGTAAGAGAAT

GCCAGCTTTGGAATTGACTACTGCTACTAGAGAAACATCTTCTACTG

AATTATTACACACTGCTAGAGGTGTTTTGTTGGACTTGGCTGACAACC

CAAGATTGAGAGCTAGAGCTGCTGCTTGGTCTGACAGAGTTGACATC

GTTACTGCTGTTCCAGGTGAAGTTTCTGCTACTTCTGGTTTGAGAGAC

ACTACTGCTGTTTTGATAAGACCAGACGGTCACGTTGCTTGGGCTGCT

CCAGGTTCTCACCACGACTTGCCAATGGCTTTGGAAAGATGGTTCGG

TGCTCCATTGACTGGTgtatcgatggattacaaggatgacgacgataagatctgagctcttaattaac

aattcttcgccagaggtttggtcaagtctccaatcaaggttgtcggct

pTEF1-

ssfO1-

tADH1 in

pSP-G1

JCAT

codon opt.

tcaagtttcagtttcatttttcttgttctattacaactttttttacttcttgctcattagaaagaaagcatagcaatctaatctaa

gttttaattacaagcggccgcATGGAACCAGAAGTTGTTGTTGCTGGTGCTGGTCC

AGTTGGTTTGATGTTGGCTTGTGAATTGAGAAGGCAAGGTGTTGGTG

TTGTTTTGGTTGAAAGAACTACTGAACCAGCTAACGACAACAGAGCG

CAAGCTCTCCACGGTAGAACTATCCCAGTTTTGGACAGAAGAGGTTT

GTTGCCAAGATTCAGAGCTGCTGAAAGAGAATTGAACGGTGGTGGTT

CTGGTGGTACTGAATCTCACAGAAAAAGACCATTGCCAAGAGGTCAC

112

(AL-1-239-

B-C3)

TTCGCTGGTATCACTGGTTTATTACCATCTGCTCCAGACCCAGACCCA

GACGTTCCACCAGTTGTTTTCGTTCCACAATGGGTTACTACTAGATTG

TTGACTGAACACGCTGCTGAATTGGGTGTTGTTTTGCACAGAGGTTCT

GAAATCACTGCTGTTGAACAAGACGCTGACGGTGTTACTGTTGCTGT

TTCTGGTGGTACTGCTCCAGCTAGATTGAGAGCTAGATACTTGATAG

GTTGTGACGGTGCTAGATCGGTTGTTAGAAGATCGGCTGGTATCGAC

TTCGCTGGTACTGGTGCTACTTTGTCTACTTTGGCTGCTGAAGTTCAC

TTGGACGACCCAGCTGACTTGCCAGTTGGTTGGCACAGAACTCAACA

CGGTTGTACTGTTATCTCTCCACACCCAGAAGGTGGTCACTCTCGTGT

TGTTGTTATCGACTTCTCTGGTCCAGACGCTGACAGAGAAGCTCCAGT

TACTTTGACTGAATTGGAAGAAAGAATCGCTGGTGTTTTGGGTAGAA

AGGTTGCTATGTCTGACTTGAAGAACGCTCAAAGATACGGTGACGCT

GCTAGATTGGCTGACAGATACAGAGTTGGTAGAGTTTTCTTGGCTGG

TGACGCTGCTCACATCCACTACCCAGTTGGTGGTCAAGGTTTGAACA

TCGGTTTGCAAGACGCTGTTAACTTGGCTTGGAAGTTGGCTGCTGACT

TGAGAGGTTGGGCTCCAGACGGTTTGTTGGACACTTACCACACTGAA

AGATACCCAGTTGCTGACTCTGTTTTGACTCACACTAGAGCGCAAGTT

GCTTTGTTGGCTCCAGGTCCAAGAATCGACGCTTTGAGAACTTTGTTC

ACTGACTTGATCGGTTTGGCTGACGTTTCTCGTTACTTGTCTGAAAAG

ATGTCTGCTGCTGACGTTAGATACGACATGGGTGAAGCTGAACCACA

CCCATTGACTGGTAGATTCGCTCCATCTTTGAGATTGGCTACTGAACA

AGGTGAAAGAAGATTGGCTGACTTGTTGGCTGGTGGTAGACCATTGT

TGTTGGACTTGTCTGGTAGAGCTGCTACTGCTGCTGTTGCTGCTAGAT

GGGCTGACAGAGTTGACGTTCACGCTGCTACTTGTCCAGCTGAACCA

GCTTTGGGTTCTTTGTTGATCAGACCAGACTCTTACGTTGCTTGGGCT

TGTGCTCCAGGTGACGCTCCAGACCAAGTTGAAAGAGGTTTGGAAGC

TGCTTTGAGAAGATGGTTCGGTGCTCCAGACCACCAAgtatcgatggattacaa

ggatgacgacgataagatctgagctcttaattaacaattcttcgccagaggtttggtcaagtctccaatcaaggttgtc

ggct

pTEF1-

UBI4-

dacO1-

tADH1 in

pSP-G1

(EH-5-49-

A)

tcaagtttcagtttcatttttcttgttctattacaactttttttacttcttgctcattagaaagaaagcatagcaatctaatctaa

gttttaattacaagcggccgccattATGCAGATCTTCGTCAAGACGTTAACCGGTAA

AACCATAACTCTAGAAGTTGAATCTTCCGATACCATCGACAACGTTA

AGTCGAAAATTCAAGACAAGGAAGGCATTCCACCTGATCAACAAAG

ATTGATCTTTGCCGGTAAGCAGCTCGAGGACGGTAGAACGCTGTCTG

ATTACAACATTCAGAAGGAGTCGACCTTACATCTTGTCTTAAGACTA

AGAGGTGGTATGTTAGTAGCCGGTGGTGGACCAACAGGGTTATGGTT

GGCTGGTGAACTAAGGTTGGCCGGGGTTAGACCTCTTGTTTTAGAGC

GTAGGACAGAACCCTTTCCACATAGCAAAGCACTGGGTATCCATCCA

AGAACTATTGAAATGTTGGAGTTGAGAGGCCTTGCTGGTAGATTTTT

AGATGGCGCCCCGAAGCTGCCGAAAGGTCACTTTGCATCGCTTCCTG

TGCCCTTAGACTATGGTGCAATGGATACTCGACATCCGTACGGTGTCT

ACCGTAAACAAGTGGAAACGGAAGCTTTATTAGCCGGCCACGCAACC

AGATTAGGAGTTCCAGTCCGCAGAGGTCATGAACTTGTAGGCCTAAG

ACAGCACGATGATGCTGTAGTCGGTACTGTTCAAGGTCCTCAAGGTA

GATACGAAGTCAGAGCCGCTTATCTTGTTGGTTGTGATGGAGGAGGT

TCTACCACAAGACGCCTGGCTGGGATAGATTTCCCTGGACAAGATCC

113

TGGTGTTGCCGTTTTGATGGCTGACGCAAGATTCCGGGACCCATTACC

CTCTGATCCAGCTATGGGTCCATTCAGACGCTATGGAGTCCTGAGAC

CTGATTTAAGGGCATGGTTTAGTGCTATTCCTTTACCAGAAGAACAG

GGTTTATGCAGAGTTATGGTTTTTTGGTATGGACGACAATTTGAAGAT

AGACGTGCTCCAGTAACTGAGGATGAAATGAGAGCAGCACTTGTTGA

TCTAGCGGGAACTGACTTTGGTATGTACGATGTTCAATGGTTGACCA

GATTCACAGACGCTTCCAGGCAAGCTGCAAGGTATAGATCTGGAAGG

GTTTTCTTAGCTGGAGATGCTGCTCATACTGTCTTCCCCACTGGTGGC

CCAGGTTTGAATGCAGGTTTACAAGATGCCATGAATTTAGGTTGGAA

GTTAGCTGGAGCTGTTCATGGCTGGGGTGGTGCTTTACTAGATTCATA

TCATGATGAGCGTTACCCTGTAGCTGAACAAGTTTTGCGTGACACAA

GAACACAATGTCTATTGTTAGATCCAGACCCTCGCTTTGTTCCATTAA

GAGAGACGTTAGCAGAACTATTGAGACTAGAACCCGTAAACAGATA

TTTCACCGGACACAACACGGCTCTAGCAGTTAGATACGATTTGGGTG

GTGCCCATCCTGCAACGGGAGGCAGAATGCCAGATGTCTTGGTCGCA

ACTGGTGCTGGTCAAAGAAGGTTTTCAGATTGTTGTGTTACCGGGAG

AGGGGTTCTTTTAGTCACTTCTGGTGCAGATAGGGGGGAACTGTTAC

GAGGATGGAAAGATAGAGTGGATGTGGTAACAGGTACCGTGGCAGG

ACCTCTTGATGCAGCTGATCATTTAGTTAGACCTGATGGTTATGTTGC

ATGGGCCGGTGGAGCTGGACACGATGAACCTGGTTTAGAAACTGCTC

TTAGATTATGGTTTGGTGAACCAGTTAGTgtatcgatggattacaaggatgacgacgat

aagatctgagctcttaattaacaattcttcgccagaggtttggtcaagtctccaatcaaggttgtcggct

pTEF1-

SOD1-

dacO1-

tADH1 in

pSP-G1

(EH-5-49-

B)

tcaagtttcagtttcatttttcttgttctattacaactttttttacttcttgctcattagaaagaaagcatagcaatctaatctaa

gttttaattacaagcggccgccattATGGCGACGAAGGCCGTGTGCGTGCTGAAGGG

CGACGGCCCAGTGCAGGGCATCATCAATTTCGAGCAGAAGGAAAGT

AATGGACCAGTGAAGGTGTGGGGAAGCATTAAAGGACTGACTGAAG

GCCTGCATGGATTCCATGTTCATGAGTTTGGAGATAATACAGCAGGC

TGTACCAGTGCAGGTCCTCACTTTAATCCTCTATCCAGAAAACACGGT

GGGCCAAAGGATGAAGAGAGGCATGTTGGAGACTTGGGCAATGTGA

CTGCTGACAAAGATGGTGTGGCCGATGTGTCTATTGAAGATTCTGTG

ATCTCACTCTCAGGAGACCATTGCATCATTGGCCGCACACTGGTGGT

CCATGAAAAAGCAGATGACTTGGGCAAAGGTGGAAATGAAGAAAGT

ACAAAGACAGGAAACGCTGGAAGTCGTTTGGCTTGTGGTGTAATTGG

GATGTTAGTAGCCGGTGGTGGACCAACAGGGTTATGGTTGGCTGGTG

AACTAAGGTTGGCCGGGGTTAGACCTCTTGTTTTAGAGCGTAGGACA

GAACCCTTTCCACATAGCAAAGCACTGGGTATCCATCCAAGAACTAT

TGAAATGTTGGAGTTGAGAGGCCTTGCTGGTAGATTTTTAGATGGCG

CCCCGAAGCTGCCGAAAGGTCACTTTGCATCGCTTCCTGTGCCCTTAG

ACTATGGTGCAATGGATACTCGACATCCGTACGGTGTCTACCGTAAA

CAAGTGGAAACGGAAGCTTTATTAGCCGGCCACGCAACCAGATTAGG

AGTTCCAGTCCGCAGAGGTCATGAACTTGTAGGCCTAAGACAGCACG

ATGATGCTGTAGTCGGTACTGTTCAAGGTCCTCAAGGTAGATACGAA

GTCAGAGCCGCTTATCTTGTTGGTTGTGATGGAGGAGGTTCTACCAC

AAGACGCCTGGCTGGGATAGATTTCCCTGGACAAGATCCTGGTGTTG

CCGTTTTGATGGCTGACGCAAGATTCCGGGACCCATTACCCTCTGATC

CAGCTATGGGTCCATTCAGACGCTATGGAGTCCTGAGACCTGATTTA

114

AGGGCATGGTTTAGTGCTATTCCTTTACCAGAAGAACAGGGTTTATG

CAGAGTTATGGTTTTTTGGTATGGACGACAATTTGAAGATAGACGTG

CTCCAGTAACTGAGGATGAAATGAGAGCAGCACTTGTTGATCTAGCG

GGAACTGACTTTGGTATGTACGATGTTCAATGGTTGACCAGATTCAC

AGACGCTTCCAGGCAAGCTGCAAGGTATAGATCTGGAAGGGTTTTCT

TAGCTGGAGATGCTGCTCATACTGTCTTCCCCACTGGTGGCCCAGGTT

TGAATGCAGGTTTACAAGATGCCATGAATTTAGGTTGGAAGTTAGCT

GGAGCTGTTCATGGCTGGGGTGGTGCTTTACTAGATTCATATCATGAT

GAGCGTTACCCTGTAGCTGAACAAGTTTTGCGTGACACAAGAACACA

ATGTCTATTGTTAGATCCAGACCCTCGCTTTGTTCCATTAAGAGAGAC

GTTAGCAGAACTATTGAGACTAGAACCCGTAAACAGATATTTCACCG

GACACAACACGGCTCTAGCAGTTAGATACGATTTGGGTGGTGCCCAT

CCTGCAACGGGAGGCAGAATGCCAGATGTCTTGGTCGCAACTGGTGC

TGGTCAAAGAAGGTTTTCAGATTGTTGTGTTACCGGGAGAGGGGTTC

TTTTAGTCACTTCTGGTGCAGATAGGGGGGAACTGTTACGAGGATGG

AAAGATAGAGTGGATGTGGTAACAGGTACCGTGGCAGGACCTCTTGA

TGCAGCTGATCATTTAGTTAGACCTGATGGTTATGTTGCATGGGCCGG

TGGAGCTGGACACGATGAACCTGGTTTAGAAACTGCTCTTAGATTAT

GGTTTGGTGAACCAGTTAGTgtatcgatggattacaaggatgacgacgataagatctgagctctt

aattaacaattcttcgccagaggtttggtcaagtctccaatcaaggttgtcggct

pTEF1-

GAL1-

dacO1-

tADH1 in

pSP-G1

(EH-5-49-

C)

tcaagtttcagtttcatttttcttgttctattacaactttttttacttcttgctcattagaaagaaagcatagcaatctaatctaa

gttttaattacaagcggccgcATGACTAAATCTCATTCAGAAGAAGTGATTGTACC

TGAGTTCAATTCTAGCGCAAAGGAATTACCAAGACCATTGGCCGAAA

AGTGCCCGAGCATAATTAAGAAATTTATAAGCGCTTATGATGCTAAA

CCGGATTTTGTTGCTAGATCGCCTGGTAGAGTCAATCTAATTGGTGAA

CATATTGATTATTGTGACTTCTCGGTTTTACCTTTAGCTATTGATTTTG

ATATGCTTTGCGCCGTCAAAGTTTTGAACGAGAAAAATCCATCCATT

ACCTTAATAAATGCTGATCCCAAATTTGCTCAAAGGAAGTTCGATTT

GCCGTTGGACGGTTCTTATGTCACAATTGATCCTTCTGTGTCGGACTG

GTCTAATTACTTTAAATGTGGTCTCCATGTTGCTCACTCTTTTCTAAA

GAAACTTGCACCGGAAAGGTTTGCCAGTGCTCCTCTGGCCGGGCTGC

AAGTCTTCTGTGAGGGTGATGTACCAACTGGCAGTGGATTGTCTTCTT

CGGCCGCATTCATTTGTGCCGTTGCTTTAGCTGTTGTTAAAGCGAATA

TGGGCCCTGGTTATCATATGTCCAAGCAAAATTTAATGCGTATTACG

GTCGTTGCAGAACATTATGTTGGTGTTAACAATGGCGGTATGGATCA

GGCTGCCTCTGTTTGCGGTGAGGAAGATCATGCTCTATACGTTGAGTT

CAAACCGCAGTTGAAGGCTACTCCGTTTAAATTTCCGCAATTAAAAA

ACCATGAAATTAGCTTTGTTATTGCGAACACCCTTGTTGTATCTAACA

AGTTTGAAACCGCCCCAACCAACTATAATTTAAGAGTGGTAGAAGTC

ACTACAGaattccggATGTTAGTAGCCGGTGGTGGACCAACAGGGTTATG

GTTGGCTGGTGAACTAAGGTTGGCCGGGGTTAGACCTCTTGTTTTAG

AGCGTAGGACAGAACCCTTTCCACATAGCAAAGCACTGGGTATCCAT

CCAAGAACTATTGAAATGTTGGAGTTGAGAGGCCTTGCTGGTAGATT

TTTAGATGGCGCCCCGAAGCTGCCGAAAGGTCACTTTGCATCGCTTC

CTGTGCCCTTAGACTATGGTGCAATGGATACTCGACATCCGTACGGT

GTCTACCGTAAACAAGTGGAAACGGAAGCTTTATTAGCCGGCCACGC

115

AACCAGATTAGGAGTTCCAGTCCGCAGAGGTCATGAACTTGTAGGCC

TAAGACAGCACGATGATGCTGTAGTCGGTACTGTTCAAGGTCCTCAA

GGTAGATACGAAGTCAGAGCCGCTTATCTTGTTGGTTGTGATGGAGG

AGGTTCTACCACAAGACGCCTGGCTGGGATAGATTTCCCTGGACAAG

ATCCTGGTGTTGCCGTTTTGATGGCTGACGCAAGATTCCGGGACCCAT

TACCCTCTGATCCAGCTATGGGTCCATTCAGACGCTATGGAGTCCTGA

GACCTGATTTAAGGGCATGGTTTAGTGCTATTCCTTTACCAGAAGAA

CAGGGTTTATGCAGAGTTATGGTTTTTTGGTATGGACGACAATTTGAA

GATAGACGTGCTCCAGTAACTGAGGATGAAATGAGAGCAGCACTTGT

TGATCTAGCGGGAACTGACTTTGGTATGTACGATGTTCAATGGTTGA

CCAGATTCACAGACGCTTCCAGGCAAGCTGCAAGGTATAGATCTGGA

AGGGTTTTCTTAGCTGGAGATGCTGCTCATACTGTCTTCCCCACTGGT

GGCCCAGGTTTGAATGCAGGTTTACAAGATGCCATGAATTTAGGTTG

GAAGTTAGCTGGAGCTGTTCATGGCTGGGGTGGTGCTTTACTAGATT

CATATCATGATGAGCGTTACCCTGTAGCTGAACAAGTTTTGCGTGAC

ACAAGAACACAATGTCTATTGTTAGATCCAGACCCTCGCTTTGTTCCA

TTAAGAGAGACGTTAGCAGAACTATTGAGACTAGAACCCGTAAACA

GATATTTCACCGGACACAACACGGCTCTAGCAGTTAGATACGATTTG

GGTGGTGCCCATCCTGCAACGGGAGGCAGAATGCCAGATGTCTTGGT

CGCAACTGGTGCTGGTCAAAGAAGGTTTTCAGATTGTTGTGTTACCG

GGAGAGGGGTTCTTTTAGTCACTTCTGGTGCAGATAGGGGGGAACTG

TTACGAGGATGGAAAGATAGAGTGGATGTGGTAACAGGTACCGTGG

CAGGACCTCTTGATGCAGCTGATCATTTAGTTAGACCTGATGGTTATG

TTGCATGGGCCGGTGGAGCTGGACACGATGAACCTGGTTTAGAAACT

GCTCTTAGATTATGGTTTGGTGAACCAGTTAGTgtatcgatggattacaaggatgac

gacgataagatctgagctcttaattaacaattcttcgccagaggtttggtcaagtctccaatcaaggttgtcggct

pGAL1-

dacO1-

tCYC1 in

pRS416

and

pGAL1-

dacO1-

tCYC1 in

pRS426

(EH-5-80-1

and EH-5-

80-2)

ccctcactaaagggaacaaaagctggagctctagtacggattagaagccgccgagcgggtgacagccctccgaa

ggaagactctcctccgtgcgtcctcgtcttcaccggtcgcgttcctgaaacgcagatgtgcctcgcgccgcactgct

ccgaacaataaagattctacaatactagcttttatggttatgaagaggaaaaattggcagtaacctggccccacaaac

cttcaaatgaacgaatcaaattaacaaccataggatgataatgcgattagttttttagccttatttctggggtaattaatc

agcgaagcgatgatttttgatctattaacagatatataaatgcaaaaactgcataaccactttaactaatactttcaacat

tttcggtttgtattacttcttattcaaatgtaataaaagtatcaacaaaaaattgttaatatacctctatactttaacgtcaag

gagaaaaaaccccggattctagaactagtATGTTAGTAGCCGGTGGTGGACCAACAGG

GTTATGGTTGGCTGGTGAACTAAGGTTGGCCGGGGTTAGACCTCTTG

TTTTAGAGCGTAGGACAGAACCCTTTCCACATAGCAAAGCACTGGGT

ATCCATCCAAGAACTATTGAAATGTTGGAGTTGAGAGGCCTTGCTGG

TAGATTTTTAGATGGCGCCCCGAAGCTGCCGAAAGGTCACTTTGCAT

CGCTTCCTGTGCCCTTAGACTATGGTGCAATGGATACTCGACATCCGT

ACGGTGTCTACCGTAAACAAGTGGAAACGGAAGCTTTATTAGCCGGC

CACGCAACCAGATTAGGAGTTCCAGTCCGCAGAGGTCATGAACTTGT

AGGCCTAAGACAGCACGATGATGCTGTAGTCGGTACTGTTCAAGGTC

CTCAAGGTAGATACGAAGTCAGAGCCGCTTATCTTGTTGGTTGTGAT

GGAGGAGGTTCTACCACAAGACGCCTGGCTGGGATAGATTTCCCTGG

ACAAGATCCTGGTGTTGCCGTTTTGATGGCTGACGCAAGATTCCGGG

ACCCATTACCCTCTGATCCAGCTATGGGTCCATTCAGACGCTATGGA

GTCCTGAGACCTGATTTAAGGGCATGGTTTAGTGCTATTCCTTTACCA

116

GAAGAACAGGGTTTATGCAGAGTTATGGTTTTTTGGTATGGACGACA

ATTTGAAGATAGACGTGCTCCAGTAACTGAGGATGAAATGAGAGCA

GCACTTGTTGATCTAGCGGGAACTGACTTTGGTATGTACGATGTTCAA

TGGTTGACCAGATTCACAGACGCTTCCAGGCAAGCTGCAAGGTATAG

ATCTGGAAGGGTTTTCTTAGCTGGAGATGCTGCTCATACTGTCTTCCC

CACTGGTGGCCCAGGTTTGAATGCAGGTTTACAAGATGCCATGAATT

TAGGTTGGAAGTTAGCTGGAGCTGTTCATGGCTGGGGTGGTGCTTTA

CTAGATTCATATCATGATGAGCGTTACCCTGTAGCTGAACAAGTTTTG

CGTGACACAAGAACACAATGTCTATTGTTAGATCCAGACCCTCGCTT

TGTTCCATTAAGAGAGACGTTAGCAGAACTATTGAGACTAGAACCCG

TAAACAGATATTTCACCGGACACAACACGGCTCTAGCAGTTAGATAC

GATTTGGGTGGTGCCCATCCTGCAACGGGAGGCAGAATGCCAGATGT

CTTGGTCGCAACTGGTGCTGGTCAAAGAAGGTTTTCAGATTGTTGTGT

TACCGGGAGAGGGGTTCTTTTAGTCACTTCTGGTGCAGATAGGGGGG

AACTGTTACGAGGATGGAAAGATAGAGTGGATGTGGTAACAGGTAC

CGTGGCAGGACCTCTTGATGCAGCTGATCATTTAGTTAGACCTGATG

GTTATGTTGCATGGGCCGGTGGAGCTGGACACGATGAACCTGGTTTA

GAAACTGCTCTTAGATTATGGTTTGGTGAACCAGTTAGTgtatcgatggattac

aaggatgacgacgataagatctgaaagcttatcgataccgtcgacctcgagtcatgtaattagttatgtcacgcttac

attcacgccctccccccacatccgctctaaccgaaaaggaaggagttagacaacctgaagtctaggtccctatttatt

tttttatagttatgttagtattaagaacgttatttatatttcaaatttttcttttttttctgtacagacgcgtgtacgcatgtaaca

ttatactgaaaaccttgcttgagaaggttttgggacgctcgaaggctttaatttgcggccggtacccaat

pTEF1-

dacO1-

tADH1 in

pSP-G1 (no

antibody

tag) (EH-5-

80-3)

tcaagtttcagtttcatttttcttgttctattacaactttttttacttcttgctcattagaaagaaagcatagcaatctaatctaa

gttttaattacaagcggccgCATGTTAGTAGCCGGTGGTGGACCAACAGGGTTAT

GGTTGGCTGGTGAACTAAGGTTGGCCGGGGTTAGACCTCTTGTTTTA

GAGCGTAGGACAGAACCCTTTCCACATAGCAAAGCACTGGGTATCCA

TCCAAGAACTATTGAAATGTTGGAGTTGAGAGGCCTTGCTGGTAGAT

TTTTAGATGGCGCCCCGAAGCTGCCGAAAGGTCACTTTGCATCGCTTC

CTGTGCCCTTAGACTATGGTGCAATGGATACTCGACATCCGTACGGT

GTCTACCGTAAACAAGTGGAAACGGAAGCTTTATTAGCCGGCCACGC

AACCAGATTAGGAGTTCCAGTCCGCAGAGGTCATGAACTTGTAGGCC

TAAGACAGCACGATGATGCTGTAGTCGGTACTGTTCAAGGTCCTCAA

GGTAGATACGAAGTCAGAGCCGCTTATCTTGTTGGTTGTGATGGAGG

AGGTTCTACCACAAGACGCCTGGCTGGGATAGATTTCCCTGGACAAG

ATCCTGGTGTTGCCGTTTTGATGGCTGACGCAAGATTCCGGGACCCAT

TACCCTCTGATCCAGCTATGGGTCCATTCAGACGCTATGGAGTCCTGA

GACCTGATTTAAGGGCATGGTTTAGTGCTATTCCTTTACCAGAAGAA

CAGGGTTTATGCAGAGTTATGGTTTTTTGGTATGGACGACAATTTGAA

GATAGACGTGCTCCAGTAACTGAGGATGAAATGAGAGCAGCACTTGT

TGATCTAGCGGGAACTGACTTTGGTATGTACGATGTTCAATGGTTGA

CCAGATTCACAGACGCTTCCAGGCAAGCTGCAAGGTATAGATCTGGA

AGGGTTTTCTTAGCTGGAGATGCTGCTCATACTGTCTTCCCCACTGGT

GGCCCAGGTTTGAATGCAGGTTTACAAGATGCCATGAATTTAGGTTG

GAAGTTAGCTGGAGCTGTTCATGGCTGGGGTGGTGCTTTACTAGATT

CATATCATGATGAGCGTTACCCTGTAGCTGAACAAGTTTTGCGTGAC

ACAAGAACACAATGTCTATTGTTAGATCCAGACCCTCGCTTTGTTCCA

117

TTAAGAGAGACGTTAGCAGAACTATTGAGACTAGAACCCGTAAACA

GATATTTCACCGGACACAACACGGCTCTAGCAGTTAGATACGATTTG

GGTGGTGCCCATCCTGCAACGGGAGGCAGAATGCCAGATGTCTTGGT

CGCAACTGGTGCTGGTCAAAGAAGGTTTTCAGATTGTTGTGTTACCG

GGAGAGGGGTTCTTTTAGTCACTTCTGGTGCAGATAGGGGGGAACTG

TTACGAGGATGGAAAGATAGAGTGGATGTGGTAACAGGTACCGTGG

CAGGACCTCTTGATGCAGCTGATCATTTAGTTAGACCTGATGGTTATG

TTGCATGGGCCGGTGGAGCTGGACACGATGAACCTGGTTTAGAAACT

GCTCTTAGATTATGGTTTGGTGAACCAGTTAGTTGAgagctcttaattaacaattct

tcgccagaggtttggtcaagtctccaatcaaggttgtcggcttgtctaccttgccagaaatttacgaaaagatggaaa

agggtcaaatcgttggtagatacgttgttgacacttctaaataagcgaatttcttatgatttatgatttttattattaaataa

gttataaaaaaaataagtgtatacaaattttaaagtgactcttaggttttaaaacgaaaattcttattcttgagtaactcttt

cctgtaggtcaggttgctttctcaggtatagcatgaggtcgctccaattcagctggcgtaatagcgaaga

pADH2-

dacO1-

tCYC1 in

pRS416

and

pADH2-

dacO1-

tCYC1 in

pRS426

(EH-5-80-4

and EH-5-

80-4)

ccctcactaaagggaacaaaagctggagctcGCAAAACGTAGGGGCAAACAAACGGA

AAAATCGTTTCTCAAATTTTCTGATGCCAAGAACTCTAACCAGTCTTA

TCTAAAAATTGCCTTATGATCCGTCTCTCCGGTTACAGCCTGTGTAAC

TGATTAATCCTGCCTTTCTAATCACCATTCTAATGTTTTAATTAAGGG

ATTTTGTCTTCATTAACGGCTTTCGCTCATAAAAATGTTATGACGTTT

TGCCCGCAGGCGGGAAACCATCCACTTCACGAGACTGATCTCCTCTG

CCGGAACACCGGGCATCTCCAACTTATAAGTTGGAGAAATAAGAGA

ATTTCAGATTGAGAGAATGAAAAAAAAAAAAAAAAAAAAGGCAGAG

GAGAGCATAGAAATGGGGTTCACTTTTTGGTAAAGCTATAGCATGCC

TATCACATATAAATAGAGTGCCAGTAGCGACTTTTTTCACACTCGAA

ATACTCTTACTACTGCTCTCTTGTTGTTTTTATCACTTCTTGTTTCTTCT

TGGTAAATAGAATATCAAGCTACAAAAAGCATACAATCAACTATCAA

CTATTAACTATATCGTAATACCATATGGCTAtctagaactagtATGTTAGTAG

CCGGTGGTGGACCAACAGGGTTATGGTTGGCTGGTGAACTAAGGTTG

GCCGGGGTTAGACCTCTTGTTTTAGAGCGTAGGACAGAACCCTTTCC

ACATAGCAAAGCACTGGGTATCCATCCAAGAACTATTGAAATGTTGG

AGTTGAGAGGCCTTGCTGGTAGATTTTTAGATGGCGCCCCGAAGCTG

CCGAAAGGTCACTTTGCATCGCTTCCTGTGCCCTTAGACTATGGTGCA

ATGGATACTCGACATCCGTACGGTGTCTACCGTAAACAAGTGGAAAC

GGAAGCTTTATTAGCCGGCCACGCAACCAGATTAGGAGTTCCAGTCC

GCAGAGGTCATGAACTTGTAGGCCTAAGACAGCACGATGATGCTGTA

GTCGGTACTGTTCAAGGTCCTCAAGGTAGATACGAAGTCAGAGCCGC

TTATCTTGTTGGTTGTGATGGAGGAGGTTCTACCACAAGACGCCTGG

CTGGGATAGATTTCCCTGGACAAGATCCTGGTGTTGCCGTTTTGATGG

CTGACGCAAGATTCCGGGACCCATTACCCTCTGATCCAGCTATGGGT

CCATTCAGACGCTATGGAGTCCTGAGACCTGATTTAAGGGCATGGTT

TAGTGCTATTCCTTTACCAGAAGAACAGGGTTTATGCAGAGTTATGG

TTTTTTGGTATGGACGACAATTTGAAGATAGACGTGCTCCAGTAACT

GAGGATGAAATGAGAGCAGCACTTGTTGATCTAGCGGGAACTGACTT

TGGTATGTACGATGTTCAATGGTTGACCAGATTCACAGACGCTTCCA

GGCAAGCTGCAAGGTATAGATCTGGAAGGGTTTTCTTAGCTGGAGAT

GCTGCTCATACTGTCTTCCCCACTGGTGGCCCAGGTTTGAATGCAGGT

TTACAAGATGCCATGAATTTAGGTTGGAAGTTAGCTGGAGCTGTTCA

118

TGGCTGGGGTGGTGCTTTACTAGATTCATATCATGATGAGCGTTACCC

TGTAGCTGAACAAGTTTTGCGTGACACAAGAACACAATGTCTATTGT

TAGATCCAGACCCTCGCTTTGTTCCATTAAGAGAGACGTTAGCAGAA

CTATTGAGACTAGAACCCGTAAACAGATATTTCACCGGACACAACAC

GGCTCTAGCAGTTAGATACGATTTGGGTGGTGCCCATCCTGCAACGG

GAGGCAGAATGCCAGATGTCTTGGTCGCAACTGGTGCTGGTCAAAGA

AGGTTTTCAGATTGTTGTGTTACCGGGAGAGGGGTTCTTTTAGTCACT

TCTGGTGCAGATAGGGGGGAACTGTTACGAGGATGGAAAGATAGAG

TGGATGTGGTAACAGGTACCGTGGCAGGACCTCTTGATGCAGCTGAT

CATTTAGTTAGACCTGATGGTTATGTTGCATGGGCCGGTGGAGCTGG

ACACGATGAACCTGGTTTAGAAACTGCTCTTAGATTATGGTTTGGTG

AACCAGTTAGTgtatcgatggattacaaggatgacgacgataagatctgaaagcttatcgataccgtcga

cctcgagtcatgtaattagttatgtcacgcttacattcacgccctccccccacatccgctctaaccgaaaaggaagga

gttagacaacctgaagtctaggtccctatttatttttttatagttatgttagtattaagaacgttatttatatttcaaatttttctt

ttttttctgtacagacgcgtgtacgcatgtaacattatactgaaaaccttgcttgagaaggttttgggacgctcgaagg

ctttaatttgcggccggtacccaat

pTEF1-

UBI4-

IFNG-

dacO1-

tADH1 in

pSP-G1

(EH-5-80-

6)

tcaagtttcagtttcatttttcttgttctattacaactttttttacttcttgctcattagaaagaaagcatagcaatctaatctaa

gttttaattacaagcggccgccattATGCAGATCTTCGTCAAGACGTTAACCGGTAA

AACCATAACTCTAGAAGTTGAATCTTCCGATACCATCGACAACGTTA

AGTCGAAAATTCAAGACAAGGAAGGCATTCCACCTGATCAACAAAG

ATTGATCTTTGCCGGTAAGCAGCTCGAGGACGGTAGAACGCTGTCTG

ATTACAACATTCAGAAGGAGTCGACCTTACATCTTGTCTTAAGACTA

AGAGGTGGTATGCAAGACCCATATGTAAAAGAAGCAGAAAACCTTA

AGAAATATTTTAATGCAGGTCATTCAGATGTAGCGGATAATGGAACT

CTTTTCTTAGGCATTTTGAAGAATTGGAAAGAGGAGAGTGACAGAAA

AATAATGCAGAGCCAAATTGTCTCCTTTTACTTCAAACTTTTTAAAAA

CTTTAAAGATGACCAGAGCATCCAAAAGAGTGTGGAGACCATCAAG

GAAGACATGAATGTCAAGTTTTTCAATAGCAACAAAAAGAAACGAG

ATGACTTCGAAAAGCTGACTAATTATTCGGTAACTGACTTGAATGTC

CAACGCAAAGCAATACATGAACTCATCCAAGTGATGGCTGAACTGTC

GCCAGCAGCTAAAACAGGGAAGCGAAAAAGGAGTCAGATGCTGTTT

CGAGGTCGAAGAGCATCCCAGATGTTAGTAGCCGGTGGTGGACCAAC

AGGGTTATGGTTGGCTGGTGAACTAAGGTTGGCCGGGGTTAGACCTC

TTGTTTTAGAGCGTAGGACAGAACCCTTTCCACATAGCAAAGCACTG

GGTATCCATCCAAGAACTATTGAAATGTTGGAGTTGAGAGGCCTTGC

TGGTAGATTTTTAGATGGCGCCCCGAAGCTGCCGAAAGGTCACTTTG

CATCGCTTCCTGTGCCCTTAGACTATGGTGCAATGGATACTCGACATC

CGTACGGTGTCTACCGTAAACAAGTGGAAACGGAAGCTTTATTAGCC

GGCCACGCAACCAGATTAGGAGTTCCAGTCCGCAGAGGTCATGAACT

TGTAGGCCTAAGACAGCACGATGATGCTGTAGTCGGTACTGTTCAAG

GTCCTCAAGGTAGATACGAAGTCAGAGCCGCTTATCTTGTTGGTTGT

GATGGAGGAGGTTCTACCACAAGACGCCTGGCTGGGATAGATTTCCC

TGGACAAGATCCTGGTGTTGCCGTTTTGATGGCTGACGCAAGATTCC

GGGACCCATTACCCTCTGATCCAGCTATGGGTCCATTCAGACGCTAT

GGAGTCCTGAGACCTGATTTAAGGGCATGGTTTAGTGCTATTCCTTTA

CCAGAAGAACAGGGTTTATGCAGAGTTATGGTTTTTTGGTATGGACG

119

ACAATTTGAAGATAGACGTGCTCCAGTAACTGAGGATGAAATGAGA

GCAGCACTTGTTGATCTAGCGGGAACTGACTTTGGTATGTACGATGTT

CAATGGTTGACCAGATTCACAGACGCTTCCAGGCAAGCTGCAAGGTA

TAGATCTGGAAGGGTTTTCTTAGCTGGAGATGCTGCTCATACTGTCTT

CCCCACTGGTGGCCCAGGTTTGAATGCAGGTTTACAAGATGCCATGA

ATTTAGGTTGGAAGTTAGCTGGAGCTGTTCATGGCTGGGGTGGTGCT

TTACTAGATTCATATCATGATGAGCGTTACCCTGTAGCTGAACAAGTT

TTGCGTGACACAAGAACACAATGTCTATTGTTAGATCCAGACCCTCG

CTTTGTTCCATTAAGAGAGACGTTAGCAGAACTATTGAGACTAGAAC

CCGTAAACAGATATTTCACCGGACACAACACGGCTCTAGCAGTTAGA

TACGATTTGGGTGGTGCCCATCCTGCAACGGGAGGCAGAATGCCAGA

TGTCTTGGTCGCAACTGGTGCTGGTCAAAGAAGGTTTTCAGATTGTTG

TGTTACCGGGAGAGGGGTTCTTTTAGTCACTTCTGGTGCAGATAGGG

GGGAACTGTTACGAGGATGGAAAGATAGAGTGGATGTGGTAACAGG

TACCGTGGCAGGACCTCTTGATGCAGCTGATCATTTAGTTAGACCTG

ATGGTTATGTTGCATGGGCCGGTGGAGCTGGACACGATGAACCTGGT

TTAGAAACTGCTCTTAGATTATGGTTTGGTGAACCAGTTAGTgtatcgatgg

attacaaggatgacgacgataagatctgagctcttaattaacaattcttcgccagaggtttggtcaagtctccaatcaa

ggttgtcggct

pTEF1-

IFNG-

dacO1-

tADH1 in

pSP-G1

(EH-5-80-

7)

tcaagtttcagtttcatttttcttgttctattacaactttttttacttcttgctcattagaaagaaagcatagcaatctaatctaa

gttttaattacaagcggccgccattATGCAAGACCCATATGTAAAAGAAGCAGAAAA

CCTTAAGAAATATTTTAATGCAGGTCATTCAGATGTAGCGGATAATG

GAACTCTTTTCTTAGGCATTTTGAAGAATTGGAAAGAGGAGAGTGAC

AGAAAAATAATGCAGAGCCAAATTGTCTCCTTTTACTTCAAACTTTTT

AAAAACTTTAAAGATGACCAGAGCATCCAAAAGAGTGTGGAGACCA

TCAAGGAAGACATGAATGTCAAGTTTTTCAATAGCAACAAAAAGAA

ACGAGATGACTTCGAAAAGCTGACTAATTATTCGGTAACTGACTTGA

ATGTCCAACGCAAAGCAATACATGAACTCATCCAAGTGATGGCTGAA

CTGTCGCCAGCAGCTAAAACAGGGAAGCGAAAAAGGAGTCAGATGC

TGTTTCGAGGTCGAAGAGCATCCCAGATGTTAGTAGCCGGTGGTGGA

CCAACAGGGTTATGGTTGGCTGGTGAACTAAGGTTGGCCGGGGTTAG

ACCTCTTGTTTTAGAGCGTAGGACAGAACCCTTTCCACATAGCAAAG

CACTGGGTATCCATCCAAGAACTATTGAAATGTTGGAGTTGAGAGGC

CTTGCTGGTAGATTTTTAGATGGCGCCCCGAAGCTGCCGAAAGGTCA

CTTTGCATCGCTTCCTGTGCCCTTAGACTATGGTGCAATGGATACTCG

ACATCCGTACGGTGTCTACCGTAAACAAGTGGAAACGGAAGCTTTAT

TAGCCGGCCACGCAACCAGATTAGGAGTTCCAGTCCGCAGAGGTCAT

GAACTTGTAGGCCTAAGACAGCACGATGATGCTGTAGTCGGTACTGT

TCAAGGTCCTCAAGGTAGATACGAAGTCAGAGCCGCTTATCTTGTTG

GTTGTGATGGAGGAGGTTCTACCACAAGACGCCTGGCTGGGATAGAT

TTCCCTGGACAAGATCCTGGTGTTGCCGTTTTGATGGCTGACGCAAG

ATTCCGGGACCCATTACCCTCTGATCCAGCTATGGGTCCATTCAGACG

CTATGGAGTCCTGAGACCTGATTTAAGGGCATGGTTTAGTGCTATTCC

TTTACCAGAAGAACAGGGTTTATGCAGAGTTATGGTTTTTTGGTATGG

ACGACAATTTGAAGATAGACGTGCTCCAGTAACTGAGGATGAAATGA

GAGCAGCACTTGTTGATCTAGCGGGAACTGACTTTGGTATGTACGAT

120

GTTCAATGGTTGACCAGATTCACAGACGCTTCCAGGCAAGCTGCAAG

GTATAGATCTGGAAGGGTTTTCTTAGCTGGAGATGCTGCTCATACTGT

CTTCCCCACTGGTGGCCCAGGTTTGAATGCAGGTTTACAAGATGCCA

TGAATTTAGGTTGGAAGTTAGCTGGAGCTGTTCATGGCTGGGGTGGT

GCTTTACTAGATTCATATCATGATGAGCGTTACCCTGTAGCTGAACAA

GTTTTGCGTGACACAAGAACACAATGTCTATTGTTAGATCCAGACCC

TCGCTTTGTTCCATTAAGAGAGACGTTAGCAGAACTATTGAGACTAG

AACCCGTAAACAGATATTTCACCGGACACAACACGGCTCTAGCAGTT

AGATACGATTTGGGTGGTGCCCATCCTGCAACGGGAGGCAGAATGCC

AGATGTCTTGGTCGCAACTGGTGCTGGTCAAAGAAGGTTTTCAGATT

GTTGTGTTACCGGGAGAGGGGTTCTTTTAGTCACTTCTGGTGCAGATA

GGGGGGAACTGTTACGAGGATGGAAAGATAGAGTGGATGTGGTAAC

AGGTACCGTGGCAGGACCTCTTGATGCAGCTGATCATTTAGTTAGAC

CTGATGGTTATGTTGCATGGGCCGGTGGAGCTGGACACGATGAACCT

GGTTTAGAAACTGCTCTTAGATTATGGTTTGGTGAACCAGTTAGTgtatc

gatggattacaaggatgacgacgataagatctgagctcttaattaacaattcttcgccagaggtttggtcaagtctcca

atcaaggttgtcggct

pPGK1-

dacO4-

dacO1-

tCYC1 in

pSP-G1

(EH-5-80-

8)

tggaatggcgggaaagggtttagtaccacatgctatgatgcccactgtgatctccagagcaaagttcgttcgatcgt

actgttactctctctctttcaaacagaattgtccgaatcgtgtgacaacaacagcctgttctcacacactcttttcttctaa

ccaagggggtggtttagtttagtagaacctcgtgaaacttacatttacatatatataaacttgcataaattggtcaatgc

aagaaatacatatttggtcttttctaattcgtagtttttcaagttcttagatgctttctttttctcttttttacagatcatcaagga

agtaattatctactttttacaacaaatataaaacaaggatccATGTCATTCACTGCCAAGGAAGT

TCAATATTTGCGTTCTCAGCAATTAGGAAGATTAGCCACCGTTGGTGC

TGATGGTACACCACATAATGTTCCAGTAGGCTACAGATACAACGCTG

ACTTGGGGACCATTGACATCACAGGAAGGGACTTAAGAAGAAGTAG

GAAATATAGAGATGTTCAGGCTGGCTCGCGGGTGGCATTTATTGTTG

ATGATCTACCGAGCACAGCACCTATAGTCGCCCGCGGGCTGGAGGTG

AGGGGAACAGCTGAGGCGCTTTCTGGTGAAGAAGATTTGATACGTGT

ACGACCTGCAAGAATCGTCACTTGGGGTATTGAAGCAGATTGGCAAG

CTGGTCCTACTGGTAGAACGGTAAGGCCCACCACTCCAAGATCCGCG

ACGATGTTAGTAGCCGGTGGTGGACCAACAGGGTTATGGTTGGCTGG

TGAACTAAGGTTGGCCGGGGTTAGACCTCTTGTTTTAGAGCGTAGGA

CAGAACCCTTTCCACATAGCAAAGCACTGGGTATCCATCCAAGAACT

ATTGAAATGTTGGAGTTGAGAGGCCTTGCTGGTAGATTTTTAGATGG

CGCCCCGAAGCTGCCGAAAGGTCACTTTGCATCGCTTCCTGTGCCCTT

AGACTATGGTGCAATGGATACTCGACATCCGTACGGTGTCTACCGTA

AACAAGTGGAAACGGAAGCTTTATTAGCCGGCCACGCAACCAGATTA

GGAGTTCCAGTCCGCAGAGGTCATGAACTTGTAGGCCTAAGACAGCA

CGATGATGCTGTAGTCGGTACTGTTCAAGGTCCTCAAGGTAGATACG

AAGTCAGAGCCGCTTATCTTGTTGGTTGTGATGGAGGAGGTTCTACC

ACAAGACGCCTGGCTGGGATAGATTTCCCTGGACAAGATCCTGGTGT

TGCCGTTTTGATGGCTGACGCAAGATTCCGGGACCCATTACCCTCTGA

TCCAGCTATGGGTCCATTCAGACGCTATGGAGTCCTGAGACCTGATTT

AAGGGCATGGTTTAGTGCTATTCCTTTACCAGAAGAACAGGGTTTAT

GCAGAGTTATGGTTTTTTGGTATGGACGACAATTTGAAGATAGACGT

GCTCCAGTAACTGAGGATGAAATGAGAGCAGCACTTGTTGATCTAGC

121

GGGAACTGACTTTGGTATGTACGATGTTCAATGGTTGACCAGATTCA

CAGACGCTTCCAGGCAAGCTGCAAGGTATAGATCTGGAAGGGTTTTC

TTAGCTGGAGATGCTGCTCATACTGTCTTCCCCACTGGTGGCCCAGGT

TTGAATGCAGGTTTACAAGATGCCATGAATTTAGGTTGGAAGTTAGC

TGGAGCTGTTCATGGCTGGGGTGGTGCTTTACTAGATTCATATCATGA

TGAGCGTTACCCTGTAGCTGAACAAGTTTTGCGTGACACAAGAACAC

AATGTCTATTGTTAGATCCAGACCCTCGCTTTGTTCCATTAAGAGAGA

CGTTAGCAGAACTATTGAGACTAGAACCCGTAAACAGATATTTCACC

GGACACAACACGGCTCTAGCAGTTAGATACGATTTGGGTGGTGCCCA

TCCTGCAACGGGAGGCAGAATGCCAGATGTCTTGGTCGCAACTGGTG

CTGGTCAAAGAAGGTTTTCAGATTGTTGTGTTACCGGGAGAGGGGTT

CTTTTAGTCACTTCTGGTGCAGATAGGGGGGAACTGTTACGAGGATG

GAAAGATAGAGTGGATGTGGTAACAGGTACCGTGGCAGGACCTCTTG

ATGCAGCTGATCATTTAGTTAGACCTGATGGTTATGTTGCATGGGCCG

GTGGAGCTGGACACGATGAACCTGGTTTAGAAACTGCTCTTAGATTA

TGGTTTGGTGAACCAGTTAGTgtcgacatggaacagaagttgatttccgaagaagacctcgagt

aagcttggtaccgcggctagctaagatccgctctaaccgaaaaggaaggagttagacaacctgaagtctaggtcc

ctatttatttttttatagttatgttagtattaagaacgttatttatatttcaaatttttcttttttttctgtacagacgcgtgtacgc

atgtaacattatactgaaaaccttgcttgagaaggttttgggacgctcgaagatccagctgcattaatgaatcggcca

acgcgcggggagaggcggtttgcgtattgggcgctcttccgcttcctcgctcactgactcgctgcgctcgg

pPGK1-

dacO1-

dacO4-

tCYC1 in

pSP-G1

(EH-5-80-

9)

tggaatggcgggaaagggtttagtaccacatgctatgatgcccactgtgatctccagagcaaagttcgttcgatcgt

actgttactctctctctttcaaacagaattgtccgaatcgtgtgacaacaacagcctgttctcacacactcttttcttctaa

ccaagggggtggtttagtttagtagaacctcgtgaaacttacatttacatatatataaacttgcataaattggtcaatgc

aagaaatacatatttggtcttttctaattcgtagtttttcaagttcttagatgctttctttttctcttttttacagatcatcaagga

agtaattatctactttttacaacaaatataaaacaaggatccATGTTAGTAGCCGGTGGTGGACC

AACAGGGTTATGGTTGGCTGGTGAACTAAGGTTGGCCGGGGTTAGAC

CTCTTGTTTTAGAGCGTAGGACAGAACCCTTTCCACATAGCAAAGCA

CTGGGTATCCATCCAAGAACTATTGAAATGTTGGAGTTGAGAGGCCT

TGCTGGTAGATTTTTAGATGGCGCCCCGAAGCTGCCGAAAGGTCACT

TTGCATCGCTTCCTGTGCCCTTAGACTATGGTGCAATGGATACTCGAC

ATCCGTACGGTGTCTACCGTAAACAAGTGGAAACGGAAGCTTTATTA

GCCGGCCACGCAACCAGATTAGGAGTTCCAGTCCGCAGAGGTCATGA

ACTTGTAGGCCTAAGACAGCACGATGATGCTGTAGTCGGTACTGTTC

AAGGTCCTCAAGGTAGATACGAAGTCAGAGCCGCTTATCTTGTTGGT

TGTGATGGAGGAGGTTCTACCACAAGACGCCTGGCTGGGATAGATTT

CCCTGGACAAGATCCTGGTGTTGCCGTTTTGATGGCTGACGCAAGAT

TCCGGGACCCATTACCCTCTGATCCAGCTATGGGTCCATTCAGACGCT

ATGGAGTCCTGAGACCTGATTTAAGGGCATGGTTTAGTGCTATTCCTT

TACCAGAAGAACAGGGTTTATGCAGAGTTATGGTTTTTTGGTATGGA

CGACAATTTGAAGATAGACGTGCTCCAGTAACTGAGGATGAAATGAG

AGCAGCACTTGTTGATCTAGCGGGAACTGACTTTGGTATGTACGATG

TTCAATGGTTGACCAGATTCACAGACGCTTCCAGGCAAGCTGCAAGG

TATAGATCTGGAAGGGTTTTCTTAGCTGGAGATGCTGCTCATACTGTC

TTCCCCACTGGTGGCCCAGGTTTGAATGCAGGTTTACAAGATGCCAT

GAATTTAGGTTGGAAGTTAGCTGGAGCTGTTCATGGCTGGGGTGGTG

CTTTACTAGATTCATATCATGATGAGCGTTACCCTGTAGCTGAACAAG

122

TTTTGCGTGACACAAGAACACAATGTCTATTGTTAGATCCAGACCCTC

GCTTTGTTCCATTAAGAGAGACGTTAGCAGAACTATTGAGACTAGAA

CCCGTAAACAGATATTTCACCGGACACAACACGGCTCTAGCAGTTAG

ATACGATTTGGGTGGTGCCCATCCTGCAACGGGAGGCAGAATGCCAG

ATGTCTTGGTCGCAACTGGTGCTGGTCAAAGAAGGTTTTCAGATTGTT

GTGTTACCGGGAGAGGGGTTCTTTTAGTCACTTCTGGTGCAGATAGG

GGGGAACTGTTACGAGGATGGAAAGATAGAGTGGATGTGGTAACAG

GTACCGTGGCAGGACCTCTTGATGCAGCTGATCATTTAGTTAGACCT

GATGGTTATGTTGCATGGGCCGGTGGAGCTGGACACGATGAACCTGG

TTTAGAAACTGCTCTTAGATTATGGTTTGGTGAACCAGTTAGTATGTC

ATTCACTGCCAAGGAAGTTCAATATTTGCGTTCTCAGCAATTAGGAA

GATTAGCCACCGTTGGTGCTGATGGTACACCACATAATGTTCCAGTA

GGCTACAGATACAACGCTGACTTGGGGACCATTGACATCACAGGAAG

GGACTTAAGAAGAAGTAGGAAATATAGAGATGTTCAGGCTGGCTCG

CGGGTGGCATTTATTGTTGATGATCTACCGAGCACAGCACCTATAGT

CGCCCGCGGGCTGGAGGTGAGGGGAACAGCTGAGGCGCTTTCTGGTG

AAGAAGATTTGATACGTGTACGACCTGCAAGAATCGTCACTTGGGGT

ATTGAAGCAGATTGGCAAGCTGGTCCTACTGGTAGAACGGTAAGGCC

CACCACTCCAAGATCCGCGACGgtcgacatggaacagaagttgatttccgaagaagacctcg

agtaagcttggtaccgcggctagctaagatccgctctaaccgaaaaggaaggagttagacaacctgaagtctaggt

ccctatttatttttttatagttatgttagtattaagaacgttatttatatttcaaatttttcttttttttctgtacagacgcgtgtac

gcatgtaacattatactgaaaaccttgcttgagaaggttttgggacgctcgaagatccagctgcattaatgaatcggc

caacgcgcggggagaggcggtttgcgtattgggcgctcttccgcttcctcgctcactgactcgctgcgctcgg

pTEF1-

oxyS-

dacO1-37-

tADH1 in

pSP-G1

(EH-5-80-

10)

tcaagtttcagtttcatttttcttgttctattacaactttttttacttcttgctcattagaaagaaagcatagcaatctaatctaa

gttttaattacaagcggccgcATGAGGTACGATGTTGTTATAGCTGGTGCAGGAC

CCACCGGTTTGATGTTAGCATGTGAACTTCGGCTGGCGGGTGCCAGA

ACTTTGGTTTTAGAAAGATTAGCCGAGCCTTTTCCACATAGCAAAGC

ACTGGGTATCCATCCAAGAACTATTGAAATGTTGGAGTTGAGAGGCC

TTGCTGGTAGATTTTTAGATGGCGCCCCGAAGCTGCCGAAAGGTCAC

TTTGCATCGCTTCCTGTGCCCTTAGACTATGGTGCAATGGATACTCGA

CATCCGTACGGTGTCTACCGTAAACAAGTGGAAACGGAAGCTTTATT

AGCCGGCCACGCAACCAGATTAGGAGTTCCAGTCCGCAGAGGTCATG

AACTTGTAGGCCTAAGACAGCACGATGATGCTGTAGTCGGTACTGTT

CAAGGTCCTCAAGGTAGATACGAAGTCAGAGCCGCTTATCTTGTTGG

TTGTGATGGAGGAGGTTCTACCACAAGACGCCTGGCTGGGATAGATT

TCCCTGGACAAGATCCTGGTGTTGCCGTTTTGATGGCTGACGCAAGA

TTCCGGGACCCATTACCCTCTGATCCAGCTATGGGTCCATTCAGACGC

TATGGAGTCCTGAGACCTGATTTAAGGGCATGGTTTAGTGCTATTCCT

TTACCAGAAGAACAGGGTTTATGCAGAGTTATGGTTTTTTGGTATGG

ACGACAATTTGAAGATAGACGTGCTCCAGTAACTGAGGATGAAATGA

GAGCAGCACTTGTTGATCTAGCGGGAACTGACTTTGGTATGTACGAT

GTTCAATGGTTGACCAGATTCACAGACGCTTCCAGGCAAGCTGCAAG

GTATAGATCTGGAAGGGTTTTCTTAGCTGGAGATGCTGCTCATACTGT

CTTCCCCACTGGTGGCCCAGGTTTGAATGCAGGTTTACAAGATGCCA

TGAATTTAGGTTGGAAGTTAGCTGGAGCTGTTCATGGCTGGGGTGGT

GCTTTACTAGATTCATATCATGATGAGCGTTACCCTGTAGCTGAACAA

123

GTTTTGCGTGACACAAGAACACAATGTCTATTGTTAGATCCAGACCC

TCGCTTTGTTCCATTAAGAGAGACGTTAGCAGAACTATTGAGACTAG

AACCCGTAAACAGATATTTCACCGGACACAACACGGCTCTAGCAGTT

AGATACGATTTGGGTGGTGCCCATCCTGCAACGGGAGGCAGAATGCC

AGATGTCTTGGTCGCAACTGGTGCTGGTCAAAGAAGGTTTTCAGATT

GTTGTGTTACCGGGAGAGGGGTTCTTTTAGTCACTTCTGGTGCAGATA

GGGGGGAACTGTTACGAGGATGGAAAGATAGAGTGGATGTGGTAAC

AGGTACCGTGGCAGGACCTCTTGATGCAGCTGATCATTTAGTTAGAC

CTGATGGTTATGTTGCATGGGCCGGTGGAGCTGGACACGATGAACCT

GGTTTAGAAACTGCTCTTAGATTATGGTTTGGTGAACCAGTTAGTgtatc

gatggattacaaggatgacgacgataagatctgagctcttaattaacaattcttcgccagaggtttggtcaagtctcca

atcaaggttgtcggct

pTEF1-

oxyS-

dacO1-87-

tADH1 in

pSP-G1

(EH-5-80-

11)

tcaagtttcagtttcatttttcttgttctattacaactttttttacttcttgctcattagaaagaaagcatagcaatctaatctaa

gttttaattacaagcggccgcATGAGGTACGATGTTGTTATAGCTGGTGCAGGAC

CCACCGGTTTGATGTTAGCATGTGAACTTCGGCTGGCGGGTGCCAGA

ACTTTGGTTTTAGAAAGATTAGCCGAGCCTGTTGACTTCTCGAAAGCT

CTAGGAGTCCACGCTCGCACTGTTGAACTATTAGATATGAGAGGCCT

CGGTGAAGGATTCCAGGCTGAAGCACCAAAGTTAAGAGGTGGTAATT

TTGCCTCATTAGGCGTCCCCCTGGATTTCTCATCATTTGATACTCGAC

ATCCGTACGGTGTCTACCGTAAACAAGTGGAAACGGAAGCTTTATTA

GCCGGCCACGCAACCAGATTAGGAGTTCCAGTCCGCAGAGGTCATGA

ACTTGTAGGCCTAAGACAGCACGATGATGCTGTAGTCGGTACTGTTC

AAGGTCCTCAAGGTAGATACGAAGTCAGAGCCGCTTATCTTGTTGGT

TGTGATGGAGGAGGTTCTACCACAAGACGCCTGGCTGGGATAGATTT

CCCTGGACAAGATCCTGGTGTTGCCGTTTTGATGGCTGACGCAAGAT

TCCGGGACCCATTACCCTCTGATCCAGCTATGGGTCCATTCAGACGCT

ATGGAGTCCTGAGACCTGATTTAAGGGCATGGTTTAGTGCTATTCCTT

TACCAGAAGAACAGGGTTTATGCAGAGTTATGGTTTTTTGGTATGGA

CGACAATTTGAAGATAGACGTGCTCCAGTAACTGAGGATGAAATGAG

AGCAGCACTTGTTGATCTAGCGGGAACTGACTTTGGTATGTACGATG

TTCAATGGTTGACCAGATTCACAGACGCTTCCAGGCAAGCTGCAAGG

TATAGATCTGGAAGGGTTTTCTTAGCTGGAGATGCTGCTCATACTGTC

TTCCCCACTGGTGGCCCAGGTTTGAATGCAGGTTTACAAGATGCCAT

GAATTTAGGTTGGAAGTTAGCTGGAGCTGTTCATGGCTGGGGTGGTG

CTTTACTAGATTCATATCATGATGAGCGTTACCCTGTAGCTGAACAAG

TTTTGCGTGACACAAGAACACAATGTCTATTGTTAGATCCAGACCCTC

GCTTTGTTCCATTAAGAGAGACGTTAGCAGAACTATTGAGACTAGAA

CCCGTAAACAGATATTTCACCGGACACAACACGGCTCTAGCAGTTAG

ATACGATTTGGGTGGTGCCCATCCTGCAACGGGAGGCAGAATGCCAG

ATGTCTTGGTCGCAACTGGTGCTGGTCAAAGAAGGTTTTCAGATTGTT

GTGTTACCGGGAGAGGGGTTCTTTTAGTCACTTCTGGTGCAGATAGG

GGGGAACTGTTACGAGGATGGAAAGATAGAGTGGATGTGGTAACAG

GTACCGTGGCAGGACCTCTTGATGCAGCTGATCATTTAGTTAGACCT

GATGGTTATGTTGCATGGGCCGGTGGAGCTGGACACGATGAACCTGG

TTTAGAAACTGCTCTTAGATTATGGTTTGGTGAACCAGTTAGTgtatcgat

124

ggattacaaggatgacgacgataagatctgagctcttaattaacaattcttcgccagaggtttggtcaagtctccaatc

aaggttgtcggct

pTEF1-

oxyS-

dacO1-

191-tADH1

in pSP-G1

(EH-5-80-

12)

tcaagtttcagtttcatttttcttgttctattacaactttttttacttcttgctcattagaaagaaagcatagcaatctaatctaa

gttttaattacaagcggccgcATGAGGTACGATGTTGTTATAGCTGGTGCAGGAC

CCACCGGTTTGATGTTAGCATGTGAACTTCGGCTGGCGGGTGCCAGA

ACTTTGGTTTTAGAAAGATTAGCCGAGCCTGTTGACTTCTCGAAAGCT

CTAGGAGTCCACGCTCGCACTGTTGAACTATTAGATATGAGAGGCCT

CGGTGAAGGATTCCAGGCTGAAGCACCAAAGTTAAGAGGTGGTAATT

TTGCCTCATTAGGCGTCCCCCTGGATTTCTCATCATTTGATACTAGAC

ACCCATATGCATTGTTTGTTCCACAAGTACGAACTGAAGAACTGCTA

ACAGGTAGAGCTTTGGAGCTAGGGGCGGAGCTGCGTCGTGGTCATGC

CGTGACCGCCTTGGAACAAGATGCTGATGGTGTTACTGTGAGCGTGA

CAGGCCCTGAAGGCCCATACGAAGTAGAATGTGCTTATTTGGTGGGC

TGCGACGGTGGAGGCAGTACGGTGAGGAAACTATTGGGCATAGATTT

TCCAGGTCAAGACCCACATATGTTTGCTGTCATCGCAGACGCAAGAT

TCAGAGAAGAGCTTCCTCACGATCCAGCTATGGGTCCATTCAGACGC

TATGGAGTCCTGAGACCTGATTTAAGGGCATGGTTTAGTGCTATTCCT

TTACCAGAAGAACAGGGTTTATGCAGAGTTATGGTTTTTTGGTATGG

ACGACAATTTGAAGATAGACGTGCTCCAGTAACTGAGGATGAAATGA

GAGCAGCACTTGTTGATCTAGCGGGAACTGACTTTGGTATGTACGAT

GTTCAATGGTTGACCAGATTCACAGACGCTTCCAGGCAAGCTGCAAG

GTATAGATCTGGAAGGGTTTTCTTAGCTGGAGATGCTGCTCATACTGT

CTTCCCCACTGGTGGCCCAGGTTTGAATGCAGGTTTACAAGATGCCA

TGAATTTAGGTTGGAAGTTAGCTGGAGCTGTTCATGGCTGGGGTGGT

GCTTTACTAGATTCATATCATGATGAGCGTTACCCTGTAGCTGAACAA

GTTTTGCGTGACACAAGAACACAATGTCTATTGTTAGATCCAGACCC

TCGCTTTGTTCCATTAAGAGAGACGTTAGCAGAACTATTGAGACTAG

AACCCGTAAACAGATATTTCACCGGACACAACACGGCTCTAGCAGTT

AGATACGATTTGGGTGGTGCCCATCCTGCAACGGGAGGCAGAATGCC

AGATGTCTTGGTCGCAACTGGTGCTGGTCAAAGAAGGTTTTCAGATT

GTTGTGTTACCGGGAGAGGGGTTCTTTTAGTCACTTCTGGTGCAGATA

GGGGGGAACTGTTACGAGGATGGAAAGATAGAGTGGATGTGGTAAC

AGGTACCGTGGCAGGACCTCTTGATGCAGCTGATCATTTAGTTAGAC

CTGATGGTTATGTTGCATGGGCCGGTGGAGCTGGACACGATGAACCT

GGTTTAGAAACTGCTCTTAGATTATGGTTTGGTGAACCAGTTAGTgtatc

gatggattacaaggatgacgacgataagatctgagctcttaattaacaattcttcgccagaggtttggtcaagtctcca

atcaaggttgtcggct

pPGK1-

dacJ-

tCYC1 in

pSP-G1

(EH-5-90-

A)

tggaatggcgggaaagggtttagtaccacatgctatgatgcccactgtgatctccagagcaaagttcgttcgatcgt

actgttactctctctctttcaaacagaattgtccgaatcgtgtgacaacaacagcctgttctcacacactcttttcttctaa

ccaagggggtggtttagtttagtagaacctcgtgaaacttacatttacatatatataaacttgcataaattggtcaatgc

aagaaatacatatttggtcttttctaattcgtagtttttcaagttcttagatgctttctttttctcttttttacagatcatcaagga

agtaattatctactttttacaacaaatataaaacaaggatccATGACCGCAGGACAGCTGCATCT

TGACGTTCCGGAACCGCCTGGTTCATGGGTAGATGAAGCCACATTTA

GGGCTGTTATGGGTGCTTTGCCATCCGGTGTTACAGTCGTGACGACCT

TGGACCCAGATGGCAGACCAGTAGGCTTAACTTGTTCTGCGGCTTGT

TCAGTTTCTCAAAGGCCTCCCTTATTACTTGTCTGCTTAAACGATAGA

125

AGCAGGGTACTAGATGCAATACTCAGAAGAGGTCAGTTCATAGTAAA

TGTCCTACGCGACCGTAGAGAAGCAACATCGGCAGTCTTTGCCAGTA

GAGCCGAAGATAGATTCGCAGCTGTGCCTTGGCAACCTGGGAGGCGA

ACTGGTGTCCCTTGGTTGTTAGCAGACACGGTTGCTCATGCAGAATGT

GAGGTGGCAGGTACTTTGCATGCTGGCGATCACGTTATCGTTGTTGG

AGGTATTGTTGCGGGTGATGCGCACCCCGACCGTGTGGGGCCATTAA

TGTATTGGAGACAAAGATACGGTGGATGGCCAGTAACAGATGATGCT

AGAGAAATTGCATTGACTCTAGCTGCCGAGGGAgtcgacatggaacagaagttga

tttccgaagaagacctcgagtaagcttggtaccgcggctagctaagatccgctctaaccgaaaaggaaggagttag

acaacctgaagtctaggtccctatttatttttttatagttatgttagtattaagaacgttatttatatttcaaatttttcttttttttc

tgtacagacgcgtgtacgcatgtaacattatactgaaaaccttgcttgagaaggttttgggacgctcgaagatccagc

tgcattaatgaatcggccaacgcgcggggagaggcggtttgcgtattgggcgctcttccgcttcctcgctcactgac

tcgctgcgctcgg

pGPD-

dacM2-

tCYC1 in

pRS413

(EH-5-105-

Α)

ccctcactaaagggaacaaaagctggagctcAGTTTATCATTATCAATACTGCCATTTC

AAAGAATACGTAAATAATTAATAGTAGTGATTTTCCTAACTTTATTTA

GTCAAAAAATTAGCCTTTTAATTCTGCTGTAACCCGTACATGCCCAAA

ATAGGGGGCGGGTTACACAGAATATATAACATCGTAGGTGTCTGGGT

GAACAGTTTATTCCTGGCATCCACTAAATATAATGGAGCCCGCTTTTT

AAGCTGGCATCCAGAAAAAAAAAGAATCCCAGCACCAAAATATTGT

TTTCTTCACCAACCATCAGTTCATAGGTCCATTCTCTTAGCGCAACTA

CAGAGAACAGGGGCACAAACAGGCAAAAAACGGGCACAACCTCAAT

GGAGTGATGCAACCTGCCTGGAGTAAATGATGACACAAGGCAATTG

ACCCACGCATGTATCTATCTCATTTTCTTACACCTTCTATTACCTTCTG

CTCTCTCTGATTTGGAAAAAGCTGAAAAAAAAGGTTGAAACCAGTTC

CCTGAAATTATTCCCCTACTTGACTAATAAGTATATAAAGACGGTAG

GTATTGATTGTAATTCTGTAAATCTATTTCTTAAACTTCTTAAATTCTA

CTTTTATAGTTAGTCTTTTTTTTAGTTTTAAAACACCAAGAACTTAGTT

TCGACGGATACTAGTAAAATGACTGATGTATCCGCCCATACTACAAC

CGATGGGGCGGTTGCAAGGGCTCGCGCTGTGGTCAGACTAAATACTG

CATATTTTCGGGCTAAGGTACTTCAAAGTGCTGTCGAACTAGGTGTTT

TTGATCTTTTAGATGGAGGCCCACAACCTGCCGCTGACATTTGCGGA

AAGTTAGGTATTAGACATAGACTCGCAGTGGACTATTTGGACGCATT

GACGGGTTTAGGGTTGCTTGAAAGGGATGGTGAACGTTACAGAAACT

CGCCAACAGCGGCTGAGTTCTTGGTCCAAGATCGACCCGCCTATTTG

GGTGGAACCGCGAGGCAACATGCCAAATTACATTATCACGCTTGGGG

TAAACTGGCCGATGCAATGAGAGAAGGCCGTGCTCAGAGCGCCGTG

GCAGCTCAAGGTGCAAAAGCCTATGTAAATTACTATGAAGATTTGGG

CCAGGCCAGGAGGGTCATGACACATATGGATGCTCATAACGGTTTCA

CAGCGGATGAAATCGCTAGACAATTGGACTGGAGAGGATACGCCAC

AGTAACAGATGTGGGCGGCGCTCGTGGGAATGTTGCAGCTCGAATTG

TTCAAGCAGTGCCTCATTTGAGAGCAAATGTAGTTGACCTACCCGCA

TTGCGACCTTTATTCGATGAGCTGATGGCCCAACTAGGAACTGCTGG

TCAGGTTGAGTTTCACGGTGGTGACTTCTTTGCGGACCCGATACCAG

AGTCTGACGTTATCGTTGTGGGCCACGTCCTGGCGGATTGGCCACAG

CCAAGACGTGTGGAATTACTTAGAAGATGTCACGCAGCATTAAGACC

TGGTGGAACGGTAGTTGTTTACGATGTTATGGTTGATGATGATAGAG

126

ATGACGTCGAAGCACTCCTACAAAGATTAAACTCTGCAATGATAAGG

GACGATATGGGTGCTTACGCTGTTACTGAATGTGCTGGATATTTAAG

AGAAGCTGGATTTACCGTAGAAAGGGCCCTGCGTACGGATACCATAA

CTCGCGATCATTTTGTTATTGGTAGAAAAGCTGCATCATAACTCGAGt

catgtaattagttatgtcacgcttacattcacgccctccccccacatccgctctaaccgaaaaggaaggagttagaca

acctgaagtctaggtccctatttatttttttatagttatgttagtattaagaacgttatttatatttcaaatttttcttttttttctgt

acagacgcgtgtacgcatgtaacattatactgaaaaccttgcttgagaaggttttgggacgctcgaaggctttaatttg

cggccggtacccaat

127

3.8 References

1. Wang P, Bashiri G, Gao X, Sawaya MR, Tang Y. Uncovering the Enzymes that Catalyze

the Final Steps in Oxytetracycline Biosynthesis. J Am Chem Soc. 2013;135(19):7138-7141. doi:

10.1021/ja403516u.

2. Wang P, Kim W, Pickens LB, Gao X, Tang Y. Heterologous Expression and Manipulation

of Three Tetracycline Biosynthetic Pathways. Angewandte Chemie-International Edition.

2012;51(44):11136-11140. doi: 10.1002/anie.201205426.

3. SDS gel-loading buffer (2X). Cold Spring Harbor Protocols. 2006;2006(1). doi:

10.1101/pdb.rec407.

4. Gallagher SR. One-Dimensional SDS Gel Electrophoresis of Proteins. Curr Protoc Mol

Biol. 2006;75(1):10.12.11-10.12A.37. doi: 10.1002/0471142727.mb1002as75.

5. Ecker DJ, Stadel JM, Butt TR, Marsh JA, Monia BP, Powers DA, Gorman JA, Clark PE,

Warren F, Shatzman A, et al. Increasing gene expression in yeast by fusion to ubiquitin. J Biol

Chem. 1989;264(13):7715-7719.

6. Ecker DJ, Khan MI, Marsh J, Butt TR, Crooke ST. Chemical synthesis and expression of

a cassette adapted ubiquitin gene. J Biol Chem. 1987;262(8):3524-3527.

7. Schuster M, Einhauer A, Wasserbauer E, Süßenbacher F, Ortner C, Paumann M, Werner

G, Jungbauer A. Protein expression in yeast; comparison of two expression strategies regarding

protein maturation. J Biotechnol. 2000;84(3):237-248. doi: 10.1016/s0168-1656(00)00355-2.

8. Stȩpień PP, Brousseau R, Wu R, Narang S, Thomas DY. Synthesis of a human insulin gene

VI. expression of the synthetic proinsulin gene in yeast. Gene. 1983;24(2-3):289-297. doi:

10.1016/0378-1119(83)90089-6.

9. Cousens LS, Shuster JR, Gallegos C, Ku L, Stempien MM, Urdea MS, Sanchez-Pescador

R, Taylor A, Tekamp-Olson P. High level expression of proinsulin in the yeast, Saccharomyces

cerevisiae. Gene. 1987;61(3):265-275. doi: 10.1016/0378-1119(87)90190-9.

10. Steimer KS, Higgins KW, Powers MA, Stephans JC, Gyenes A, George-Nascimento C,

Luciw PA, Barr PJ, Hallewell RA, Sanchez-Pescador R. Recombinant polypeptide from the

128

endonuclease region of the acquired immune deficiency syndrome retrovirus polymerase (pol)

gene detects serum antibodies in most infected individuals. J Virol. 1986;58(1):9-16.

11. Barr P, Steimer K, Sabin E, Parkes D, Georgenascimento C, Stephens J, Powers M, Gyenes

A, Vannest G, Miller E. Antigenicity and immunogenicity of domains of the human

immunodeficiency virus (HIV) envelope polypeptide expressed in the yeast Saccharomyces

cerevisiae. Vaccine. 1987;5(2):90-101. doi: 10.1016/0264-410x(87)90053-3.

12. Barr PJ, Inselburg J, Green KM, Kansopon J, Hahm BK, Gibson HL, Lee-Ng CT, Bzik DJ,

Li W-b, Bathurst IC. Immunogenicity of recombinant Plasmodium falciparum SERA proteins in

rodents. Mol Biochem Parasitol. 1991;45(1):159-170. doi: 10.1016/0166-6851(91)90038-8.

13. Bachmair A, Finley D, Varshavsky A. In vivo half-life of a protein is a function of its

amino-terminal residue. Science. 1986;234(4773):179-186. doi: 10.1126/science.3018930.

14. Peng B, Williams TC, Henry M, Nielsen LK, Vickers CE. Controlling heterologous gene

expression in yeast cell factories on different carbon substrates and across the diauxic shift: a

comparison of yeast promoter activities. Microbial Cell Factories. 2015;14(1). doi:

10.1186/s12934-015-0278-5.

15. Ma SM, Li JWH, Choi JW, Zhou H, Lee KKM, Moorthie VA, Xie X, Kealey JT, Da Silva

NA, Vederas JC, Tang Y. Complete Reconstitution of a Highly Reducing Iterative Polyketide

Synthase. Science. 2009;326(5952):589-592. doi: 10.1126/science.1175602.

16. Jones EW. Tackling the protease problem in Saccharomyces cerevisiae. Methods Enzymol.

1991;194:428-453. doi: 10.1016/0076-6879(91)94034-A.

17. Romanos MA, Scorer CA, Clare JJ. Foreign gene expression in yeast: a review. Yeast.

1992;8(6):423-488. doi: 10.1002/yea.320080602.

18. Tøttrup HV, Carlsen S. A process for the production of human proinsulin inSaccharomyces

cerevisiae. Biotechnol Bioeng. 1990;35(4):339-348. doi: 10.1002/bit.260350403.

19. Rix U, Wang C, Chen Y, Lipata FM, Remsing Rix LL, Greenwell LM, Vining LC, Yang

K, Rohr J. The Oxidative Ring Cleavage in Jadomycin Biosynthesis: A Multistep Oxygenation

Cascade in a Biosynthetic Black Box. ChemBioChem. 2005;6(5):838-845. doi:

10.1002/cbic.200400395.

129

20. Perić-Concha N, Borovička B, Long PF, Hranueli D, Waterman PG, Hunter IS. Ablation

of theotcC Gene Encoding a Post-polyketide Hydroxylase from the Oxytetracyline Biosynthetic

Pathway inStreptomyces rimosusResults in Novel Polyketides with Altered Chain Length. J Biol

Chem. 2005;280(45):37455-37460. doi: 10.1074/jbc.M503191200.

21. Kharel MK, Pahari P, Shepherd MD, Tibrewal N, Nybo SE, Shaaban KA, Rohr J.

Angucyclines: Biosynthesis, mode-of-action, new natural products, and synthesis. Nat Prod Rep.

2012;29(2):264-325. doi: 10.1039/c1np00068c.

22. Koskiniemi H, Metsä-Ketelä M, Dobritzsch D, Kallio P, Korhonen H, Mäntsälä P,

Schneider G, Niemi J. Crystal Structures of Two Aromatic Hydroxylases Involved in the Early

Tailoring Steps of Angucycline Biosynthesis. J Mol Biol. 2007;372(3):633-648. doi:

10.1016/j.jmb.2007.06.087.

23. Kallio P, Liu Z, Mäntsälä P, Niemi J, Metsä-Ketelä M. Sequential Action of Two

Flavoenzymes, PgaE and PgaM, in Angucycline Biosynthesis: Chemoenzymatic Synthesis of

Gaudimycin C. Chem Biol. 2008;15(2):157-166. doi: 10.1016/j.chembiol.2007.12.011.

24. Dairi T, Nakano T, Aisaka K, Katsumata R, Hasegawa M. Cloning and Nucleotide

Sequence of the Gene Responsible for Chlorination of Tetracycline. Biosci, Biotechnol, Biochem.

2014;59(6):1099-1106. doi: 10.1271/bbb.59.1099.

25. Kong F, Yuan L, Zheng YF, Chen W. Automatic Liquid Handling for Life Science. Journal

of Laboratory Automation. 2012;17(3):169-185. doi: 10.1177/2211068211435302.

26. Dörr M, Fibinger MPC, Last D, Schmidt S, Santos-Aberturas J, Böttcher D, Hummel A,

Vickers C, Voss M, Bornscheuer UT. Fully automatized high-throughput enzyme library screening

using a robotic platform. Biotechnol Bioeng. 2016;113(7):1421-1432. doi: 10.1002/bit.25925.

27. Oliva B, Gordon G, McNicholas P, Ellestad G, Chopra I. Evidence that tetracycline analogs

whose primary target is not the bacterial ribosome cause lysis of Escherichia coli. Antimicrob

Agents Chemother. 1992;36(5):913-919. doi: 10.1128/aac.36.5.913.

28. Gossen M, Bujard H. Anhydrotetracycline, a novel effector for tetracycline controlled gene

expression systems in eukaryotic cells. Nucleic Acids Res. 1993;21(18):4411-4412. doi:

10.1093/nar/21.18.4411.

29. Asai M. Studies on Pillaromycin A. II. The Structure of Pillaromycinone. Chem Pharm

Bull (Tokyo). 1970;18(9):1706-1712. doi: 10.1248/cpb.18.1706.

130

30. Wells JS, O'Sullivan J, Aklonis C, Ax HA, Tymiak AA, Kirsch DR, Trejo WH, Principe

P. Dactylocyclines, novel tetracycline derivatives produced by a Dactylosporangium sp. I.

Taxonomy, production, isolation and biological activity. J Antibiot. 1992;45(12):1892-1898. doi:

10.7164/antibiotics.45.1892.

31. Tymiak AA, Aklonis C, Bolgar MS, Kahle AD, Kirsch DR, O'Sullivan J, Porubcan MA,

Principe P, Trejo WH. Dactylocyclines: novel tetracycline glycosides active against tetracycline-

resistant bacteria. J Org Chem. 1993;58(3):535-537. doi: 10.1021/jo00055a002.

131

4 Towards the biosynthesis of 6-demethyl-6-

epitracyclines from TAN-1612 in Saccharomyces

cerevisiae

*The contents of this chapter will be published in:

E. Herbst, V. W. Cornish, et al “Biosynthesis of 6-demethyl-6-epitetracycline analogs of TAN-

1612 in Saccharomyces cerevisiae”, in preparation.

132

4.1 Chapter outline

This chapter describes the work towards 6-demethyl-6-epitracycline derivatives of the fungal

anhydrotetracycline TAN-1612 in Saccharomyces cerevisiae. Building on the previous chapters

describing anhydrotetracycline hydroxylations, this chapter begins by exploring the product

formed by coexpressing the TAN-1612 pathway and the bacterial hydroxylase PgaE (Section

4.4.1). The testing of fungal hydroxylase homologs to DacO1, the 6α-hydroxylase of

anhydrodactylocyclinone is then described (Section 4.4.2) followed by the description of a method

for bacterial monooxygenase saturation mutagenesis near the anhydrotetracycline binding site,

with OxyS as an example (Section 4.4.3). Finally, the screening process of this natural diversity

and generated diversity by mutagenesis is described (Section 4.4.4) with future directions for

followup discussed. A key result described in this chapter is the isolation and characterization of

a new major product in a strain coexpressing PgaE and the TAN-1612 pathway that differs from

the TAN-1612 major product in the strain expressing the TAN-1612 pathway without PgaE

(Figure 4-2, Figure 4-3 and Figure 4-5). The followup plan on this result and on the screen for

TAN-1612 hydroxylases is outlined in the Discussion.

4.2 Introduction

TAN-1612, a fungal anhydrotetracycline derivative originally produced by Aspergillus niger, has

been introduced to S. cerevisiae by the Tang laboratory and our laboratory has further improved

its titers in synthetic media, thus allowing the heterologous production of TAN-1612

derivatives.(1) TAN-1612 differs from anhydrotetracycline in five functional groups, an A-ring

methyl ketone instead of the A-ring amide, a 4α-proton instead of the 4α-dimethylamino, a 4a-

hydroxy instead of the 4a-proton, a 6-proton instead of the 6-methyl and an 8-methoxy instead of

133

the 8-proton (Scheme 4-2). The stereochemistry of TAN-1612 at the 4a,12a positions has not yet

been verified, although the stereochemistry of the product of the homologous fungal polyketide

pathway, viridicatumtoxin, has been determined to be the same as anhydrotetracycline in these

positions.(2, 3) As outlined in the introduction chapter, the synthesis of 6-demethyl-6-

epiglycotetracyclines is highly desirable (Section 1.6). TAN-1612 presents a unique scaffold for

the biosynthesizing these derivatives in S. cerevisiae due to its ready biosynthesis in this

heterologous host and its unique functional groups among anhydrotetracycline (Section 1.6.4).

Much of the knowledge generated in the process of anhydrotetracycline hydroxylation by OxyS

and PgaE, as well as DacO1 expression optimization, described in the previous chapters, is directly

relevant to the hydroxylation of TAN-1612 in S. cerevisiae as outlined in this chapter.

4.3 Materials and methods

See Chapter 5 for general methods. See Chapter 3 for general protocol for high throughput assay

of hydroxylation/reduction assay in whole cells in microtiter plates and a general protocol for

western blots.

4.3.1 General protocol for high throughput assay of TAN-1612 hydroxylation

in whole cells in microtiter plates

Fresh colonies of strains harboring a plasmid encoding the TAN-1612 pathway and a hydroxylase

and control strains (EH-5-212-2 encoding PgaE and the TAN-1612 pathway as positive control

and EH-5-212-4 encoding the TAN-1612 pathway and no hydroxylase as a negative control) were

inoculated in 200 μL selective media (UT -) in 96-well plates (Corning 353072) and placed in

shaker overnight. Overnight cultures were used to inoculate 300 μL selective media (UT-) cultures

in deep well plates (Corning P-2ML-SQ-C-S) with an average starting OD of 0.01. The plate was

covered with two layers of SealMate film (Excel Scientific, SM-KIT-BS) and placed in shaker at

134

800 rpm for three nights. 200 μL H2O were then added and after resuspension 20 μL were diluted

into 180 μL of H2O in Corning 3603 plates before UV/VIS spectroscopic measurements. UV/VIS

spectroscopic measurements taken were as follows: (i) absorption spectrum 350 nm – 600 nm, (ii)

emission spectrum 450 nm – 650 nm (λexcitation = 400 nm), (iii) excitation spectrum 300 nm – 450

nm (λemission = 500 nm), (iii) excitation spectrum 300 nm – 450 (λemission = 560 nm). Spectral steps

in the spectra were 50 nm.

4.3.2 Protocol for selecting amino acids for mutagenesis in hypothesized

binding pockets of monooxygenases

The structure of aklavinone-11 hydroxylase with FAD and aklavinone (PDB ID 3IHG) was loaded

on PyMOL. Residues that are within 5 Å from the substrate aklavinone were selected as follows:

PyMOL>hide everything, all

PyMOL>select contacts, (resn VAK and chain A) around 5

Selector: selection "contacts" defined with 88 atoms.

PyMOL>select contacts_res, byres contacts

Selector: selection "contacts_res" defined with 268 atoms.

PyMOL>show sticks, contacts_res

Setting: seq_view set to on.

PyMOL>show sticks, resn vak

135

4.4 Results

4.4.1 Testing PgaE and other bacterial hydroxylases for TAN-1612

hydroxylation

Considering the mass spectrometry support for hydroxylation of anhydrotetracycline by OxyS and

PgaE (Chapter 3, Section 3.4.5) it was logical to test them for TAN-1612 hydroxyaltion (Scheme

4-1). SsfO1 was tested as well because of its efficient expression in S. cerevisiae and 6α-

hydroxylation of its native substrate (Section 3.4.5 and Scheme 4-2).

Scheme 4-1 Biosynthetic plan for TAN-1612 6α-hydroxylation

136

Scheme 4-2 Bacterial monooxygenases and their hypothesized substrates and products

Bacterial flavin-dependent monooxygenases OxyS, DacO1, SsfO1, CtcN and PgaE and their

native substrates. Marked in blue are differences between the native substrate and TAN-1612.

The strain expressing the TAN-1612 pathway without an additional hydroxylase has an excitation

peak at 420 nm for 560 nm emission and an emission peak of 530 nm for 400 nm excitation (Figure

4-1). This strain was confirmed to produce TAN-1612 as the major compound of 400 nm

absorption (Figure 4-5-a and Figure 4-2). The strains expressing OxyS and SsfO1 have similar

excitation and emission peaks, although both are increased for the sample encoding OxyS.

Notably, the strain encoding PgaE has a significantly reduced emission in 560 nm when excited at

137

420 +/- 30 nm (Figure 4-1), indicating that TAN-1612 is either not produced or produced in much

lower quantities than in the other strains.

Figure 4-1 UV/Vis analysis of hydroxylation attempt of TAN-1612 by bacterial hydroxylases

(a) Excitation spectrum (λemission = 560 nm) (b) emission spectrum (λexcitation = 400 nm) of strains

EH-5-212-1, EH-5-212-2, EH-5-212-3 and EH-5-212-4 harboring a plasmid encoding the TAN-

1612 pathway as well as a plasmid encoding OxyS, PgaE, SsfO1 and no hydroxylase, respectively.

Emission and excitation spectra were taken after diluting cultures that were incubated for 3 nights

in UT- media (4 mL) in 15 mL culture tubes (Corning 352059). Each data point and error bar

represents the average and standard error of three biological replicates, respectively.

138

In order to test which compound or compounds are being produced instead of TAN-1612 in the

strain expressing the TAN-1612 pathway and PgaE, both that strain and the control strain

expressing no hydroxylase were cultured in 500 mL scale to allow purification and further analysis

by mass spectrometry and NMR. Following three nights of culturing each culture was extracted

twice with EtOAc and the combined organic extract was washed with H2O and dried with Na2SO4

with the organic solvent removed under reduced pressure. The extract was then purified by

semiprepative HPLC prior to NMR and mass spectrometry analysis.

As the absorption chromatogram of the semipreparative HPLC separation shows, the main product

with regards to both 400 nm absorption and 254 nm absorption is modified in the +PgaE sample

relative to the -PgaE control. While the main product in the -PgaE control elutes after 37 minutes,

the main product in the +PgaE sample elutes after 34 minutes (Figure 4-2). Interestingly the -PgaE

sample has a minor product eluting after 34 minutes, which could be the same product as the main

product of the +PgaE sample, or a different one.

139

Figure 4-2 UV/Vis chromatogram of the HPLC separation of extracts from +/- PgaE strains

encoding the TAN-1612 pathway.

Strains EH-5-212-2 and EH-5-212-4 harboring a plasmid encoding the TAN-1612 pathway as well

as a plasmid encoding PgaE and no hydroxylase, respectively are indicated as +PgaE and -PgaE,

respectively. Both strains were cultured in 500 mL scale for three nights. Prior to HPLC separation

the culture was extracted twice with EtOAc and the combined organic extract was washed with

H2O and dried with Na2SO4, with the organic solvent removed under reduced pressure. Absorption

chromatograms are shown for 254 and 400 nm for an HPLC separation method of 10:90 to 90:10

MeCN in 99.9% H2O/0.1% TFA over 1 h.

140

The main product of each HPLC separation was isolated and analyzed by NMR and mass

spectrometry. The mass spectrometry analysis of the -PgaE compound clearly supports that TAN-

1612 was isolated (MS (ES+): m/z calc’d for C21H19O9+, 415.1029; found 415.1026 [M + H]+

(Figure 4-3-a) and the NMR spectrum matches the published TAN-1612 spectrum (Figure 4-5-

a).(1) For the +PgaE sample, the mass spectrum shows four major peaks for an elution peak with

absorbance at both 254 nm and 400 nm: 593.1218, 298.0743, 279.0966 and 278.0483 (Figure 4-3-

b). The NMR spectrum of the sample does not show the methoxy and methyl ketone protons of

shifts 3.89 and 2.58 ppm as well as the two aromatic protons of chemical shifts 7.05 and 6.45 ppm

and does show additional 7 aromatic protons in their stead (Figure 4-5-b). The possible identity of

the newly isolated major compound of the +PgaE +TAN-1612 pathway sample is discussed in

Section 4.5.1.

141

142

Figure 4-3 Mass spectrometry analysis of isolate from +/-PgaE strain encoding the TAN-

1612 pathway.

Liquid chromatography mass spectrometry analyses for isolates from strains EH-5-212-2 and EH-

5-212-4 harboring a plasmid encoding the TAN-1612 pathway as well as a plasmid encoding PgaE

and no hydroxylase, respectively are shown in (b) and (a), respectively. Both strains were cultured

in 500 mL scale for three nights. Prior to HPLC separation the culture was extracted twice with

EtOAc and the combined organic extract was washed with H2O and dried with Na2SO4, with the

organic solvent removed under reduced pressure. The LCMS separation method is 5:95 to 95:5

MeCN in 99.9% H2O/0.1% formic acid over 2 min.

Figure 4-4 MSMS analysis of isolate from +PgaE strain encoding the TAN-1612 pathway.

Liquid chromatography MSMS analyses for isolate from strain EH-5-212-2 harboring a plasmid

encoding the TAN-1612 pathway as well as a plasmid encoding PgaE with ion selection at mass

593. The LCMSMS separation method is 5:95 to 95:5 MeCN in 99.9% H2O/0.1% formic acid over

2 min.

143

Figure 4-5 1H-NMR spectra of isolates from +/-PgaE strains encoding the TAN-1612

pathway.

144

1H-NMR spectra of isolates from strains EH-5-212-2 and EH-5-212-4 harboring a plasmid

encoding the TAN-1612 pathway as well as a plasmid encoding PgaE and no hydroxylase, are

shown in (b) and (a), respectively. Both strains were cultured in 500 mL scale for three nights.

Prior to HPLC separation the culture was extracted twice with EtOAc and the combined organic

extract was washed with H2O and dried with Na2SO4, with the organic solvent removed under

reduced pressure. NMR spectra shown are in MeOD-d4 for the major product of each strain

according to HPLC chromatogram (Figure 4-2). 1H NMR (for (b), δ>4 ppm, 500 MHz, MeOD-

d4): δ 7.61 (dd, J = 8.0, 1.0 ,1H), 7.47 (d, J = 8.6, 2H), 7.22 (dd, J = 8.0, 1.1, 1H), 6.88 (dd, J =

8.6, 2H), 6.85 (dd, J = 8.0, 1H), 6.72 (s, 1H).

Figure 4-6 COSY NMR spectrum of isolate from +PgaE strain encoding the TAN-1612

pathway.

Strain EH-5-212-2 harboring a plasmid encoding the TAN-1612 pathway as well as a plasmid

encoding PgaE. Both strains were cultured in 500 mL scale for three nights. Prior to HPLC

separation the culture was extracted twice with EtOAc and the combined organic extract was

145

washed with H2O and dried with Na2SO4, with the organic solvent was removed under reduced

pressure. NMR spectra shown is in MeOD-d4 for the major product according to the HPLC

chromatogram (Figure 4-2).

4.4.2 Searching the fungal hydroxylase space for 6α-hydroxylation of TAN-

1612

Given the expression challenges with the bacterial monooxygenase DacO1 (Section 3.4.1) it was

logical to test fungal monooxygenases as well. Fungal monooxygenases were chosen based on

homology to DacO1 and/or based on the presence of anhydrotetracycline monooxygenase in their

CDS product name. Table 4-1 shows the first twelve such enzymes attempted. Western blot

analysis revealed that expression challenges could be prevalent with fungal hydroxylases as well,

with only 25% of the fungal hydroxylases assayed displaying a band at the expected size (Figure

4-7). The strains that did not indicate any protein expression at the expected size encoded fungal

monooxygenases of entries 1, 2, 4, 5, 6, and 11 from Table 4-1. The two strains that did exhibit

protein expression at the expected size encoded fungal hydroxylases entries 8 and 12 from Table

4-1 (Figure 4-7). For the small sample tested, the two codon optimized fungal monooxygenases

did not present a band of the expected size in S. cerevisiae. Two of the six non-codon optimized

did presented such a band. The two strains that exhibited a protein of the expected size had a higher

identity %, positives % and query cover with both DacO1 and OxyS than the six strains that did

not exhibit a band of the expected size but not necessarily to a statistically significant degree

(Figure 4-7, Table 4-1 and data not shown).

146

Table 4-1 Fungal monooxygenases biomined for TAN-1612 6α-hydroxylation in S. cerevisiae

Entry Gene product

label

Source organism GenBank

entry

Codon

optimi

zed

Identity

(%) /

positives

(%) /

cover

with

DacO1a

Identity

(%) /

positives

(%) /

cover

with

OxySb

1 Anhydrotetracy

cline

monooxygenas

e

Valsa mali strain

03-8

CM00310

4.1

Y 30/44/266 25/39/388

2 Anhydrotetracy

cline

monooxygenas

e

Penicillium

subrubescens

strain CBS 132785

MNBE01

000128.1

N 32/43/533 35/46/541

3 Anhydrotetracy

cline

monooxygenas

e

Cercospora

beticola

NC_036772

NC_0367

72.1

N 30/41/510 34/45/537

4 related to

tetracenomycin

polyketide

synthesis

hydroxylase

tcmG

Phialocephala

subalpina strain

UAMH 11012

FJOG010

00008.1

N 31/46/370 30/43/547

5 Anhydrotetracy

cline

monooxygenas

e

Golovinomyces

cichoracearum

MCBR01

007648.1

N 28/43/398 30/40/564

6 Anhydrotetracy

cline

monooxygenas

e

Valsa mali var.

pyri strain

SXYL134

KN71467

1.1

N 23/37/345 24/38/405

7 tetracycline 6-

hydroxylase

protein

Aureobasidium

pullulans var.

pullulans

KL584983

.1

N 26/40/416 25/39/455

8 pentachlorophe

nol 4-

monooxygenas

e

Hypoxylon sp.

EC38

KZ111246

.1

N 34/45/520 38/49/531

9 probable 2-

polyprenyl-6-

methoxyphenol

Phialocephala

subalpina strain

UAMH 11012

FJOG010

00031.1

N 32/46/505 39/51/506

147

hydroxylase

and related

FAD-

dependent

oxidoreductase

s

10 2-polyprenyl-6-

methoxyphenol

hydroxylase

Aspergillus oryzae

100-8

AMCJ010

00140.1

N 32/47/501 34/50/503

11 pentachlorophe

nol 4-

monooxygenas

e

Colletotrichum

orbiculare MAFF

240422

KB72597

6.1

Y 34/44/531 34/47/531

12 unnamed

protein product;

2-polyprenyl-6-

methoxyphenol

hydroxylase

and related

FAD-

dependent

oxidoreductase

s"

Aspergillus oryzae

RIB40

AP007159

.1

N 32/45/521 34/47/529

aDacO1 [Dactylosporangium sp. SC14051] GenBank: AFU65892.1

b OxyS [Streptomyces rimosus] GenBank AAZ78342.1

148

Figure 4-7 Western blot analysis of fungal hydroxylase expression in BJ5464-NpgA

Strain cultures were lysed with Y-PER and labeled with Monoclonal ANTI-FLAG HRP antibody.

The size (kDa) indicated below the gels is the expected size of the protein based on the amino acid

sequence. Strains EH-5-227-1 through EH-5-227-9 encode Fungal monooxygenase entry 6, 5, 1,

4, 2, 2, 8, 11, 12, from Table 4-1, respectively.

149

4.4.3 Evolving monooxygenases for 6α-hydroxylation of anhydrotetracyclines

– OxyS example

Considering the effective hydroxylation of anhydrotetracycline in yeast by OxyS (Chapter 2), its

apparent lack of hydroxylation activity on TAN-1612 (Section 4.4.1) and an effective UV/Vis

assay for anhydrotetracyclines hydroxylation (Sections 3.4.3-3.4.5), directed evolution of OxyS to

accept TAN-1612 as a substrate seemed logical. In addition, OxyS structure was previously probed

by X-ray crystallography, as well as the structure of a homologous protein, Aklavinone-11-

Hydroxylase, along with its native substrate, aklavinone (42% homology, PDB ID 4K2X and

3IHG, respectively).(4, 5) Thus, the substrate binding pocket of OxyS could be identified with

some confidence and mutated accordingly.

In order to perform saturation mutagenesis on residues that are in proximity to the hypothesized

substrate binding pocket of OxyS, residues of Aklavinone-11-Hydroxylase that are within 5 Å of

aklavinone were listed (Section 4.3.2). Then the homologous residues in OxyS were noted as well

(Table 4-2). Some of these homologous residues in OxyS indeed sit in proximity to a cavity that

is a continuation of the cavity in which FAD is situated in the OxyS crystal structure (Figure 4-8).

Table 4-2 Residues chosen for mutagenesis in OxyS

Residue

number

in 3IHG

Residue context in

3IHG

Residue in

OxyS

Residue context

in OxyS

Distance

from VAK

(Å)

1 R45 PYPRAAG K42 FSKALGV 4-5

2 A47 PRAGQN A43 SKALGVH <4

3 G48 AAGQNPR G45 KALGVHAR 4-5

4 I72 ADDIRG - - <4

5 F79 QGDFVIR - - <4

6 I81 QGDFVIR - - 4-5

7 M101 DDMVAA - - 4-5

8 W113 PAGWAM L95 PYALFV 4-5

9 M115 PAGWAMLSQD F96 PYAL-FVPQV <4

10 M201 LTHMVGV M176 --HMFAV <4

11 W221 GTTGWYYL W211 DLRAWFAAFP <4

150

12 Y223 TGWYYLH F212 DLRAWFAAFPL <4

13 T232 PEFKGTFGPTDRP T225X ATVA <4

14 P235 TFGPTDRP F228 TVAFFDRP 4-5

15 F245 RHTLFVEYDPDEG V240X RRAPVTEED <4

16 W287 VDIQGWEMAARIA - - <4

17 M289 WEMAARIA - - 4-5

18 P314 VTPPTGGMSG P295X IHLPAGGQGL <4

19 T315 VTPPTGGMSG A296X IHLPAGGQGL <4

20 G316 VTPPTGGMSG G297X IHLPAGGQGL <4

21 G317 VTPPTGGMSG G298X IHLPAGGQGL <4

22 R373 AIYAQRMAP P357 LIDPDPRYE 4-5

23 M374 AIYAQRMAP R358 LIDPDPRYE 4-5

24 Y387 SVGYPET V372 LLHVPET 4-5

151

Figure 4-8 PyMOL illustrations of amino acids in proximity to the anhydrotetracycline

substrate in aklavinone-11-Hydroxylase and in OxyS

PyMOL illustration of (a) alkavinone and FAD (stick representation, carbons colored white)

surrounded by amino acids of aklavinone-11-Hydroxylase within 5 Å of alkavinone (stick

representation, carbons colored green, PDB ID 3IHG) and (b) FAD (carbons colored white)

surrounded by OxyS (surface representation, carbons colored green, PDB ID 4K2X) and its amino

152

acids that are homologous to those of aklavinone-11-Hydroxylase amino acids that are within 5 Å

of alkavinone in the structure shown in (a).

4.4.4 Library screening for TAN-1612 hydroxylation in S. cerevisiae

Following the assembly of the libraries of fungal monooxygenases (Section 4.4.2) and OxyS

saturation mutagenesis (Section 4.4.3) the libraries were screened for TAN-1612 hydroxylation

using a variant of the microtiter plate assay for anhydotetracycline hydroxylation (Section 3.4.3

and 4.3.1). As negative control a strain encoding the TAN-1612 pathway without an additional

hydroxylase was used. As positive control a strain encoding the TAN-1612 pathway and PgaE was

used, as that strain was already shown to produce an alternative major product instead of TAN-

1612 (Figure 4-2).

Given that the positive control strain expressing PgaE showed a significantly reduced emission at

560 nm upon 400 and 450 nm excitation, colonies were plotted according to their absorbance at

these wavelengths and normalized to OD6oo: (400 + 450 nm) / 600 nm. When colonies of each 96-

well plate was ranked according to this absorption criteria the PgaE positive control was ranked

on average 83.6 (out of 96) with a standard deviation of 10.0 (Table 4-3 and Figure 4-9). A low

rank of PgaE is expected in the likely scenario that most strains screened do not encode an efficient

TAN-1612 hydroxylase. Wells 84 and 96 containing no cells consistently ranked the lowest in the

measurement of (400 + 450)/600 nm (Figure 4-9).

Table 4-3 Rank of PgaE positive control in screen for TAN-1612 hydroxylase

Plate Strain Absorbance C1

(400 + 450)/600

Absorbance C2

(400 + 450)/600

a EH-5-217-2 89 59

b EH-5-217-2 91 92

c EH-5-217-2 94 68

d EH-5-217-4 88 79

e EH-5-217-4 78 85

f EH-5-217-4 93 87

153

g EH-5-217-1 and

EH-217-3

81 87

Average (all) 83.6

Standard

deviation (all)

10.0

Figure 4-9 Library screening for TAN-1612 hydroxylation in S. cerevisiae

Y axes shows the sum of absorption at 400 and 450 nm divided by absorption at 600 nm while the

X axes show the colony number. Plates a, b and c are screens of strain library EH-5-217-2. Plates

d, e and f are screens of strain library EH-5-217-4. Both strain libraries EH-5-217-2 and EH-5-

217-4 encode the same OxyS saturation mutagenesis library and differ in the background TAN-

1612 producing strain. Plate g contains strain libraries EH-5-217-1 and EH-5-217-3 encoding

bacterial and fungal hydroxylases, including selected DacO1 fusion proteins. Strains EH-5-212-1,

154

EH-5-212-2 and EH-5-212-4 encoding the TAN-1612 pathway along with OxyS, PgaE and no

hydroxylase, are labeled in blue, red and white, respectively.

4.5 Discussion

4.5.1 Testing PgaE and other bacterial hydroxylases for TAN-1612

hydroxylation

Despite the structural dissimilarities between UWM6, the native susbstrate of PgaE and TAN-

1612 (Scheme 4-2) it is PgaE and not OxyS or SsfO1 whose coexpression with the TAN-1612

pathway has led to the production of a different major product of 400 and 254 nm absorption

instead of TAN-1612. Both the chromatograms for the HPLC separation of the +PgaE and -PgaE

samples, as well as the excitation and emission spectra support that PgaE is converting TAN-1612

or an intermediate in its biosynthesis (Figure 4-2, Figure 4-1). It would therefore be of interest to

test other enzymes involved in the 12-position hydroxylation of UWM6, such as CabE, LanE,

UrdE and GilOI.(6, 7)

The protons of chemical shifts 7.47 and 6.88 ppm with coupling constant of 8.6 Hz are likely two

pairs of equivalent aromatic protons at ortho to each other and they are shown to interact in the

COSY spectrum as well (Figure 4-5 and Figure 4-6); the protons of chemical shifts 7.61, 7.22 and

6.85 ppm are likely three adjacent protons in one aromatic ring that has three other substituents,

with the protons of chemical shifts 7.22 and 7.61 ppm likely meta to each other. Interactions

between the protons of chemical shifts 7.61 and 6.85 as well as 7.22 and 6.85 ppm are also readily

observed in the COSY spectrum and the coupling constants, characteristic of ortho interactions,

are 8.0 Hz each (Figure 4-5 and Figure 4-6). The singlet at 6.72 ppm is likely a single proton in an

aromatic ring, possibly the only remaining proton in the D-ring of the substituted TAN-1612 or its

155

intermediate (Figure 4-5). The absence of the methoxy shift in the new major product at 3.89 ppm

(Figure 4-5) can indicate that PgaE derivatization occurs prior to AdaD methylation, which is

assumed to be the last step in TAN-1612 biosynthesis.(1) It could be that following hydroxylation

by PgaE and a potential further derivatization by a yeast endogenous molecule, substrate affinity

of AdaD to the modified product is too low to allow methylation. Examples for the moieties that

might be part of the newly formed major compound are shown in Figure 4-10. Importantly, not all

moieties are necessarily part of the same molecule, as can be verified by larger scale fermentation

followed by further purification and NMR analysis. The aromatic moiety of Figure 4-10-c could

theoretically represent a doubly hydroxylated TAN-1612 or any of the TAN-1612 intermediates

of Figure 4-11. However such an assignment is questionable, given the lack of the protons of

chemical shift 3.89 and 2.58 ppm corresponding to the methoxy and methyl ketone at the NMR

spectrum of the newly formed major compound (compare Figure 4-5-a and Figure 4-5-b).

Figure 4-10 Possible moieties in the product isolated from +PgaE strain coexpressing the

TAN-1612 bioysnthetic pathway

Either of the two aromatic moieties of (a) could be responsible for generating the protons of

chemical shifts 7.61, 7.22 and 6.85 ppm, the aromatic moiety of (b) could be responsible for

generating protons of chemical shifts 7.47 and 6.88 ppm and either of the aromatic moieties of (c)

could be responsible for generating a proton of chemical shifts 6.72 ppm in the 1H-NMR spectrum

of the product isolated from +PgaE strain coexpressing the TAN-1612 bioysnthetic pathway

(Figure 4-5). The squiggly lines represent any non-proton substituent.

156

Figure 4-11 Possible intermediates in the biosynthesis of TAN-1612

TAN-1612 (1) and its intermediates 2-10 that can occur in the case of exclusion or disfunction of

AdaB, AdaC or AdaD, the three post PKS biosynthetic enzymes of the TAN-1612 biosynthetic

pathway, or combinations thereof.(1)

157

The mass spectrum of the isolated compound from the +PgaE culture has major peaks with m/z

values of 593.1218, 298.0743, 279.0966 and 278.0483 (Figure 4-3). The 593.1218 ion has an

elution peak after 1.60 and after 1.47 min, but an absorption peak at 254 nm and 400 +/- 60 nm is

noted only slightly before the 1.60 min (Figure 4-3) and not slightly before the 1.47 peak (the ions

are first detected at the diode array before mass spectrometry scanning). Assuming that the two

593.1218 ions could be related stereoisomers of the same exact mass, it is perhaps doubtful

whether any of them corresponds to a derivative of the TAN-1612 intermediates shown in Figure

4-11. This is because at any stereoisomeric form such compounds would be expected to have

absorption at 400 and/or 254.

The 298.0743 and 278.0483 ions are four protons and one oxygen different in their m/z values.

Possibly a molecule of H2O + oxidation / reduction apart. In the MSMS spectrum for the ions of

m/z 593 both the ions 278.0452 and 296.0551 appear (Figure 4-4), the former being the mass of

H2O less than the latter and the latter being H2 away from 298.0743 in its mass. Also of note is

that the 593.1218 ion could be a result of a dimer of the 296.0551 ion plus the mass of a proton.

Thus, the 593.1218, 298.0743, 296.0551 and 278.0483 ions could correspond to chemical formulas

of C32H21N2O10+, C16H12NO5

+, C16H10NO5+, C16H8NO4

+ as they differ in 0.0022, 0.0028, 0.0008

and 0.0030 u, respectively, from the expected masses of these ions. These could be ions of some

xanthurenic acid derivatives that PgaE might or might not have a contribution to their biosynthesis

(Figure 4-12). While the derivatives of xanthurenic acid shown in Figure 4-12 are not known in

yeast, xanthurenic acid is a known yeast metabolite.(8) This proposal of structure or any other

structure that involves only yeast metabolites and their derivatives could be explaining the mass

and NMR spectra of the isolated major compound in the +PgaE sample (Figure 4-3, Figure 4-4,

Figure 4-5 and Figure 4-6). However, a major compound that does not involve a derivative of

158

TAN-1612 or its intermediate still does not directly explain how the expression of PgaE leads to

the reduction in the TAN-1612 absorbance and fluorescence (Figure 4-1 and Figure 4-2).

Figure 4-12 xanthurenic acid and its derivatives that could correspond to experimental mass

and NMR spectra

The xanthurenic acid derivatives presented in this figure have [M + H]+ values of 593.1196,

298.0715 and 296.0559 that are 0.0022, 0.0028 and 0.0008 amu from the experimental values

detected 593.1218, 298.0743 and 296.0551 (Figure 4-3 and Figure 4-4). In addition, the protons

of a molecule such as the one shown with chemical formula C16H11NO5 can correspond to the 6

proton types identified in the NMR spectrum (Figure 4-5 and Figure 4-6) and identified to

potentially belong to the moieties described in Figure 4-10.

In order to determine the structure of the newly formed major compound with regards to 254 nm

and 400 nm absorption in the +PgaE sample coexpressing the TAN-1612 pathway, the reaction

would be repeated in larger scale to isolate enough of the newly formed compound for HSBC and

HMBC proton coupled carbon NMR spectra. Furthermore, the minor compound eluting at 34 min

for the -PgaE sample will be isolated and analyzed as well to identify whether it is identical or at

all related to the product isolated from the +PgaE sample that elutes at 34 min (Figure 4-2). This

159

characterization can assist in understanding the role PgaE has on the biosynthesis of the newly

formed major compound with respect to 400 nm and 254 nm absorption in the +PgaE sample

(Figure 4-2). Another source of information would be the culturing and isolation of another control

strain encoding PgaE but not encoding the TAN-1612 pathway and examining whether such strain

displays a similar major peak in the HPLC chromatogram of 254 and 400 nm absorption as the

strain that encodes the TAN-1612 pathway.

4.5.2 Searching the fungal hydroxylase space for 6α-hydroxylation of TAN-

1612

Importantly, the library generation approaches used to generate 6α-hydroxylases of

anhydrotetracycline (Subsections 3.4.2 and 3.4.4) could be used for generating TAN-1612

hydroxylases (Subsections 4.4.2 and 4.4.3) and vice versa. The expression analysis of fungal

monooxygenases showed that while branching towards fungal monooxygenases might be a useful

strategy to find 6α-hydroxylases and specifically 6α-hydroxylases of TAN-1612, the fungal

monooxygenases do not necessarily present an advantage over their bacterial counterparts as far

as expression challenges in S. cerevisiae are concerned (Figure 4-7 and Chapter 3).

4.5.3 Evolving monooxygenases for 6α-hydroxylation of anhydrotetracyclines

Amino acids in OxyS were chosen for mutagenesis based on homology to amino acids in the

proximity of the substrate in the crystal structure of RdmE and this approach could be implemented

in other monooxygenases, such as PgaE or expression optimized DacO1 (Chapter 3). Importantly,

implementing this approach does not require an existing crystal structure of the monooxygenase

of interest. Also, as seen in the case of OxyS, at least some of the amino acids chosen for

mutagenesis according to this strategy actually sit in proximity to the anhydrotetracycline cavity

of OxyS (Figure 4-8). However, in the case of OxyS, an approach that uses the published crystal

160

structure of OxyS to choose additional amino acids for mutagenesis or to avoid mutating some of

the amino acids that are not in proximity to the cavity might have been preferred. Further

mutagenesis is being performed to explore the effect of additional mutations of OxyS in positions

that are homologous to amino acids in RdmE structure that are up to 8 Å from aklavinone in 3IHG.

Finally, similar followup mutagenesis studies on stably expressed DacO1 fussion proteins, as well

as PgaE will be directly relevant for testing 6α-hydroxylation of TAN-1612. Specifically for PgaE,

much structural information is available to guide the amino acid choice for saturation mutagenesis

from its crystal structure and from biochemical analyses.(6, 9-11)

4.5.4 Library screening for TAN-1612 hydroxylation in S. cerevisiae

In the screen for TAN-1612 hydroxylation, a variant of the microtiter plate assay for

anhydrotetracycline hydroxylation described in Chapter 3 was used. Chapter 3 also discusses non-

microtiter plate avenues for the anhydrotetracycline hydroxylation (Section 3.5.3) and these would

be especially interesting to examine in the context of TAN-1612 hydroxylation, as TAN-1612 does

not need to be exogenously supplied to the cells.

Similarly to the followup on the +PgaE +TAN-1612 encoding strain (Sections 4.4.1 and 4.5.1),

strains with a lower (400 + 450)/600 nm absorption than the PgaE positive control (Figure 4-9)

will be assayed for TAN-1612 hydroxylation by UV/Vis and mass spectrometry. If data supports

production of an alternative major product to TAN-1612 these strains will be cultured in larger

scale to allow purification and analysis of the product by NMR. If data does not support TAN-

1612 hydroxylation in any of the assayed strains, strain selection for further assay will be attempted

based on other criteria such as 550 + 600 – 450 nm emission upon 400 nm excitation or 400 + 450

– 350 nm excitation for 560 nm emission. These criteria are based on the reduced emission and

excitation peaks observed for the PgaE encoding positive control strain (Figure 4-1).

161

4.6 Conclusion

This chapter describes the progress towards TAN-1612 hydroxylation in S. cerevisiae, the first

step in the synthesis of 6-demethyl-6-epitetracycline from TAN-1612 in S. cerevisiae. This chapter

shows that a strain expressing both PgaE and the TAN-1612 pathway produces a major compound

with regards to 400 nm and 254 nm absorption that differs from TAN-1612 (Sections 4.4.1 and

4.5.1, Figure 4-1, Figure 4-2, Figure 4-3 and Figure 4-5). Future work towards TAN-1612

hydroxylation in S. cerevisiae should include a larger scale culturing of the +PgaE +TAN-1612

pathway strain to enable followup NMR experiments. In addition, the minor compound eluting at

34 min for the -PgaE sample (Figure 4-2) will be isolated and analyzed to verify whether it is

identical or at all related to the product isolated from the +PgaE sample that elutes at 34 min.

Another control strain encoding PgaE but not encoding the TAN-1612 pathway will be cultured

and examined to discover whether it displays a similar major peak in the HPLC chromatogram to

that of the +PgaE +TAN-1612 pathway strain.

This chapter also describes a hydroxylase harvesting and mutagenesis strategy as well as a UV/Vis

assay as a platform for identifying TAN-1612 hydroxylation. This platform was applied to the

screening for TAN-1612 hydroxylation (Section 4.4.4) by OxyS mutants (Section 4.4.3), DacO1

fusion proteins (Sections 3.4.1 and 3.4.5), and other hydroxylases from bacterial (Sections 3.4.2

and 3.4.5) and fungal sources (Section 4.4.2). Followup culturing and spectroscopic analysis will

be done on strains coexpressing the TAN-1612 pathway and one of the above mentioned

hydroyxlases, identified as potentially hydroxylating TAN-1612 in the UV/Vis assay (Figure 4-9).

In addition to the followup on PgaE and on the other hydroxylases described in this chapter, other

enzymes involved in the 12-position hydroxylation of UWM6, such as CabE, LanE, UrdE and

GilOI should also be tested for TAN-1612 hydroxylation.(6, 7) With a mutagenesis strategy that

162

does not require a crystal structure that could be used for DacO1 (Figure 4-8) and with the

additional benefit of the PgaE crystal structure,(9) followup mutagenesis studies such as shown on

OxyS (Section 4.4.3) and on DacO1 (Section 3.4.4) on stably expressed DacO1 fusion proteins

(Section 3.4.5), as well as on PgaE could prove beneficial for 6α-hydroxylation of TAN-1612.

Identification and overexpression of a reductase responsible for 5a(11a)-dehydrotetracycline

reduction in S. cerevisiae (Section 2.4.2 and 2.5.2) will be potentially useful for the reduction of

the hydroxylation product of TAN-1612 as would be other efforts in the reduction of 5a(11a)-

dehydrotetracycline in S. cerevisiae (Section 3.4.5 and 3.5.5). Finally, the anhydrotetracycline

hydroxylations described in this chapter and in Chapters 2 and 3 relies heavily on the UV/Vis assay

developed for the hydroxylation of anhydrotetracyclines (Section 3.4.3, 3.4.5, and 4.4.4). More

broadly, high-throughput assays are of very high importance to small molecule production in

microorganisms because of the inability to predict in advanced the successful strain modifications

needed in the complex environment of the cell.(12) TAN-1612 and anhydrotetracycline

hydroxylation belong to a special group of biochemical transformations that are easy to assay in

high throughput because of the inherent change in the chromophore of the CD ring (Scheme 4-2).

However, the next steps for 6-demtehyl-6-epiglycotetracyclines from TAN-1612 will require a

general, readily implemented, high-throughput assay for tetracycline biosynthesis in yeast. The

development and characterization of such an assay is described in Chapter 5.

163

4.7 Appendix

Table 4-4 Strains used in this study

Strain Genotype Source/Reference

BJ5464-

NpgA

MATα ura3-52 his3-Δ200 leu2-Δ1

trp1 pep4::HIS3 δ:: pADH2-npgA-tADH2 prb1Δ1.6R

can1 GAL

a / (13)

PBA-482 BJ5464 ΔPEP4::pTDH3-Sb-NpgA-tENO2

ΔPRB1::pTDH3-Sb-NpgA-tENO2

P. A. Baldera-

Aguayob

PBA-1277 BJ5464 ΔPEP4::pHSP26-Sb-NpgA-tENO2

ΔPRB1::pHSP26-Sb-NpgA-tENO2

P. A. Baldera-

Aguayob

PBA-1333 PBA482 pPBA-L2BWG9 P. A. Baldera-

Aguayob

PBA-1337 PBA1277 pPBA-L6BWG9 P. A. Baldera-

Aguayob

EH-5-212-1 PBA-1337 AL-1-101-C10 This Study

EH-5-212-2 PBA-1337 AL-234-C-C1 This Study

EH-5-212-3 PBA-1337 AL-239-B This Study

EH-5-212-4 PBA-1337 pSP-G1 (Addgene 64736) This Study

EH-5-217-1 PBA-1337 Plasmid Libraries A + C This Studyc

EH-5-217-2 PBA-1337 Plasmid Library D This Studyc

EH-5-217-3 PBA-1333 Plasmid Libraries B + C This Studyc

EH-5-217-4 PBA-1333 Plasmid Library D This Studyc

EH-5-227-11 BJ5464-NpgA AL-2-72-B-C6 This Study

EH-5-227-12 BJ5464-NpgA AL-2-72-D-C5 This Study

EH-5-227-13 BJ5464-NpgA AL-2-72-F-C4 This Study

EH-5-227-14 BJ5464-NpgA AL-2-79-J-C4 This Study

EH-5-227-15 BJ5464-NpgA AL-2-79-K-C5 This Study

EH-5-227-16 BJ5464-NpgA AL-2-79-L-C6 This Study

EH-5-227-17 BJ5464-NpgA AL-2-86-D-C5 This Study

EH-5-227-18 BJ5464-NpgA AL-2-86-I-C5 This Study

EH-5-227-19 BJ5464-NpgA AL-2-86-A-C1 This Study a Y. Tang, University of California, Los Angeles, Department of Chemical and Biomolecular

Engineering & Department of Chemistry and Biochemistry, Department of Bioengineering

b Integrated Program in Cellular, Molecular and Biomedical Studies, Cornish Laboratory,

Columbia University, New York, NY 10032, USA

c Plasmid libraries compositions are described below

164

Table 4-5 Plasmids used in this study

Description Source/Reference

pSP-G1 pTEF1-FLAG-tADH1, pPGK1-MYC-tCYC1, 2 μ, Ura Addgene 64736

AL-1-101-C10 pTEF1-oxyS-tADH1 in pSP-G1 Chapter 2

AL-1-234-C-C1 pTEF1-pgaE-tADH1 in pSP-G1 JCAT codon opt. Chapter 3

AL-1-239-B-C3 pTEF1-ssfO1-tADH1 in pSP-G1 JCAT codon opt. Chapter 3

AL-2-72-B-C6 pTEF1-Fungal-monooxygenase-6-tADH1 in pSP-G1c This Study

AL-2-72-D-C5 pTEF1-Fungal-monooxygenase-5-tADH1 in pSP-G1c This Study

AL-2-72-F-C4 pTEF1-Fungal-monooxygenase-1-tADH1 in pSP-G1c This Study

AL-2-79-J-C4 pTEF1-Fungal-monooxygenase-4-tADH1 in pSP-G1c This Study

AL-2-79-K-C5 pTEF1-Fungal-monooxygenase-2-tADH1 in pSP-G1c This Study

AL-2-79-L-C6 pTEF1-Fungal-monooxygenase-2-tADH1 in pSP-G1c This Study

AL-2-86-D-C5 pTEF1-Fungal-monooxygenase-8-tADH1 in pSP-G1c This Study

AL-2-86-I-C5 pTEF1-Fungal-monooxygenase-11-tADH1 in pSP-G1c This Study

AL-2-86-A-C1 pTEF1-Fungal-monooxygenase-12-tADH1 in pSP-G1c This Study

EH-5-49-C pTEF1-GAL1-dacO1-tADH1 in pSP-G1 Chapter 3

EH-5-80-6 pTEF1-UBI4-IFNG-dacO1-tADH1 in pSP-G1 Chapter 3

EH-5-80-7 pTEF1-IFNG-dacO1-tADH1 in pSP-G1 Chapter 3

EH-5-80-10 pTEF1-oxyS-dacO1-37-tADH1 in pSP-G1 Chapter 3

EH-5-80-11 pTEF1-oxyS-dacO1-87-tADH1 in pSP-G1 Chapter 3

pAV124 Trp1, 2 μ acceptor vector for yeast golden gate Addgene

63190/(14)

pPBA-AV124 Modified Trp1, 2 μ acceptor vector for yeast golden gate P. A. Baldera-

Aguayoa

pPBA-

L2BWG9

pHSP26-adaA-tPGK1_pPGK1-adaB-tADH1_pTEF1-

adaC_tENO1_pTDH3-adaD_-tRPL15A on pPBA-

AV124

P. A. Baldera-

Aguayoa

pPBA-

L6BWG9

pHSP26-adaA-tPGK1_pHSP26-adaB-tADH1_pTEF1-

adaC_-tENO1_pTDH3-adaD-tRPL15A on pPBA-

AV124

P. A. Baldera-

Aguayoa

a Integrated Program in Cellular, Molecular and Biomedical Studies, Cornish Laboratory,

Columbia University, New York, NY 10032, USA

b Fungal-monooxygenase-X refers to the fungal monooxygenase from Table 4-1 entry X. Thus,

for example Fungal-monooxygenase-2 refers to the fungal monooxgenase from Table 4-1 entry 2.

165

Plasmid libraries used for making strains EH-5-217-1 through EH-5-217-4

Table 4-6 Plasmid Library A – Strain EH-5-217-1

Library component

no.

Plasmid Plasmid common

name

Colony #

1 EH-5-49-C Gal1-DacO1 1

2 EH-5-80-6 9 - ubiIFN H

3 EH-5-80-7 9 - IFN H

4 EH-5-80-10 11-OxyS-DacO1-37 G

5 EH-5-80-11 11-OxyS-DacO1-87 H

Table 4-7 Plasmid Library B – Strain EH-5-217-3

Library component

no.

Plasmid Plasmid common

name

Colony #

1 EH-5-49-C Gal1-DacO1 1

2 EH-5-80-6 9 - ubiIFN H

3 EH-5-80-7 9 - IFN H

4 EH-5-80-10 11-OxyS-DacO1-37 G

5 EH-5-80-11 11-OxyS-DacO1-87 H

6 AL-234-C PgaE in pSPG1 1

7 AL-1-101 OxyS 10

8 AL-239-B SsfO1 3

Table 4-8 Plasmid Library C – Strain EH-5-217-1 and EH-5-217-3

* AL-2-79-K and AL-2-86-L are the same plasmid but one of them does and the other does not

encode a mutation in the sequence.

no. Plasmid Plasmid common name Colony #

1 AL-2-72-B Atc Moase Valsa Mali Var Pyr C6

2 AL-2-72-D Atc Moase G Cichoaracearumi C5

3 AL-2-72-F Atc Moase Valsa Mali CM C4

4 AL-2-79-J Related to tetra hydroxylase P subalpina C4

5 AL-2-79-K Atc Moase P subrubescens C5

6 AL-2-79-L* Atc Moase P subrubescens C6

7 AL-2-86-D Pentachlorophenol-4-Moase Hypoxylon C5

8 AL-2-86-I Pentachlorophenol-4-Moase Colletotrichum orbiculare C5

9 AL-2-86-A 2-ployprenyl-6-methoxyphenol-hydroxylase Aspergillus

Oryzae RIB40

C1

166

Table 4-9 Plasmid Library D – Strains EH-5-217-2 and EH-217-4

no. Plasmid Plasmid common name

1 AL-2-57-A F96X OxyS Library

2 AL-2-57-B M176X OxyS Library

3 AL-2-57-C W211X OxyS Library

4 AL-2-57-D F212X OxyS Library

5 AL-2-57-E T225X OxyS Library

6 AL-2-57-F V240X OxyS Library

7 AL-2-57-G P295X OxyS Library

8 AL-2-57-H A296X OxyS Library

9 AL-2-83-A A43X OxyS Library

10 AL-2-83-B G297X OxyS Library

11 AL-2-83-C G298X OxyS Library

Table 4-10 Sequences used in this study

Sequences for the hydroxylases fungal hydroxylases of Table 4-1 are shown capitalized within the

context of the pSP-G1 backbone containing pTEF1, the FLAG tag and tADH1 (partial,

decapitalized).

pTEF1-

CM003104

.1-tADH1-

in-pSP-G1

(Entry 1

Table 4-1,

AL-2-72-

F-C4)

tcaagtttcagtttcatttttcttgttctattacaactttttttacttcttgctcattagaaagaaagcatagcaatctaatctaa

gttttaattacaagcggccgcATGATGGGTGCAAAACACGTTATTAGAAGAGTGA

CCAATTCGGAAAATGATACAGGATCAAGTTCGAAATCCACTACTTCA

TCTAATTCCGACAGGGACCATAATCCCTCTTCCTCTAATAGAGACAG

ATATCACAATGAAACATCACCTGCCAATATGATATTGATAAACGAGC

CACCTCCTACCACCTCCACAGAATTGGAGAGCGACACTCATATTAAC

GTACGTAGCGTTTCTCCATGTTTGGAAACTGGTTCCCTAAAAGCAGA

GCAAGCGCCTCAGAATGTCCAGAACCCACCACAGACACAGGACCAA

GCTACTGAAGATACTGCCACCAACTCTCAACCGGCCTCAACGCAGTT

CAGAGCTATCATTGTGGGAGGTGGCCCAAACGGTTTGTGTCTGGCAC

ATGCGTTATACCTAGCTGGTATTGATTATGTTTTGCTAGAGCGCGGTG

GTGAAATCATCAACCAATCTGGAGCCGGACTTGCTTTATGGCCTCATT

CAGTTCGTATATTAGATCAATTGGGTCTGCTAGACGAAGCTAGGAAA

CATTATTTCCCGATTAAGACAAAGCATAACCACCGCCCCGACGGGTC

AGTCAGAGATGTTAACGATATGTTTGCCAAGGTAGAAGTTAATCACG

GTCACCCCTGGATGCTGTTCCATCGAGTAACATTGCTAGAACTCTTGT

GGGAAAATTTGCCTGATAAAGAGACTAGAGTAAAAGTCAACAAGAA

GGTGGAAAGTGTAGTCTCCAACCATGATGGCGTCGTGGTTACATGTT

CAGATGGAACATCTGAAGCAGGAAGTATAGTGATAGGTTGCGATGGT

GTACACTCGACTGTTAGACAAGTCATGCATGACTTGAGAAGCGAGAA

AAAGAAGAAAAGGCCAGGCCGAAAGCTCTCTCTGGGAAGGTCGCCG

TCACAAGACAAGCCAATGAGAGCCCACTACTATGGGCTTATTGGTTG

GATTCCGCTTCTTGATGGGTTACAACCTGCTGCTTGCTACGAAGTGAG

AAGTGAGCCTAAAGGTAAAACCTTTACCATTTTAACGGGCGAAGATA

CCGCTTACTTCATCGTGTATATTCATTTAGAAAAACCCACGAGAGAG

167

CGCAGCAGGCACACAGACGAGGATGCCGAAAGGTTGGCAGCCGCAT

TGGCCGAGAACAAGATAACGAGGGAAATTACTTTTGGTGACCTATGG

AGATCAAGGCGTTGGGGCAAAATGCTTGATTTTCAAGAAGGGTTCGT

AGATAAATGGTATCATGAAAGAATCGTTTTAGTTGGCGATGCTGTCC

ATAAGATGACTCCAAATGCGGGACTAGGCTTGAATGCTGGCTGGCAA

GGTATCGCTGAATTAACGAACAGATTGCGGAGATTAGTTGTTGCAGA

AGGACAAAGACCAGATGCAAGGTGCGTTGAAAAAGTTTTTCGAGGTT

ACCAAGATAGTAGAAAGGGTATGGCAAAGAAAACTATGAGGTTTTCT

TCTCTGTACACCCGTGTGGTAGCTAATCAGAGCTTACTGTATCGTTTT

TGTGATAGAATGACACCAGCGGTTGGTGGGGATGTCGCATTATTAAA

TACAATGGCTAGTCCTATTGTTAAAAAAGGTGTCACTTTCGACTTTGT

AGCGGAACGTGATCATAAAGAAGGTAGAGTAAGATGGGTTCATCCA

CAACATGTTCCAGCTGAAgtatcgatggattacaaggatgacgacgataagatctgagctcttaatt

aacaattcttcgccagaggtttggtcaagtctccaatcaaggttgtcggct

pTEF1-

MNBE010

00128.1-

tADH1-in-

pSP-G1

(Entry 2

Table 4-1,

AL-2-79-

K-C5 or

AL-2-79-

L-C6)

tcaagtttcagtttcatttttcttgttctattacaactttttttacttcttgctcattagaaagaaagcatagcaatctaatctaa

gttttaattacaagcggccgcATGACCAAGTCCAAGTTATACGACGTGATAATCG

CGGGCGCCGGACCCGTCGGCCTCTTCCTCGCCCACGAACTCAGTCTC

CGCCAAATAGCAGTCCTAGTCCTAGAACGAACAACCCAACAAGACTC

GCCCTGGAAATCCGGACCTCTAGGCCTCCGAGGCCTAAACATCCAAT

CCATCGAAGCATTCTATCGCCGCGGTTTACTAGGCCAATTATTCAATC

TGGACGAAAGGCAAAAGTACCAACCGAGCAAGAAACCAGGTTTCCA

ATTCGGCGGCCACTTCGCCGGCATCATAATAAATGCCAATCAATTCG

ATTTGCAACGCTGGAAGTACCGCCTCTCAGGTCCGGCGCTTGGTCCG

GGTCCAACGACGATGGATCAAGTGGAGAAGGTCCTAACGGCGCGGG

TGGAGAGTCTAGGCGTGAAGATTCTCCGCGGTCGATCTGTTAGCCGT

ATCTCGCAGGATGAGGCAGGCGTTACAGTCGAAACAGACGATGACC

ATTCCGAGTCATTTCGGGGCAAATGGCTCGTTGGCTGTGACGGTGGG

CGCAGCGTTGTTCGAAAAGAAGCCGGCTTCGAATTCGTCGGTACAGA

TGCCCAATTCACCGGCTACGCAACGCATGTCGAGATAGAAGGTCGCG

AGAAGTTGAAGCCTGGTCTCAACGTTTCTGAAACGGGGATGTATATC

AACGTAGTGCAGAATGGCGCTTTTCATCTCCTTGAATTTGACGGGGC

GAACTTCGATCGCGCGGGTGATATCACGATCGATCACCTTCAGCGGG

TCCTTGATCGGGCTATTGGTCGGACAGATGTGAAGATTACAAATGTG

CATCTTGCTTCTTGTTATACCGATCGTTCAATGCAGGCGGCAAGTTAC

CGGAGGAACCGCGTTCTTCTTGCCGGTGATGCAGCGCATATTCATTCT

CCGCTCGGCGGGCAAGGACTGAATGTCGGACTAGGTGATGCGATGA

ATTTGGGGTGGAAGCTTGCGGCTATAGTTCAGCGGGACCAAGGCCAG

GGAGAGGATTCTGGGAATACGCATGATCTGACTCTTCTCGATACCTA

CGAGAGCGAGAGACACCCCATTGCTGCGTCGGTTCTTGATTGGACTC

GTGCGCAGGTCACGGCGCTGAGGCCGGATGCCTTTGGTCGGGCTACG

CGCGCCCTGACGGAAGATCTGATCAAGACGGATGATGGGGCTAATCT

GTTTATTGATCGGATCTGGGGACTTTCGCAGCGATATAAACTTGGCG

AAGAGTCGAGGCCTACACATCCGCTTCTTGGGTCTAGTGCGCCTGAT

TTCGAGTTTGGGAATGGGTCTAGGCTTGGCTCGAAGCTTGAGGAGGG

ACGGGGGTTACTTCTTGATTTTGGGAACAGTAGTGAACTGGAATCAG

TTGTTGATAGAAAATATTCTGGGAAGGTAGATTATCTTGCTCCAGAG

168

GCAAAGGAGAACTGTGGGCTGAGTGCCTTGTTGATTCGACCTGATGG

AATCGTTGCATGGGTGGTGGAAGAAGATGCGCAGCACGATATTGATG

CTTTGAAAGCTGCATTAGGGGATTGGTTCACTTTGgtatcgatggattacaaggat

gacgacgataagatctgagctcttaattaacaattcttcgccagaggtttggtcaagtctccaatcaaggttgtcggct

pTEF1-

NC_03677

2.1-

tADH1-in-

pSP-G1

(Entry 3

Table 4-1)

tcaagtttcagtttcatttttcttgttctattacaactttttttacttcttgctcattagaaagaaagcatagcaatctaatctaa

gttttaattacaagcggccgcATGACAACTAATCCAGAAGTCATCATCGTGGGCG

GCGGAGCAGTCGGCCTGGCCGCTGCATGCGAATTGGCCATCAAGAAT

GTCTCTGTTACGATCTTGGAGCGGGAATCTGGACCTGCAGCGCCATG

GAAGAACGAGAAGCTGGGCTTTCGCGAGCTGTTTGTGCCTGCCATCG

AGCACTTGTACCGCCGTGGCCTGCTGAACGACATGTTCCGCAATGAA

GAACGACCACAGACTATGCCGGAGAAGAAGGGTGGCTTTGAGTTTGC

TGGTCATTTTGCTGGTATGATGATCAATGGAAACGATGTCAACTATTC

ACACTGGGACGATACACACCTTCCCGGGCCTGCTTGTGTACCTGGAT

CTTCGACACTGGGTCACATTGAGAATGTTCTGCGTGCACGTGCTGAG

CAACTCAAAGTGCCTATTCTTACTGGCAAGAATGTGACTGGCATCGA

AGACACAGACAGCGGCGTCAAGGTGTTCTGCGGCGAGGAAACATTC

GAGGCTCAATATCTTGTCGGATGCGATGGTGGTCGGAGCACCATTCG

CAAGAAAGCTGGGATTCCATTCATCGGCACAGAGCCCGAGTTCACTG

GCTATGCTGTCCATTGCAAGCTCGACAACCCCTTTTTGCTCAAGCCCG

GCTTCAATCGATGCCCAAATGGCGGACTCTACATCATGGCCGGCCCA

CAGCACATCTATGCTGTAGATCACGACATCAGCTTTGACCGAGACCA

AGAAGTAACAGCGGAACACTTTCAAAAGGTTCTGAGAAAGCTTTCCG

GTACCAACGTGGTTGTTGAGCAGGTCGTTCTCGCATCCGCTTTCACCG

ATCGCTGCAGACAAGCTGAGCAGTACCGCAAAGGCCGAGTCTTCCTT

GCTGGCGATGCTGCCCACATCTACCCTCCATTCAGCGCCCAAGGACT

CAACTGCGGTTACATCGATGCAGTGAATCTCGGTTGGAAGCTCGCTG

CTTTGGTCAAGGGCTATGCTACTCCGGGCTTGATCGATACATACCAG

CAAGAGCAACACCCAATGGCTGCGCGAGTACTTGATTGGGTGCGAGC

ACAAATTGTTACCCTTCGACCTGACGCTCATGGCCGGGCTGTGGGCA

ATGTTATGCGCGAGTTCCTCTACACCGAAGCTGGTGTGACATACTGC

ATCGGTCGACAATGGGGTCTTGATGTGCGGTATGAGATCGGAGAGGG

ACATAAGCTTGTCGGTCGCAGCGCTCTGGATTACGAGCTGGACGATG

GCTCTCGACTGGGAACTAAGCTCGGCGCCGCGAACTTCACACTCGTG

GCATTTGGTGACTGCAGTTCAGAGCTCACCAGCTCCGTCAACGAGCT

TAACCCAGTGCTCGGATTCTGCAGAGCGAAGGGCGAGGAGAAGTCA

GGCCTCCAGGGCGTCCTTGTACGTCCTGACGGGATCGTGTCGTGGGC

ATCTGCAGATGCCCTCAACATCGAGACGATCAAGGCCAGCCTCGAAC

GCTGGGTCGCGCTGCCAGCTGCTGTTGAGAAATTTGAATCTGTGTCTT

ACGAGGCCAACCAGTTGTCCAACATTCAGACGCCATTGACGACTACT

GTTCGATCGAATATCGGACGTGTATTTGGCAAGGATGGAGAGAAGTA

CACTGGCCTTCCGACAGTGGTGACGGAAGTCCCGGCAACGGCTGATG

CAgtatcgatggattacaaggatgacgacgataagatctgagctcttaattaacaattcttcgccagaggtttggtc

aagtctccaatcaaggttgtcggct

pTEF1-

FJOG0100

0008.1-

tcaagtttcagtttcatttttcttgttctattacaactttttttacttcttgctcattagaaagaaagcatagcaatctaatctaa

gttttaattacaagcggccgcATGGGCGAGACCGATGTTCTGATAGTTGGTGGAG

GGCCCACTGGATTGATGCTCGCCCTCGAGCTTACCAGCCAAGACATC

169

tADH1-in-

pSP-G1

(Entry 4

Table 4-1,

AL-2-79-J-

C4)

CCATTCCGCATCATCGACTCAAGTCCTATTCGAAGCGACAAAAGCCG

GGCCTTGGTTCTCCATTCACGGACTCTGGAACTTCTCAATCGCCATGG

AATTGCTCAGGAGTTCGTTGACCGTGGAAATTTCAATTTGGCCGTGC

GCATCTTCGCCAACCAGAAGTTCGTGTTTGAGAACGATTTCAACATC

ATCGCTTTCAATGACACTATGTACCAGAACCCCTTGATTATCAGTCAG

GCAGAGATTGAATCGATCCTGGACGAAGTCTTGATGAAACATCAAGT

GAAGGTGGAACGGCCCGTGACAGCGGAGAAGATATCACAAGATGAG

ACTGGAGTTACTGCCGTACTACGCCACGAGGATGGAAGCGAAGAGTC

ACTCCGTTGCAAATATGCTGTAGGCTGTGACGGCCCTCGCAGCATTG

TACGAAATTCGGCAGGTCTACAATTCGAAGGAGCGGCGTACCCGCAG

GATTTCATCCTCGCAGATGTGCACATGAAATGGGAGACTAGAGAATG

CCTTCATATCTATCTTGGCGCCTCTGGGTTCATGATTGTCTTTCCGATG

AAGGACAATGTCTGGCGTCTGATCTGTTCTCGCAGGGAAGCACTGGG

CGCTGACGCTGAGCCAACTCTTGAGAATTTCCAGGAGATGTTGGACA

AGCTGCTTCCTGAACCTGTTCAAATATTTGATCCAGTTTGGATCAGTC

GATTTAAATTACACCATCGAATTGCAGACAATTACCGCGCTGGTCGA

ATGCTCGTCGCGGGCGATGCAGCCCATGTCCATTCTCCTGCTGGTGG

GCAAGGGATGAACACCGGTATGCACGATGCAGTCAATCTTGGCTGGA

AGCTTGCAAGTGTCATTCGGGGCGAGAACGATGACTCTCTGTTGGAC

TCGTACAATATCGAACGGCGTAGAGTTGGCCAGACTCTCCTGCAAGG

CACAGATAGACTTTTCGAGTTTATGGCAACAACAAATCCTTTGTATCT

CTTCATAAGGAACTACGTCATGCCTTTCATTATGCCCTGGGCCATGGC

GATACCTGGCCGTCGAGCTCTTGCATATAGATTTGTATCCGAGTTGGG

GATTCGATATCGCAAAAGTCCCATCGTTGGACAGGCAACGACTTGGA

AAGGAACACTGAAAGGTGGAGATCGAGTACCCGACGGGAGACTCCT

GAAGGGGAATATTGAGATCACTGTTCACTCGTTGCTTGGAGCACGCA

AGCACAGTCTACTTCTGTTCTCGGGCGTGGACGGGGTCACCAGCACA

GACGATCTGGAGAAAGCTCATGTGGAGTTCATCGAGGCCAGTGGTCG

GTCGATCCCAGTATACACAATCACGAAATCATCCTCGGAGAACATAG

AGATCATGGATCCGGAGGGGCTAGTGCATAGGCTGTTTGGCTTTACA

GCATCCGGCTTTGTGTTAGTTCGACCCGATGGTCACATTGCGTTCATC

GGGCCTCTCACGTCAATGGACGAGCTCAAGACGTGGATGGAGAGAgta

tcgatggattacaaggatgacgacgataagatctgagctcttaattaacaattcttcgccagaggtttggtcaagtctc

caatcaaggttgtcggct

pTEF1-

MCBR010

07648.1-

tADH1-in-

pSP-G1

(Entry 5

Table 4-1,

AL-2-72-

D-C5)

tcaagtttcagtttcatttttcttgttctattacaactttttttacttcttgctcattagaaagaaagcatagcaatctaatctaa

gttttaattacaagcggccgcATGGATTGTGAAGTATTAATTATCGGTGCTGGGCC

CACAGGACTTATGCTTGCCCTCGAACTTTCCCGCGAACAAATCCCCTT

CCGAATTCTCGATAGTCATTATTCTACTAAGACAGCCCATCAATCTCG

GTCTATAGTGGTTCACTCACGTTCCCTGGAGTTACTTGCCCGGCATGA

CTTAGCCGAAAGCTTCATCGCTAAAGGATTACTCATGGAAGAGATTC

GCTTCTTCTTGAAGCAAACCCTTAATGGGAAGAGTAGTTACCCTGGA

AGTCTCATGAATGATACAATTTATAAAAAACCACTTATCATTAGACA

ACCACTATCAGAGCAGCTTCTGGAAAAACGTCTTTGCGAGTATGGAG

GCATTATTGAAAGGGGGGCAAGCGCTAAAAAAATGGTGATAGATGA

AGATGGCGAGCGGGTGTCTGTTTTGGTACAGCATAGAATTCAGGAAG

AAGAAGAGAAAGTAAGAGAGGAATTAATCAATTGTAAGTATGTTGT

170

AGGTTGTGATGGAGCTAACAGTATGGTACGAAAATTTGCGGGTATCG

ACTTTCCGGGTACTTTATACCCTCAAGAGCTCATTCTCATTGATGCAC

AGGTAGACTGGAACAATAAAATATGTCCACATGTTTTTATTGATAAG

GAATTCGTGATCTTTATACCGCTAGACGAAACAGGTATCAACCGAAT

CATCTGCCGGCGTCCTCCAAATGACACCAGCACTATCTCATCCAGAT

CAAATCTGTTATCAGGATCATCGCCTTACCAAGAGCCTAGCCTATCC

GAGTTTCATGATACAGTTATTTCTTCCGTTCCTGGTACGACTCGTATT

CACAGCCCGATTTGGTCTTCCTCATTCCGTGTCTCACGTCGACTTGCT

AGTACATATAGTGTGGGGCGGATCTTCCTTGCTGGTGACGCCGCTCA

TGTCCATGCACCTATTGGCGGTCAAGGTATGAATACCGGTATTCAAG

ATGCCGTAAACCTGGGTTGGAAACTTGCGCGTGTTATAAAATCAACT

GCTCCGTCCTCTTTACTCAACTCATATAATGTAGAGCGATATAAAGTT

GGTAGTGACACTGTTGAAAATACAGATCGCATGTTCAATATTTTAAT

AACTACAAACCCTGTAAAAATCTGGTTACGAAACTTTCTTTTTCGCTG

GGTTTTGCCATATTTCCTCAAACCAGATTTTGCAGACAGAAAAATTCG

TTACATATCACAATTGTCAATTCGATATCGAAATAGTCCCGTTGTAGG

CACGGCTTCTGTGTGGAAAGGAATTCTAAGAGGCGGAGATCGGGCAC

CAGATGGATGGATGATGAATCCTGAAGGAGAAAATATCACTTTGCAC

AGGCTTTTTAGGACATCAAATGATCATCTTTTCTTATTCAGCGGCCTA

GACACTCTTGAATCGCGGATGGTCAATGATACTGTCCAGCAGCTAAA

ACTCTGCAGCTCAGTTCTTGTGGTACACAAAATATATGACGGGAATT

TTAAAGATAAAACAATTATCGATGGTTACATAGACCCAGGGGGTAAA

GTTCACACCTTGTATCAGTTTATTGAGCCTGGTTACGTTCTTGTTCGA

CCCGACGGATATATCTCATTTATAGGACCTATGACTTCACTAGATGA

ACTGAAGAATTGGATGAATTCATATATGCAGGGAgtatcgatggattacaaggat

gacgacgataagatctgagctcttaattaacaattcttcgccagaggtttggtcaagtctccaatcaaggttgtcggct

pTEF1-

KN714671.

1-tADH1-

in-pSP-G1

(Entry 6

Table 4-1,

AL-2-72-

B-C6)

tcaagtttcagtttcatttttcttgttctattacaactttttttacttcttgctcattagaaagaaagcatagcaatctaatctaa

gttttaattacaagcggccgcATGGAGGCTAAGAAATCTTTCAAAGTTATCATTGC

TGGTGGCAGTATCGTCGGCCTAACGCTGGCCAACGCGCTCGAGAAGG

CGGGTATTGATTTTCTTGTCCTCGAGAGGGGCGACATTGCTCCACAGC

TCGGCGCATCAGTCTCAATCTACTGCAATGCGTCCAGAGTCCTCGAC

CAGCTGGGAGTTTGGAAAGGCATTCACCAGAGCACAATCCCGCTGAC

TGATCGGCTATACTTCGATGAACATGGCCGCTTGTTTGAGGACAACA

AGATGATGAGGCTCATAGACGACAAGACTAAGCGGCCTATCACGTTC

TTGAGAAGGGAGGCATATATCAGGGCTCTTTACGAAAACCTGAAGGA

CAAGTCGAAGGTGAAGGGTCACTCTGGGCTGGTGTCCTTTGCCGAAG

ATGATGAAGGCGTTTCAGTATTCACCAGTACCGGCGAGGAAATCAGG

GGCAGCATTCTTGTGGGAGCAGACGGCATCCATAGTACCGTCCGGAA

GCTCATGGCTGAGTCTGTCGCCAAGTCGGACCCTCAGAGGGCTAAGA

ATCTCATCGAGGGCTTCACGGCAAACTACCGGACGATCTTCGGAACG

TCCAAAAACCAGCGAAAAGACGACCCGAGCGTGCAGATGGTGGCAG

ATAGTCTTTCCCATACCTCCTATTACCGTGGCGTCACGGGGGTCTGCG

CCGTCGGCAACAGTGGGGTGATCTACTGGTTCCTCTTGGTGAAGGAG

GACACGCCTTCGACAATGCCGAACTGCCCGCGCTACTCGGAGGCGGA

TGCCGAAGAGACGCTTCGTAAATTCGGCCACCTTCATATGGGACCGG

GATATACTTTCAATGACCTCTGGGAACTGAGGGAGAAAGGAGTCATG

171

GTACCGTTGCAGGAAGGCATCGTCGAAGGAAGCTGGAATAGTAGCG

GGAGGGTTGTACTCATGGGCGACGCCGTGCACAAGTTCACTATAAGC

GCGGGGCTGGGCGCCAATTTAGGCGTAGAAGGCGCTTGCCACCTGGT

GAATGAGCTCCTCCCTGTCCTCAAAGAGACGGAAAACCCCGGAAAGC

AAGAGATAAAGGATGTGCTCGACAGATACGAGGAGAAGCATCGTCC

TCGAACTAAGATCTGTGCCGTATTGTCTAACTATTTGACCAAGTACGA

GGCGATGGAGACCTGGTGGCTCAGACTTCTGAGGTTCATTATTCCTC

GGATCCCGGACAGTTACAAGGCGAAGTCATTTGTGGACTTCATGTAT

GGTGCTCCGATTCTCGATTTCCTGCCTCACCCGGGCAAGAGCTCTGCG

gtatcgatggattacaaggatgacgacgataagatctgagctcttaattaacaattcttcgccagaggtttggtcaagt

ctccaatcaaggttgtcggct

pTEF1-

KL584983.

1-tADH1-

in-pSP-G1

(Entry 7

Table 4-1)

tcaagtttcagtttcatttttcttgttctattacaactttttttacttcttgctcattagaaagaaagcatagcaatctaatctaa

gttttaattacaagcggccgcATGTCGGTCTTACAATTCAATCTGAACGGCTTCGT

GCCTGGCGACCCTCATGATCTCTACCGTGTCCACCAAGAAACCGCCG

AGATCGCAAACAATGTCCCGCAAGAAGTCGACGTCCTCATCATTGGC

TCTGGTCCAGCTGGTCTGGCTCTAGCAGCACAGCTATCTCTCTTTCCA

AGCATCACCACAAGAATCATTGAACAAAAGGCTGAACGACTAATCCT

AGGCCAAGCCGATGGCCTCCAAGTCCGAAGCATCGAAATGTTCGAAG

CATATGGCTTCAGCGAAAGAGTCCTCAAGGAAGCGTACTACATCAAC

GAAACCACCTTCTGGAAGCCTGACGAGCAGCAACCGGACCGCATCGT

GAGAAGTGGCAGGATTAAGGATACTGTAGACGATCTCTCCGAGTTCC

CGCACGTCATCCTCAATCAGGCGAGAGTTCATGACTTCTTCCTTGATG

TCATGCTCATGTCGCCGACAAGATTGCAACCGAGCTATTCACGCAAA

CTCAAGCAGCTCAGCCTTCAACCAGCACAGGGCAACTCCTCGCACGC

TACGTCCTCCGTGCAAGTTCAACTCGAGAGAACAGACTCCAGCCACG

AGGGCGAAGTCGAGGATGTACGTGCTCGCTACGTCGTAGGCTGTGAT

GGAGCCCGCAGTGCCGTCCGTCAAGCCCTCGGTCTCAAGCTGGAAGG

CGACTCCGCAAACCAAGCTTGGGGTGTCATGGACGTACTTGCTGTAA

CCGACTTCCCAGACTGGAGGACCAAGGTTGCAATCCACTCCGCGACA

CAAGGCAGCATCTTGATCATCCCCAGAGAAGGAGGTTACCTTGTGCG

CCTCTACATTGAACTCGACAAGCTCGACGCCAACGAACGTATCGCAA

ACAAGAAGATTACCGCCGACAAGCTCATCGAGACCGCACAGCGCAT

CTTCCATCCTTATGCTTTGGATGTCAAAGAAGTAGTCTGGCACTCTGT

CTACGAGATTGGCCAGCGCCTGTGTACCAGATTCGACAACCTCACTC

CGGAGACCGCTACAACAGAATCCCCAAACATCTTTATCGCAGGCGAC

GCATGCCACACCCACAGCCCTAAAGCAGGTCAAGGCATGAATGTCAG

CATGCAAGATGGCTACAATCTAGGCTGGAAGCTCGCCTCCGTGCTCA

ATGGAATCTCTAACCCGGCCATCCTCCACACCTACTCCGCCGAACGT

CAAAAAGTCGCTCAAGACCTCATCAACTTTGACCGAGAGATTGCTTC

AATGTTTAGCGCCAGACCCAAGAGCCATGCCAAAGACACAGAGGGA

GTCGACCCTGCTGAATTCCAAAAGTACTTTGTCAAGCAGGGCCTCTTC

ATGGCTGGACTGGGCACAGCTTACTCGACTTCTGCTATCACCGCTGG

ATCAGAGCATCAACACCTGGCCAAAGGTTTCCCCATCGGCATGAGAT

TGCACTCTTCTCCAGTCATAAGATTGGCAGATGCAAAGCCGATCCAA

CTAGGTCATGTGGTAAAAGCAGATGGAAGATGGAGAGTCTTCGTCTT

CGCATCAGCCGAGGATCCCGCTTCGACCTCGTCAAGCTTCCACTTGG

172

CCTGCGAATCCCTGACCGAGCTTGTCAACAAGGTCAACCCTACTGGT

GCTGATATCGACTCCGTCATCGATGTCAGAGGTATTCTGCAGCAGGG

CCATGCTTCACTGAACATCAACCACATGCCTGCTATCGTGTGGCCGC

AAAAGGGAAAGTATGGACTCAGAGATTACGAAAAGGTCTTCTGTGCT

GAAGATGTGGATGGACATAGGGATATCTTTAATGCTAGGGATATCGA

TCGCAGAGGCTGTATCGTGGTCGTACGACCTGACCAGTACGTCGCCA

ACGTTCTGCCGTTGTGCGATCATGATGGTTTGTCAAGGTTCTTCGGAG

GTTTCATGTTTGCAAGGGATgtatcgatggattacaaggatgacgacgataagatctgagctctt

aattaacaattcttcgccagaggtttggtcaagtctccaatcaaggttgtcggct

pTEF1-

KZ111246.

1-tADH1-

in-pSP-G1

(Entry 8

Table 4-1,

AL-2-86-

D-C5)

tcaagtttcagtttcatttttcttgttctattacaactttttttacttcttgctcattagaaagaaagcatagcaatctaatctaa

gttttaattacaagcggccgcATGACCAAGACTTACGATGTTATCGTTGCCGGCGC

AGGCCCCATCGGCTTGTTTCTCGCCTGTGAGCTGAAGTTGGCGAACG

TTGACGTGCTAGTACTAGAGCGTGACACCAACCCCGAATCCCCCTGG

AAAAGCGAACCACTCGGCATACGAGGCTTGAATACGGTATCTGTCGA

GTCCTTCTATCGACGTGGGATGCTAGACAAGGTCACCTCCATGGAGC

GACCGAACTTCTTCAAGAAAGGCCCGGGGTTCCAATTCGGAGGCCAC

TTCGCCGGCATGCTACTGAATGCCAACAAGCTCGAAATGTCCCGTTA

TAAGTACCGCCTCCAAGGCCCAGCATTGGTTCCCAACGGATCAAGTA

TGGAACGTGTTGAGAAGGCCCTAACAGAGCGCGCTGAGAGCATTGG

AGTAGATATTCTCAGGGGGAAGGGAGTCACGGAAGTCTCGCAGGAT

GACAACGGCGTTACGGTGGAAGCAGGAGGCGAGACTTTCCATGCGA

AATGGCTTGTCGGATGTGATGGCGGGCGGAGCACAGTTCGCAAGGCC

GCTGGCTTCGACTTCGTCGGAACAAATCCTGAATGTACCGGTTACGC

TTTTTTGGCCGATTTAGACCATCCCGAAAAGCTGAAGCCGGGGTTCC

AACCCTCTGTAGGCGGCATGTACATCATAGGACATTGGGGTAACATG

TACTTGCTAGACTTCGATGGTGGCGCTTTCGATCGTACGCAGGAGAT

CACACCCGAGCACCTTAACGAAGTCATGGCCCGTGTGACAGGCACCG

ACGTCAAGGTCACCAAGATTCATCAGGCATCCTCGTTTACCGACCGA

TCTAAACAAGCGTCGACTTATCGAAAAGGCCGCGTTCTCTTAGCCGG

CGACGCGGCGCACATCCATTCGCCTCTGGGAGCACAAGGATTGAATC

TAGGGTTGGGAGATGCCGTGAACCTTGGCTGGAAATTGGCAGCGACA

GTTCAGGCTTCTAAATCTGGAGAGGAACCAAAGGACCTCAGCCTACT

CGATTCCTACGAGAAAGAGCGATATCCGATCGGGTCTTGGGTTCTTG

AATGGACACGCGCTCAGATCTCGACGCTAAAGCCCGACATGTTCGGT

ATCGCTCTTCGTAGGCTCATGAGCGATTTATTGGAGACGACTGATGG

AACGAATTTATTCATCGATCGCATCTGGGGTCTGTCGCTGCGGTACG

ATCTTGGCGACGAACACCCCGCCGTCGGCTGTAGCGCTCCCGATTTC

GAATTCCACGACGGAACGAGGCTAGGTCCCAAGCTAGCGACGGGGC

GAGGTCTATTGCTCGATTTCGGATCTAATGCAGAACTCAAGGATCTT

GTCGGGAAATACGAGAGCAAGGTTGATTATCTTAGCACAGAGGCAA

AGGACCAGCTGGGCCTGAAATCCTTGTTGATCCGCCCCGATGGCGTT

GTGGCTTGGGTGGCTGAAGAGAAGCCTGATATGGACTCGGCGAAGG

CCGCTCTGGAGAGATGGTTTGGGCTGGGTTCGAAAgtatcgatggattacaagga

tgacgacgataagatctgagctcttaattaacaattcttcgccagaggtttggtcaagtctccaatcaaggttgtcggc

t

173

pTEF1-

FJOG0100

0031.1-

tADH1-in-

pSP-G1

(Entry 9

Table 4-1)

tcaagtttcagtttcatttttcttgttctattacaactttttttacttcttgctcattagaaagaaagcatagcaatctaatctaa

gttttaattacaagcggccgcATGGAAAACCAGCTCTACGACGTCATCATTGTTGG

CCGCGGTCCTGTTGGGCTATTTTTAGCATGCGAACTTCGCCTCACAGG

GCTCTCTGTCTTGGTTGTTGAGCGCCGCTCGAGCTCAGAGGGAGTGG

CGGAGACTCGCGCTTTTGTTGTCCATTGCCGCACACTCGAGATCCTCG

CATTCCGTGGTCTTCTTGGCAAGTTCCTCCAGGAGGGCACGAAGTCTC

CTTGGTGGCACTATGGCGTCTTGGACACCCGTTTGGATTTTAGCAGAT

TTGGCCACGAAACGAAGCAGAATTTTGTCCTGTTAAGCCCACAGTTC

AGAACTGAGGAGATTTTTCTGGAGCGTGCGAAGGAATTAGGCGTCGA

CATTGTTCTGGGAATTCGCGTCGAGTCCGTGGAGCCTTCGACTACGTC

TGTTACGATATATGGAACATCAAGCAGTCCAGATGGCGGCGCTAGAA

GTTCGTTCAAGGCGAGAGGAAAGTATCTTGTCGGTGCGGATGGAGTG

CGCAGTACCATCCGGAAGGCTACGGGCTTTGAATTCCCAGGTACGCC

GGCAACACACACGGGTTTGACCGGCGAGGCAACATTGGGAGTGCCC

ATGCCCTATCCCTACATTGCCAAAAACGAAGCAGGGCTTGTGATCGC

CGTACAGCTGAATATTCCCAGCGGCCGGGCTCGCCTCAATGTTTTCAC

CCCTGCTCGAGCTAATGTACCCGATTCGACCCCGGTAACGCTGGAAG

ATTTCAACGAAGCATTACAAGAAGTCACTGGTGTTGACTACAAGCTC

TCCAACCCTTGCATGCTTGGACGCTTCAACAACGAAGCCCGCTGCGT

CGATACTTACCGGAAGGGACGCATCTTTCTCGCCGGTGATGCAGGCC

ACCAGCACCTTCCAGCTGGCGGCCAAGGGCTCAACGTCGGTATTCAG

GAAGCAACTAACCTTGGATGGAAACTTGGGGCCGTGATTCGCGGCTA

TGCTCCGGACTCGCTATTGGATACTTATGAGACCGAAAGACTTCCAA

TCGCACAAGGCGTCGTGCAGAGCACCACTGCTCAGTCTGTTCTCTTTT

TTGCGCACAGCGGGCCGGAGTTGGCAATACGAAGTGTGGTGAACAC

GTTGCTCAAAATACCAACAGCAAACCACGACATTGCTGTAGGAGTTA

GTGGTTTCGGCGTCTCATACCCAAAGCTTCTTGACATGATACTTCCAG

CTGGATGGGAGGCACTTCCAGAGGCGATCGCCGGGAAGCGAGCCCT

AGATGTGAAGTTACGCGTCGGGAATGGCGAGGAAAAGCAGCTAAGC

GACTATATGAGGGGAGGGAAGTGGGTTCAACTTCGGTTCCTTGACAA

ACTCGCAGGAAGGCCGCCACTGCCGGCGTTCGAGGGGGCGACAGAG

GTTGTGGATGTCGCTGAAGTTCTGGAGGGGAAAGGGAGCATGTACCG

GGGCGCCTTGAGCGAACTGCTAATTCGCCCGGACGGTTATCTAGGGT

TTGGAAAGCGCgtatcgatggattacaaggatgacgacgataagatctgagctcttaattaacaattcttcg

ccagaggtttggtcaagtctccaatcaaggttgtcggct

pTEF1-

AMCJ0100

0140.1-

tADH1-in-

pSP-G1

(Entry 10

Table 4-1)

tcaagtttcagtttcatttttcttgttctattacaactttttttacttcttgctcattagaaagaaagcatagcaatctaatctaa

gttttaattacaagcggccgcATGACCTACACTACTCATGACGTGGTGATTGTGGG

AGCTGGTCCAGTTGGACTCTTCCTCGCTTGCGAACTACGCCTTGCCGG

TCTCTCCGTTCTGGTGGTCGAGAAACGAACAAATTCGGACGGCATGG

CGGAAACGCGTGCCTTTGTGATGCATGGTCGATCTTTGGAAATCTTCG

CCTCTCGCGGTCTGCTCGACTCTTTCGTTGAAGCGGGTCAAAAGACC

GACTGGTGGCATTACGGTGTTCTCGACACTCGTCTTGATTACAGTGTT

TTCGGCCGTGAAACGGACCAGAACTACGTGTTACTCGTGCCTCAGTA

CAAAACCGAGATGATCTTATTTCAACGCGCCGTGGATCTGGGTGCAG

TGATTATCAAGGGTGTCCAGGTCGATTCAATACACGAATCCGGGCCT

TATGTCGTTGCGAGGGGTTTCTACTGCAATAACAAACCTTTCATTGCC

174

AGCGGGAAATACCTCGTCGGTGCGGACGGTGTTCGCAGCACGATCCG

CAAAATCGCCAACATCGAGTTCACTGGCAATCCACCTGTCAATACAG

TCATGAGTGGCGAGGCTACACTAGGCACGGCCATGCCGAATCCCTAC

ATTGTTCACAACGAGCACGGCCTAGTTATCGCCGCTGACCTGAGGGT

CCCCAGCGGAAGAACCCGCCTAAATGTGTTCGCGAGTGATCGCGGCA

CGGTTCCTGAGTCAGTTGAAGTGACTCTTGAGGAGATGAATCAGAGC

TTGCAGAAAATAACCGGCGTTGACTACAAGCTTTCTAACCCTTGCAT

GCTAAAGCGTTTCAGCAACGAACAACGGCTAGCGACAACGTATCGCC

AAAATCGGATTTTCATCGTCGGAGATGCATGCCATAAACACCTCCCA

GCCGGCGGCCAGGGCTTAAATGTCGGTCTCCAAGAAGCTCTGAATTT

AGGATGGAAACTTGCCGCAGTCATCTCCAAGTCCGCCCCTGCTTCGTT

ACTCGATACGTACGAAGAACGATGGCCAGTTGCCAAAGCTGTTGTAC

AAAACACAACTTCACAGTCACTCTTATTTTTTGCCTCGTCTGGCCCAG

AATGGGCGGTTAGGGAAGCGATCGACAAGCTTCTGCGTGTACCAGAG

GCCAACAAGCGTTTGGCTAGGGAGATCAGCGGATTTTCTGTTGCCTA

CCCCAAATCGCTGGATATGATACTTCCAGACGGATGGCGGGCCCTAC

CAGAGAACATCCAGGGAAAGCGTGCATTGAATGTAAAGATGAGATT

ACCAGATGGGATGGTAACCGAACTTCGTGATTACACCCAAGACGGAC

GATGGATCCAGCTGCACCTTCCTGGAAAGCATATCATTGAACTCCGG

CCTCCTCCGGCATTTGGTAACTGGACGACGGTAGTGGAAGTTGTCGA

CATGCCAGATGAAGAAGAGAAGACGAGCTTGTATATGTGCGGAGTG

AGAGAGATGCTGATCCGCCCGGATGGCTATTTGGCCTTTGGTCGAAT

GGACGACgtatcgatggattacaaggatgacgacgataagatctgagctcttaattaacaattcttcgccaga

ggtttggtcaagtctccaatcaaggttgtcggct

pTEF1-

KB725976.

1-tADH1-

in-pSP-G1

(Entry 11

Table 4-1,

AL-2-86-I-

C5)

tcaagtttcagtttcatttttcttgttctattacaactttttttacttcttgctcattagaaagaaagcatagcaatctaatctaa

gttttaattacaagcggccgcATGACTAATCAATATGATGTTGTAATTATTGGAGC

TGGGCCAGTTGGATTATTATTGGCTTGTGAATTAGCTTTGGGAAAAA

CAAGCGTCATAGTTCTGGAAAGAGAAGCGTCGCCCGTATCTCCGTGG

AAAGAAGAACCGACTGGTATGAGAGGTTTGCACCTGCCTTCTAGTGA

AATTTTGTGGCGTAGGGGTTTGTTAGAAAAAATCTGGACATTGGAGG

GTAGACCACATGGCCCGACTAAGACTCCTGGATTTCAATTTGCAGGC

CATTTCGCCGGGATACCTCTTAACCTATCTCAGCTAGACTTAGATAGG

TGGAAATATCGATTGAGGGGTCCATCATTAATGGGGGGTCCAATAAC

AATTGCATTAATTGAGAGAGTGTTAGCAGAACGCGCTGAATCATTGG

GAGTCGAAATTCATCGGGGTTGCGGGTTCGAGAGGATAGTCCAACAA

TCCGCTGACGGCGTAACAATAGAAGCGGGTGAAGAAACCCAGACAT

TTCATGCAAAATGGCTTATTGGTTGCGATGGCGGTCGTTCCCAAGTCC

GTAAGGCTGCCGAAATCGACTTTCCTGGCACTGACGCGACCTTAACT

GGCTACGTGATCCATTGTGATCTTGACCATCCTGAACGATTGTCACCT

GGTTTCCAACCTACCAAACAGGGAATGTATATTTTTAGAAAGCCCGG

CGCTCTCTACCTGATGGATTTCGACGGTGGTGCCGGCCAAACAAGAG

AACCAAGCCAGCAGAGACTACAAGAAGTGATGGAGAGGGTTGTAGG

GAGAACTGATATCAAGCTAGAGAAGGTTCACCTAGCAAGTGCGTTTA

CGGACAGGGGCAAACAAGCCACAACGTATAGGAAGGGAAGAGTCCT

CTTGGCTGGTGACGCAGCGCACATCCACCCACCACTTGGTGCACAAG

GTATGAACGCCGGTTTGGGTGATGCCATGAATCTAGGATGGAAGTTA

175

GCTGCAACCGTTCGTGGAGAAGAGAAAGGAGTCCGGGCTTTCACCGT

GTTGGATACCTATACTTCAGAGAGACATCCCGTTGGTCAATGGGTCTT

AGAATGGAATTACGCACAAGTTGCTGCTCTAAAACCCGATGTAACAG

GTTACGCCGTACAAAAATTAATGAGGGATTTGATTGCAACTGATGAC

GGTACGAATTACTTCATAGATAATGTGTGGGGATTGTCTCAGCGCTA

TGGTGACGGTGAAGGTGTTCATCCAGTAGTTGGAAGATCGGCCCCTG

ATTTCACATTTAAAGATGGTAGTAGACTTGGGCCAAAATTAGAGGGT

GGCAGAGGACTTTTTATAGATTTTGAAGATGTGGATCTGGAGAAAGC

AGTTAAGCGATTCGAAAGAGTTGATTATCTCGGACAAAACGTGGAAG

ATAGACGTGGAATCAGAGCTTTACTTATTAGACCAGATGGATTTGTT

GCTTGGGCTGTAGAAGAGGGAGATGAACCAGACGCAGAGGGTTTAG

AAACTGAATTAAAAAAGTGGTTTTCTTACgtatcgatggattacaaggatgacgacgat

aagatctgagctcttaattaacaattcttcgccagaggtttggtcaagtctccaatcaaggttgtcggct

pTEF1-

AP007159.

1-tADH1-

in-pSP-G1

(Entry 12

Table 4-1,

AL-2-86-

A-C1)

tcaagtttcagtttcatttttcttgttctattacaactttttttacttcttgctcattagaaagaaagcatagcaatctaatctaa

gttttaattacaagcggccgcATGGGCAAGACCTGGGACGTTATTATTGCTGGCG

CTGGGCCGGTCGGTCTTTTCCTCGCCTGTGAACTGGCCATAGCTGGCG

TGTCGGTACTTGTACTGGAGCGAGAGATGCAGCCAGAGTCGCCTTGG

AAAGAGGGATTGTTTGGGCGTCGCGGTCTCTATACCCCGGCGGTCGA

GGCATTTTACCGCCGTGGCATCTTGAAGAGAATTTTCGGTGATGATG

AGCGTCCGACGCATTTGAAGAAGACTGAAGGCTTTCAATATGGAGGA

CACTTTGCGGGCTTGGTGCTTAATGCAAACAACATCGAGTTTTCACGC

TGGCCATATCGCTTGCCAGGACCGTCCTTTCTTCCTGGTCCGACTTCT

CTTGGACGTCTTGAGGCTGTGTTGTCGGAACGTGCAGAGACGCTGGG

TGTCCAGATCCTTAGAGGTATGGAAGTCTCCCGGCTCGCTGATGAAG

GTGAATCAGTCAAGGTATGGGCGGGCGACCAGTGGTTTACGGCTCAA

TGGCTTGTCGGATGTGACGGTGGACGCAGCACAGTTCGGAAAACTGC

TGGATTCGAGTTTGTCGGGACCGAAGCAGAATTCACTGGTTATATTG

CGGTATGTGATCTCGACCGACCAGACTTATTGAAGTCAGGTTTCAAG

CACACAAACTCGGGGATGTACATAGTTAGCGGACCTGGACAGTTGCA

TGTTATCGATTTTGATAGGAGCTTTGATCGGTCACAAACTATCACGCG

GGAACACTTCCAGGAAGTGCTACGGCGAGTGTCGGGCACAAACATC

GTTGTCGAAGCGCTTCACCTAGCCTCCTCCTTCACCGATCGCTCGAAG

CAAGCAACAGAATATCGTAGAGGACGCATCCTTCTGGCTGGTGACAG

TGCCCACACACACTCTCCACTGGGCGCGCAAGGATTGAGTACAGGAA

TTGGCGATGCGATGAATCTCGGCTGGAAGCTTGCGGCTACCGTCAAG

GGGTTTGCATCTCCTGGTCTGCTAGACACCTACCACCAAGAGCGGCA

TCCAGAGGCAGCTCGAGTGCTTGAGTGGACTCGCGCTCAGGTGGCGG

CCTTACGCCCTGATCCATATGGCCAGGCTATTGCGAGTCTCATGCGG

GACATGATCAATACCCAAGATGGGGCGACTTATCTCGCCGATCGCAT

CTGGGGACTTTCAGTGCGCTATGGCCCGGGCGACGCGCATCCACTTG

TCGGTTCTAGCGCTCCGGATTTTGAATTCGATGATGGAATGCGACTTG

GAGCTAAGTTAGAGACAGGATCCTTCTTGGTGATTGACTTTGGGAGC

AACAACCAGGTGGCAGAGCATGTGCAGTCCTTGCAGTCTCTGCAGTT

CATGATTCAATACTGTGCATGTAGCGCAAAGGAGGAATTCGGGCTCA

AGGGGTTACTCTTGCGACCTGATGGTGTGGTCGCATGGGTGTCGACG

GAGGAGATAAATATAATACGACTGCATGTTGCATTGTCTCGTTGGAT

176

AAGTCTCCCTAGTTTCGAGGCTgtatcgatggattacaaggatgacgacgataagatctgagct

cttaattaacaattcttcgccagaggtttggtcaagtctccaatcaaggttgtcggct

177

Table 4-11 Primers used for OxyS saturation mutagenesis in this study

To create a saturation mutagenesis of the positions outlined in Table 4-2 the even numbered

primers were used for PCR amplification with EH414 and the odd numbered primers were used

for PCR amplification with EH745. The resulting amplicons were gel purified and used for Gibson

Assembly with pSP-G1 (empty) digested with SpeI and NotI.

EH414 GCTCATTAGAAAGAAAGCATAGC

EH745 CTGGCGAAGAATTGTTAATTAAGAGC

EH723 AGTGCGAGCGTGGACTCCTAGMNNTTTCGAGAAGTCAACAGGC

TC

A43X

EH724 CTAGGAGTCCACGCTCGC A43X

EH725 CAATGCATATGGGTGTCTAGTATCAA F96X

EH726 CATCATTTGATACTAGACACCCATATGCATTGNNKGTTCCACAA

GTACGAACTGAAGAAC

F96X

EH727 ATGTGGGTCTTGACCTGGAAAATC M176X

EH728 AACTATTGGGCATAGATTTTCCAGGTCAAGACCCACATNNKTTT

GCTGTCATCGCAGACG

M176X

EH729 TGCACGAAGGTCATGTCTCATG W211X

EH730 TCATGAGACATGACCTTCGTGCANNKTTCGCAGCATTTCCGCTA

G

W211X

EH731 CCATGCACGAAGGTCATGTCTC F212X

EH732 GAGACATGACCTTCGTGCATGGNNKGCAGCATTTCCGCTAGAA

CC

F212X

EH733 CAGCATACGGTCTATCGAAAAAGGCGACMNNTGCTCTGTAGAC

GTCTGGTTC

T225X

EH734 GTCGCCTTTTTCGATAGACCG T225X

EH735 CGGCGCCCTCCTGTCA V240X

EH736 GTATGCTGACAGGAGGGCGCCGNNKACGGAGGAGGATGTCAG

AGC

V240X

EH737 CTAAGTTCAGTCCCTGGCCACCAGCMNNCAAATGAATATGGCA

TGCATCACC

P295X

EH738 GCTGGTGGCCAGGGACTG P295X

EH739 ATCTTGAAATCCTAAGTTCAGTCCCTGGCCACCMNNGGGCAAA

TGAATATGGCATGCATC

A296X

EH740 GGTGGCCAGGGACTGAACTTAG A296X

EH741 TCCTAAGTTCAGTCCCTGGCCMNNAGCGGGCAAATGAATATGG

CA

G297X

EH742 GGCCAGGGACTGAACTTAGGA G297X

EH743 TTAACGGCATCTTGAAATCCTAAGTTCAGTCCCTGMNNACCAG

CGGGCAAATGAATATGG

G298X

EH744 CAGGGACTGAACTTAGGATTTCAAG G298X

EH812 GTGCGAGCGTGGACTCCTAGAGCMNNCGAGAAGTCAACAGGC

TCG

K42X

EH813 GCTCTAGGAGTCCACGCTCGCACTG K42X

178

EH814 CAACAGTGCGAGCGTGGACMNNTAGAGCTTTCGAGAAGTCAA

CAG

G45X

EH815 GTCCACGCTCGCACTGTTG G45X

EH816 TGGGTGTCTAGTATCAAATGATGAG L95X

EH817 TCTCATCATTTGATACTAGACACCCATATGCANNKTTTGTTCCA

CAAGTACGAACTGAAG

L95X

EH818 GGCGACTGTTGCTCTGTAGA F228X

EH819 TCTACAGAGCAACAGTCGCCNNKTTCGATAGACCGTATGCTGA

CA

F228X

EH820 CATTAATTCCCTTAAACCCTCGTAGCGMNNATCTGGATCAATT

AGAACAGCTTGAG

P357X

EH821 CGCTACGAGGGTTTAAGGGA P357X

EH822 AGGATCTGGATCAATTAGAACAGCT R358X

EH823 CTCAAGCTGTTCTAATTGATCCAGATCCTNNKTACGAGGGTTTA

AGGGAATTAATGATTG

R358X

EH824 TAGCCCCGCTAAATATCTATTAGTCTCAGGMNNGTGCAACAAT

TCAATCATTAATTCCCT

V372X

EH825 CCTGAGACTAATAGATATTTAGCGG V372X

179

4.8 References

1. Li YR, Chooi YH, Sheng YW, Valentine JS, Tang Y. Comparative Characterization of

Fungal Anthracenone and Naphthacenedione Biosynthetic Pathways Reveals an alpha-

Hydroxylation-Dependent Claisen-like Cyclization Catalyzed by a Dimanganese Thioesterase. J

Am Chem Soc. 2011;133(39):15773-15785. doi: 10.1021/ja206906d.

2. Chooi YH, Cacho R, Tang Y. Identification of the viridicatumtoxin and griseofulvin gene

clusters from Penicillium aethiopicum. Chem Biol. 2010;17(5):483-494. doi:

10.1016/j.chembiol.2010.03.015.

3. Nicolaou KC, Hale CR, Nilewski C, Ioannidou HA, ElMarrouni A, Nilewski LG, Beabout

K, Wang TT, Shamoo Y. Total synthesis of viridicatumtoxin B and analogues thereof: strategy

evolution, structural revision, and biological evaluation. J Am Chem Soc. 2014;136(34):12137-

12160. doi: 10.1021/ja506472u.

4. Lindqvist Y, Koskiniemi H, Jansson A, Sandalova T, Schnell R, Liu Z, Mäntsälä P, Niemi

J, Schneider G. Structural Basis for Substrate Recognition and Specificity in Aklavinone-11-

Hydroxylase from Rhodomycin Biosynthesis. J Mol Biol. 2009;393(4):966-977. doi:

10.1016/j.jmb.2009.09.003.

5. Wang P, Bashiri G, Gao X, Sawaya MR, Tang Y. Uncovering the Enzymes that Catalyze

the Final Steps in Oxytetracycline Biosynthesis. J Am Chem Soc. 2013;135(19):7138-7141. doi:

10.1021/ja403516u.

6. Patrikainen P, Kallio P, Fan K, Klika Karel D, Shaaban Khaled A, Mäntsälä P, Rohr J,

Yang K, Niemi J, Metsä-Ketelä M. Tailoring Enzymes Involved in the Biosynthesis of

Angucyclines Contain Latent Context-Dependent Catalytic Activities. Chem Biol.

2012;19(5):647-655. doi: 10.1016/j.chembiol.2012.04.010.

7. Kharel MK, Pahari P, Shepherd MD, Tibrewal N, Nybo SE, Shaaban KA, Rohr J.

Angucyclines: Biosynthesis, mode-of-action, new natural products, and synthesis. Nat Prod Rep.

2012;29(2):264-325. doi: 10.1039/c1np00068c.

8. Kim J, Kim KH. Effects of minimal media vs. complex media on the metabolite profiles

of Escherichia coli and Saccharomyces cerevisiae. Process Biochem. 2017;57:64-71. doi:

10.1016/j.procbio.2017.04.003.

180

9. Koskiniemi H, Metsä-Ketelä M, Dobritzsch D, Kallio P, Korhonen H, Mäntsälä P,

Schneider G, Niemi J. Crystal Structures of Two Aromatic Hydroxylases Involved in the Early

Tailoring Steps of Angucycline Biosynthesis. J Mol Biol. 2007;372(3):633-648. doi:

10.1016/j.jmb.2007.06.087.

10. Kallio P, Liu Z, Mäntsälä P, Niemi J, Metsä-Ketelä M. Sequential Action of Two

Flavoenzymes, PgaE and PgaM, in Angucycline Biosynthesis: Chemoenzymatic Synthesis of

Gaudimycin C. Chem Biol. 2008;15(2):157-166. doi: 10.1016/j.chembiol.2007.12.011.

11. Kallio P, Patrikainen P, Belogurov GA, Mäntsälä P, Yang K, Niemi J, Metsä-Ketelä M.

Tracing the Evolution of Angucyclinone Monooxygenases: Structural Determinants for C-12b

Hydroxylation and Substrate Inhibition in PgaE. Biochemistry. 2013;52(26):4507-4516. doi:

10.1021/bi400381s.

12. Rogers JK, Taylor ND, Church GM. Biosensor-based engineering of biosynthetic

pathways. Curr Opin Biotechnol. 2016;42:84-91. doi: 10.1016/j.copbio.2016.03.005.

13. Ma SM, Li JWH, Choi JW, Zhou H, Lee KKM, Moorthie VA, Xie X, Kealey JT, Da Silva

NA, Vederas JC, Tang Y. Complete Reconstitution of a Highly Reducing Iterative Polyketide

Synthase. Science. 2009;326(5952):589-592. doi: 10.1126/science.1175602.

14. Agmon N, Mitchell LA, Cai Y, Ikushima S, Chuang J, Zheng A, Choi W-J, Martin JA,

Caravelli K, Stracquadanio G, Boeke JD. Yeast Golden Gate (yGG) for the Efficient Assembly of

S. cerevisiae Transcription Units. ACS Synthetic Biology. 2015;4(7):853-859. doi:

10.1021/sb500372z.

181

5 A yeast three hybrid assay for metabolic engineering

of tetracycline derivatives

*The contents of this chapter were published in:

E. Herbst, P. A. Baldera-Aguayo, H. Lee and V. W. Cornish “A Yeast Three Hybrid Assay for

Metabolic Engineering of Tetracycline Derivatives” Biochemistry, 2018, 57, 4726-4734.

https://pubs.acs.org/doi/abs/10.1021/acs.biochem.8b00419

182

5.1 Chapter outline

Metabolic engineering stands to transform the discovery and production of a wide range of

chemicals, but metabolic engineering currently demands considerable resource investments that

restrict commercial application. To facilitate the applicability of metabolic engineering, general

high-throughput and readily-implemented technologies are needed to assay vast libraries of strains

producing desirable chemicals. Towards this end we describe here the development of a yeast three

hybrid (Y3H) assay as a general, high-throughput, versatile and readily-implemented approach for

the detection of target molecule biosynthesis. Our system detects target molecule biosynthesis

through a change in reporter gene transcription that results from the binding of the target molecule

to a modular protein receptor. We demonstrate the use of the Y3H assay for detecting the

biosynthesis of tetracyclines, a major class of antibiotics, based on the interaction between

tetracyclines and the tetracycline repressor protein (TetR). Various tetracycline derivatives can be

detected using our assay, whose versatility enables its use both as a screen and a selection to match

the needs and instrumentation of a wide range of end-users. We demonstrate the applicability of

the Y3H assay to metabolic engineering by differentiating between producer and non-producer

strains of the natural product anhydrotetracycline TAN-1612. The Y3H assay is superior to state-

of-the-art HPLC-MS methods in throughput and limit of detection of tetracycline derivatives.

Finally, our establishment of the Y3H assay for detecting the biosynthesis of a tetracycline

supports the generality of the Y3H assay for detecting the biosynthesis of many other target

molecules.

183

5.2 Introduction

Metabolic engineering is a potentially greener and more economical approach than traditional

organic synthesis for the production of a wide range of organic chemicals. As opposed to organic

synthesis, metabolic engineering is capable of producing valuable chemicals from readily available

and renewable carbon sources and significantly reducing the use of organic solvents and

reagents.(1, 2) Traditional methods in DNA mutagenesis and increasingly, advances in DNA

synthesis, DNA assembly and genome engineering are enabling high throughput strain

construction in metabolic engineering.(3-8) High-throughput strain construction is necessary

because not only the multi-gene biosynthetic pathway for the natural product, but also the

underlying metabolism of the host must be optimized.(9) Assuming a biosynthetic pathway of 5

genes and 10 interacting host genes, to test just 5 variants for each of these genes results already

in a library size exceeding 1010. However, the throughput of the most highly employed assaying

methods such as LC-MS is only ~102-103 samples per day and is thus heavily limiting in assaying

strains for successful metabolic engineering.(5, 9)

The currently employed methods for assaying metabolite production in metabolic engineering are

either low-throughput and general or high-throughput but not general. LC-MS methods are general

but low-throughput, enabling the screening of only about 102-103 samples per day.(9) Colorimetric,

fluorometric and growth assays for molecules that are colored, fluorescent, essential for producer

strain growth or could be easily converted to one of these, are high-throughput but not general.(10)

Attempts have been made to develop assays that are both high-throughput and general, but it is yet

to be determined whether or not these assays could be widely implemented.(10, 11) Versatile,

general, readily implemented high-throughput assays are required to transform metabolic

184

engineering from a field with a high potential to a field with an extensive real-world impact on the

way chemicals are made.

We set out to adapt the Y3H assay, previously employed for various basic science applications by

our laboratory and others,(12-18) to a general, readily implemented, high-throughput versatile

assay for metabolic engineering. We wished to develop an assay that can be readily adapted to

new target molecules, used as either a screen or a selection, and minimizes handling, to match

varying needs and instrumentation of end-users.

Here, we demonstrate the application of the Y3H assay to detect the biosynthesis of tetracyclines,

one of the major classes of antibiotics. We describe the engineering of the Y3H system to detect

tetracyclines by synthesizing a minocycline-methotrexate chemical inducer of dimerization (CID)

and cloning a LexA-TetR (tetracycline repressor) fusion protein.(14) Reporter gene expression

responds to tetracycline in our system by competitive binding of tetracycline and the CID to the

LexA-TetR protein fusion (Figure 5-1). Our assay can report the presence of tetracyclines using

various reporter gene outputs, enabling the use of the assay both as a screen and a selection to

match the needs and instrumentation of a variety of end-users. We show the applicability of our

assay to metabolic engineering through the differentiation between producer and non-producer

strains of the natural product anhydrotetracycline, TAN-1612.

Figure 5-1 Detection of a target molecule biosynthesis by the yeast three hybrid system.

185

Low/no production of a target molecule is detected by high reporter gene expression (left). High

production titers of the target molecule outcompete the chemical inducer of dimerization (CID)

from the fusion protein receptor and are detected by lower gene expression (right). DBD = DNA

binding domain; AD = activation domain; PR = protein receptor (e.g., TetR); DHFR =

dihydrofolate reductase.

5.3 Materials and methods

General methods. Absorption and fluorescence spectra were recorded on Infinite-M200

fluorescent spectrometer. DNA sequences were purchased from IDT. Polymerases, restriction

enzymes and Gibson Assembly mix were purchased from New England Biolabs. Sanger

sequencing was performed by Genewiz. Yeast strains were grown at 30 °C and shaker settings

were 200 rpm, unless otherwise indicated. Yeast transformations were done using the lithium

acetate method.(19) Plasmids were cloned and amplified using Gibson Assembly and cloning

strain C3040 (New England Biolabs).(20) Unless otherwise indicated, yeast strains were grown on

synthetic minimal media lacking histidine and/or uracil and/or tryptophan and/or leucine, as

indicated by the abbreviation HUTL-.(21) Yeast strain patches were obtained from glycerol stocks

by streaking on an agar plate of synthetic medium lacking the appropriate amino acid markers,

incubating at 30 °C for 3 days, patching single colonies onto a fresh agar plate and incubating at

30 °C overnight. Unless otherwise indicated, when 96 well plates were placed in the yeast shaker,

95% of the interface between the plate and its cover was wrapped with laboratory film (Parafilm,

Bemis PM999). All Y3H assays were performed with sterile components and three biological

replicates. Protein homology was calculated by BLAST (https://blast.ncbi.nlm.nih.gov) using the

standard sequence alignment parameters. DataExpress was used to analyze mass spectrometry

186

peaks. Protein visualization was performed using The PyMOL Molecular Graphics System,

Version 2.3 Schrödinger, LLC.

Codon optimization by COOL (http://cool.syncti.org/index.php) was used for all hydroxylases and

reductases unless noted explicitly that JCAT or no optimization was used (http://www.jcat.de/).

Optimization parameters chosen were individual codon use, codon context GC content of 39.3%

and S. cerevisiae organism. The following restriction sites were generally excluded: GAGACC,

GGTCTC, GGATCC, GAGCTC, CTCGAG, GAAGAC, GTCTTC, CGTCTC, GAGACG,

GAATTC, TTAATTAA, TCTAGA, ACTAGT, CCCGGG, CTGCAG, AAGCTT, GTCGAC,

ACGCGT, GGTACC, GCGGCCGC, AGATCT, GGCCGGCC, CCGCGG, GCTAGC,

CCATGG.

All chemical reactions were carried out under Argon atmosphere with anhydrous solvents unless

otherwise indicated. Analytical high-pressure liquid chromatography (HPLC) was performed with

a C-18 column, 2.6 μ, 250x4.6 mm, eluent given in parentheses. Unless stated otherwise, reactions

were monitored using analytical HPLC with a separation gradient of 10% to 90% MeCN in 0.1%

(v/v) TFA aqueous solution over 20 minutes. Preparative HPLC was carried out with a C-18 5 μ

column, 250x10 mm, eluent given in parentheses. NMR spectra were obtained using Bruker 400

MHz or 500 MHz instruments, as indicated. Unless stated otherwise, mass spectroscopy

measurements were performed on Advion CMS mass spectrometer equipped with atmospheric

pressure chemical ionization (APCI) and electrospray ionization (ESI) sources. TAN-1612

quantification was performed using a phenyl-hexyl 1.7 μ 100x3 mm column, on a Waters SQD2

quadrupole mass spectrometer equipped with a UPC2 SFC inlet, a photodiode array (PDA) UV-

Vis detector, and a dual ESI/APCI probe. HRMS spectra were taken on a Waters ACQUITY UPLC

BEH C18 column (2.1 x 50 mm) at 30 °C with a flow rate of 0.8 mL/min. Unless stated otherwise,

187

all reagents, salts and solvents were purchased from commercial sources and used without further

purification.

Synthesis of the Min-Mtx CID (1, Scheme 5-1). Step a. To 9-NH2-minocycline (100 mg, 0.2

mmol), Boc-11-aminoundecanoic acid (89 mg, 0.3 mmol), EDC (75 mg, 0.4 mmol) and HOBt (60

mg, 0.4) were added DMF (1 mL) and Et3N (139 μL, 1.0 mmol) and the reaction was mixed at r.t

for 6 h. The reaction progress was monitored by RP-HPLC. Aqueous NaHCO3 (0.05 M)(22) was

then added and the aqueous phase was extracted three times with CHCl3. The combined organic

fraction was extracted with brine, the combined brine extracts were extracted twice with CHCl3,

the combined organic fraction was dried with Na2SO4 and the solvent was removed under reduced

pressure. The crude product was purified in two batches by preparative RP-HPLC (30-35% MeCN

in 99.9%:0.1% H2O:TFA, 45 min) to afford compound 3 (53.4 mg, 36%) as a yellow solid. 1H-

NMR (400 MHz, MeOD d4) δ 8.60 (s, 1H), 4.11 (s, 1H), 3.24 (dd, J = 15.4, 4.2, 1H), 3.14 (s, 6H),

3.08 (m, 3H), 3.02 (s, 6H), 2.50 (t, J = 7.4, 2H), 2.44 (t, J = 14.3, 1H), 2.24 (m, 1H), 1.72 (m, 1H),

1.62 (m, 1H), 1.43 (s, 9H), 1.42 (m, 4H) 1.38 (m, 2H), 1.32 (m, 10H). MS (APCI+): m/z calc. for

C39H58N5O10+: 756.42; found: 756.4 [M + H]+; MS (APCI-): m/z calc. for C39H56N5O10

-: 754.40;

found: 754.3 [M – H]-.

Steps b-d. To compound 3 (27.3 mg, 0.036 mmol) were added DCM (0.5 mL) and TFA (0.5 mL)

and the reaction was mixed at r.t. for 12 min. The solvent was removed under reduced pressure.

PhMe was added and the solvent was again removed under reduced pressure. PhMe addition and

solvent removal was repeated once more. Methotrexate-α-OtBu (27.5 mg, 0.054 mmol)(23, 24)

and PyBOP (37.5 mg, 0.072 mmol) were added, followed by DIPEA (11.5 mg, 0.09 mmol) in

DMF (1 mL) and the reaction was mixed at r.t.. After 22 h EtOAc and aqueous NaHCO3 were

added and the aqueous phase was extracted twice with EtOAc. The combined organic fraction was

188

extracted with brine and the brine extract was filtered. The solid residue was redissolved in MeOH

and the solvent was removed under reduced pressure. The crude product was purified in two

batches by preparative RP-HPLC (20-30% MeCN in 99.9%:0.1% H2O:TFA, 50 min). Following

solvent removal under reduced pressure, TFA (2 mL) was added and the reaction was stirred at r.t.

for 90 min. The reaction progress was monitored by RP-HPLC. The solvent was removed under

reduced pressure to afford compound 1 (4.1 mg, 10%) as a yellow solid. 1H-NMR (500 MHz,

MeOD d4) δ 8.64 (s, 1H), 8.25 (s, 1H), 7.76 (d, J = 8.8, 2H), 6.86 (d, J = 8.8, 2H), 4.92 (s, 2H),

4.54 (dd, J = 8.8, 4.5, 1H), 4.08 (s, 1H), 3.27 (s, 3H), 3.11 (m, 2H), 3.00 (s, 6H), 2.83 (s, 3H), 2.80,

(s, 3H), 2.47 (m, 2H), 2.41-2.05 (m, 6H), 1.70-1.59 (m, 4H), 1.48-1.28 (m, 16H) MS (ESI+): m/z

calc. for C54H71N13O122+: 1093.54; found: 546.9 [M + 2H]2+.

Y3H strain construction

LexA-TetR plasmids. Plasmids PBA-Gib5 and HL-249-1 through HL-249-6 encoding LexA-

TetR protein fusions for the various TetR classes were cloned by Gibson Assembly of the

appropriate TetR sequence with pMW103 digested with restriction enzymes BamHI-HF and

EcoRI-HF (Table S3). All TetR sequences with the exception of TetR(B) were obtained as IDT

gBlocks. TetR(B) sequence was amplified from pET21d-TetR, courtesy of A. Davidson.(25, 26)

LacZ assay strain construction. V506 was grown overnight in UT- media.(27) The culture was

then diluted 104-105X with UT- and plated on UT- plates. Single colonies were streaked on HTU-

and UT- plates and a streak growing on UT- but not on HTU-, indicating the loss of the

pMW3(GSG)2rGR2 plasmid, was glycerol stocked as PBA5. This strain was further used for the

transformation of the LexA-TetR plasmids to generate LacZ assay strains PBA8 and HL-260-1

through HL-260-7.

189

Growth assay strain construction. Yeast strain LW2635(28) was transformed with plasmid

PBA-Gib5 to generate strain PBA6. PBA6 was transformed with pMW2eDHFR(27) to generate

the Y3H growth assay strain PBA14.

mCherry reporter plasmid. Reporter plasmid HL-174-2 encoding mCherry under the

transcriptional control of LexA operators was cloned by Gibson Assembly of an mCherry sequence

amplified from yEpGAP-Cherry(29) and an 8LexA-min-pGal1 sequence amplified from

pMW112(14) into pRS416 digested with SacI and XhoI.

Fluorescent protein assay strain construction. V506 was grown overnight in T- media. The

culture was then diluted 104-105X with T- and plated on T- plates. Single colonies were streaked

on TU-, HT- and T- plates and a streak growing on T- but not on UT- or on HT-, indicating the loss

of the pMW106 and pMW3(GSG)2rGR2 plasmids, was glycerol stocked as PBA3. This strain was

transformed with plasmid HL-174-2 to generate yeast strain HL-294-7. The latter was transformed

with PBA-Gib5 to generate the Y3H fluorescent reporter assay strain HL-2-4-1.

TAN-1612 producer/non-producer strain construction. TAN-1612 producer and non-producer

strains EH-3-54-4 and EH-3-54-2 were obtained by transforming plasmids pYR342 and pYR291

or pYR342 and pRS426, respectively, into yeast strain BJ5464-NpgA (Table 5-1 and Table

5-2).(30)

Protocol for fluorescent protein Y3H assay

Fresh patches of Y3H strain HL-2-4-1 were inoculated into HTU- media (1 mL) in 24-well plates

(flat, clear bottom, Corning #3526) and placed in shaker. After 24 hours the cells were pelleted by

centrifugation at 3,250 rpm and resuspended in H2O (1 mL). Pelleting and resuspension were

repeated once more. OD600 was measured by diluting 10X into H2O and the cells were diluted

accordingly into H2O for OD600 = 1 to be used as a 10X solution. 20 μL cells of OD600 = 1 were

190

added to the assay plate as the last component in the assay plate (flat, clear bottom, black 96-well

plate, Corning #3603). Each well of the assay plate contained 200 μL total liquid volume, 20 μL

cells, 100 μL of 2X HTU- media of 4% raffinose and no glucose, 20 μL of 10X 20% galactose

aqueous solution, 20 μL of 10X CID solution of 100 μM in 10% DMSO aqueous solution, 20 μL

supernatant and 20 μL purified TAN-1612 in a 10% DMSO aqueous solution,(30) or a 10% DMSO

aqueous solution. Starting conditions in the assay plate were OD600 = 0.1, 2% galactose, 2%

raffinose, 0% glucose, 1X HTU-, 10 μM CID, 2% DMSO. The assay plate was placed in the 30 °C

shaker overnight before measuring OD600 and 620 nm fluorescence (λex = 588 nm).(31)

TAN-1612 supernatants from flask cultures were obtained by inoculating TAN-1612 producing

and non-producing strains, EH-3-54-4 and EH-3-54-2 respectively, in UT- media (5 mL) in 15 mL

culture tubes (Corning 352059) and placing in shaker overnight; Overnight cultures were used to

inoculate 100 mL YPD in 500 mL conical flasks with a starting OD600 of 0.08. After 68 h, 1 mL

of culture was pelleted in 1.5 mL Eppendorf tubes at 14,000 rpm, sterile filtered and diluted 20X

into H2O to be used as a 10X stock solution.

TAN-1612 supernatants from 96-well plate cultures were obtained by inoculating 12 and 84

colonies of TAN-1612 producing and non-producing strains, EH-3-54-4 and EH-3-54-2

respectively, in UT- media (0.5 mL) in a 96-well plate (Corning P-2ML-SQ-C-S). The plate was

covered with two layers of SealMate film (Excel Scientific, SM-KIT-BS) and placed in a shaker

overnight. Overnight cultures were used to inoculate 300 μL YPD in an identical setup with a

starting OD600 of 0.1 and placed in 800 rpm shaker overnight. After 54 h, 150 μL of H2O were

added to the cultures to prevent drying of cultures due to evaporation and two new layers of

Sealmate film were replaced. After 78 h, the cultures were pelleted at 3,250 rpm and the

supernatant was diluted 20X into H2O to be used as a 10X stock solution.

191

Protocol for Y3H growth assay

Fresh patches of Y3H strain PBA14 were inoculated in HT- media (1 mL) and placed in shaker

overnight. Cultures were then centrifuged at 3,000 rpm for 3 min and the pellets were resuspended

with H2O (1 mL). Pelleting and resuspension was repeated twice more. OD600 was measured and

the cells were diluted accordingly into H2O to OD600 = 1 to be used as a 10X stock solution. 20 μL

cells of OD600 = 1 were added as the last component to the assay plate (flat, clear bottom, black

96-well plate, Corning #3603). Each well of the assay plate contained 200 μL total liquid volume,

20 μL cells, 100 μL of 2X HT- media with 4% raffinose, 4% galactose, 0% glucose, 0.4% 5-FOA,

20 μL of 0/250 μM 10X CID in 10% DMSO aqueous solution, 20 μL of doxycycline (0/50 μM in

10% EtOH aqueous solution) and 40 μL H2O. Starting conditions in the assay plate were OD600 =

0.1, 2% galactose, 2% raffinose, 0% glucose, 0.2% 5-FOA, 1X HT-, 25 μM CID, 1% DMSO, 1%

EtOH. The assay plate was placed in the 30 °C shaker for 5 days with daily measurements of

OD600.

Protocol for Y3H LacZ assay

Fresh patches of Y3H strains PBA-8 and HL-260-1 through HL-260-7 were inoculated into HTU-

media (1 mL) in 24-well plates (flat, clear bottom, Corning #3526) and place at 30 °C shaker. After

24 hours the cells were pelleted by centrifugation at 3000 rpm for 5 min and resuspended in H2O

(1 mL). Pelleting and resuspension were repeated once more. OD600 was measured and the cells

were diluted accordingly into H2O to OD600 = 1 to be used as a 10X solution. 20 μL cells of OD600

= 1 were added to the assay plate as the last component in the assay plate (clear v-bottom, clear

96-well plate, Corning #3894). Each well of the assay plate contained 200 μL total liquid volume,

20 μL cells, 100 μL of 2X HTU- media of 4% raffinose and no glucose, 20 μL of 10X 20%

galactose aqueous solution, 20 μL of 10X CID solution in 10% DMSO aqueous solution or a 10%

192

DMSO aqueous solution, 20 μL of the tetracycline ligand in 10% EtOH aqueous solution and 20

μL water. Starting conditions in the assay plate are OD600 = 0.1, 2% galactose, 2% raffinose, 0%

glucose, 1X HTU-, varying concentration CID, varying concentration of tetracycline ligand, 1%

DMSO and 1% EtOH. The assay plate was placed in the shaker overnight. OD measurements were

then taken after 24 h and the cells were pelleted and resuspended in 100 μL Z-buffer (60 mM

Na2HPO4, 40 mM NaH2PO4, 10 mM KCl, 2 mM MgSO4, pH adjusted to 7, 2.7 mL/L β-

mercaptoethanol added fresh the day of the assay). The cells were pelleted again, resuspended in

100 μL Y-PER buffer (Fisher Scientific, #78990) and lysed for 30 min at r.t. before the addition

of o-Nitrophenyl β-D-galactopyranoside (ONPG) in Z-buffer (8.5 μL, 10 mg/mL). After 90 min a

Na2CO3 solution (1 M, 110 μL) was added to quench the reaction. After pelleting at 3,000 rpm,

175 μL of the supernatant was transferred into flat clear-bottom 96-well plates (Corning #353072)

and OD420 was measured. The limit of detection was calculated as the concentration of molecule

at which the signal-to-noise ratio of the assay was > 3:1.(32, 33)

5.4 Results

5.4.1 Y3H system design and CID design and synthesis

For a Y3H system detecting tetracyclines, we cloned a Y3H strain encoding LexA-TetR and

DHFR-B42 fusion proteins and synthesized a minocycline-methotrexate (Min-Mtx) CID (Figure

5-1). We chose to base our Y3H system on well-studied components. Namely, the LexA DNA

binding domain (DBD) and B42 activation domain (AD),(34) respectively, introduced by Brent

and coworkers,(14, 27, 35, 36) as well as the DHFR- methotrexate and TetR-tetracycline

interactions. The latter two are highly characterized receptor-ligand interactions of picomolar

affinity,(37) and the methotrexate-DFHR interaction has been used numerous times by our

laboratory.(27, 35) We used LexA-TetR fusion proteins of TetR variants that were highly studied,

193

such as the variants from classes B and D,(37-39) as well TetR variants from the much less studied

TetR classes A,(40, 41) C,(42, 43) E,(44, 45) G,(46) and H.(47-50) TetR and LexA form stable

homodimers, with each of the TetR monomers binding tetracyclines and LexA binding DNA as a

dimer.(38, 51) Thus it is possible that the monomer ratio of TetR / LexA in the LexA-TetR fusion

protein is unequal to 1, affecting accordingly the local concentrations of the Min-Mtx CID and

DHFR-B42 as well. In any case, increased levels of tetracyclines are expected to outcompete the

CID from the LexA-TetR protein fusion and lower reporter protein expression (Figure 5-1).

Our design and synthesis of the Min-Mtx CID is based on the wealth of structure activity

relationship (SAR) data for the interaction of tetracyclines with TetR, as well as our laboratory’s

previously reported CIDs.(27, 35) It is known from inspection of the high-resolution structure of

TetR bound to tetracycline derivatives as well as biochemical characterization that tetracyclines

can be derivatized at the D ring without disrupting binding to TetR (Scheme 5-1).(37, 38, 52)

Moreover, the FDA-approved analog tigecycline and the clinical candidates eravacycline and TP-

271 are all 9-amidotetracyclines;(53, 54) thus, synthesis of a 9-amidotetracycline CID was logical.

Specifically, 9-aminominocycline was chosen as the starting material because of its high acid

stability relative to tetracycline as well as its commercial availability.(55) The design and synthesis

of the Mtx component of the Min-Mtx CID was based on our laboratory’s previously reported

methotrexate CIDs (Scheme 5-1).(27, 35)

Scheme 5-1 Synthesis of the chemical inducer of dimerization (CID) Min-Mtxa

194

aReagents and conditions: (a) Boc-11-aminoundecanoic acid (1.5 equiv), EDC (2 equiv), HOBt (2

equiv), Et3N (5 equiv), DMF, 22 oC, 6 h, 36%; (b) TFA:DCM (1:1), 22 oC, 12 min; (c)

Methotrexate-α-OtBu (1.5 equiv),(23, 24) PYBOP (2 equiv), DIPEA (2.5 equiv), DMF, 22 oC, 22

h; (d) TFA, 22 oC, 1.5 h, 10% over three steps. EDC = N-(3-Dimethylaminopropyl)-N′-

ethylcarbodiimide hydrochloride, HOBt = 1-hydroxybenzotriazole, TFA = trifluoroacetic acid,

PyBOP = (Benzotriazol-1-yloxy)tripyrrolidinophosphonium hexafluorophosphate, DIPEA = N-

Ethyldiisopropylamine.

5.4.2 Initial characterization of the Y3H assay for tetracycline derivatives

With the Y3H strains and Min-Mtx CID at hand we verified the functionality of our Y3H system

and determined the optimal CID concentration and TetR variant to further use in the assay. First,

we confirmed that reporter gene output in our system is dependent upon the presence of the CID

and determined the desired range of CID concentration to use in the assay (Figure 5-2a). We then

tested candidates from each of the TetR classes as protein receptors and observed that the Y3H

system is responsive to the CID with all known TetR classes except for the TetR(C) variant we

used (Figure 5-2b). Given that TetR(B) and TetR(G) showed the best response to the CID and to

maintain consistency with the highly studied TetR(B),(37-39) we focused our further experiments

on TetR(B).

195

We show that our assay can differentiate sub-μM concentrations of various tetracyclines (Figure

5-2c), key for its applicability to differentiate between high and low producer strains. Our Y3H

system differentiates doxycycline, tetracycline and 9-NH2-minocycline concentrations in the range

of 2 – 200 nM, 0.02 – 2 μM and 0.2 – 20 μM, respectively. The limit of detection in our Y3H assay

is ≤ 0.2 μM for doxycycline and ≤ 2 μM for tetracycline and 9-NH2-minocycline.(32)

Figure 5-2 Characterization of the dynamic range of the Y3H assay for tetracyclines

Characterization of the dynamic range of the Y3H assay for tetracyclines in (a) varying

concentrations of the Min-Mtx CID, (b) varying TetR classes and (c) varying target molecule

structure and concentrations. LacZ was used as a reporter gene. Miller units were calculated as

196

(1000*OD420)/(well-volume(mL)*reaction-time(min)*OD600).(56) “+TetR” and “-TetR” are

strains HL-260-5 and HL-260-7 encoding LexA-TetR(G) and LexA, respectively; Error bars

represent the standard error of the mean (s.e.m) from three biological replicates.

After confirming the functionality of the Y3H system for tetracyclines we proceeded to show that

it can report the presence of tetracyclines with a modular output, key for enabling versatile use by

various end-users. Using a URA3 reporter gene and growing the yeast in the presence of 5-

fluorouracil (5-FOA) we show that yeast growth is doxycycline-dependent in the presence, but not

in the absence of the Min-Mtx CID (Figure 5-3a). We additionally show that the Y3H assay for

tetracyclines can be used as a colorimetric as well as a fluorometric assay, by employing a LacZ

or and mCherry reporter gene, respectively (Figure 5-3b, Figure 5-3c). As expected, while reporter

gene activity is dependent on CID and doxycycline in the Y3H strain, such dependence is not

detected in a control strain excluding the eDFHR-AD fusion protein (Figure 5-5).

197

Figure 5-3 The modularity of the yeast three hybrid assay enables both screening and

selecting for biosynthesis of target tetracyclines

The modularity of the yeast three hybrid assay enables both screening and selecting for

biosynthesis of target tetracyclines by enabling (a) a growth assay, (b) a colorimetric assay and a

(c) fluorometric assay. (a) Yeast growth, (b) LacZ transcription and (c) mCherry transcription are

dependent on doxycycline (Dox) in the presence, but not in the absence of the CID. CID, Dox

concentrations = (a) 25 μM, 5 μM (b) 25 μM, 0.2 μM (c) 10 μM, 1 μM. Error bars represent the

standard error of the mean (s.e.m) from three biological replicates. Miller units were calculated as

(1000*OD420)/(well-volume(mL)*reaction-time(min)*OD600).(56) mCherry (620 nm)/OD600

was calculated by dividing mCherry fluorescence (620 nm, λex = 588 nm) by OD600.

198

5.4.3 Applying the Y3H assay to the detection of target molecule biosynthesis

We demonstrate the applicability of the Y3H assay to metabolic engineering by differentiating

between producer and non-producer strains of TAN-1612 (Figure 5-4a).(30) For the TAN-1612

producer strain we used S. cerevisiae strain EH-3-54-4, encoding all four genes of the TAN-1612

biosynthetic pathway as previously reported by Tang and coworkers and producing ~60 μM TAN-

1612 (Supplementary methods).(30) The non-producer control we used, S. cerevisiae strain EH-

3-54-2, does not encode the polyketide synthase AdaA and has no measurable production of TAN-

1612 (data not shown).

The Y3H assay for tetracyclines can differentiate between supernatants of producer and non-

producer strains of TAN-1612 (Figure 5-4b). When purified TAN-1612 is spiked into non-

producer supernatants, the resulting Y3H signal resembles the signal obtained with producer

supernatant (Figure 5-4b). These results agree with our hypothesis that TAN-1612, present in the

producer strain supernatant but not in the non-producer strain supernatant, outcompetes the CID

from the LexA-TetR fusion protein.

The Y3H assay for tetracyclines also differentiates TAN-1612 producer colonies from a non-

producing population cultured in a 96-well plate (Figure 5-4c). The 12 producer colonies gave an

average mCherry fluorescence / OD600 signal of 132 with a standard deviation of 17 and the 84

non-producer colonies gave an average mCherry fluorescence / OD600 signal of 210 with a standard

deviation of 16. Thus, assuming normal distribution there is less than 2.5% overlap in the Y3H

signal from producer and non-producer colonies.

199

Figure 5-4 Differentiation of a producer and a non-producer strain of a target molecule by

the Y3H assay for tetracyclines, demonstrating the applicability of the Y3H assay to

metabolic engineering.

(a) Workflow for screening colonies for production of a target molecule using the Y3H assay. (b)

The Y3H assay for tetracyclines differentiates between the supernatants of TAN-1612 producer

and TAN-1612 non-producer cultured in shake flasks. (c) The Y3H assay for tetracyclines

differentiates the supernatants of TAN-1612 producer colonies from a non-producing population

cultured in a 96-well plate. The non-producer strain does not encode AdaA, the polyketide

synthase of the TAN-1612 biosynthetic pathway, while the producer strain encodes the complete

200

pathway (Table 5-1 and Table 5-2). The concentration of TAN-1612 in the Y3H assay well from

the supernatant and from the purified sample is ~0.3 μM and 0.5 μM, respectively (Supplementary

methods). mCherry (620 nm)/OD600 was calculated by dividing mCherry fluorescence (620 nm,

λex = 588 nm) by OD600. Error bars represent the standard error of the mean (s.e.m) from three

biological replicates of the Y3H strain and two biological replicates of the TAN-1612

producer/non-producer.

5.5 Discussion

The Y3H assay for tetracyclines is advantageous compared to the state-of-the-art methods for

tetracycline detection in terms of throughput, cost, instrumentation requirements and limit of

detection. Being readily performed in 96-well microtiter plates, the Y3H assay throughput is >1000

samples / hour when measured by a standard laboratory plate reader (Infinite-M200). By

comparison, the throughput of the previous state-of-the-art HPLC-based methods for tetracycline

detection is <10 samples/hour.(33, 57) The use of a plate reader to measure the Y3H assay output

is also advantageous in terms of cost to state-of-the-art chromatography-based methods such as

HPLC and even more so, LCMS. The price difference is both in the initial investment in

instrumentation, as spectrophotometers are cheaper than LC instruments, and in the reagent costs

of 96 well plates vs LC-grade solvents. In terms of limit of detection the Y3H assay has a limit of

detection of ≤ 200 nM for doxycycline (Figure 5-2c), which is better than the state-of-the-art HPLC

limit of detection of ≥ 4.5 μM for doxycycline.(33, 57) The low limit of detection our assay exhibits

enables it to differentiate producer from non-producer strains even in low titers of initial strain

development (Figure 5-4 and Section 5.7.1 5.7.2, ~60 μM). Differentiating between production

levels in higher titer ballparks in later stages of producer strain optimization would simply require

a higher dilution factor of supernatants into the Y3H strain media. Future generations of this Y3H

201

assay might include TetR mutants that are more specific and have a lower detection limit to

tetracycline derivatives of interest.

The Y3H assay for metabolic engineering also compares well with our laboratory’s recently

published FP assay,(58) because of two key differences in setup requirements and output. First,

since the Y3H assay does not require the purification of the protein receptor, it is used much more

readily to screen for the ultimate protein receptor for the assay. Thus, we show here the first

comparison of all TetR classes for binding of a small molecule in an attempt to identify the best

TetR variant for differentiating between + and – CID (Figure 5-2b). Further optimization of a

protein receptor for the assay of a specific target molecule is readily enabled in the Y3H assay

simply by cloning the protein variants into the same plasmid backbone and transforming into yeast.

The versatility in choice of output in the Y3H assay is a key enabling factor for the advantages the

Y3H assay has over the FP assay since it makes the assay accessible to a wider range of users. The

fluorescent output in the Y3H assay enables measurements with a standard laboratory plate reader,

eliminating the requirement for anisotropy capacities (Figure 5-3c). The LacZ output could also

be measured by a standard plate reader as well as by the naked eye (Figure 5-3b).

If the Y3H system and the metabolic pathway for the small molecule of interest are cloned into

the same strain, colonies can be assayed for production even more directly, increasing throughput

and minimizing handling. The Y3H assay can then enable screening colonies for production of the

target molecule directly on an agar plate without the need for further handling of the colonies in

microtiter plates. The agar plate screen could be performed with the naked eye or a simple camera

by using X-Gal plates and the LacZ reporter or fluorescent camera/microscope by using the

mCherry reporter. Finally, the Y3H assay throughput could be vastly increased by selecting for

high producing strains en masse in flask or fermenter culturing using the growth assay or

202

fluorescence assisted cell sorting (FACS). Such en masse screening methods not only greatly

enhance the throughput but also minimize the labor, instrumentation, cost and time, required to

obtain results. The Y3H assay for tetracyclines can report the presence of a target molecule with

any of the above mentioned outputs (Figure 5-3), enabling metabolic engineering applications for

a range of end users with varying needs and instrumentation availabilities.

While a TetR-tetO-based assay could conceivably be developed for the metabolic engineering of

tetracycline derivatives,(59) our TetR-based Y3H assay is advantageous for two main reasons. For

one, the Y3H assay can potentially be applied to a greater variety of tetracycline derivatives as

TetR-binding to the tetracycline suffices and there is no demand for a TetR conformational change.

By comparison, signal transduction in the TetR-tetO system is dependent on coupling between

tetracycline binding and a conformational change in TetR.(26, 38) It has been hypothesized that

high-affinity binding may be a necessary but not sufficient property for TetR induction in the TetR-

TetO system.(60) In practice, while it was possible to detect binding to a tetracycline analog, 4-

de(dimethylamino)-anhydrotetracycline, no induction was detected using a TetR-TetO-based

assay.(37) Thus, the use of TetR in a Y3H system can facilitate the evolvement of TetR mutants

with specific binding to tetracycline derivatives of interest in future generations of the assay.

Furthermore, the Y3H system for metabolic engineering can be readily modified to apply for many

metabolites of interest beyond tetracyclines, as detailed below. In contrast, the TetR-TetO system

is much more limited in the scope of molecules it could be applied to, namely transcriptional

regulators.(61)

A Y3H system as described here for the metabolic engineering of tetracycline derivatives can be

designed for any target molecule with a receptor. Given T, a desirable target, PR, a protein receptor

for T, and T’, a readily derivatizable analog of T, two new components need to be generated for a

203

Y3H assay to detect biosynthesis of T. The first is a Mtx-T’ CID and the second is a LexA-PR

fusion protein (Figure 5-1). This approach is especially amenable for the development of assays

for molecules of pharmacological interest. The reason is that such molecules often have a known

protein target as well as an established derivatization chemistry and SAR that greatly support the

design and synthesis of a CID. More broadly, building from the Cornish Laboratory’s early reports

of chemical complementation,(17, 62, 63) this work advances the enabling technology of the Y3H

system for the emerging field of synthetic biology for handling the diversity of engineering multi-

gene pathways and assaying for chemistry not natural to the cell.

5.6 Conclusion

This chapter describes the development of a yeast three hybrid (Y3H) assay as a general, high-

throughput, versatile and readily-implemented approach for the detection of target molecule

biosynthesis. We demonstrate the use of the Y3H assay for detecting the biosynthesis of

tetracyclines, a major class of antibiotics, based on the interaction between tetracyclines and the

tetracycline repressor protein (TetR). We demonstrate the applicability of the Y3H assay to

metabolic engineering by differentiating between producer and non-producer strains of the natural

product anhydrotetracycline TAN-1612. The Y3H assay is superior to state-of-the-art HPLC-MS

methods in throughput and limit of detection of tetracycline derivatives. Finally, our establishment

of the Y3H assay for detecting the biosynthesis of an anhydrotetracycline supports the generality

of the Y3H assay for detecting the biosynthesis of many other target molecules. Specifically, future

versions of this assay to screen or select for further modifications of TAN-1612 that could not be

assayed by the UV/Vis assay for hydroxylation of anhydrotetracycline used in the previous

chapters are envisioned.

204

5.7 Appendix

5.7.1 Supplementary figures

Figure 5-5 LacZ readout of strain harboring plasmid encoding for LexA-TetR (PBA-8) and

strain without the plasmid (PBA-5).

Only the strain with the LexA-TetR fusion protein responds to the CID. Addition of Dox lowers

reporter gene expression only in the strain with LexA-TetR in the presence of the CID.

Concentrations of CID and Dox are 25 μM and 200 nM, respectively. Miller units were calculated

as (1000*OD420)/(well-volume(mL)*reaction-time(min)*OD600).(56) Error bars represent the

standard error of the mean (s.e.m) from three biological replicates.

0

100

200

300

400

500

+ CID+Dox

+ CID-Dox

− CID +Dox

− CID -Dox

+ CID+Dox

+ CID-Dox

− CID +Dox

− CID -Dox

+LexA-TetR -LexA-TetR

Mill

er

Un

its

205

Figure 5-6 standard curve for TAN-1612 quantification in cultures.

A Purified TAN-1612 was analyzed at different concentrations on a separation gradient of 5% to

95% MeCN in supercritical CO2 over 5.5 minutes. Absorption between 390 nm to 410 nm was

integrated at retention times of 0.87 – 1.50 min, which corresponds to TAN-1612 (MS (AI-): m/z

calc. for C21H17O9-: 413.09; found: 413.4 [M - H]-; MS (EI-): m/z calc. for C21H17O9

-: 413.09;

found: 413.4 [M - H]-).

5.7.2 Supplementary methods

Quantification of TAN-1612 concentration in cultures.

TAN-1612 cultures for quantification were obtained by inoculating two biological replicates of

each of TAN-1612 producing and non-producing strains, EH-3-54-4 and EH-3-54-2 respectively,

in UT- media (5 mL) in 15 mL culture tubes (Corning 352059) and placing in shaker overnight.

Overnight cultures were used to inoculate 100 mL YPD in 500 mL conical flasks with a starting

OD600 of 0.08. After 68 h, the 100 mL from the two biological replicates were combined and

extracted with 200 mL of EtOAc in 8 x 50 mL falcon tubes (Corning 352098) by mixing at

maximal speed in Orbital Shaker (Bellco Glass) at r.t. for 1.5 h and centrifuging at 3,300 rpm at

r.t. for 15 min. The solvent of the combined organic phase was removed under reduced pressure

y = 0.016x + 8.1328R² = 0.9933

0

2

4

6

8

10

12

14

0 50 100 150 200 250 300

Ln

(ab

so

rpti

on

39

0-4

10

nm

)

TAN-1612 (μM)

206

and the sample was redissolved in MeOH (1.5 mL) and filtered (Acrodisc® Syringe Filters, 13mm,

0.2 μm, Pall Laboratory, PTFE Membrane, 4422). The filtered sample was diluted 50X in MeOH

and analyzed by Supercritical Fluid Chromatography / Mass spectrometry (SFC/MS) equipped

with a photodiode array (PDA) UV-vis detector. Absorption between 390 nm to 410 nm was

integrated at retention times of 0.87 – 1.50 min, which corresponds to TAN-1612 (MS (AI-): m/z

calc. for C21H17O9-: 413.09; found: 413.4 [M - H]-. The TAN-1612 concentration in the extract

was calculated to be 161 μM according to the following linear function:

concentration=[ln(absorption 390-410 nm)-8.1328]/0.016 (Figure 5-6). The concentration of

TAN-1612 in the culture is therefore estimated to be ~60 μM, assuming a quantitive yield for the

extraction process.

5.7.3 Supplementary tables

Table 5-1 Strains used in this study

Strain Genotype Source/Reference

FY250 MATα ura3-52 trp1 63 his3 200 leu2 1 gal L.P. Guarentea

LW2635 MATa trp1Δ63 his3Δ200 8lexAop-Spo13-URA3

leu2Δ1 Gal+

/(28)

BJ5464-

NpgA

MATα ura3-52 his3-Δ200 leu2-Δ1

trp1 pep4::HIS3 δ:: pADH2-npgA-tADH2 prb1Δ1.6R can1

GAL

Y. Tangb/(64)

V506 FY250 pMW106, pMW3(GSG)2rGR2, pMW2eDHFR /(27)

PBA-5 FY250 pMW106, pMW2eDHFR This study

HL-260-1 FY250 pMW106, pMW103TetR(A), pMW2eDHFR This study

PBA-8 FY250 pMW106, pMW103TetR(B), pMW2eDHFR This study

HL-260-2 FY250 pMW106, pMW103TetR(C), pMW2eDHFR This study

HL-260-3 FY250 pMW106, pMW103TetR(D), pMW2eDHFR This study

HL-260-4 FY250 pMW106, pMW103TetR(E), pMW2eDHFR This study

HL-260-5 FY250 pMW106, pMW103TetR(G), pMW2eDHFR This study

HL-260-6 FY250 pMW106, pMW103TetR(H), pMW2eDHFR This study

HL-260-7 FY250 pMW106, pMW103(empty), pMW2eDHFR This study

PBA-6 LW2635 PBA-Gib5 This study

PBA-14 LW2635 PBA-Gib5, pMW2eDHFR This study

PBA-3 FY250 pMW2eDHFR This study

HL-294-7 FY250 pMW2eDHFR HL-174-2 This study

HL-2-4-1 FY250 pMW2eDHFR pMW103TetR(B), HL-174-2 This study

207

EH-3-54-4 BJ5464-NpgA pYR291, pYR342 This study

EH-3-54-2 BJ5464-NpgA pRS426, pYR342 This study a L. P. Guarente, Massachusetts Institution of Technology, Biology Department

b Y. Tang, University of California, Los Angeles, Department of Chemical and Biomolecular

Engineering & Department of Chemistry and Biochemistry, Department of Bioengineering

Table 5-2 Plasmids used in this study

Plasmid Description Source/Reference

pMW2eDHFR pGal1-eDHFR-B42, trp marker, 2 μ /(27)

pMW106 LacZ under control of 8 tandem LexA operators, Ura

marker

/(27, 65)

pMW103 pADH1-LexA (empty), His marker, 2 μ R. Brenta/(65)

pMW112 LacZ under control of 8 tandem LexA operators, Ura

marker

R. Brent/(14, 65)

HL-249-1 pADH1-LexA-TetR(A)-tADH1 on pMW103 This study

PBA-Gib5 pADH1-LexA-TetR(B)-tADH1 on pMW103 This study

HL-249-2 pADH1-LexA-TetR(C)-tADH1 on pMW103 This study

HL-249-3 pADH1-LexA-TetR(D)-tADH1 on pMW103 This study

HL-249-4 pADH1-LexA-TetR(E)-tADH1 on pMW103 This study

HL-249-5 pADH1-LexA-TetR(G)-tADH1 on pMW103 This study

HL-249-6 pADH1-LexA-TetR(H)-tADH1 on pMW103 This study

pRS416 Shuttle vector (empty), uracil marker, CEN ATCC87521

HL-174-2 8LexAOp-minpGal1-mCherry-tCyc1, Ura marker, CEN This study

pYR291 AdaA expression in S. cerevisiae, uracil marker, 2 μ Y. Tangb/(30)

pYR342 AdaB-D expression in S. cerevisiae, Tryp marker, 2 μ Y. Tang/(30)

pRS426 Shuttle vector (empty), uracil marker, 2 μ ATCC77107

pET21d-TetR TetR(B) with C-term Histidine tag A.

Davidsonc/(25)

yEpGAP-

Cherry

mCherry, uracil marker, 2 μ Neta Deand/(29)

a R. Brent, Fred Hutchinson Cancer Research Center, Division of Basic Sciences

b Y. Tang, University of California, Los Angeles, Department of Chemical and Biomolecular

Engineering & Department of Chemistry and Biochemistry, Department of Bioengineering

c A. Davidson, Dept. of Molecular Genetics, Dept. of Biochemistry, Faculty of Medicine,

University of Toronto

d Neta Dean, Department of Biochemistry and Cell Biology, Stony Brook University

Table 5-3 Sequences of the various TetR classes in the within the pMW103 backbone.

208

Sequences for the TetR classes are shown decapitalized within the context of LexA-DBD (partial,

capitalized) in the 5’ end and the tADH1 terminator at the 3’ end (partial, capitalized).

The NCBI and GenBank Reference Sequences used to order/clone DNA for the different TetR

classes A, B, C, D, E, G and H are NC_006143.1, CP012140.1, NC_002056.1, CP000650.1,

LKJQ01000048.1, CP003225.1 and AJ514834.1, respectively.

TetR(A) –

HL-249-1

GCTGCGCGTCAGCGGGATGTCGATGAAAGATATCGGCATTATGGATG

GTGACTTGCTGGCAGTGCATAAAACTCAGGATGTACGTAACGGTCAG

GTCGTTGTCGCACGTATTGATGACGAGGTTACCGTTAAGCGCCTGAA

AAAACAGGGCAATAAAGTCGAACTGTTGCCAGAAAATAGCGAGTTT

AAACCAATTGTCGTAGATCTTCGTCAGCAGAGCTTCACCATTGAAGG

GCTGGCGGTTGGGGTTATTCGCAACGGCGACTGGCTGGAATTCatgacaa

agttgcagccgaatacagtgatccgtgccgccctggacctgttgaacgaggtcggcgtagacggtctgacgacac

gcaaactggcggaacggttgggggttcagcagccggcgctttactggcacttcaggaacaagcgggcgctgctc

gacgcactggccgaagccatgctggcggagaatcatacgcattcggtgccgagagccgacgacgactggcgct

catttctgatcgggaatgcccgcagcttcaggcaggcgctgctcgcctaccgcgatggcgcgcgcatccatgccg

gcacgcgaccgggcgcaccgcagatggaaacggccgacgcgcagcttcgcttcctctgcgaggcgggtttttcg

gccggggacgccgtcaatgcgctgatgacaatcagctacttcactgttggggccgtgcttgaggagcaggccgg

cgacagcgatgccggcgagcgcggcggcaccgttgaacaggctccgctctcgccgctgttgcgggccgcgata

gacgccttcgacgaagccggtccggacgcagcgttcgagcagggactcgcggtgattgtcgatggattggcgaa

aaggaggctcgttgtcaggaacgttgaaggaccgagaaagggtgacgattagGATCCGTCGACCAT

GGCGGCCGCTCGAGTCGACCTGCAGCCAAGCTAATTCCGGGCGAATT

TCTTATGATTTATGATTTTTATTATTAAATAAGTTATAAAAAAAATAA

GTGTATACAAATTTTAAAGTGACTCTTAGGTTTTAAAACGAAAATTCT

TATTCTTGAGTAACT

TetR(B) –

PBA-Gib5

GCTGCGCGTCAGCGGGATGTCGATGAAAGATATCGGCATTATGGATG

GTGACTTGCTGGCAGTGCATAAAACTCAGGATGTACGTAACGGTCAG

GTCGTTGTCGCACGTATTGATGACGAGGTTACCGTTAAGCGCCTGAA

AAAACAGGGCAATAAAGTCGAACTGTTGCCAGAAAATAGCGAGTTT

AAACCAATTGTCGTAGATCTTCGTCAGCAGAGCTTCACCATTGAAGG

GCTGGCGGTTGGGGTTATTCGCAACGGCGACTGGCTGGAATTCatgtcta

gattagataaaagtaaagtgattaacagcgcattagagctgcttaatgaggtcggaatcgaaggtttaacaacccgt

aaactcgcccagaagctaggtgtagagcagcctacattgtattggcatgtaaaaaataagcgggctttgctcgacg

ccttagccattgagatgttagataggcaccatactcacttttgccctttagaaggggaaagctggcaagattttttacgt

aataacgctaaaagttttagatgtgctttactaagtcatcgcgatggagcaaaagtacatttaggtacacggcctaca

gaaaaacagtatgaaactctcgaaaatcaattagcctttttatgccaacaaggtttttcactagagaatgcattatatgc

actcagcgctgtggggcattttactttaggttgcgtattggaagatcaagagcatcaagtcgctaaagaagaaaggg

aaacacctactactgatagtatgccgccattattacgacaagctatcgaattatttgatcaccaaggtgcagagccag

ccttcttattcggccttgaattgatcatatgcggattagaaaaacaacttaaatgtgaaagtgggtcttagGATCC

GTCGACCATGGCGGCCGCTCGAGTCGACCTGCAGCCAAGCTAATTCC

GGGCGAATTTCTTATGATTTATGATTTTTATTATTAAATAAGTTATAA

AAAAAATAAGTGTATACAAATTTTAAAGTGACTCTTAGGTTTTAAAA

CGAAAATTCTTATTCTTGAGTAACT

TetR(C) –

HL-249-2

GCTGCGCGTCAGCGGGATGTCGATGAAAGATATCGGCATTATGGATG

GTGACTTGCTGGCAGTGCATAAAACTCAGGATGTACGTAACGGTCAG

209

GTCGTTGTCGCACGTATTGATGACGAGGTTACCGTTAAGCGCCTGAA

AAAACAGGGCAATAAAGTCGAACTGTTGCCAGAAAATAGCGAGTTT

AAACCAATTGTCGTAGATCTTCGTCAGCAGAGCTTCACCATTGAAGG

GCTGGCGGTTGGGGTTATTCGCAACGGCGACTGGCTGGAATTCatgaaca

agctccaacgcgaggccgtgatccgaaccgcgctcgaactgcttaacgacgtgggcatggaaggtctaacgacg

cgccgactggctgagcgcctcggggtgcaacagccagcgctctactggcatttcaagaacaagcgtgcgttgctc

gacgcacttgccgaagccatgctgacgataaatcacacgcattcgacgccaagggatgacgacgactggcgttc

gttcctgaagggcaatgcatgcagttttcgacgggcgttgctcgcttatcgcgatggcgcgcgtattcatgccggga

cgcggccagccgcgccgcagatggaaaaagccgacgcgcagcttcgcttcctttgcgatgctggcttttcggcag

gtgacgcgacctatgcgttgatggcaatcagctacttcaccgtcggcgctgttcttgagcagcaagctagcgaggc

agacgccgaggagcggggcgaagatcagttgaccacctcagcgtctacgatgccggcgcgcctacagagcgc

gatgaaaatcgtctacgaaggcggtccggacgcggcattcgagcgaggcctggctctcatcatcggcggtcttga

aaaaatgaggctcactacgaacgacattgaggtgctgaagaatgttgacgaatagGATCCGTCGACCA

TGGCGGCCGCTCGAGTCGACCTGCAGCCAAGCTAATTCCGGGCGAAT

TTCTTATGATTTATGATTTTTATTATTAAATAAGTTATAAAAAAAATA

AGTGTATACAAATTTTAAAGTGACTCTTAGGTTTTAAAACGAAAATT

CTTATTCTTGAGTAACT

TetR(D) –

HL-249-3

GCTGCGCGTCAGCGGGATGTCGATGAAAGATATCGGCATTATGGATG

GTGACTTGCTGGCAGTGCATAAAACTCAGGATGTACGTAACGGTCAG

GTCGTTGTCGCACGTATTGATGACGAGGTTACCGTTAAGCGCCTGAA

AAAACAGGGCAATAAAGTCGAACTGTTGCCAGAAAATAGCGAGTTT

AAACCAATTGTCGTAGATCTTCGTCAGCAGAGCTTCACCATTGAAGG

GCTGGCGGTTGGGGTTATTCGCAACGGCGACTGGCTGGAATTCatggcac

ggctgaacagagaatcggttattgatgcggcactggaactgctgaatgagacagggattgacgggctgacgaccc

gcaagctggcgcagaagctgggaatagaacagccgacactttactggcatgtgaaaaataaacgggcgttactgg

atgcgctggcggtggagatcctggcgcgtcatcatgattattcactgcctgcggcgggggaatcctggcagtcattt

ctgcgcaataatgcaatgagtttccgccgggcgctgctgcgttaccgtgacggggcaaaagtgcacctcggcacc

cgccctgatgaaaaacagtatgatacggtggaaacccagttacgctttatgacagaaaacggcttttcactgcgcga

cgggttatatgcgatttcagcggtcagtcattttacccttggtgccgtactggagcagcaggagcatactgccgccct

gaccgaccgccctgcagcaccggacgaaaacctgccgccgctattgcgggaagcgctgcagattatggacagt

gatgatggtgagcaggcctttctgcatggcctggagagcctgatccgggggtttgaggtgcagcttacggcactgt

tgcaaatagtcggtggtgataaacttatcatccccttttgctagGATCCGTCGACCATGGCGGCC

GCTCGAGTCGACCTGCAGCCAAGCTAATTCCGGGCGAATTTCTTATG

ATTTATGATTTTTATTATTAAATAAGTTATAAAAAAAATAAGTGTATA

CAAATTTTAAAGTGACTCTTAGGTTTTAAAACGAAAATTCTTATTCTT

GAGTAACT

TetR(E) –

HL-249-4

GCTGCGCGTCAGCGGGATGTCGATGAAAGATATCGGCATTATGGATG

GTGACTTGCTGGCAGTGCATAAAACTCAGGATGTACGTAACGGTCAG

GTCGTTGTCGCACGTATTGATGACGAGGTTACCGTTAAGCGCCTGAA

AAAACAGGGCAATAAAGTCGAACTGTTGCCAGAAAATAGCGAGTTT

AAACCAATTGTCGTAGATCTTCGTCAGCAGAGCTTCACCATTGAAGG

GCTGGCGGTTGGGGTTATTCGCAACGGCGACTGGCTGGAATTCatggcac

gactaagcttggacgacgtaatttcaatggcgctcaccctgctggacagcgaagggctagagggcttgactacgc

gtaagctggcgcagtccctaaaaattgagcaaccgactctgtattggcacgtgcgcaacaagcagactcttatgaa

catgctttcagaggcaatactggcgaagcatcacacccgttcagcaccgttaccgactgagagttggcagcagtttc

tccaggaaaatgctctgagtttccgtaaagcattactggtccatcgtgatggagcccgattgcatatagggacctctc

210

ctacgcccccccagtttgaacaagcagaggcgcaactacgctgtctatgcgatgcagggttttcggtcgaggagg

ctcttttcattctgcaatctatcagccattttacgttgggtgcagtattagaggagcaagcaacaaaccagatagaaaa

taatcatgtgatagacgctgcaccaccattattacaagaggcatttaatattcaggcgagaacctctgctgaaatggc

cttccatttcgggctgaaatcattaatatttggattttctgcacagttagatgaaaaaaagcatacacccattgaggatg

gtaataaatagGATCCGTCGACCATGGCGGCCGCTCGAGTCGACCTGCAGC

CAAGCTAATTCCGGGCGAATTTCTTATGATTTATGATTTTTATTATTA

AATAAGTTATAAAAAAAATAAGTGTATACAAATTTTAAAGTGACTCT

TAGGTTTTAAAACGAAAATTCTTATTCTTGAGTAACT

TetR(G) –

HL-249-5

GCTGCGCGTCAGCGGGATGTCGATGAAAGATATCGGCATTATGGATG

GTGACTTGCTGGCAGTGCATAAAACTCAGGATGTACGTAACGGTCAG

GTCGTTGTCGCACGTATTGATGACGAGGTTACCGTTAAGCGCCTGAA

AAAACAGGGCAATAAAGTCGAACTGTTGCCAGAAAATAGCGAGTTT

AAACCAATTGTCGTAGATCTTCGTCAGCAGAGCTTCACCATTGAAGG

GCTGGCGGTTGGGGTTATTCGCAACGGCGACTGGCTGGAATTCatgacca

aactggacaagggcaccgtgatcgcggcggcgctagagctgttgaacgaggttggcatggacagcctgacgac

gcggaagctcgctgaacgcctcaaggttcagcagcctgcgctttactggcatttccagaacaagcgagcgctgctt

gatgcgctcgccgaggcgatgctggcggaacgccatacccgctcgctacccgaagagaatgaggactggcggg

tgttcctgaaagagaatgccctgagcttcagaacggcgttgctctcttatcgggacggcgcgcgtatccatgccgg

cactcgaccgacagaaccgaattttggcaccgccgagacgcaaatacgctttctctgcgcggagggcttttgtccg

aagcgcgccgtttgggcgctccgggcggtcagtcactatgtggtcggttccgttctcgagcagcaggcatctgatg

ccgatgagagagttccggacaggccagatgtgtccgagcaagcaccgtcgtccttcctgcacgatctgtttcacga

gttggaaacagacggcatggatgctgcgttcaacttcggactcgacagcctcatcgctggtttcgagcggctgcgtt

catctacaacagattagGATCCGTCGACCATGGCGGCCGCTCGAGTCGACCTGC

AGCCAAGCTAATTCCGGGCGAATTTCTTATGATTTATGATTTTTATTA

TTAAATAAGTTATAAAAAAAATAAGTGTATACAAATTTTAAAGTGAC

TCTTAGGTTTTAAAACGAAAATTCTTATTCTTGAGTAACT

TetR(H) –

HL-249-6

GCTGCGCGTCAGCGGGATGTCGATGAAAGATATCGGCATTATGGATG

GTGACTTGCTGGCAGTGCATAAAACTCAGGATGTACGTAACGGTCAG

GTCGTTGTCGCACGTATTGATGACGAGGTTACCGTTAAGCGCCTGAA

AAAACAGGGCAATAAAGTCGAACTGTTGCCAGAAAATAGCGAGTTT

AAACCAATTGTCGTAGATCTTCGTCAGCAGAGCTTCACCATTGAAGG

GCTGGCGGTTGGGGTTATTCGCAACGGCGACTGGCTGGAATTCatggcaa

agctagataaagaacaagttattgatgatgcgttgattttacttaatgaagttggtattgaaggattaacaacgcgtaac

gtggcgcaaaaaataggtgtggaacaacccacattgtattggcatgtaaaaaataaacgcgctttgttagatgcatta

gcagaaactattttgcaaaagcaccatcatcatgttttgccattgccgaatgaaacatggcaggactttttgcgaaata

acgcgaaaagcttccgccaagccttattaatgtatcgtgatggtggcaaaattcatgcgggaacacgcccctctgaa

agtcaatttgagacatcagaacagcaactacagtttttgtgtgatgctgggtttagtctatctcaagccgtgtatgcatt

aagctctattgcgcattttacattaggctccgtactggaaactcaagagcatcaagaaagccaaaaagagcgtgaaa

aagtagagacggatactgttgcctatccgccattattaacccaagccgttgcaattatggatagtgataatggtgatg

ctgcatttttgtttgtccttgatgtgatgatctctggacttgaaacagtattaaagagcgctaaatagGATCCGTC

GACCATGGCGGCCGCTCGAGTCGACCTGCAGCCAAGCTAATTCCGGG

CGAATTTCTTATGATTTATGATTTTTATTATTAAATAAGTTATAAAAA

AAATAAGTGTATACAAATTTTAAAGTGACTCTTAGGTTTTAAAACGA

AAATTCTTATTCTTGAGTAACT

211

5.8 References

1. Fasciotti M. Perspectives for the use of biotechnology in green chemistry applied to

biopolymers, fuels and organic synthesis: from concepts to a critical point of view. Sustainable

Chem Pharm. 2017;6:82-89. doi: 10.1016/j.scp.2017.09.002.

2. Keasling JD. Manufacturing Molecules Through Metabolic Engineering. Science.

2010;330(6009):1355-1358. doi: 10.1126/science.1193990.

3. Chao R, Yuan Y, Zhao H. Recent advances in DNA assembly technologies. FEMS Yeast

Res. 2014;15(1):1-9. doi: 10.1111/1567-1364.12171.

4. Kosuri S, Church GM. Large-scale de novo DNA synthesis: technologies and applications.

Nat Methods. 2014;11(5):499-507. doi: 10.1038/nmeth.2918.

5. Jullesson D, David F, Pfleger B, Nielsen J. Impact of synthetic biology and metabolic

engineering on industrial production of fine chemicals. Biotechnol Adv. 2015;33(7):1395-1402.

doi: 10.1016/j.biotechadv.2015.02.011.

6. Richardson SM, Mitchell LA, Stracquadanio G, Yang K, Dymond JS, DiCarlo JE, Lee D,

Huang CLV, Chandrasegaran S, Cai Y, Boeke JD, Bader JS. Design of a synthetic yeast genome.

Science. 2017;355(6329):1040-1044. doi: 10.1126/science.aaf4557.

7. Wang HH, Isaacs FJ, Carr PA, Sun ZZ, Xu G, Forest CR, Church GM. Programming cells

by multiplex genome engineering and accelerated evolution. Nature. 2009;460(7257):894-898.

doi: 10.1038/nature08187.

8. Doudna JA, Charpentier E. The new frontier of genome engineering with CRISPR-Cas9.

Science. 2014;346(6213):1258096-1258096. doi: 10.1126/science.1258096.

9. Rogers JK, Taylor ND, Church GM. Biosensor-based engineering of biosynthetic

pathways. Curr Opin Biotechnol. 2016;42:84-91. doi: 10.1016/j.copbio.2016.03.005.

10. Schallmey M, Frunzke J, Eggeling L, Marienhagen J. Looking for the pick of the bunch:

high-throughput screening of producing microorganisms with biosensors. Curr Opin Biotechnol.

2014;26:148-154. doi: 10.1016/j.copbio.2014.01.005.

212

11. Feng J, Jester BW, Tinberg CE, Mandell DJ, Antunes MS, Chari R, Morey KJ, Rios X,

Medford JI, Church GM, Fields S, Baker D. A general strategy to construct small molecule

biosensors in eukaryotes. eLife. 2015;4. doi: 10.7554/eLife.10606.

12. Licitra EJ, Liu JO. A three-hybrid system for detecting small ligand-protein receptor

interactions. Proc Natl Acad Sci U S A. 1996;93(23):12817-12821. doi:

10.1073/pnas.93.23.12817.

13. Belshaw PJ, Ho SN, Crabtree GR, Schreiber SL. Controlling protein association and

subcellular localization with a synthetic ligand that induces heterodimerization of proteins. Proc

Natl Acad Sci U S A. 1996;93(10):4604-4607. doi: 10.1073/pnas.93.10.4604.

14. Golemis EA, Serebriiskii I, Finley RL, Kolonin MG, Gyuris J, Brent R. Interaction

Trap/Two-Hybrid System to Identify Interacting Proteins. Curr Protoc Cell Biol.

2008;53(1):17.13.11-17.13.35. doi: 10.1002/0471142727.mb2001s82.

15. Bernstein DS, Buter N, Stumpf C, Wickens M. Analyzing mRNA-protein complexes using

a yeast three-hybrid system. Methods. 2002;26(2):123-141. doi: 10.1016/S1046-2023(02)00015-

4.

16. Henthorn DC, Jaxa-Chamiec AA, Meldrum E. A GAL4-based yeast three-hybrid system

for the identification of small molecule-target protein interactions. Biochem Pharmacol.

2002;63(9):1619-1628. doi: 10.1016/S0006-2952(02)00884-5.

17. Baker K, Bleczinski C, Lin H, Salazar-Jimenez G, Sengupta D, Krane S, Cornish VW.

Chemical complementation: a reaction-independent genetic assay for enzyme catalysis. Proc Natl

Acad Sci U S A. 2002;99(26):16537-16542. doi: 10.1073/pnas.262420099.

18. Fields S, Song OK. A Novel Genetic System to Detect Protein Protein Interactions. Nature.

1989;340(6230):245-246. doi: DOI 10.1038/340245a0.

19. Morita T, Takegawa K. A simple and efficient procedure for transformation of

Schizosaccharomyces pombe. Yeast. 2004;21(8):613-617. doi: 10.1002/yea.1104.

20. Gibson DG, Young L, Chuang RY, Venter JC, Hutchison CA, 3rd, Smith HO. Enzymatic

assembly of DNA molecules up to several hundred kilobases. Nat Methods. 2009;6(5):343-345.

doi: 10.1038/nmeth.1318.

213

21. Green SR, Moehle CM. Media and Culture of Yeast. Curr Protoc Cell Biol.

2001;4(1):1.6.1-1.6.12. doi: 10.1002/0471143030.cb0106s04.

22. Krishnan L, Sum P-e, Daigneault S, Bernatchez M, Pilcher AS, Horne JM, Tuper AJ,

Mccauley III JJ, Michaud AP, inventorsMethods of purifying tigecycline patent U.S. Patent

2007/0049561 A1. 2006 March 1, 2017.

23. Nagy A, Szoke B, Schally AV. Selective Coupling of Methotrexate to Peptide-Hormone

Carriers through a Gamma-Carboxamide Linkage of Its Glutamic-Acid Moiety - Benzotriazol-1-

Yloxytris(Dimethylamino)Phosphonium Hexafluorophosphate Activation in Salt Coupling. Proc

Natl Acad Sci U S A. 1993;90(13):6373-6376. doi: 10.1073/pnas.90.13.6373.

24. Castex U, Lalanne C, Mouchet P, Lemaire M, Lahana R. Regioselective synthesis of

peptidic derivatives and glycolamidic esters of methotrexate. Tetrahedron. 2005;61(4):803-812.

doi: 10.1016/j.tet.2004.11.049.

25. Reichheld SE, Davidson AR. Two-way interdomain signal transduction in tetracycline

repressor. J Mol Biol. 2006;361(2):382-389. doi: 10.1016/j.jmb.2006.06.035.

26. Reichheld SE, Yu Z, Davidson AR. The induction of folding cooperativity by ligand

binding drives the allosteric response of tetracycline repressor. Proc Natl Acad Sci U S A.

2009;106(52):22263-22268. doi: 10.1073/pnas.0911566106.

27. Abida WM, Carter BT, Althoff EA, Lin H, Cornish VW. Receptor-dependence of the

transcription read-out in a small-molecule three-hybrid system. ChemBioChem. 2002;3(9):887-

895. doi: 10.1002/1439-7633(20020902)3:9<887::AID-CBIC887>3.0.CO;2-F.

28. Harton MD, Wingler LM, Cornish VW. Transcriptional regulation improves the

throughput of three-hybrid counter selections in Saccharomyces cerevisiae. Biotechnol J.

2013;8(12):1485-1491. doi: 10.1002/biot.201300186.

29. Keppler-Ross S, Noffz C, Dean N. A New Purple Fluorescent Color Marker for Genetic

Studies in Saccharomyces cerevisiae and Candida albicans. Genetics. 2008;179(1):705-710. doi:

10.1534/genetics.108.087080.

30. Li YR, Chooi YH, Sheng YW, Valentine JS, Tang Y. Comparative Characterization of

Fungal Anthracenone and Naphthacenedione Biosynthetic Pathways Reveals an alpha-

Hydroxylation-Dependent Claisen-like Cyclization Catalyzed by a Dimanganese Thioesterase. J

Am Chem Soc. 2011;133(39):15773-15785. doi: 10.1021/ja206906d.

214

31. Ostrov N, Jimenez M, Billerbeck S, Brisbois J, Matragrano J, Ager A, Cornish VW. A

modular yeast biosensor for low-cost point-of-care pathogen detection. Science Advances.

2017;3(6):e1603221. doi: 10.1126/sciadv.1603221.

32. ICH Expert Working Group, editor. Validation of analytical procedures: text and

methodology Q2 (R1). International Conference on Harmonization, Geneva, Switzerland; 2005.

33. Monser L, Darghouth F. Rapid liquid chromatographic method for simultaneous

determination of tetracyclines antibiotics and 6-Epi-doxycycline in pharmaceutical products using

porous graphitic carbon column. J Pharm Biomed Anal. 2000;23(2-3):353-362. doi:

10.1016/s0731-7085(00)00329-0.

34. Ruden DM, Ma J, Li Y, Wood K, Ptashne M. Generating Yeast Transcriptional Activators

Containing No Yeast Protein Sequences. Nature. 1991;350(6315):250-252. doi:

10.1038/350250a0.

35. Lin H, Abida WM, Sauer RT, Cornish VW. Dexamethasone−Methotrexate:  An Efficient

Chemical Inducer of Protein Dimerization In Vivo. J Am Chem Soc. 2000;122(17):4247-4248.

doi: 10.1021/ja9941532.

36. Gyuris J, Golemis E, Chertkov H, Brent R. Cdi1, a human G1 and S phase protein

phosphatase that associates with Cdk2. Cell. 1993;75(4):791-803. doi: 10.1016/0092-

8674(93)90498-F.

37. Lederer T, Kintrup M, Takahashi M, Sum PE, Ellestad GA, Hillen W. Tetracycline analogs

affecting binding to Tn10-Encoded Tet repressor trigger the same mechanism of induction.

Biochemistry. 1996;35(23):7439-7446. doi: 10.1021/bi952683e.

38. Hinrichs W, Kisker C, Duvel M, Muller A, Tovar K, Hillen W, Saenger W. Structure of

the Tet repressor-tetracycline complex and regulation of antibiotic resistance. Science.

1994;264(5157):418-420. doi: 10.1126/science.8153629.

39. Saenger W, Hinrichs W, Orth P, Schnappinger D, Hillen W. Structural basis of gene

regulation by the tetracycline inducible Tet repressor–operator system. Nat Struct Biol.

2000;7(3):215-219. doi: 10.1038/73324.

40. Waters SH, Rogowsky P, Grinsted J, Altenbuchner J, Schmitt R. The tetracycline

resistance determinants of RP1 and Tnl721: nucleotide sequence analysis. Nucleic Acids Res.

1983;11(17):6089-6105. doi: 10.1093/nar/11.17.6089.

215

41. Rhodes G, Parkhill J, Bird C, Ambrose K, Jones MC, Huys G, Swings J, Pickup RW.

Complete Nucleotide Sequence of the Conjugative Tetracycline Resistance Plasmid pFBAOT6, a

Member of a Group of IncU Plasmids with Global Ubiquity. Appl Environ Microbiol.

2004;70(12):7497-7510. doi: 10.1128/aem.70.12.7497-7510.2004.

42. Brow MAD, Pesin R, Sutcliffe JG. The Tetracycline Repressor of Psc101. Mol Biol Evol.

1985;2(1):1-12. doi: 10.1093/oxfordjournals.molbev.a040334.

43. Bernardi A, Bernardi F. Complete sequence of pSC101. Nucleic Acids Res.

1984;12(24):9415-9426. doi: 10.1093/nar/12.24.9415.

44. Tovar K, Ernst A, Hillen W. Identification and Nucleotide-Sequence of the Class-E Tet

Regulatory Elements and Operator and Inducer Binding of the Encoded Purified Tet Repressor.

Mol Gen Genet. 1988;215(1):76-80. doi: 10.1007/Bf00331306.

45. Gao F, Webb HE, Bugarel M, den Bakker HC, Nightingale KK, Granier SA, Scott HM,

Loneragan GH. Carbapenem-Resistant Bacteria Recovered from Faeces of Dairy Cattle in the

High Plains Region of the USA. PLoS One. 2016;11(1):e0147363. doi:

10.1371/journal.pone.0147363.

46. Liu P, Li P, Jiang X, Bi D, Xie Y, Tai C, Deng Z, Rajakumar K, Ou HY. Complete Genome

Sequence of Klebsiella pneumoniae subsp. pneumoniae HS11286, a Multidrug-Resistant Strain

Isolated from Human Sputum. J Bacteriol. 2012;194(7):1841-1842. doi: 10.1128/jb.00043-12.

47. Hansen LM, Mcmurry LM, Levy SB, Hirsh DC. A New Tetracycline Resistance

Determinant, Tet H, from Pasteurella-Multocida Specifying Active Efflux of Tetracycline.

Antimicrob Agents Chemother. 1993;37(12):2699-2705. doi: 10.1128/Aac.37.12.2699.

48. Kehrenberg C, Tham NTT, Schwarz S. New Plasmid-Borne Antibiotic Resistance Gene

Cluster in Pasteurella multocida. Antimicrob Agents Chemother. 2003;47(9):2978-2980. doi:

10.1128/aac.47.9.2978-2980.2003.

49. Mendez B, Tachibana C, Levy SB. Heterogeneity of tetracycline resistance determinants.

Plasmid. 1980;3(2):99-108. doi: 10.1016/0147-619x(80)90101-8.

50. Hillen W, Berens C. Mechanisms Underlying Expression of TN10 Encoded Tetracycline

Resistance. Annu Rev Microbiol. 1994;48(1):345-369. doi:

10.1146/annurev.mi.48.100194.002021.

216

51. Mohana-Borges R, Pacheco ABF, Sousa FJR, Foguel D, Almeida DF, Silva JL. LexA

Repressor Forms Stable Dimers in Solution. J Biol Chem. 2000;275(7):4708-4712. doi:

10.1074/jbc.275.7.4708.

52. Kisker C, Hinrichs W, Tovar K, Hillen W, Saenger W. The Complex Formed between Tet

Repressor and Tetracycline-Mg2+ Reveals Mechanism of Antibiotic-Resistance. J Mol Biol.

1995;247(2):260-280. doi: 10.1006/jmbi.1994.0138.

53. Greer ND. Tigecycline (Tygacil): the first in the glycylcycline class of antibiotics. Proc

(Bayl Univ Med Cent). 2006;19(2):155-161. doi: 10.1080/08998280.2006.11928154.

54. Liu F, Myers AG. Development of a platform for the discovery and practical synthesis of

new tetracycline antibiotics. Curr Opin Chem Biol. 2016;32:48-57. doi:

10.1016/j.cbpa.2016.03.011.

55. Rogalski W. Chemical Modification of the Tetracyclines. In: Hlavka JJ, Boothe JH, editors.

The Tetracyclines. Berlin, Heidelberg: Springer Berlin Heidelberg; 1985. p. 179-316.

56. Zhang X, Bremer H. Control of the Escherichia coli rrnB P1 promoter strength by ppGpp.

J Biol Chem. 1995;270(19):11181-11189. doi: 10.1074/jbc.270.19.11181.

57. Kogawa AC, Salgado HRN. Quantification of Doxycycline Hyclate in Tablets by HPLC–

UV Method. J Chromatogr Sci. 2013;51(10):919-925. doi: 10.1093/chromsci/bms190.

58. Ng YZ, Baldera-Aguayo PA, Cornish VW. Fluorescence Polarization Assay for Small

Molecule Screening of FK506 Biosynthesized in 96-Well Microtiter Plates. Biochemistry.

2017;56(40):5260-5268. doi: 10.1021/acs.biochem.7b00602.

59. Gossen M, Bujard H. Tight Control of Gene-Expression in Mammalian-Cells by

Tetracycline-Responsive Promoters. Proc Natl Acad Sci U S A. 1992;89(12):5547-5551. doi:

10.1073/pnas.89.12.5547.

60. Scholz O, Kostner M, Reich M, Gastiger S, Hillen W. Teaching TetR to recognize a new

inducer. J Mol Biol. 2003;329(2):217-227. doi: 10.1016/S0022-2836(03)00427-3.

61. Ramos JL, Martinez-Bueno M, Molina-Henares AJ, Teran W, Watanabe K, Zhang X,

Gallegos MT, Brennan R, Tobes R. The TetR Family of Transcriptional Repressors. Microbiol

Mol Biol Rev. 2005;69(2):326-356. doi: 10.1128/mmbr.69.2.326-356.2005.

217

62. Lin H, Tao H, Cornish VW. Directed evolution of a glycosynthase via chemical

complementation. J Am Chem Soc. 2004;126(46):15051-15059. doi: 10.1021/ja046238v.

63. Peralta-Yahya P, Carter BT, Lin H, Tao H, Cornish VW. High-throughput selection for

cellulase catalysts using chemical complementation. J Am Chem Soc. 2008;130(51):17446-17452.

doi: 10.1021/ja8055744.

64. Ma SM, Li JWH, Choi JW, Zhou H, Lee KKM, Moorthie VA, Xie X, Kealey JT, Da Silva

NA, Vederas JC, Tang Y. Complete Reconstitution of a Highly Reducing Iterative Polyketide

Synthase. Science. 2009;326(5952):589-592. doi: 10.1126/science.1175602.

65. Watson MA, Buckholz R, Weiner MP. Vectors encoding alternative antibiotic resistance

for use in the yeast two-hybrid system. BioTechniques. 1996;21(2):255-259. doi:

10.2144/96212st02.