selective ablation of transgenic neural precursor cells- a ......pgk: phosphoglycerate kinase...

155
Selective Ablation of Transgenic Neural Precursor Cells- A Cellular and Molecular Tool for Use in Neural Regeneration Studies by Christopher Edward Rodgers A thesis submitted in conformity with the requirements for the degree of Master of Science Institute of Medical Science University of Toronto © Copyright by Christopher Edward Rodgers 2018

Upload: others

Post on 22-Aug-2021

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

Selective Ablation of Transgenic Neural Precursor Cells- A Cellular and Molecular Tool for Use in Neural

Regeneration Studies

by

Christopher Edward Rodgers A thesis submitted in conformity with the requirements

for the degree of Master of Science

Institute of Medical Science University of Toronto

© Copyright by Christopher Edward Rodgers 2018

Page 2: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

ii

Selective Ablation of Transgenic Neural Precursor Cells- A

Cellular and Molecular Tool for Use in Neural Regeneration

Studies

Christopher Edward Rodgers

Master of Science

Institute of Medical Science

University of Toronto

2018

Abstract

Induced pluripotent-stem cell derived-neural precursor cell (iPSC-NPC) transplantation

represents a promising potential therapy for repair and recovery of the devastating condition of

spinal cord injury (SCI). Three critical questions on NPC transplantation are (1) "Which neural

cell type provides optimal regeneration?" (2) "Is long term cell integration critically necessary

for recovery or only short term trophic support/immunomodulation?" and (3) “How much

proliferation of transplanted iPSC-NPCs is required for neuroregeneration?” Selective cell

ablation, using enzyme-prodrug systems, such as herpes-simplex virus thymidine kinase (HSV-

TK) and ganciclovir (GCV) or brivudine (BVDU), or nitroreductase (NTR) and metronidazole

(Mtz), is a cellular/molecular tool that may assist in answering the above questions via targeted

ablation of transplanted cells in SCI-model rodents and assessment of repair/recovery. This

project demonstrates successful creation (through transposon gene transfer) and the first

GCV/BVDU-mediated in vitro ablation (>80% reduction over controls) of HSV-TK-expressing

transgenic human iPSC-NPCs in an SCI transplantation context.

Page 3: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

iii

Acknowledgments

I wish to express special and particular recognition for my close family members and relatives

who provided constant loving support: my mother, Marilyn Snell; my father, David Rodgers; my

sister, Laura Rodgers; and my grandmothers, Stella Rodgers and Jean Peters.

Additional acknowledgements include, but are not limited to:

My committee members- Dr. Cindi Morshead and Dr. Andras Nagy: For their continued

thoughtful guidance and nurturing support

My supervisor- Dr. Michael Fehlings: For providing the opportunity, environment, and stipend

financial support to conduct my MSc research

My laboratory mentors- Dr. Mahmood Chamankhah, for his exceptional tutelage in conducting

my molecular biology subcloning experiments; and Dr. Mohamad Khazaei for his mentorship in

relation to cell culture techniques.

Page 4: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

iv

Table of Contents

Acknowledgments.......................................................................................................................... iii

Table of Contents ........................................................................................................................... iv

List of Abbreviations .................................................................................................................... vii

List of Figures ................................................................................................................................xv

List of Tables .............................................................................................................................. xvii

List of Appendices ..................................................................................................................... xviii

Chapter 1 : Literature Review ..........................................................................................................1

Traumatic Spinal Cord Injury .....................................................................................................1

1.1 Background and Definitions ................................................................................................1

1.1.1 Historical Background and Injury Phases ................................................................1

1.1.2 Temporal Phases of Injury .......................................................................................2

1.2 Mechanisms of Damage in SCI ...........................................................................................3

1.2.1 Demyelination ..........................................................................................................3

1.2.2 Neuronal Loss/Damage ............................................................................................4

Therapeutic Strategies: Neuroprotection and Neuroregeneration ...............................................7

2.1 Cell Transplantation Strategies ..........................................................................................10

2.1.1 Remyelination ........................................................................................................12

2.1.2 Neuronal Regeneration/Axonal-Synaptic Plasticity ..............................................14

Cell Ablation Strategies ............................................................................................................19

3.1 Ablation Studies .................................................................................................................19

3.1.1 Diptheria Toxin Receptor ......................................................................................20

3.1.2 Herpes-Simplex Virus Thymidine Kinase and Ganciclovir ..................................23

3.1.3 Herpes-Simplex Virus Thymidine Kinase and Brivudine .....................................27

3.1.4 E. coli Nitroreductase (NfSA/NfsB) and CB1954 .................................................30

3.1.5 E. coli Nitroreductase (NfsB/NTR) & Metronidazole (Mtz) .................................33

Page 5: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

v

3.2 Gene Transfer Vectors .......................................................................................................39

3.2.1 Lentiviral Vectors ..................................................................................................39

3.2.2 Transposon Vectors ...............................................................................................41

Chapter 2: Research Aims .............................................................................................................46

Overarching Goals, Questions, & Technical Aims ........................................................................46

Chapter 3: Materials and Methods .................................................................................................48

1) NTR Insert Subcloning: Lentivirus->PiggyBac ..................................................................48

2) NTR-NPC Monoclonal Lines and Mtz Ablations ...............................................................57

3) NTR-Mtz Ablation Troubleshooting ...................................................................................60

4) HSV-TK Plasmid DNA Purification and Confirmation ......................................................62

5) Mixed NTR and HSV-TK NPC GCV Ablation ..................................................................63

6) HSV-TK-NPC GCV and BVDU Bystander Ablations .......................................................64

7) HSV-TK-NPC GCV and BVDU Quantitative Ablations ....................................................65

Chapter 4: Results ..........................................................................................................................67

1) NTR-PiggyBac Subcloning .................................................................................................67

2) Mtz Treatment of CMV-NTR NPCs ...................................................................................76

3) NTR-Mtz Ablation System Troubleshooting ......................................................................77

4) HSV-TK Plasmids and the Creation of HSV-TK NPCs .....................................................79

5) GCV Bystander Effect: Mixed HSV-TK & HCO-NTR-NPCs ...........................................81

6) GCV/BVDU Bystander Effects: Mixed HSV-TK & WT-NPCs .........................................82

7) GCV and BVDU Successfully Ablate HSV-TK hiPSC-NPCs ...........................................84

Chapter 5: General Discussion, Future Directions & Conclusion .................................................89

5.1 Summary of HSV-TK+GCV/BVDU Results .....................................................................89

5.2 HSV-TK+GCV/BVDU Ablation in Context ......................................................................90

5.3 Advantages and Challenges of HSV-TK+GCV/BVDU Ablation ......................................91

5.4 Reflections on NTR-Mtz Ablation Approaches ..................................................................93

Page 6: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

vi

5.5 Potential Applications and Future Directions of HSV-Tk+GCV/BVDU hiPSC-

Derived NPC Ablation .......................................................................................................96

5.6 Conclusion ........................................................................................................................101

References ....................................................................................................................................102

1.1 102

1.1.1 102

1.1.2 104

1.2 104

1.2.1 104

1.2.2 105

2 106

2.1 107

2.1.1 108

2.1.2 110

3.1 114

3.1.1 114

3.1.2 116

3.1.3 121

3.1.4 123

3.1.5 127

3.2 130

3.2.1 130

3.2.2 131

Chapter 3 - 4) ..........................................................................................................................135

Chapter 3 - 7) ..........................................................................................................................136

Appendix I: List of Contributions ................................................................................................137

Page 7: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

vii

List of Abbreviations

(In Alphabetical Order)

1O2: singlet oxygen

a.a.: amino acid

AcMNPV: Autographica californica nuclear polyhedrosis virus

adpNSC: adult primitive neural stem cell (GFAP-, Oct-4+)

AIF: apoptosis-inducing factor

ANOVA: analysis of variance (statistical test)

AP: action potential

ASIC: acid-sensing ion channel

Bcl-2: B-cell lymphoma 2

Bcl-xL: B-cell lymphoma-extra large

BDNF: brain-derived neurotrophic factor

BFGF/FGF-2: basic fibroblast growth factor

bp: base pair

BVDU: (E)-5-(2-bromovinyl)-2'-deoxyuridine/brivudine

C11-C14: CMV-NTR-piggybac clones 11-14

CB1954: 5-(aziridin-1-yl)-2,4-dinitrobenzamide (NTR ablation cancer prodrug)

CD95: cluster of differentiation 95

chABC: chondroitinase ABC/chondroitin ABC-lyase

Page 8: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

viii

cIAP2/BIRC3: cellular inhibitor of apoptosis protein 2/baculoviral IAP repeat-containing

protein3

CMV-NTR-NPC: ubiquitious promoter-driven-nitroreductase (E. coli NfsB)-expressing neural

precursor cells

CNS: central nervous system

CNTF: ciliary neurotrophic factor

Cre: Causes recombination/Cyclicization recombination

CSPG: Chondroitin-sulphate proteoglycan

CST: cortical spinal tract

Cx43: connexin 43

D5: DCX-NTR-piggyBac clone 5

DAPI: 4′,6-diamidino-2-phenylindole

DCs: dendritic cells

DCX: doublecortin

dCyd: 2'-deoxycytidine

DDH2O: Double-Distilled Water

DISC: death-inducing signalling complex

DMSO: dimethyl sulfoxide

dNSC: definitive neural stem cell (GFAP+, Oct-4-)

dNTP: deoxynucleotide triphosphate

DT: diptheria toxin

Page 9: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

ix

dThd: deoxythymidine

DTR: diptheria toxin receptor

dUrd: 2(1)-deoxyuridine

EAA: extracellular amino acid

ECM: extracellular matrix

EDTA: ethylenediaminetetraacetic acid

EGF: epidermal growth factor

EM: electron microscopy

ER: endoplasmic reticulum

ESC: embryonic stem cell

EtOH: Ethanol

FADD: Fas-associated death domain protein

FAF1: Fas-associated factor 1

Fas/FasR: first apoptosis signal/first apoptosis signal receptor

FBS: fetal bovine serum

FMN: flavin mononucleotide

FP: few polyhedral

gag: group antigens lentiviral gene

Gal-1: galectin-1

GCV: ganciclovir = 9-[[2-hydroxy-1-(hydroxymethyl)ethoxy]methyl] guanine

Page 10: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

x

GDEPT: Gene-directed enzyme-prodrug therapy

GDNF: glial-derived neurotrophic factor

GFAP: glial fibrillary-acidic protein

GFP: green fluorescent protein

GJIC: gap junction intercellular channel

hat: hobo/Ac/Tam3 transposon family

HB-EGF: heparin-binding EGF-like growth factor

HCO-NTR: human-codon-optimized nitroreductase/NfsB

HCO-NTR-NPCs: Human-codon-optimized-NPCs (expressing NTR under the ubiquitous CMV

promoter)

hCTL: human cytoxic T lymphocyte

HEK-293T: human embryonic kidney 293T cell line

HeLa cells: Henrietta Lacks’ cells

hiPSC: human induced pluripotent stem cell

HIV-1: human immunodeficiency virus 1

hpf: hours post-fertilization

HSV-1: human herpes virus 1

HSV-TK/HSV-TK 1: herpes-simplex virus thymidine kinase 1

•HO: hydroxyl radical

ID2: inhibitor of DNA-binding protein two

ID50/IC50: 50% inhibitory dose/concentration (of viral plaque formation)

Page 11: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

xi

iDTR: inducible diptheria toxin receptor

IL-1: interleukin 1

Il-18: interleukin 18

IL-1β: interleukin 1 Beta

IL-6: interleukin 6

INS: preproinsulin gene

IP3: inositol trisphosphate receptor-type 3

iPSC: induced pluripotent stem cell

IR: internal repeat

ITR: inverted terminal repeat

JAK/STAT3: janus kinase/signal transducers and activators of transcription-3

KA: kainite

kb: kilo-base pairs

LB: Lennox-broth

LCs: Langerhans cells

LF: lateral funiculus

LINES: long-interspersed nuclear elements

LTR: long terminal repeat

M1-M5: MB-NTR-piggyBac clones 1-5

MAG: myelin-associated glycoprotein

Page 12: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

xii

MAPK10/JNK3: mitogen-activated protein kinase 10/C-Jun-N-terminal kinase 3

MAPK8/JNK1: mitogen-activated protein kinase 8/C-Jun-N-terminal kinase 1

MBP: myelin basic protein

MGE: medial ganglionic eminence

MSC: mesenchymal stem/stromal cell

MTT: 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide

Mtz: metronidazole

NAD (NAD+/NADH = oxidized vs reduced forms): nicotinamide adenine dinucleotide

NADPH: nicotinamide adenine dinucleotide phosphate

nef: negative factor

NG2: neuron-glial antigen 2

NPC: neural precursor cell (heterogenous mixture of neural progenitor and neural stem cells)

NRP-1: neuropilin-1

NSC: neural stem cell (“upstream” of NPC in differentiation/development)

NTR: Nitroreductase (E. Coli NfsA/NfsB)

NTR-NPC: nitroreductase (E. coli NfsB)-expressing neural precursor cells

OEC: olfactory ensheathing cell

ONOO−: peroxynitrite

OPC: oligodendrocyte progenitor cell

OPI: overproduction inhibition

Page 13: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

xiii

P/S: penicillin-streptomycin mixture

p53/TRP53: transformation/tumour-suppressor-related protein 53

PARP1: Poly [ADP-ribose] polymerase 1

PDGF-A: platelet-derived growth factor subunit A

PFA: paraformaldehyde

PFT-μ: pifithrin-μ

PGK: phosphoglycerate kinase

PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

pNSC: primitive neural stem cell (GFAP-, Oct-4+)

pol: reverse transcriptase and integrase lentiviral gene

PTEN: Phosphatase and tensin homolog

PTK: puromycin-resistance-HSV-TK fusion protein gene

RCF: relative centrifugal force (often referred to as “g”s)

RCL: replication competent lentivirus

RFP: red fluorescent-protein

RIPA: radioimmunoprecipitation assay

RRE: Rev response element

SAPs: self-assembling peptides

SB: Sleeping Beauty transposon

SCI: spinal cord injury

Page 14: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

xiv

SDS-PAGE: sodium-dodecyl-sulfate polyacrylamide gel electrophoresis

Sema3A: semaphorin 3A

SF9: serum free media 9 (components described in Chapter 3: Materials and Methods,

Section 2)

Shh: Sonic Hedgehog

SIN: Self-inactivating

SINES: short-interspersed nuclear elements

1O2: singlet oxygen

•O2-: superoxide

tat: trans-activator lentiviral gene

TE: transposable element

TNF-α: tumor necrosis factor alpha

TUNEL: Terminal deoxynucleotidyl transferase dUTP nick end labeling

vif: viral infectivity

VLF: ventrolateral funiculus

vpr: virus protein R

vpu: virus protein U

VSV-G: vesicular stomatitis Indiana virus G-protein

WT: wild-type

WT-NPC: wild-type hiPSC-derived NPC (no transgene)

Page 15: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

xv

List of Figures

Figure C1-I: An Overview of SCI Pathophysiology (pg. 6)

Figure 1a: CMV-NTR Lentiviral Plasmid and Subcloning Insert Gene Maps (pg. 68)

Figure 1b: DCX-NTR Lentiviral Plasmid and Subcloning Insert Gene Maps (pg. 69)

Figure 1c: MBP-NTR Lentiviral Plasmid and Subcloning Insert Gene Maps (pg. 70)

Figure 1d: PiggyBac Transposon Plasmid Vector Gene Map (pg. 71)

Figure 1e: CMV-NTR-PiggyBac Subcloned Plasmid (pg. 72)

Figure 1f: DCX-NTR- PiggyBac Subcloned Plasmid (pg. 73)

Figure 1g: MBP-NTR- PiggyBac Subcloned Plasmid (pg. 74)

Figure 2: NTR Inserts/PiggyBac Transposon Agarose DNA Gel Restriction Digests (pg. 75)

Figure 3: CMV-NTR-NPC Mtz Ablation Attempts (pg. 76)

Figure 4: CMV-NTR-NPC Coomassie Blue Protein Stain (pg. 77)

Figure 5: HCO-NTR-NPC Mtz Ablation Attempts (pg. 78)

Figure 6a: pCMV-Tol2 Plasmid Gene Map (pg. 79)

Figure 6b: pKTol2P-PTK Plasmid Gene Map (pg. 80)

Figure 7: HSV-TK + GCV Bystander Effect with HSV-TK and HCO-NTR NPCs (pg. 81)

Figure 8: HSV-TK + GCV Bystander Effect with HSV-TK and WT-GFP NPCs (pg. 82)

Figure 9: HSV-TK + BVDU Bystander Effect with HSV-TK and WT-GFP NPCs (pg. 83)

Figure 10: Evaluation of GCV ablational efficiency of HSV-TK NPCs (pg. 84)

Figure 11: Evaluation of BVDU ablational efficiency of HSV-TK NPCs (pg. 85)

Figure 12: The Comparison of GCV & BVDU ablational efficiency of HSV-TK NPCs (pg. 86)

Page 16: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

xvi

Figure 13: The Evaluation of DMSO treatment of HSV-TK NPCs (pg. 88)

Page 17: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

xvii

List of Tables

Table 1: An Overview of key therapeutic neuroprotective and neuroregenerative strategies for

SCI (pg. 9)

Table 2: An Overview of Cell Ablation Systems (pg. 38)

Page 18: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

xviii

List of Appendices

Appendix I: List of Contributions: (pg. 137)

Page 19: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

1

Chapter 1 : Literature Review

Traumatic Spinal Cord Injury

1.1 Background and Definitions

1.1.1 Historical Background and Injury Phases

The vertebrate spinal cord, or medulla spinalis as it was once referred to, which in humans spans

the cerebrospinal axis from the medulla oblongata to the lowest lumbar vertebra, has long been

known to provide connections between the brain and the peripheral nervous system, including

the motor, sensory, and autonomic nerves as evidenced by forms of paralysis and loss of bladder

function accompanying injury to the cord (Barlow, 1832; Wilson, 1911; Budd, 1839; Ott, 1879;

Nichols, 1899; Pollock et al., 1951; Garstang and Miller-Smith, 2007; Stanley, 1833; Thorburn,

1888). Spinal cord injury (SCI) is defined simply as any damage causing a temporary or

permanent alteration in the cord’s function and can be divided into two principal classes of injury

- namely traumatic injuries encompassing all physical external sources (falls, motor-vehicle

accidents, projectile-wounds, etc.) and non-traumatic injuries encompassing internal disease

sources (vertebral disc degeneration, cancerous tumour, etc.) (reviewed in Ahuja et al.,

2017a&b). With regards to traumatic spinal cord injury, there exist two main phases: primary

and secondary injury (Tator and Fehlings, 1991).

The primary phase of traumatic SCI contains the initial physical damage from such forces as

laceration, shearing, and often most prominently, compression/contusion and is notable

specifically for marking the injury severity (Tator, 1995; Tator and Fehlings, 1991; reviewed in

Ackery et al., 2004). Depending on trauma severity, nerve fiber neuronal axons are either

demyelinated or disintegrated entirely (McDonald, 1975). Subsequently, a secondary phase of

injury, defined as a “delayed and progressive tissue energy” is generated consisting of a cascade

cycle of neuronal and glia cell death as well as inflammation and ischemia (reviewed in Ahuja et

al., 2017a&b). Secondary injury is known to involve both a buildup of extracellular amino acids

(EAAs), particularly glutamate and aspartate that alter neural and glial metabolism, and are

correlated with apoptosis, and inflammation, as well as a disruption of the blood-spinal cord

barrier involving the recruitment of cells of the immune system-including neutrophils,

Page 20: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

2

macrophages/microglia and mast cells, which produce pro-inflammatory cytokines, such as

interleukin 6 (IL-6), interleukin 1-Beta (IL-1β), and tumor necrosis factor alpha (TNF-α)

(Hachem et al., 2017; Saghazadeh and Rezaei, 2017; reviewed in Ahuja et al., 2017a&b). The

levels of pro-inflammatory cytokines have been shown to reach a climax between six to twelve

hours post-injury and plateau for as long as an additional four days (Nakamura et al., 2003;

Ulndreaj et al., 2016).

1.1.2 Temporal Phases of Injury

The time length post injury determines the injury phase, of which in current classifications exist

four: acute, sub-acute, intermediate, and chronic, which occur at periods of under forty-eight

hours, forty-eight hours to fourteen days, fourteen days to six months, and beyond six months

respectively (reviewed in Ahuja et al., 2017a&b). During the acute phase, the aforementioned

secondary injury begins to occur as the disruption of the blood-spinal cord barrier from the initial

primary insult produces swelling and hemorrhage as well as vascular disruption and permits the

entry of immune cells and subsequent production of proinflammatory, as well as pro-apoptotic

factors (reviewed in Ahuja and Fehlings, 2016). As a result of the initial inflammation produced

in the acute phase, degradation and death of neurons and oligodendrocytes occur, thus inducing

both demylination and reduced neuronal signaling (reviewed in Ahuja and Fehlings, 2016).

For the twelve days of the subacute phase, arterial spasms, and resultant vasoconstriction, in

addition to vesicular clotting, and fluid-buildup swelling occur, leading to further ischemia,

cutting off vital oxygen and glucose to neurons and glia, further promoting necrosis (reviewed in

Ahuja and Fehlings, 2016; reviewed in Ahuja et al., 2017a&b). The effects of microglia and

other immune cells at the two-week mark post-injury (late subacute-early intermediate phase)

additionally include extracellular damage, leading to the development of cystic microcavities

(reviewed in Ahuja and Fehlings, 2016; reviewed in Ahuja et al., 2017a&b). Further secondary

injury damage and regeneration barriers occur from the result of rapid astrocytic proliferation

proximal to the lesion, as they secrete both proinflammatory factors and extracellular matrix

(ECM) molecules known as chondroitin sulphate proteoglycans (CSPGs) (reviewed in Ahuja

and Fehlings, 2016; reviewed in Fehlings et al., 2017; Rhodes & Fawcett, 2004). The CSPGS

and perilesional astrocytes eventually produce a structure known as the glial scar, which

continues to mature throughout the intermediate and into the chronic SCI phase and presents a

Page 21: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

3

critical barrier to neural repair and regeneration (reviewed in Ahuja and Fehlings, 2016;

reviewed in Ahuja et al., 2017a&b). Apoptosis in SCI secondary injury is further promoted due

to an excessive cellular calcium buildup occurring due to loss of ionic homeostasis (Schanne et

al., 1979). These overly high calcium levels activate calcium-dependent proteases, and

negatively impact mitochondria, culminating in programmed cell death (Schanne et al., 1979).

As time progresses to the intermediate phase (two weeks), there occurs a continuation of axonal

death and a coalescence of cystic microcavities to form a larger macrocavity bounded by the

astrocytes and CSPGs of the glial scar, which significantly impedes the regrowth of neuronal

axons, remyelinating projections of oligodendrocytes, and neuronal/glial cell migration to

replace injured or destroyed cells (reviewed in Ahuja and Fehlings, 2016; reviewed in Ahuja et

al., 2017a&b). Despite the hurdles to regeneration created through the maturation of the glial

scar, there is however evidence of some beneficial properties of the scar existing, including the

sealing and protection of the blood-spinal cord barrier, partially-sequestering inflammation, and

preventing further increase of lesion volume (Saghazadeh and Rezaei, 2017).

1.2 Mechanisms of Damage in SCI

1.2.1 Demyelination

The loss and degradation of myelin sheaths coating neuronal axons ranks highly among the most

critical neurological damage conditions induced by SCI (Huang et al., 2014; Wu and Ren, 2009).

Demyelination in SCI is understood to be largely a result of oligodendrocyte apoptosis, which is

believed to be initiated through cellular and biochemical pathways involving the organelles of

the mitochondria and endoplasmic reticulum (ER) of oligodendrocytes (Huang et al., 2014).

Evidence for ER and mitochondrial involvement in oligodendrocyte apoptosis post-SCI can also

be inferred from similar interaction that occur in other forms of central nervous system (CNS)

demyelination, such as that occurring from intracerebral hemorrhage (Zhuo et al., 2016).

In observations of oligodendrocyte death taking place after SCI, evidence of the occurrence of

apoptotic events has been demonstrated from detection of upregulated expression of the proteins

caspase-12, known to promote activation of the inflammatory cytokines interleukin 1 (IL-1) and

interleukin 18 (IL-18), and cytochrome C, known to stimulate apoptosis through binding with

the inositol trisphosphate receptor-type 3 (IP3) receptor in the ER, and inducing the release of

Page 22: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

4

stored excess intracellular calcium (Huang et al., 2014; Fisher et al., 2002; Liu et al., 1996). One

major regulator of oligodendrocyte apoptosis has been hypothesized to be the inhibitor of DNA-

binding protein two (ID2), as significant upregulations of ID2 that increase with time post-injury

have been detected in the white matter in rodent models of compressive SCI (Huang et al.,

2014). As ID2 is also thought to be involved in suppressing cellular differentiation, it is possible

that ID2 may also act to prevent oligodendrocyte progenitor cell (OPC) differentiation and

inhibit remyelination during SCI secondary injury (Hara et al., 1994).

In addition to ID2, upregulation of the transformation/tumour-suppressor-related protein 53

(p53/TRP53) appears to play a critical role in oligodendrocyte apoptosis, and thus demyelination

in SCI (Ma et al., 2017). Levels of the transcription factor, E2F1, itself understood to be involved

in both cell cycle control and tumor suppression, have been shown to be upregulated along with

caspase-12 and cytochrome C during oligodendrocyte apoptosis (Ma et al., 2017; Stevens and La

Thangue, 2005). E2F1 expression appears to be partly regulated by p53, as demonstrated by

experiments involving the treatment of SCI-model rats with the p53 chemical inhibitor, pifithrin-

μ(PFT-μ), which resulted in both genetic/protein-based changes: the suppression of caspase-12,

cytochrome C and E2F1, along with physiological improvements: the increase in prevalence,

thickness, and length of myelin sheaths of spared axons (Ma et al., 2017). Demyelination clearly

represents a formidable, yet hopefully not insurmountable challenge along the road to repair and

recovery in traumatic SCI.

1.2.2 Neuronal Loss/Damage

The damage to and loss of neurons following traumatic SCI is an issue of serious pathological

consideration (reviewed in Ahuja et al., 2017a&b). The marked increase of extracellular

glutamate is known to be one of the factors of neuronal excitotoxicity and resultant apoptosis

(Mazzone et al., 2017; reviewed in Ahuja et al., 2017a&b). Glutamate excitotoxicity in spinal

neurons has been shown to be a slow and gradual process, with a correlation between increasing

cell loss of grey matter motor and premotor spinal neurons with increasing concentrations of the

administered glutamate analog kainite (KA; between 0.01mM-0.05mM and beyond) (Mazzone

et al., 2010).

Recent work has provided insight into the mechanism of extracellular glutamate-related neuronal

loss/damage, showing that the glutamate surge may activate acid-sensing ion channels (ASICs)

Page 23: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

5

on SCI neurons, through an extracellular pH reduction (Mazzone et al., 2017). Treatment of

mouse spinal cord slices with KA at a concentration of 0.01mM, right at the threshold for

excitotoxic damage, has been demonstrated to increase gene expression of the ASICs: ASIC1a,

ASIC1b, ASIC2 and ASIC3, whereas a treatment with a ten-fold increase in KA concentration

actually reduced gene expression of the ASICs: ASIC1a and ASIC2 (Mazzone et al., 2017).

Taken together, the aforementioned results would appear to indicate that the excessive buildup of

extracellular glutamate in traumatic SCI secondary injury may facilitate a reduction in the

excitoxicity threshold of spinal neurons, through downregulation of ASIC expression, and thus

make them more susceptible to undergo excitotoxic apoptosis in the presence of high levels

(>0.01mM) of extracellular glutamate (Mazzone et al., 2017).

Oxidative stress presents an additional mechanism behind the neuronal loss exhibited in cases of

traumatic SCI, as it does in other forms of CNS damage/degeneration (reviewed in Smith et al.,

2013). Reactive oxygen species, including such molecules as hydroxyl radicals (•HO),

peroxynitrite (ONOO−), superoxide (•O2-), and singlet oxygen (1O2), which are naturally

produced during cellular metabolism of oxygen, rise in concentration as part of the secondary

injury cascade of traumatic SCI (as similarly occurs in other forms of CNS traumatic injury), and

induce cellular damage/death of spinal neurons through such mechanisms as lipid peroxidation

of the cell membrane, DNA damage, and protein oxidation (reviewed in Smith et al., 2013;

reviewed in Catala, 2009; Ansari et al., 2008).

Necrosis, or delayed cell death of neurons is generally more prevalent than apoptosis as a result

of oxidative stress in traumatic SCI, and acts in a “spreading” manner leading to necrosis in

neighbouring neurons (Muthaiah et al., 2017). In necrosis, it is understood that the DNA binding

protein, Poly [ADP-ribose] polymerase 1 (PARP1) is upregulated, and recent research has

discovered that the cellular enzymes mitogen-activated protein kinase 8/C-Jun-N-terminal kinase

1 (MAPK8/JNK1) and mitogen-activated protein kinase 10/C-Jun-N-terminal kinase 3

(MAPK10/JNK3) are involved in PARP1-induced neuronal necrosis (Muthaiah et al., 2017).

It was previously reported that Fas-associated factor 1 (FAF1) stimulates PARP1 expression, via

reactive oxygen species interaction with FAF1 resulting in its transport to the nucleus and

catalytic induction of PARP1 gene expression (Yu et al., 2016). In addition to its presence

promoting PARP1 expression and necrosis, FAF1 depletion was observed to prevent the

Page 24: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

6

downstream PARP1-mediated necrotic event of energetic collapse (e.g., mitochondrial

depolarization and nuclear translocation of apoptosis-inducing factor (AIF)) (Yu et al., 2016). In

order to provide neural repair for traumatic SCI, and the resultant neuronal damage and loss from

secondary injury, mitigating or reducing necrosis due to oxidative stress would appear to be a

potentially beneficial, and efficient strategy.

Figure C1-I: An Overview of SCI Pathophysiology. SCI consists of an initial physical trauma

(primary injury, shown in a) on the right-middle), followed by an extended damaging process of

secondary injury that is subdivided into 4 temporal phases post-injury (a-c). The acute phase (a): 0-

48hrs) involves vesicular damage and blood-spinal cord barrier disruption resulting in hemorrhage,

ischemia, edema, as well as the infiltration of immune cells (e.g., neutrophils, monocytes, etc.), and

apoptotic/necrotic neuronal and glial cell death (leading to demyelination and axonal

severing/degeneration). During the subacute phase (b): 48hrs-14 days), demyelination, axonal

degeneration, and neuroinflammation (e.g., macrophage maturation from monocytes) continue and

the processes of cavity formation and reactive astrogliosis begin with the first deposition of CSPGs.

Throughout the intermediate phase (c): 14 days- 6 months) microcavities grow and coalesce as the

lesion matures to form a central macro-cavity bounded by a glial scar composed of astrocytes and

CSPGs. By the chronic phase (c): 6 months and beyond), glial scar maturation and lesion

stabilization occur, in addition to cyst formation due to clearance of microglial debris, and the gradual

removal of damaged neuronal axons and soma.

Image reproduced with permission from: Ahuja et al., 2017b. Neurosurgery. PMID: 28350947

C1-I

a) b) c)

Page 25: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

7

Therapeutic Strategies: Neuroprotection and Neuroregeneration

Two major strategies towards pursuing treatment for traumatic SCI are neuroprotection, defined

as “the attenuation of pathophysiological processes triggered by acute injury to minimize

secondary damage,” and neuroregeneration, which alternatively seeks to repair and replace

damaged neural tissue and provide functional recovery (reviewed in Hilton et al., 2017; reviewed

in Ahuja and Fehlings, 2016). The three major neuroprotective treatments currently in use

clinically are: surgical decompression, methylprednisolone (or other neuroprotective agents)

administration, and blood pressure augmentation (reviewed in Ahuja et al., 2017). There are

additionally several experimental treatments currently being explored (reviewed in Ahuja et al.,

2017b; reviewed in Hilton et al., 2017).

Although the strategy of neuroprotection has and likely will continue to provide strong

therapeutic benefit for reducing secondary injury damage in traumatic SCI, its main drawback

and limitation lies in the simple fact that it can only preserve neural tissue, and not repair or

restore damaged or lost tissue, nor provide full functional recovery (reviewed in Hilton et al.,

2017; reviewed in Ahuja et al., 2017b; reviewed in Ahuja and Fehlings, 2016; reviewed in Ahuja

et al., 2016). While neuroprotection aims to mitigate the neuronal and glial damage contributing

to sensory and motor loss in traumatic SCI, neuroregeneration proposes to replace damaged or

dead neurons and oligodendrocytes and/or provide restored neuronal connectivity and synaptic

activity through outgrowth, rewiring, and remyelination of existing axons (reviewed in Hilton et

al., 2017; reviewed in Ahuja et al., 2017b; reviewed in Ahuja and Fehlings, 2016; reviewed in

Ahuja et al., 2016; reviewed in O’Donovan, 2016).

Three critical neuroregenerative strategies for traumatic SCI being investigated in animal models

include (1) remyelination of surviving neuronal axons, (2) neuronal plasticity enhancement

through neuronal replacement and/or rewiring of existing axons and synaptic connections, and

(3) glial scar degradation/modification (reviewed in Ahuja et al., 2017b; reviewed in Ahuja and

Fehlings, 2016; reviewed in Ahuja et al., 2016).

Page 26: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

8

Cell replacement for neurorepair can involve either endogenous activation (mobilization,

migration/transportation, and differentiation) of resident neural stem/progenitor (the mixture

hereafter referred to as neural precursor cells (NPCs)) cells, or transplantation of exogenous

NPCs or OPCs (for purely remyelination based approaches) (reviewed in Ahuja et al., 2016;

reviewed in Franklin, 2015; Najm et al., 2015). With regards to exogenous NPC/OPC

transplantation, it is additionally not currently understood whether short-term trophic support

from transplanted-cell-secreted factors/molecules/proteins etc., or long-term integration of

transplanted cells represent the more critical mechanism of regeneration, and thus the more

efficient approach for neurorepair (reviewed in Ahuja et al., 2017b; reviewed in Ahuja and

Fehlings, 2016; reviewed in Ahuja et al., 2016). The precise amount and required time length of

transplanted cell (e.g., NPC, OPC, etc.) proliferation (e.g., how long must proliferation

occur/what cell quantity is necessary for providing physical repair/functional recovery?) is

similarly not entirely understood.

Page 27: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

9

A) Neuroprotection B) Neuroregeneration

Pharmacological treatment:

-methylprednisolone (currently used)

-riluzole (in clinical trial)

-minocycline (in clinical trial)

-glyburide (in clinical trial)

Endogenous cell (NPC/OPC) activation:

-Olig2 upregulation

-galectin-1 administration

-cyclic AMP

-rho-associated protein kinase inhibition

-Nogo-A inhibition

Surgical decompression (currently used)

Exogenous NPC-based cell transplantation-

remyelination and/or axonal regeneration:

-NPCs

-OPCs

-neuronal precursors

Moderate hypothermia: -reducing systemic body

temperature to 330C by intravascular cooling

strategies (in clinical trial)

Exogenous cell transplantation- additional cell types:

-olfactory-ensheathing cells

-Schwann cells

-mesenchymal stromal cells

Blood pressure-modulation

(currently used)

Glial Scar modification-

-chABC treatment

-neuron-glial antigen 2 knockdown

Table 1: An Overview of key therapeutic neuroprotective and neuroregenerative

strategies for SCI. While many neuroprotective options (A) are currently employed clinically

or are undergoing/have undergone clinical trials and produce modest sensorimotor benefit,

they are incapable of providing full physical repair and functional recovery. A plethora of pre-

clinical neuroregenerative strategies (B) are currently under investigation in animal models

with widely varying, yet greatly promising results.

Page 28: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

10

2.1 Cell Transplantation Strategies

Cell transplantation therapies for traumatic SCI represent a promising option in order to promote

neuronal/axonal regeneration and improved plasticity and synapse formation (reviewed in Nori

et al., 2017). Two critical goals in cell transplantation-based strategies for SCI repair include: (1)

remyelination of injured axons; and, (2) axonal/neuronal regeneration and synaptic

restoration/rewiring (reviewed in Nori et al., 2017; reviewed in Ahuja et al., 2017b).

The microenvironment niche of the injured spinal cord is understood to be a major obstacle for

cell-transplantation based-neuronal regeneration, both in terms of limiting exogenous cell

survival as well as reducing the differentiation of NPCs towards a neuronal lineage, making

axonal regeneration a more difficult task than remyelination with cell transplantation (reviewed

in Nori et al., 2017; reviewed in Ruff et al., 2012). The combination of cell transplantation (e.g.,

NPCs, OPCs, mesenchymal stromal cells, olfactory ensheathing cells, etc.) with

immunomodulation and trophic support, to ameliorate the injured cord microenvironment’s

permissiveness to cell integration and survival as well as promote remyelination, axonal

regeneration, synapse formation, collateral sprouting, and neurogenesis as well as lessen the

degradation of retrograde axons has been proposed and is currently under evaluation in animal

SCI models (reviewed in Ruff et al., 2012; reviewed in Liu et al., 2012; reviewed in Sun and He,

2010; reviewed in McKerracher, 2001).

An elusive major question, in addition to that of the debate of remyelination vs neuronal/axonal

regeneration and synaptic rewiring, that has remained unsolved as of yet, in regards to cell

transplantation-based therapies for promoting remyelination, axonal regeneration and plasticity

for traumatic SCI, is namely, which of the two mechanisms, (1) microenvironment modification

(e.g., immunomodulation) and trophic support, or (2) long-term survival, migration,

differentiation (in the case of NPCs) and integration of transplanted cells, represents the most

critical mechanism that can provide physiological regeneration/repair and functional recovery

(reviewed in Badner et al., 2017; reviewed in Iyer et al., 2017; reviewed in Ruff et al., 2012;

reviewed in Kim et al., 2007).

Page 29: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

11

The mechanisms of long-term transplanted cell survival and short-term support (trophic support,

immunomodulation, etc.) are heavily intertwined, wherein current studies cannot usually

separate either as being the primary mediator of reported physical repair (remyelination, axonal

regeneration, reduced astrogliosis, etc) and functional recovery (hindlimb motor performance,

BBB scores etc.) and, as such it is difficult to determine which of the two may provide the most

effective and efficient direction for therapeutic benefit and eventual translation (reviewed in

Bonner and Steward, 2015; reviewed in Lu et al., 2014; reviewed in Li and Lepski, 2013;

reviewed in Ruff et al., 2012; reviewed in Fehlings and Vawda, 2011).

Evidence specifically focusing on the importance of the long-term survival and integration (e.g.,

synaptogenesis etc.) for transplanted stem cells in mediating physical repair and functional

recovery in SCI animal models is generally scarce, but still has been demonstrated through such

experiments as those that displayed the survival of a majority of adult-human spinal-cord derived

neurospheres up to 6 weeks after transplantation in rats, correlating with behavioural

improvements (Akesson et al., 2007). An additional study reported survival of up to 8 weeks of

human autologous, reprogrammed, iPSC-derived NPCs transplanted into thoracic-injured mice,

as well as both their successful differentiation into neurons and glia, and functional integration

(synaptic-connections with surviving endogenous axons and remyelination), all without the

additional use of extra neurotrophic factors, biomaterials-scaffolding, or immune modulation

(Liu et al., 2017).

Additionally, for transplanted proliferating cells, such as human iPSC-derived NPCs, it is

uncertain precisely how much cell proliferation is necessary and for how long it must occur in

order to provide adequate and sufficient physical repair and functional recovery. Previous

research using a rat model of middle cerebral artery occlusion stroke has indicated that

transplanted human iPSC-derived NPCs continue to proliferate (as evidenced by nestin

expression) up to at least 2 weeks post transplantation and can improve neurobehavioural

outcomes, however it is unknown if shorter-term proliferation may be sufficient for providing

adequate neuroregeneration through trophic support and immunomodulation in CNS conditions

(Yuan et al., 2013).

Page 30: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

12

2.1.1 Remyelination

In order to reduce demyelination and promote remyelination after traumatic SCI, it may be

worthwhile to develop methods to replace dead oligodendrocytes through NPC or OPC

transplantation, as well as reduce/inhibit oligodendrocyte apoptosis, and promote endogenous

OPC differentiation.

In terms of promoting endogenous remyelination, the b-HLH transcription factor and prominent

oligodendrocyte marker, Olig2, known to be significant in mammalian CNS development, has

been identified as a strong candidate for promoting endogenous remyelination (Tan et al., 2017).

Via the use of lentiviral vector gene therapy, Olig2 overexpression was recently shown in a

rodent SCI model to increase differentiation of OPCs to mature oligodendrocytes, induce

remyelination, reverse gene expression changes of several transcription factors whose expression

is altered in secondary injury, and even provide significant improvement in hind limb motor

performance (Tan et al., 2017).

With regards to cell transplantation approaches for promoting remyelination, NPCs of either

adult (endogenous or induced pluripotent) or embryonic origin, present promising transplantation

cell types for neuroregeneration (reviewed in Ahuja et al., 2017b; reviewed in Ahuja and

Fehlings, 2016; reviewed in Ahuja et al., 2016; reviewed in Lu, 2017; reviewed in Lopez Juarez

et al., 2016; reviewed in Doulames, 2016). One previous study from over a decade ago

determined that NPC transplantations in combination with Sonic hedgehog (Shh) were capable

of promoting white matter-tract sparing and functional motor improvements (BBB scores and

motor evoked potentials) in thoracic (T9-T10) contusion injured rats (Bambakidis and Miller,

2004). The work of Bambakidis and Miller particularly found that the combinatorial treatment

provided improved survival, proliferation, and migration of transplanted NPCs.

In order to determine the most effective timepoint for cell transplantation, one set of rodent

model SCI experiments compared the transplantation of OPCs (pre-differentiated from human

NPCs) at either 7 days or 10 months, both of which permitted survival, migration, and

differentiation of OPCs; however only in the 1 week-post injury transplantation animals was

enhanced remyelination or improved motor performance observed (Keirstead et al., 2005).

Another study from the mid 2000’s compared transplantations of NPCs at 2 weeks (subacute

Page 31: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

13

phase) and 8 weeks (chronic phase) post-T7 23-gram clip compression injury (Karimi-

Abdolrezaee et al., 2006).

While the above study found that 2 week-post injury transplanted NPCs showed strong survival,

proliferation, migration (>5mm rostrally and caudally of injection point), and differentiation-

prominently into OPCs and subsequently myelinating and myelin basic protein (MBP)-

expressing oligodendrocytes, 10 week-post injury injected NPCs faired relatively poorly in terms

of survival in comparison (Karimi-Abdolrezaee et al., 2006). In addition to NPC survival,

proliferation, migration, and differentiation, Karimi-Abdolrezaee et al. demonstrated axon

ensheathment and even improved functional recovery as detected by improved BBB scores, and

other behavioural grading systems (grid walk) for rats receiving 2-week post-SCI NPC

transplantions compared to vehicle controls.

Subsequent experiments with the same model showed that transplanted-NPC-derived-

oligodendrocytes could within 6 weeks, additionally provide such functional morphological

benefits as forming compact myelin and Nodes of Ranvier (detected via electron micrscopy

(EM)), primarily along white matter tracts and improving conduction speed of action potentials

(APs) on ensheathed axons (observed through electrophysiological probes) (Eftekharpour et al.,

2007). The additional combination of NPC transplantations with epidermal growth factor

(EGF), basic fibroblast growth factor (BFGF), Platelet-derived growth factor subunit A (PDGF-

A) and chondroitinase ABC (ChABC) was later shown to strongly improve survival and

migration of transplanted NPC-derived OPCs and oligodendrocytes (Karimi-Abdolrezaee et al.,

2010). By lentivirally-transducing NSCs with the gene, Olig2, a critical regulator of

oligodendrocyte development, after 7 weeks transplantation, one group was able to show

significantly improved migration towards the white matter, white matter sparing, lesion cavity

volume reduction, OPC proliferation, myelin sheath thickness, and hindlimb motor functional

recovery compared to control rats receiving either NPCs alone or pure vehicle controls (Hwang

et al., 2009).

Ciliary neurotrophic factor (CNTF) has been additionally used as a successful combinatorial

remyelination treatment in rodent constusive SCI models along with OPC transplantation (Cao et

al., 2010). In a study using transgenic-CNTF-expressing OPC transplantations, the

combinatorial treatment was shown to greatly improve: a) OPC survival, proliferation, and

Page 32: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

14

differentiation into oligodendrocytes; b) remyelination of axons at the lesion area near the

ventrolateral funiculus (VLF) or lateral funiculus (LF); c) motor-evoked potential recovery; and

d) hindlimb motor performance scores (Cao et al., 2010).

Two more recent studies that made use of either induced-pluripotent stem cell (iPSC)-derived

OPCs, or iPSC-derived NPCs for intraspinal cell transplantation post-contusive thoracic (T10)

SCI in rat models reported major improvements in: a) myelin formation and axon ensheathment;

b) electrophysiological improvements in axon conduction; and c) motor performance (BBB

scores etc.) when compared to vehicle control animals (Salewski et al., 2015; Kawabata et al.,

2016).

Critical among the results of Salewski et al. and Kawabata et al. was the demonstration of some

of the earliest evidence that iPSC-derived NPCs and OPCs can similarly provide successful and

efficient neuroregeneration through remyelination as well as functional motor recovery, just as

previous work proved was possible for NPCs and OPCs of embryonic stem cell (ESC) or

endogenous adult origins (reviewed in Khazaei, et al., 2017; Salewski et al., 2015; Kawabata et

al., 2016; reviewed in Lee-Kubli and Lu, 2015; reviewed in Khazaei et al., 2014). As powerful a

strategy for traumatic SCI neuroregeneration/neurorepair as remyelination may present itself to

be, it may still not be able to provide complete functional recovery, as it does not address the

issue of dead or damaged neuronal axons and destroyed synapses surrounding the lesion area

(reviewed in Ahuja et al., 2017a&b; reviewed in Ahuja et al., 2016). The restoration of full

motor, sensory and autonomic recovery may also require supporting neuronal plasticity- the

regrowth of axons/neurites and re-establishment of synaptic connections.

2.1.2 Neuronal Regeneration/Axonal-Synaptic Plasticity

The restoration of full motor, sensory and autonomic recovery for traumatic SCI may, in

addition to remyelination, require supporting neuronal plasticity- the regrowth or replacement of

dead/damaged neurons, the outgrowth of axons and re-establishment of synapses affected by the

injury (reviewed in Nori et al., 2017; reviewed in Ahuja et al., 2017a&b; reviewed in Ahuja and

Fehlings, 2016; reviewed in Ahuja et al., 2016). In order to accomplish such neuronal plasticity

recovery, a wide variety of strategies, from endogenous activation with administration of growth

factors/hormones, regulatory proteins, or stimulatory/inhibitory molecules, to cell transplantation

(NPCs, Schwann cells, mesenchymal stem/stromal cells (MSCs), olfactory ensheathing cells

Page 33: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

15

(OECs), etc.) with methods taken to promote neurogenesis, axonal regeneration, and both

trophic support, and the survival and integration of transplanted cells have been pursued

(reviewed in Nori et al., 2017; reviewed in Ruff et al., 2012; reviewed in Hu and Selzer 2017;

reviewed in Kwon et al., 2002).

Natural, endogenous neuronal regeneration after traumatic SCI is minimal and only occurs for

neurons farther away from the lesion, as has been recently modeled in rats to only occur as close

as 3-5mm away from the SCI lesion area, and thus methods to stimulate endogenous repair, or

transplant cells to replace neurons may be necessary to establish functional recovery (He and

Nan, 2016; reviewed in Ahuja et al., 2017b; reviewed in Bähr and Bonhoeffer, 1994). As myelin

debris, particularly in the form of myelin-associated glycoprotein (MAG) is known to induce

inhibitory effects upon neuronal regeneration, it may be particularly important to examine the

interactions occurring between treatments for neuronal regeneration and remyelination, as there

is some evidence that the latter may potentially alter the former, even possibly detrimentally

(reviewed in Rao and Pearse, 2016).

Various animal SCI model experiments have shown promise in terms of promoting axonal

regeneration and plasticity with transplanted cell types including MSCs, Schwann cells, and

NPCs (of ESC, iPSC, or adult endogenous origin), however of these cell types, only NPCs

possess the capacity to differentiate into all three main neural cell types (neurons,

oligodendrocytes, and astrocytes) and thus they will subsequently represent the primary focus for

this review section (reviewed in Nori et al., 2017; reviewed in Ruff et al., 2012; reviewed in Kim

et al., 2007; Yang et al., 2017; Liu et al., 2013; Ban et al., 2011; reviewed in Oliveri et al., 2014).

Despite the lack of full endogenous regeneration and functional recovery in human (and

mammalian in general) SCI, there is evidence that some endogenous repair does occur, both in

the form of reorganizing surviving axonal connections and synapses and the minor proliferation

of endogenous NPCs; thus one potential focus for cell transplantation based strategies for

neuronal/axonal regeneration and plasticity is to capitalize on and support these endogenous

mechanisms, through trophic support, scaffolding and microenvironment/immune modulation

(reviewed in Bareyre, 2008).

Research involving transgenic NPCs expressing a modified form of the rabies virus (TVA and

G-protein) for synaptic tracing provides perhaps one of the most comprehensive views of the

Page 34: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

16

possibilities of long-term synaptic integration of grafted NPC-derived neurons, as there is

evidence of synaptic input from not only corticospinal and reticulospinal neurons of the host

animal, but also from neurons of the cortex, dorsal root ganglion, and medulla oblongata (Adler

et al., 2017). Recent work using mouse ESC-derived NPC grafts to partially regenerate cortical

spinal tract (CST) neurons/axons and recreate functional excitatory synapses after C4 CST

lesions further support the key value of long-term NPC-transplant integration; however the

translational relevance of the study involved may be considered questionable, due to not using a

contusion or compression SCI model, and thus not completely modelling the inhibitory

microenvironment produced by compressive/contusive SCI (Kadoya et al., 2016; reviewed in

Ahuja et al., 2017a&b).

The specific benefits of survival and long-term integration of transplanted NPCs and derived

cells (addressed in Section 2.1 above) have perhaps been highlighted in greater depth in other

CNS-injury models, including recent mouse neuropathic pain model experiments that showed

survival and integration of large numbers of ESC-derived, medial ganglionic eminence (MGE)-

like interneuron precursors up to 6 months after transplantation into the T10-T11 spinal cord,

which not only differentiated largely in to GABA-ergic interneurons and received synaptic inputs

from endogenous axons (primary afferent and spinal cord neurons), but also promoted improved

bladder function, and reduced pain hypersensitivity (Fandel et al., 2016; Etlin et al., 2016;

Hwang et al., 2016; Bráz et al., 2012).

Although such neuropathic pain studies provide valuable insight into the regenerative benefits of

long-term integration of neuronal precursors into the injured spinal cord, stressing the significant

value of focusing efforts on the integration sub-strategy for CNS damage, they do not model

contusive SCI, particularly with its inhibitory inflamed microenvironment, and thus do not

indicate whether integration for other neuronal subtypes (e.g., motor neurons as opposed to

inhibitory interneurons) may be capable of providing similar reparative benefits and functional

recovery (e.g., hindlimb motor function) (reviewed in Ahuja et al., 2017a&b; Fandel et al.,

2016).

The significance of neurotrophic factors in supporting axonal regeneration in addition to other

forms of physical repair is evident from a marmoset SCI model study involving the

transplantation of transgenic NPCs expressing galectin-1 (Gal-1), which not only improved the

Page 35: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

17

survival and neuronal differentiation of transplanted NPCs, but also resulted in higher BBB

motor scores (Yamane et al., 2010). The mechanism for Gal-1 mediated axonal regeneration

was determined to occur through Gal-1’s selective binding to the neuropilin-1 (NRP-

1)/PlexinA4 receptor complex, blocking the interaction between semaphorin 3A (Sema3A) and

the NRP-1 receptor complex, and preventing downstream inhibitory signaling-primarily in the

form of excess hydrogen peroxide production and destabilization of F-actin (Quintá et al., 2014;

Quintá et al., 2016).

In a similar vein, there is compelling evidence that microenvironment modifications, such as: (1)

STAT-3 overexpression; (2) inhibition/suppression of the axon guidance molecules and

developmental attractants and repellants of ascending and descending axons respectively, Wnts

1, 4, and 5a (usually absent and known to be reintroduced in the injured cord milieu); (3)

phosphate and tensin homolog (PTEN) inhibition (demonstrated to promote CST axonal axonal

sprouting); (4) IL-6 activation (to stimulate janus kinase/signal transducers and activators of

transcription-3 (JAK/STAT3) and phosphoinositide 3-kinase/protein kinase B/mechanistic

target of rapamycin (PI3K/Akt/mTor); as well as (5) the use of biomaterial-based scaffolds,

including self-assembling peptides (SAPs) to support axonal regrowth may separately be

effective strategies for inducing axonal regeneration and improved plasticity after SCI (Mehta et

al., 2016; Luo et al., 2016; Leibinger et al., 2013a; Leibinger et al., 2013b; Liu et al., 2008; Du et

al., 2015; Liu et al., 2010; Zweckberger et al., 2016; Zweckberger et al., 2015).

Both the directions of (1) long-term survival, differentiation, and integration of transplanted

NPCs/NSCs, and (2) trophic support/scaffolding/immune modulation have shown strong

potential for promoting neuronal regeneration and plasticity in animal SCI models; however

further study may be necessary to separate and compare the differential benefits of each strategy

(reviewed in Nori et al., 2017; reviewed in Ruff et al., 2012; Badner et al., 2017; reviewed in Iyer

et al., 2017). A combination of both strategies, such as shown by studies combining the

transplantation of neuronal and glial-restricted progenitors (a stage between NPCs and mature

neurons/glia) to reconstitute excitatory and inhibitory neurons as well as oligodendrocytes, with

lentiviral-vector administration of brain-derived neurotrophic factor (BDNF) to guide axonal

regeneration in the desired direction (rostral or caudal), will likely be necessary in order to fully

re-establish axonal/neuronal regeneration and synaptic connections and functional motor and

Page 36: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

18

sensory recovery (reviewed in Nori et al., 2017; reviewed in Ruff et al., 2012; Bonner et al.,

2011; Bonner et al., 2010).

Furthermore, the issue of the timing and amount of transplanted NPC proliferation required in

order to provide effective physical repair (e.g., remyelination of axons, axonal regeneration or

neuronal replacement and rewiring of synapses, etc.) and functional sensorimotor recovery is

additionally uncertain. Although previous work in rodent CNS injury/condition (e.g., spinal cord

injury and stroke) models has shown functional behavioural improvements using transplanted

iPSC- or ESC-derived NPCs that continued to proliferate up to at least the 2-week post-

transplanation timepoint, what remains unclear is whether such neurological improvements

occurred largely due to trophic and immunomodulatory factor secretion and thus activation of

endogenous regeneration processes (e.g., central canal NPC proliferation, migration, and

differentiation), or actual cell replacement with the integration of the transplanted NPC-derived

neurons and glia themselves (Yuan et al., 2013; Salewski et al., 2013; Karimi-Abdolrezaee et al.,

2010; Kawabata et al., 2016; reviewed in Lee-Kubli and Lu, 2015).

Page 37: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

19

Cell Ablation Strategies

3.1 Ablation Studies

The concept of cell ablation represents a potential method for pursuing a better understanding of

the mechanisms of regeneration, as well as developmental roles of specific cells and their lineage

relationships over the course of development (reviewed in Curado et al., 2008). Pertaining to

SCI neurorepair/regeneration, cell ablation studies may be particularly attractive for use in

combination with cell transplantation, for the purpose of providing a method to conduct loss of

function experiments via selectively ablating grafted cells and comparing physical repair and

functional recovery with control animals not receiving ablations. As mentioned previously in

Section 2.1.2, three unanswered questions with regards to neural regeneration are:

1) “Is a) remyelination, or b) neuronal regeneration (neurite outgrowth)/axonal and synaptic

plasticity reestablishment the more effective and efficient strategy for SCI neurorepair (and

eventually pursuing translational therapeutic treatments)?”

2) “Is long-term integration of transplanted cells (e.g., establishment of synaptic connectivity

from transplanted NPC-derived neurons, or large-scale remyelination directly by transplanted

NPC/OPC-derived oligodendrocytes) necessary for providing significant physical repair and

functional recovery, or is the combination of the short-term mechanisms of: a) trophic support

(e.g., secretion of positive growth factors – BDNF/ glial-derived neurotrophic factor (GDNF)

etc.), and b) immune modulation (e.g., downregulating production of Il-6, IL-β, or TNF-α)

sufficient?”

and 3) “How much proliferation of transplanted hiPSC-NPCs is necessary and for how long must

it occur to provide effective SCI physical repair and functional recovery?” (reviewed in Ahuja et

al., 2017b; reviewed in Ahuja and Fehlings, 2016; reviewed in Siddiqui et al., 2015; reviewed in

Badner et al., 2017; reviewed in Khazaei et al., 2017).

Page 38: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

20

Studies centered around selective cell ablation of specific types of transplanted cells (e.g.,

neurons and oligodendrocytes), such as those making use of enzyme-prodrug combinations may

provide one avenue for answering, or at least partially clearing up the above questions. Possible

systems for cell ablation include: a) transgenic expression of the diptheria toxin receptor in

combination with diptheria toxin treatment, b) the herpes-simplex virus thymidine kinase-

ganciclovir/brivudine combination, and c) the E. coli nitroreductase (NfsB or NfsA genes)-

metronidazole or CB1954 combinations (reviewed in Curado et al., 2008).

3.1.1 Diptheria Toxin Receptor

Diptheria toxin (DT) is a 535-amino acid dipeptide exotoxin encoded by a bacteriophage

that infects Corynebacterium diphtheriae (Wagner et al., 2002; Pappenheimer 1977; Freeman,

1951). DT’s crystal structure has been determined to be that of a Y-shaped molecule, containing

three domains, that splits into two fragments: (1) fragment A possessing the functional catalytic

(C) domain that transfers an ADP ribose from nicotinamide adenine dinucleotide (NAD) to a

diphthamide residue of eukaryotic elongation factor 2 (eEF-2), thus blocking translation in

infected cells- resulting in the molecule’s toxicity in eukaryotes expressing the receptor; and (2)

fragment B possessing an endosomal translocation (T/TM) domain and a receptor binding (R)

domain on its carboxy-terminal (Bell and Eisenberg, 1997). The cell surface receptor for

diptheria toxin: heparin-binding EGF-like growth factor (HB-EGF), is expressed in humans, but

not in mice, thus rendering the exotoxin largely ineffective against endogenous mouse cells, as it

cannot normally be endocytosed (Saito et al., 2001; Cha et al. 2003). Through transgenic

expression of HB-EGF, the diphtheria toxin cell-surface receptor (DTR) in mouse cells, a

selective/cell-specific ablation system can be set up, whereby DTR-expressing mouse cells (from

embryonic genetic modification or transplantation) or transplanted human cells (naturally DTR-

expressing) can be selectively ablated with DT treatment of experimental mice (Saito et al.,

2001).

The DTR ablation system was first termed a "toxin receptor-mediated cell knockout” by Saito et

al., who used a hepatocyte-specific promoter to induce fulminant hepatitis in transgenic,

hepatocyte-DTR expressing mice (Saito et al., 2001). Saito et al. were able to display a

correlation between hepatocyte sensitivity to DT and transgenic DTR/HB-EGF expression, using

as many as three cell lines (Saito et al., 2001). An inducible variant of transgenic DTR

Page 39: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

21

expression (iDTR) was later developed using the Causes recombination/Cyclicization

recombination (Cre)-Lox recombination system, in which mice containing the HB-EGF gene

under the control of a ubiquitous promoter containing a transcriptional stop cassette, were

crossed with a cell-specific (CD4 T cell and CD19 B cell) Cre-expressing mouse line, thus

enabling cell specific removal of the stop cassette and cell-specific DTR/HB-EGF expression

(Buch et al., 2005).

For CD4 T-cell-DTR-expressing mice, Buch et al. demonstrated that 100ng daily injections of

DT over the course of 3 days was sufficient to reduce the mean splenic T:B cell ratio from 0.5 to

0.1 (thus ablating approximately 80% of splenic CD4-T cells); similarly for CD19-B cells, they

displayed that daily doses of 100ng DT reduced the splenic B:T cell ratio from 2.1 to 0.6 after 3

days, and 0.3 after 7 days, and finally 100ng DT doses provided every 8 hours for 7 days resulted

in near 100% ablation of CD19-B cells (Buch et al., 2005).

In regards to neural cell type ablation, Buch et al. not only reconfirmed that DT is capable of

crossing the BBB in mice, but also generated an oligodendrocyte-specific DTR-expressing

mouse line by crossing the ubiquitous-promoter DTR line with a MOG promoter-CRE mouse

line (Buch et al., 2005; Wrobel et al., 1990). Treatment of the MOG-iDTR mice with 100ng of

DT (peritoneally injected) thrice per day, over a course of 7 days consecutively, reportedly

induced: (1) massive destruction of cerebellar myelin (determined via immunohistochemical

staining); (2) axonal integrity losses; (3) inflammatory cell recruitment; and (4) mild reactive

astrogliosis (evidenced through glial fibrillary-acidic protein (GFAP) staining); as well as (5)

functional motor deficits including: hindlimb paralyses, tremors, and even body weight

reductions (Buch et al., 2005). Minor limitations of the study of Buch et al., may be that

researchers did not specifically quantify the approximate amount of oligodendrocyte ablation

with the MOG-iDTR line, and that they were unable to explicitly confirm that the apoptosis they

detected in cerebellar granular layers via fluorescein-12-terminal deoxynucleotidyl transferase

dUTP nick end labeling (TUNEL) staining was indeed from oligodendrocytes, and not granule

cells (Buch et al., 2005; Mathis et al., 2003).

Transgenic, cell-specific DTR expression has similarly shown promise as an effective ablation

model for other immune cells of mice, such as CD11c dendritic cells (DCs) and epidermal

Langerhans cells (LCs) (reviewed in Bennet and Clausen, 2007). More recently, the DTR

Page 40: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

22

ablation system has been used to target Pet-1 expressing, 5-HT serotonergic neurons in a mouse

model to study the role of 5-HT neurons in thermoregulation, and particularly to compare

functional deficits of adult loss of 5HT-neurons vs those observed in 5-HT developmental

knockout mice (Lmx1b(f/f/p)) (Cerpa et al., 2014). The above study reported that a total dose of

as low as 1.5-2µg of DT, administered systemically (50 μg/kg DT once a week for 2–3 weeks)

was sufficient to selectively ablate approximately 80% of 5-HT neurons in the ventrolateral

medulla and, the raphe nuclei of the medulla oblongata (Cerpa et al., 2014). One limitation of

the work of Cerpa et al., could arguably be that they were apparently unable to determine a DT

dosage and schedule that could effectively ablate approximately 99-100% of 5-HT neurons

without incurring unwanted side effects (sickliness, weight loss, mortality etc.), even in control

wild-type (WT) animals (Cerpa et al., 2014).

Despite the potential as a cell-specific suicide gene tool displayed by the DTR ablation system in

the aforementioned studies, the system is not without severe drawbacks and limitations, foremost

of which is its inherent species limitation. Most human and other primate cells express HB-EGF,

the cell-surface receptor for DT endocytosis, and thus the DTR ablation system cannot be used to

selectively target any specific primate cell type (Fen et al., 1993; Vaughan et al., 1992).

Additionally, there exists the possibility for minor bystander effects to occur with the use of DTR

ablation, as there has been evidence presented that the HB-EGF receptor may not be vital for the

entry of DT into, but may instead only vastly accelerate the rate of DT uptake via endocytosis

(10-50% toxin uptake/minute with the receptor vs. 1-2% uptake/minute without the receptor), as

the toxin can apparently still enter cells through interaction with clathrin-coated pits which

enable it to be endocytosed during a bulk turnover of cell-surface proteins (Almond and Eidels,

1994). Although the slower, alternative entry of DT may not initially appear entirely relevant,

only a single DT molecule is necessary to induce cell death, and thus the possibility for a

bystander effect with DTR ablation is nevertheless is a serious concern (Yamaizumi et al., 1978).

Finally, and most significantly for any researcher considering the use of the DTR ablation system

for selective cell-killing, the obvious issue of toxicity to the user in the event of any emergency

(accidental self injection etc.) is the most vital concern- as 0.1µg/kg body mass is fatal for a

human user (Pappenheimer et al., 1982).

Page 41: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

23

3.1.2 Herpes-Simplex Virus Thymidine Kinase and Ganciclovir

Gene-directed enzyme-prodrug therapies (GDEPTs) represent an umbrella category for a

variety of cell-ablation strategies (reviewed in Zhang et al., 2015). The combination of the

herpes-simplex virus thymidine kinase 1 (HSV-TK/HSV-TK 1), transgenically expressed in a

cell-type designated for ablation and one of its specific substrates- namely ganciclovir (GCV, or

9-[[2-hydroxy-1-(hydroxymethyl)ethoxy]methyl] guanine), a deoxyguanosine (purine

nucleoside) analog, has been demonstrated to be an effective method of targeted ablation, both in

anti-cancer and regeneration studies (reviewed in Zhang et al., 2015; Chou and Hong, 2014;

Ikeshima-Kataoka and Yuasa, 2008; Balzarini et al., 1993; Oliver et al., 1985; Nishiyama and

Rapp, 1979).

The mechanism of GCV-mediated cytotoxicity/ablation occurs in several steps: (1)

phosphorylation of GCV first to GCV-monophosphate by HSV-TK (or other viral thymidine

kinases, such as that of cytomegalovirus), and subsequently to GCV-triphosphate via

endogenous cellular kinases in two steps; (2) incorporation of GCV-triphosphate into cellular

DNA strands during replication, inducing chain termination- as GCV-triphosphate is a weak

substrate for elongation; and finally – (3) apoptosis, due to the aforementioned failure of DNA

replication (reviewed in Bagó et al., 2016; reviewed in Zhang et al., 2015; Balzarini et al., 1993;

Beltinger et al., 1999; Fischer et al., 2005). HSV-TK is understood to have approximately 1000-

fold greater affinity for the initial phosphorylation step than endogenous cellular thymidine

kinases, preventing general cytotoxicity in non-HSV-TK expressing cells at concentrations of

GCV lethal to cells expressing the viral thymidine kinase (reviewed in Bagó et al., 2016;

reviewed in Zhang et al., 2015; Balzarini et al., 1993).

Investigations into the precise apoptotic pathway involved in HSV-TK and ganciclovir mediated

ablation determined that the enzyme-prodrug combination first initiates expression of a cell death

receptor- “the first apoptosis signal receptor” (Fas/FasR; also referred to as cluster of

differentiation 95 (CD95)) and p53- which translocates CD95 to the cell outer-surface, where a

cell-death-inducing signalling complex (DISC), composed of Fas-associated death domain

protein (FADD) and caspase-8, is formed (Beltinger et al., 1999). The previous results showing

a p53 and CD95-dependent mechanism of apoptosis have been controversial, as further studies

demonstrated that in certain cell types, such as glioma cells, the HSV-TK and ganciclovir

Page 42: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

24

apoptotic process occurs independently of p53 or death receptor expression, and instead involves

the down-regulation of expression of B-cell lymphoma 2 (Bcl-2) proteins, known apoptotic

regulators, involved as part of a series of separate mitochondrial pathways (Fischer et al., 2005).

Perhaps the most critical drawback of the HSV-TK and GCV cell ablation system is the presence

of a significant “bystander effect,” referring to the lack of complete selectivity of the GDEPT to

only target HSV-TK-expressing cells for apoptosis (reviewed in van Dillen et al., 2002; reviewed

in Mesnil et al., 2000). While the presence/expression of the viral thymidine kinase is required

for significant phosphorylation of GCV to GCV-monophosphate, and initiation of the latter two

cellular-kinase mediated phosphorylations to produce the active, DNA replication-chain

termination and apoptosis-inducing form of GCV-triphosphate, apoptosis can still occur in cells

neighbouring HSV-TK expressing cells, via the diffusion of GCV-monophosphate (as well as

GCV-di and triphosphate to a lesser degree) across cell membranes, a process mediated by gap

junction intercellular channels (GJICs), particularly connexin 43 (Cx43) (Asklund et al., 2003;

Burrows et al., 2002; Dilber et al., 1997).

Such a bystander effect mechanism has shown not to be universally homogenous among cell

types/cell lines, as some cells, such as those of the rat colon, DHD/K12/TRb cell line, have been

demonstrated to retain a bystander effect even when cultured in lower densities without physical

contact, or when treated with blockers of GJICs, such as 18 alpha-glycyrrhetinic acid or 1-

octanol; the retention of a bystander effect in DHD/K12 cells has been suggested to occur due to

the presence of a soluble factor encapsulating GCV-triphosphate, allowing its diffusion through

neighbouring cells, independent of the presence of Cx43 or other GJICs (Princen et al., 1999).

The soluble factor that may allow a gap-junction independent bystander effect to occur for

certain cell types has been suggested to be apoptotic vesicles, as evidenced by electron

microscopy studies (Freeman et al., 1993). In the study of gliomas, dexamethasone has been

indicated as an additional inhibitor of the HSV-TK/GCV ablation system’s bystander effect, via

the downregulation of Cx43 expression, resulting in: (1) reduced GJIC-mediated cell-cell

communication; in addition to (2) growth inhibition; (3) incorporation of thymidine; and (3)

apoptotic cascade modulation- a process accomplished through reducing CD95 expression, and

increasing expression of the two anti-apoptotic proteins, cellular inhibitor of apoptosis protein

2/baculoviral IAP repeat-containing protein3 (cIAP2/BIRC3) and B-cell lymphoma-extra large

Page 43: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

25

(Bcl-xL), which interfere with caspases and prevent cytochrome c release respectively (Robe et

al., 2005).

Previous research has displayed ablation success with the HSV-TK and ganciclovir GDEPT for a

variety of cancer cell lines, including: murine lymphomas, murine mammary carcinomas, and

murine sarcomas (Moolten et al., 1990; Balzarini et al., 1993; Moolten, 1986). The range of

animal cancer models in which the HSV-TK and ganciclovir system has been employed further

range from leukemia, glioma, liver cancer, bladder cancer, oral cancer, and adenocarcinoma

(reviewed in Zhang et al., 2015; Bondanza et al., 2011; Ribot et al., 2011; Staquicini et al., 2011;

Kakinoki et al., 2010; Tang et al., 2009; Greish et al., 2010; Chen et al., 2010; Ambade et al.,

2010).

In regards to the ablation of neural cell types, the HSV-TK/GCV system has been utilized to

ablate rat glioma cells (C6 cell line), as well as murine cerebellar neural stems cells (C17.2 cell

line) (Li et al., 2005; Pu et al., 2011). For C17.2 cells, using the MTT colorimetric, cell-

metabolic activity and viability assay- wherein the activity of NAD(P)H-dependent cellular

oxidoreductase enzymes to reduce the tetrazolium dye, 3-(4,5-dimethylthiazol-2-yl)-2,5-

diphenyltetrazolium bromide (MTT), to the insoluble, purple-coloured formazan is employed as

a metric of cell viability, a final concentration of 1µg/mL of GCV was determined to be

sufficient to yield approximately 99% ablation of HSV-TK expressing C17.2 cells after a time

period of 7 days (Pu et al., 2011). In both the studies of C6, and C17.2 cells, a strong bystander

effect was detected, whereby mixed HSV-TK-expressing to endogenous (non-HSV-TK-

expressing) cell ratios of 1:16 were sufficient to yield ablation of approximately 25% of all cells

(Li et al., 2005; Pu et al., 2011).

A second study that evaluated HSV-TK/GCV ablation as well as bystander effect with C17.2

cells showed that 25µM was sufficient to kill ~95% of HSV-TK-expressing cells after 48hrs of

treatment, and when grown in mixed populations with two non HSV-TK-expressing, glioma cell

lines (U87-MG and LN-18), the bystander effects were identified to be cell-type dependent, with

one apoptotic HSV-TK+ C17.2 cell accompanying ten U87-MG cells, or five LN-18 cells (Uhl et

al., 2005). A third study involving the use of the HSV-TK/GCV ablation system showed nearly

100% death of human-ESC-derived, Ki67+ NPCs with 40µM (~10µg/mL) GCV treatment after

200hrs, and virtually no toxicity on control NPCs in a separate population (Tieng et al., 2016).

Page 44: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

26

The HSV-TK+GCV ablation system has additionally been previously utilized to selectively kill

murine astrocytes in the central, as well as peripheral and enteric nervous systems using a GFAP

promoter (Sofroniew et al., 1999; Bush et al., 1999; Bush et al., 1998). Through subcutaneous

osmotic minipump administration of 100mg/kg/day of GCV for a period of fourteen days, one

study demonstrated the selective ablation of GFAP-expressing astrocytes of the small intesntinal

jejunum and ileum and subsequent development of fulminant jejuno-ileitis (Bush et al., 1998). A

year later, Bush et al. similarly displayed the targeted cell-killing of reactive astrocytes in

forebrain-stab-injured mice, with a GCV dose of 100mg/kg/day administered for one week,

which similar to experiments attempting to prevent glial scar formation in SCI with STAT3

knockouts, led to increased immune cell (CD45+ leukocyte) infiltration, and blood-brain-barrier

repair failure, yet also increased neurite outgrowth (Bush et al., 1999; Herrmann et al., 2008).

In addition to astrocytes, definitive neural stem cells (dNSCs) have additionally been selectively

ablated with a GFAP promoter (Sachewsky et al., 2014; Imura et al., 2003; Morshead et al.,

2003). dNSCs, as separate from primitive, Oct-4-expressing, neural stem cells (pNSCs) were

identified partly through work that demonstrated that GCV treatment could selectively kill only

adult and postnatal, yet not early embryonic (pre-12.5 days) GFAP-expressing NSCs of the

murine CNS (Imura et al., 2003). Research from the same year as Imura et al. additionally used

GCV to ablate GFAP-expressing, dNSCs, and resultantly showed that while GFAP-expressing

dNSCs are present in the murine forebrain, they are not found in the retina (Morshead et al.,

2003).

A more recent study employing a similar, GFAP-promoter-driven HSV-TK transgenic mouse

model confirmed the presence of a rare population of adult, Oct-4-expressing (and GFAP-non-

expressing) pNSCs (AdpNSCs) in the mouse forebrain peri-ventricular region via the GCV-

mediated selective ablation of GFAP+ dNSCs (Sachewsky et al., 2014). Sachewsky et al.

additionally discovered that GCV can remain stable and still induce cell death of HSV-TK-

expressing, GFAP+, adult murine dNSCs as late as 10 days after an initial 16-hour treatment

(Sachewsky et al., 2014). In regards to SCI model usages, the HSV-TK+GCV ablation system

has been used in mice to successfully target both endogenous neuron-glial antigen 2+ (NG2+)

pericytes and OPCs, as well as endogenous GFAP+ reactive astrocytes (Hesp et al., 2018;

Faulkner et al., 2004).

Page 45: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

27

As the HSV-TK/GCV ablation system’s mechanism of inducing apoptosis requires incorporation

of GCV-triphosphate into replicating DNA to prevent chain-elongation, it would be natural to

assume at first glance that an additional limitation of the system would be an inability to target

all non-dividing, post-mitotic or senescent cells; however, such a limitation has been

demonstrated to be false (Tieng et al., 2016; Laberge et al., 2013; Beltinger et al., 2000).

Cellular and molecular studies have determined that the principal mechanism of driving

apoptosis in the HSV-TK/GCV combination is in fact the disruption of mitochondrial DNA

synthesis, rather than genomic/nuclear DNA synthesis, and as many post-mitotic cells, such as

mature neurons, still undergo mitochondrial turnover, they still remain potential targets for HSV-

TK/GCV (Tieng et al., 2016; Laberge et al., 2013; Beltinger et al., 2000). After the GCV-

triphosphate-mediated interruption of mitochondrial DNA replication, it has been demonstrated

that affected cells (e.g., GCV-treated, HSV-TK-expressing, senescent human fibroblasts) suffer

mitochondrial membrane integrity losses, followed by cytochrome c release from the

mitochondria into the cytosol, and eventually caspase activation and nuclear fragmentation

(Laberge et al., 2013; Beltinger et al., 2000).

The HSV-TK/GCV GDEPT has certainly demonstrated promise as an effective and efficient

method of cell ablation in vitro and in vivo, as mentioned previously in numerous animal models

of cancer, and a few regeneration models (reviewed in Zhang et al., 2015; Ikeshima-Kataoka and

Yuasa, 2008). The bystander effect inherent in HSV-TK/GCV ablation, useful in cancer-based

utilizations for the ability to target non-transfected tumour cells, remains its principal detractor

for studies of regeneration, in particular for traumatic SCI models to selectively target neurons or

oligodendrocytes derived from transplanted NPCs (reviewed in Zhang et al., 2015; reviewed in

Ahuja et al., 2017b).

3.1.3 Herpes-Simplex Virus Thymidine Kinase and Brivudine

One option for mitigating the aforementioned bystander effect in HSV-TK/GCV ablation, is to

pair the viral enzyme with an alternative prodrug, particularly a pyrimidine, rather than purine

analog (reviewed in Dachs et al., 2009; reviewed in Degrève et al., 1999). Pyrimidine analogs,

such as (E)-5-(2-bromovinyl)-2'-deoxyuridine, also known as brivudine (BVDU), unlike purine

analogs (e.g., GCV), require the activity of HSV-TK or another viral thymidine kinase for not

Page 46: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

28

only the first, but also the second phosphorylation step towards producing their active, cytotoxic

triphosphate form (reviewed in Dachs et al., 2009).

As Cx43 and other gap junctions are less permeable to diphosphate forms of pyrimidine (or

purine) analogs than to monophosphate forms, smaller quantities of the diphosphate form can

accumulate in neighbouring cells, resulting in a reduction in the number of neighbouring cells

that are able to accumulate the pyrimidine analog-triphosphate and undergo chain termination

during DNA replication and subsequently apoptose, than in respective neighbouring cells with

the use of a purine analog (reviewed in Dachs et al., 2009).

To compare the bystander effect of pyrimidine analogs, such as BVDU, with purine analogs,

such as GCV, one set of experiments using mixed populations of various ratios of HSV-TK+ and

HSV-TK- osteosarcoma cells found that: only by using a BVDU concentration of at least

250µM, one ablated HSV-TK+ cell was found to be capable of killing only one additional

neighbouring HSV-TK- cell; whereas even by using a GCV dose of at least 50µM, one ablated

HSV-TK+ cell was found to be capable of killing between seven to thirteen additional

neighbouring HSV-TK- cells (reviewed in Degrève et al., 1999). Such a reduced bystander

effect, although perhaps detrimental for some cancer-based uses, would appear on the surface to

make pyrimidine analog prodrugs, namely BVDU, sufficiently more useful for more selective

ablation approaches in SCI and other neuroregeneration studies.

The inhibitory effects of brivudine on the replication of human herpes simplex virus 1 (HSV-1)

have been understood since the 1980s, and were swiftly harnessed towards cell ablation methods

using transgenic/HSV-TK-expressing cells, primarily in cancer studies, such as those with

murine models of mammary carcinoma and, leukemia, in which brivudine was found to induce

significant apoptosis of HSV-TK-expressing cells at concentrations ranging from 5000-20,000

times below the equivalent requirements for wild type cytotoxicity (reviewed in De Clercq,

2005; reviewed in De Clercq, 2004; Balzarini et al., 1985a; Balzarini et al., 1985b; Balzarini et

al., 1985c; Balzarini et al., 1986; Balzarini et al., 1989; Balzarini et al., 1993).

Mechanistically, it has been elucidated that in contrast to purine analogs, such as GCV, the

primary target of BVDU in its cytotoxic effects on HSV-TK-expressing cells is thymidylate

synthase, rather than purely DNA incorporation during replication (Balzarini et al., 1987). One

ablation study involving murine mammary carcinoma cells transgenically expressing HSV-TK

Page 47: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

29

along with rigorous exploration of intracellular biochemical pathways demonstrated thymidylate

synthase to be the principal BVDU target via showing that: a) cytotoxic effects could be more

easily reversed via the addition of deoxythymidine (dThd) than 2(1)-deoxyuridine (dUrd); b)

BVDU had a greater effect on preventing dUrd incorporation than dThd incorporation into

replicating cellular DNA; and, c) BVDU exhibited a strong inhibitory effect on the pathway

involved in incorporating 2'-deoxycytidine (dCyd) into cellular DNA thymidylate (Balzarini et

al., 1987). In regards to apoptosis, it has been determined that BVDU initiates the process via

activation of the c-Jun/activator protein-1 and Fas ligand/caspase-8 pathway, as opposed to the

Fas-ligand-independent pathway initiated by GCV (reviewed in De Clercq, 2005; Tomicic et al.,

2003). Although not explicitly determined, it is hypothesized that BVDU, like GCV, also

activates the mitochondrial damage pathway involving a caspase-mediated cleavage of Bcl-2

(reviewed in De Clercq, 2005; Tomicic et al., 2003).

The optimal concentration of BVDU necessary to induce effective total cell ablation (arbitrarily

defined hereafter as ~90% in under 5 days) is not perfectly understood but is known to be cell

type dependent (Balzarini et al., 1985 (3)). For murine mammary carcinoma (FM3A) HSV-TK+

cells, the 50% inhibitory dose/concentration (of viral plaque formation- ID50/IC50 for tumor cells

inhibition) BVDU dose has been shown to be 0.5ng/mL, versus 11µg/mL for normal (non-HSV-

TK expressing) FM3A cells (Balzarini et al., 1985 (3)).

In contrast, the equivalent BVDU ID50 was found to be: (1) between 0.01-0.02µM (~0.00333-

~0.00666 µg/mL) for primary rabbit testes (PRT) cells; (2) between 0.02-0.06 µM (~0.00666-

~0.020 µg/mL) for MDA-MB-435 human breast carcinoma cells; (3) between 0.045-0.105 µM

(0.015-0.035 µg/mL) for rat gliosarcoma (9L) cells; and (4) as low as 0.01 µg/mL for baby

hamster kidney 21 (BHK-21) cells (as evaluated in a plaque reduction test of plated monolayers)

(Reefschläger et al., 1982; Grignet-Debrus et al., 2000; Field et al., 1984). For osteosarcoma

cells, previous work determined the IC50 for BVDU to be 0.035µM (0.012µg/mL) for HSV-TK-

expressing cells vs. ~900µM (~300µg/mL) for HSV-TK negative/normal tumor cells (reviewed

in Degrève et al., 1999).

BVDU uses in relation to neural cells appear to be rarer, however most principally it has shown

success in treating herpetic encephalitis, wherein infected neurons contain viral (as opposed to

transgenic, cellular-driven) HSV-TK expression (reviewed in Rosato and Leib, 2015; Wigdahl et

Page 48: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

30

al., 1984; Wigdahl et al., 1983). In one of the aforementioned studies of BVDU’s effects on

treating encephalitis, a dosage of 10µg/mL was shown to produce near 100% elimination of HSV

plaques after 3 days in human fetal DRG neurons (Wigdahl et al., 1984).

Similar to GCV, BVDU has most certainly shown potential as an efficient prodrug for inducing

cell ablation in combination with transgenic HSV-TK expression. Although it has often been

overshadowed by GCV in cancer research, due to its reduced bystander effect limiting its ability

to target neighbouring, non-HSV-TK-expressing tumor cells, this very same flaw could

theoretically represent a significant strength in regards to its use in studies of neuroregeneration,

such as targeting transplanted NPC-derived cells in traumatic SCI models, due to its enhanced

selectivity over GCV.

3.1.4 E. coli Nitroreductase (NfSA/NfsB) and CB1954

The combination of the E. coli oxygen-insensitive NAD(P)H, flavin mononucleotide (FMN)-

containing nitroreductase enzyme (either NfSA- 240 amino acids (a.a.), or NfsB- 217a.a.;

hereafter referred to as “NTR”) with the nitroaromatic alkylating agent prodrug, CB1954 [5-

(aziridin-1-yl)-2,4-dinitrobenzamide], is a GDEPT that has demonstrated considerable promise

in both cancer and regeneration research for over two decades (reviewed in Zhang et al., 2015;

reviewed in Searle et al., 2004; reviewed in Denny 2002; reviewed in Williams et al., 2015; Vass

et al., 2009; Drabek et al., 1997; Clark et al., 1997). CB1954 was first identified for the purpose

of tumor cell ablation in the late 1960s due to its cytotoxic potency against the Chester Beatty

Walker rat carcinoma cell line, WS, during screens of various nitrophenylaziridine compounds

(reviewed in Knox et al., 2003; reviewed in Knox et al., 1993; reviewed in Workman et al.,

1986; Connors and Melzack, 1971; Cobb et al., 1969; Khan and Ross 1969). The sensitivity of

Walker rat carcinoma cells to CB1954 was later identified to be due to the expression of NQO1

(DT-diaphorase), a similar type-I (oxygen-independent) nitroreductase enzyme to NfsA/NfsB

(reviewed in Knox et al., 2003). In contrast to rat tumors, human tumors were indentified to be

largely insensitive to CB1954 due to the severely reduced CB1954-catalyzing efficiency of the

human form of NQO1 (reviewed in Knox et al., 2003).

Mechanistically, type-I nitroreductases, such as NTR and NQO1, catalyze a two-electron transfer

to CB1954, using NADH or nicotinamide adenine dinucleotide phosphate (NADPH) as an

intermediate substrate, and reduce the 4-nitro group of CB1954 to a 4-hydroxylamine, which

Page 49: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

31

subsequently undergoes a non-enzymatic reaction with a thioester (e.g., acetyl coenzyme A)

producing a difunctional, DNA-interstrand crosslinking agent (reviewed in Williams et al., 2015;

reviewed in Knox et al., 2003; reviewed in Knox et al., 1993; Knox et al., 1988; Roberts et al.,

1986; Connors and Melzack, 1971). After DNA interstrand crosslinking from the reduced form

of CB1954, affected (NTR-expressing) cells are understood to undergo apoptosis in a p53-

independent manner (Cui et al., 1999).

In regards to applications (both in vitro and in vivo), the NTR/CB1954 ablation system has been

used to target and successfully kill mouse fibroblasts in vitro using a concentration of 20µM with

no toxicity observed in control mouse fibroblasts below 500 µM, as well as CD2-promoter-

driven-NTR-expressing mouse T cells in an in vivo setting, with no significant systemic toxicity

(Drabek et al., 1997). Similarly, NTR/CB1954 has demonstrated similar selectivity and

ablational efficiency in targeting mouse mammary gland luminary cells using ovine beta-

lactoglobulin promoter-driven NTR expression, as well as with mouse pre-adipocytes (Clark et

al., 1997; Felmer and Clark, 2004). With NTR-expressing mouse fibroblast NIH3T3 cells, one

study reported approximately 99% ablation using a treatment of 0.1mM CB1954 for an exposure

period of only 24 hours (Bridgewater et al., 1995).

Further evidence has demonstrated CB1954’s ablational efficiency against human cell types

genetically engineered to express NTR, such as the LS174T colorectal cancer cell line, or the

SUIT2, BxPC3, and AsPC1 pancreatic cancer cell lines, which have been shown to be as much

as 500 times more sensitive (in terms of apoptosis and cell killing) to CB1954 treatment than

their respective control parent lines (Green et al., 1997). Additionally, in human ovarian

carcinoma cells of the cell lines SK-OV-3 or IGROV-1, transgenic NTR expression has been

shown to increase CB1954 sensitivity by up to 2,000-fold over control parent line cells (Weedon

et al., 2000).

Attempts have been made to further improve the sensitivity of NTR-expressing cells to CB1954

treatment, such as one using six mutant forms of NTR each with amino acid substitutions, which

identified one specific mutant NTR variant with an amino acid substitution at residue F124 that

conferred a 5-fold increase in CB1954 sensitivity in human SK-OV-3 ovarian carcinoma cells

(Grove et al., 2003). A follow up study examining double mutants of NTR identified an even

more CB1954-sensitive variant than the F124-NTR mutant, which rendered SK-OV3 cells up to

Page 50: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

32

14-17 times more sensitive to CB1954 ablation treatment than their regular (NfsB) NTR-

expressing counterparts (Jaberipour et al., 2010). Additional optimizations of NTR/CB1954

ablation have reported improvements in ablational efficiency/prodrug sensitivity by: (1) using

alternative forms of NTR, such as using the other oxygen-insensitive E. coli nitroreductase,

NfsA, which was demonstrated to be 10 times more efficient in the catalytic efficiency of

CB1954 reduction; or (2) by developing a human-codon preference-adapted version of the NfsB

gene using 144 silent base changes to enhance NTR transcription and subsequently translation in

human cell types, which reportedly rendered several human cell lines up to 10 times more

sensitive to CB1954 ablation than their regular NTR/NfsB counterparts (Prosser et al., 2010;

Grohmann et al., 2009).

Relatively few neural uses of the NTR/CB1954 have been applied, however one study using the

nestin promoter to express NTR in neonatal rat cerebellar NPCs in an in vivo setting reported

significant cerebellar degeneration and volume reduction, in addition to ataxia as well as

sensorimotor functional losses after three days of daily intracerebellar treatment with 0.5mg/kg

CB1954, starting at the five-day age point (Kwak et al., 2007). Using 20µL

intracerebroventricular injections of 2mg/mL of CB1954 once daily for a period of 2 days, the

same study additionally reported significant degeneration in the hilus and granule cell layer of

the hippocampal dentate gyrus of similar nestin promoter-driven, NTR-expressing transgenic

neonatal rats (Kwak et al., 2007). CB1954 reportedly showed no relevant toxicity for control,

non-NTR-expressing animals in either neonatal rat NPC ablation experiment (Kwak et al., 2007).

The NTR/CB1954 enzyme-prodrug combination is not without its drawbacks, as similar to the

HSV-TK/GCV cell ablation system, it has been shown to possess a moderate, yet significant

bystander effect (Felmer and Clark 2004; Green et al., 1997; Bridgewater et al., 1997). The

bystander effect for NTR/CB1954 is understood to occur due to the presence of at least two cell

permeable metabolites, specifically the reduced forms of CB1954 possessing the 4-

hydroxylamine group, both before and after thioester activation (Bridgewater et al., 1997). In

practical application, the bystander effect of NTR/CB1954 has been demonstrated to be cell

density and type dependent, and for the specific instances of human pancreatic (SUIT2, BxPC3,

and AsPC1) as well as human colorectal (LS174T) cancer cell lines, a bystander effect was

detected by only 10% of NTR-expressing cells in one study (Green et al., 1997; Grove et al.,

1999).

Page 51: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

33

Overall, the NTR/CB1954 enzyme-prodrug ablation system has been demonstrated to be a

promising cellular/molecular tool for selective cell ablation (reviewed in Zhang et al., 2015;

reviewed in Williams et al., 2015; reviewed in Denny, 2002). The primary hurdle with regards

to its use in neuroregeneration studies, such as those to examine the restorative functions of

transplanted NPCs (and derived cells, such as oligodendrocytes and various types of neurons) at

different timepoints after spinal cord injury, is of course the system’s moderate, yet significant

bystander effect, which could inevitably result in unintended abnormalities and inabilities to

distinguish the specific repair brought on by the transplanted NPCs (Bridgewater et al., 1997).

Nevertheless, the NTR/CB1954 enzyme-prodrug combination for cell ablation still represents a

more promising alternative than species-restricted options such as transgenic-diptheria toxin

receptor expression, previously stated to be unusable for the selective ablation of human cell

types (reviewed in Zhang et al., 2015; Bennett and Clausen, 2007).

3.1.5 E. coli Nitroreductase (NfsB/NTR) & Metronidazole (Mtz)

Building on the success of the NTR/CB1954 enzyme prodrug cell ablation system, other

prodrugs have been evaluated in combination with NTR for a variety of purposes, including

finding a prodrug with a reduced bystander effect to improve the specificity of NTR cell ablation

in the areas of regeneration and development studies (reviewed in Zhang et al., 2015; reviewed

in Williams et al., 2015; reviewed in Dachs et al., 2009; reviewed in Denny 2002). One specific

prodrug identified to possess a reduced bystander effect in combination with NTR/NfsB is the

nitroimidazole anti-protozoan compound, metronidazole (Mtz) (reviewed in Zhang et al., 2015;

reviewed in Williams et al., 2015; reviewed in White and Mumm, 2013; reviewed in Curado et

al., 2008; Bailey et al., 1996).

The NTR/Mtz combination reportedly offers a plethora of benefits over other cell ablation

systems/enzyme-prodrug combinations, including: (1) increased efficacy; (2) greater specificity;

(3) resistance to promoter leakiness (e.g., off-target expression of the enzyme in non-target cell

types); and (4) quicker inducibility; in addition to (5) the ability to efficiently target non-

proliferating cells (reviewed in Curado et al., 2008; Curado et al., 2007; Pisharath et al., 2007).

The use of Mtz with NTR has specifically been described as “cell-type specific, inducible,

reversible, rapid and scaleable” (reviewed in Curado et al., 2008). The mechanism of ablation

induced in NTR-expressing cells by Mtz is understood to be largely similar, if not nearly

Page 52: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

34

identical to that of the NTR/CB1954 combination, whereby NADH/NADPH first reduces NTR

itself, then subsequently NTR catalyzes the reduction of the non-cyclic nitrogen of Mtz,

producing a cytotoxic radical-containing product that acts as a DNA-interstrand crosslinking

agent (reviewed in Curado et al., 2008; Edwards, 1993; Lindmark and Müller, 1976). The DNA-

crosslinking action of the activated/reduced form of Mtz then induces apoptosis in a p53-

independent mechanism (believed to be along the intrinsic/mitochondrial pathway), involving

caspase releases, plasma-membrane integrity losses, and nuclear fragmentation (reviewed in

Curado et al., 2008; Cui et al., 1999; Edwards, 1993; Lindmark and Müller, 1976).

One of the first cell ablation usages of the NTR/Mtz ablation was to target human cytoxic T

lymphocytes (hCTLs) in an in vitro setting (Verdijk et al., 2004). At a concentration of 1mM,

Mtz was found to produce a 79% proliferation inhibition in hCTLs, and additionally was

reportedly toxic for both NTR-expressing and control hCTLs at higher concentrations (Verdijk et

al., 2004). In its first reported in vivo utilization, in the Danio rerio zebrafish model, NTR-

ubiquitously-expressing (under the EF1α promoter) embryos were treated with 7.5-10mM of

Mtz, inducing muscular necrosis, cardiac damage, and necrosis over a 24-hour timeframe (56

hours post-fertilization (hpf)-80hpf) (reviewed in Pisharath and Parsons, 2009; Pisharath et al.,

2007; Pisharath 2007).

In the same work of Pisharath et al. in 2007, a treatment of 10mM of Mtz for a period of 24

hours was found to completely ablate embryonic, NTR-expressing, zebrafish pancreatic beta

cells that had previously been developed to use the preproinsulin gene promoter (ins) to drive

expression of an NTR-mCherry (a red fluorescent marker similar to red-fluorescent protein

(RFP)) fusion protein; the ablation of pancreatic beta cells was confirmed both by examining

fluorescence and insulin transcript levels (reviewed in Pisharath and Parsons, 2009; Pisharath et

al., 2007; Pisharath 2007). The very same Mtz treatment (10mM for 24 hours) was found to

have no off-target effects or toxicity in control zebrafish embryos, and additionally show no

apparent bystander effects on neighbouring cells of the developing exocrine pancreas, or in any

other endocrine pancreatic cells (e.g., alpha cells or delta cells, etc.) (reviewed in Pisharath and

Parsons, 2009; Pisharath et al., 2007; Pisharath 2007).

Subsequent in vivo examination of the NTR/Mtz system has included the successful complete

ablation: (1) of embryonic zebrafish cardiomyocytes (10mM for 24 hours); (2) embryonic

Page 53: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

35

zebrafish hepatocytes (10mM for 72 hours); (3) zebrafish primordial germ cells (10mM for 48

hours); (4) zebrafish hoxa13a/hoxd13a-expressing fin-fold mesenchyme (20mM for 3 day); (5)

zebrafish podocytes (10mM for 12 hours); (6) zebrafish xanthophores (10mM for 24 hours); as

well as (7) a reproduction of the targeting of zebrafish pancreatic beta cells (5mM for 12 hours)

(reviewed in Curado et al., 2008; Curado et al., 2007; Zhou et al. 2018; Lalonde and Akimenko,

2018; Huang et al., 2013; Zhou and Hildebrandt, 2012; Walderich et al., 2016). Upon removal

of Mtz, several embryonic zebrafish cell types have been shown to regenerate, such as zebrafish

pancreatic beta cells after 36 hours, and fin-fold mesenchyme after only 48 hours (Pisharath et

al., 2007; Lalonde and Akimenko, 2018). The NTR/Mtz system has additionally been used for

the purpose of creating inducible infertility models of both male and female zebrafish, by

targeting NTR-expressing spermatagonia and oocytes respectively (Hsu et al., 2010a, Hsu et al.,

2010b).

In order to improve the ablational efficiency of the NTR/Mtz system, attempts have been made

to develop a form of the NTR (NfsB) enzyme with a greater catalytic efficiency for reducing Mtz

(or other nitrimidazole/dinitrobenzamide substrates) (Mathias et al., 2014). The most successful

reported attempt at developing an improved version of NTR involved the discovery of a triple-

mutant (T41Q/N71S/F124T) form of the enzyme that has been demonstrated to induce

significant (compared to regular WT-NfsB/NTR controls) Mtz-induced ablation in vivo in both:

a) zebrafish spinal motor neurons after only 4 hours of 10mM Mtz treatment (only slightly less

than 24 hours of 10mM Mtz treatment on WT-NTR+ cells); and b) zebrafish spinal interneurons

after only 4 hours of as low as 4mM Mtz treatment (comparable to 24 hours of 10mM Mtz

treatment on WT-NTR+ cells) (Mathias et al., 2014).

The very same triple mutant form of NTR even permitted efficient in vivo ablation of zebrafish

cranial motor neurons, a more internal/deeper cell type than spinal inter- or motor neurons, with

only 4-hour treatments with 10mM Mtz (comparable to 24 hours of 10mM Mtz treatment on

WT-NTR+ cells) (Mathias et al., 2014). Taken together, the triple mutant form of NTR has

shown to be an enticing option for improving the ablation efficiency of the NTR/Mtz system

(Mathias et al., 2014).

In addition to zebrafish spinal motor and interneurons as well as cranial motor neurons, a

plethora of other neural cell types have been assessed in regards to NTR/Mtz ablation.

Page 54: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

36

Additional neural cell types successfully ablated using the NTR/Mtz system in a zebrafish in vivo

setting include: (a) brachiomotor neurons (significantly reducing food intake); (b) dopaminergic

neurons (wherein 24h of 5mM Mtz followed by 48h of 7.5mM Mtz induced caspase 3 release,

ablation, and sensorimotor deficits); (c) radial glial cells (wherein 1h of 5mM Mtz treatment

applied daily for 3 days induced significant apoptosis in GFAP-positive telencephalic cells); and

(d) rod photoreceptors (complete ablation after 24h of 10mM Mtz treatment with no damage to

neighbouring cones) (Allen et al., 2017; Mathias et al., 2014; Godoy et al., 2015; Shimizu et al.,

2015; Montgomery et al., 2010). Remyelination mechanisms have similarly been studied in a

zebrafish model, as evidenced by a study that used NTR/Mtz to induce demyelination, via total

ablation of CNS oligodendrocytes after 48 hours of 10mM Mtz treatment, but yet observed

complete remyelination after 7 days of prodrug removal (Chung et al., 2013).

In addition to zebrafish, other model organisms that have been used with the NTR/Mtz system

have included: (1) Oryzias latipes, more commonly known as Medaka or the Japanese rice fish,

with which osteoblasts and pancreatic beta cells have been among target cell types successfully

ablated; and (2) Xenopus Laevis, more commonly known as the African clawed frog, with which

rods and oligodendrocytes have been selectively ablated (Willems et al., 2012; Otsuka and

Takeda, 2017; Choi et al., 2011; Kaya et al., 2012). As in zebrafish, retinal degeneration has

been studied in the frog model using the NTR/Mtz system to target rod photoreceptors (Choi et

al., 2011). Although in the work of Coi et al., cone degeneration occurred subsequently after rod

ablation, it is not understood to have occurred through any bystander effect of NTR/Mtz, but

rather through as a separate result of the process of retinal degeneration, as NTR expression was

controlled by the rhodopsin promoter, not found in cones (Choi et al., 2011).

Similar to the 2013 work of Chung et al., remyelination has been investigated in frogs, using

MBP-promoter-driven expression of NTR in mature oligodendrocytes followed by Mtz treatment

at the tadpole stage (Sekizar et al., 2015; Kaya et al. 2012). Sekizar et al. monitored cell division

and regeneration after removal of Mtz and suggested based on their findings that optic nerve-

residing, Sox10+ OPCs may be the source of the newly formed mature oligodendrocytes that

provided remyelination, and additionally that little replication of OPCs is necessary for

remyelination in Xenopus (Sekizar et al., 2015). Sekizar et al.’s findings indicate that one of the

critical differences in the capability of remyelination in amphibians versus mammals may be the

Page 55: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

37

presence or absence respectively of sufficient quantities of endogenous OPCs in the optic nerve

(Sekizar et al., 2015).

Taken as a whole, the NTR/Mtz system has proven itself countless times as a simple and

efficient enzyme-prodrug combination for cell ablation (reviewed in Williams et al., 2015).

Despite the success of the NTR/Mtz, its usages have rarely extended beyond the fish and

amphibian model organisms, save for the 2004 work of Verdijk et al. using hCTLs in an in vitro

setting (reviewed in Williams et al., 2015; Verdijk et al., 2004). One of the only other known

mammalian usages of the NTR/Mtz consisted of ablating human lymphoma NB4 cells both in

vitro and in vivo transplanted in mice to generate tumors (McCormack et al., 2013). No

mammalian neural usages (in vitro or in vivo) of the NTR/Mtz system are known to have yet

occurred, however there is no obvious indication from the cellular and molecular aspects of the

system that such a feat should not be possible, nor that the system should intrinsically be limited

to neural cells of fish or amphibians (reviewed in Williams et al., 2015). As CB1954 has been

previously used to ablate NTR-expressing neural progenitors in rodents, it would seem feasible

that Mtz-mediated ablation should similarly be achievable in NPCs or NPC-derived cells (Kwak

et al., 2007).

Additionally, although the NTR/Mtz system has been demonstrated to lack any considerable

bystander effect, it still possesses drawbacks such as: (a) the availability of a suitable promoter

for expression in target cell types (a drawback seen in any gene-driven ablation system); (b) the

resistance of specific tissue/cell types to ablation (depending on the in vivo accessibility of the

cell type to the prodrug, regeneration capacity of the cell type, or ability of the cell type to

metabolize Mtz- as seen in hepatocytes); and (c) the requirement of longer treatments for certain

cell types depending on regenerative capacity among other factors (reviewed in Williams et al.,

2015; reviewed in Curado et al., 2008). The NTR/Mtz ablation system would appear to be an

ideal candidate choice for pursuing neuroregeneration loss/gain of function studies in rodent

models of SCI, most of all due to its lack of an observable bystander effect, however as

previously stated, the fact that no studies have yet pursued mammalian neural in vivo usages of

the system should nevertheless remain as a caution and caveat moving forward (reviewed in

Williams et al., 2015).

Page 56: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

38

Cell Ablation

System

Advantages Disadvantages

Diptheria Toxin Receptor

(DTR)

-Minimal bystander effects

(though potential for one to exist)

-Unusable with human cells

(non-selective),

-general toxicity,

-extremely dangerous to user

HSV-TK+ Ganciclovir

(GCV)

-widely used/highly understood,

-high substrate affinity (GCV for

HSV-TK)

-no species restrictions

-Considerable bystander effects

-Potential difficulties ablating

non-proliferative/post-mitotic

cells

HSV-TK+Brivudine

(BVDU)

-Reduced bystander effects,

-reduced toxicity (over GCV),

-low carcinogenicity

-Minorly reduced BVDU

substrate affinity for HSV-TK

(vs. GCV)

-BVDU ablation is less widely

used/understood (vs. GCV)

-Potential difficulties ablating

non-proliferative/post-mitotic

cells

NTR+CB1954 -no species restrictions

-not reliant on cell division

-considerable bystander effects

NTR+Metronidazole

(Mtz)

-no species restrictions

-not reliant on cell division

-no known bystander effects

-highly proliferative cell types

may be slightly resistant

-potential difficulties with low

Mtz solubility

Table 2: An Overview of Cell Ablation Systems. A wide variety of gene-directed enzyme-

prodrug therapies (GDEPT) exist for achieving the goal of selective cell ablation, ranging from

transgenic expression of the diptheria toxin receptor (+administration of the toxin itself) for

use with rodent cells, to the herpes simplex virus thymidine kinase (HSV-TK) +GCV/BVDU

system, and the E. coli NfsB nitroreductase (NTR)+CB1954/metronidazole (Mtz) systems.

Page 57: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

39

3.2 Gene Transfer Vectors

The first step towards creating lines of selectively ablatable NPCs for SCI neuroregeneration

studies is to successfully transfer the gene encoding an enzyme intended for use with an

apoptosis-inducing prodrug into the NPC genome (reviewed in Kantor et al., 2014; reviewed in

Manfredsson, 2014). In order to accomplish the task of NPC genomic transformation, a suitable

gene transfer vector must be selected. Two such vectors include: (1) lentiviruses; and (2)

transposons (reviewed in Kantor et al., 2014; reviewed in Manfredsson, 2014; reviewed in Joshi

et al., 2017).

3.2.1 Lentiviral Vectors

Lentiviruses (“slow-viruses” from the latin “lente”), which are composed of a dual copy,

positive sense single-stranded RNA genome covered by both a protein capsid and a lipid

envelope, are representatives of a genus of the retroviridae family (Baltimore Classification

Class VI) (reviewed in Sharon and Kamen, 2018). Genomic integration, a task accomplished by

the viral enzymes – reverse transcriptase (used for creating a DNA copy of the RNA viral

genome) and integrase (used for integrating the reverse transcribed viral DNA into the host cell

genome), has allowed lentiviruses to be developed as gene transfer vectors of transgenes

(reviewed in Sharon and Kamen, 2018).

While lentiviruses as gene transfer vectors offer such benefits as: (a) the ability to transfect cells

regardless of cell-cycle status (including transfecting post-mitotic cells); (b) the maintenance of

transgene expression in daughter cells (a feature not possessed by adenovirus/adeno-associated

viruses, as they are incapable of genomic integration of transgenes); (c) low immunogenicity;

and, (d) high transformation efficiency, they are not without considerable drawbacks (reviewed

in Sharon and Kamen, 2018; reviewed in Segura et al., 2013; reviewed in Durand and Cimarelli,

2011). Lentiviral vectors are unfortunately capable of inducing tumorigenicity due to proto-

oncogene activation, general insertional mutagenesis of target cells, and perhaps most critically

possess the risk, however minute, of a significant safety hazard (reviewed in Sharon and Kamen,

2018; reviewed in Durand and Cimarelli, 2011).

Page 58: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

40

Most lentiviruses are derived from the exceptionally dangerous and incurable human

immunodeficiency virus 1 (HIV-1), although they have undergone considerable modifications,

deletions of protein coding genes, and mutations over three “generations” to minimize the risk

considerably (reviewed in Sharon and Kamen, 2018; reviewed in Stellberger et al., 2017). The

only remnants of HIV-1 in lentiviral vectors are the protein binding regions (involved in reverse

transcription and packaging), packaging signal (ψ), and long terminal repeat (LTR) (reviewed in

Sharon and Kamen, 2018). First generation lentiviruses, which were divided into a three plasmid

system consisting of: (1) a transfer plasmid; (2) a packaging plasmid; and, (3) an envelope

plasmid, were shown to possess both a large cloning capacity (~8.5 kilo-base pairs (kb)), and a

high transfection efficiency, but yet also too high a risk of recombination to produce pathogenic

replication competent lentiviruses (RCL) (reviewed in Sharon and Kamen, 2018; reviewed in

Schlimgen et al., 2016; reviewed in Cornetta et al., 2011).

In the next generation, the unneeded virulence factor genes virus protein U (vpu), viral

infectivity (vif), negative factor (nef), and virus protein R (vpr) were removed, in order to

attenuate any replication-competent virus produced, however rare (reviewed in Sharon and

Kamen, 2018). Finally in the third generation of lentiviral vectors, an additional plasmid was

added to the system, as the packaging plasmid was divided into two- one of which contained the

gene rev (encoding the Rev response element (RRE) mRNA transcript binding protein to allow

nuclear export), and the other of which contained a combination of the gene, group antigens

(gag), encoding a structural protein component of the virion’s capsid, nucleocapsid, and matrix)

and pol (encoding the reverse transcriptase and integrase) (reviewed in Sharon and Kamen, 2018;

Farley et al., 2015).

Additionally, in third generation lentiviruses the gene trans-activator (tat), encoding a

transcription-activating, 5’ LTR binding protein, was rendered unnecessary and removed, as the

5’ LTR-situated tat promoter was replaced with a CMV promoter (reviewed in Sharon and

Kamen, 2018). Self-inactivating (SIN) lentiviral vectors were additionally developed, in which

modifications were made to remove the promoter/enhancer regions of the viral 3’ LTR,

rendering the viral vector unable to perform complete RNA transcription (reviewed in Sharon

and Kamen, 2018; reviewed in Stellberger et al., 2017; reviewed in Segura et al., 2013). The

modifications employed to generate third generation lentiviruses enabled the risk of

recombination to be almost entirely eliminated (reviewed in Sharon and Kamen, 2018).

Page 59: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

41

Lentiviral production generally consists of transiently transfecting human embryonic kidney

293T (HEK-293T) cells (or a similarly adherent cell type) through calcium phosphate

precipitation and harvesting the subsequently produced virus via collecting culture media

(reviewed in Sharon and Kamen, 2018). The harvested lentiviral culture media is then processed

through ultra-high-speed centrifugation (often above 90,000 g of relative centrifugal force

(RCF)), and virus pellets are resuspended at low volumes (several microlitres, depending on the

desired titer, starting culture media volume, time of transient transfection, density of adherent

cells used, etc.) and frozen at -80 degrees Celsius for long-term storage (reviewed in Sharon and

Kamen, 2018; Jiang et al., 2015). Although the titers of virus that can be produced through the

aforementioned method can be very efficient (up to 5x107 in unprocessed lentiviral media), the

serious drawbacks hindering the use of lentivirus are the length of time and labour involved in

production, high learning curve of production techniques with the possibility of failing to

generate sufficient titer (e.g., via accidentally suctioning off the viral pellet), and as mentioned

previously, the notorious, if actually minute, safety risks (if using first and second generation

lentiviral vectors specifically) (reviewed in Sharon and Kamen, 2018).

Overall, lentiviral vectors have demonstrated considerable success as gene transfer vehicles

(reviewed in Sharon and Kamen, 2018; reviewed in Stellberger et al., 2017). Although they

possess the beneficial properties of the ability to integrate within host cell genomes, the ability to

target both dividing and non-dividing cells, the ability to be passed on to daughter cells, and

strong transfection efficiency, lentiviral vectors are nevertheless hampered by the issues of

safety, as well as complexity and lengthiness in regards to production (reviewed in Sharon and

Kamen, 2018; reviewed in Stellberger et al., 2017).

3.2.2 Transposon Vectors

Non-viral vectors, such as transposons, are a generally more recent advancement in gene

transfer technology and offer a number of benefits over lentiviral vectors (reviewed in Vargas et

al., 2016; reviewed in Di Matteo et al., 2012). Some of these aforementioned benefits include:

(1) even lower immunogenicity, as the viral entry protein frequently used by lentiviral vectors,

vesicular stomatitis Indiana virus G-protein (VSV-G) can still incur an immune response from

some target cell types; (2) reduced development costs; (3) shorter production times, due to the

lack of additional required procedures such as transfecting adherent cells and spending additional

Page 60: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

42

days collecting the produced virus particles, testing for RCL, and titrating the produced virus;

and perhaps most importantly, (4) increased safety, as transposon vector production does not

contain the rare, yet dangerous risk of recombination to form RCL that is inherent to lentiviral

production (reviewed in Vargas et al., 2016). Cellular uptake difficulty is one minor

disadvantage of transposon gene vectors, as unlike viral vectors, they cannot independently enter

target cells, and thus require an additional assistance method, such as electroporation (reviewed

in Vargas et al., 2016). The efficiency of cellular uptake and resultant transduction of transposon

vectors is known to be influenced by: (1) plasmid size; (2) cell type; and (3) transfection method

(reviewed in Vargas et al., 2016).

Transposons are simple mobile genetic elements, present in plants, animals, fungi and even some

bacteria, that either undergo: (a) a “copy and paste” mechanism, as in the case of Class I

retrotransposons (e.g., LTR-containing retrotransposons, long-interspersed nuclear elements

(LNES), and short-interspersed nuclear elements (SINES), etc.) wherein the transposable

element (TE)’s mRNA transcript is reverse transcribed back to DNA, then integrates elsewhere

in the host genome; or (b) a “cut and paste” mechanism, as in the case of Class II DNA

transposons, which utilize the enzyme, transposase to mediate their excision from a host genome,

prior to reinsertion at a new site (reviewed in Vargas et al., 2016; reviewed in Skipper et al.,

2013; reviewed in Di Matteo et al., 2012). Class II DNA transposons consist of the transposase

gene flanked by inverted terminal repeats (ITRs) that act as recognition sites for the transposase

enzyme (reviewed in Vargas et al., 2016; reviewed in Skipper et al., 2013).

As far as gene transfer vectors, Class II, “cut and paste” DNA transposons are the exclusive

choice used, in order to avoid excessive additional random insertions of a target gene (as usually

only one insertion is required in most procedures), yet some additional ones (depending on the

transposon vector used) are almost always unavoidable to some degree (reviewed in Vargas et

al., 2016; reviewed in Skipper et al., 2013; reviewed in Ivics et al., 2009). When used for gene

transfer, transposons are often, but not always, split into a two-plasmid system, with the first

plasmid containing the delivery gene of interest flanked by ITR recognition sites, and the second

plasmid containing the appropriate transposase gene (reviewed in Vargas et al., 2018). The two-

plasmid system reduces the chance of integration of the transposase gene itself, usually limiting

the number of integrations below an excessive/unwated amount (reviewed in Vargas et al.,

2018). Transposons and transposase enzymes are known to be self-regulatory, based on their

Page 61: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

43

protein expression levels and transposon copy numbers (numbers of transposon genes present),

reducing activity at too high or low expression levels, thus unwanted excisions and integrations

are generally avoided with regards to gene transfer applications (reviewed in Vargas et al.,

2018). Two commonly used transposon vector systems are: (I) the piggyBac transposon; and

(II) the Tol2 transposon (reviewed in Vargas et al., 2016; reviewed in Ivics and Izsvák, 2010;

reviewed in Ivics et al., 2009; reviewed in Zhao et al., 2016).

The piggyBac transposon, previously known as IFP2, was originally discovered during

experiments studying Autographica californica nuclear polyhedrosis virus (AcMNPV) and other

baculovirus infections of cells of the cabbage looper moth, Trichoplusia ni (Fraser et al., 1996;

Fraser et al., 1995; Beames and Summers, 1990; Cary et al., 1989; Beames and Summers, 1988;

Fraser et al., 1985; Fraser et al., 1983). Investigators discovered two unique host-cell derived

2.4kb DNA sequences that had integrated into a specific region composed of two adjacent sites,

termed the few polyhedra (FP) locus of AcMNPV, wherein the four base-pair sequence “TTAA”

had been duplicated (Wang and Fraser, 1993; Beames and Summers, 1990; Cary et al., 1989;

Beames and Summers, 1988; Fraser et al., 1985; Fraser et al., 1983). The TTAA site was

determined to be a recognition site for piggyBac transposon insertion (Wang and Fraser, 1993).

Additionally, two 13-base pair (bp) ITRs, a set of two assymetrically-located 19bp internal

repeats (IRs), and another 7bp IR were detected in the piggyBac-transposed AcMNPV cells;

these features were eventually determined to be other key features of piggyBac transposon

integration (Carey et al., 1989; Fraser et al., 1985; Fraser et al., 1983).

PiggyBac transposons possess the ability to transfer very large (up to approximately 100kb)

insert regions and, have only shown minor losses of efficiency for inserts beyond 14kb (reviewed

in Vargas et al., 2018; reviewed in Zhao et al., 2016; Li et al., 2011; Wilson et al., 2007).

Previous uses of piggyBac transposons have included mouse germ line cells and HEK-293T cells

(reviewed in Vargas et al., 2018; Wilson et al., 2007; Ding et al., 2005). A significant advantage

of the piggyBac transposon system over other transposons, such as the extinct teleost fish-

derived, reconstructed/synthetic transposon system, Sleeping Beauty (SB), is the lack of any

“footprint” or base changes left behind after transposition (reviewed in Vargas et al., 2018;

reviewed in Zhao et al., 2016; Ivics et al., 1997; Radice et al., 1994). While SB is known to

leave behind a 3bp footprint in its insertion sites, piggyBac leaves its sites completely intact

(reviewed in Vargas et al., 2018; reviewed in Zhao et al., 2016).

Page 62: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

44

One further advantage of the piggyBac system over SB and a few other transposon systems are a

reduced susceptibility to a phenomenon known as “overproduction inhibition” (OPI): a self-

regulatory feature of some transposases involving the decrease of transposase activity, suggested

to occur via transposase monomers forming dimers, preventing excision, that often occurs when

the copy number of a transposon and thus its transposase expression reach beyond a threshold

level (reviewed in Vargas et al., 2018; reviewed in Muñoz-López and García-Pérez, 2010;

Wilson et al., 2007). The piggyBac system has additionally been demonstrated to be modifiable,

with experiments demonstrating altered features such as: (a) increased excision and integration

efficiencies; (b) removed integration capabilities; and (c) drug inducibility (reviewed in Vargas

et al., 2018; Yusa et al., 2011; Li et al., 2013; Cadiñanos and Bradley, 2007). Despite the

significant advantages of the piggyBac transposon system, it nevertheless contains several

drawbacks, including a possible preference for integration in transcription start sites and areas of

transcription in target/host cells, as well as the potential for promoter disruption adjacent to the

integration area, due to minor transcriptional activity of the piggyBac 5’LTR (reviewed in

Vargas et al., 2018; Gogol-Döring et al., 2016; Cadiñanos and Bradley, 2007).

The Tol2 transposon, itself a member of the hobo/Ac/Tam3 (hAT) transposon family, and the

only naturally active vertebrate transposon, was originally isolated from the medaka fish

(reviewed in Zhao et al., 2016; Kawakami et al., 2000; Kawakami and Shima, 1999; Kawakami

et al., 1998). Tol2-affected fish were albino, due to the preference for Tol2 integration into a

tyronsinase gene in the medaka genome (Kawakami et al., 2000; Kawakami and Shima, 1999;

Kawakami et al., 1998). The original Tol2 element is 4.7kb in size and contains a four exon

transposase gene with flanking ITRs of varying length (reviewed in Muñoz-López and García-

Pérez, 2010; reviewed in Koga, 2004). The Tol2 transposon system has been successfully used

for gene transfer in a variety of frog (Xenpous laevis), zebrafish, threespine stickleback

(Gasterosteus aculeatus), and human cell types (reviewed in Kawakami, 2007; Howes et al.,

2017; Kikuta and Kawakami, 2009; Hamlet et al., 2006; Balciunas et al., 2006; Wu et al., 2006).

Unlike piggyBac, Tol2 is known to have no specific preference for integration and produces an

8bp duplication on integration (Grabundzija et al., 2010; reviewed in Muñoz-López and García-

Pérez, 2010; reviewed in Ivics et al., 2009; reviewed in Koga, 2004). Additionally, Tol2

excisions do leave behind one of two types of footprints, either the removal of the 5’ or 3’

terminal region, which has suggested the possibility of two distinct excision methods (reviewed

Page 63: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

45

in Muñoz-López and García-Pérez, 2010; reviewed in Koga, 2004). Tol2 tranposons, much like

piggyBac transposons, have been demonstrated to similarly have a preference for integration

within transcriptional units and start sites (reviewed in Ivics and Izsvák, 2010; Grabundzija et al.,

2010). The cargo capacity of the Tol2 transposon is known to be at least 11kb, possibly higher,

however, this capacity is considerably dwarfed by the gargantuan 100kb capacity of the

piggyBac system (reviewed in Skipper et al., 2013; Balciunas et al., 2006; Urasaki et al., 2006).

Tol2’s transposition efficiency has been shown to be considerably lower than piggyBac’s, as

indicated by an experiment tracking the transposition frequencies of each in Henrietta Lacks

(HeLa) cells (Grabundzija et al., 2010). Tol2’s transposition efficiency at its peak activity was

only 1% in transposed HeLa cells, vs 3.5% for piggyBac (Grabundzija et al., 2010). Similarly,

the same study by Grabundzija et al. reported a higher OPI for Tol2 in HeLa cells (Grabundzija

et al., 2010). While the piggyBac transposon used by Grabundzija et al. required only 50ng of

“helper plasmid” (the transposase-encoding plasmid) to reach its peak transpositional activity,

Tol2 required as much as 125ng of its own helper plasmid (Grabundzija et al., 2010). Overall,

the work of Grabundzija et al. generally demonstrated the Tol2 transposon system to be less

efficient than the piggyBac system, in a variety of factors ((Grabundzija et al., 2010). An

additional study comparing Tol2 and piggyBac transposon efficiencies for gene transfer to

human primary T cells reached similar conclusions as Grabundzija et al. (Huang et al., 2010).

In summary, transposons have certainly shown promise in a variety of species and a plethora of

cell types as gene transfer vehicles (reviewed in Vargas et al., 2018; reviewed in Skipper et al.,

2013; reviewed in Ivics et al., 2009). Although they are limited by such drawbacks as OPI,

tendencies for integration within transcriptional units, and in the case of Tol2 and SB, excision

footprints, they nevertheless present themselves as largely more suitable gene transfer vectors in

comparison to viral vectors (e.g., lentivirus), both due to increased cargo capacity and reduced

biosafety among other factors (reviewed in Vargas et al., 2018; reviewed in Skipper et al., 2013;

reviewed in Ivics et al., 2009).

Page 64: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

46

Chapter 2: Research Aims

Overarching Goals, Questions, & Technical Aims

As discussed in Chapter 1- Sections 2.1.2 & 3.1, my MSc project of creating transgenic,

targetable NPCs for selective prodrug ablation exists in the overarching context of the

unanswered questions in SCI neuroregeneration of:

(1) “Is (a) remyelination, or (b) neuronal regeneration (neurite outgrowth)/axonal and synaptic

plasticity reestablishment the more effective and efficient strategy for SCI neurorepair (and

eventually pursuing translational therapeutic treatments)?”

(2) “Is long-term integration of transplanted cells (e.g., establishment of synaptic connectivity

from transplanted NPC-derived neurons, or large-scale remyelination directly by transplanted

OPC-derived oligodendrocytes) necessary for providing significant physical repair and

functional recovery, or is the combination of the short-term mechanisms of: (i) trophic support

(e.g., secretion of positive growth factors – BDNF/GDNF etc.), and (ii) immune modulation

(e.g., downregulating production of Il-6, IL-1β, or TNF-α) sufficient?”

and (3) “How much proliferation of transplanted hiPSC-NPCs is necessary and for how long

must it occur to provide effective SCI physical repair and functional recovery?”

(reviewed in Ahuja et al., 2017a&b; Ahuja and Fehlings, 2016; Siddiqui et al., 2015; Badner et

al., 2017; Khazaei et al., 2017).

Cell ablation may be used to attempt to answer the above three questions, in addition to several

others in regards to NPC transplants in a neuroregeneration context. Future experiments beyond

the scope of my current work could potentially make use of my suicide-gene-expressing NPCs in

transplant studies into SCI-model rodents, with prodrug-mediated ablations and evaluations both

of the loss of physical, cellular and tissue repair in the SCI lesion, as well as the reductions in

functional recovery gained (if any) from non-ablated NPC-transplanted SCI rats over control

(sham and vehicle injured) rodents. Although such experiments are far beyond the scope of my

current MSc project, I nevertheless wish to frame my work in its larger picture for future

potential uses.

Page 65: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

47

As my MSc project is a more methods focused approach, in lieu of traditional hypotheses, I have

developed a set of three key research aims to break down the fundamental aspects of my work.

The research aims I have developed towards creating selectively-targetable NPCs are:

1) Obtain an effective and efficient gene transfer vector (namely the piggyBac and Tol2

transposons for my NTR-NPC and HSV-TK NPCs respectively).

2) Generate Suicide-Gene expressing NPCs (an especially lengthy process with regards to the

NTR-construct NPCs, due to the two month-long culturing time for the monoclonal lines, and

only a very short (approximately 8-hour timeframe) for the puromycin purification of HSV-TK-

NPCs); and,

3) Perform drug directed ablations (accomplished using Mtz and GCV/BVDU for the NTR

and HSV-TK NPCs respectively).

Page 66: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

48

Chapter 3: Materials and Methods

1) NTR Insert Subcloning: Lentivirus->PiggyBac

The initial steps undertaken in my MSc project involved the subcloning of the three promoter-

NTR inserts (CMV, DCX, and MBP) from a pre-existing lentiviral plasmid vector into a

piggyBac transposon vector (displayed in Figures 1a-1d in Chapter 4: Results). Template

DNA of each NTR-promoter construct was produced through plating and streaking to acquire

single colonies of DH5α E. coli from bacterial stabs previously made available, followed by

culturing the single colonies using antibiotic selection with kanamycin (50µg/mL), and finally

maxiprep plasmid purification (alkaline lysis-neutralization-silica column elution).

Bacterial stabs were streaked on 10cm culture plates (Sarstedt TC Dish, 100 Standard Catalogue

(Cat.) #83.3902) containing 15mL of solidified agar (1.2%; Difco Cat. #0140-01) with Lennox-

broth (LB; 20g per L of H2O; Sigma Aldrich Cat. #L3022) and grown overnight at 370C in a

Lab Companion IB-11E 150L Incubator (Jeio Tech Company) to acquire single colonies of

plasmid-bearing E. coli. Single colonies of each plasmid-containing bacteria were picked using

10µL micropipette tips and cultured in 2L Erlenmeyer flasks containing 500mL of LB media

(20g per L of H2O; Sigma Aldrich Cat. #L3022) overnight at thirty-seven degrees Celsius with

mild shaking (225rpm) in a floor-model MaxQ5000 bacterial culture incubator/shaker

(Thermofisher Scientific Cat. #SHKE5000). Purified plasmid DNA was acquired via alkaline-

lysis-neutralization, silica gel column adherence, elution and isopropanol precipitation with 70%

ethanol washes (GeneAid Maxiprep Kit Cat. #PM010; 1 maxiprep per 250mL of culture, 2

maxipreps per plasmid). Final plasmid DNA was resuspended in 1mL of sterile molecular grade

H2O (Wisent Multicell Cat. #809-115-CL) per maxiprep (250mL or one-half of each plasmid’s

culture) in 1.7mL graduated centrifuge tubes (Froggabio Cat. #LMCT1.7B).

For the first portion of NTR subcloning, I performed PCR amplification of each of the three

NTR-promoter constructs using three pairs of primers (ACGT Corporation) designed to not only

amplify the constructs, but also additionally extend them with flanking MluI restriction enzyme

recognition sites. The primers used for PCR amplification of the NTR constructs were as

follows:

Page 67: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

49

1) CMV-NTR:

a) Forward (5’-> 3’) Primer: MluI-CMV-F:

ACGTACGCGTtccctatcagtgatagagatctccc

b) Reverse (3’-5’) Primer: MluI-CMV-R:

TCGAACGCGTactttccacaccTCTAGAAAAGTCG

2) DCX-NTR:

a) Forward (5’->3’) Primer: MluI-DCX-F:

ACGTACGCGTactccctatcagtgatagagatctccc

b) Reverse (3’-5’) Primer: MluI-CMV-R:

TGCAACGCGTcggagttaggggcgggac

2) MBP-NTR:

a) Forward (5’->3’) Primer: MluI-MBP-F:

ACGTACGCGTactccctatcagtgatagagatctccc

b) Reverse (3’-5’) Primer: MluI-MBP-R:

TGCAACGCGTactttccacaccTCTAGAAAACAATCCC

The MluI enzyme restriction digest site added was as follows (whereby | lines between bases

indicate the precise position of the asymmetric restriction digest cut):

5’ … A| C G C G T … 3’

3’ … T G C G C | A … 5’

The PCR amplification of NTR-promoter constructs was performed using a Mastercycler® pro

thermocycler (Eppendorf Cat. #950030010) using the following PCR programs (all identical):

Page 68: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

50

1) CMV-NTR:

980 C - 2 minutes (Initiation)

35x Cycles of [section enclosed within bolded brackets]:

[ 980 C - 15 seconds (Denaturation)

650 C - 30 seconds (Annealing)

720 C - 1 minute + 10 seconds (Elongation) ]

720 C - 5 minutes (Final Elongation)

40 C - hold (Final Hold)

2) DCX-NTR:

980 C - 2 minutes (Initiation)

35x Cycles of [section enclosed within bolded brackets]:

[ 980 C - 15 seconds (Denaturation)

650 C - 30 seconds (Annealing)

720 C - 1 minute + 10 seconds (Elongation) ]

720 C - 5 minutes (Final Elongation)

40 C - hold (Final Hold)

3) MBP-NTR:

980 C - 2 minutes (Initiation)

35x Cycles of [section enclosed within bolded brackets]:

[ 980 C - 15 seconds (Denaturation)

Page 69: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

51

650 C - 30 seconds (Annealing)

720 C - 1 minute + 10 seconds (Elongation) ]

720 C - 5 minutes (Final Elongation)

40 C - hold (Final Hold)

For each NTR-insert PCR amplification, 400µL master mixes were set up, then aliquoted into 16

x 25µL reactions. The PCR master mixes for all NTR constructs were created as follows:

a) Molecular Biology Grade, Sterile, Double-Distilled Water (DDH2O) (Wisent Multicell Cat.

#809-115-CL): 200µL (12.5µL per 25µL reaction)

b) 5x Q5® Reaction Buffer (New England Biolabs Cat. #B9027S): 80µL (5uL per 25µL

reaction)

c) 10mM Deoxynucleotide (dNTP) Solution Mix (New England Biolabs Cat. #N0447S): 8µL

(0.5µL per 25µL reaction)

d) Forward Primer (MluI-CMV/DCX/MBP-F): 4µL (0.25µL per 25µL reaction)

e) Reverse Primer (MluI-CMV/DCX/MBP-R): 4µL (0.25µL per 25µL reaction)

f) Template DNA (lentiviral CMV/DCX/MBP NTR- 1ng/µL): 16µL (1µL per 25µL reaction)

g) 5x Q5® High GC Enhancer (New England Biolabs Cat. #M0491S): 80µL (5µL per 25µL

reaction)

h) Q5® High-Fidelity DNA Polymerase (New England Biolabs Cat. #M0491S, 2,000 units/µL):

8µL (0.5µL per 25µL reaction)

PCR amplified NTR-promoter inserts (20µL volume of each 25µL reaction) were then run on

1% agarose (Invitrogen Ultrapure Agarose Cat. #16500-500) DNA gels with 20 lanes, containing

2.5µL of SYBRTM Safe DNA gel stain (Invitrogen Cat. #S33102) at 80V for 100 minutes in a

Wide Mini-Sub® Cell GT Horizontal Electrophoresis System with a 15 x 10 cm tray (Bio-Rad

Cat. #1704468) powered by a PowerPac™ HC High-Current Power Supply (Bio-Rad Cat.

Page 70: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

52

#1645052). Finished gels were imaged using the ChemiDoc™ XRS+ Imaging System (Bio-Rad

Cat. #1708265) on a Universal Hood III gel dock (Bio-Rad Cat. #731BR02333) with exposure

times ranging from 0.5 seconds to 4 seconds.

Once the NTR-promoter insert fragments had been PCR amplified with added flanking MluI

sites, the subsequent steps in NTR subcloning entailed fragment isolation (from an agarose

electrophoresis DNA gel) of a large quantity (400µL of each PCR reaction mix) of each PCR-

amplified NTR insert, followed by subsequent steps of purification, resuspension, MluI-

digestion, and repeated purification+resuspension.

Each PCR-amplified, NTR insert was run on a 1% agarose preparatory gel (precise

materials/catalogue numbers as described directly above) at 60V for 100 minutes. Each

preparatory gel consisted of 4 large wells, each loaded with 2x (50µL PCR NTR-fragment

reaction mix + 10µL gel loading dye, purple (6x - New England Biolabs Cat. #B7024S), as well

as 3uL (150ng) of a diluted (50ng/µL) 1kb plus DNA ladder (ThermoFisher Scientific Cat. #

10787018).

PCR-amplified-NTR insert fragments were then isolated from each preparatory gel using fresh,

single edge Fisherbrand™ razor blades (ThermoFisher Scientific Cat. #12-640) by placing each

gel under UV illumination with a benchtop UV transilluminator. Purification of the NTR

fragments from the extracted agarose gel slabs was accomplished first through column

purification using homemade columns composed of Fisherbrand™ Premium Microcentrifuge

Tubes: 0.6mL (ThermoFisher Scientific Cat. #502GRDBFP) with extremely small holes poked

through the bottom centre of each tube with a flame-heated, 25-gauge needle (Sigma-Aldrich BD

Precisionglide® syringe needles Cat. #Z19406) mounted on a 1mL syringe (Sigma-Aldrich Cat.

# Z683531) and placed inside 2mL collection tubes (borrowed from a plasmid miniprep kit-

QIAGEN Cat. #27104) stuffed approximately 1/3 full with glass wool (Sigma-Aldrich Cat. #

18421). The NTR fragment agarose-gel isolated slabs were then loaded into 2 columns each

(based on agarose slab size/volume) and centrifuged at 8000g for 2 minutes.

The aqueous flow-throughs containing each of the NTR fragments were then purified via phenyl-

chloroform (Sigma-Aldrich Cat. #P1944) purification. NTR fragments were mixed with phenyl-

chloroform in a volume 1:1 ratio in 1.7mL graduated centrifuge tubes (Froggabio Cat.

#LMCT1.7B), mixed vigorously via shaking and inverting, then centrifuged at 14,000g in an

Page 71: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

53

ultracentrifuge, then subsequently the top/clear layer of each fragment DNA (approximately

700µL) was pipetted out and transferred to a fresh 1.7mL centrifuge tube. Post-purified NTR

fragment DNA was then precipitated using 95% anhydrous ethanol (EtOH) (Sigma-Aldrich Cat.

#676829) added at a 2.5:1 ratio of EtoH:fragment DNA with the addition of 10% total final

volume worth of sodium acetate (Sigma-Aldrich S2889), and frozen at -200C for one hour. NTR

fragment DNA was then pelleted via centrifugation at 14,000g for 20 minutes, and the

supernatant of each tube was drained, followed by the addition of 100µL of 70% EtOH with

gentle mixing (tapping of tubes), then centrifuged again at 14,000g for 10 minutes. After the

final centrifugation step, the 70% EtOH supernatant was drained carefully, and the NTR

fragment DNA tubes were left to dry until the ethanol smell dissipated, then finally resuspended

in 28µL of molecular grade DDH2O. For each purified NTR fragment, two tubes of 28µL were

obtained and the tubes were combined into one tube of 56µL per fragment.

After resuspension, the purified DNA of each NTR fragment was then run on a 1% agarose DNA

electrophoresis gel for 100 minutes at 80V for visual inspection of the purified fragments. For

the DNA gel, 2µL of each purified NTR fragment DNA was mixed with 4µL of molecular grade

sterile DDH2O and 4µL of 6x gel loading dye; 3µL (150ng) of 1kb plus DNA ladder was

additionally included in the gel. MluI digestions of the PCR-amplified, purified NTR fragments

(amplified to contain flanking MluI restriction sites) were then undertaken in 100µL total

volume, using a mixture of 68µL of molecular grade sterile DDH2O, 20µL of each NTR

fragment, 10µL of 10x buffer 3.1 (New England Biolabs Cat. #B7203S), and 2µL of MluI

enzyme (New England Biolabs Cat. #R0198S). The NTR fragment MluI digestions were carried

out at 370C overnight (approximately 15 hours). Post-MluI digestion, the NTR fragments were

purified via phenyl chloroform purification and resuspended in 20 µL molecular grade DDH2O

each (identical methods as described above). 2µL of each of the purified and resuspended, MluI-

digested NTR fragments (with 4µL of molecular grade sterile DDH2O and 4µL of 6x gel loading

dye) was then run on a 1% agarose DNA gel at 80V for 100 minutes.

The piggyBac vector plasmid (into which the MluI-digested NTR fragments were to be cloned)

was subsequently then itself digested with MluI. A reaction mix consisting of a total volume of

100µL was created using 82.3µL of molecular grade sterile DDH2O, 10µL of buffer 3.1, 5.7µL

(approximately 2µg) of piggyBac plasmid DNA, and 2µL MluI enzyme. The piggyBac vector

plasmid MluI digestion was carried out at 370C for 4 hours.

Page 72: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

54

Ligations of the MluI digested (and purified & resuspended) NTR fragments+piggyBac vector

were then set up using 23µL of molecular grade sterile DDH2O, 4µL of 10x T4 ligase buffer

(New England Biolabs Cat. #M0202S), 10µL of insert DNA (the MluI-digested/purified NTR

fragments), 1µL of vector DNA (the MluI-digested, linearized piggyBac plasmid vector), and

2µL of T4 DNA ligase (New England Biolabs Cat. #M0202S). The four 40µL ligation reactions

(3 for each NTR fragment insert plus 1 control ligation of just the MluI-digested piggyBac

vector) were each split into 2x20µL reactions in 200µL PCR tubes (Bio-Rad Cat. #TFI0201) and

run overnight (approximately 15 hours) at 160C in a Mastercycler® pro thermocycler (Eppendorf

Cat. #950030010).

The next morning, the NTR-piggyBac ligation reaction mixes were precipitated with 100%

EtOH and resuspended in 12µL of molecular grade sterile DDH2O (protocol as described above),

then used for electroporation to transform DH10β E. coli. Transformations were performed

using 35 µL of 1/5th diluted (in 10% glycerol) ElectroMaxTM DH10β E. coli cells (Invitrogen

Cat. #18290-015), with 5µL of each ligation reaction mix in an electroporator (Eppendorf

Multiporator Cat. # 940000602) using 2500V electroshocks for 5ms. 400µL of SOC media

(New England Biolabs Cat. #B9020S) was added to the electroporated bacterial cells, which

were then incubated at 370C for one hour. After the recovery incubation, the transformed

bacterial cells were pelleted gently at 500g for 5 minutes, and the volume of SOC media was

adjusted to 200µL, followed by gentle resuspension by pipetting. Transformed DH10β cells

were then plated on 10cm dishes with LB+agar (1.2%). For each ligation reaction’s transformed

DH10β cells, 50µL was used per each of 4 plates containing 50µg/mL of kanamycin. The cells

on each plate were distributed via shaking with glass plating beads (Novagen Cat. #71013-4),

then left overnight (approximately 15 hours) for a 300C incubation.

Single colony DH10β clones were picked from each NTR-fragment insert + piggyBac vector

ligation-reaction plate and cultured in 14mL plastic culture tubes (Corning Life Sciences Cat.

#C352006) with 6mL of LB media containing 50 µg/mL of kanamycin at 300C with 225rpm of

shaking in a benchtop shaking incubator (Corning Life Sciences Cat. #EF18354) for 15 hours.

The next day, DNA from the single-colony-derived clones for each NTR insert + piggyBac

vector was miniprepped using 2.1mL of each clone (QIAGEN Cat. #27104, protocol contained

therein). Each clone’s miniprep DNA was eluted using 35µL of molecular grade sterile DDH2O.

Page 73: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

55

The miniprepped DNA from each NTR-piggyBac clone (14 for CMV-NTR-piggyBac, 5 for each

of DCX- and MBP-NTR-piggyBac) was then digested with MluI in order to check for the

presence of each insert fragment (as the MluI site was preserved during the ligation and now

flanked each NTR insert in the piggyBac vector (see Chapter 4: Results, Figure 1e-g). MluI

reaction mixes were setup using 10.5µL of molecular grade sterile DDH2O, 2µL of buffer 3.1,

7µL of each clone’s miniprep DNA, and 0.5µL of MluI enzyme, and digested at 370C for 1 hour.

The MluI-digested miniprep clone DNA (20 µL per digest + 5µL of purple 6x loading dye) was

then run on two 0.8% agarose gels with 20 wells each (due to the high number of clones) at 80V

for 100 minutes, then imaged (using the same gel imager described earlier in this chapter). For

CMV-NTR-piggyBac, clones #11-14 (termed C11-C14) were found to possess the CMV-NTR

insert (via release of a 2.1kb fragment after MluI digestion). For DCX-NTR-piggyBac and

MBP-NTR-piggyBac, clones #5 (D5) and #1-5 (M1-M5) respectively were found to possess the

DCX- and MB-NTR inserts (via release of the 3.5kb fragments after MluI digestions).

Additional amounts of each positive clone’s plasmid DNA was acquired via adding 5mL of LB

(with 50µg/mL kanamycin) to the remaining cultured clone stock in 14mL culture tubes,

growing overnight at 300C with shaking (225rpm), followed by plasmid miniprep (as described

earlier).

A series of follow up restriction digests (25µL each), using 5µL (of 35µL total) of miniprep

clone DNA, and 0.5µL of each restriction enzyme (listed below) were set up for the positive

clones C11-C14, D5, and M1-M5, using:

NdeI single digest (New England Biolabs Cat. #R0111S) (C11-C14, M1-M5, and D5);

SnaBI (New England Biolabs Cat. #R0130S) single digest (C11-C14);

SnaBI + Bstz17I (New England Biolabs Cat. #R3594S) double digest (C11-C14);

HindIII (New England Biolabs Cat. #R0104S) single digest (C11-C14);

EcoRV (New England Biolabs Cat. #R0195S) (M1-M5);

XhoI (New England Biolabs Cat. #R0146S) single digest (M1-M5);

XhoI + EcoRV double digest (M1-M5);

Page 74: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

56

XhoI + NdeI (M1-M5, and D5);

Bstz17I single digest (M3);

NdeI + Bstz17I double digest (M3); and,

EcoRV + NdeI souble digest (D5).

The follow-up restriction digests for the positive clones were run on 0.8% agarose gels and

confirmed CMV-NTR-piggyBac clones- C13 and C14, DCX-NTR-piggyBac clones D5, and

MBP-NTR-piggyBac clone M3 to not only possess the appropriate respective NTR insert, but

also be the correct expected size of each plasmid restriction fragment, so as to rule out the

possibility of any major (~1kb+) deletions through hyper-recombination during DH10β

incubation after electroporation and plating single colony clones.

The final steps of subcloning of the NTR-promoter constructs from their original lentiviral

plasmid vectors, into the piggyBac transposon vector, entailed the use of BsaI (New England

Biolabs Cat. #R0535S) digests to remove an unrelated insert in the plasmid, which otherwise

could have potentially interfered with NTR expression or transposon insertion in host NPC

genomes. The unrelated insert was flanked by BsaI restriction sites, thus allowing simple single

BsaI digests followed by ligations to re-circularize each linearized remaining, NTR-inserted,

piggyBac vector plasmid. BsaI digests of 100µL total volume were performed on clones C13,

C14, M3, and D5 using: 50µL (500ng) of miniprepped clone DNA (multiple minipreps were

combined per clone to acquire enough plasmid DNA), 3µL of BsaI enzyme, 10µL of 10x

CutSmart® buffer (New England Biolabs Cat. #B7204S), and 37µL of molecular grade sterile

DDH2O. All BsaI digests were incubated at 370C for approximately 15 hours overnight,

however 5µL of each reaction mix was removed after 1.5 hours.

The next morning, the BsaI-digested NTR-piggyBac clone DNA samples were purified with

phenyl chloroform, precipitated with 95% EtOH, and resuspended in 15µL molecular grade

sterile DDH2O. Additionally, the 5µL samples of each clone’s BsaI digest were run on a 0.8%

agarose gel at 80V for 100 minutes for visual inspection of linearization and removal of the

unrelated insert. Ligations were then set up to re-circularize the NTR-piggyBac clone plasmids,

using 40µL total volumes (later split into 2x 20µL reactions) per BsaI-digested clone composed

Page 75: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

57

of a reaction mix of 2 µL of clone DNA, 2µL of T4 DNA ligase, 4µL of 10x T4 buffer, and

32µL of molecular grade sterile DDH2O. As described previously, ligation reactions were

carried out at 160C overnight (approximately 15 hours) in a thermocycler.

After precipitation and resuspension, the next day, the BsaI digested plasmid DNA of clones

C13, C14, D5, and M3 was electroporated (2500V for 5ms) into 35µL (per clone BsaI digest) of

1/5th diluted (in 10% glycerol) ElectroMaxTM DH10β E. coli cells. The same protocol for post-

electroporation, plating, and culturing of transformed bacterial cells was used as previously, with

the one difference that instead of dividing the final remaining 200µL of cells into 4 kanamycin

plates, all 200µL was plated onto one plate (per each BsaI-digested NTR-piggyBac clone-

transformation). Several single colonies were obtained the next day from each plate, which were

then cultured and miniprepped (protocol as described previously), and the resultant plasmid

DNA was screened for positive clones via BsaI (to check for the absence of the unrelated insert)

and MluI digests (to check for the presence of each promoter-NTR insert) (protocols as described

for diagnostic restriction digest sets above).

Final plasmid DNA of the promoter-NTR-piggyBac clones with the unrelated insert removed

was acquired via plasmid maxiprep protocol using an endotoxin-free purification kit (FroggaBio

Cat. #PM010, protocol described therein). Plasmid DNA concentration was quantified with a

nanodrop spectrophotometer (ThermoFisher Scientific Cat. #ND-2000). Two DNA pellets were

obtained per each of the three NTR-piggyBac clones (500mL of LB media with 50µg/mL

kanamycin culture volume per clone, 250mL culture per pellet), each of which was eluted in

1mL of molecular grade sterile DDH2O. Additionally, glycerol stocks comprised of 1mL of each

clone’s LB culture mix and 1mL of 50% glycerol (Sigma-Aldrich 56-81-5) were created and

stored at -800C as backups. Sequencing was additionally performed on the NTR-piggyBac

clones for complete confirmation using several primer sets (ACGT corporation).

2) NTR-NPC Monoclonal Lines and Mtz Ablations

The second major step in my MSc project, as indicated earlier in aim 2 of Chapter 2: Research

Aims, entailed transforming a line of human iPSC (hiPSC)-derived NPCs (supplied by Dr.

Mohamad Khazaei in our laboratory) with each of the three promoter (CMV/DCX/MBP)-NTR-

Page 76: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

58

piggyBac transposon plasmids separately. The goal of such a step was to create three

independent NTR-NPC lines (CMV/DCX/MBP) with the NTR-promoter constructs integrated

into host cell genomes. Due to the nature of piggyBac transposons, and time constraints, random

integration was pursued, although site-directed integration (preferably into a vital/cell-life-

dependent gene) as achievable through Gateway cloning® (ThermofisherScientific) may have

been preferable (Grabundzija et al., 2010). In order to minimize the variation in NTR gene

expression (and thus Mtz susceptibility), monoclonal lines (derived from single NTR-NPCs)

were created via FACS and extremely-low (single cell) density plating was undertaken.

For the transformation of NPCs with the NTR-piggyBac plasmids, electroporation was

accomplished via the use of an Amaxa NucleofectorTM 2b (Lonza Cat. #AAB-1001). For each of

the three NTR-piggyBac plasmids, 1.0x106 NPCs in 1mL of serum free 9 media (SF9-

containing: Gibco™ Dulbecco's Modified Eagle Medium: Nutrient Mixture F-12 (DMEM/F-12 -

ThermoFisher Scientific Cat. # 11320033); 1% of a penicillin-streptomycin mixture (P/S, 10,000

units/mL; Thermofisher Scientific # 15140122); 10nM B27 supplement - vitamin A

(ThermoFisher Scientific Cat. #12587010); 10nM BFGF, 10nM EGF; and 10nM heparin sodium

salt (Sigma-Aldrich Cat. # H3149)) were loaded into a curved glass electroporation cuvette

(accessory of Amaxa NucleofectorTM 2b) with 1µg (5µL) of NTR-piggyBac plasmid DNA, and

100µL of Nucleofector® Solution. Program A33 on the electroporator was selected, and after

completion of the transformations, 500µL of pre-warmed (370C) SF9 media was added to each

cuvette, and the transformed, NTR-NPCs were then taken for FACS.

Through FACS, accomplished with a flow cytometer (Beckman-Coulter Cat. #FC500),

successfully NTR-transfected NPCs (1-3%) were selected based on either mCherry (red)

fluoresence (for the ubiquitous CMV-NTR-NPCs), or green fluorescent protein (GFP)

fluorescence (for DCX- and MBP-NTR NPCs, as all NTR-piggyBac transposon plasmids

possessed a GFP marker for visual identification of successful transformation) and single cells of

each of the three types were plated with SF9 media in multiple wells of one 96 well plate

(Sarstedt Cat. #83.3924.005) per each of the three promoter-NTR combinations. Monoclonal-

derived cell groupings (referring to each plated well) exhibiting the brightest mCherry (CMV-

NTR-NPCs) or GFP (DCX- and MBP-NTR NPCs) were selected and continually cultured and

expanded (and gradually transferred to larger volume cell culture plates) over a two-month

timeframe to eventually produce monoclonal, NTR-NPC lines consisting of approximately 1.5-

Page 77: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

59

2.0x107 cells each per line. Ten backups of approximately 1.0x106 cells each per NTR-NPC

monoclonal line were created and placed in liquid nitrogen for long-term storage.

For ablation experiments with Mtz (Santa Cruz Biotechnology Cat. #CAS 443-48-1), 1.25x104

CMV-NTR-NPCs were plated in each well of a 24 well plate (Sarstedt Cat. #83.3922.005) in

SF9 media (500µL per well) using 15mm thickness, round glass cover slips (Bellco Glass Cat.

#1203J82) coated with Matrigel™ (Corning™ Matrigel™ Membrane Matrix Cat. #CB-40234A).

Twenty-four hours later, the media of each well was changed and switched to: SF9 media with

no Mtz (top row/row 1- wells 1-6, as a negative control); SF9 media with 2.5mM dissolved Mtz

(row 2- wells 7-12); SF9 media with 5mM dissolved Mtz (row 3- wells 13-18); and SF9 media

with 10mM dissolved Mtz (row 4- wells 19-24). DCX- and MBP- NTR NPCs (which would not

be capable of expressing NTR until differentiation into neurons and oligodendrocytes

respectively) were also plated at the same density in one 24-well plate per each cell line using the

same protocol as per CMV-NTR NPCs, however with the minor difference of using DMEM/F-

12 media containing 1% fetal bovine serum (FBS) (Sigma-Aldrich Cat. #F1051) and 1% P/S,

instead of SF9 media. DCX- and MBP-NTR NPCs were left in the serum-containing F-12 media

for approximately two weeks in order to induce differentiation (into a mixture of neurons,

oligodendrocytes, and astrocytes).

After each of a set of six timepoints (24-well plate vertical “columns”: column 1- 4 hours,

column 2- 12 hours, column 3- 24 hours, column 4- 48 hours, column 5- 72 hours, and column 6-

96 hours), the cover slips for CMV-NTR-NPCs for a corresponding vertical column (4 wells

each) were removed, transferred to the corresponding vertical column and well of a fresh 24 well

plate. The transferred cover-slip cells were then fixed with 1mL of 4.0% of paraformaldehyde

(PFA, Sigma-Aldrish Cat. #P6148), followed by 3x5 minute washes in 1mL of 1x (diluted 1:10

from 10x) phosphate-buffered saline (PBS, Wisent Bioproducts Cat. #311-012-LL), and finally

left in 1mL of 1x PBS. The same process described above for CMV-NTR NPCs (plating, Mtz

treatments, fixation, and qualitative visual observation) was additionally performed with a set of

WT-NPCs (hiPSC-derived NPCs with no NTR transgene) (supplied by Dr. Mohamad Khazaei

in our laboratory). Imaging of cells was performed once daily for 72 hours using an EVOS

FLoid™ Cell Imaging Station (ThermoFisher Scientific Cat. #4471136). Due to the lack of any

obvious visually-identifiable changes in cell density (and thus Mtz-mediated ablation) for any of

the Mtz concentrations or timepoints relative to each timepoint’s corresponding control well,

Page 78: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

60

further quantitative analysis (e.g., 4′,6-diamidino-2-phenylindole (DAPI) staining and

cytometry) was not pursued. A second attempt of the previously described Mtz ablation

experiment with CMV-NTR-NPCs was performed using an alternative source of Mtz (Sigma-

Aldrich Cat. #M3761).

3) NTR-Mtz Ablation Troubleshooting

Due to the unforeseen lack of ablational capability observed for any of the Mtz concentrations

and timepoints for the CMV-NTR-NPC line, two main avenues were pursed in order to attempt

to troubleshoot the inconclusive results observed (addressed further in Chapter 4: Results,

Figures 4-5). The first of these troubleshooting attempts entailed the evaluation of NTR enzyme

protein expression, while the second pursued the possibility of using a version of the NTR/NfsB

gene that had been optimized for human codon-preference usage (containing 144 silent

mutations) (Grohman et al., 2009).

There was unfortunately no readily available, commercial antibody for NTR, and thus a

comparison of the total protein extract of CMV-NTR-NPCs and WT-NPCs was made via

sodium-dodecyl-sulfate polyacrylamide gel electrophoresis (SDS-PAGE) followed by staining

with Coomassie Brilliant Blue G-250 Dye (ThermoFisher Scientific Cat. #20279). The protein

extract of both CMV-NTR-NPCs and WT-NPCs was acquired via scraping cells (using

Corning® cell scrapers, Sigma-Aldrich Cat. #CLS3010) off 8 individual wells per each of the

two cell types, combining their media into one 15mL polypropylene conical centrifuge tube

(FroggaBio Cat. #TB15-500) per cell type, centrifuging at 290Gs for 5 minutes to pellet cells in

an Eppendorf 5810R 5 Amp version centrifuge with an A-4-81, 4x250mL swing bucket rotor

(Eppendorf Cat. #022627023) followed by removal of media supernatants, transferring to 1.7mL

ultracentrifuge tubes, resuspending first in 300µL of 1xPBS (with an extra identical

centrifugation step in between), then in a 300µL (per each cell type) lysis solution composed of

294µL radioimmunoprecipitation assay (RIPA) buffer (Sigma Aldrich Cat. # R0278), 3µL of

ethylenediaminetetraacetic acid (EDTA) (Sigma Aldrich Cat. #E6758), and 3µL of protease

inhibitor cocktail (Sigma-Aldrich Cat. #P8340 SIGMA), and finally via sonication.

The lysis-buffer-resuspended CMV-NTR- and WT-NPCs were placed on ice, and 3x10 second

cycles of sonication with a setting of 25W and 50% amplitude (with 45 second recovery periods

on ice in between in order to avoid overheating and denaturing proteins) was performed with a

Page 79: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

61

Vibra-Cell™ model VC50 AT sonicator (Sonics & Materials Inc., Dannbury, CT- discontinued

model) in a Fisherbrand™ Sound Enclosure (ThermoFisher Scientific Cat. #432B2). After

sonication, the lysis buffer CMV-NTR- and WT-NPC samples were centrifuged at 18,630g and

40C in a Scilogex refrigerated microcentrifuge D3024R (Scilogex Cat. #922015139999), and the

protein-extract-containing supernatant of each sample was aliquoted into 6x50µL aliquots in

1.7mL microcentrifuge tubes, then stored at -800C.

Prior to running SDS-PAGE and staining of the total protein of the CMV-NTR-NPC and WT-

NPC protein extracts, protein extract concentrations needed to be determined. Protein extract

concentrations were evaluated via performing a protein assay using spectrophotometry to

measure absorbances at 562nm with both the protein extract samples and protein samples of

known concentration, in order to establish a standard curve (using the Micro BCATM Protein

Assay Kit - ThermofisherScientific Cat. #23235, protocol described in detail within). For SDS-

PAGE, 20µg samples of each NPC protein extract was run on a gel composed of a 4%

acrylamide stacking gel affixed ontop of a 10% acrylamide resolving/separating gel in a

CriterionTM Cell (Bio-Rad Cat. #1656001) electrophoresis system for 30 minutes at 80V (for

protein samples to clear the stacking gel), followed by one hour and 30 minutes at 100V (for

protein samples to resolve).

Final staining of the total protein of each NPC extract on the gel was accomplished via 3x5

minute washes in DDH2O, followed by 30 minutes in Coomassie Brilliant Blue G-250 Dye, and

finally an overnight (approximately 15 hours) destaining wash in DDH2O. All washing, staining,

and destaining steps were performed with gentle rocking agitation on a Bio-Rad Mini Rocker

(BioRad Cat. # 1660710EDU). After staining, the gel was imaged using the ChemiDoc™ XRS+

System (Bio-Rad Cat. #1708265) on a Universal Hood III gel dock (Bio-Rad #731BR02333)

with exposure times ranging from 30 to 180 seconds.

The second portion of troubleshooting the NTR-Mtz ablation system for human iPS-derived

NPCs, as mentioned previously, entailed the use of a mutated variant of the NfsB/NTR gene that

had been optimized for human codon preference usage with 144 silent mutations (Grohman et

al., 2009). A construct composed of the human-codon optimized NfsB/NTR gene (HCO-NTR)

driven by the ubiquitous CMV promoter was cloned into the CMV-NTR-piggyBac plasmid

vector. Unlike the previous CMV-NTR construct used to generate the monoclonal line in

Page 80: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

62

Chapter 3: Section 2, the HCO-NTR construct did not possess a fluorescent marker tag/fusion-

protein gene, yet still possessed a GFP marker to identify transformed cells. Human-codon-

optimized-NPCs expressing NTR under the ubiquitous CMV promoter (HCO-NTR-NPCs) were

created using electroporation (as described previously) with 1µg of HCO-CMV-NTR plasmid

DNA, and successfully transformed cells were purified via antibiotic selection with puromycin

(Sigma-Aldrich #P8833) treatments of 1µg/mL for 5 hours, followed by 3 washes of 10 minutes

each using regular DMEM/F-12 media (with 10% FBS and 1% P/S).

For ablation experiments of HCO-NTR-NPCs, 1x106 cells were plated in 10cm, Matrigel™-

coated culture plates in SF9 media. Twenty-four hours later, the media of plates was replaced

with SF9 media mixed with pre-dissolved, pharmaceutical-grade Mtz to a final concentration of

6mM, from plastic intravenous sacks (IV Solution, Metronidazole, Metro-IV, 500mg, PAB®,

100mL, B.Braun Cat. # D5353-5224). The same procedure was repeated using non-

transformed/WT-NPCs. The HCO-NTR-NPC plates were then imaged daily for two days with

both brightfield and green-fluorescent channels using an EVOS FLoid™ Cell Imaging Station

(ThermoFisher Scientific Cat. #4471136).

4) HSV-TK Plasmid DNA Purification and Confirmation

HSV-TK plasmids used were obtained from Addgene (http://www.addgene.org), including the

Tol2 transposon plasmid, pKTol2P-PTK (Addgene #85599) containing a gene for a fusion

protein of HSV1-TK and puromycin resistance (puDeltatk) under the control of the ubiquitous

PGK promoter as well as the Tol2 transposase containing plasmid, pCMV-Tol2 (Addgene

#31823) (Clark et al., 2007; Balciunas et al., 2006; Chen and Bradley, 2000). Bacterial stabs

were streaked on 10cm culture plates (Sarstedt TC Dish, 100 Standard #83.3902) containing

15mL of solidified agar (1.2%; Difco #0140-01) with LB (20g per L of H2O; Sigma Aldrich Cat.

#L3022) and grown overnight at 370C in a Lab Companion IB-11E 150L Incubator (Jeio Tech

Company) to acquire single colonies of plasmid-bearing E. coli. Single colonies of each plasmid-

containing bacteria were picked using 10µL micropipette tips and cultured in 2L Erlenmeyer

flasks containing 500mL of LB media (20g per L of H2O; Sigma Aldrich L3022) overnight at

370C with mild shaking (225rpm) in a floor-model MaxQ5000 bacterial culture incubator/shaker

(Thermofisher Scientific #SHKE5000). Purified plasmid DNA was acquired via alkaline-lysis-

neutralization, silica gel column adherence, elution, and isopropanol precipitation with 70%

Page 81: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

63

ethanol washes (GeneAid Maxiprep Kit PM010; 4x maxi-preparations, 1 maxiprep per 250mL of

culture, 2 maxipreps per plasmid). Final plasmid DNA was resuspended in 1mL of sterile

molecular grade H2O (Wisent Multicell 809-115-CL) per maxiprep (250mL or one-half of each

plasmid’s culture) in 1.7mL graduated centrifuge tubes (Froggabio LMCT1.7B).

Restriction digests of plasmid pCMV-Tol2 were performed with 200ng of plasmid DNA using

the enzyme combinations of 1.) BsaI (New England Biolabs #R0535S) and SnaBI (New England

Biolabs #R0130S); and 2.) BsaI (New England Biolabs #R0535S) and BglII (ThermoFisher

Scientific # ER0081). Similarly, restriction digests of plasmid pKTol2P-PTK were performed

with 200ng of plasmid DNA using the enzyme combinations of 1.) NheI (New England Biolabs

#R0131S) and SnaBI (New England Biolabs #R0130S); and 2.) BsaI (New England Biolabs

#R0535S) and BglII (ThermoFisher Scientific # ER0081). For all restriction digests, enzyme-

DNA-buffer mixtures were incubated for one hour at thirty-seven degrees Celsius in a 150L

incubator (Jeio Tech Company Lab Companion IB-11E).

Subsequently, the restriction digests of each plasmid were run on a 1% agarose (Invitrogen

Ultrapure Agarose #16500-500) gel with 20 lanes at 80V for 100 minutes in a Wide Mini-Sub®

Cell GT Horizontal Electrophoresis System with a 15 x 10 cm tray (Bio-Rad #1704468) powered

by a PowerPac™ HC High-Current Power Supply (Bio-Rad #1645052). Finished gels were

imaged using the ChemiDoc™ MP Imaging System (Bio-Rad) on a Universal Hood III gel dock

(Bio-Rad #731BR02333) with exposure times ranging from 0.5 seconds to 4 seconds. Similarly,

to the NTR-piggyBac plasmids created through subcloning in Chapter 3: Section 2, both

plasmids CMV-Tol2 and KTol2P-PTK were further confirmed via sequencing (primers supplied

by ACGT Corporation).

5) Mixed NTR and HSV-TK NPC GCV Ablation

The initial usage of the HSV-TK+GCV ablation system in my MSc project attempted to assess

bystander effects through applying GCV to a mixed population of HSV-TK+ NPCs and non-

HSV-TK-expressing NPCs. For the HSV-TK- NPCS, HCO-NTR-NPCs (from Chapter 3:

Section 3) were selected due to their immediate availability. The same line of hiPSC-derived

NPCs was used to generate HSV-TK+ NPCs, and the transformations with both plasmids CMV-

Tol2 (Tol2 transposase) and KTol2P-PTK (Tol-2 transposon carrying a puromycin-

resistance+HSV-TK fusion protein gene) were carried out identically as described for the three

Page 82: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

64

promoter NTR-NPC lines (Chapter 3, Section 2) and HCO-NTR-NPCs (Chapter 3, Section 3).

500ng of each plasmid of the two-plasmid, Tol2 system was used during the transformation,

rather than 1µg for the single-plasmid systems described earlier (Chapter 3, Sections 2 & 3).

For the GCV bystander evaluations, 1.0x106 cells, of either: HCO-NTR-NPCs (GFP+); HSV-TK-

NPCs; or a 1:1 mixture of both cell types, were plated in one 10cm plastic culture plate (Sarstedt

TC Dish, 100 Standard Cat. #83.3902) each (per cell type used) in 10mL of SF9 media. One

additional plate of mixed HSV-TK and HCO-NTR NPCs was also set up as a negative control.

Twenty-four hours later, the media of each plate was changed and replaced with either: 1) three

experimental plates: SF9 containing 1µg/mL of dissolved GCV (powder dissolved in alkaline

DDH2O as described within InvivoGen protocol; InvivoGen Cat. # sud-gcv); or 2) negative

control mixed culture plate: regular SF9 media. All four plates were imaged once daily with

brightfield and green fluorescent channels of an EVOS FLoid™ Cell Imaging Station for a

period of 96 hours. One media change for all plates occurred at the 72-hour timepoint.

6) HSV-TK-NPC GCV and BVDU Bystander Ablations

In order to reduce bystander effects for future targeted cell killing studies beyond the scope of

this project, BVDU was selected as an additional ablation prodrug in combination with HSV-

TK-NPCs (reviewed in Dachs et al., 2009; Degrève et al., 1999). A mixed culture of HSV-TK-

NPCs and GFP+-WT-NPCs (a separate line provided by Dr. Mohamad Khazaei in our

laboratory) in a 1:1 ratio was set up wherein 5.0x104 cells (half HSV-TK-NPCs, and half GFP+-

WT-NPCs) were plated in each well (pre-coated with MatrigelTM) of two, six-well culture plates

(Sarstedt TC Plate 6 Well,Standard,F, Cat. #83.3920.005). After 24 hours, the wells of each plate

received the following 6 concentrations of GCV or BVDU respectively into one well each:

0µg/mL (negative control well); 0.0625µg/mL; 0.125µg/mL; 0.25µg/mL; 0.50µg/mL; and

1.0µg/mL. For the next four days, the media from each well of each plate was replaced with

fresh media containing the appropriate concentration of GCV/BVDU (Sigma-Aldrich Cat.

#B9647). All wells were imaged once daily with brightfield and green fluorescent channels of

an EVOS FLoid™ Cell Imaging Station for a period of 96 hours.

During the daily media changes with SF9 (containing the appropriate concentration of

GCV/BVDU), the old media of each well was collected in 15mL polypropylene conical

centrifuge tubes (FroggaBio #TB15-500). The 15mL tubes of collected media containing

Page 83: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

65

floating dead cells were centrifuged at 290Gs for 5 minutes to pellet cells in an Eppendorf 5810R

5 Amp version centrifuge with an A-4-81, 4x250mL swing bucket rotor (Eppendorf

#022627023). After centrifugation, the tubes were drained of media manually by gently pouring

into a 250mL waste beaker, then left inverted on a clean bench surface with paper towel for 2

minutes.

The cell pellets were then resuspended in 100µL of BD Cytofix/Cytoperm™

fixation/permeabilization solution containing 4.2% formaldehyde (BD Biosciences #554714)

using repeated pipetting to fully resuspend pelleted cells, then left for 20 minutes of fixation.

After fixation, the 15mL tubes of resuspended dead cells were centrifuged again at 290Gs for 5

minutes in the same Eppendorf 5810R swing-bucket rotor centrifuge as stated previously. The

supernatant of fixation solution was again drained manually, and the tubes were left inverted for

2 minutes to remove the remaining fixation solution. The fixed cell pellets were each finally

resuspended in 100µL of 1x PBS (Wisent Multicell #311-012-CL) with repeated pipetting, then

transferred to 1.7mL graduated centrifuge tubes (Froggabio LMCT1.7B), then stored at 40C.

Due to time constraints on my MSc project, as well as the apparent death of essentially all GFP+-

WT-NPCs after only twenty-four hours (as examined qualitatively in n=2 replicates), FACS and

cytometry of the fixed cells collected from the four timepoints of the two drug treatments was

not pursued.

7) HSV-TK-NPC GCV and BVDU Quantitative Ablations

The final step in the ablational studies carried out within this MSc project was the simple

quantitative evaluation of the ablational capabilities of GCV and BVDU against the line of HSV-

TK-NPCs (described earlier in Chapter 3: Sections 4-6). For both GCV and BVDU, four six

well culture plates (Standard Cat. #83.3920.005) were set up wherein 5.0x104 HSV-TK-NPCs

were plated in 2mL of SF9 media in each well (pre-coated with MatrigelTM). After 24 hours, the

media of all plates (8 total, 4 per prodrug representing one day treatment each) was changed, and

the wells of each plate received the following 6 concentrations of GCV or BVDU respectively

into one well each: 0µg/mL (negative control well); 0.0625µg/mL; 0.125µg/mL; 0.25µg/mL;

0.50µg/mL; and 1.0µg/mL (all dissolved in 2mL SF9 media).

Once daily for a period of 96 hours, one plate of HSV-TK-NPCs of both prodrug treatments (2

plates per prodrug/day) was removed from 370C incubation and the cells were fixed with 2mL of

Page 84: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

66

4.0% of PFA (Sigma-Aldrich Cat. #P6148), followed by 2x5 minute washes in 1mL of 1xPBS

(Wisent Bioproducts Cat. #311-012-LL), and finally left in 1mL of 1x PBS. All wells were then

stained with 1-2µg/mL of DAPI for 10-30 minutes (later optimization required longer staining

times for improved fluorescence, image quality, and cytometry). All wells were imaged with the

brightfield channel of an EVOS FLoid™ Cell Imaging Station before fixation, as well as with

the brightfield and blue fluorescent channels post-fixation. A minimum of five representative

images were acquired per well. Estimation of the total remaining attached cells/well was

accomplished through “peak” analysis, whereby blue fluorescent, DAPI-stained nuclei (“peaks”)

were counted in ImageJ photo-analysis software (NIH). The mean number of total attached

cells/well was determined through extrapolating the mean number of cells in the surface area

(SA) covered by a representative image (approximately 0.37mm2) to the total SA per well

(approximately 1134mm2), a factor of ~3,057 times the SA of a representative image. The same

procedure was repeated for the 48, 72, and 96-hour timepoints, and n=3 replicates were

performed for the entire experiment.

As both the GCV and BVDU prodrugs used during the previous ablation experiments had been

diluted in dimethyl-sulfoxide (DMSO), up to a maximum final concentration of 0.4% DMSO in

culture media (the highest concentration of DMSO used for the 1.0µg/mL GCV/BVDU

concentrations), I believed it to be important to rule out the possibility of DMSO-induced cell

death, a minor possibility as indicated by previous literature (Yuan et al., 2014). In order to fully

determine if DMSO, and not prodrug treatment could be culpable for inducing NPC death, I

plated 5.0x104 NPCs in two wells of a six well culture plate on MatrigelTM with 2mL of SF9

media. Twenty-four hours later, the media of both wells was changed and replaced with either a)

SF9 containing 0.4% DMSO (the highest DMSO concentration used for prodrug dilution), or b)

0.4% PBS then returned to 370C incubation. After 48 hours, both wells were imaged with the

brightfield channel of an EVOS FLoid™ Cell Imaging Station and qualitatively evaluated for

obvious changes in cell density.

Page 85: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

67

Chapter 4: Results

1) NTR-PiggyBac Subcloning

The earliest portion of my MSc project entailed the subcloning of three promoter- (CMV, DCX,

and MBP) NTR inserts (containing an NTR-mcherry fusion protein) from a lentiviral plasmid

vector (Figures 1a-1c) into a piggyBac transposon vector (Figure 1d). After several

unsuccessful attempts at removing the NTR inserts via restriction digests at flanking regions,

blunting both the insert and vector ends via Klenow (E. coli DNA polymerase I) fragment action

(not shown), eventual success in obtaining positive clones was achieved through adding flanking

MluI restriction sites via PCR amplification of the NTR inserts. Both the NTR inserts and

piggyBac vector plasmid were then digested with the MluI restriction endonuclease and ligated

with T4 DNA ligase (described in depth in Chapter 3: Materials and Methods). DH10β E.

coli were transformed through electroporation and grown at 300C on 10cm agar plates to obtain

single colonies. Plasmid DNA from cultured single colony clones was then extracted via

miniprep protocol and positive clones containing the successfully ligated piggyBac vector

plasmid with promoter-NTR inserts were confirmed both by MluI restriction digests and

sequencing (Figure 2a-d).

Page 86: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

68

Figure 1a: CMV-NTR: original lentiviral plasmid ~10.9kb

(top) and subcloning insert form ~2.1kb (bottom). The

original subcloning attempt involved cutting the CMV-NTR

insert out of the lentiviral plasmid via an NheI+XbaI double

restriction digest with blunt end cloning. The successful attempt

instead entailed the use of Q5-PCR amplification to add MluI

sites (near the XbaI (5113) and NheI (3062) sites respectively)

for “sticky-end” (5’ and 3’ overhangs) subcloning into the

piggyBac vector (see Figure 1d).

1a

Page 87: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

69

Figure 1b: DCX-NTR: original lentiviral plasmid ~12.3kb

(top) and subcloning insert form ~3.5kb (bottom). The

original subcloning attempt involved cutting the DCX-NTR

insert out of the lentiviral plasmid via an NheI+XbaI double

restriction digest with blunt end cloning. The successful attempt

instead entailed the use of Q5-PCR amplification to add MluI

sites (near the XbaI (6531) and NheI (3062) sites respectively)

for “sticky-end” (5’ and 3’ overhangs) subcloning into the

piggyBac vector (see Figure 1d).

1b

Page 88: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

70

Figure 1c: MBP-NTR: original lentiviral plasmid ~12.3kb

(top) and subcloning insert form ~3.5kb (bottom). The

original subcloning attempt involved cutting the MBP-NTR

insert out of the lentiviral plasmid via an NheI+XbaI double

restriction digest with blunt end cloning. The successful attempt

instead entailed the use of Q5-PCR amplification to add MluI

sites (near the XbaI (6526) and NheI (3062) sites respectively)

for “sticky-end” (5’ and 3’ overhangs) subcloning into the

piggyBac vector (see Figure 1d).

1c

Page 89: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

71

Figure 1d: PiggyBac Transposon Vector Plasmid. All three

promoter-NTR inserts (Figures 1a-c) were subcloned into the

above vector at the MluI restriction site (7336, outlined in red)

via PCR amplification to add flanking MluI restriction sites to

each NTR insert, followed by the MluI digest of the vector and

all inserts to create compatible 5’ and 3’ overhang “sticky-ends”.

An unrelated 600bp insert (5335 -> 6022, outlined in blue) was

removed following ligation of the NTR inserts from each of the

three resultant NTR-piggyBac plasmids via BsaI restriction

digests (due to sites flanking the insert) and self-ligation.

1d

Page 90: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

72

1e

Figure 1e: PiggyBac Transposon Vector containing the

subcloned CMV-NTR insert. The primers used to PCR-amplify

the CMV-NTR insert and add flanking MluI restriction sites (8768

and 6653) are indicated in purple (MluI-CMV-F (6653-6683) and

MluI-CMV-R (8743-8774)). Positive clones were confirmed both

through MluI restriction digest to release the CMV-NTR insert (see

Figure 2c) and through sequencing of a region spanning 500bp both

up and downstream of the CMV-NTR insert (indicated in blue

above).

Page 91: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

73

Figure 1f: PiggyBac Transposon Vector containing the subcloned

DCX-NTR insert. The primers used to PCR-amplify the DCX-NTR

insert and add flanking MluI restriction sites (10,340 and 6653) are

indicated in purple (MluI-DCX-F (6653-6685) and MluI-DCX-R

(10,322-10,346)). Positive clones were confirmed both through MluI

restriction digest to release the DCX-NTR insert (~3.5kb, see Figure 2d)

and through sequencing of a region spanning 500bp both up and

downstream of the DCX-NTR insert (indicated in blue above).

1f

Page 92: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

74

Figure 1g: PiggyBac Transposon Vector containing the subcloned

MBP-NTR insert. The primers used to PCR-amplify the MBP-NTR

insert and add flanking MluI restriction sites (10,175 and 6645) are

indicated in purple (MluI-MBP-F (6645-6677) and MluI-MBP-R

(10,147-10,181)). Positive clones were confirmed both through MluI

restriction digest to release the MBP-NTR insert (~3.5kb, see Figure 2b)

and through sequencing of a region spanning 500bp both up and

downstream of the MBP-NTR insert (indicated in blue above).

1g

Page 93: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

75

2c 2d

Figure 2: Agarose DNA Gel Restriction Digests of NTR Inserts and PiggyBac Transposon Vector. a)

PCR amplified and MluI-digested: 1=CMV-NTR insert; 2&3=DCX&MBP-NTR inserts; 4=piggyBac vector;

5=1kb+ DNA ladder. b) Successfully ligated MBP-NTR-piggyBac positive clone confirmations via MluI

digest and release of ~3.5kb MBP-NTR insert: 2,4,6,8,10=uncut MBP-NTR-piggyBac plasmid;

3,57,9,11=MluI-cut MBP-NTR-piggyBac; 12->13=uncut & MluI cut piggyBac vector respectively;

1&15=1kb+ DNA ladder. c) Successfully ligated CMV-NTR-piggyBac positive clone confirmations via

MluI digest and release of ~2.1kb CMV-NTR insert: 1,3,5,7,9,11,15=uncut CMV-NTR-piggyBac plasmid;

2,4,6,8,10,12,16=MluI cut CMV-NTR-piggyBac; 13->14=uncut & MluI cut negative clone (no CMV-NTR

insert); 17-18=uncut & MluI cut piggyBac vector; 19=1kb+ DNA ladder. d) Successfully ligated DCX-NTR-

piggyBac positive clone confirmations via MluI digest and release of ~3.5kb DCX-NTR insert: 1=MluI cut

DCX-NTR-piggyBac plasmid; 2= uncut DCX-NTR-piggyBac; 3= 1kb+ DNA ladder.

2a 2b

Page 94: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

76

2) Mtz Treatment of CMV-NTR NPCs

After successful subcloning and confirmation of positive NTR-piggyBac plasmid clones (as well

as separate removals of the unrelated 600bp insert followed by self-ligation (not shown for

brevity)), endotoxin-free purified plasmid DNA was obtained via culturing a large volume

(500mL) of each plasmid’s single colony clone and extracting plasmid DNA through maxiprep

protocol. Three separate monoclonal lines of human iPSC-derived NPCs were generated via

electroporation with each of the NTR-piggyBac plasmids separately, FACS sorting, single cell

selection and prolonged expansion. CMV-NTR-NPCs would theoretically have been already

expressing NTR, and thus preliminary ablations using four concentrations of Mtz and six

timepoints were set up and evaluated qualitatively (Figure 3).

Figure 3: Preliminary attempts at Mtz ablation of CMV-NTR NPCs (e-h) in comparison with

WT-NPCs as a negative control (a-d). 1.25x104 NPCs were originally plated in each well of

separate 24-well plates, and after 24 hours, Mtz treatments began via media changes. Left columns

(a, c; & e, g ) depict the 4-hour timepount post-Mtz treatment, while the right columns (b, d; & f, h)

depict the 72 hour timepoints. Top rows (a-b; & e-f) indicate control wells receiving no Mtz

treatment, while the bottom rows (c-d; & g-h) received 10mM Mtz. For both WT-NPC controls and

CMV-NTR NPCs, cell proliferation and general cell quantities were relatively unchanged with

10mM Mtz (bottom rows) in comparison to control NPCs receiving no Mtz treatment (top rows).

Scale bars = 100µm.

3

Page 95: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

77

Mtz treatment up to 10mM did not appear to have any noticeably significant effects on total

remaining cell density of CMV-NTR NPCs after 72 hours. Due to such an unexpected result,

two principal attempts at troubleshooting these difficulties with NTR-Mtz ablation of my hiPSC-

derived, CMV-NTR-NPC line were pursued.

3) NTR-Mtz Ablation System Troubleshooting

Both (I) total protein staining (Figure 4- for investigating NTR translation, due to the lack of an

existing antibody), and (II) the generation of human codon-preference usage-optimized CMV-

NTR-NPCs (HCO-NTR-NPCs) (Figure 5), wherein the potential issue of low NTR transcription

due to prokaryotic (E. coli) codon-optimization could potentially be tackled were pursued as

options for troubleshooting NTR-Mtz -CMV-NTR-NPC ablation.

Figure 4: SDS PAGE and Coomassie Brilliant Blue G-250 Dye total protein stain of 20µg of protein

extract obtained from CMV-NTR-NPCs and WT-NPCs respectively by sonication. The polyacrylamide

gel was composed of a 4% stacking gel and a 10% resolving/separating gel, and was run for 30 minutes at

80V, followed by 1 hour and 30 minutes at 100V. A faint single band appeared (outlined in red) at the

approximate molecular mass (~50kDA) of the NTR-mcherry fusion protein for the CMV-NTR protein

extract, compared to the less clear band smear evident from the negative control WT-NPC protein extract.

4

Page 96: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

78

5

Figure 5: Attempts at ablating a polyclonal (puromycin-purified) line of CMV-NTR-NPCs using a

human codon preference usage-optimized form of the NfsB/NTR gene (144 silent mutations) referred to

as “HCO-NTR-NPCs”. Unlike the monoclonal line of CMV-NTR-NPCs (see Figure 3e-h), the HCO-NTR

construct did not contain an NTR-mcherry fusion protein, but only a GFP reporter gene as part of the piggyBac

transposon cargo (confirming successful NPC transformation). 1.0x106 HCO-NTR-NPCs were plated on a

10cm culture dish, and treated with 6mM of pre-dissolved, pharmaceutical grade Mtz through a media change.

The plate was imaged once daily at the 24-hour (a) and 48-hour (b) timepoints, and remaining cell density was

monitored qualitatively. Despite potential atrophy and “blebbing” of some cells at the 24-hour timepoint (a)

(circled in red), no major changes in cell density appeared to occur after 48 hours (b) of Mtz treatment. Scale

bars = 122µm.

Page 97: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

79

4) HSV-TK Plasmids and the Creation of HSV-TK NPCs

Due to the negative findings in selectively killing my line of hiPSC-derived NPCs with the

NTR-Mtz ablation system, an alternative approach using the HSV-TK + GCV/BVDU ablation

system was instead pursued- the first step of which entailed finding an appropriate gene transfer

vector (Research Aim 1). A two plasmid, Tol2 transposon-based gene transfer vector for HSV-

TK was selected from plasmids available from the Addgene repository. The Tol2 plasmids that

were selected consisted of: pCMV-Tol2 (Figure 6a) - containing the Tol2 transposase, and

pkTol2P-PTK (Figure 6b) - containing the actual Tol2 transposon carrying a puromycin-

resistance-HSV-TK fusion protein gene driven by a PGK promoter.

6a

Figure 6a: pCMV-Tol2 plasmid encoding the Tol2 transposase enzyme. Important

features of interest include an ampicillin resistance gene (top left in light green) for

bacterial selection, and a CMV-enhancer-promoter combination (top centre in white)

driving a Tol2 transposase gene (right side, downstream (below) the Kozak consensus

sequence (supporting eukaryotic translation)), and SV40 small T intron (mammalian gene

expression/transcription). pCMV-Tol2 was required for providing the Tol2 transposase

enzyme in order to excise the Tol2 transposon cargo (puromycin-resistance-HSV-TK

fusion protein gene- PTK) from plasmid pKTol2-PTK using the flanking Tol2

recognition sites and support integration into the genomes of target NPCs.

Page 98: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

80

6b

Figure 6b: pKTol2P-PTK plasmid encoding the Tol2 transposon and puromycin-

resistance-HSV-TK fusion protein gene (PTK). Important features of interest include a

kanamycin resistance gene (top left in light green) for bacterial selection, a Kozak consensus

sequence (supporting eukaryotic translation), and the Tol2 transposon itself carrying the PTK

gene (~500bp downstream/below primer Tol2-PTK-F1 (539-558) marked in purple) driven

by a PGK promoter. The main sequencing primers (marked in purple) indicating the relative

locations of each respective feature (~500bp downstream of each primer) were: Puro-F

(1244-1264, for the puromycin-resistance gene portion of PTK), mPGK-F (658-677, for the

PGK promoter), and Tol2-PTK-F1 (539-558, for the main portion of the Tol2 transposon

recognition sites and PTK gene).

Page 99: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

81

5) GCV Bystander Effect: Mixed HSV-TK & HCO-NTR-NPCs

After plasmid DNA purification, and confirmation through restriction digest and sequencing

(described in depth in Chapter 3: Materials and Methods- Section 4), the two plasmid, Tol2

transposon system (Figures 6a-b) was used to transform cells from the same original line of

hiPSC-derived NPCs (used to generate the CMV-NTR-NPCs described earlier) and purified

through puromycin. Bystander effects were evaluated as HSV-TK NPCs were plated along with

HCO-NTR-NPCs in 10cm culture plates, both separately (Figure 7a-b) and together at equal

ratios (Figure 7c) and treated with 1µg/mL GCV dissolved in media for 96 hours (three 10cm

plates total: HCO-NTR-NPCs + GCV (Figure 7a, e), HSV-TK NPC + GCV (Figure 7b, f), and

1:1 mixed HSV-TK and HCO-NTR-NPCs + GCV (Figure 7c,g ). A fourth control plate of

equal-ratio-mixed HSV-TK and HCO-NTR-NPCs (Figure 7d, h) was additionally maintained.

7

Figure 7: The evaluation of bystander effects of the HSV-TK + GCV ablation system in human-iPSC-

derived NPCs. 1.0x106 cells were plated on each of four separate, 10cm culture plates and subjected to

1µg/mL GCV (or regular media for the control plate) through media changes after 24 hours. Top (a-d) and

bottom row (e-h) images represent the 24-hour and 96-hour timepoints respectively. The cell composition of

each plate was as follows: (a&e) HCO-CMV-NTR GFP+ NPCs; (b&f) HSV-TK NPCs; (c&g) and (d&h)

(negative control) equal ratio-mixed HSV-TK and HCO-NTR-NPCs. Scale bars = 122µm (a-d), 125µm (e-h).

Page 100: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

82

After 96 hours of treatment with 1µg/mL of GCV, HCO-NTR-NPCs were as expected (since

they should be unable to phosphorylate GCV into its active, triphosphate form), relatively

unaffected (Figure 7e), while considerable cell death was observed qualitatively for both HSV-

TK NPCs (Figure 7f), and the mixed cells (Figure 7g). Especially noteworthy was the large-

scale death of GFP+, HCO-NTR-NPCs in the mixed cell population (Figure 7g), indicating the

obvious presence of a bystander cell killing effect for the HSV-TK NPCs after GCV treatment.

6) GCV/BVDU Bystander Effects: Mixed HSV-TK & WT-NPCs

8

Figure 8: The evaluation of bystander effects of the HSV-TK + GCV ablation system in hiPSC-

derived NPCs. 5.0x104 cells (1:1 mixed HSV-TK and GFP+ WT-NPCs) were plated on each well of two

separate, 6 well culture plates and subjected to increasing concentrations of GCV (0.0625-1µg/mL) through

media changes after 24 hours. The left half (a-f) represent the 0-hour timepoint, wherein GFP+ cells were

present in all wells, whereas the right half (g-l) represent the 96-hour timepoint, wherein considerable cell

death of both cell types (including virtually all GFP+ WT-NPCs) was observed for all GCV concentrations.

Scale bars = 99µm (a-c), 100µm (d-l).

Page 101: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

83

Due to the substantial bystander effects observed with GCV ablation of HSV-TK NPCs,

targeted cell elimination with an additional prodrug, BVDU, understood to exert a reduced

bystander effect upon ablation of HSV-TK-expresing cells, was pursued (reviewed in Dachs et

al., 2009). For the comparison of bystander effects between GCV and BVDU, two six well

culture plates were established wherein 5x104 cells consisting of equal ratio mixed HSV-TK

NPCS, and GFP+ WT-NPCs (also hiPSC-derived) were plated per well and subjected to

increasing concentrations (0.0625-1µg/mL) of either GCV (Figure 8) or BVDU (Figure 9).

9

Figure 9: The evaluation of bystander effects of the HSV-TK + BVDU ablation system in hiPSC-derived

NPCs. 5.0x104 cells (1:1 mixed HSV-TK and GFP+ WT-NPCs) were plated on each well of two separate, 6

well culture plates and subjected to increasing concentrations of BVDU (0.0625-1µg/mL) through media

changes after 24 hours. The left half (a-f) represent the 0-hour timepoint, wherein GFP+ cells were present in

all wells, whereas the right half (g-l) represent the 96-hour timepoint, wherein considerable cell death of both

cell types (including virtually all GFP+ WT-NPCs) was observed for all BVDU concentrations. Scale bars =

100µm.

Page 102: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

84

Both GCV and BVDU treatment of the mixed NPC populations for 96 hours (with all

concentrations) led to the noticeable depletion (in terms of remaining attached cells) of nearly all

GFP+ WT-NPCs (Figures 8-9). As such, a bystander effect was clearly apparent for both

prodrugs in combination with HSV-TK-expressing hiPSC-derived NPCs. I next attempted to

quantitatively evaluate the ablational/cell-killing efficiency of both the GCV and BVDU

prodrugs solely on HSV-TK NPCs (Figures 10-12).

7) GCV and BVDU Successfully Ablate HSV-TK hiPSC-NPCs

Figure 10: The ablation of proliferating hiPSC-derived HSV-TK+ NPCs with GCV (0.0625-

1µg/mL) for 96 hours. Four 6-well plates of 5.0x104 initial NPCs were set up and each well received

increasing concentrations of GCV (b-g). Quantification of attached cells/well was accomplished via

DAPI staining and peak analysis image processing in ImageJ, displayed above as: a) after 96 hours, h)

from 24-96 hours; statistical comparisons were performed via analysis of variance (ANOVA). Scale

bars = 100µm. Data was collected from the means of three replicate experiments (N=3).

10

Page 103: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

85

11

Figure 11: The ablation of proliferating hiPSC-derived HSV-TK+ NPCs with BVDU (0.0625-

1µg/mL) for 96 hours. Four 6-well plates of 5.0x104 initial NPCs were set up and each well received

increasing concentrations of BVDU (b-g). Quantification of attached cells/well was accomplished via

DAPI staining and peak analysis image processing in ImageJ, displayed above as: a) after 96 hours,

h) from 24-96 hours; statistical comparisons were performed via analysis of variance (ANOVA).

Scale bars = 100µm. Data was collected from the means of three replicate experiments (N=3).

Page 104: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

86

Figure 12: Comparison of the ablation of hiPSC-derived HSV-TK+ NPCs with GCV and BVDU

(0.0625-1µg/mL for both) after a) 96 hours, and b) 24-96 hours. Eight (4 per prodrug) 6-well

plates of 5.0x104 initial NPCs were set up and each well received increasing concentrations of

GCV/BVDU. Quantification of attached cells/well was accomplished via DAPI staining and peak

analysis image processing in ImageJ; statistical comparisons were performed via ANOVA. No

significant differences were detected between a) equal concentrations of GCV & BVDU after 96

hours, nor b) equal concentrations of GCV & BVDU across time points.

12

Page 105: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

87

Quantification of the ablational efficiencies of both GCV and BVDU was set up as described in

Chapter 3: Materials and Methods and displayed above in Figures 10-12. After 96 hours of

GCV treatment (Figure 10a-g), all concentrations used were found to produce significant

(p<0.01) differences (detected via ANOVA) over the control well in terms of the mean estimated

number of remaining attached cells. BVDU ablation produced similar results (Figure 11a-g),

albeit slightly more gradual, as the lowest concentration used (0.0625µg/mL) did not produce a

significant difference in remaining attached cells over the control. The 1 µg/mL concentration of

both prodrugs resulted in a reduction in attached cells of over 80% compared to controls

(Figures 10a &11a). No significant differences were detected in the estimated number of

attached cells between 96-hour treatments of GCV and BVDU (Figure 12a). Similarly, no

significance was detected comparing the quantities of attached cells of each prodrug’s treatments

individiually across timepoints (Figures 10h and 11h), nor for comparing the same treatment

concentration of both prodrugs across timepoints (Figure 12b).

As the cells were proliferative, even in the highest concentration of each prodrug used (1µg/mL),

there were still many more cells remaining than initially plated (~2x105 vs 5 x104) however the

proliferative ability was certainly significantly reduced compared to controls (1.2-1.4 x106cells

remaining). Additionally, as the method used for cell quantification only took into account

attachment to each well, and did not assess viability, it is considerably possible that the vast

majority of cells remaining in the wells of the highest concentrations of each prodrug treatment

were in fact non-viable (possibly undergoing apoptosis), as may be discerned from Figures 10g

and 11g.

The final step in assessing GCV and BVDU ablation lay in the elimination of a potential

confounding variable- namely that of DMSO, the solvent in which both prodrugs had been diluted

in prior to mixing with the culture media used for the experiments in Figures 10-12. In order to

determine that the cell-killing/proliferative inhibition effects seen for both GCV and BVDU had

not been caused by DMSO cytotoxicity, a minor experiment was set up whereby 5.0x104 HSV-TK

NPCs were plated in each of two wells of a 6-well plate, and after 24-hours, subjected to media

Page 106: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

88

changes containing either 0.4% dissolved DMSO (the highest concentration used for GCV/BVDU

dilution) (Figure 13c), or 0.4% dissolved PBS (Figure 13a). Each well was imaged at the starting

(0-hour) and 48-hour timepoints to obtain a general qualitative determination of any potential

DMSO-related toxicity. No obvious discernable effects on cell density/proliferation were detected

with 0.4% DMSO treatment of HSV-TK NPCs (Figure 13d).

13

Figure 13: Assessing the potential for DMSO cytotoxicity with HSV-TK NPCs.

5.0x104 HSV-TK-NPCs each were plated in two wells of a 6-well culture plate and

after 24-hours subjected to media treatments containing: a) 0.4% PBS, or c) 0.4%

DMSO. After 48 hours of treatment (b and d), the wells were imaged, and general cell

density was assessed qualitatively. Both the treatments of 0.4% PBS (b) and 0.4%

DMSO (d) appeared to have no effect on the general proliferation of HSV-TK-NPCs.

Scale bars = 100µm.

Page 107: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

89

Chapter 5: General Discussion, Future Directions & Conclusion

Overall, a simple, “proof of concept” has now been demonstrated for the successful ablation of

hiPSC-derived NPCS with the HSV-TK + GCV/BVDU ablation system through the preceeding

work in Chapter 4: Results. Numerous challenges and pitfalls were overcome throughout the

course of my MSc project and I shall attempt to address and elaborate on the most germane and

critical points relating to each experiment, as well as the broader extrapolations of my work in

relation to the grander picture within which it should be framed. There are numerous

possibilities for building upon my work and various avenues and directions to embark on for

future investigations.

5.1 Summary of HSV-TK+GCV/BVDU Results

The most critical results obtained in this project are the effective ablation/proliferation reduction

of proliferating human iPSC-derived NPCs using the HSV-TK +GCV/BVDU ablation system.

A reduction in the estimated total number of attached cells of 80% or greater over controls was

detected for both the GCV and BVDU prodrugs at the highest concentrations used (1µg/mL)

after 96 hours (Figures 10a &11a). As mentioned previously in Chapter 5: Results, no

significant differences were detected between equivalent concentrations of the GCV and BVDU

prodrugs after 96 hours of treatment in terms of the estimated quantity of attached HSV-TK

NPCs per well of each plate (Figures 10h and 11h). Additionally, no significant differences in

regards to the estimated number of remaining attached cells were detected among the four-day

time points for equivalent concentrations of either GCV and BVDU alone, nor for GCV and

BVDU compared together (Figures 10h, 11h, 12b).

In terms of bystander effects, the obvious presence of one was first detected for GCV when

HSV-TK NPCs were cultured together with HCO-NTR NPCs (Figure 7g), as the vast majority

(e.g., all but 2 cells in the representative image depicted) of GFP+ HCO-NTR NPCs no longer

remained attached to the plate after four days of 1µg/mL GCV treatment. Additional bystander

effects were detected after 96 hours for all concentrations of GCV employed (0.0625-1µg/mL)

from the combined equal culture of HSV-TK NPCs and GFP+ WT-NPCs (Figure 8h-l). All

Page 108: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

90

BVDU treatments (0.0625-1µg/mL) similarly to GCV, also displayed bystander effects after 96

hours of treatment (Figure 9h-l), as identical to GCV, they also resulted in the elimination of all

GFP+ WT-NPCs.

5.2 HSV-TK+GCV/BVDU Ablation in Context

Concerning the HSV-TK+ GCV/BVDU ablation system, my results have indicated the existence

of a bystander effect for both prodrugs when utilized to ablate hiPSC-derived, HSV-TK-

transformed NPCs (as indicated by Figures 7g, 8h-l, and 9h-l). My personal findings, although

only rudimentary and qualitative with relation to bystander effect, nevertheless are partially as

expected by literature, as both prodrugs are known to possess some degree of bystander effect

(due to GCV-monophosphate and BVDU-diphosphate “spillover” to neighbouring cells through

Cx43 channels); however, it was not possible to detect an obvious difference between the

bystander effects of GCV and BVDU (reviewed in Dachs et al., 2009).

Although BVDU was predicted to possess a reduced bystander effect over GCV, such an

investigation was deemed to be beyond the scope of this project, and thus future studies will be

necessary for firmly establishing any significant differences in prodrug bystander effects in

regards to ablating hiPSC-derived HSV-TK NPCs (reviewed in Degrève et al., 1999).

As mentioned briefly in Chapter 4: Results, Section 7, many if not most of the remaining

attached cells may not have been viable, and either in the process of, or recently underwent

apoptosis, and simply not yet detached (possibly inferred from Figure 10e-g and 11e-g). No

significant differences were detected in terms of estimated attached cell number between

equivalent treatments of GCV and BVDU (Figure 12), and thus although a difference may exist

between the ablational efficiencies of each prodrug, my personal experiments could not discern

any. Perhaps future studies employing wider prodrug concentration ranges and longer

timepoints may be able to determine if any ablational efficiencies exist between GCV and

BVDU specifically for hiPSC-NPCs.

Although the mechanisms of apoptosis induced by GCV and BVDU are indeed understood to be

similar (as both are nucleotide analogs inducing apoptosis primarily through incorporation into

replicating genomic or mitochondrial DNA causing chain-termination), there are most certainly

minor, yet perhaps critical differences between these mechanisms, such as BVDU’s ability to

Page 109: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

91

target thymidylate synthase, and GCV’s ability to induce extrinsic, Fas-pathway apoptosis

(involving DISC-formation) without the activation of Fas ligand (Balzarini et al., 1987; Beltinger

et al., 1999). These aforementioned differences in the apoptotic pathways induced for GCV and

BVDU could play a role in ablational efficiency differences, if any are yet detected for hiPSC-

derived NPCs.

Although significance was determined for the difference in the estimated number of attached

cells remaining after 96 hours for all treatments of GCV (Figure 10a) and for all but the lowest

concentration of BVDU (Figure 11a), there were still large quantities of cells remaining

(~2x105), even for the highest concentrations used. Since the HSV-TK NPCs were cultured and

treated with GCV and BVDU in SF9 media, they were certainly proliferative, and it is possible

that the concentrations of GCV/BVDU used may not have been completely sufficient to

overcome proliferation and accomplish both cell death and detachment from wells for all HSV-

TK NPCs. Such a result is understandable considering the previous reported difficulties in

achieving complete ablation and removal of proliferating NPCs (alebit ones of ESC origin) with

the HSV-TK+GCV ablation system (Tieng et al., 2016).

5.3 Advantages and Challenges of HSV-TK+GCV/BVDU Ablation

The proof of concept tool for the ablation of hiPSC-derived NPCs using the HSV-

TK+GCV/BVDU system herein demonstrated offers several critical advantages for further

usages, along with the caveat of significant challenges. Through the application of 1µg/mL of

either GCV or BVDU for a time duration of 96 hours, it is possible to reduce the number of

remaining cells in vitro by at least 80% over controls, allowing for targeted cell death of

proliferating NPCs at the very least. For future in vivo usages, effective concentrations and

timepoints established will likely require modification, however as a GCV dosage of

100mg/kg/day has previously been shown to effectively ablate GFAP+ endogenous murine

astrocytes without resulting in any serious general toxicity related side effects, reaching a similar

internal GCV concentration as demonstrated in my in vitro work should likely be achievable

without serious toxicity related side effects (Bush et al., 1998).

Both GCV and BVDU are understood to be able to cross the blood brain barrier/blood spinal

cord barrier and are additionally FDA approved substances, allowing for potential future clinical

usages of either prodrug, if it is deemed effective and necessary to employ them for controlling

Page 110: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

92

the proliferation of transplanted hiPSC-derived NPCs in order accomplish improvements in

terms of physical repair and functional recovery (Tieng et al., 2016; reviewed in De Clercq

2005). Additionally, while GCV is considered a potential carcinogen and posses a number of

potentially serious side effects, such as anaemia, nausea, vomiting, diarrhea, headaches, and

confusion, BVDU offers the advantage of carcinogenic-free, and relatively side-effect free

ablation, with only mild nausea as an uncommon adverse reaction (reviewed in Zhang et al.,

2015; reviewed in De Clerq 2005).

Two major, yet certainly not insurmountable challenges exist for the HSV-TK+GCV/BVDU

ablation system with the targeted elimination of hiPSC-derived NPCs. These challenges are

specifically: (1) the presence of an obvious bystander cell-killing effect, and (2) the potential

difficulty in ablating non-proliferating/post-mitotic cells. The bystander effect for both the GCV

and BVDU prodrugs as observed in the results of this thesis (Figure 8h-l & 9h-l) presents an

obstacle in terms of the selectivity of ablation- (e.g., when employed for in vivo SCI

neuroregeneration studies with transplanted HSV-TK hiPSC-derived NPCs, will potential side-

effects occur and will neurorepair be hampered due to the unwanted death of neighbouring

cells?). The bystander effects for both the GCV and BVDU prodrugs are understood to be

dependent on Cx43 gap junction expression, cell type, and proximity among other factors

(reviewed in Dachs et al., 2009; reviewed in Degrève et al., 1999).

Realistically, a bystander effect may be unavoidable in HSV-TK+GCV/BVDU ablation,

however as previous studies employing both prodrugs have demonstrated successful targeted

ablation in vivo of neural cell types without obvious adverse reactions due to bystander effects,

achieving similar results should theoretically not be an overly arduous task (Tieng et al., 2016;

Field et al., 1984). As BVDU ablation of HSV-TK hiPSC-derived NPCs has been demonstrated

in this thesis to be as effective GCV ablation, and as BVDU has been previously demonstrated to

exert a greatly reduced bystander effect, the challenge of bystander effects in vivo could partially

be mitigated, albeit not removed entirely through the use of BVDU instead GCV (reviewed in

Dachs et al., 2009; reviewed in Degrève et al., 1999). Additionally, bystander effects for in vivo

transplantations of my HSV-TK hiPSC-derived NPCs could be potentially be further reduced

through the administration of substances known to block Cx43 channels, such as 18 alpha-

glycyrrhetinic acid or 1-octanol (Princen et al., 1999).

Page 111: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

93

Concerning challenge (2): the potential difficulty in ablating non-dividing/post-mitotic cells

(e.g., NPC-derived neurons), increasing the dosage of GCV/BVDU should theoretically allow

for effective cell ablation through the incorporation of GCV/BVDU-triphosphate into replicating

mitochondrial DNA during mitochondrial turnover, and the resultant induction of the intrinsic,

mitochondrial apoptotic pathway (Beltinger et al., 1999, Laberge et al., 2013). As previous

research has achieved successful ablation of HSV-TK expressing, senescent human fibroblasts

using mildly increased concentrations of GCV, and confirmed that it occurred through the

mitochondrial pathway via the detection of mitochondrial death signals, it would in the opinion

of this author, be a fair deduction that such should be accomplishable for targeting post-mitotic

neurons differentiated from transplanted hiPSC-derived HSV-TK NPCs (Laberge et al., 2013).

Nevertheless, if the concentrations required for achieving post-mitotic cell ablation in vivo in SCI

model rats is determined to be in excess of the general toxicity range (e.g., greatly in excess of

100mg/kg/day), then the ablation tool will still be effective for controlling transplanted hiPSC-

derived NPC proliferation (overarching question (3) from Chapter 2: Research Aims), thus

allowing for studies to occur in which the optimal amount of proliferation/NPC quantity required

for effective neuroregeneration may be determined.

5.4 Reflections on NTR-Mtz Ablation Approaches

With regards to the sub-cloning of each of the NTR-inserts into the piggyBac transposon plasmid

vector, considerable difficulties were encountered initially, largely believed to be inherent to the

large sizes of all three inserts, as well as the target vector plasmid. As the early attempts at NTR-

piggyBac subcloning involved the use of blunt-ends (no compatible 5’ or 3’ overhangs), relying

merely on random collisions of molecules for ligation of insert and vector, it is understandable

for the continued lack of success in obtaining positive clones, considering that even the smallest

NTR insert fragment (CMV-NTR) was over 2kb in size. Previous research has indicated similar

difficulties with regards to blunt-end subcloning of transgenes into gene transfer vectors wherein

both inserts and vectors were of such aforementioned (2-3kb for inserts, 10kb+ for vectors)

(reviewed in Kantor et al, 2014; reviewed in Manfredsson et al., 2016).

Even with the initial “eureka-moment” successes of obtaining positive NTR-piggyBac clones

through PCR-amplification adding MluI-restriction sites for compatible end cloning of the NTR

inserts and piggyBac vector, additional steps and verifications were required, in the form of

Page 112: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

94

further restriction digests to remove the unrelated insert, which itself proved to be a more

difficult task than previously expected, as self-ligation of the linearized, NTR-piggyBac

plasmids, or less likely, successful transformation of DH10β E. coli also required multiple

attempts for success. Although in hindsight, possibly additional planning and experimental

design of the overall strategic steps for NTR-piggyBac subcloning may have considerably

shortened the timeframe with which that section of my MSc project elapsed, the strategies and

steps that were pursued nevertheless provided valuable skills and experience.

Additionally, despite the negative findings with the Mtz ablation of CMV-NTR-NPCs created

through the use of the subcloned CMV-NTR-piggyBac plasmid, the considerable effort and time

spent creating the plasmid through extensive molecular biology should not perhaps be considered

wasted, as in addition to providing the opportunity for learning techniques, it created a safe, and

efficient gene-transfer vector containing a useful suicide gene for potential future attempts at

achieving success with the NTR-Mtz ablation system of hiPSC-derived NPCs or other human

neural cell types in the broader context of neuroregeneration loss-of-function studies (reviewed

in Kantor et al., 2014). As there have yet to be any human or other mammalian neural cell

reported usages of the NTR-Mtz ablation system, the plausible future success of the system with

the targeted killing of hiPSC-derived NPCs would certainly still be a novel and noteworthy

technique (reviewed in Williams et al., 2015).

Gene expression of NTR/NfsB in either the CMV-NTR-NPC monoclonal line, or HCO-NTR-

NPC polyclonal line was not assessed during the course of my MSc work, and thus may be a

potential culprit for the lack of Mtz ablational success demonstrated. In hindsight, rather than

pursuing the possible issue of codon-optimization, the hurdle for which HCO-NTR-NPCs were

created as an attempt to solve, a more immediate solution of investigating NTR/NfsB gene

expression should likely have been taken, which would have theoretically been simple through

quantitative real-time PCR (qRT-PCR). The obstacle towards NTR-Mtz ablational success may

have also been located downstream, in the form of protein expression, however there is currently

no readily available, commercial antibody for NfsB, and thus Coomassie blue total protein

staining of CMV-NTR-NPC protein extract was pursued instead. Despite the possible presence

of a faint band (Chapter 4: Results, Section 3, Figure 4), it is not immediately obvious that

NTR protein expression was present, as not only could the band represent a different protein of

Page 113: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

95

equivalent size, the control sample still showed a blurry/very faint band at the same mass of

~50kDa, obfuscating any certainty of detected NTR protein.

As mentioned briefly earlier in Chapter 3: Materials and Methods, Section 2, targeted

transformation (such as using Gateway cloning® (ThermofisherScientific)) of hiPSC-derved

NPCs with the promoter-NTR constructs may have been a more suitable strategy for pursuing

success, as previous work has indicated that transgene stability is considerably increased

(maintaining genomic integration and transcription over multiple passages) through selective

integration of transgenes into cell-life-vital genes and regions (reviewed in Kantor et al., 2014;

reviewed in Manfredsson et al., 2016; reviewed in Joshi et al., 2017).

If the lack of NTR/NfsB gene/transcript expression was the primary reason behind the apparent

lack of Mtz-mediated ablational success for CMV-NTR NPCs, such a process may have

occurred through repression, silencing or interference (e.g., through methylation, etc.) of the

integrated NTR transgene from NPC genomic elements, which may still be possible despite

multiple piggyBac transposon insertions (~15) likely occurring on average per electroporated

NPC, due to the lack of targeted insertion within cell-life-dependent genes (reviewed in Zhao et

al., 2016; reviewed in Li et al., 2013; reviewed in Grabundzija 2010; reviewed in Wu et al.,

2006). Interference with NTR transgene expression in CMV-NTR NPCs may even have

occurred as a result of the piggyBac transposon’s inherent preference for integration within

transcriptional regions, or due to promoter repression from the minor transcriptional activity of

the piggyBac 5’LTR (reviewed in Vargas et al., 2018; Gogol-Döring et al., 2016; Cadiñanos and

Bradley, 2007).

The strategy of pursuing human codon optimization for NTR expression perhaps should not have

been the immediate first or second choice for troubleshooting the NTR-Mtz ablation system, as

gene transcript expression of CMV-NTR NPCs, and transforming additional cell types as

controls may likely in hindsight have been more efficient and relevant choices for effective NTR

troubleshooting. If an additional cell type had been transformed with the CMV-NTR-piggyBac

plasmid and ablation pursued with Mtz, the possibility of an NPC-specific issue (e.g., resistance

due to exceptionally rapid proliferation, etc.) preventing ablation could perhaps have been

discovered or ruled out. Perhaps issues relating to high NPC proliferation of my hiPSC-NPC

Page 114: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

96

line (especially in serum free hormone mix/SF9 media) or prodrug uptake may have been the

answer to the enigma of unsuccessful ablation.

The concentrations of Mtz (up to 10mM) and timepoints (up to 96 hours) were more than has

been demonstrated sufficient with various zebrafish and frog cell types, yet it is possible that

human-specific differences may have warranted higher concentrations or longer timepoints

(reviewed in Williams et al., 2015; reviewed Sekizar et al., 2015; reviewed in Chung et al.,

2013). Solubility of the Mtz prodrug was considered to be a potential obstacle to ablation,

however it should likely be ruled out, as the ablation of HCO-NTR-NPCs was attempted using a

pre-dissolved, pharmaceutical intravenous sack of Mtz solution. Although potential apoptotic

“blebbing” or atrophy appeared to have been detected after 24 hours of Mtz treatment in HCO-

NTR-NPCs (Figure 5a), after 48 hours no obvious indications of reduced cell density/cell death

were apparent (Figure 5b), indicating that for whichever reason of any of the above stated

(transgene repression, translational issues, cell-type-dependent Mtz resistance due to rapid

proliferation) HCO-NTR-NPC ablation with Mtz had proven more challenging than originally

forseen.

5.5 Potential Applications and Future Directions of HSV-Tk+GCV/BVDU hiPSC-Derived NPC Ablation

There are numerous possibilities for potential applications and future directions of the HSV-TK

hiPSC-derived NPCs developed in this thesis. An obvious usage as outlined earlier is the in vivo

ablation of the cell line post-transplantation in SCI rats at different time points (e.g.,

administering GCV/BVDU at 2h, 12h, 24h, 48h, 72h, 96h post transplantation and 14 days post-

injury etc.) and comparing the changes in physical repair (as could be detected via histology

using human nuclear antigen as a potential marker of transplanted cells) and functional recovery

(as detected via behavioural tests- e.g., catwalk, grip strength, etc.) to animals receiving the same

hiPSC-derived NPC transplantions without ablations. Bystander effects could theoretically be

detected in vivo through Western blots comparing apoptosis markers (e.g., caspase 3 etc.) from

spinal cord sections of both ablated and non-ablated, hiPSC-NPC transplant animals.

Although the HSV-TK+GCV/BVDU ablation approach developed in this thesis used only a

constitutive PGK promoter allowing for ablation of all transplanted cell types, through

subcloning work, cell-type specific promoters (e.g., DCX, MBP, GFAP, etc.) could be added

Page 115: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

97

instead and thus potentially allow for cell-type-specific ablation (e.g., neurons vs.

oligodendrocytes vs. astrocytes). Such a cell-specific promoter-based approach could potentially

allow for pursuing overarching question (1) (from Chapter 2: Research Aims) in regards to the

effectiveness of oligodendrocytes/remyelination vs. neurons/synaptic re-establishment in

achieving SCI physical repair and functional recovery. Furthermore, the HSV-TK hiPSC-

derived NPCs developed in this thesis could theoretically be additionally employed for pursuing

neuroregeneration studies for other (non-SCI) CNS disorders/conditions/diseases that could

benefit from cell transplantation therapies (e.g., stroke, Parkinson’s, multiple sclerosis, epilepsy

etc.).

An immediately obvious direction for further evaluation of hiPSC-HSV-TK NPC ablation with

GCV and BVDU would be assessing not only the remaining cell density after treatment, but also

the viability of remaining cells, such as could be pursued through MTT assays (discussed in

Chapter 1, Section 3.1.2), or live-dead assays with a combination of markers including calcein

AM (for live cells, due to the requirement for cellular esterase activity to produce a fluorescent

metabolite) and propidium iodide (for dead/non-viable cells due to lack of membrane

permeability in most live cells).

Quantitative differences in bystander effects of GCV and BVDU in ablation of hiPSC-derived

NPCs could likely be accomplished through continuing with the strategy that I had initially

pursued- FACS of both live and dead NPCs after GCV/BVDU treatments at specific timepoints.

The quantitative results of this thesis (Figures 10-12) could indeed be useful for further studies

of GCV and BVDU bystander effects, as effective concentrations and timepoints for ablation

(likely a minimum of 1µg/mL for a minimum of 4 days, the highest timepoint and concentration

used) have already been determined for hiPSC-derived HSV-TK NPCs.

Furthermore, for fine-tuning of hiPSC-HSV-TK NPC ablation, it would likely be necessary to

fully confirm both gene/mRNA transcript expression and protein translation of HSV-TK, which

could be achieved through qRT-PCR and Western blotting respectively. Unlike the NTR/NfsB

protein, HSV-TK does indeed have commercially available antibodies, enabling the possibility

of evaluating and confirming HSV-TK protein expression for the hiPSC-HSV-TK NPC line

created over the course of this MSc project. Determining if the cell death observed in

GCV/BVDU ablation of hiPSC-HSV-TK NPCs is indeed occurring from apoptosis as expected,

Page 116: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

98

and not necrosis would presumably be yet another important potential future direction, and as

such could be investigated through Western blot antibody staining of apoptotic markers, such as:

caspase 3, Bcl-2, cytochrome C, or annexin V (Ma et al., 2017; Zhuo et al., 2016).

As the HSV-TK Tol2 transposon gene-transfer vector system used did not possess a fluorescent

reporter gene, it could be potentially useful for future uses of hiPSC-HSV-TK NPCs to subclone

in a GFP gene for confirmation of successfully transformed NPCs, or even an mCherry gene for

the creation of a puromycin-resistance-HSV-TK-mCherry tri-fusion protein-encoding gene to

allow for visual identification of HSV-TK-expressing NPCs; however mChery fluorescence

should not perhaps be used as the sole indicator of HSV-TK expression, due to the challenges

faced with ablational success of CMV-NTR-NPCs, which similarly were expected to express

(yet may not have) NTR due to visual mCherry expression (faintly visible in Figure 3).

In terms of novelty, although the HSV-TK + GCV enzyme-prodrug ablation system has been

utilized extensively for decades, and has previously been used to target such neural cell types as:

C17.2 immortalized mouse cerebellar NPCs; C6 rat gliomas; GFAP-expressing mouse dNSCs;

and, even human ESC-derived NPCs, there have not currently been any reported usages of the

system for ablating hiPSC-derived NPCs (Tieng et al., 2016; Pu et al., 2011; Uhl et al., 2005; Li

et al., 2005; Morshead et al., 2003). Additionally, there have only been two reported usages of

the HSV-TK+GCV ablation system in an SCI context, specifically from studies that ablated:

endogenous murine NG2+ pericytes and OPCs, and murine GFAP+ reactive astrocytes (Hesp et

al., 2018; Faulkner et al., 2004). BVDU-mediated ablation of hiPSC-derived NPCs is further

considerably even more novel than the use of GCV for the same cell type, as apart from the

treatment of herpetic encephalitis for neurons of both mouse and human origin, there have not

currently been any reported uses of HSV-TK+BVDU ablation for other human neural cell types,

including NPCs (Field et al., 1984; Wigdahl et al., 1984; Wigdhal et al., 1983).

Ultimately, hiPSC-derived HSV-TK NPCs of the line developed in this MSc project should be

used for in vivo ablations after NPC transplants post-SCI in rodent models, which as described in

Chapter 2: Research Aims, is the larger picture within which my own MSc project is framed.

Perhaps through ablation of transplanted HSV-TK NPCs in SCI-model rats, questions (2) and (3)

of the primary unanswered questions with relation to NPC transplantation for SCI

neurorepair/neuregeneration, specifically the issues of: (2) short-term trophic

Page 117: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

99

support/immunomodulation vs. long term migration, differentiation, and integration; and (3) the

amount/timing of proliferation required to occur for transplanted hiPSC-derived NPCs in order to

yield sufficient NPC and NPC-derived cell quantities (or quantities of trophic/immune-

modulating factors) to provide effective SCI physical repair and functional recovery, may yet be

at least partially answered.

It may be pertinent to perform a transcriptomic analysis on the HSV-TK NPC line developed in

this thesis in terms of the gene expression for trophic factors (e.g., BDNF, GDNF, CNTF, etc.)

at various timepoints when subjected to general differentiation conditions as would be

experienced in vivo. Such an experiment may allow for a clearer understanding of the levels of

various trophic factors expressed at various timepoints (e.g., 24h, 48h, 72h, 96h, 1 week, 2

weeks, and finally 3 weeks, etc.) of NPC proliferation and differentiation of the HSV-TK hiPSC-

derived NPC line, which could then assist in determining the most appropriate time to ablate

cells without losing significant trophic/immunomodulatory factor secretion. Additionally, it

would be vital to determine the conversion ratio between in vivo dosage (e.g., 100mg/kg/day

etc.) and the final internal concentration of GCV/BVDU that accumulates resultantly in the

injured cord niche.

As GCV is known to remain stable for up to 10 days, such a task may be achievable (Sachewsky

et al., 2014). Subsequently, in vivo ablations of the HSV-TK hiPSC-derived NPCs post

transplantation in SCI rodents could be performed with administrations of both GCV and BVDU,

using a final concentration range of 0.125µg/mL (the second lowest concentration used of each,

and the lowest concentration showing ablational significance over controls for both prodrugs) up

to 10µg/mL (as such higher concentrations have previously been used to ablate human ESC-

derived NPCs without detectable general toxicity) at timepoints ranging from 24h-post

transplantation up to 3 (or even 6) weeks post-transplantation. As iterated previously,

sensorimotor behavioural changes (reductions in functional recovery) could then be detected via

behavioural test comparisons to animals receiving NPC transplantations without ablations.

Similarly, alterations in the physical repair (e.g., remyelination, endogenous axonal regrowth or

neuronal replacement from transplanted hiPSC-derived NPCs and subsequent synaptic plasticity

reestablishment, etc.) could be detected via performing spinal cord ultra-microtome sectioning

and histology with immunohistochemical staining for for oligodendrocytes (e.g., MBP), and

neurons (e.g., TuJ1, etc.). Differences in synaptic rewiring between transplantation+ablation vs

Page 118: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

100

transplantation-alone animal cords could more specifically be detected via pseudorabies virus

tracing in tract regions (e.g., cortical, dorsal, rubrospinal tracts etc.) adjacent to the injury level

(e.g., C7-T1 injury).

Although the HSV-TK +GCV/BVDU ablation system is expected to be effective at ablating even

non-dividing/post-mitotic cell types, such as mature neurons, through disruption of

mitochondrial DNA replication, in the event that such a process is deemed impractical for

hiPSC-derived NPCs in rodent model SCI usage (perhaps due to the necessary prodrug

concentrations possibly approaching the upper limit of general toxicity), the ablation in vitro

proof of concept demonstration that I have developed could still be used to assess the

aforementioned overarching question (3) (e.g., required amount/timing of transplanted iPSC-

derived NPC proliferation).

Likewise, through future subcloning work to add the DCX and MBP promoters to plasmid

pKTol2P-PTK (carrying the Tol2 transposon and puromycin resistance-HSV-TK fusion protein

gene) in place of the present PGK promoter to allow for selective ablation of early neurons and

myelinating oligodendrocytes respectively (as planned for NTR-Mtz ablation), question (1) in

regards to NPC transplants for SCI neuroepair/regeneration namely the issue of remyelination

versus neuronal regeneration/synaptic connectivity reestablishment (addressed in Chapter 2:

Research Aims) may additionally be elucidated. The three research aims of Chapter 2, namely:

(1) acquiring an effective gene-transfer vector for a suicide gene (originally NTR, and later

HSV-TK), (2) generating a cell line of suicide-gene expressing hiPSC-NPCs; and (3) performing

drug-directed ablations (Mtz originally and later GCV/BVDU) were in the opinion of this author,

realistic and reasonable, especially considering the difficulties and challenges faced with

achieving cell death for the NTR-Mtz ablation system.

Page 119: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

101

5.6 Conclusion

In summary, I have generated a rudimentary proof of concept study of the HSV-TK +

GCV/BVDU ablation system in relation to hiPSC-derived NPCs. A set of three primary research

aims, namely: (1) acquiring an effective gene-transfer vector for a suicide gene (originally NTR,

and later HSV-TK), (2) generating a cell line of suicide-gene expressing hiPSC-NPCs; and (3)

performing drug-directed ablations (Mtz originally and later GCV/BVDU) were developed and

subsequently accomplished. Both the GCV and BVDU prodrugs have been shown to

significantly reduce the number of attached cells remaining after 96-hour treatments with

concentrations up to 1µg/mL (<80% reduction over controls for the highest concentrations used).

I was unable to detect any significant differences between prodrugs in regards to ablational

efficiency, nor in terms of the magnitude of bystander effects, however the presence of one was

obviously discernable for both prodrugs.

Initially the NTR-Mtz ablation system, with constructs involving three promoters, for not only

general NPC ablation, but also targeted ablation of early neurons and myelinating

oligodendrocytes was considered and pursued, however due to challenges and difficulties in

achieving cell death with the NTR-Mtz system, the goals and aims of my MSc research project

were narrowed to the more reasonable and realistic approach of providing initial evaluation of

solely hiPSC-derived NPC ablation. Considerable future work beyond the scope of my own

MSc project will be necessary to evaluate GCV and BVDU-mediated ablation of hiPSC-derived

NPCs, including cell viability determinations and, further bystander effect evaluations, however

HSV-TK-expressing hiPSC-derived NPCs may yet prove successful for examining the larger

picture within which I have attempted to frame their future intended uses, concerning the effects

of NPC-transplant-mediated neurorepair and regeneration for future SCI therapeutic treatment.

Page 120: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

102

References

1.1

1.1.1

Barlow, W.R. (1832). “History of the Case of a Man whose Spine was Fractured, with the

Dissection.” Medico-Chirurgical Transactions. 17:115-20. PMID: 20895582

Wilson, G.E. (1911). “Traumatic Lesions of the Cord without Injury to the Spinal Column.” The

Canadian Medical Association Journal. 1(11):1054-66. PMID: 20310218

Budd, W. (1839). “Contributions to the pathology of the spinal cord.” Medico-Chirurgical

Transactions. 1839;22:153-90. PMID: 20895680

Ott, I. (1879). “Observations on the Physiology of the Spinal Cord.” The Journal of Physiology.

2(1):42-90.8. PMID: 16991274

Nichols, J.L. (1899). “A Study of the Spinal Cord by Nissl's Method in Typhoid Fever and in

Experimental Infection with the Typhoid Bacillus.” The Journal of Experimental Medicine.

4(2):189-215. PMID: 19866906

Pollock LJ, Boshes B, Chor H, Finkelman I, Arieff AJ, Brown M. (1951). “Defects in regulatory

mechanisms of autonomic function in injuries to spinal cord.” The Journal of Neurophysiology.

14(2):85-93. PMID: 14814570

Garstang S.V., Miller-Smith S.A. (2007). “Autonomic nervous system dysfunction after spinal

cord injury.” Physical Medicine and Rehabilitation Clinics of North America. 18(2):275-96.

PMID: 17543773

Stanley, E. (1833). “On Irritation of the Spinal Cord and its Nerves, in connection with Disease

in the Kidneys.” Medico-Chirurgical Transactions. 18(Pt 1):260-80. PMID: 20895605

Thorburn, W. (1888). “The Distribution of Paralysis and Anaesthesia in Injuries of the Cervical

Region of the Spinal Cord.” The British Medical Journal. 2(1460):1382-5. PMID: 20752529

Page 121: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

103

Ahuja, C.S., Wilson, J.R., Nori, S., Kotter, M.R.N., Druschel, C., Curt, A., Fehlings. M.G.

(2017a). “Traumatic spinal cord injury.” Nature Reviews- Disease Primers. 3:17018. PMID:

28447605

Ahuja, C.S., Nori, S., Tetreault, L, Wilson, J., Kwon, B., Harrop, J., Choi, D., Fehlings, M.G.

(2017b). “Traumatic Spinal Cord Injury—Repair and Regeneration.” Neurosurgery. 2017; 80

(3S): S9-S22. PMID: 28350947

Tator CH1, Fehlings MG. (1991). “Review of the secondary injury theory of acute spinal cord

trauma with emphasis on vascular mechanisms.” The Journal of Neurosurgery. 75(1):15-26.

PMID: 2045903

Tator, C.H., (1995). “Update on the pathophysiology and pathology of acute spinal cord injury.”

Brain Pathology. 5(4):407-13. PMID: 8974623

Ackery, A., Tator, C., Krassioukov, A. (2004). “A global perspective on spinal cord injury

epidemiology.” The Journal of Neurotrauma. 21, 10:1355–1370. PMID: 15672627

McDonald, W.I. (1975). “Mechanisms of functional loss and recovery in spinal cord damage.”

Ciba Foundation Symposium. (34):23-33. PMID: 177252

Hachem, L.D., Ahuja, C.S., Fehlings, M.G. (2017). “Assessment and management of acute

spinal cord injury: From point of injury to rehabilitation.” 1:1-11. The Journal of Spinal Cord

Medicine. PMID: 28571527

Saghazadeh A., Rezaei N. (2017). “The role of timing in the treatment of spinal cord injury.”

Biomedicine and Pharmacotherapy. 92:128-139. PMID: 28535416

Nakamura M, Houghtling RA, MacArthur L, Bayer BM, Bregman BS. (2003). “Differences in

cytokine gene expression profile between acute and secondary injury in adult rat spinal cord.”

Experimental Neurology. 184, 1:313–325. PMID: 14637102

Ulndreaj, A., Chio, J.C., Ahuja, C.S., Fehlings, M.G. (2016). “Modulating the immune response

in spinal cord injury.” Expert Review of Neurotherapeutics. 16, 10:1–3. PMID: 27352883

Page 122: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

104

1.1.2

Ahuja C.S., Fehlings M. G. (2016). “Concise Review: Bridging the Gap: Novel

Neuroregenerative and Neuroprotective Strategies in Spinal Cord Injury.” Stem Cells

Translational Medicine. 5(7):914-24. PMID: 27130222

Rhodes, K. E.; Fawcett, J. W. (2004). “Chondroitin sulphate proteoglycans: Preventing plasticity

or protecting the CNS?”. Journal of Anatomy. 204 (1): 33–48. PMID 14690476

Schanne, F.A., Kane, A.B., Young, E.E., Farber. J.L. (1979). “Calcium dependence of toxic cell

death: a final common pathway.” Science. 206, 4419:700–702. PMID: 386513

1.2

1.2.1

Huang, S.Q., Tang, C.L., Sun, S.Q., Yang, C., Xu, J., Wang, K.J., Lu, W.T., Huang, J., Zhuo, F.,

Qiu, G.P., Wu, X.Y., Qi, W. (2014). “Demyelination initiated by oligodendrocyte apoptosis

through enhancing endoplasmic reticulum-mitochondria interactions and Id2 expression after

compressed spinal cord injury in rats.” CNS Neuroscience Therapeutics. 20(1):20-31. PMID:

23937638

Wu, B., Ren, X. (2009). “Promoting axonal myelination for improving neurological recovery in

spinal cord injury.” The Journal of Neurotrauma. 26(10):1847-56. PMID: 19785544

Zhuo, F., Qiu, G., Xu, J., Yang, M., Wang, K., Liu, H., Huang, J., Lu, W., Liu, Q., Xu, S., Huang

S., Sun, S. (2016). “Both endoplasmic reticulum and mitochondrial pathways are involved in

oligodendrocyte apoptosis induced by capsular hemorrhage.” Molecular and Cellular

Neurosciences. 72:64-71. PMID: 26808219

Fischer, H., Koenig, U., Eckhart, L., Tschachler, E. (2002). "Human caspase 12 has acquired

deleterious mutations." Biochemical and Biophysical Research Communications. 293 (2): 722–6.

PMID: 12054529

Page 123: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

105

Liu, X., Kim, C.N., Yang, J., Jemmerson, R., Wang, X. (1996). "Induction of apoptotic program

in cell-free extracts: requirement for dATP and cytochrome c." Cell. 86 (1): 147–57. PMID:

8689682

Hara, E., Yamaguchi, T., Nojima, H., Ide, T., Campisi, J., Okayama, H., Oda, K. (1994). "Id-

related genes encoding helix-loop-helix proteins are required for G1 progression and are

repressed in senescent human fibroblasts." The Journal of Biological Chemistry. 269 (3): 2139–

45. PMID: 8294468

Ma, L., Yu, H.J., Gan, S.W., Gong, R., Mou, K.J., Xue, J., Sun, S.Q. (2017). “p53-Mediated

oligodendrocyte apoptosis initiates demyelination after compressed spinal cord injury by

enhancing ER-mitochondria interaction and E2F1 expression.” Neuroscience Letters. 644:55-61.

PMID: 28237798

Stevens, C., La Thangue, N.B. (2005). "The emerging role of E2F-1 in the DNA damage

response and checkpoint control." DNA Repair (Amst.). 3 (8-9): 1071–9. PMID: 15279795

1.2.2

Mazzone, G.L., Margaryan, G., Kuzhandaivel, A., Nasrabady, S.E., Mladinic, M., Nistri, A.

(2010). “Kainate-induced delayed onset of excitotoxicity with functional loss unrelated to the

extent of neuronal damage in the in vitro spinal cord.” Neuroscience. 168(2):451-62. PMID:

20362644

Mazzone, G.L., Veeraraghavan, P., Gonzalez-Inchauspe, C., Nistri, A., Uchitel, O.D. (2017).

“ASIC channel inhibition enhances excitotoxic neuronal death in an in vitro model of spinal cord

injury.” Neuroscience. 20;343:398-410. PMID: 28003157

Smith, J.A., Park, S., Krause, J.S., Banik, N.L. (2013). “Oxidative stress, DNA damage, and the

telomeric complex as therapeutic targets in acute neurodegeneration.” Neurochemistry

International. 62(5):764-75. PMID: 23422879

Catala, A., (2009). “Lipid peroxidation of membrane phospholipids generates hydroxy-alkenals

and oxidized phospholipids active in physiological and/or pathological conditions.” Chemistry

and Physics of Lipids. 157(1):1-11. PMID: 18977338

Page 124: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

106

Ansari, M.A., Roberts, K.N., Scheff, S.W. (2008). “Oxidative stress and modification of synaptic

proteins in hippocampus after traumatic brain injury.” Free Radical Biology and Medicine.

45(4):443-52. PMID: 18501200

Muthaiah, V.P.K., Michael, F.M., Palaniappan, T., Rajan, S.S., Chandrasekar, K.,

Venkatachalam, S. (2017). “JNK1 and JNK3 play a significant role in both neuronal apoptosis

and necrosis. Evaluation based on in vitro approach using tert-butylhydroperoxide induced

oxidative stress in neuro-2A cells and perturbation through 3-aminobenzamide.” Toxicology in

Vitro. 41:168-178. PMID: 28257805

Yu, C., Kim, B.S., Kim, E. (2016). “FAF1 mediates regulated necrosis through PARP1

activation upon oxidative stress leading to dopaminergic neurodegeneration.” Cell Death and

Differentiation. 23(11):1873-1885. PMID: 27662363

2

Hilton, B.J., Moulson, A.J., Tetzlaff, W. (2017) “Neuroprotection and secondary damage

following spinal cord injury: concepts and methods.” Neuroscience Letters. 652:3-10. PMID:

27939975

Ahuja, C.S., Martin, A.R., Fehlings, M. (2016). “Recent advances in managing a spinal cord

injury secondary to trauma.” F1000Research. 5. pii: F1000 Faculty Rev-1017. PMID: 27303644

O'Donovan, K.J. (2016). “Intrinsic Axonal Growth and the Drive for Regeneration”. Frontiers in

Neuroscience. 10:486. PMID: 27833527

Franklin, R.J. (2015). “Regenerative Medicines for Remyelination: From Aspiration to Reality.”

Cell Stem Cell. 16(6):576-7. PMID: 26046755

Najm, F.J., Madhavan, M., Zaremba, A., Shick, E., Karl, R.T., Factor, D.C., Miller, T.E., Nevin,

Z.S., Kantor, C., Sargent, A., Quick, K.L., Schlatzer, D.M., Tang, H., Papoian, R., Brimacombe,

K.R., Shen, M., Boxer, M.B., Jadhav, A., Robinson, A.P., Podojil, J.R., Miller, S.D., Miller,

Tesar, P.J. (2015). “Drug-based modulation of endogenous stem cells promotes functional

remyelination in vivo.” Nature. 522(7555):216-20. PMID: 25896324

Page 125: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

107

2.1

Nori S., Nakamura M., Okano, H. (2017). “Plasticity and regeneration in the injured spinal cord

after cell transplantation therapy.” Progress in Brain Research. 231:33-56. PMID: 28554400

Ruff, C.A., Wilcox, J.T., Fehlings, M.G. (2012). “Cell-based transplantation strategies to

promote plasticity following spinal cord injury.” Experimental Neurology. 235(1):78-90. PMID:

21333647

Liu, J., Yang, X., Jiang, L., Wang, C., Yang, M. (2012). “Neural plasticity after spinal cord

injury.” Neural Regeneration Research. 7(5):386-91. PMID: 25774179

Sun, F., He, Z. (2010). “Neuronal intrinsic barriers for axon regeneration in the adult CNS.”

Current Opinion in Neurobiology. 20(4):510-8. PMID: 20418094

McKerracher, L. (2001). “Spinal cord repair: strategies to promote axon regeneration.”

Neurobiology of Disease. 8(1):11-8. PMID: 11162236

Badner, A., Siddiqui, A.M., Fehlings, M.G. (2017). “Spinal cord injuries: how could cell therapy

help?.” Expert Opinion on Biological Therapy. 17(5):529-541. PMID: 28306359

Iyer, N.R., Wilems, T.S., Sakiyama-Elbert, S.E. (2017). “Stem cells for spinal cord injury:

Strategies to inform differentiation and transplantation.” Biotechnology and Bioengineering.

114(2):245-259. PMID: 27531038

Kim, B.G., Hwang, D.H., Lee, S.I., Kim, E.J., Kim, S.U. (2007). “Stem cell-based cell therapy

for spinal cord injury.” Cell Transplantation. 16(4):355-64. PMID: 17658126

Bonner, J.F., Steward, O. (2015). “Repair of spinal cord injury with neuronal relays: From fetal

grafts to neural stem cells.” Brain Research. 1619:115-23. PMID: 25591483

Lu, P., Kadoya, K., Tuszynski, M.H. (2014). “Axonal growth and connectivity from neural stem

cell grafts in models of spinal cord injury.” Current Opinion in Neurobiology. 27:103-9. PMID:

24709371

Li, J., Lepski, G. (2013). “Cell transplantation for spinal cord injury: a systematic review.”

Biomed Research International. 2013:786475. PMID: 23484157

Page 126: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

108

Fehlings, M.G., Vawda R. (2011). “Cellular treatments for spinal cord injury: the time is right

for clinical trials.” Neurotherapeutics. 8(4):704-20. PMID: 22002087

Akesson, E., Piao, J.H., Samuelsson, E.B., Holmberg, L., Kjaeldgaard, A., Falci, S., Sundström,

E., Seiger, A. (2007). “Long-term culture and neuronal survival after intraspinal transplantation

of human spinal cord-derived neurospheres.” Physiology and Behaviour. 92(1-2):60-6. PMID:

17610915

Liu, Y., Zheng, Y., Li, S., Xue, H., Schmitt, K., Hergenroeder, G.W., Wu, J., Zhang, Y., Kim,

D.H., Cao, Q. (2017). “Human neural progenitors derived from integration-free iPSCs for SCI

therapy.” Stem Cell Research. 19:55-64. PMID: 28073086

Yuan T, Liao W, Feng NH, Lou YL, Niu X, Zhang AJ, Wang Y, Deng ZF. (2013). “Human

induced pluripotent stem cell-derived neural stem cells survive, migrate, differentiate, and

improve neurologic function in a rat model of middle cerebral artery occlusion.” Stem Cells

Research and Therapy. 4(3):73. PMID: 23769173

2.1.1

Tan B.T., Jiang, L., Liu, L., Yin, Y., Luo, Z.R., Long, Z.Y., Li, S., Yu, L.H., Wu, Y.M., Liu, Y.

(2017). “Local injection of Lenti-Olig2 at lesion site promotes functional recovery of spinal cord

injury in rats.” CNS Neuroscience & Therapeutics. 23(6):475-487. PMID: 28452182

Lu, P. (2017). “Stem cell transplantation for spinal cord injury repair.” Progress in Brain

Research. 231:1-32. PMID: 28554393

Lopez Juarez, A., He, D., Richard Lu, Q. (2016). “Oligodendrocyte progenitor programming and

reprogramming: Toward myelin regeneration.” Brain Research. 1638(Pt B):209-20. PMID:

26546966

Doulames, V.M., Plant, G.W. (2016). “Induced Pluripotent Stem Cell Therapies for Cervical

Spinal Cord Injury.” International Journal of Molecular Sciences. 17(4):530. PMID: 27070598

Page 127: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

109

Bambakidis, N.C., Miller, R.H. (2004). “Transplantation of oligodendrocyte precursors and sonic

hedgehog results in improved function and white matter sparing in the spinal cords of adult rats

after contusion.” The Spine Journal: The Official Journal of the North American Spine Society.

4(1):16-26. PMID: 14749190

Keirstead, H.S., Nistor, G., Bernal, G., Totoiu, M., Cloutier, F., Sharp, K., Steward, O. (2005).

“Human embryonic stem cell-derived oligodendrocyte progenitor cell transplants remyelinate

and restore locomotion after spinal cord injury.” The Journal of Neuroscience. 25(19):4694-705.

PMID: 15888645

Karimi-Abdolrezaee, S., Eftekharpour, E., Wang, J., Morshead, C.M., Fehlings, M.G. (2006).

“Delayed transplantation of adult neural precursor cells promotes remyelination and functional

neurological recovery after spinal cord injury.” The Journal of Neuroscience. 26(13):3377-89.

PMID: 16571744

Eftekharpour E1, Karimi-Abdolrezaee S, Wang J, El Beheiry H, Morshead C, Fehlings MG.

(2007). “Myelination of congenitally dysmyelinated spinal cord axons by adult neural precursor

cells results in formation of nodes of Ranvier and improved axonal conduction.” The Journal of

Neuroscience. 27(13):3416-28. PMID: 17392458

Karimi-Abdolrezaee, S., Eftekharpour, E., Wang, J., Schut, D., Fehlings, M.G. (2010).

“Synergistic effects of transplanted adult neural stem/progenitor cells, chondroitinase, and

growth factors promote functional repair and plasticity of the chronically injured spinal cord.”

30(5):1657-76. PMID: 20130176

Hwang, D.H., Kim, B.G., Kim, E.J., Lee, S.I., Joo, I.S., Suh-Kim, H., Sohn, S., Kim, S.U.

(2009). “Transplantation of human neural stem cells transduced with Olig2 transcription factor

improves locomotor recovery and enhances myelination in the white matter of rat spinal cord

following contusive injury.” BMC Neuroscience. 10:117. PMID: 19772605

Cao, Q., He, Q., Wang, Y., Cheng, X., Howard, R.M., Zhang, Y., DeVries, W.H., Shields, C.B.,

Magnuson, D.S., Xu, X.M., Kim, D.H., Whittemore, S.R. (2010). “Transplantation of ciliary

neurotrophic factor-expressing adult oligodendrocyte precursor cells promotes remyelination and

Page 128: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

110

functional recovery after spinal cord injury.” BMC Neuroscience. 30(8):2989-3001. PMID:

20181596

Salewski, R.P., Mitchell, R.A., Li, L., Shen, C., Milekovskaia, M., Nagy. A., Fehlings, M.G.

(2015). “Transplantation of Induced Pluripotent Stem Cell-Derived Neural Stem Cells Mediate

Functional Recovery Following Thoracic Spinal Cord Injury Through Remyelination of Axons.”

Stem Cells Translational Medicine. 4(7):743-54. PMID: 25979861

Kawabata, S., Takano, M., Numasawa-Kuroiwa, Y., Itakura, G., Kobayashi, Y., Nishiyama, Y.,

Sugai, K., Nishimura, S., Iwai, H., Isoda, M., Shibata, S., Kohyama, J., Iwanami, A., Toyama,

Y., Matsumoto, M., Nakamura, M., Okano, H. (2016). “Grafted Human iPS Cell-Derived

Oligodendrocyte Precursor Cells Contribute to Robust Remyelination of Demyelinated Axons

after Spinal Cord Injury.” Stem Cell Reports. 6(1):1-8. PMID: 26724902

Khazaei, M., Ahuja, C.S., Fehlings, M.G. (2017). “Induced Pluripotent Stem Cells for Traumatic

Spinal Cord Injury.” Frontiers in Cell and Developmental Biology. 4:152. PMID: 28154814

Lee-Kubli, C.A., Lu, P. (2015). “Induced pluripotent stem cell-derived neural stem cell therapies

for spinal cord injury.” Neural Regeneration Research. 10(1):10-6. PMID: 25788906

Khazaei, M., Siddiqui, A.M., Fehlings, M.G. (2014). “The Potential for iPS-Derived Stem Cells

as a Therapeutic Strategy for Spinal Cord Injury: Opportunities and Challenges.” The Journal of

Clinical Medicine. 4(1):37-65. PMID: 26237017

2.1.2

Hu, J., Selzer, M.E. (2017). “RhoA as a target to promote neuronal survival and axon

regeneration.” Neural Regeneration Research. 12(4):525-528. PMID: 28553321

Kwon, B.K., Borisoff, J.F., Tetzlaff, W. (2002). “Molecular targets for therapeutic intervention

after spinal cord injury.” Molecular Interventions. 2(4):244-58. PMID: 14993395

He, B., Nan, G. (2016). “Neuronal regeneration after acute spinal cord injury in adult rats.” Spine

Journal: The Official Journal of the North American Spine Society. 16(12):1459-1467. PMID:

27349629

Page 129: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

111

Bähr, M., Bonhoeffer, F. (1994). “Perspectives on axonal regeneration in the mammalian CNS.”

Trends in Neurosciences. 17(11):473-9. PMID: 7531889

Rao, S.N., Pearse, D.D. (2016). “Regulating Axonal Responses to Injury: The Intersection

between Signaling Pathways Involved in Axon Myelination and The Inhibition of Axon

Regeneration.” Frontiers in Molecular Neuroscience. 9:33. PMID: 27375427

Yang, E.Z., Zhang, G.W., Xu, J.G., Chen, S., Wang, H., Cao, L.L., Liang, B., Lian, X.F. (2017).

“Multichannel polymer scaffold seeded with activated Schwann cells and bone mesenchymal

stem cells improves axonal regeneration and functional recovery after rat spinal cord injury.”

Acta Pharmacologica Sinica. 38(5):623-637. PMID: 28392569

Liu, J., Chen, J., Liu, B., Yang, C., Xie, D., Zheng, X., Xu, S., Chen, T., Wang, L., Zhang, Z.,

Bai, X., Jin, D. (2013). “Acellular spinal cord scaffold seeded with mesenchymal stem cells

promotes long-distance axon regeneration and functional recovery in spinal cord injured rats.”

Journal of the Neurological Sciences. 325(1-2):127-36. PMID: 23317924

Ban, D.X., Ning, G.Z., Feng, S.Q., Wang, Y., Zhou, X.H., Liu, Y., Chen, J.T. (2011).

“Combination of activated Schwann cells with bone mesenchymal stem cells: the best cell

strategy for repair after spinal cord injury in rats.” Regenerative Medicine. 6(6):707-20. PMID:

22050523

Oliveri, R.S., Bello, S., Biering-Sørensen, F. (2014). “Mesenchymal stem cells improve

locomotor recovery in traumatic spinal cord injury: systematic review with meta-analyses of rat

models.” Neurobiology of Disease. 62:338-53. PMID: 24148857

Bareyre, F.M. (2008). “Neuronal repair and replacement in spinal cord injury.” Journal of the

Neurological Sciences. 265(1-2):63-72. PMID: 17568612

Adler, A.F., Lee-Kubli, C., Kumamaru, H., Kadoya, K., Tuszynski, M.H. (2017).

“Comprehensive Monosynaptic Rabies Virus Mapping of Host Connectivity with Neural

Progenitor Grafts after Spinal Cord Injury.” Stem Cell Reports. 8(6):1525-1533. PMID:

28479302

Page 130: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

112

Kadoya, K., Lu, P., Nguyen, K., Lee-Kubli, C., Kumamaru, H., Yao, L., Knackert, J., Poplawski,

G., Dulin, J.N., Strobl, H., Takashima, Y., Biane, J., Conner, J., Zhang, S.C., Tuszynski, M.H.

(2016). “Spinal cord reconstitution with homologous neural grafts enables robust corticospinal

regeneration.” Nature Medicine. 22(5):479-87. PMID: 27019328

Fandel, T.M., Trivedi, A., Nicholas, C.R., Zhang, H., Chen, J., Martinez, A.F., Noble-

Haeusslein, L.J., Kriegstein, A.R. (2016). “Transplanted Human Stem Cell-Derived Interneuron

Precursors Mitigate Mouse Bladder Dysfunction and Central Neuropathic Pain after Spinal Cord

Injury.” Cell Stem Cell. 19(4):544-557. PMID: 27666009

Etlin, A., Bráz, J.M., Kuhn, J.A., Wang, X., Hamel, K.A., Llewellyn-Smith, I.J., Basbaum, A.I.

(2016). “Functional Synaptic Integration of Forebrain GABAergic Precursors into the Adult

Spinal Cord.” The Journal of Neuroscience. 36(46):11634-11645. PMID: 27852772

Hwang, I., Hahm, S.C., Choi, K.A., Park, S.H., Jeong, H., Yea, J.H., Kim, J., Hong, S. (2016).

“Intrathecal Transplantation of Embryonic Stem Cell-Derived Spinal GABAergic Neural

Precursor Cells Attenuates Neuropathic Pain in a Spinal Cord Injury Rat Model.” Cell

Transplantation. 25(3):593-607. PMID: 26407027

Bráz, J.M., Sharif-Naeini, R., Vogt, D., Kriegstein, A., Alvarez-Buylla, A., Rubenstein, J.L.,

Basbaum, A.I. (2012). “Forebrain GABAergic neuron precursors integrate into adult spinal cord

and reduce injury-induced neuropathic pain.” Neuron. 74(4):663-75. PMID: 22632725

Yamane, J., Nakamura, M., Iwanami, A., Sakaguchi, M., Katoh, H., Yamada, M., Momoshima,

S., Miyao, S., Ishii, K., Tamaoki, N., Nomura, T., Okano, H.J., Kanemura, Y., Toyama, Y.,

Okano, H. (2010). “Transplantation of galectin-1-expressing human neural stem cells into the

injured spinal cord of adult common marmosets.” Journal of Neuroscience Research.

88(7):1394-405. PMID: 20091712

Quintá, H.R., Wilson, C., Blidner, A.G., González-Billault, C., Pasquini, L.A., Rabinovich, G.A.,

Pasquini, J.M. (2016). “Ligand-mediated Galectin-1 endocytosis prevents intraneural H2O2

production promoting F-actin dynamics reactivation and axonal re-growth.” Experimental

Neurology. 283(Pt A):165-78. PMID: 27296316

Page 131: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

113

Quintá, H.R., Pasquini, J.M., Rabinovich, G.A., Pasquini, L.A. (2014). “Glycan-dependent

binding of galectin-1 to neuropilin-1 promotes axonal regeneration after spinal cord injury.” Cell

Death and Differentiation. 21(6):941-55. PMID: 24561343

Mehta ST1, Bixby JL1,2,3,4, Lemmon VP1,2,3. (2017). “Genetically modifying transcription

factors to promote CNS axon regeneration.” Neural Regeneration Research. 12(5):737-738.

PMID: 28616023

Luo, X., Ribeiro, M., Bray, E.R., Lee, D.H., Yungher, B.J., Mehta, S.T., Thakor, K.A., Diaz,

F.2., Lee, J.K., Moraes, C.T., Bixby, J.L., Lemmon, V.P., Park, K.K. (2016). “Enhanced

Transcriptional Activity and Mitochondrial Localization of STAT3 Co-induce Axon Regrowth in

the Adult Central Nervous System.” Cell Reports. 15(2):398-410. PMID: 27050520

Leibinger, M., Andreadaki, A., Diekmann, H., Fischer, D. (2013a). “Neuronal STAT3 activation

is essential for CNTF- and inflammatory stimulation-induced CNS axon regeneration.” Cell

Death and Disease. 4:e805. PMID: 24052073

Leibinger, M., Müller, A., Gobrecht, P., Diekmann, H., Andreadaki, A., Fischer, D. (2013b).

“Interleukin-6 contributes to CNS axon regeneration upon inflammatory stimulation.” Cell

Death and Disease. 4:e609. PMID: 23618907

Liu, Y., Wang, X., Lu, C.C., Kerman, R., Steward, O., Xu, X.M., Zou, Y. (2008). “Repulsive

Wnt signaling inhibits axon regeneration after CNS injury.” The Journal of Neuroscience.

28(33):8376-82. PMID: 18701700

Du, K., Zheng, S., Zhang, Q., Li, S., Gao, X., Wang, J., Jiang, L., Liu, K. (2015). “Pten Deletion

Promotes Regrowth of Corticospinal Tract Axons 1 Year after Spinal Cord Injury.” The Journal

of Neuroscience. 35(26):9754-63. PMID: 26134657

Liu, K., Lu, Y., Lee, J.K., Samara, R., Willenberg, R., Sears-Kraxberger, I., Tedeschi, A., Park,

K.K., Jin, D., Cai, B., Xu, B., Connolly, L., Steward, O., Zheng, B., He, Z. (2010). “PTEN

deletion enhances the regenerative ability of adult corticospinal neurons.” Nature Neuroscience.

13(9):1075-81. PMID: 20694004

Page 132: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

114

Zweckberger, K., Ahuja, C.S., Liu, Y., Wang, J., Fehlings, M.G. (2016). “Self-assembling

peptides optimize the post-traumatic milieu and synergistically enhance the effects of neural

stem cell therapy after cervical spinal cord injury.” Acta Biomateriala. 42:77-89. PMID:

27296842

Zweckberger, K., Liu, Y., Wang, J., Forgione, N., Fehlings, M.G. (2015). “Synergetic use of

neural precursor cells and self-assembling peptides in experimental cervical spinal cord injury.”

The Journal of Visualized Experiments: JoVE. (96):e52105. PMID: 25742521

Bonner, J.F., Connors, T.M., Silverman, W.F., Kowalski, D.P., Lemay, M.A., Fischer, I. (2011).

“Grafted neural progenitors integrate and restore synaptic connectivity across the injured spinal

cord.” The Journal of Neuroscience. 31(12):4675-86. PMID: 21430166

Bonner, J.F., Blesch, A., Neuhuber, B., Fischer, I. (2010). “Promoting directional axon growth

from neural progenitors grafted into the injured spinal cord.” The Journal of Neuroscience

Research. 88(6):1182-92. PMID: 19908250

3.1

Curado S1, Stainier DY, Anderson RM. (2008). “Nitroreductase-mediated cell/tissue ablation in

zebrafish: a spatially and temporally controlled ablation method with applications in

developmental and regeneration studies.” Nature Protocols. 3(6):948-54. PMID: 18536643

Siddiqui AM1, Khazaei M1, Fehlings MG2. (2015). “Translating mechanisms of

neuroprotection, regeneration, and repair to treatment of spinal cord injury.” Progress in Brain

Research. 218:15-54. PMID: 25890131

3.1.1

Wagner PL1, Waldor MK. (2002). “Bacteriophage control of bacterial virulence.” Infection and

Immunity. 70(8):3985-93. PMID: 12117903

Pappenheimer AM Jr. (1977). “Diptheria toxin.” Annual Review of Biochemistry. 46:69-94.

PMID: 20040

Page 133: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

115

Freeman VJ. (1951). " Studies on the virulence of bacteriophage-infected strains of

Corynebacterium diphtheriae.” Journal of Bacteriology. PMID: 14850426

Bell CE1, Eisenberg D. (1997). “Crystal structure of nucleotide-free diphtheria toxin.”

Biochemistry. 36(3):481-8. PMID: 9012663

Saito M1, Iwawaki T, Taya C, Yonekawa H, Noda M, Inui Y, Mekada E, Kimata Y, Tsuru A,

Kohno K. (2001). “Diphtheria toxin receptor-mediated conditional and targeted cell ablation in

transgenic mice.” Nature Biotechnology. 19(8):746-50. PMID: 11479567

Cha JH1, Chang MY, Richardson JA, Eidels L. (2003). “Transgenic mice expressing the

diphtheria toxin receptor are sensitive to the toxin.” Molecular Microbiology. 49(1):235-40.

PMID: 12823824

Buch T1, Heppner FL, Tertilt C, Heinen TJ, Kremer M, Wunderlich FT, Jung S, Waisman A.

(2005). “A Cre-inducible diphtheria toxin receptor mediates cell lineage ablation after toxin

administration.” Nature Methods. 2(6):419-26. PMID: 15908920

Wrobel CJ1, Wright DC, Dedrick RL, Youle RJ. (1990). “Diphtheria toxin effects on brain-

tumor xenografts. Implications for protein-based brain-tumor chemotherapy.” The Journal of

Neurosurgery. 72(6):946-50. PMID: 2159988

Mathis C1, Collin L, Borrelli E. (2003). “Oligodendrocyte ablation impairs cerebellum

development.” Development. 130(19):4709-18. PMID: 12925596

Bennett CL1, Clausen BE. (2007). “DC ablation in mice: promises, pitfalls, and challenges.”

Trends in Immunology. 28(12):525-31. PMID: 17964853

Cerpa V1, Gonzalez A2, Richerson GB3. Diphtheria toxin treatment of Pet-1-Cre floxed

diphtheria toxin receptor mice disrupts thermoregulation without affecting respiratory

chemoreception. Neuroscience. 279:65-76. PMID: 25171790

Fen Z1, Dhadly MS, Yoshizumi M, Hilkert RJ, Quertermous T, Eddy RL, Shows TB, Lee ME.

(1993). Structural organization and chromosomal assignment of the gene encoding the human

heparin-binding epidermal growth factor-like growth factor/diphtheria toxin receptor.

Biochemistry. 32(31):7932-8. PMID: 8347598

Page 134: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

116

Vaughan TJ1, Pascall JC, Brown KD. (1992). “Tissue distribution of mRNA for heparin-binding

epidermal growth factor.” Biochemistry. 287 (Pt 3):681-4. PMID: 1445231

Almond BD1, Eidels L. (1994). “The cytoplasmic domain of the diphtheria toxin receptor (HB-

EGF precursor) is not required for receptor-mediated endocytosis.” The Journal of Biological

Chemistry. 269(43):26635-41. PMID: 7929396

Yamaizumi M, Mekada E, Uchida T, Okada Y. (1978). “One molecule of diphtheria toxin

fragment A introduced into a cell can kill the cell.” Cell. 15(1):245-50. PMID: 699044

Pappenheimer AM Jr, Harper AA, Moynihan M, Brockes JP. (1982). “Diphtheria toxin and

related proteins: effect of route of injection on toxicity and the determination of cytotoxicity for

various cultured cells.” The Journal of Infectious Diseases. 145(1):94-102. PMID: 6798133

3.1.2

Zhang J1, Kale V, Chen M. (2015). “Gene-directed enzyme prodrug therapy.” The AAPS

Journal. 17(1):102-10. PMID: 25338741

Chou TY1, Hong BY1. (2014). “Ganciclovir ophthalmic gel 0.15% for the treatment of acute

herpetic keratitis: background, effectiveness, tolerability, safety, and future applications.”

Therapeutics and Clinical Risk Management. 10:665-81. PMID: 25187721

Ikeshima-Kataoka H1, Yuasa S. (2008). Selective ablation of an astroglial subset by toxic gene

expression driven by tenascin promoter. Neurological Research. 30(7):701-9. PMID: 18489816

Balzarini J1, Bohman C, De Clercq E. (1993). “Differential mechanism of cytostatic effect of

(E)-5-(2-bromovinyl)-2'-deoxyuridine, 9-(1,3-dihydroxy-2-propoxymethyl)guanine, and other

antiherpetic drugs on tumor cells transfected by the thymidine kinase gene of herpes simplex

virus type 1 or type 2.” The Journal of Biological Chemistry. 268(9):6332-7. PMID: 8384209

Oliver S, Bubley G, Crumpacker C. (1985). “Inhibition of HSV-transformed murine cells by

nucleoside analogs, 2'-NDG and 2'-nor-cGMP: mechanisms of inhibition and reversal by

exogenous nucleosides.” Virology. 145(1):84-93. PMID: 2990104

Page 135: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

117

Nishiyama Y, Rapp F. (1979). “Anticellular effects of 9-(2-hydroxyethoxymethyl) guanine

against herpes simplex virus-transformed cells.” Molecular Pharmacology. 28(6):581-7. The

Journal of General Virology. 45(1):227-30. PMID: 230303

Bagó JR1, Sheets KT1, Hingtgen SD2. (2016). “Neural stem cell therapy for cancer.” Methods

(San Diego, Calif.). 99:37-43. PMID: 26314280

Beltinger C1, Fulda S, Kammertoens T, Meyer E, Uckert W, Debatin KM. (1999). “Herpes

simplex virus thymidine kinase/ganciclovir-induced apoptosis involves ligand-independent death

receptor aggregation and activation of caspases.” Proceedings of the National Academy of

Sciences of the United States of America. 96(15):8699-704. PMID: 10411938

Fischer U1, Steffens S, Frank S, Rainov NG, Schulze-Osthoff K, Kramm CM. (2005).

“Mechanisms of thymidine kinase/ganciclovir and cytosine deaminase/ 5-fluorocytosine suicide

gene therapy-induced cell death in glioma cells.” Oncogene. 24(7):1231-43. PMID: 15592511

van Dillen IJ1, Mulder NH, Vaalburg W, de Vries EF, Hospers GA. (2002). “Influence of the

bystander effect on HSV-tk/GCV gene therapy. A review.” Current Gene Therapy. 2(3):307-22.

PMID: 12189718

Mesnil M1, Yamasaki H. (2000). “Bystander effect in herpes simplex virus-thymidine

kinase/ganciclovir cancer gene therapy: role of gap-junctional intercellular communication.”

Cancer Research. 60(15):3989-99. PMID: 10945596

Asklund T1, Appelskog IB, Ammerpohl O, Langmoen IA, Dilber MS, Aints A, Ekström TJ,

Almqvist PM. (2003). “Gap junction-mediated bystander effect in primary cultures of human

malignant gliomas with recombinant expression of the HSVtk gene.” Experimental Cell

Research. 284(2):185-95. PMID: 12651152

Burrows FJ1, Gore M, Smiley WR, Kanemitsu MY, Jolly DJ, Read SB, Nicholas T, Kruse CA.

(2002). “Purified herpes simplex virus thymidine kinase retroviral particles: III. Characterization

of bystander killing mechanisms in transfected tumor cells.” Current Gene Therapy. 9(1):87-95.

PMID: 11916247

Page 136: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

118

Dilber MS1, Abedi MR, Christensson B, Björkstrand B, Kidder GM, Naus CC, Gahrton G,

Smith CI. (1997). “Gap junctions promote the bystander effect of herpes simplex virus thymidine

kinase in vivo.” Cancer Research. 57(8):1523-8. PMID: 9108455

Princen F1, Robe P, Lechanteur C, Mesnil M, Rigo JM, Gielen J, Merville MP, Bours V. (1999).

“A cell type-specific and gap junction-independent mechanism for the herpes simplex virus-1

thymidine kinase gene/ganciclovir-mediated bystander effect.” Clinical Cancer Research: an

official journal of the American Association for Cancer Research. 5(11):3639-44. PMID:

10589781

Freeman SM1, Abboud CN, Whartenby KA, Packman CH, Koeplin DS, Moolten FL, Abraham

GN. (1993). “The "bystander effect": tumor regression when a fraction of the tumor mass is

genetically modified.” Cancer Research. 53(21):5274-83. PMID: 8221662

Robe PA1, Nguyen-Khac M, Jolois O, Rogister B, Merville MP, Bours V. (2005).

“Dexamethasone inhibits the HSV-tk/ ganciclovir bystander effect in malignant glioma cells.”

BMC Cancer. 5:32. PMID: 15804364

Moolten FL1, Wells JM, Heyman RA, Evans RM. (1990). “Lymphoma regression induced by

ganciclovir in mice bearing a herpes thymidine kinase transgene.” Human Gene Therapy.

1(2):125-34. PMID: 1964092

Moolten FL. (1986). “Tumor chemosensitivity conferred by inserted herpes thymidine kinase

genes: paradigm for a prospective cancer control strategy.” Cancer Research. 46(10):5276-81.

PMID: 3019523

Bondanza A1, Hambach L, Aghai Z, Nijmeijer B, Kaneko S, Mastaglio S, Radrizzani M,

Fleischhauer K, Ciceri F, Bordignon C, Bonini C, Goulmy E. (2011). “IL-7 receptor expression

identifies suicide gene-modified allospecific CD8+ T cells capable of self-renewal and

differentiation into antileukemia effectors.” Blood. 117(24):6469-78. PMID: 21531977

Ribot EJ1, Miraux S, Konsman JP, Bouchaud V, Pourtau L, Delville MH, Franconi JM,

Thiaudière E, Voisin PJ. (2011). “In vivo MR tracking of therapeutic microglia to a human

glioma model.” NMR in Biomedicine. 24(10):1361-8. PMID: 21387452.

Page 137: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

119

Staquicini FI1, Ozawa MG, Moya CA, Driessen WH, Barbu EM, Nishimori H, Soghomonyan S,

Flores LG 2nd, Liang X, Paolillo V, Alauddin MM, Basilion JP, Furnari FB, Bogler O, Lang FF,

Aldape KD, Fuller GN, Höök M, Gelovani JG, Sidman RL, Cavenee WK, Pasqualini R, Arap

W. (2011). “Systemic combinatorial peptide selection yields a non-canonical iron-mimicry

mechanism for targeting tumors in a mouse model of human glioblastoma.” The Journal of

Clinical Investigation. 121(1):161-73. PMID: 21183793

Kakinoki K1, Nakamoto Y, Kagaya T, Tsuchiyama T, Sakai Y, Nakahama T, Mukaida N,

Kaneko S. (2010). “Prevention of intrahepatic metastasis of liver cancer by suicide gene therapy

and chemokine ligand 2/monocyte chemoattractant protein-1 delivery in mice.” The Journal of

Gene Medicine. 12(12):1002-13. PMID: 21157824

Tang W1, He Y, Zhou S, Ma Y, Liu G. (2009). “A novel Bifidobacterium infantis-mediated

TK/GCV suicide gene therapy system exhibits antitumor activity in a rat model of bladder

cancer.” The Journal of Experimental & Clinical Cancer Research : CR. 28:155. PMID:

20015348

Greish K1, Frandsen J, Scharff S, Gustafson J, Cappello J, Li D, O'Malley BW Jr, Ghandehari H.

(2010). “Silk-elastinlike protein polymers improve the efficacy of adenovirus thymidine kinase

enzyme prodrug therapy of head and neck tumors.” The Journal of Gene Medicine. 12(7):572-9.

PMID: 20603862

Chen LS1, Wang M, Ou WC, Fung CY, Chen PL, Chang CF, Huang WS, Wang JY, Lin PY,

Chang D. (2010). “Efficient gene transfer using the human JC virus-like particle that inhibits

human colon adenocarcinoma growth in a nude mouse model.” Gene Therapy. 17(8):1033-41.

PMID: 20410928

Ambade AV1, Joshi GV, Mulherkar R. (2010). “Effect of suicide gene therapy in combination

with immunotherapy on antitumour immune response & tumour regression in a xenograft mouse

model for head & neck squamous cell carcinoma.” The Indian Journal of Medical Research.

132:415-22. PMID: 20966520

Page 138: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

120

Li S1, Tokuyama T, Yamamoto J, Koide M, Yokota N, Namba H. (2005). “Potent bystander

effect in suicide gene therapy using neural stem cells transduced with herpes simplex virus

thymidine kinase gene.” Oncology. 69(6):503-8. PMID: 16424680

Pu K1, Li SY, Gao Y, Ma L, Ma W, Liu Y. (2011). “Bystander effect in suicide gene therapy

using immortalized neural stem cells transduced with herpes simplex virus thymidine kinase

gene on medulloblastoma regression.” Brain Research. 1369:245-52. PMID: 21073865

Uhl M1, Weiler M, Wick W, Jacobs AH, Weller M, Herrlinger U. (2005). “Migratory neural

stem cells for improved thymidine kinase-based gene therapy of malignant gliomas.”

Biochemical and Biophysical Research Communications. 328(1):125-9. PMID: 15670759

Tieng V1, Cherpin O2, Gutzwiller E2, Zambon AC3, Delgado C2, Salmon P4, Dubois-Dauphin

M2, Krause KH1. (2016). “Elimination of proliferating cells from CNS grafts using a Ki67

promoter-driven thymidine kinase.” Molecular Therapy. Methods & Clinical Development.

6:16069. PMID: 27990449

Sofroniew MV1, Bush TG, Blumauer N, Lawrence Kruger, Mucke L, Johnson MH. (1999).

“Genetically-targeted and conditionally-regulated ablation of astroglial cells in the central,

enteric and peripheral nervous systems in adult transgenic mice.” Brain Research. 835(1):91-5.

PMID: 10448200

Bush TG1, Puvanachandra N, Horner CH, Polito A, Ostenfeld T, Svendsen CN, Mucke L,

Johnson MH, Sofroniew MV. (1999). “Leukocyte infiltration, neuronal degeneration, and neurite

outgrowth after ablation of scar-forming, reactive astrocytes in adult transgenic mice.” Neuron.

23(2):297-308. PMID: 10399936

Bush TG1, Savidge TC, Freeman TC, Cox HJ, Campbell EA, Mucke L, Johnson MH, Sofroniew

MV. (1998). “Fulminant jejuno-ileitis following ablation of enteric glia in adult transgenic

mice.” Cell. 93(2):189-201. PMID: 9568712

Sachewsky N1, Leeder R2, Xu W1, Rose KL3, Yu F1, van der Kooy D2, Morshead CM4.

(2014). “Primitive neural stem cells in the adult mammalian brain give rise to GFAP-expressing

neural stem cells.” Stem Cell Reports. PMID: 24936468

Page 139: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

121

Imura T1, Kornblum HI, Sofroniew MV. (2003). “The predominant neural stem cell isolated

from postnatal and adult forebrain but not early embryonic forebrain expresses GFAP.” The

Journal of Neuroscience. 23(7):2824-32. PMID: 12684469

Morshead CM1, Garcia AD, Sofroniew MV, van Der Kooy D. (2003). “The ablation of glial

fibrillary acidic protein-positive cells from the adult central nervous system results in the loss of

forebrain neural stem cells but not retinal stem cells.” The European Journal of Neuroscience.

18(1):76-84. PMID: 12859339

Hesp ZC1,2, Yoseph RY1,2, Suzuki R3, Jukkola P3, Wilson C2, Nishiyama A3, McTigue

DM4,5. (2018). “Proliferating NG2-Cell-Dependent Angiogenesis and Scar Formation Alter

Axon Growth and Functional Recovery After Spinal Cord Injury in Mice.” The Journal of

Neuroscience. 38(6):1366-1382. PMID: 29279310

Faulkner JR1, Herrmann JE, Woo MJ, Tansey KE, Doan NB, Sofroniew MV. (2004). “Reactive

astrocytes protect tissue and preserve function after spinal cord injury.” The Journal of

Neuroscience. 24(9):2143-55. PMID: 14999065

Laberge RM1, Adler D, DeMaria M, Mechtouf N, Teachenor R, Cardin GB, Desprez PY,

Campisi J, Rodier F. (2013). “Mitochondrial DNA damage induces apoptosis in senescent cells.”

Cell Death and Disease. 4:e727. PMID: 23868060

Beltinger C1, Fulda S, Kammertoens T, Uckert W, Debatin KM. (2000). “Mitochondrial

amplification of death signals determines thymidine kinase/ganciclovir-triggered activation of

apoptosis.” Cancer Research. 60(12):3212-7. PMID: 10866313

3.1.3

Dachs GU1, Hunt MA, Syddall S, Singleton DC, Patterson AV. (2009). “Bystander or no

bystander for gene directed enzyme prodrug therapy.” Molecules. 14(11):4517-45. PMID:

19924084

Degrève B1, De Clercq E, Balzarini J. (1999). “Bystander effect of purine nucleoside analogues

in HSV-1 tk suicide gene therapy is superior to that of pyrimidine nucleoside analogues.” Gene

Therapy. 6(2):162-70. PMID: 10435100

Page 140: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

122

De Clercq E1. (2005). “(E)-5-(2-bromovinyl)-2'-deoxyuridine (BVDU).” Medicinal Research

Reviews. 25(1):1-20. PMID: 15389733

De Clercq E1 (2004). “Discovery and development of BVDU (brivudin) as a therapeutic for the

treatment of herpes zoster.” Biochemical Pharmacology. 68(12):2301-15. PMID: 15548377

Balzarini J, De Clercq E, Ayusawa D, Shimizu K, Seno T. (1985a). “Selective inhibition of the

proliferation of herpes simplex virus type 1 thymidine kinase gene-transformed murine

mammary FM3A carcinoma cells by (E)-5-(2-bromovinyl)-2'-deoxyuridine and related

compounds.” Nucleic Acids Symposium Series. (16):283-6. PMID: 3003708

Balzarini J, de Clercq E, Ayusawa D, Seno T. (1985b). “Murine mammary FM3A carcinoma

cells transformed with the herpes simplex virus type 1 thymidine kinase gene are highly sensitive

to the growth-inhibitory properties of (E)-5-(2-bromovinyl)-2'-deoxyuridine and related

compounds.” FEBS Letters. 85(1):95-100. PMID: 2987041

Balzarini J, De Clercq E, Verbruggen A, Ayusawa D, Seno T. (1985c). “Highly selective

cytostatic activity of (E)-5-(2-bromovinyl)-2'-deoxyuridine derivatives for murine mammary

carcinoma (FM3A) cells transformed with the herpes simplex virus type 1 thymidine kinase

gene.” Molecular Pharmacology. 28(6):581-7. PMID: 3001499

Balzarini J, De Clercq E, Verbruggen A, Crumpacker C, Ayusawa D, Seno T. (1986). “Increased

sensitivity of thymidine kinase-deficient (TK-) tumor cell lines to the cell growth inhibitory

effects of (E)-5-(2-bromovinyl)-2'-deoxyuridine (BVDU) and related compounds.” Anticancer

Research. 6(5):1077-84. PMID: 2432829

Balzarini J1, De Clercq E. (1989). “Inhibitory effects of (E)-5-(2-bromovinyl)-2'-deoxyuridine

(BVDU) and related compounds on herpes simplex virus (HSV)-infected cells and HSV

thymidine kinase gene-transformed cells.” Methods and Findings in Experimental and Clinical

Pharmacology. 11(6):379-89. PMID: 2545982

Balzarini J1, De Clercq E, Verbruggen A, Ayusawa D, Shimizu K, Seno T. (1987).

“Thymidylate synthase is the principal target enzyme for the cytostatic activity of (E)-5-(2-

bromovinyl)-2'-deoxyuridine against murine mammary carcinoma (FM3A) cells transformed

Page 141: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

123

with the herpes simplex virus type 1 or type 2 thymidine kinase gene.” Molecular

Pharmacology. 32(3):410-6. PMID: 2823092

Tomicic MT1, Friedrichs C, Christmann M, Wutzler P, Thust R, Kaina B. (2003). “Apoptosis

induced by (E)-5-(2-bromovinyl)-2'-deoxyuridine in varicella zoster virus thymidine kinase-

expressing cells is driven by activation of c-Jun/activator protein-1 and Fas ligand/caspase-8.”

Molecular Pharmacology. 63(2):439-49. PMID: 12527816

Reefschläger J, Wutzler P, Thiel KD, Bärwolff D, Langen P, Sprössig M, Rosenthal HA. (1982).

“Efficacy of (E)-5-(2-bromovinyl)-2'-deoxyuridine against different herpes simplex virus strains

in cell culture and against experimental herpes encephalitis in mice.” Antiviral Research.

2(5):255-65. PMID: 6295273

Grignet-Debrus C1, Cool V, Baudson N, Degrève B, Balzarini J, De Leval L, Debrus S, Velu T,

Calberg-Bacq CM. (2000). “Comparative in vitro and in vivo cytotoxic activity of (E)-5-(2-

bromovinyl)-2'-deoxyuridine (BVDU) and its arabinosyl derivative, (E)-5-(2-bromovinyl)-1-

beta-D-arabinofuranosyluracil (BVaraU), against tumor cells expressing either the Varicella

zoster or the Herpes simplex virus thymidine kinase.” Cancer Gene Therapy. 7(2):215-23.

PMID: 10770629

Field HJ, Anderson JR, Efstathiou S. (1984). “A quantitative study of the effects of several

nucleoside analogues on established herpes encephalitis in mice.” The Journal of General

Virology. 65 (Pt 4):707-19. PMID: 6323620

3.1.4

Searle PF1, Chen MJ, Hu L, Race PR, Lovering AL, Grove JI, Guise C, Jaberipour M, James

ND, Mautner V, Young LS, Kerr DJ, Mountain A, White SA, Hyde EI. (2004). “Nitroreductase:

a prodrug-activating enzyme for cancer gene therapy.” Clinical and Experimental Physiology

and Pharmacology. 31(11):811-6. PMID: 15566399

Page 142: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

124

Grove JI1, Searle PF, Weedon SJ, Green NK, McNeish IA, Kerr DJ. (1999). “Virus-directed

enzyme prodrug therapy using CB1954.” Anticancer Drug Design. 14(6):461-72. PMID:

10834268

Denny WA1. (2002). “Nitroreductase-based GDEPT.” Current Pharmaceutical Design.

8(15):1349-61. PMID: 12052212

Williams EM1, Little RF1, Mowday AM2, Rich MH1, Chan-Hyams JV1, Copp JN3, Smaill

JB4, Patterson AV4, Ackerley DF5. (2015). “Nitroreductase gene-directed enzyme prodrug

therapy: insights and advances toward clinical utility.” The Biochemical Journal. 471(2):131-53.

PMID: 26431849

Vass SO1, Jarrom D, Wilson WR, Hyde EI, Searle PF. (2009). “E. coli NfsA: an alternative

nitroreductase for prodrug activation gene therapy in combination with CB1954.” The British

Journal of Cancer. 100(12):1903-11. PMID: 19455141

Drabek D1, Guy J, Craig R, Grosveld F. (1997). “The expression of bacterial nitroreductase in

transgenic mice results in specific cell killing by the prodrug CB1954.” Gene Therapy. 4(2):93-

100. PMID: 9081711

Clark AJ1, Iwobi M, Cui W, Crompton M, Harold G, Hobbs S, Kamalati T, Knox R, Neil C,

Yull F, Gusterson B. (1997). “Selective cell ablation in transgenic mice expression E. coli

nitroreductase.” Gene Therapy. 4(2):101-10. PMID: 9081700

Knox RJ1, Burke PJ, Chen S, Kerr DJ. (2003). “CB 1954: from the Walker tumor to NQO2 and

VDEPT.” Current Pharmaceutical Design. 9(26):2091-104. PMID: 14529407

Knox RJ1, Friedlos F, Boland MP. (1993). “The bioactivation of CB 1954 and its use as a

prodrug in antibody-directed enzyme prodrug therapy (ADEPT).” Cancer Metastasis Reviews.

12(2):195-212. PMID: 8375021

Workman P, Morgan JE, Talbot K, Wright KA, Donaldson J, Twentyman PR. (1986). “CB 1954

revisited. II. Toxicity and antitumour activity.” Cancer Chemotherapy and Pharmacology.

16(1):9-14. PMID: 3940225

Page 143: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

125

Connors TA, Melzack DH. (1971). “Studies on the mechanism of action of 5-aziridinyl-2,4-

dinitrobenzamide (CB 1954), a selective inhibitor of the Walker tumour.” International Journal

of Cancer. PMID: 5546147

Cobb LM, Connors TA, Elson LA, Khan AH, Mitchley BC, Ross WC, Whisson ME. (1969).

“2,4-dinitro-5-ethyleneiminobenzamide (CB 1954): a potent and selective inhibitor of the growth

of the Walker carcinoma 256.” Biochemical Pharmacology. 18(6):1519-27. PMID: 4307990

Khan AH, Ross WC. (1969). “Tumour-growth inhibitory nitrophenylaziridines and related

compounds: structure-activity relationships.” Chemico-biological Interactions. 1(1):27-47.

PMID: 5407053

Knox RJ1, Friedlos F, Jarman M, Roberts JJ. (1988). “A new cytotoxic, DNA interstrand

crosslinking agent, 5-(aziridin-1-yl)-4-hydroxylamino-2-nitrobenzamide, is formed from 5-

(aziridin-1-yl)-2,4-dinitrobenzamide (CB 1954) by a nitroreductase enzyme in Walker carcinoma

cells.” Biochemical Pharmacology. 37(24):4661-9. PMID: 3202902

Roberts JJ, Friedlos F, Knox RJ. (1986). CB 1954 (2,4-dinitro-5-aziridinyl benzamide) becomes

a DNA interstrand crosslinking agent in Walker tumour cells. Biochemical and Biophysical

Research Communications. 140(3):1073-8. PMID: 3778483

Cui W1, Gusterson B, Clark AJ. (1999). “Nitroreductase-mediated cell ablation is very rapid and

mediated by a p53-independent apoptotic pathway.” Gene Therapy. 6(5):764-70. PMID:

10505099

Felmer RN1, Clark JA. (2004). “The gene suicide system Ntr/CB1954 causes ablation of

differentiated 3T3L1 adipocytes by apoptosis.” Biological Research. 37(3):449-60. PMID:

15515969

Bridgewater JA1, Springer CJ, Knox RJ, Minton NP, Michael NP, Collins MK. (1995).

“Expression of the bacterial nitroreductase enzyme in mammalian cells renders them selectively

sensitive to killing by the prodrug CB1954.”

Green NK1, Youngs DJ, Neoptolemos JP, Friedlos F, Knox RJ, Springer CJ, Anlezark GM,

Michael NP, Melton RG, Ford MJ, Young LS, Kerr DJ, Searle PF. (1997). “Sensitization of

Page 144: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

126

colorectal and pancreatic cancer cell lines to the prodrug 5-(aziridin-1-yl)-2,4-dinitrobenzamide

(CB1954) by retroviral transduction and expression of the E. coli nitroreductase gene.” Cancer

Gene Therapy. 4(4):229-38. PMID: 9253508

Weedon SJ1, Green NK, McNeish IA, Gilligan MG, Mautner V, Wrighton CJ, Mountain A,

Young LS, Kerr DJ, Searle PF. (2000). “Sensitisation of human carcinoma cells to the prodrug

CB1954 by adenovirus vector-mediated expression of E. coli nitroreductase.” International

Journal of Cancer. 86(6):848-54. PMID: 10842200

Grove JI1, Lovering AL, Guise C, Race PR, Wrighton CJ, White SA, Hyde EI, Searle PF.

(2003). “Generation of Escherichia coli nitroreductase mutants conferring improved cell

sensitization to the prodrug CB1954.” Cancer Research. 63(17):5532-7. PMID: 14500391

Jaberipour M1, Vass SO, Guise CP, Grove JI, Knox RJ, Hu L, Hyde EI, Searle PF. (2010).

“Testing double mutants of the enzyme nitroreductase for enhanced cell sensitisation to

prodrugs: effects of combining beneficial single mutations.” Biochemical Pharmacology.

79(2):102-11. PMID: 19665450

Prosser GA1, Copp JN, Syddall SP, Williams EM, Smaill JB, Wilson WR, Patterson AV,

Ackerley DF. (2010). “Discovery and evaluation of Escherichia coli nitroreductases that activate

the anti-cancer prodrug CB1954.” Biochemical Pharamcology. 79(5):678-87. PMID: 19852945

Grohmann M1, Paulmann N, Fleischhauer S, Vowinckel J, Priller J, Walther DJ. (2009). “A

mammalianized synthetic nitroreductase gene for high-level expression.” BMC Cancer. 9:301.

PMID: 19712451

Kwak SP1, Malberg JE, Howland DS, Cheng KY, Su J, She Y, Fennell M, Ghavami A. (2007).

“Ablation of central nervous system progenitor cells in transgenic rats using bacterial

nitroreductase system.” Journal of Neuroscience Research. 85(6):1183-93. PMID: 17304579

Bridgewater JA1, Knox RJ, Pitts JD, Collins MK, Springer CJ. (1997). “The bystander effect of

the nitroreductase/CB1954 enzyme/prodrug system is due to a cell-permeable metabolite.”

Human Gene Therapy. 8(6):709-17. PMID: 9113510

Page 145: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

127

3.1.5

White DT1, Mumm JS. (2013). “The nitroreductase system of inducible targeted ablation

facilitates cell-specific regenerative studies in zebrafish.” Methods (San Diego, Calif.).

62(3):232-40. PMID: 23542552

Bailey SM1, Knox RJ, Hobbs SM, Jenkins TC, Mauger AB, Melton RG, Burke PJ, Connors TA,

Hart IR. (1996). “Investigation of alternative prodrugs for use with E. coli nitroreductase in

'suicide gene' approaches to cancer therapy.” Gene Therapy. 3(12):1143-50. PMID: 8986441

Curado S1, Anderson RM, Jungblut B, Mumm J, Schroeter E, Stainier DY. (2007). “Conditional

targeted cell ablation in zebrafish: a new tool for regeneration studies.” Developmental

Dynamics: an official publication of the American Association of Anatomists. 236(4):1025-35.

PMID: 17326133

Pisharath H1, Rhee JM, Swanson MA, Leach SD, Parsons MJ. (2007). “Targeted ablation of beta

cells in the embryonic zebrafish pancreas using E. coli nitroreductase.” Mechanisms of

Development. 124(3):218-29. PMID: 17223324

Edwards DI1. (1993). “Nitroimidazole drugs--action and resistance mechanisms. I. Mechanisms

of action.” The Journal of Antimicrobial Chemotherapy. 31(1):9-20. PMID: 8444678

Lindmark DG, Müller M. (1976). “Antitrichomonad action, mutagenicity, and reduction of

metronidazole and other nitroimidazoles.” Antimicrobial Agents and Chemotherapy. PMID:

791102

Verdijk RM1, Wilke M, Beslier V, Kloosterman A, Brand A, Goulmy E, Mutis T. (2004).

“Escherichia coli-nitroreductase suicide gene control of human telomerase reverse transcriptase-

transduced minor histocompatibility antigen-specific cytotoxic T cells.” Bone Marrow

Transplant. 33(9):963-7. PMID: 15048139

Pisharath H1, Parsons MJ. (2009). “Nitroreductase-mediated cell ablation in transgenic zebrafish

embryos.” Methods in Molecular Biology (Clifton, N.J.). 546:133-43. PMID: 19378102

Page 146: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

128

Pisharath H1. (2007). “Validation of nitroreductase, a prodrug-activating enzyme, mediated cell

death in embryonic zebrafish (Danio rerio).” Comparative Medicine. 57(3):241-6. PMID:

17605338

Zhou L1, Feng Y1, Wang F1, Dong X2, Jiang L1, Liu C1, Zhao Q3, Li K4. (2018). “Generation

of all-male-like sterile zebrafish by eliminating primordial germ cells at early development.”

Scientific Reports. 8(1):1834. PMID: 29382876

Lalonde RL1, Akimenko MA1. (2018). “Effects of fin fold mesenchyme ablation on fin

development in zebrafish.” PLoS One. 13(2):e0192500. PMID: 29420592

Huang J1, McKee M, Huang HD, Xiang A, Davidson AJ, Lu HA. (2013). “A zebrafish model of

conditional targeted podocyte ablation and regeneration.” Kidney International. 83(6):1193-200

Zhou W1, Hildebrandt F. (2012). “Inducible podocyte injury and proteinuria in transgenic

zebrafish.” Journal of the American Society of Nephrology: JASN. 23(6):1039-47. PMID:

22440901

Walderich B1, Singh AP1, Mahalwar P1, Nüsslein-Volhard C1. (2016). “Homotypic cell

competition regulates proliferation and tiling of zebrafish pigment cells during colour pattern

formation.” Nature Communications. PMID: 27118125

Hsu CC1, Hou MF, Hong JR, Wu JL, Her GM. (2010a). “Inducible male infertility by targeted

cell ablation in zebrafish testis.” Marine Biotechnology (New York, N.Y.). 12(4):466-78. PMID:

19936986

Hu SY1, Lin PY, Liao CH, Gong HY, Lin GH, Kawakami K, Wu JL. (2010b). “Nitroreductase-

mediated gonadal dysgenesis for infertility control of genetically modified zebrafish.” Marine

Biotechnology (New York, N.Y.). 12(5):569-78. PMID: 19941022

Mathias JR1, Zhang Z, Saxena MT, Mumm JS. (2014). “Enhanced cell-specific ablation in

zebrafish using a triple mutant of Escherichia coli nitroreductase.” Zebrafish. 11(2):85-97.

PMID: 24428354

Page 147: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

129

Allen JR1, Bhattacharyya KD2, Asante E1, Almadi B1, Schafer K1, Davis J1, Cox J3, Voigt

M3, Viator JA2,4, Chandrasekhar A1. (2017). “Role of branchiomotor neurons in controlling

food intake of zebrafish larvae.” Journal of Neurogenetics. 31(3):128-137. PMID: 28812416

Godoy R1, Noble S1, Yoon K1, Anisman H2, Ekker M1. (2015). “Chemogenetic ablation of

dopaminergic neurons leads to transient locomotor impairments in zebrafish larvae.” Journal of

Neurochemistry. 135(2):249-60. PMID: 26118896

Shimizu Y1, Ito Y1, Tanaka H1, Ohshima T1. (2015). “Radial glial cell-specific ablation in the

adult Zebrafish brain.” Genesis. 53(7):431-9. PMID: 26045148

Montgomery JE1, Parsons MJ, Hyde DR. (2010). “A novel model of retinal ablation

demonstrates that the extent of rod cell death regulates the origin of the regenerated zebrafish rod

photoreceptors.” The Journal of Comparative Neurology. 518(6):800-14. PMID: 20058308

Chung AY1, Kim PS, Kim S, Kim E, Kim D, Jeong I, Kim HK, Ryu JH, Kim CH, Choi J, Seo

JH, Park HC. (2013). “Generation of demyelination models by targeted ablation of

oligodendrocytes in the zebrafish CNS.” Molecules and Cells. 36(1):82-7. PMID: 23807048

Willems B1, Büttner A, Huysseune A, Renn J, Witten PE, Winkler C. (2012). “Conditional

ablation of osteoblasts in medaka.” Developmental Biology. 364(2):128-37. PMID: 22326228

Otsuka T1, Takeda H1,2. (2017). “Targeted Ablation of Pancreatic β Cells in Medaka.”

Zoological Science. 34(3):179-184. PMID: 28589841

Choi RY1, Engbretson GA, Solessio EC, Jones GA, Coughlin A, Aleksic I, Zuber ME. “Cone

degeneration following rod ablation in a reversible model of retinal degeneration.” Investigative

Ophthalmology and Visual Science. 52(1):364-73. PMID: 20720220

Kaya F1, Mannioui A, Chesneau A, Sekizar S, Maillard E, Ballagny C, Houel-Renault L,

Dupasquier D, Bronchain O, Holtzmann I, Desmazieres A, Thomas JL, Demeneix BA, Brophy

PJ, Zalc B, Mazabraud A. (2012). “Live imaging of targeted cell ablation in Xenopus: a new

model to study demyelination and repair.” The Journal of Neuroscience: the official journal of

the Society for Neuroscience. 32(37):12885-95. PMID: 22973012

Page 148: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

130

Sekizar S1, Mannioui A, Azoyan L, Colin C, Thomas JL, Du Pasquier D, Mallat M, Zalc B.

(2015). “Remyelination by Resident Oligodendrocyte Precursor Cells in a Xenopus laevis

Inducible Model of Demyelination.” Developmental Neuroscience. 37(3):232-42. PMID:

25896276

McCormack E1, Silden E, West RM, Pavlin T, Micklem DR, Lorens JB, Haug BE, Cooper ME,

Gjertsen BT. (2013). “Nitroreductase, a near-infrared reporter platform for in vivo time-domain

optical imaging of metastatic cancer.” Cancer Research. 73(4):1276-86. PMID: 23233739

3.2

Kantor B1, Bailey RM2, Wimberly K2, Kalburgi SN2, Gray SJ3. (2014). “Methods for gene

transfer to the central nervous system.” Advances in Genetics. 87:125-97. PMID: 25311922

Manfredsson FP1,2. (2016). “Introduction to Viral Vectors and Other Delivery Methods for

Gene Therapy of the Nervous System.” Methods in Molecular Biology (Clifton, N.J.) 1382:3-18.

PMID: 26611575

Joshi CR1, Labhasetwar V2, Ghorpade A3,4. (2017). “Destination Brain: the Past, Present, and

Future of Therapeutic Gene Delivery.” Journal of Neuroimmune Pharmacology: The Official

Journal of the Society on Neuroimmune Pharmacology. 12(1):51-83. PMID: 28160121

3.2.1

Sharon D1, Kamen A1. (2018). “Advancements in the design and scalable production of viral

gene transfer vectors.” Biotechnology and Bioengineering. PMID: 28941274

Segura MM1, Mangion M, Gaillet B, Garnier A. (2013). “New developments in lentiviral vector

design, production and purification.” Expert Opinion on Biological Therapy. 13(7):987-1011.

PMID: 23590247

Durand S1, Cimarelli A. (2011). “The inside out of lentiviral vectors.” Viruses. 3(2):132-59.

PMID: 22049307

Page 149: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

131

Stellberger T1, Koehler N1, Dinkelmeier A1, Draxler J1, Haase M1, Hellinckx J1, Pavlovic M1,

Busch U1, Baiker A2. (2017). “Strategies and methods for the detection and identification of

viral vectors.” Virus Genes. 53(5):749-757. PMID: 28725978

Schlimgen R1, Howard J, Wooley D, Thompson M, Baden LR, Yang OO, Christiani DC,

Mostoslavsky G, Diamond DV, Duane EG, Byers K, Winters T, Gelfand JA, Fujimoto G,

Hudson TW, Vyas JM. (2016). “Risks Associated With Lentiviral Vector Exposures and

Prevention Strategies.” Journal of Occupational and Environmental Medicine. 58(12):1159-

1166. PMID: 27930472

Cornetta K1, Yao J, Jasti A, Koop S, Douglas M, Hsu D, Couture LA, Hawkins T, Duffy L.

(2011). “Replication-competent lentivirus analysis of clinical grade vector products.” Molecular

Therapy. The Journal of the American Society of Gene Therapy. 19(3):557-66. PMID: 21179010

Farley DC1, McCloskey L1, Thorne BA2, Tareen SU2, Nicolai CJ2, Campbell DJ2, Bannister

R1, Stewart HJ1, Pearson LJ1, Moyer BJ2, Robbins SH2, Zielinski L2, Kim T2, Radcliffe PA1,

Mitrophanous KA1, Gombotz WR2, Miskin JE1, Kelley-Clarke B2. (2015). “Development of a

replication-competent lentivirus assay for dendritic cell-targeting lentiviral vectors.” Molecular

Therapy. Methods and Clinical Development. 2:15017. PMID: 26029728

Jiang W1,2, Hua R1,2, Wei M1,2, Li C2, Qiu Z3, Yang X2, Zhang C1. (2015). “An optimized

method for high-titer lentivirus preparations without ultracentrifugation.” Scientific Reports.

5:13875. PMID: 26348152

3.2.2

Vargas JE1, Chicaybam L2,3, Stein RT1, Tanuri A4, Delgado-Cañedo A5, Bonamino MH6,7.

(2016). “Retroviral vectors and transposons for stable gene therapy: advances, current challenges

and perspectives.” The Journal of Translational Medicine. 14(1):288. PMID: 27729044

Di Matteo M1, Belay E, Chuah MK, Vandendriessche T. (2012). “Recent developments in

transposon-mediated gene therapy.” Expert Opinion on Biological Therapy. 12(7):841-58.

PMID: 22679910

Page 150: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

132

Skipper KA, Andersen PR, Sharma N, Mikkelsen JG1. (2013). “DNA transposon-based gene

vehicles - scenes from an evolutionary drive.” Journal of Biomedical Science. 20:92. PMID:

24320156

Ivics Z1, Li MA, Mátés L, Boeke JD, Nagy A, Bradley A, Izsvák Z. (2009). “Transposon-

mediated genome manipulation in vertebrates.” Nature Methods. 6(6):415-22. PMID: 19478801

Ivics Z1, Izsvák Z. (2010). “The expanding universe of transposon technologies for gene and cell

engineering.” Mobile DNA. 1(1):25. PMID: 21138556

Zhao S1, Jiang E1, Chen S1, Gu Y1, Shangguan AJ1, Lv T1, Luo L1, Yu Z1. (2016). “PiggyBac

transposon vectors: the tools of the human gene encoding.” Translational Lung Cancer

Research. 5(1):120-5. PMID: 26958506

Fraser MJ1, Ciszczon T, Elick T, Bauser C. (1996). “Precise excision of TTAA-specific

lepidopteran transposons piggyBac (IFP2) and tagalong (TFP3) from the baculovirus genome in

cell lines from two species of Lepidoptera.” Insect Molecular Biology. 5(2):141-51. PMID:

8673264

Fraser MJ1, Cary L, Boonvisudhi K, Wang HG. (1995). “Assay for movement of Lepidopteran

transposon IFP2 in insect cells using a baculovirus genome as a target DNA.” Virology.

211(2):397-407. PMID: 7645244

Beames B1, Summers MD. (1990). “Sequence comparison of cellular and viral copies of host

cell DNA insertions found in Autographa californica nuclear polyhedrosis virus.” Virology.

174(2):354-63. PMID: 2154879

Cary LC1, Goebel M, Corsaro BG, Wang HG, Rosen E, Fraser MJ. (1989). “Transposon

mutagenesis of baculoviruses: analysis of Trichoplusia ni transposon IFP2 insertions within the

FP-locus of nuclear polyhedrosis viruses.” Virology. 172(1):156-69. PMID: 2549707

Beames B1, Summers MD. (1988). “Comparisons of host cell DNA insertions and altered

transcription at the site of insertions in few polyhedra bacilovirus mutants.” Virology.

162(1):206-20. PMID: 2829419

Page 151: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

133

Fraser MJ, Brusca JS, Smith GE, Summers MD. (1985). “Transposon-mediated mutagenesis of a

baculovirus.” Virology. 145(2):356-61. PMID: 2992159

Fraser MJ1, Smith GE, Summers MD. (1983). “Acquisition of Host Cell DNA Sequences by

Baculoviruses: Relationship Between Host DNA Insertions and FP Mutants of Autographa

californica and Galleria mellonella Nuclear Polyhedrosis Viruses.” Journal of Virology.

47(2):287-300. PMID: 16789244

Li MA1, Turner DJ, Ning Z, Yusa K, Liang Q, Eckert S, Rad L, Fitzgerald TW, Craig NL,

Bradley A. (2011). “Mobilization of giant piggyBac transposons in the mouse genome.” Nucleic

Acids Research. 39(22):e148. PMID: 21948799

Wilson MH1, Coates CJ, George AL Jr. (2007). “PiggyBac transposon-mediated gene transfer in

human cells.” Molecular Therapy: The Journal of the American Society of Gene Therapy.

15(1):139-45. PMID: 17164785

Ding S1, Wu X, Li G, Han M, Zhuang Y, Xu T. (2005). “Efficient transposition of the piggyBac

(PB) transposon in mammalian cells and mice. Cell. 122(3):473-83. PMID: 16096065

Ivics Z1, Hackett PB, Plasterk RH, Izsvák Z. (1997). “Molecular reconstruction of Sleeping

Beauty, a Tc1-like transposon from fish, and its transposition in human cells.” Cell. 91(4):501-

10. PMID: 9390559

Radice AD1, Bugaj B, Fitch DH, Emmons SW. (1994). “Widespread occurrence of the Tc1

transposon family: Tc1-like transposons from teleost fish.” Molecular and General Genetics:

MGG. 244(6):606-12. PMID: 7969029

Muñoz-López M1, García-Pérez JL. (2010). “DNA transposons: nature and applications in

genomics.” Current Genomics. 11(2):115-28. PMID: 20885819

Yusa K1, Zhou L, Li MA, Bradley A, Craig NL. (2011). “A hyperactive piggyBac transposase

for mammalian applications.” Proceedings of the National Academy of Sciences of the United

States of America. 108(4):1531-6. PMID: 21205896

Li MA1, Pettitt SJ, Eckert S, Ning Z, Rice S, Cadiñanos J, Yusa K, Conte N, Bradley A. (2013).

“The piggyBac transposon displays local and distant reintegration preferences and can cause

Page 152: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

134

mutations at noncanonical integration sites.” Molecular and Cellular Biology. 33(7):1317-30.

PMID: 23358416

Cadiñanos J1, Bradley A. “Generation of an inducible and optimized piggyBac transposon

system.” Nucleic Acids Research. 35(12):e87. PMID: 17576687

Gogol-Döring A1,2, Ammar I3, Gupta S4, Bunse M3, Miskey C5, Chen W3, Uckert W3, Schulz

TF4, Izsvák Z3, Ivics Z5. (2016). “Genome-wide Profiling Reveals Remarkable Parallels

Between Insertion Site Selection Properties of the MLV Retrovirus and the piggyBac

Transposon in Primary Human CD4(+) T Cells.” Molecular Therapy: The Journal of the

American Society of Gene Therapy. 24(3):592-606. PMID: 26755332

Kawakami K1, Shima A, Kawakami N. (2000). “Identification of a functional transposase of the

Tol2 element, an Ac-like element from the Japanese medaka fish, and its transposition in the

zebrafish germ lineage.” Proceedings of the National Academy of Sciences of the United States

of America. 97(21):11403-8. PMID: 11027340

Kawakami K1, Shima A. (1999). “Identification of the Tol2 transposase of the medaka fish

Oryzias latipes that catalyzes excision of a nonautonomous Tol2 element in zebrafish Danio

rerio.” Gene. 240(1):239-44. PMID: 10564832

Kawakami K1, Koga A, Hori H, Shima A. (1998). “Excision of the tol2 transposable element of

the medaka fish, Oryzias latipes, in zebrafish, Danio rerio.” Gene. 225(1-2):17-22. PMID:

9931412

Koga A1. (2004). “Transposition mechanisms and biotechnology applications of the medaka fish

Tol2 transposable element.” Advances in Biophysics. 38:161-80. PMID: 15493333

Kawakami K1. (2007). “Tol2: a versatile gene transfer vector in vertebrates.” Genome Biology.

8 Suppl 1:S7. PMID: 18047699

Howes TR1, Summers BR2, Kingsley DM3,4. (2017). “Dorsal spine evolution in threespine

sticklebacks via a splicing change in MSX2A.” BMC Biology. 15(1):115. PMID: 29212540

Kikuta H1, Kawakami K. (2009). “Transient and stable transgenesis using tol2 transposon

vectors.” Methods in Molecular Biology (Cifton, N.J.). 546:69-84. PMID: 19378098

Page 153: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

135

Hamlet MR1, Yergeau DA, Kuliyev E, Takeda M, Taira M, Kawakami K, Mead PE. (2006).

“Tol2 transposon-mediated transgenesis in Xenopus tropicalis.” Genesis. 44(9):438-45. PMID:

16906529

Balciunas D1, Wangensteen KJ, Wilber A, Bell J, Geurts A, Sivasubbu S, Wang X, Hackett PB,

Largaespada DA, McIvor RS, Ekker SC. (2006). “Harnessing a high cargo-capacity transposon

for genetic applications in vertebrates.” PLoS Genetics. 2(11):e169. PMID: 17096595

Wu SC1, Meir YJ, Coates CJ, Handler AM, Pelczar P, Moisyadi S, Kaminski JM. (2006).

“piggyBac is a flexible and highly active transposon as compared to sleeping beauty, Tol2, and

Mos1 in mammalian cells.” Proceedings of the National Academy of Sciences of the United

States of America. 103(41):15008-13. PMID: 17005721

Grabundzija I1, Irgang M, Mátés L, Belay E, Matrai J, Gogol-Döring A, Kawakami K, Chen W,

Ruiz P, Chuah MK, VandenDriessche T, Izsvák Z, Ivics Z. (2010). “Comparative analysis of

transposable element vector systems in human cells.” Molecular Therapy: The Journal of the

American Society of Gene Therapy. 18(6):1200-9. PMID: 20372108

Urasaki A1, Morvan G, Kawakami K. (2006). “Functional dissection of the Tol2 transposable

element identified the minimal cis-sequence and a highly repetitive sequence in the subterminal

region essential for transposition.” Genetics. 174(2):639-49. PMID: 16959904

Huang X1, Guo H, Tammana S, Jung YC, Mellgren E, Bassi P, Cao Q, Tu ZJ, Kim YC, Ekker

SC, Wu X, Wang SM, Zhou X. (2010). “Gene transfer efficiency and genome-wide integration

profiling of Sleeping Beauty, Tol2, and piggyBac transposons in human primary T cells.”

Molecular Therapy: The Journal of the American Society of Gene Therapy. 18(10):1803-13.

PMID: 20606646

Chapter 3 - 4)

Clark KJ1, Carlson DF, Foster LK, Kong BW, Foster DN, Fahrenkrug SC. (2007). “Enzymatic

engineering of the porcine genome with transposons and recombinases.” BMC Biotechnology.

7:42. PMID: 17640337

Page 154: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

136

Chen YT1, Bradley A. (2000). “A new positive/negative selectable marker, puDeltatk, for use in

embryonic stem cells.” Genesis. 28(1):31-5. PMID: 11020714

Chapter 3 - 7)

Yuan C1, Gao J1, Guo J1, Bai L1, Marshall C2, Cai Z3, Wang L1, Xiao M1. (2014). “Dimethyl

sulfoxide damages mitochondrial integrity and membrane potential in cultured astrocytes.” PLoS

One. 9(9):e107447. PMID: 25238609

Page 155: Selective Ablation of Transgenic Neural Precursor Cells- A ......PGK: phosphoglycerate kinase PI3K/Akt/mTOR: phosphoinositide 3-kinase/ Protein kinase B/mechanistic target of rapamycin

137

Appendix I: List of Contributions

Dr. Mohamad Khazei: Providing the hiPSC-derived NPC line, as well as assistance with

learning many cell culture techniques and general guidance with regards to strategies for my

overall MSc project.

Dr. Chris Ahuja: Providing advice and suggestions for developing the monoclonal NTR-NPC

line, as well as the human-codon-optimized (HCO)-NTR-NPC cell line.

Dr. Mahmood Chamankhah: Aiding in developing the strategies for successful NTR-piggyBac

subcloning, as well as guiding and mentoring with subcloning experiments

Jian Wang: Additional assistance with learning and improving cell culture techniques

Priscilla Chan: Assisting with providing a protocol for the steps involved in NTR protein

extraction, quantification, and protein staining.

James Hong: Performing “peak analysis” image processing for quantification of remaining

attached cells in the context of HSV-TK NPC ablation experiments (Figures 10-12).