<<

Stem cells from patients with Congenital

Hyperinsulinism

A thesis submitted to the University of Manchester

for the degree of doctor of philosophy in the Faculty of Biology, Medicine and Health

School of Medical Sciences

Sophie Grace Kellaway

2016 Contents

0.1 Abbreviations ...... 9

0.2 Abstract ...... 11

0.3 Declaration ...... 12

0.4 Copyright statement ...... 12

0.5 Acknowledgements ...... 13

0.6 Thesis organisation ...... 14

0.7 Author note ...... 15

1 Literature Review 16

1.1 Introduction ...... 16

1.2 The ...... 17

1.2.1 Anatomy ...... 17

1.2.2 Development ...... 20

1.2.3 Post developmental endocrine proliferation ...... 23

1.2.4 Cell cycle in the pancreas ...... 23

1.2.5 Signalling and markers ...... 25

1.3 Congenital (CHI) ...... 27

1.3.1 Symptoms and causes ...... 27

1.3.2 Hyperproliferation ...... 30

1.3.3 Treatment ...... 30

1.3.4 Experimental models ...... 30

1.4 ...... 31

1.4.1 Epidemiology ...... 31

1.4.2 Pathogenesis ...... 32

1.4.3 Treatment ...... 33

1.5 Stem Cells ...... 34

1.5.1 Pancreatic stem cells ...... 34

1.5.2 Mesenchymal stem cells ...... 35

2 1.5.3 Pluripotent stem cells ...... 38

1.5.4 Generation of iPSCs ...... 41

1.6 Aims ...... 43

1.7 References ...... 43

2 A Novel Source of Mesenchymal Stem Cells derived from the Pancreas of Patients

with Congenital Hyperinsulinism. 64

2.1 Abstract ...... 65

2.2 Introduction ...... 66

2.3 Methods ...... 68

2.3.1 Derivation of CHI pancreatic mesenchymal stem cell (CHI pMSC) lines . . . . 68

2.3.2 Cell culture ...... 68

2.3.3 Karyotyping ...... 69

2.3.4 Short tandem repeat (STR) profiling ...... 69

2.3.5 RT-PCR and qRT-PCR ...... 69

2.3.6 Genotyping ...... 70

2.3.7 Adipogenesis, osteogenesis and chondrogenesis ...... 72

2.3.8 Flow Cytometry ...... 73

2.3.9 Immunocytochemistry (ICC) ...... 73

2.3.10 Bisulfite sequencing ...... 75

2.4 Results ...... 76

2.4.1 MSCs were derived from three CHI patients ...... 76

2.4.2 Derived cells were karyotypically stable, unique and contained the CHI causing

mutations ...... 83

2.4.3 CHI derived MSCs express pancreatic markers ...... 84

2.4.4 CHI pMSCs have altered gene expression and methylation patterns compared

to bmMSCs ...... 88

2.5 Discussion ...... 94

2.6 References ...... 97

3 Increased Proliferation and Altered Cell Cycle Regulation in Pancreatic Stem

Cells derived from Patients with Congenital Hyperinsulinism 104

3.1 Abstract ...... 105

3.2 Introduction ...... 106

3.3 Methods ...... 110

3.3.1 Cell culture ...... 110

3 3.3.2 Derivation of adult pancreatic mesenchymal stem cell (A pMSC) lines . . . . . 111

3.3.3 Flow Cytometry (propidium iodide) ...... 111

3.3.4 Immunocytochemistry (ICC) ...... 111

3.3.5 Western Blotting ...... 112

3.3.6 Ki67 stain and count ...... 112

3.4 Results ...... 114

3.4.1 Increased proliferation in CHI tissue and CHI-derived pMSCs ...... 114

3.4.2 Cell cycle regulation in CHI MSCs ...... 116

Kip1 3.4.3 Distribution of G1/S molecules - decreased nuclear p27 in CHI-derived pMSCs ...... 118

3.4.4 Distribution of G2/M molecules - possible increase of CDK1 in CHI-derived pMSCs ...... 120

3.4.5 and affect p27Kip1 protein levels and localisation ...... 122

3.5 Discussion ...... 125

3.6 References ...... 128

4 Stem Cell Derived β-cells: a new resource for Congenital Hyperinsulinism? 134

4.1 Abstract ...... 135

4.2 Introduction ...... 136

4.3 Methods ...... 140

4.3.1 MSC culture ...... 140

4.3.2 Feeder dependent iPSC culture ...... 140

4.3.3 Feeder free iPSC culture ...... 141

4.3.4 Derivation of iPSCs ...... 141

4.3.5 Three germ layer iPSC differentiation ...... 141

4.3.6 MSC differentiation ...... 142

4.3.7 Differentiation: Melton/Kieffer ...... 142

4.3.8 RT-PCR and qRT-PCR ...... 144

4.3.9 Bisulfite sequencing ...... 146

4.3.10 Immunocytochemistry (ICC) ...... 147

4.3.11 Western blot ...... 148

4.3.12 Glucose-stimulated insulin secretion (GSIS) assay ...... 148

4.4 Results ...... 151

4.4.1 Serum withdrawal was insufficient to induce differentiation of CHI MSCs . . . 151

4.4.2 A step-wise differentiation protocol shows stage specific gene induction, en-

hanced by the addition of DKK1 ...... 152

4 4.4.3 Phenotypic analysis of differentiated CHI pMSCs ...... 156

4.4.4 Spheroid formation was essential for differentiation ...... 160

4.4.5 Derivation of iPSCs ...... 161

4.4.6 Differentiation of CHI-iPSCs to definitive endoderm ...... 167

4.4.7 Differentiation of CHI-iPSCs to pancreatic endoderm ...... 168

4.4.8 Differentiation of CHI-iPSCs to hormone positive islet cells ...... 169

4.5 Discussion ...... 174

4.6 References ...... 180

5 Conclusions 189

5.1 CHI pMSCs were derived and have a pancreatic phenotype ...... 189

5.2 CHI pMSCs were differentiated to immature β-like cells ...... 190

5.3 A CHI iPSC line could be differentiated to pancreatic cells ...... 192

5.4 Increased proliferation in CHI ...... 193

5.5 Conclusion ...... 194

5.6 Future work ...... 194

5.7 References ...... 197

5 List of Figures

1.1 Islet structure ...... 17

1.2 Regulation of insulin secretion ...... 18

1.3 Development of the pancreas ...... 20

1.4 Cell cycle in the pancreas ...... 24

1.5 Histological differences between focal and diffuse CHI ...... 29

1.6 Insulin secretion in CHI ...... 29

1.7 Sources of MSCs ...... 36

1.8 EMT in pMSCs ...... 37

2.1 Derived cell lines showed characteristic features of MSCs ...... 77

2.2 Derived cell lines showed multipotency, characteristic of MSCs ...... 79

2.3 Mesenchymal proteins were expressed in CHI pMSCs ...... 81

2.4 CHI pMSCs are genetically stable and unique ...... 82

2.5 Pancreatic genes are expressed in CHI pMSCs ...... 84

2.6 Growing cells on vitronectin and fibronectin increased cell number and insulin gene

expression ...... 86

2.7 Pancreatic proteins are expressed in CHI pMSCs ...... 87

2.8 Comparison of gene expression between CHI pMSCs and bmMSCs ...... 88

2.9 Comparison of gene methylation between CHI pMSCs and bmMSCs ...... 91

2.10 Bisulfite sequencing and gene sequencing of SPP1 ...... 93

3.1 Increased proliferation in CHI tissue compared to age-matched controls ...... 114

3.2 Increased proliferation in CHI-derived MSCs compared to adult controls ...... 115

3.3 Alterations to cell cycle control in CHI-derived MSCs compared to adult controls . . . 117

3.4 Altered levels of nuclear p27Kip1 in CHI MSCs ...... 119

3.5 Levels of G2/M regulators in CHI and adult MSCs ...... 121

3.6 Glucose and insulin concentrations determine p27Kip1 localisation ...... 124

4.1 Melton/Kieffer differentiation protocols ...... 143

6 4.2 Serum withdrawal enhanced spheroid formation but was not sufficient alone ...... 151

4.3 Step-wise differentiation induces and reduces gene expression ...... 153

4.4 Spheroids treated with differentiation inducers were of a similar size to human pan-

creatic islets ...... 155

4.5 Some untreated spheroids after 21 days have a capsule ...... 156

4.6 Differentiated CHI pMSCs express INS and PDX1 ...... 158

4.7 Differentiated spheroids contained and secreted more insulin than control spheroids . . 159

4.8 Cells differentiated as monolayer do not have the same expression profile as spheroids 160

4.9 MSCs were transduced and cleared the SeV ...... 162

4.10 CHI-iPSCs were phenotypically and functionally pluripotent ...... 164

4.11 Transition of iPSCs to feeder-free culture ...... 165

4.12 CHI-iPSCs did not maintain hypomethylation seen in CHI-pMSCs ...... 170

4.13 CHI iPSCs efficiently differentiate to definitive endoderm ...... 171

4.14 CHI iPSCs differentiate to definitive endoderm by D’amour protocol only when cul-

tured on feeders ...... 172

4.15 CHI iPSCs efficiently differentiate to pancreatic endoderm ...... 172

4.16 CHI iPSCs differentiated to cells with characteristics of immature α and β-cells . . . . 173

7 List of Tables

1.1 Cross-section of MSC to β-cell differentiation protocols ...... 39

1.2 Cross-section of MSC to β-cell differentiation protocols ...... 40

2.1 Primers used for RT-PCR, details on design and use are in Section 2.3.5. Primers

indicated with * from Ellard et al. [2007] ...... 72

2.2 Antibodies used in the study ...... 74

2.3 Primers used for bisulfite sequencing ...... 75

2.4 Details of cell lines and patients ...... 76

2.5 CD marker expression ...... 78

3.1 Details of cell lines and patients ...... 110

3.2 Antibodies used in the study ...... 113

4.1 Details of cell lines and patients ...... 140

4.2 Primers used for RT-PCR ...... 146

4.3 Primers used for bisulfite sequencing ...... 147

4.4 Antibodies used in the study ...... 150

8 0.1 Abbreviations

AU - arbitrary units bFGF - basic fibroblast growth factor bmMSCs - bone marrow mesenchymal stem cells

BSA - bovine serum albumin

Btc - betacellulin

CDK - cyclin dependent kinase

CHI - congenital hyperinsulinism

CKI - cyclin dependent kinase inhibitor

CT - cycle threshold DAPI - 4’,6-diamidino-2-phenylindole

ECL - enhanced chemiluminescent

EGF - epidermal growth factor

ELISA - enzyme-linked immunosorbent assay

EMT - epithelial-mesenchymal transition

Ex-4 - exendin-4

FBS - foetal bovine serum

G6P - glucose-6-phosphate

GLP - glucagon-like peptide

GSIS - glucose stimulated insulin secretion hESCs - human embryonic stem cells

HGF - hepatocyte growth factor

HRP - horse radish peroxidase

IBMX - 3-isobutyl-1-methylxanthine

ICC - immunocytochemistry

IGFR - insulin-like growth factor receptor

INS - insulin iPSCs - induced pluripotent stem cells

IR - insulin receptor

ITS - insulin/transferrin/selenium

KGF - keratinocyte growth factor

KRH - Krebs-Ringer-HEPES

LoH - loss of heterozygosity

MET - mesenchymal-epithelial transition

Mol - moles

9 MPCs - multipotent progenitor cells

MSCs - mesenchymal stem cells

NaB - sodium butyrate

NEAA - non-essential amino acids

NGS - normal goat serum

Nic - nicotinamide

P - passage

PBS - phosphate buffered saline

PCR - polymerase chain reaction

PdBU - phorbol 12,13-dibutyrate

PDL - pancreatic duct ligation PI - propidium iodide pMSCs - pancreatic mesenchymal stem cells

PVDF - polyvinylidene fluoride

SDS - sodium dodecyl sulphate

STR - short tandem repeat

TPB - ((2S,5S)-(E,E)-8-(5-(4-(trifluoromethyl)phenyl)-2,4-pentadienoylamino)benzolactam

TSA - trichostatin-A

10 0.2 Abstract

The University of Manchester Sophie Grace Kellaway Doctor of Philosophy 2016

Stem cells from patients with Congenital Hyperinsulinism

Diabetes and congenital hyperinsulinism (CHI) are severe diseases affecting the pancreas. Cur- rent models for testing drugs to treat these diseases are in vivo in rodents or isolated rodent islets.

Differences between the human and rodent pancreas, and ethical issues, mean that in vitro human models are needed. To develop a novel in vitro model for pancreatic diseases, mesenchymal stem cells

(MSCs) and induced pluripotent stem cells (iPSCs) were derived from the pancreas of patients with

CHI. MSCs from three forms of CHI were phenotypically normal for MSCs, and maintained the CHI- causing mutation. When compared to MSCs from bone marrow, the CHI pancreatic MSCs expressed pancreas-specific gene ISL1 and showed promoter hypomethylation of other pancreatic genes, includ- ing PDX1. The CHI pMSCs could be differentiated to cells resembling immature β-cells, with some

β-cell gene expression (INS, PDX1 ), but no glucose responsive insulin secretion. CHI associated hypersecretion of insulin was not seen as the ATP-sensitive potassium KATP channels were not being expressed. Addition of the Wnt inhibitor DKK1 markedly enhanced differentiation via induction of neuronal genes. Alongside high insulin secretion, CHI also features increased proliferation. CHI

MSCs were also hyperproliferative, and showed alterations to the cell cycle. These changes were related to p27Kip1 localisation, a known affected protein in CHI tissue, and CDK1, a novel regulator for CHI. iPSCs were also derived from focal CHI MSCs and were also phenotypically normal, but did not maintain the pancreatic hypomethylation present in MSCs. The CHI iPSCs were efficiently differentiated to definitive endoderm and PDX1 positive cells. Terminally differentiated iPSCs were endocrine, but were not mature β-cells. In conclusion, authentic MSCs and iPSCs were derived for the first time from patients with CHI. These stem cells could be differentiated towards β-cells, but mature glucose responsive β-cells were not produced. MSC derived β-like cells secreted insulin but did not have KATP channels, whereas iPSC derived β-like cells had KATP channel gene expression but not INS. With further optimisation to resolve these, CHI stem cell derived β-cells may be used for in vitro modelling. Further, the undifferentiated MSCs only show hyperproliferation associated with p27Kip1 and CDK1 and so can be a useful resource for modelling hyperproliferation seen in CHI.

11 0.3 Declaration

I declare that no portion of the work referred to in the thesis has been submitted in support of an application for another degree or qualification of this or any other university or other institute of learning.

0.4 Copyright statement i. The author of this thesis (including any appendices and/or schedules to this thesis) owns certain copyright or related rights in it (the Copyright) and s/he has given The University of Manchester certain rights to use such Copyright, including for administrative purposes.

ii. Copies of this thesis, either in full or in extracts and whether in hard or electronic copy, may be made only in accordance with the Copyright, Designs and Patents Act 1988 (as amended) and regulations issued under it or, where appropriate, in accordance with licensing agreements which the

University has from time to time. This page must form part of any such copies made.

iii. The ownership of certain Copyright, patents, designs, trade marks and other intellectual property (the Intellectual Property) and any reproductions of copyright works in the thesis, for ex- ample graphs and tables (Reproductions), which may be described in this thesis, may not be owned by the author and may be owned by third parties. Such Intellectual Property and Reproductions cannot and must not be made available for use without the prior written permission of the owner(s) of the relevant Intellectual Property and/or Reproductions.

iv. Further information on the conditions under which disclosure, publication and commer- cialisation of this thesis, the Copyright and any Intellectual Property University IP Policy (see http://documents.manchester.ac.uk/display.aspx?DocID=24420), in any relevant Thesis restriction declarations deposited in the University Library, The University Librarys regulations (see http://www.library.manchester.ac.uk/about/regulations/) and in The Universitys policy on Presen- tation of Theses

12 0.5 Acknowledgements

First and foremost, I would like to thank Mark and Karen for having me in their lab for the last

3 years, and for setting up this project. Thanks also, to my colleagues for their helpful discussions and assistance: Karolina, Bing, Amna, Helga, Saba, Aleks and Alex. Particularly to Karolina, who has worked closely with me on getting the cells to where they needed to be, and for the many cell culture swaps when I needed a day off, and to Alex for some superb comments on the manuscripts!

For their technical assistance essential to this project, my thanks to: Roger Meadows (slides- canner), Steve Marsden (confocal microscopy), Fraser Coombe and Paul Fuller (DNA sequencing),

Graeme Fox (STR profiling), Mike Jackson (flow cytometry). Similarly, thank you to the surgical team for helping us with obtaining the tissue in a viable state, and to the parents of the patients who gave their consent for the tissue to be used for research.

Thank you for the NC3Rs for funding this project.

Finally, thank you to my mum and Ste, who have kept me going throughout, especially when I found out that the first cells were not what I thought they were!

13 0.6 Thesis organisation

My thesis is based on derivation, characterisation and differentiation of both mesenchymal stem cells

(MSCs) and induced pluripotent stem cells (iPSCs) derived from patients with congenital hyperin- sulinism (CHI).

I have written the thesis in the ‘alternative format’, comprising three paper-style chapters, a general literature review and overall discussion, which connects and compares the results chapters.

None of these chapters are published yet, although a manuscript based on the first chapter is being submitted for publication. I have chosen this format as:

ˆ My research easily separates into these main sections

ˆ Some work included only makes sense in the context of my colleagues’ unpublished work

ˆ I hope that having the work written in a format suitable for publication will allow dissemination

of the results

ˆ This format has allowed practice of writing manuscripts, a skill I will need going forward

In the first chapter I describe the derivation and characteristics of three MSC lines from patients with CHI. All work in this chapter, including deriving cell lines from primary tissue was done by me, except for some replicates of flow cytometry for CD markers, which were done by Karolina Mosinska.

The second chapter is based on exploration of the hypothesis that hyperproliferation seen in CHI will also be seen in CHI stem cells. In this chapter, the data for the first figure was generated and analysed by Bing Han, who has characterised proliferation in multiple CHI patients including those which I derived cells from. All other data, experimental design and analysis was my own. The final results chapter is on differentiation of the CHI MSCs to β-cells. The protocol used was based on a published protocol, and was chosen based on screening of multiple published protocols by Karolina and myself. The negative results of this screening are not included but were essential for the work that followed, hence the attribution to Karolina. The assessment of the final protocol and discovery of enhancement of β-cell differentiation by DKK1 was my own work. This chapter also explains the derivation and differentiation of iPSCs, derived from the previously discussed MSCs. This was all my own work, except for one experimental replicate of figure 5.5C, SOX17, contributed by Karolina where I found mine had failed after ceasing lab work. Other authorships are attributed to the medical team who care for the CHI patients, Alexander Ryan for assistance with preparation and proofing the manuscripts, and to Karen Cosgrove and Mark Dunne who conceived the project and provided guidance throughout.

14 The thesis therefore forms a body of work based on my own hypotheses and work, including the following contributions to knowledge:

ˆ Proof of principle of deriving MSCs from CHI tissue, and a replicable method to do so

ˆ Demonstration that CHI pancreatic MSCs (pMSCs) have gene expression and methylation not

seen in non-pancreatic (bone marrow) MSCs

ˆ The observation that CHI pMSCs show innately higher rates of proliferation than healthy adult

pMSCs, correlated with the CHI tissue

ˆ The finding that increased CHI pMSC proliferation may be linked to p27Kip1 localisation

(known from CHI tissue) and CDK1 (not previously studied)

ˆ That the use of DKK1 in differentiation of pMSCs enhances generation of β-like cells, via

induction of neuronal genes which can be switched to pancreatic fate with further factors

ˆ Published differentiation protocols used on CHI pMSCs and iPSCs can be used to produce

endocrine cells, but further work is needed to develp them for in vitro modelling of CHI or

diabetes

ˆ iPSCs can be generated from pMSCs, but this does not allow maintenance of hypomethylation

Word count: 55,093

0.7 Author note

This thesis involves characterisation of novel cell lines, and includes a strong focus on confirming the identity and origins of the cells. The level to which this is done, using multiple methods for each cell line may seem above what is normally expected. This is because for the first year of the project, I was working on cell lines identified as CHI pancreatic progenitors, which had been cross-contaminated or misidentified by a former student without our knowledge. A recent article in Nature News (Butler,

2015 1) suggests that despite the known phenomenon of misidentification of large numbers of cell lines, scientists are still lax in confirming the identity of what they are working with, and so many results may be invalidated. As such, I wanted to be sure that the cell lines I derived were the correct cell type, from the patients that they were supposed to be and had a genetic fingerprint for future use.

Some journals require this information now with manuscript submission, although not necessarily in the main text of the paper. With this caveat, I have chosen to include this information in the main text of these papers as I feel it is important for reporting novel cell lines after our experiences.

1D. Butler. Biomedical researchers lax in checking for imposter cell lines. Nature News, 2015

15 Chapter 1

Literature Review

1.1 Introduction

Diabetes is the most common, and fastest growing endocrine disease; it is treatable with insulin injections, medications and lifestyle changes, but not curable. Islet transplantation can offer a cure in some cases, but relies on more donors than are available (Shapiro et al., 2000). Therefore hu- man embryonic stem cells (hESCs), induced pluripotent stem cells (iPSCs) and pancreatic endocrine progenitors have been considered to provide a means of producing a large volume of β-cells for trans- plantation. Research into other cures for diabetes is ongoing, options being explored include drugs to cause β-cell proliferation or neogenesis, using stem cells (Godfrey et al., 2012). This research is largely dependent on rodent β-cell lines, many of which are poorly responsive to glucose and so not ideal models (Skelin et al., 2010). In order to maintain proper glucose-stimulated insulin secretion

(GSIS), β-cells should be derived from animals immediately prior to experimentation (Rees and Al- colado, 2005). Unfortunately, this leads to large numbers of animals being sacrificed, and still yields an imperfect model as there are differences between human and rodent islets such as structure and metabolism (MacDonald et al., 2011, Pan and Wright, 2011, Cabrera et al., 2006). In line with the

‘3Rs’ - reduction, refinement and replacement of animals in scientific research (Russell and Burch,

1959), and to reduce the effect of differences between human and rodent cells, a human stem cell model which can be induced to differentiate to β-cells as required could be used. HESCs and iP-

SCs have been researched for this purpose too, but differentiation protocols are long and inefficient

(D’Amour et al., 2006, Maehr, 2011, Lumelsky et al., 2001), and final differentiated cells can have genetic or proteomic profiles not consistent with true β-cells (Liew et al., 2008, Hansson et al., 2004,

Hori et al., 2002).

We hypothesise that by using human pancreatic stem cells, a number of these problems will be

16 negated. The pancreatic origin means differentiated cells are likely to be as authentic as possible

(Bar-Nur et al., 2011), there will be none of the differences associated with rodent islets, differ- entiation will not require numerous intermediate stages and a reduction in the number of animals sacrificed should occur. Differentiation of stem cells typically can rely on matching the developmental processes (Best et al., 2008, Hanley, 2014) as such this review will cover both the development and architecture of the pancreas, as well as current stem cell based research.

1.2 The Pancreas

1.2.1 Anatomy

Gross Anatomy

Figure 1.1: Islet structure Schematic showing how human and rodent islet structure differs; rodents have a core of β-cells and a mantle of other cell types whilst humans have all the cells mixed throughout (Cabrera et al., 2006).

The pancreas is a glandular organ comprised of exocrine and endocrine tissue. The exocrine tissue is made up of acinar cells clustered at the end of ducts, the purpose of which is to secrete and channel digestive enzymes, mucins and bicarbonate to the duodenum. The endocrine tissue is arranged in islets of Langerhans and made up of 5 major cell types, along with vessels, neurons and mesenchymal support tissue. Islets in rodents are made up of a core and mantle whilst in humans are more mixed (Cabrera et al., 2006); this is shown in Figure 1.1. The cells are α-cells which se- crete glucagon, β-cells which secrete insulin, δ-cells which secrete somatostatin, -cells which secrete ghrelin and PP-cells which secrete pancreatic polypeptide (Elayat et al., 1995, Brissova et al., 2005).

All of these secretions are into the surrounding microvasculature and in response to signals from the blood such as glucose levels. The endocrine and exocrine are compartmentally, morphologically and functionally discrete, and do not interact except in cases of extreme stresses, as reviewed by (Pan and Wright, 2011).

17 β-cells

The role of β-cells is to synthesise, package and release insulin in response to varying blood glucose levels, in a process known as GSIS. Insulin is a protein synthesised in the cells as preproinsulin, this is cleaved to active insulin and C-peptide. Transcription of the INS gene is caused by several transcription factors including NEUROD1, PDX1, PAX6 and ISL1 (Melloul et al., 2014, Petersen et al., 1994, Naya et al., 1995, Sander et al., 1997, Zhang et al., 2009). Glucose sensing occurs due to a signalling cascade triggered when the glucose transporters (GLUT1, 2, 4) move glucose from neighbouring blood vessels into the cells. This stimulates ATP generation which closes ATP- sensitive potassium (KATP) channels made of Kir6.2/Sur1 subunits (Aguilar-Bryan et al., 1998), causing depolarisation of the cell (Petit and Loubatieres-Mariani, 1992, Ashcroft and Rorsman, 1990,

Koster et al., 2005). Intracellular calcium rises, which causes exocytosis of stored insulin granules into the bloodstream (Petit and Loubatieres-Mariani, 1992, Braun et al., 2008, Fridlyand et al., 2013).

This is shown schematically in Figure 1.2. This signal is amplified by GCK which phosphorylates intracellular glucose to glucose-6-phosphate (G6P); this process is not inhibited by G6P and so metabolism is driven constantly by any glucose available, amplifying insulin release (Matschinsky,

2002). As stored insulin is quickly secreted in response to glucose sensing, glucose also causes further transcription of the insulin genes (Leibiger et al., 2002, Evans-Molina et al., 2007). Glucose modulates

PDX1 which binds and activates the promoters of INS, GLUT2 and glucokinase (GCK ), whilst also affecting chromatin accessiblity (Evans-Molina et al., 2007). This glucose dependent transcription is independent of secretion (Macfarlane et al., 2000).

Figure 1.2: Regulation of insulin secretion Insulin is secreted in response to glucose due to calcium influx caused by depolarisation when potassium is pumped out of ATP-sensitive channels, this occurs because of increased levels of ATP generated by glucose metabolism after transport by GLUT receptors (Koster et al., 2005, Braun et al., 2008).

18 β-cells are essential for the regulation of glucose levels in the body by their secretion of insulin, and dysfunction and/or destruction of β-cells causes diabetes, reviewed in Atkinson et al. (2013),

Kahn et al. (2014). One method that has been considered to cure diabetes therefore, is replacement of islets, β-cells, differentiation of progenitor cells within the pancreas to β-cells or drugs to cause proliferation of β-cells (Shapiro et al., 2000, Godfrey et al., 2012, Yi et al., 2013). This will be discussed further throughout the literature review.

Structure and Extracellular Matrix

Extracellular matrix (ECM) is a network of proteins and sugars which surrounds all cells in the body.

Cells interact with the ECM primarily via surface receptors called integrins. These interactions are important for .

Adult islets typically exist in an ECM capsule composed of fibroblasts and secreted collagen, which is in turn surrounded by the periinsular basement membrane, composed of laminin and collagen

(Stendahl et al., 2009). During development, however, the ECM is mostly composed of vitronectin,

fibronectin and collagen IV, particularly around progenitor cells. Vitronectin and fibronectin levels decrease as progenitors develop into islets (Stendahl et al., 2009).

There are many different integrins which recognise unique motifs within ECM components. For example, the α5 integrin recognises fibronectin and the αvβ3 heterodimeric integrin acts as a recep- tor for vitronectin. Most integrins are present in the adult and/or foetal pancreas, allowing binding to varied ECM. The αvβ3 integrin is highly expressed in the developing foetal pancreas but is down regulated as islets begin to form, suggesting an involvement with progenitor cell maintenance (Cirulli et al., 2000).

Culturing cells on extracellular matrix enhances survival. Whilst cells secrete their own ECM in order to prevent anoikis, it is often not the native matrix that is best for them. Isolated islets or purified β-cells show improved survival and maintenance of islet structure when cultured with ECM, whilst incomplete islets with native ECM show better survival than complete islets with no ECM

(Stendahl et al., 2009). However, β-cells cultured on collagen-IV, fibronectin or vitronectin show diminished insulin secretion (Kaido et al., 2006), perhaps due to the fact these matrix components are mainly seen in early differentiation, so may be involved in anti-differentiation signalling.

19 1.2.2 Development

Organogenesis

Figure 1.3: Development of the pancreas The graphical representation of the development of the pancreas shows each stage as follows. The primitive gut tube forms dorsal and ventral buds, these become the early pancreas. This is followed by cluster budding in the epithelium in the primary transition, then the secondary transition, where microlumina form containing the progenitor cells. The committed endocrine progenitors delaminate from the microlumina and assemble into the early islets. The blue, green and yellow circles represent progenitor cells which are committed to becoming β, α and δ cells, the branched structure represents the microlumen.

The pancreas is derived from the endoderm (Wells and Melton, 1999), and as with all organs goes through a number of stages during formation before all cells are mature and functional. The development has been well characterised in rodents but due to technical and ethical challenges less is known in humans. From what has been observed though, there seem to be strong similarities in developmental processes and signalling, just with different time scales. Several signalling pathways

20 are involved in the development process, with most of them being involved in multiple stages in context or level dependent functions (Wilson et al., 2003). The final organ size, potential for cell division and specific differentiation seems to be pre-determined early, potentially through epigenetic mechanisms and may affect how signalling pathways function on individual cells (Pan and Wright,

2011).

Initially the pancreas is derived from the embryonic foregut (primitive gut tube) and separates from cells destined to be intestine or (Wells and Melton, 1999). Specification of the pancreas is by expression of PDX1, which commits the cells early on to a pancreatic fate (Bouwens and Rooman,

2005, Puri and Hebrok, 2010). Dorsal and ventral buds are formed, which elongate, rotate and fuse to become the definitive pancreas (Piper et al., 2004, Gittes, 2009). The first major stage of pancreatic development is known as the primary transition. In this stage there is a large amount of proliferation in pancreatic progenitors to generate stratified epithelium, and endocrine cells begin to develop by cluster budding (Pan and Wright, 2011). Most of the endocrine cells at this stage are short lived, but Ngn3 lineage tracing shows at least some of the cells become incorporated into the newborn islet mantle (Gu et al., 2002). These Ngn3+ cells may last to adulthood, based on the assumption (and evidence seen in this experiment and others by the group) that Ngn3 cells do not renew themselves like stem cells; this has been disputed by other groups (Pan and Wright, 2011).

In the secondary transition, microlumina are formed and segregate into distinct areas. The trunk of the microlumen contains endocrine biased progenitors and the tip contains multipotent progenitor cells (MPCs) which become exocrine cells (Kopp et al., 2011). At this stage the endocrine biased progenitors in the trunk become committed and delaminate from the epithelium whilst exocrine destined cells remain (Puri and Hebrok, 2010). This delamination process resembles epithelial-to- mesenchymal transition (EMT) and allows the cells to assemble into islets as they divide. The endocrine progenitors divide to become endocrine precursors, then the distinct endocrine cell types.

The progenitors form the separate cell types whilst also forming into the eventual islet structures, which become histologically evident and express hormones approximately 12-13 weeks post concep- tion. Most of the mature endocrine cells are derived from the early endocrine progenitors, but around

20% of β-cells are derived from β-cell replication which becomes a predominate, but not exclusive mechanism for increasing mass after birth (Bonner-Weir et al., 2010).

The stages of development are shown graphically in Figure 1.3.

21 β-cells

β-cells develop from early progenitors, through to endocrine precursors, immature β-cells, then fi- nally mature β-cells. Foetal progenitors are stimulated to divide by FGF, and to differentiate by

Notch signalling in a process known as neogenesis. Foetal neogensis is the main method used to increase β-cell mass at these early stages and is stimulated by glucose, Glp-1, gastrin, exendin-4 and insulin; glucose also acts to suppress apoptosis during neogensis. In contrast to the adult pancreas, neogenesis contributes around 90% of the β-cell mass, with only 10% being produced by β-cell repli- cation (Bouwens and Rooman, 2005). It is unclear whether the adult pancreas loses the ability to respond to or to produce these signals, or whether the effect is dependent on levels of expression of the signals or other factors.

The best marker of a mature β-cell is functional i.e. GSIS. Immature cells are not able to ade- quately respond to glucose challenges, so maturity is important for generating β-cells in vitro. Insulin is an end stage β-cell marker not generally expressed along the differentiation pathway; but insulin is not a suitable marker of β-cell maturity for two reasons. INS can be expressed in endocrine pre- cursors and immature β-cells (Blum et al., 2012, Pan and Wright, 2011), and if in vitro cell cultures are examined many cells absorb insulin from medium (Hansson et al., 2004). β-cell populations are heterogeneous, with various levels of maturity seen (Pipeleers, 1992, Szabat et al., 2009, 2012).

When working with β-cells a marker of maturity is essential, as it is not always practical to measure

GSIS. The transcription factors MafA/B are one option. In rodents, immature β-cells are MafB positive, mature β-cells are MafA positive, mature alpha cells remain MafB positive, and during the maturation process the β-cells also go through a MafA/MafB double positive stage related to Pdx1 upregulation (Nishimura et al., 2006, Artner et al., 2007). The case in humans is less clear though, although MAFA is still used as a marker of β-cells in many cases. Urocortin 3 (UCN3) is also a good marker of mature β-cells, positive cells are seen in adult islets and authentically differentiated in vitro β-cells, whilst those in vitro cells which demonstrate poor GSIS are UCN3 negative (Blum et al., 2014). It is important to note though, that UCN3 is a marker only and not an instigator of maturation - it may be that it is upregulation by transcription factors involved in maturation, or acts in combination with other maturation factors (Blum et al., 2012). UCN3 may also be expressed in

α-cells, particularly when derived from ESCs although this may be suggestive of poor differentiation

(van der Meulen et al., 2012).

22 1.2.3 Post developmental endocrine proliferation

In order to increase β-cell mass in diabetic patients, a source for the new β-cells must be found.

The neonatal developmental pathways may not be applicable to adults, if the stem/progenitor cells present during embryogenesis have been terminally differentiated. To this end, experiments have been done to ascertain if the adult pancreas is capable of regeneration.

After a major insult it is possible for the rodent pancreas to regenerate by neogenesis from duct-like progenitors (Li et al., 2010). Depending on the type of injury, the pancreas regenerates differently - pancreatic duct ligation only allows neogenesis of exocrine cells, not β-cells (Rankin et al., 2013, Cavelti-Weder et al., 2013). This regeneration requires signalling from EGF, Glp-1 and

INGAP, reviewed by Bonner-Weir and Weir (2005), but without insult these factors are not enough to trigger neogenesis, although INGAP can initiate duct proliferation (Pittenger et al., 2009). Ad- ditionally after injury, Ngn3+ cells can convert to islet cells, seemingly via a duct-like progenitor role due to gastrin signalling (Rooman et al., 2002, Bouwens and Rooman, 2005). It is possible that the progenitors involved in these processes are not distinct progenitors, but just plastic and less differentiated β-cells. Alongside neogenesis from progenitor cells, experiments have shown β-cells duplicate (Dor et al., 2004, Yi et al., 2013). All of these experiments were performed in rodents, and there is little evidence of whether the same phenomena occur in humans and so ex vivo human mod- els are used to investigate if endocrine cells can be produced from the adult pancreas (see section 1.5).

1.2.4 Cell cycle in the pancreas

Control of the cell cycle is important for regulating the number and size of cells. Quiescent cells are said to be in G0, but all cells which are mitotically active can be allocated to a stage of the cell cycle. These cell cycle stages are Gap 1 or G1-phase where cells are resting or growing, Synthesis or

S-phase where DNA is duplicated, Gap 2 or G2-phase where the cell prepares to divide and Mitosis or M-phase where cell division occurs and two daughter cells are produced, reviewed by Harashima et al. (2013). The progression through the G1/S checkpoint commits a cell to division (Bertoli et al.,

2013), the G2/M checkpoint controls both DNA integrity and cell size at the point of division (Stark and Taylor, 2004). Cell cycle progression, particularly through G1/S, is controlled by both positive and negative regulator proteins, with members of the cyclin and cyclin-dependent kinase (CDK) families of proteins being involved in positive regulation, and members of the Cip/Kip and Ink fam- ilies being negative cell cycle regulators, via repression of the cyclins and CDKs as cyclin-dependent kinase inhibitors (CKIs).

23 Figure 1.4: Cell cycle in the pancreas Cell cycle control differs between cell types: In A-D green shapes indicate cell cycle activators and red shapes indicate cell cycle inhibitors. (A) In proliferative cells, cell cycle activators reside in the nucleus and promote cell cycle entry and progression. (B) In non-proliferative cells such as adult β-cells, cell cycle activators reside in the cytoplasm and inhibitors can be found in the nucleus. (C) In proliferative β-cells, the cell cycle activators shuttle to the nucleus and (D) In non-proliferative β-cells subjected to the same stimuli as the cells in panel C, both activators and inhibitors shuttle to the nucleus, maintaining the quiescent state. Figure taken from Wang et al. (2015).

24 One problem with studying pancreatic tissue, or attempting to regrow damaged pancreatic tissue is that it is a largely non-proliferative organ. This quiescence is due to the cell cycle regulators, which differ between pancreatic cells and more proliferative cell types. In actively proliferating cells, generally, CDK4/6 binds to cyclinD causing phosphorylation of retinoblastoma (Rb) which leads to transcription and promotion of entry to the G1 stage of the cell cycle. CDK4 and CDK6 both act in a similar capacity, showing redundancy in most cells, similarly cyclinD1, 2 and 3 can partner

CDK4/6. However, in β-cells specifically, only CDK6 is active, partnered to cyclinD3 (Fiaschi-Taesch et al., 2013). On the whole, β-cells are non-proliferative, with cell cycle entry prevented by p16INK4a, p57KIP2 and p27KIP2. On the occasions when β-cells are induced to duplicate these CKIs move to the cytoplasm allowing the nuclear cell cycle promoters to function. This nuclear/cytoplasmic shuttling is a major method for cell cycle regulation. Indeed, in the majority of β-cells, most of the time, the only cell cycle molecules in the nucleus are Rb and p57KIP2 which is why they are considered a non-proliferative cell type (Wang et al., 2015). The different molecules involved in regulation, both in proliferative and non-proliferative pancreatic tissues are shown in Figure 1.4.

1.2.5 Signalling and markers

PDX1

PDX1 is a transcription factor which is one of the key drivers of the pancreatic lineage. Whilst all pancreatic developmental factors are also used for specifying other lineages, PDX1 is the most specific for the pancreatic lineage. The importance of PDX1 in islet development is evidenced by the fact that

Pdx1 null mice can only produce immature endocrine cells, with a mature pancreas not fully develop- ing (Ahlgren et al., 1996). This is likely because PDX1 is a master transcription factor, with the gene having cis-regulatory binding sites for HNF1α, FOXA2, PAX6, HNF6, MAFA and PTF1α (Gerrish et al., 2001, Lee et al., 2002, Samaras et al., 2002, 2003, Jacquemin et al., 2003, Wiebe et al., 2007).

In turn, PDX1 drives transcription of other developmental factors, and of insulin itself (Iype et al.,

2005). Forced PDX1 expression is sufficient for α- to β-cell transdifferentiation and in anti-apoptosis signalling in β-cells (Johnson et al., 2006). Together these experiments strongly implicate PDX1 in development of the endocrine pancreas, and specifically β-cells and also in the maintenance of β-cells.

SOX9

SOX9 is a transcription factor used in many cells throughout development, in the pancreas it is used in multipotent stem cells during pancreas formation as well as in ductal cells in the mature pancreas

(Furuyama et al., 2011, Seymour, 2013, Kawaguchi, 2013). During development, or pancreas differ- entiation from stem cells, SOX9 is essential for stem/progenitor cell maintenance, where inactivation

25 causes progenitor apoptosis and decreased proliferation (Seymour et al., 2007).

GATA4/6

The GATA factors are also used in numerous developmental processes. In the pancreas they act as a switch for specification of endocrine cells, rather than other lineages. GATA6 marks endocrine cells and GATA4 exocrine (Ketola et al., 2004). Null mice for either Gata4 or Gata6 show early lethality due to hyperglycaemia, but a single allele is sufficient for a normal pancreas (Carrasco et al., 2012) so further specifics of their functions are still relatively unknown.

NEUROD1, NGN3 and the NOTCH pathway

The NOTCH pathway is heavily involved in the development of endocrine progenitors, during the secondary transition, and functions downstream through main NOTCH target HES1, followed by

NGN3 and NEUROD1. The NOTCH ligands are involved with lateral inhibition, preventing neigh- bouring NGN3 cells both becoming endocrine cells. The NOTCH pathway activates HES1, which represses NGN3 expression. Knockout mice of Hes1, downstream of the NOTCH ligands, show early differentiation and loss of the progenitor pool (Jensen et al., 2000), and overexpression inhibits differentiation of the progenitors and maintenance of the progenitor pool (Murtaugh et al., 2003).

Similarly premature Ngn3 expression causes over differentiation to endocrine cells, particularly α- cells (Schwitzgebel et al., 2000) and Ngn3 knock out mice contain no endocrine cells (Gradwohl et al.,

2000). These data shown Ngn3 to be essential during rodent pancreatic development, and it is similar in humans, but it is key that NGN3 is only expressed for a short window during development (Salis- bury et al., 2014). After this, NGN3 cells produce SNAI2 in order to undergo epithelial-mesenchymal transition, to escape the trunk and make islets (Rukstalis and Habener, 2007). Together these show that expression and correct timing of NOTCH and its downstream targets are essential for correct endocrine development, or differentiation.

ISL1

Islet1 (ISL1), as the name suggests, is essential for islet cell formation. Isl1 knock out mice do not contain islet cells (Ahlgren et al., 1997). It is present in Ngn3+ precursors, and is thought to trigger their proliferation and differentiation (Du et al., 2009). Isl1 is also present in the mesenchyme of mice (Ahlgren et al., 1997) and in MSCs derived from the human pancreas (Eberhardt et al., 2006).

One function of ISL1 is to enhance insulin gene expression (Zhang et al., 2009); other functions,

26 including in MSCs are still being investigated.

PAX4 and ARX

The divide between β-cells and α-cells is mainly decided by a switch of PAX4 or ARX expression.

Endocrine precursors are remarkably plastic, if 99% of β-cells are destroyed α-cells are able to con- vert to β-cells via an insulin/glucagon double positive intermediate (Thorel et al., 2010). In isolated endocrine precursors it is possible to determine the fate to either α- or β-cells using only the transcrip- tion factors Arx or Pax4 respectively. Despite this plasticity, there is evidence that differentiation is affected or predetermined by methylation patterns. In a β-cell, if Dnmt1 is ablated preventing methylation patterns being maintained during division, during the next round of mitosis the cell will become an α-cell. This appears to happen via a mechanism in which Arx is derepressed due to the loss of CpG methylation (Collombat et al., 2003). Conversely forced expression of PAX4 markedly enhances differentiation to β-cells rather than α-cells (Liew et al., 2008). Finally, ex vivo expanded

β-cells can be induced to re-differentiate to β-cells by inhibition of ARX (Friedman-Mazursky et al.,

2016).

NES

The role of Nestin (NES) in the pancreas has been the subject of much debate. NES is a cytoskeletal protein, which has been suggested as putative marker of pancreatic stem cells (Hunziker and Stein,

2000). The evidence on this is mixed though, as NES is seen in multiple cells (Street et al., 2004) and is not required for β-cell generation during normal development (Piper et al., 2002). NES is also seen in MSCs derived from the pancreas (Eberhardt et al., 2006), it is not clear if these MSCs are derived from NES postive cells within the pancreas though.

1.3 Congenital hyperinsulinism (CHI)

1.3.1 Symptoms and causes

CHI is an inherited disorder, characterised by high levels of disregulated insulin secretion (Dunne et al., 2004). It affects neonatal patients, being a genetic disorder. There are three forms of CHI - focal, diffuse and atypical. All three forms show the main phenotype of excessive insulin secretion, but have histological and genetic differences, the histological differences are shown in Figure 1.5.

The most common cause of CHI is defects in the KATP channels, which cause constitutive efflux of

27 potassium ions and influx of calcium ions causing a release of insulin granules, shown schematically in Figure 1.6.

The inappropriately high levels of insulin cause numerous problems, resulting in most patients being hospitalised within a few days of being born (Hussain and Aynsley-Green, 2004). The insulin results in severely low blood glucose, glucose is essential for metabolism in every cell. This results in lethargy, seizures, cognitive problems, feeding problems or coma (De Leon and Stanley, 2007,

Hussain, 2008). If blood glucose is not swiftly normalised it will lead to degradation of body tissues, including the brain and eventually death (Hussain, 2008). This means that lifelong management of the condition is essential.

Focal CHI is named for a focal lesion, comprising almost exclusively β-cells. It is due to a reces- sive heterozygous mutation, combined with loss of heterozygosity in the lesion alone. The mutation is paternally inherited, and the maternal allele lost in a sporadic fashion, resulting in expression changes in several maternally imprinted genes (Giurgea et al., 2006). ABCC8, encoding the SUR1 subunit of the KATP channel, is one of the genes deleted which causes the loss of regulation of insulin secretion. Other genes are affected, including p57Kip2 (James et al., 2009), which may lead to the observed increase in islet-cell proliferation in the lesion (Kassem et al., 2000).

Diffuse CHI, unlike focal CHI, affects every β-cell in the pancreas. It is caused by a variety of gene mutations, most commonly ABCC8, KCNJ11, GCK and HNF4α (Dunne et al., 1997, Glaser et al.,

2000). An homozygous mutation, or two different inactivation mutations on each allele inactivate the insulin secretion pathway in every β-cell. With diffuse CHI being predominately caused by homozy- gous mutations, it is relatively rare in some populations such as the Netherlands (1 in 50,000), but common in regions with high rates of consanguinity such as Saudi Arabia (1 in 2500) (Bruining, 1990).

Atypical CHI is markedly different from focal and diffuse. Where focal and diffuse CHI present shortly after birth, atypical has a later onset (Arnoux et al., 2011). The genetic background of focal and diffuse CHI is well characterised, whereas for atypical CHI it is not (Arnoux et al., 2011). It may be caused by a wide range of mutations in different patients which would make linkage studies impossible, as pancreatic development is controlled by a myriad of genes which could be causing the disease (Pan and Wright, 2011). One factor that is known to be associated with atypical CHI is altered hexokinase expression, but the role of this has not been fully worked out yet (Henquin et al.,

2013).

28 Figure 1.5: Histological differences between focal and diffuse CHI In diffuse CHI every β-cell is affected, in focal CHI only one area of β-cells is affected. Adapted from (Dunne et al., 2004).

Figure 1.6: Insulin secretion in CHI Despite low glucose levels and therefore low intracellular ATP levels, KATP channels cannot open, β-cells remain depolarised with constant calcium influx and therefore insulin is constitutively secreted (Koster et al., 2005, Braun et al., 2008, Dunne et al., 2004).

29 1.3.2 Hyperproliferation

Aside from altered insulin secretion, CHI also features increased proliferation in the pancreas, com- pared to healthy age-matched pancreas. In the lesion of focal CHI this is caused by loss of p57Kip2

(Fournet et al., 2001, Kassem et al., 2001, James et al., 2009). In diffuse CHI, increased nuclear

CDK6 and p27Kip1 is also associated with increased proliferation although the ultimate cause is un- known (Salisbury et al., 2015). Unpublished results from our lab also show increased proliferation in atypical CHI and outside of the lesion in focal CHI. As this is universal to all forms, it may be that the increased proliferation is due to the high levels of insulin. In this context insulin could be acting as a growth factor (Denley et al., 2004, Belfiore and Malaguarnera, 2011, Ish-Shalom et al., 1997), causing a cycle of proliferation leading to more insulin-secreting cells, leading to more proliferation.

1.3.3 Treatment

Due to the variability in causes and presentation, treatment of CHI can be very different. First line treatments for CHI are pharmaceutical, with immediate glucose and glucagon infusions (Moens et al.,

2002) to stabilise blood glucose and prevent permanent damage. More permanent treatments for ongoing management of the condition include (Glaser et al., 1989) and (Touati et al., 1998), which in some cases prevent insulin secretion. Where pharmaceutical treatments fail, for example, when the precise mutation renders the drugs non-functional, the only remaining option is surgery (Arnoux et al., 2010). For atypical and diffuse CHI this involves a subtotal

(Dunne et al., 2004, Hussain, 2008), but this can lead to diabetes later in life (Leibowitz et al., 1995) and so is a last resort. In focal CHI, (18)F-L-dopa positron emission tomography allows identification of the lesion, allowing precise surgical excision (Otonkoski et al., 2006). This makes surgery a good option for focal CHI, with it being a first line treatment at some centres (Hussain, 2008), although surgery on neonates is still not an ideal situation. The excessive proliferation in CHI can lead to regrowth of the pancreas following surgery in some cases. When this occurs repeated surgery is required to further resect the pancreas (Jack et al., 2003, Meissner et al., 2003). Due to these problems with medical management, novel treatments are desired (Meissner et al., 2003).

1.3.4 Experimental models

CHI research primarily relies on looking at post-operative pancreatic tissue. This has the advantage of allowing study of authentic diseased tissue. On the other hand, because CHI is a rare disease and surgery is last resort, fresh tissue is limited. This restricts the number of experiments which can be performed. Studies on fixed tissue are more readily achievable but only provide a snapshot, not a dynamic system which can be probed. To combat these difficulties, other models have been

30 generated. One option is transfection of constitutively active genes containing the CHI causing mutations, into HEK293 cells which do not normally express KATP channels (Arya et al., 2014). This allows study of the effect of the mutations on ion flux, using radioactive rubidium, but no way to understand the effects on insulin secretion. One other option for studying CHI is using rodent models. Generation of an ABCC8 mutant knock in mouse has allowed study of the effect on channels and insulin secretion (Shimomura et al., 2013). However, the differences between human and rodent islet structure (Cabrera et al., 2006, Balboa and Otonkoski, 2015), discussed in section 1.2.1, may mean that rodent experiments cannot be extrapolated to humans. Further, there are differences between cell cycle in humans and rodents (Kulkarni et al., 2012, Fiaschi-Taesch et al., 2009), so it is likely that hyperproliferation in CHI cannot be accurately studied in these rodent models. As such, novel, human models of CHI to study channel ion flux and insulin secretion are needed.

1.4 Diabetes

Diabetes is a chronic disease of the pancreas which leads to life-long therapeutic treatment and lifestyle management. There are 2 major forms of the disease, known as Type 1 and Type 2 dia- betes; is further subdivided into Type 1A and Type 1B. Type 1A diabetes is the most common form and is an autoimmune disease which results in an absolute insulin deficiency due to β-cell destruction (Atkinson et al., 2013). This β-cell destruction means that as yet Type

1A diabetes is not curable and so is the focus of islet replacement research. Type 1B diabetes is non-immune mediated but also results in a severe insulin deficiency, the causes are largely unknown

(Atkinson and Eisenbarth, 2001). is usually a result of lifestyle and diet and is char- acterised by (insulin produced in the body does not function as well as it should) and a gradual progression to insulin deficiency (Kahn et al., 2014). Whilst Type 1 diabetes requires daily insulin supplementation for survival, in early and well managed cases Type 2 diabetes can be treated by a change in diet and a reduction in a weight - over time this can progress to needing insulin supplementation though and the disease symptoms converge.

1.4.1 Epidemiology

Diabetes is one of the most common chronic diseases; in the UK the rate is estimated to be around 1 in 16 people having the disease (Diabetes UK, 2015). There are approximately 2.9 million diagnosed cases of diabetes in England alone (Diabetes UK, 2015), with the rate increasing to 1 in 5 in adults over 85 years old, due to the disease being permanent.

31 Type 2 diabetes is primarily a disease of adults due to the nature of its development from lifestyle and diet - obesity is the major risk factor (Hu et al., 2001, Mokdad et al., 2003, CDC, 2004). Type

2 accounts for around 85% of diabetes cases in the UK and 90% of adult cases of diabetes. Type

2 diabetes is most common worldwide in more developed countries where obesity is prevalent but within this there are separate ethnic predispositions - non-whites have a lower threshold for obesity related risk of diabetes (Ntuk et al., 2014).

Type 1 diabetes, being an autoimmune disease, is usually diagnosed in children. In the UK 18-20 children per 100,000 are diagnosed with type 1 diabetes each year (Daneman, 2006). 50-60% of Type

1 diabetes patients are under 18 years old at diagnosis (Daneman, 2006). Geographical variance in this disease is also high for incidence rates. The highest incidences worldwide are in Finland (where

48-49 members of the population are diagnosed per 100,00 each year), Sardinia, Kuwait and Puerto

Rico (Atkinson and Eisenbarth, 2001). The disease also shows strong familial links alongside age and ethnicity - the risk of developing the disease is 15 times higher for siblings of patients than in the general population (Redondo et al., 2001). Despite the disease usually being diagnosed in children, incidence of patients is highest in adults due to the disease being highly treatable, but not curable.

1.4.2 Pathogenesis

Type 1 diabetes as stated above is a disease caused by β-cell destruction which causes an absolute insulin deficiency. The β-cell destruction is as a result of auto immune attacks, the body incorrectly produces antibodies against its own islets. Initially this usually includes anti-insulin or glutamic acid decarbyoxylase antibody (GADA). GADA is seen in 5-30% of type 2 diabetes cases which are there- fore thought to have been incorrectly diagnosed (Atkinson and Eisenbarth, 2001). Other antibodies seen include protein tyrosine phosphatase IA2 auto-antibody (IA-2AA) and ICA512A (Atkinson and

Eisenbarth, 2001), where the presence of 2 or more of these antibodies give a 90% predictive risk of developing the disease (Devendra et al., 2004). The antibodies alone cannot cause the disease , but are seen in a latent period before the disease becomes symptomatic known as the prodrome. This period can last years before the disease graduates to the next stage, wherein more antibodies appear and β-cell destruction accelerates and becomes measurable. At this stage the first phase insulin response in which insulin is rapidly released in response to increased blood glucose levels is decreased and eventually lost as a result of β-cell mass falling below a critical threshold.

β-cell destruction continues and insulin secretion continues to fall until there is a full deficiency and the patient will have overt clinical diabetes (Daneman, 2006). triggers the β-cell destruction which causes clinical presentation due to cytotoxic T cells expressing the Fas ligand. Upon binding

32 Fas forms the death-inducing signalling complex and causes apoptosis. This Fas-mediated apoptosis is thought to account for the vast majority of β-cell destruction in type 1 diabetes (Devendra et al.,

2004).

Type 2 diabetes is largely due to lifestyle factors such as obesity, wherein adiposity of the body cavities leads to insulin resistance (Cnop et al., 2002). Insulin resistance - where the cells of the body are less sensitive to insulin, combines with β-cell dysfunction to cause the overt clinical symptoms.

Initially β-cells may still secrete insulin normally but insufficiency develops (Stancakova et al., 2009) in a feedback loop with resistance (Kahn et al., 2014), this can eventually lead to the β-cells ceasing to function (Kahn et al., 2014) and being destroyed (Butler et al., 2003).

1.4.3 Treatment

For type 2 diabetes, the first line treatment is lifestyle changes, to reduce the causes - normally reducing weight, increasing exercise and decreasing sugar intake (Kahn et al., 2014). This is usually combined with the oral drug (National Institute for Health and Care Excellence, 2015,

Maruthur et al., 2016). Metformin acts to reduce glucose released by the liver and increasing the sensitivity of cells to what insulin is being produced (Viollet et al., 2012). Another common drug treatment is the sulphonylureas which increase insulin production by remaining β-cells by acting on

KATP channels (Proks et al., 2002). All treatments for type 2 diabetes involve daily management and in many cases do not prevent progression of the disease. The long term complications due to the drugs themselves or the diabetes (which is still biologically present, but masked by the drugs) are not clear (Maruthur et al., 2016).

The treatment for type 1 diabetes is injection of insulin, which must be performed daily for life and requires careful monitoring. A combination of short and long acting insulin/insulin analogues are used to replace the insulin that the destroyed β-cells cannot produce. Typically a long acting insulin is used across the day, with rapid acting insulin taken with each meal (Atkinson and Eisen- barth, 2001). This is adjusted to allow for meal size and activity levels, with genetically engineered analogues with varying durations used to attempt to replicate the steady levels of insulin normally found in the body. Blood glucose levels are measured regularly to ensure correct dosage is achieved, but euglycaemia can be difficult to achieve and glucose/insulin spikes are common place (Daneman,

2006).

Due to the problems associated with insulin treatment, permanent cures and prevention of disease development have been investigated. Over 100 treatments prevented type 1 diabetes developing in

33 the non-obese diabetic (NOD) mouse model or in rats treated with but none of these worked in humans suggesting that this is a poor model (Devendra et al., 2004, Gale, 2004). The insufficiency of prevention or treatment belies the need for a true cure for diabetes. The closest to a cure that has been achieved is islet transplants, where cadaveric islets are transplanted into the hepatic portal vein (Shapiro et al., 2000). Originally this was attempted using immunosuppresion with glucocorticoids but this had a very low success rate, only 12.4% of transplants functioned for more than 1 week as glucocorticoids are diabetogenic. The Edmonton Protocol revolutionised this using immune suppression by antibodies - daclizumab, sirolimus and tacrolimus which showed no immune rejection and long term insulin independence. Multiple transplants were required to achieve insulin independence (2 on average, but 3 was needed in some cases), but all patients trialled did not meet clinical diagnostic criteria for diabetes 6 months on, due to adequate production of insulin and measurable C-peptide in response to oral glucose challenges. The long-term follow up suggested this was not a permanent cure but would need repeating; with refinement islet transplants could offer a permanent cure (Ryan et al., 2005). There are not enough donor sources of islets for the operation to become commonplace when combined with the difficulties, and so stem cell therapies may be put to use in this way once developed.

1.5 Stem Cells

1.5.1 Pancreatic stem cells

Stem cells are present in most organs throughout the body and are responsible for generation of the organ, as well as post-developmental growth and repair. It has therefore been considered that there are likely to be pancreatic stem cells. The evidence regarding pancreatic stem cells is, however, mixed as I will discuss. There are also differences between humans and rodents which complicates matters.

Early evidence supporting the existence of pancreatic stem cells was that performing at 95% pancreatectomy, pancreatic duct ligation (PDL) or selective β-cell ablation in rodents would lead to regeneration of the pancreas (Li et al., 2010). Attempts to discern the source of the regeneration have been mixed. Some regeneration is due to the β-cell duplication, and some due to ductal cells, as discussed in section 1.2.3. PDL has also been shown to cause an increase in nestin within the pancreas (Peters et al., 2004), which as discussed earlier (1.2.4.6), may mark stem cells including

MSCs, but is not specific (Street et al., 2004). These reports support the existence of pancreatic stem cells in rodents, but cannot of course be replicated in humans.

34 A separate phenomenon by which pancreatic mass can increase, is during pregnancy (Sorenson and Brelje, 1997). This occurs in both humans and rodents. Experimental lineage tracing, and histological studies have suggested this is due to β-cell duplication in rodents, meaning stem cells, should they exist may only respond to certain extreme stimuli.

As it has proved challenging to identify, or even demonstrate the existence of pancreatic stem cells, attempts have been made to culture and purify them. The post-developmental pancreas is not proliferative, so any proliferative cells in culture may represent progenitor cells, with the exception of fibroblasts. Fibroblasts can be cultured from most tissues and proliferate in culture for a few passages. In direct culture with serum-containing media, endocrine, exocrine and ductal tissue all produce MSCs. These will be discussed further in section 1.5.2 but express nestin, linking to the observation of increased nestin seen following PDL. Variations on this culture have produced differ- ent cells. The addition of a ROCK inhibitor and TGFβ inhibitor generates epithelial rather than mesenchymal cells, but with a much lower capacity for proliferation (Lima et al., 2013). Complex ductal structures can also be induced to proliferate and differentiate (Huch et al., 2013, Corritore et al., 2014). These are of interest as it has been hypothesised that stem cells may reside in the ducts, based on regeneration of the pancreas from ductal cells (Li et al., 2010). However, these ductal cultures have not been well studied.

It is unclear from the evidence generated to date if pancreatic stem cells exist, or are capable of pancreas regeneration. The two potential sources are ducts and MSCs, with the most readily proliferative cells being the MSCs.

1.5.2 Mesenchymal stem cells

Sources and characteristics

MSCs represent a diverse set of cells from throughout the body, with largely overlapping character- istics. Initially, and most commonly, MSCs are derived from the bone marrow, and bone marrow

MSCs (bmMSCs) have been investigated as a source for diabetes treatment (Li and Ikehara, 2013).

They have since been derived from adipose tissue, the pancreas, Wharton’s jelly, umbilical cord, periosteum and dental pulp. MSCs from each of these sources have the same defining characteristics, with some variation based on tissue of origin.

The characteristics to define MSCs, as set down by The International Society for Cellular Ther-

35 Figure 1.7: Sources of MSCs pMSCs have been derived from exocrine cells and β-cells, confirmed by lineage tracing (Lima et al., 2013, Russ et al., 2008) but may also be derived from duct cells (Corritore et al., 2014) or CD90/CD105 tissue resident cells (Carlotti et al., 2010). apy, are plastic adherence, a panel of cell surface markers and the ability to differentiate to three mesodermal lineages (Dominici et al., 2006). Differentiation and the ability to serially passage the cells, set MSCs aside from fibroblasts. Fibroblasts also show plastic adherence and may contaminate cell cultures for the first few passages. Differentiation of fibroblasts has been reported (Brohem et al., 2013) but the evidence to suggest that they were not in fact MSCs is unclear. The cell surface markers required are CD73, CD90 and CD195, with at least 95% of cells being positive for these markers. Other markers may also be used to support the identification. Cells should also be lacking any of CD14, CD19, CD34 and CD45, with less than 2% of cells expressing these, to rule out the presence of contaminating haematopoeitic stem cells. This is more of a problem when deriving MSCs from the bone marrow, which is what these characteristics were developed for - there is no reason to expect haematopoeitic stem cells to be present outside of the bone marrow. The functional assay, of differentiation to fat, bone and cartilage shows MSCs are multipotent. As with the cell surface markers, this characteristic was determined on bmMSCs. It has been shown that pancreatic MSCs

(pMSCs) demonstrate inefficient differentiation to adipocytes, whilst adipose-derived MSCs show much higher efficiency. Other differences between MSCs from various sources include the prolifer- ation rates, duration of proliferation in culture, and efficiency of differentiation to other lineages, including β-cells.

36 Figure 1.8: EMT in pMSCs Reversible EMT in pMSCs may be responsible for phenotypes in culture; serum addition and/or monolayer culture yields mesenchymal cells, whilst serum withdrawal or non-adherent culture yields epithelial-like cells (Dalvi et al., 2009, Davani et al., 2009).

PMSCs have been derived from endocrine, exocrine and ductal cells. It is nigh impossible to separate these different tissues, so pMSCs are grown from enriched, but ultimately mixed cell pop- ulations. It has been suggested that pMSCs are tissue-resident such as the observed CD90/CD105 positive cells inside islets (Carlotti et al., 2010), or that they are the product of de-differentiation via epithelial-mesenchymal transition (EMT). Lineage tracing has shown MSCs can be derived from

β-cells, but are not solely (Russ et al., 2008, 2009, Joglekar et al., 2009, Sordi et al., 2010, Lima et al., 2013). From a mixed pancreas digest, it is therefore likely that MSCs from multiple sources will be obtained, as shown in Figure 1.5. It has been suggested that MSCs, generated by EMT are an artifact of cell culture conditions (Seeberger et al., 2008). What is unclear is if that is a problem.

Particularly for in vitro modelling, so long as the terminal cells are functional, the manner in which they were derived may not matter. Further, the process seems to be at least partially reversible, with the formation of islet-like clusters (Dalvi et al., 2009). These cell clusters are unable to graft in mice though (Davani et al., 2009). The authors of the study in which this was assessed, suggested that the cues in the mouse blood may again be switching the cell phenotype back (Davani et al.,

2009), shown in Figure 1.7.

Whilst the evidence is mixed, these experiments most strongly suggest that the MSC, or MSC-like cells produced from the pancreas are a result of de-differentiation of mature cells and are an effect of the in vitro culture. Cells containing a selection of the same markers as these MSCs are present in the pancreas in vivo as discussed further in Section 1.5.2, but this is not definitive proof of the existence of MSCs in vivo as further concurrent markers would be needed, as well as the ability to isolate these cells. The production of ductal structures mentioned above shows better evidence for a stem cell, however, in vitro ductal structures are produced from mixed progenitor populations rather than isolated single cells. A lack of unique cell surface markers for pancreatic stem cells present during development has hindered identification of any remaining post-natal stem cells, but with the

37 current evidence it seems unlikely that there are stem cells present in the adult pancreas. Rather the balance of evidence suggests that cells with the potential to re-differentiate can be produced in vitro by de-differentiation of mature cells.

Differentiation

Differentiation of MSCs to β-cells or β-like cells has been documented in MSCs from multiple sources, using varied protocols, and with varied outcomes. A cross-section of these are shown in Table 1.1-2.

Whilst growth factors have been used to reprogram bone marrow, adipose, umbilical cord and Whar- ton’s jelly MSCs to β-cells, groups have reported that serum withdrawal alone induces formation of hormone-positive clusters of cells - ‘islet-like clusters’. The use of additional growth factors seems to enhance whether GSIS is seen following differentiation, and many protocols use the 3D culture.

These cell clusters are not unique to pMSCs, with most, if not all MSCs forming dense spheroids in low adherence culture, reviewed in Cesarz and Tamama (2015). As the authors discuss, spheroids may be autofluorescent and are used to aid differentiation of bmMSCs to bone and cartilage, so there are some question marks over the authenticity and accuracy of pancreatic islet-like cluster results.

1.5.3 Pluripotent stem cells

Characteristics

Whereas MSCs are multipotent, able to differentiate to several lineages, ESCs and iPSCs are pluripo- tent. Pluripotent cells are more ‘basal’ and able to generate all body tissues.

ESCs are derived from the inner cell mass of blastocysts (Thomson et al., 1998), an early stage of embryonic development. They proliferate in culture for a prolonged period (>100 passages) and generate cells of every germ layer. This means that when these cells are transplanted into immuno- compromised mice, they form teratomas. Teratomas are tumours containing multiple cell types, randomly differentiated, and may include hair and full teeth. ESCs are the gold standard pluripo- tent cells, as they are derived from the earliest stages of human development.

Pluripotent stem cells can also be derived from any somatic cell, these are iPSCs. In the land- mark study, by simply transducing cells with the pluripotency factors OCT4, NANOG, KLF4 and c-Myc, they were reprogrammed to a state similar to ESCs (Takahashi et al., 2007) There are some differences between ESCs and iPSCs but the defining characteristic of pluripotency is the same. The differences are primarily epigenetic, with incomplete reprogramming occurring in iPSCs, potentially

38 Cell type Number Number With Factors Insulin Reference of stages of days non- monolayer stage Human pMSCs (duct) 4 12 Yes 1% FBS, 10 mM nic, 10 nM Ex-4, 10 PCR only, immunofluo- Seeberger et al. P2-6 ng/ml TGFβ1 rescence negative (2006) Human bmMSCs & pla- 2 12 No 1% N2, 1% B27, 25 ng/ml bFGF, 10 mM PCR only Battula et al. cental MSCs nic, gelatin (2007) Human pMSCs (islet) 1 7-21 Yes 1% ITS, 1% BSA 8.8 µU/ml in 16.7 mM Gallo et al. glucose, 13.2 µU/ml in (2007) 3.3 mM glucose Human umbilical cord, 3 <7 No 1 mM NaB, 50 ng/ml activin a, 0.2- 0.25-0.37 µU/ml in 15 Bhandari et al. Wharton’s jelly & amni- 1% BSA, 50 ng/ml KGF, custom culture mM glucose (0.0-0.25 (2011) otic MSCs media µU/ml for controls) Human bmMSCs & pM- 3 21 Yes 5% platelet lysate, 10 µM retinoic 30 pmol for pMSCs, 11 Zanini et al. SCs (islet) acid,10 µg/ml activin a, 200 µg/ml Glp- pmol bmMSCs in 25 (2011) 39 1, 20 ng/ml EGF, 10 ng/ml FGF, 10 mM glucose µg/ml Btc, 10 mM nic Rat bmMSCs 6 12 No 2-15% FBS, 100 ng/ml activin a, 10 µM >1.5 ng/ml in 25 mM Wang et al. retinoic acid,10 ng/ml bFGF, 10 mM glucose, <0.5 ng/ml in 3 (2012) nic, 10 nM Ex-4, matrigel mM glucose Human bmMSCs, adM- 1 14-21 No 10 mM nic, 2 mM activin a, 10 nM Ex-4, 4-12 ng/mg in 25 mM Kim et al. SCs, Wharton’s jelly & 100 pM HGF, 2% B27, 2% N2 glucose, 3-4 in 3 mM (2012) periosteum glucose, insulin/total protein Mouse pMSCs (islet) 3 10 Yes 1% BSA, 0.3-3 mM taurine, 10 mM nic, 498 µIU/ml in 16.5 mM Gopurappilly 10 mM Glp-1 glucose et al. (2013)

Table 1.1: Cross-section of MSC to β-cell differentiation protocols Abbreviations used in this table: P - passage, FBS - foetal bovine serum, ITS - insulin/transferrin/selenium, BSA - bovine serum albumin, bFGF - basic fibroblast growth factor, HGF - hepatocyte growth factor, GLP - glucagon like peptide, Nic - nicotinamide, NaB - sodium butyrate, Ex-4 - Exendin-4, NEAA - non-essential amino acids, IBMX - 3-isobutyl-1-methylxanthine, Btc - betacellulin, EGF - epidermal growth factor, TSA - trichostatin-A Cell type Number Number With Factors Insulin Reference of stages of days non- monolayer stage Human adMSCs 4 17 No 5-10 ng/ml activin a, 50 ng/ml Wnt3a, Immunocytochemistry Li et al. (2012) 10 µM retinoic acid, 10 ng/ml DKK1, 20 only (Insulin, C- ng/ml EGF, 60 ng/ml bFGF, 1x NEAA, peptide, PDX1) 2% B27, 10 ng/ml Ex-4, 10 mM Nic Human bmMSCs 3 21 Yes 1% BSA, 1% ITS, 2 nM Activin A, 0.1-5 71.6 µU/ml in 25 mM Jafarian et al. mM taurine, 1 mM NaB, 100 pM HGF, glucose, 19.4 µU/ml in (2014) 10-100 nM Ex-4, 1x NEAA, 2% B27, 2% 5.5 mM glucose (both N2 with IBMX) Human bmMSCs 1 15 No 10% FBS, 10 ng/ml conophylline, 1 nM 0.005/0.017 ng/µg pro- Gabr et al.

40 Btc tein/h in 5.5/25 mM (2014) glucose Human bmMSCs 2 10 No 55 nM TSA, 10 nM GLP-1 0.006/0.019 ng/µg pro- Gabr et al. tein/h in 5.5/25 mM (2014) glucose Human bmMSCs 3 18 No 0.5 mM β-mercaptoethanol, 1% NEAA, 0.006/0.019 ng/µg pro- Gabr et al. 20 ng/ml bFGF, 20 ng/ml EGF, 2% B27, tein/h in 5.5/25 mM (2014) 2 mM L-glutamine, 10 ng/ml btc, 10 glucose ng/ml activin a, 10 mM nic

Table 1.2: Cross-section of MSC to β-cell differentiation protocols Abbreviations used in this table: P - passage, FBS - foetal bovine serum, ITS - insulin/transferrin/selenium, BSA - bovine serum albumin, bFGF - basic fibroblast growth factor, HGF - hepatocyte growth factor, GLP - glucagon like peptide, Nic - nicotinamide, NaB - sodium butyrate, Ex-4 - Exendin-4, NEAA - non-essential amino acids, IBMX - 3-isobutyl-1-methylxanthine, Btc - betacellulin, EGF - epidermal growth factor, TSA - trichostatin-A in a lineage specific fashion (Kim et al., 2011, Bar-Nur et al., 2011, Beagan et al., 2016). This may lead to a bias in differentiation of iPSCs to certain cells which is not exhibited by ESCs. Recently, it has been found that simply erasing epigenetic marks in mouse epiblast cells will also induce repro- gramming to a pluripotent state (Zhang et al., 2016). The main advantage of iPSCs over ESCs is that they can be created in a patient-specific fashion, for specific diseases (Yamanaka, 2012). There is considerable variation between iPSC lines (Koyanagi-Aoi et al., 2013, Rouhani et al., 2014), even from the same healthy or diseased background, representing the natural genetic variability in the population. To combat this, large banks are being generated of both healthy iPSCs and those from patients with monogenic disorders (Moran, 2013). It is hoped that banks, such as HipSci (Leha et al.,

2016), will enable development of more robust and repeatable differentiation protocols for cells of different lineages (Moran, 2013).

1.5.4 Generation of iPSCs

Initial generation of iPSCs was by retroviral transduction with lentiviruses, using the ‘Yamanaka factors’ (Takahashi et al., 2007). These factors as mentioned previously are OCT4, SOX2, KLF4 and c-Myc. Whilst OCT4 and SOX2 are also markers of pluripotency, KLF4 aids with MET and formation of epithelial colonies, and acts in complex with OCT4, SOX2 or NANOG (Kim et al.,

2008). C-MYC is used to aid long term growth of the cells. Alternatively LIN28 has been used instead of c-Myc (Yu et al., 2007), which is not functionally similar but rather is involved in pro- moting translation of OCT4 mRNA. OCT4 and SOX2 are the key regulators of pluripotency, acting in feed-forward loops wherein OCT4 and SOX2 together bind to an OCT4 enhancer (Chew et al.,

2005) as well as transcriptionally regulating NANOG, the third primary pluripotency marker (Rodda et al., 2005). OCT4, in concert with the other factors further causes specific gene activation (Sharov et al., 2008) and prevents differentiation by gene silencing via polycomb complexes and miRNAs

(Kashyap et al., 2009).

The retroviruses used in the initial iPSC induction are integrative viruses, and in the original study the authors noted at least three retroviral integration sites for each of the four factors. De- pending on which loci the viruses integrate at (Stocking et al., 1993), there can be impacts upon the cells normal function or induction of a cancerous phenotype via insertional mutagenesis (Li and

Dahiya, 2002, Hacein-Bey-Abina et al., 2003). Further, the viruses can reactivate causing problems such as cancers (Okita et al., 2007). Due to these issues, non-integrative transduction methods have been developed. One such methods is transduction via Sendai virus (Fusaki et al., 2009, Seki et al., 2010) which is passively cleared from the cells over time, but remains present sufficiently

41 long enough to induce reprogramming. A downside to this method is that any imbalance of factors between the different vectors can affect reprogramming and so clones must be fully assessed (Carey et al., 2011). An alternative method which is fully virus free is to reprogram by mRNA transduction

(Warren et al., 2010). This method guarantees no virus will be found in cells at any stage which is optimum when cells may be used in vivo but is labour intensive, requiring multiple rounds of transfection. Other methods have been investigated such as directly introducing the proteins, which are reviewed by Robinton and Daley (2012) but these methods have very low efficiency, with normal reprogramming already being at most around 1% efficient. Efficiency of reprogramming is generally low due to barriers, such as the difficulty in full erasure of histone modifications associated with the differentiated cell phenotype, reorganisation of chromatin and activation of the pluripotency asso- ciated genes, with numerous examples reviewed by Ebrahimi (2015). Whilst only one high quality iPSC line is required per patient from the selected clones, efficiency of reprogramming to ensure fully reprogrammed iPSCs can be boosted with modulators of epigenetics. These modulators enhance the reorganisation of the genome, an example of which is valproic acid, a histone deacetylase inhibitor which eliminates the need for c-Myc (Huangfu et al., 2008a,b). This is also desirable for in vivo applications, or applications wherein a normal cell cycle is required. Other factors including GLIS1

(Maekawa et al., 2011), SALL4 (Tsubooka et al., 2009) and NANOG (Silva et al., 2009) have also been shown to enhance reprogramming efficiency and in some cases the quality of reprogramming, but these require the use of a further vector when reprogramming.

Differentiation

Differentiation of pluripotent cells to β-cells is a long, step-wise process, which has proven very difficult to date, discussed by (Quiskamp et al., 2015). The pancreas is endodermal, and generation of definitive endoderm is relatively straight-forward. Following this, cells must be differentiated to gut tube, pancreatic endoderm, endocrine progenitors and finally immature to mature β-cells. The

first major report of generating β-cells was D’Amour et al. (2006), but these cells were not mature and appeared to be polyhormonal. The length of the protocols, use of dozens of factors and number of weakly-defined stages make improving protocols challenging. This is further complicated by the fact that seemingly small factors, such as seeding density (Gage et al., 2013), can have a big effect.

Work on defining developmental stages in human foetal tissue has helped to improve on protocols.

For example, it was shown that NGN3 is expressed only for a window during human development

(Salisbury et al., 2014). Subsequently, it was found that prolonged or premature expression of NGN3 during iPSC differentiation primes cells to become polyhormonal or glucagon expressing (Johansson et al., 2007) and addition of ascorbic acid (vitamin C) can downregulate NGN3 to prevent this and

42 enrich for β-cells (Rezania et al., 2014). This addition, and numerous other refinements led to two similar protocols being published within weeks of each other, demonstrating a major step forward in differentiation of pluripotent cells to β-cells (Rezania et al., 2014, Pagliuca et al., 2014). These papers demonstrated functional β-cells, which were glucose responsive across multiple repeated stimuli, and able to reverse diabetes in mice. These have heralded a new wave of β-cell research based on pluripotent stem cells, with the two papers amassing a cumulative 241 citations in under two years.

This means that in vitro modelling of such as diabetes and CHI, using pluripotent stem cells, may now be possible (Balboa and Otonkoski, 2015). Indeed iPSCs have now been derived from patients with type 1 diabetes, wherein the production of β-cells was not adversely affected by the genetic make-up of the patients (Millman et al., 2016).

1.6 Aims

The aims of this project were to derive stem cells (MSCs and/or iPSCs) from patients with CHI.

Following successful derivation, confirmed by phenotyping, these stem cells would be assessed for their usefulness for in vitro modelling of diabetes and CHI. To do this, the aim was to differentiate the stem cells to functional β-cells, by surveying and modifying protocols from the published literature as necessary. The success of these differentiation protocols would be assessed by gene and protein expression, and insulin secretion. Further, an investigation into whether the cells could be used for modelling hyperproliferation in CHI would be carried out, based on the recent data demonstrating this in CHI tissue. Together these separate threads would give an overall picture of whether CHI stem cells could be used for modelling CHI or diabetes in vitro.

1.7 References

L. Aguilar-Bryan, J. P. Clement, G. Gonzalez, K. Kunjilwar, A. Babenko, and J. Bryan. Toward

Understanding the Assembly and Structure of KATP Channels. Physiological Reviews, 78(1):

227–245, 1998.

U. Ahlgren, J. Jonsson, and H. Edlund. The morphogenesis of the pancreatic mesenchyme is uncou-

pled from that of the pancreatic epithelium in IPF1/PDX1-deficient mice. Development, 122(5):

1409–1416, 1996.

U. Ahlgren, S. L. Pfaff, T. M. Jessell, T. Edlund, and H. Edlund. Independent requirement for ISL1

in formation of pancreatic mesenchyme and islet cells. Nature, 385(6613):257–260, 1997.

J.-B. Arnoux, P. de Lonlay, M.-J. Ribeiro, K. Hussain, O. Blankenstein, K. Mohnike,

V. Valayannopoulos, J.-J. Robert, J. Rahier, C. Sempoux, C. Bellanne, V. Verkarre, Y. Aigrain,

43 F. Jaubert, F. Brunelle, and C. Nihoul-Fekete. Congenital hyperinsulinism. Early Human Devel-

opment, 86(5):287–294, 2010.

J.-B. Arnoux, V. Verkarre, C. Saint-Martin, F. Montravers, A. Brassier, V. Valayannopoulos,

F. Brunelle, J.-C. Fournet, J.-J. Robert, Y. Aigrain, C. Bellanne-Chantelot, and P. de Lonlay.

Congenital hyperinsulinism: current trends in diagnosis and therapy. Orphanet Journal of Rare

Diseases, 6(1):1750–1172, 2011.

I. Artner, B. Blanchi, J. C. Raum, M. Guo, T. Kaneko, S. Cordes, M. Sieweke, and R. Stein. MafB

is required for islet β cell maturation. Proceedings of the National Academy of Sciences, 104(10):

3853–3858, 2007.

V. B. Arya, Q. Aziz, A. Nessa, A. Tinker, and K. Hussain. Congenital hyperinsulinism: clinical and

molecular characterisation of compound heterozygous ABCC8 mutation responsive to Diazoxide

therapy. International Journal of Pediatric , 2014(1):1–5, 2014.

F. M. Ashcroft and P. Rorsman. ATP-sensitive K+ channels: a link between B-cell metabolism and

insulin secretion. Biochemical Society Transactions, 18(1):109–111, 1990.

M. A. Atkinson and G. S. Eisenbarth. Type 1 diabetes: new perspectives on disease pathogenesis

and treatment. The Lancet, 358(9277):221–229, 2001.

M. A. Atkinson, G. S. Eisenbarth, and A. W. Michels. Type 1 diabetes. Lancet, 383(9911):69–82,

2013.

D. Balboa and T. Otonkoski. Human pluripotent stem cell based islet models for diabetes research.

Best Practice & Research Clinical Endocrinology & Metabolism, 29(6):899–909, 2015.

O. Bar-Nur, H. Russ, S. Efrat, and N. Benvenisty. Epigenetic Memory and Preferential Lineage-

Specific Differentiation in Induced Pluripotent Stem Cells Derived from Human Pancreatic Islet

Beta Cells. Cell Stem Cell, 9(1):17–23, 2011.

V. L. Battula, P. M. Bareiss, S. Treml, S. Conrad, I. Albert, S. Hojak, H. Abele, B. Schewe, L. Just,

T. Skutella, and H.-J. Bahring. Human placenta and bone marrow derived MSC cultured in

serum-free, b-FGF-containing medium express cell surface frizzled-9 and SSEA-4 and give rise to

multilineage differentiation. Differentiation, 75(4):279 – 291, 2007.

J. Beagan, T. Gilgenast, J. Kim, Z. Plona, H. Norton, G. Hu, S. Hsu, E. Shield, X. Lyu, E. Apostolou,

K. Hochedlinger, V. Corces, J. Dekker, and J. Phillips-Cremins. Local genome topology can exhibit

an incompletely rewired 3d folding state during somatic cell reprogramming. Cell Stem Cell, 18

(5):611–624, 2016.

44 A. Belfiore and R. Malaguarnera. Insulin receptor and cancer. Endocrine-Related Cancer, 18(4):

R125–R147, 2011.

C. Bertoli, J. M. Skotheim, and R. A. M. de Bruin. Control of cell cycle transcription during G1

and S phases. Nat Rev Mol Cell Biol, 14(8):518 – 528, 2013.

M. Best, M. Carroll, N. A. Hanley, and K. Piper Hanley. Embryonic stem cells to beta-cells by

understanding pancreas development. Molecular and Cellular Endocrinology, 288(1–2):86–94, 2008.

D. R. Bhandari, K.-W. Seo, B. Sun, M.-S. Seo, H.-S. Kim, Y.-J. Seo, J. Marcin, N. Forraz, H. L.

Roy, D. Larry, M. Colin, and K.-S. Kang. The simplest method for in vitro β-cell production from

human adult stem cells. Differentiation, 82(3):144 – 152, 2011.

B. Blum, S. Hrvatin, C. Schuetz, C. Bonal, A. Rezania, and D. A. Melton. Functional beta-cell

maturation is marked by an increased glucose threshold and by expression of urocortin 3. Nat

Biotech, 30(3):261–264, 2012.

B. Blum, A. N. Roose, O. Barrandon, R. Maehr, A. C. Arvanites, L. S. Davidow, J. C. Davis, Q. P.

Peterson, L. L. Rubin, and D. A. Melton. Reversal of β cell de-differentiation by a small molecule

inhibitor of the TGFβ pathway. eLife, 3:e02809–, 2014.

S. Bonner-Weir and G. C. Weir. New sources of pancreatic β-cells. Nat Biotech, 23(7):857–861, 2005.

S. Bonner-Weir, W.-C. Li, L. Ouziel-Yahalom, L. Guo, G. C. Weir, and A. Sharma. β-Cell Growth

and Regeneration: Replication Is Only Part of the Story. Diabetes, 59(10):2340–2348, 2010.

L. Bouwens and I. Rooman. Regulation of Pancreatic Beta-Cell Mass. Physiological Reviews, 85(4):

1255–1270, 2005.

M. Braun, R. Ramracheya, M. Bengtsson, Q. Zhang, J. Karanauskaite, C. Partridge, P. R. Johnson,

and P. Rorsman. Voltage-Gated Ion Channels in Human Pancreatic β-Cells: Electrophysiological

Characterization and Role in Insulin Secretion. Diabetes, 57(6):1618–1628, 2008.

M. Brissova, M. J. Fowler, W. E. Nicholson, A. Chu, B. Hirshberg, D. M. Harlan, and A. C. Powers.

Assessment of Human Pancreatic Islet Architecture and Composition by Laser Scanning Confocal

Microscopy. Journal of Histochemistry & Cytochemistry, 53(9):1087–1097, 2005.

C. Brohem, C. de Carvalho, C. Radoski, F. Santi, M. Baptista, B. Swinka, C. de A Urban,

L. de Araujo, R. Graf, I. Feferman, and M. Lorencini. Comparison between fibroblasts and mes-

enchymal stem cells derived from dermal and adipose tissue. Int J Cosmet Sci., 35(5):448–57,

2013.

45 G. J. Bruining. Recent advances in hyperinsulinism and the pathogenesis of diabetes mellitus. Current

Opinion in Pediatrics, 2(4):758–765, 1990.

A. E. Butler, J. Janson, S. Bonner-Weir, R. Ritzel, R. A. Rizza, and P. C. Butler. β-Cell Deficit and

Increased β-Cell Apoptosis in Humans With Type 2 Diabetes. Diabetes, 52(1):102–110, 2003.

O. Cabrera, D. M. Berman, N. S. Kenyon, C. Ricordi, P.-O. Berggren, and A. Caicedo. The unique

cytoarchitecture of human pancreatic islets has implications for islet cell function. Proceedings of

the National Academy of Sciences of the United States of America, 103(7):2334–2339, 2006.

B. Carey, S. Markoulaki, J. Hanna, D. Faddah, Y. Buganim, J. Kim, K. Ganz, E. Steine, J. Cas-

sady, M. Creyghton, G. Welstead, Q. Gao, and R. Jaenisch. Reprogramming factor stoichiometry

influences the epigenetic state and biological properties of induced pluripotent stem cells. 9(6):

588–598, 2011.

F. Carlotti, A. Zaldumbide, C. J. Loomans, E. van Rossenberg, M. Engelse, E. J. de Koning, and

R. C. Hoeben. Isolated human islets contain a distinct population of mesenchymal stem cells.

Islets, 2(3):164–173, 2010.

M. Carrasco, I. Delgado, B. Soria, F. Martin, and A. Rojas. GATA4 and GATA6 control mouse

pancreas organogenesis. J Clin Invest, 122(10):3504–3515, 2012.

C. Cavelti-Weder, M. Shtessel, J. E. Reuss, A. Jermendy, T. Yamada, F. Caballero, S. Bonner-

Weir, and G. C. Weir. Pancreatic Duct Ligation After Almost Complete β-Cell Loss: Exocrine

Regeneration but No Evidence of β-Cell Regeneration. Endocrinology, 154(12):4493–4502, 2013.

CDC. Prevalence of overweight and obesity among adults with diagnosed Diabetes United States,

1988-1994 and 1999-2000. Centers for Disease Control and Prevention (CDC) (November 2004)

MMWR. Morbidity and Mortality Weekly Report, 53(45):1066–68, 2004.

Z. Cesarz and K. Tamama. Spheroid Culture of Mesenchymal Stem Cells. Stem Cells International,

2016:9176357–, 2015.

J.-L. Chew, Y.-H. Loh, W. Zhang, X. Chen, W.-L. Tam, L.-S. Yeap, P. Li, Y.-S. Ang, B. Lim, P. Rob-

son, and H.-H. Ng. Reciprocal transcriptional regulation of pou5f1 and sox2 via the oct4/sox2

complex in embryonic stem cells. Molecular and Cellular Biology, 25(14):6031–6046, 2005.

V. Cirulli, G. M. Beattie, G. Klier, M. Ellisman, C. Ricordi, V. Quaranta, F. Frasier, J. K. Ishii,

A. Hayek, and D. R. Salomon. Expression and Function of α3 and β5 Integrins in the Developing

Pancreas: Roles in the Adhesion and Migration of Putative Endocrine Progenitor Cells. The

Journal of Cell Biology, 150(6):1445–1460, 2000.

46 M. Cnop, M. J. Landchild, J. Vidal, P. J. Havel, N. G. Knowles, D. R. Carr, F. Wang, R. L. Hull,

E. J. Boyko, B. M. Retzlaff, C. E. Walden, R. H. Knopp, and S. E. Kahn. The Concurrent Ac-

cumulation of Intra-Abdominal and Subcutaneous Fat Explains the Association Between Insulin

Resistance and Plasma Leptin Concentrations: Distinct Metabolic Effects of Two Fat Compart-

ments. Diabetes, 51(4):1005–1015, 2002.

P. Collombat, A. Mansouri, J. Hecksher-Sorensen, P. Serup, J. Krull, G. Gradwohl, and P. Gruss.

Opposing actions of Arx and Pax4 in endocrine pancreas development. Genes & Development, 17

(20):2591–2603, 2003.

E. Corritore, E. Dugnani, V. Pasquale, R. Misawa, P. Witkowski, J. Lei, J. Markmann, L. Piemonti,

E. M. Sokal, S. Bonner-Weir, and P. A. Lysy. β-Cell Differentiation of Human Pancreatic Duct-

Derived Cells After In Vitro Expansion. Cellular Reprogramming, 16(6):456–466, 2014.

M. P. Dalvi, M. R. Umrani, M. V. Joglekar, and A. A. Hardikar. Human pancreatic islet progenitor

cells demonstrate phenotypic plasticity in vitro. Journal of Biosciences, 34(4):523–528, 2009.

K. A. D’Amour, A. G. Bang, S. Eliazer, O. G. Kelly, A. D. Agulnick, N. G. Smart, M. A. Moorman,

E. Kroon, M. K. Carpenter, and E. E. Baetge. Production of pancreatic hormone-expressing

endocrine cells from human embryonic stem cells. Nat Biotech, 24(11):1392–1401, 2006.

D. Daneman. Type 1 diabetes. The Lancet, 367(9513):847–858, 2006.

B. Davani, S. Ariely, L. Ikonomou, Y. Oron, and M. C. Gershengorn. Human islet-derived precursor

cells can cycle between epithelial clusters and mesenchymal phenotypes. Journal of Cellular and

Molecular Medicine, 13:2570–2581, 2009.

D. D. De Leon and C. A. Stanley. Mechanisms of Disease: advances in diagnosis and treatment of

hyperinsulinism in neonates. Nat Clin Pract End Met, 3(1):57–68, 2007.

A. Denley, E. R. Bonython, G. W. Booker, L. J. Cosgrove, B. E. Forbes, C. W. Ward, and J. C.

Wallace. Structural Determinants for High-Affinity Binding of Insulin-Like Growth Factor II to

Insulin Receptor (IR)-A, the Exon 11 Minus Isoform of the IR. Molecular Endocrinology, 18(10):

2502–2512, 2004.

D. Devendra, E. Liu, and G. S. Eisenbarth. Type 1 diabetes: recent developments. BMJ, 328(7442):

750–754, 2004.

Diabetes UK. Diabetes UK Facts and Stats, Nov 2015. URL

https://www.diabetes.org.uk/About us/What-we-say/Statistics/.

47 M. Dominici, K. Le Blanc, I. Mueller, I. Slaper-Cortenbach, F. Marini, D. Krause, R. Deans, A. Keat-

ing, D. Prockop, and E. Horwitz. Minimal criteria for defining multipotent mesenchymal stromal

cells. The International Society for Cellular Therapy position statement. Cytotherapy, 8(4):315–7,

2006.

Y. Dor, J. Brown, O. I. Martinez, and D. A. Melton. Adult pancreatic β-cells are formed by self-

duplication rather than stem-cell differentiation. Nature, 429(6987):41–46, 2004.

A. Du, C. S. Hunter, J. Murray, D. Noble, C.-L. Cai, S. M. Evans, R. Stein, and C. L. May. Islet-1

is Required for the Maturation, Proliferation, and Survival of the Endocrine Pancreas. Diabetes,

58(9):2059–2069, 2009.

M. J. Dunne, C. Kane, R. M. Shepherd, J. A. Sanchez, R. F. James, P. R. Johnson, A. Aynsley-

Green, S. Lu, J. P. Clement, K. J. Lindley, S. Seino, L. Aguilar-Bryan, G. Gonzalez, and P. J. Milla.

Familial Persistent Hyperinsulinemic of Infancy and Mutations in the Sulfonylurea

Receptor. New England Journal of Medicine, 336(10):703–706, 1997.

M. J. Dunne, K. E. Cosgrove, R. M. Shepherd, A. Aynsley-Green, and K. J. Lindley. Hyperinsulinism

in Infancy: From Basic Science to Clinical Disease. Physiological Reviews, 84(1):239–275, 2004.

M. Eberhardt, P. Salmon, M.-A. von Mach, J. G. Hengstler, M. Brulport, P. Linscheid, D. Seboek,

J. Oberholzer, A. Barbero, I. Martin, B. Muller, D. Trono, and H. Zulewski. Multipotential nestin

and Isl-1 positive mesenchymal stem cells isolated from human pancreatic islets. Biochemical and

Biophysical Research Communications, 345(3):1167–1176, 2006.

B. Ebrahimi. Reprogramming barriers and enhancers: strategies to enhance the efficiency and kinetics

of induced pluripotency. 4(1):1–12, 2015.

A. A. Elayat, M. M. el Naggar, and M. Tahir. An immunocytochemical and morphometric study of

the rat pancreatic islets. Journal of Anatomy, 186(Pt 3):629–637, 1995.

C. Evans-Molina, J. C. Garmey, R. Ketchum, K. L. Brayman, S. Deng, and R. G. Mirmira. Glucose

Regulation of Insulin Gene Transcription and Pre-mRNA Processing in Human Islets. Diabetes,

56(3):827–835, 2007.

N. Fiaschi-Taesch, T. A. Bigatel, B. Sicari, K. K. Takane, F. Salim, S. Velazquez-Garcia, G. Harb,

K. Selk, I. Cozar-Castellano, and A. F. Stewart. Survey of the Human Pancreatic β-Cell G1/S

Proteome Reveals a Potential Therapeutic Role for Cdk-6 and Cyclin D1 in Enhancing Human

β-Cell Replication and Function In Vivo. Diabetes, 58(4):882–893, 2009.

48 N. M. Fiaschi-Taesch, J. W. Kleinberger, F. G. Salim, R. Troxell, R. Wills, M. Tanwir, G. Casinelli,

A. E. Cox, K. K. Takane, D. K. Scott, and A. F. Stewart. Human pancreatic β-cell g1/s molecule

cell cycle atlas. Diabetes, 62(7):2450, 2013.

J.-C. Fournet, C. Mayaud, P. de Lonlay, M.-S. Gross-Morand, V. Verkarre, M. Castanet, M. Devillers,

J. Rahier, F. Brunelle, J.-J. Robert, C. Nihoul-Fekete, J.-M. Saudubray, and C. Junien. Unbalanced

Expression of 11p15 Imprinted Genes in Focal Forms of Congenital Hyperinsulinism. The American

Journal of Pathology, 158(6):2177–2184, 2001.

L. E. Fridlyand, D. A. Jacobson, and L. Philipson. Ion channels and regulation of insulin secretion

in human β-cells: A computational systems analysis. Islets, 5(1):1–15, 2013.

O. Friedman-Mazursky, R. Elkon, and S. Efrat. Redifferentiation of expanded human islet β cells by

inhibition of ARX. Scientific Reports, 6:20698–, 2016.

K. Furuyama, Y. Kawaguchi, H. Akiyama, M. Horiguchi, S. Kodama, T. Kuhara, S. Hosokawa,

A. Elbahrawy, T. Soeda, M. Koizumi, T. Masui, M. Kawaguchi, K. Takaori, R. Doi, E. Nishi,

R. Kakinoki, J. M. Deng, R. R. Behringer, T. Nakamura, and S. Uemoto. Continuous cell supply

from a Sox9-expressing progenitor zone in adult liver, exocrine pancreas and intestine. Nat Genet,

43(1):34–41, 2011.

N. Fusaki, B. A. N. Hiroshi, A. Nishiyama, K. Saeki, and M. Hasegawa. Efficient induction of

transgene-free human pluripotent stem cells using a vector based on sendai virus, an rna virus

that does not integrate into the host genome:. 85(8):348–362, 2009.

M. M. Gabr, M. M. Zakaria, A. F. Refaie, S. M. Khater, S. A. Ashamallah, A. M. Ismail, N. El-Badri,

and M. A. Ghoneim. Generation of Insulin-Producing Cells from Human Bone Marrow-Derived

Mesenchymal Stem Cells: Comparison of Three Differentiation Protocols. BioMed Research In-

ternational, 832736:9, 2014.

B. K. Gage, T. D. Webber, and T. J. Kieffer. Initial Cell Seeding Density Influences Pancreatic

Endocrine Development During in vitro Differentiation of Human Embryonic Stem Cells. PLoS

ONE, 8(12):e82076–, 2013.

E. A. Gale. European Nicotinamide Diabetes Intervention Trial (ENDIT): a randomised controlled

trial of intervention before the onset of type 1 diabetes. The Lancet, 363(9413):925–931, 2004.

R. Gallo, F. Gambelli, B. Gava, F. Sasdelli, V. Tellone, M. Masini, P. Marchetti, F. Dotta, and

V. Sorrentino. Generation and expansion of multipotent mesenchymal progenitor cells from cul-

tured human pancreatic islets. Cell Death Differ, 14(11):1860–1871, 2007.

49 K. E. Gerrish, M. A. Cissell, and R. Stein. The role of hepatic nuclear factor 1a and PDX-1 in

transcriptional regulation of the pdx-1 gene. Journal of Biological Chemistry, 276(51):47775–84,

2001.

G. K. Gittes. Developmental biology of the pancreas: A comprehensive review. Developmental

Biology, 326(1):4–35, 2009.

I. Giurgea, C. Bellanna-Chantelot, M. Ribeiro, L. Hubert, C. Sempoux, J.-J. Robert, O. Blankenstein,

K. Hussain, F. Brunelle, C. Nihoul-Fekete, J. Rahier, F. Jaubert, and P. de Lonlay. Molecular

Mechanisms of Neonatal Hyperinsulinism. Horm Res Paediatr, 66(6):289–296, 2006.

B. Glaser, H. Landau, A. Smilovici, and R. Nesher. Persistent hyperinsulinaemic hypoglycaemia of

infancy: long-term treatment with the somatostatin anologue sandostatin. Clinical Endocrinology,

31(1):71–80, 1989.

B. Glaser, P. Thornton, T. Otonkoski, and C. Junien. Genetics of neonatal hyperinsulinism. Archives

of Disease in Childhood - Fetal and Neonatal Edition, 82(2):F79–F86, 2000.

K. J. Godfrey, B. Mathew, J. C. Bulman, O. Shah, S. Clement, and G. I. Gallicano. Stem cell-based

treatments for Type1 diabetes mellitus: bone marrow, embryonic, hepatic, pancreatic and induced

pluripotent stem cells. Diabetic Medicine, 29(1):14–23, 2012.

R. Gopurappilly, V. Bhat, and R. Bhonde. Pancreatic tissue resident mesenchymal stromal cell

(msc)-like cells as a source of in vitro islet neogenesis. Journal of Cellular Biochemistry, 114(10):

2240–2247, 2013.

G. Gradwohl, A. Dierich, M. LeMeur, and F. Guillemot. neurogenin3 is required for the development

of the four endocrine cell lineages of the pancreas. Proceedings of the National Academy of Sciences,

97(4):1607–1611, 2000.

G. Gu, J. Dubauskaite, and D. A. Melton. Direct evidence for the pancreatic lineage: NGN3+ cells

are islet progenitors and are distinct from duct progenitors. Development, 129(10):2447–2457,

2002.

S. Hacein-Bey-Abina, C. von Kalle, M. Schmidt, F. Le Deist, N. Wulffraat, E. McIntyre, I. Radford,

J.-L. Villeval, C. C. Fraser, M. Cavazzana-Calvo, and A. Fischer. A serious adverse event after

successful gene therapy for x-linked severe combined immunodeficiency. New England Journal of

Medicine, 348(3):255–256, 2003.

N. Hanley. Closing in on pancreatic beta cells. Nat Biotech, 32(11):1100–1102, Nov. 2014.

50 M. Hansson, A. Tonning, U. Frandsen, A. Petri, J. Rajagopal, M. C. Englund, R. S. Heller, J. Hakans-

son, J. Fleckner, H. N. Skold, D. Melton, H. Semb, and P. Serup. Artifactual Insulin Release From

Differentiated Embryonic Stem Cells. Diabetes, 53(10):2603–2609, 2004.

H. Harashima, N. Dissmeyer, and A. Schnittger. Cell cycle control across the eukaryotic kingdom.

Trends in Cell Biology, 23(7):345 – 356, 2013.

J.-C. Henquin, C. Sempoux, J. Marchandise, S. Godecharles, Y. Guiot, M. Nenquin, and J. Rahier.

Congenital Hyperinsulinism Caused by Hexokinase I Expression or Glucokinase-Activating Muta-

tion in a Subset of β-Cells. Diabetes, 62(5):1689–1696, 2013.

Y. Hori, I. C. Rulifson, B. C. Tsai, J. J. Heit, J. D. Cahoy, and S. K. Kim. Growth inhibitors promote

differentiation of insulin-producing tissue from embryonic stem cells. Proceedings of the National

Academy of Sciences, 99(25):16105–16110, 2002.

F. B. Hu, J. E. Manson, M. J. Stampfer, G. Colditz, S. Liu, C. G. Solomon, and W. C. Willett. Diet,

Lifestyle, and the Risk of Type 2 Diabetes Mellitus in Women. N Engl J Med, 345(11):790–797,

2001.

D. Huangfu, R. Maehr, W. Guo, A. Eijkelenboom, M. Snitow, A. E. Chen, and D. A. Melton. Induc-

tion of pluripotent stem cells by defined factors is greatly improved by small-molecule compounds.

26(7):795–797, 2008a.

D. Huangfu, K. Osafune, R. Maehr, W. Guo, A. Eijkelenboom, S. Chen, W. Muhlestein, and D. A.

Melton. Induction of pluripotent stem cells from primary human fibroblasts with only oct4 and

sox2. 26(11):1269–1275, 2008b.

M. Huch, P. Bonfanti, S. F. Boj, T. Sato, C. J. M. Loomans, M. van de Wetering, M. Sojoodi,

V. S. W. Li, J. Schuijers, A. Gracanin, F. Ringnalda, H. Begthel, K. Hamer, J. Mulder, J. H. van

Es, E. de Koning, R. G. J. Vries, H. Heimberg, and H. Clevers. Unlimited in vitro expansion of

adult bi-potent pancreas progenitors through the Lgr5/R-spondin axis. The EMBO Journal, 32

(20):2708–2721, 2013.

E. Hunziker and M. Stein. Nestin-Expressing Cells in the Pancreatic Islets of Langerhans. Biochemical

and Biophysical Research Communications, 271(1):116–119, 2000.

K. Hussain. Diagnosis and Management of Hyperinsulinaemic Hypoglycaemia of Infancy. Horm Res

Paediatr, 69(1):2–13, 2008.

K. Hussain and A. Aynsley-Green. Hyperinsulinaemic hypoglycaemia in preterm neonates. Archives

of Disease in Childhood - Fetal and Neonatal Edition, 89(1):F65–F67, 2004.

51 D. Ish-Shalom, C. Christoffersen, P. Vorwerk, N. Sacerdoti-Sierra, R. Shymko, D. Naor, and

P. De Meyts. Mitogenic properties of insulin and insulin analogues mediated by the insulin recep-

tor. Diabetologia, 40(S2):S25–31, 1997.

T. Iype, J. Francis, J. C. Garmey, J. C. Schisler, R. Nesher, G. C. Weir, T. C. Becker, C. B. Newgard,

S. C. Griffen, and R. G. Mirmira. Mechanism of insulin Gene Regulation by the Pancreatic Tran-

scription Factor Pdx-1: Application of pre-mRNA analysis and chromatin immunoprecipitation

to assess formation of functional transcriptional complexes. Journal of Biological Chemistry, 280

(17):16798–16807, 2005.

M. M. Jack, R. M. Greer, M. J. Thomsett, R. M. Walker, J. R. Bell, C. Choong, D. M. Cowley,

A. C. Herington, and A. M. Cotterill. The outcome in Australian children with hyperinsulinism

of infancy: early extensive surgery in severe cases lowers risk of diabetes. Clinical Endocrinology,

58(3):355–364, 2003.

P. Jacquemin, F. P. Lemaigre, and G. G. Rousseau. The Onecut transcription factor HNF-6 (OC-1)

is required for timely specification of the pancreas and acts upstream of Pdx-1 in the specification

cascade. Developmental Biology, 258(1):105–116, 2003.

A. Jafarian, M. Taghikhani, S. Abroun, Z. Pourpak, A. Allahverdi, and M. Soleimani. Generation

of high-yield insulin producing cells from human bone marrow mesenchymal stem cells. Molecular

Biology Reports, 41(7):4783–4794, 2014.

C. James, R. R. Kapoor, D. Ismail, and K. Hussain. The genetic basis of congenital hyperinsulinism.

Journal of Medical Genetics, 46(5):289–299, 2009.

J. Jensen, R. S. Heller, T. Funder-Nielsen, E. E. Pedersen, C. Lindsell, G. Weinmaster, O. D. Madsen,

and P. Serup. Independent development of pancreatic alpha- and beta-cells from neurogenin3-

expressing precursors: a role for the notch pathway in repression of premature differentiation.

Diabetes, 49(2):163–176, 2000.

M. V. Joglekar, V. M. Joglekar, S. V. Joglekar, and A. A. Hardikar. Human fetal pancreatic insulin-

producing cells proliferate in vitro. Journal of Endocrinology, 201(1):27–36, 2009.

K. A. Johansson, U. Dursun, N. Jordan, G. Gu, F. Beermann, G. Gradwohl, and A. Grapin-Botton.

Temporal Control of Neurogenin3 Activity in Pancreas Progenitors Reveals Competence Windows

for the Generation of Different Endocrine Cell Types. Developmental Cell, 12(3):457–465, 2007.

J. D. Johnson, E. Bernal-Mizrachi, E. U. Alejandro, Z. Han, T. B. Kalynyak, H. Li, J. L. Beith,

J. Gross, G. L. Warnock, R. R. Townsend, M. A. Permutt, and K. S. Polonsky. Insulin protects

52 islets from apoptosis via Pdx1 and specific changes in the human islet proteome. Proceedings of

the National Academy of Sciences, 103(51):19575–19580, 2006.

S. E. Kahn, M. E. Cooper, and S. Del Prato. Pathophysiology and treatment of type 2 diabetes:

perspectives on the past, present, and future. The Lancet, 383(9922):1068–1083, 2014.

T. Kaido, M. Yebra, V. Cirulli, C. Rhodes, G. Diaferia, and A. M. Montgomery. Impact of Defined

Matrix Interactions on Insulin Production by Cultured Human β-Cells: Effect on Insulin Content,

Secretion, and Gene Transcription. Diabetes, 55(10):2723–2729, 2006.

V. Kashyap, N. C. Rezende, K. B. Scotland, S. M. Shaffer, J. L. Persson, L. J. Gudas, and N. P.

Mongan. Regulation of stem cell pluripotency and differentiation involves a mutual regulatory

circuit of the nanog, oct4, and sox2 pluripotency transcription factors with polycomb repressive

complexes and stem cell micrornas. Stem Cells and Development, 18(7):1093–1108, 2009.

S. A. Kassem, I. Ariel, P. S. Thornton, I. Scheimberg, and B. Glaser. Beta-cell proliferation and

apoptosis in the developing normal human pancreas and in hyperinsulinism of infancy. Diabetes,

49(8):1325–1333, 2000.

S. A. Kassem, I. Ariel, P. S. Thornton, K. Hussain, V. Smith, K. J. Lindley, A. Aynsley-Green, and

B. Glaser. p57KIP2 Expression in Normal Islet Cells and in Hyperinsulinism of Infancy. Diabetes,

50(12):2763–2769, 2001.

Y. Kawaguchi. Sox9 and programming of liver and pancreatic progenitors. J Clin Invest, 123(5):

1881–1886, 2013.

I. Ketola, T. Otonkoski, M.-A. Pulkkinen, H. Niemi, J. Palgi, C. M. Jacobsen, D. B. Wilson, and

M. Heikinheimo. Transcription factor GATA-6 is expressed in the endocrine and GATA-4 in the

exocrine pancreas. Molecular and Cellular Endocrinology, 226(1–2):51–57, 2004.

J. Kim, J. Chu, X. Shen, J. Wang, and S. H. Orkin. An extended transcriptional network for

pluripotency of embryonic stem cells. Cell, 132(6):1049–1061, 2008.

K. Kim, R. Zhao, A. Doi, K. Ng, J. Unternaehrer, P. Cahan, H. Hongguang, Y.-H. Loh, M. J. Aryee,

M. W. Lensch, H. Li, J. J. Collins, A. P. Feinberg, and G. Q. Daley. Donor cell type can influence

the epigenome and differentiation potential of human induced pluripotent stem cells. Nat Biotech,

29(12):1117–1119, 2011.

S.-J. Kim, Y.-S. Choi, E.-S. Ko, S.-M. Lim, C.-W. Lee, and D.-I. Kim. Glucose-stimulated insulin

secretion of various mesenchymal stem cells after insulin-producing cell differentiation. Journal of

Bioscience and Bioengineering, 113(6):771 – 777, 2012.

53 J. L. Kopp, C. L. Dubois, E. Hao, F. Thorel, P. L. Herrera, and M. Sander. Progenitor cell domains

in the developing and adult pancreas. Cell Cycle, 10(12):1921–1927, 2011.

J. C. Koster, M. A. Permutt, and C. G. Nichols. Diabetes and Insulin Secretion: The ATP-Sensitive

K+ Channel (KATP) Connection. Diabetes, 54(11):3065–3072, 2005.

M. Koyanagi-Aoi, M. Ohnuki, K. Takahashi, K. Okita, H. Noma, Y. Sawamura, I. Teramoto,

M. Narita, Y. Sato, T. Ichisaka, N. Amano, A. Watanabe, A. Morizane, Y. Yamada, T. Sato,

J. Takahashi, and S. Yamanaka. Differentiation-defective phenotypes revealed by large-scale anal-

yses of human pluripotent stem cells. Proceedings of the National Academy of Sciences, 110(51):

20569–20574, 2013.

R. N. Kulkarni, E.-B. Mizrachi, A. G. Ocana, and A. F. Stewart. Human β-Cell Proliferation and

Intracellular Signaling: Driving in the Dark Without a Road Map. Diabetes, 61(9):2205–2213,

2012.

C. S. Lee, N. J. Sund, M. Z. Vatamaniuk, F. M. Matschinsky, D. A. Stoffers, and K. H. Kaestner.

Foxa2 Controls Pdx1 Gene Expression in Pancreatic β-Cells In Vivo. Diabetes, 51(8):2546–2551,

2002.

A. Leha, N. Moens, R. Meleckyte, O. J. Culley, M. K. Gervasio, M. Kerz, A. Reimer, S. A. Cain,

I. Streeter, A. Folarin, O. Stegle, C. M. Kielty, R. Durbin, F. M. Watt, and D. Danovi. A high-

content platform to characterise human induced pluripotent stem cell lines. Methods, 96:85–96,

2016.

B. Leibiger, T. Moede, S. Uhles, P.-O. Berggren, and I. B. Leibiger. Short-term regulation of insulin

gene transcription. Biochemical Society Transactions, 30(2):312–317, 2002.

G. Leibowitz, B. Glaser, A. A. Higazi, M. Salameh, E. Cerasi, and H. Landau. Hyperinsulinemic

hypoglycemia of infancy (nesidioblastosis) in clinical remission: high incidence of diabetes mellitus

and persistent beta-cell dysfunction at long-term follow-up. The Journal of Clinical Endocrinology

& Metabolism, 80(2):386–392, 1995.

J. Li, L. Zhu, X. Qu, J. Li, R. Lin, L. Liao, J. Wang, S. Wang, Q. Xu, and R. C. Zhao. Stepwise

Differentiation of Human Adipose-Derived Mesenchymal Stem Cells Toward Definitive Endoderm

and Pancreatic Progenitor Cells by Mimicking Pancreatic Development In Vivo. Stem Cells and

Development, 22(10):1576–1587, 2012.

L.-C. Li and R. Dahiya. MethPrimer: designing primers for methylation PCRs. Bioinformatics, 18

(11):1427–1431, 2002.

54 M. Li and S. Ikehara. Bone Marrow Stem Cell as a Potential Treatment for Diabetes. Journal of

Diabetes Research, 2013:5, 2013.

W.-C. Li, J. M. Rukstalis, W. Nishimura, V. Tchipashvili, J. F. Habener, A. Sharma, and S. Bonner-

Weir. Activation of pancreatic-duct-derived progenitor cells during pancreas regeneration in adult

rats. Journal of Cell Science, 123(16):2792–2802, 2010.

C. G. Liew, N. N. Shah, S. J. Briston, R. M. Shepherd, C. P. Khoo, M. J. Dunne, H. D. Moore, K. E.

Cosgrove, and P. W. Andrews. PAX4 Enhances Beta-Cell Differentiation of Human Embryonic

Stem Cells. PLoS ONE, 3(3):e1783–, 2008.

M. J. Lima, K. R. Muir, H. M. Docherty, R. Drummond, N. W. McGowan, S. Forbes, Y. Heremans,

I. Houbracken, J. A. Ross, S. J. Forbes, P. Ravassard, H. Heimberg, J. Casey, and K. Docherty.

Suppression of Epithelial-to-Mesenchymal Transitioning Enhances Ex Vivo Reprogramming of

Human Exocrine Pancreatic Tissue Toward Functional Insulin-Producing β-Like Cells. Diabetes,

62(8):2821–2833, 2013.

N. Lumelsky, O. Blondel, P. Laeng, I. Velasco, R. Ravin, and R. McKay. Differentiation of Embryonic

Stem Cells to Insulin-Secreting Structures Similar to Pancreatic Islets. Science, 292(5520):1389–

1394, 2001.

M. J. MacDonald, M. J. Longacre, S. W. Stoker, M. Kendrick, A. Thonpho, L. J. Brown, N. M. Hasan,

S. Jitrapakdee, T. Fukao, M. S. Hanson, L. A. Fernandez, and J. Odorico. Differences between

Human and Rodent Pancreatic Islets: Low pyruvate carboxylase, atp citrate lyase, and pyruvate

carboxylation and high glucose-stimulated acetoacetate in human pancreatic islets. Journal of

Biological Chemistry, 286(21):18383–18396, 2011.

W. M. Macfarlane, R. M. Shepherd, K. E. Cosgrove, R. F. James, M. J. Dunne, and K. Docherty.

Glucose modulation of insulin mRNA levels is dependent on transcription factor PDX-1 and occurs

independently of changes in intracellular Ca2+. Diabetes, 49(3):418–423, 2000.

R. Maehr. iPS Cells in Type 1 Diabetes Research and Treatment. Clinical Pharmacology & Thera-

peutics, 89(5):750–753, 2011.

M. Maekawa, K. Yamaguchi, T. Nakamura, R. Shibukawa, I. Kodanaka, T. Ichisaka, Y. Kawamura,

H. Mochizuki, N. Goshima, and S. Yamanaka. Direct reprogramming of somatic cells is promoted

by maternal transcription factor glis1. 474(7350):225–229, 2011.

N. M. Maruthur, E. Tseng, S. Hutfless, L. M. Wilson, C. Suarez-Cuervo, Z. Berger, Y. Chu, E. Iyoha,

J. B. Segal, and S. Bolen. Diabetes Medications as Monotherapy or Metformin-Based Combination

Therapy for Type 2 Diabetes: A Systematic Review and Meta-analysisDiabetes Medications as

55 Monotherapy or Metformin-Based Combination Therapy. Annals of Internal Medicine, Epub

ahead of print, 2016.

F. M. Matschinsky. Regulation of Pancreatic β-Cell Glucokinase: From Basics to Therapeutics.

Diabetes, 51(suppl 3):S394–S404, 2002.

T. Meissner, U. Wendel, P. Burgard, S. Schaetzle, and E. Mayatepek. Long-term follow-up of 114

patients with congenital hyperinsulinism. European Journal of Endocrinology, 149(1):43–51, 2003.

D. Melloul, S. Marshak, and E. Cerasi. Regulation of insulin gene transcription. Diabetologia, 45(3):

309–326, 2014.

J. Millman, X. Chunhui, A. Van Devort, M. Gurtler, F. Pagliuca, and D. Melton. Generation of stem

cell-derived β-cells from patients with type 1 diabetes. Nature Communications, 7:11463, 2016.

K. Moens, V. Berger, J.-M. Ahn, C. Van Schravendijk, V. J. Hruby, D. Pipeleers, and F. Schuit.

Assessment of the Role of Interstitial Glucagon in the Acute Glucose Secretory Responsiveness of

In Situ Pancreatic β-Cells. Diabetes, 51(3):669–675, 2002.

A. H. Mokdad, E. S. Ford, B. Bowman, W. H. Dietz, F. Vinicor, V. S. Bales, and J. S. Marks.

Prevalence of obesity, diabetes, and obesity-related health risk factors, 2001. JAMA, 289(1):

76–79, 2003.

N. Moran. Banking iPS cells. Nat Biotech, 31(1):11–11, 2013.

L. C. Murtaugh, B. Z. Stanger, K. M. Kwan, and D. A. Melton. Notch signaling controls multiple

steps of pancreatic differentiation. Proceedings of the National Academy of Sciences, 100(25):

14920–14925, 2003.

National Institute for Health and Care Excellence. Type 2 diabetes in adults: management (NICE

guidelines NG28), December 2015. URL https://www.nice.org.uk/guidance/ng28.

F. J. Naya, C. M. Stellrecht, and M. J. Tsai. Tissue-specific regulation of the insulin gene by a novel

basic helix-loop-helix transcription factor. Genes & Development, 9(8):1009–1019, 1995.

W. Nishimura, T. Kondo, T. Salameh, I. El Khattabi, R. Dodge, S. Bonner-Weir, and A. Sharma.

A switch from MafB to MafA expression accompanies differentiation to pancreatic β-cells. Devel-

opmental Biology, 293(2):526–539, 2006.

U. E. Ntuk, J. M. Gill, D. F. Mackay, N. Sattar, and J. P. Pell. Ethnic-Specific Obesity Cutoffs for

Diabetes Risk: Cross-sectional Study of 490,288 UK Biobank Participants. Diabetes Care, 37(9):

2500–2507, 2014.

56 K. Okita, T. Ichisaka, and S. Yamanaka. Generation of germline-competent induced pluripotent

stem cells. Nature, 448(7151):313–317, 2007.

T. Otonkoski, K. Nanto-Salonen, M. Seppnen, R. Veijola, H. Huopio, K. Hussain, P. Tapanainen,

O. Eskola, R. Parkkola, K. Ekstrom, Y. Guiot, J. Rahier, M. Laakso, R. Rintala, P. Nuutila, and

H. Minn. Noninvasive Diagnosis of Focal Hyperinsulinism of Infancy With [18F]-DOPA Positron

Emission Tomography. Diabetes, 55(1):13–18, 2006.

F. Pagliuca, J. Millman, M. Gurtler, M. Segel, A. VanDervort, J. Ryu, Q. Peterson, D. Greiner, and

D. Melton. Generation of Functional Human Pancreatic β Cells In Vitro. Cell, 159(2):428–439,

2014.

F. C. Pan and C. Wright. Pancreas organogenesis: From bud to plexus to gland. Dev. Dyn., 240(3):

530–565, 2011.

K. Peters, R. Panienka, J. Li, G. Kloppel, and R. Wang. Expression of stem cell markers and

transcription factors during the remodeling of the rat pancreas after duct ligation. Virchows

Archiv, 446(1):56–63, 2004.

H. V. Petersen, P. Serup, J. Leonard, B. K. Michelsen, and O. D. Madsen. Transcriptional Regula-

tion of the Human Insulin Gene is Dependent on the Homeodomain Protein STF1/IPF1 Acting

Through the CT Boxes. Proceedings of the National Academy of Sciences of the United States of

America, 91(22):10465–10469, 1994.

P. Petit and M. Loubatieres-Mariani. Potassium channels of the insulin-secreting B cell. Fundamental

& Clinical Pharmacology, 6(3):123–134, 1992.

D. G. Pipeleers. Heterogeneity in Pancreatic β-cell Population. Diabetes, 41(7):777–781, 1992.

K. Piper, S. Ball, L. Turnpenny, S. Brickwood, D. Wilson, and N. Hanley. Beta-cell differentiation

during human development does not rely on nestin-positive precursors: implications for stem cell-

derived replacement therapy. Diabetologia, 45(7):1045–7, 2002.

K. Piper, S. Brickwood, L. Turnpenny, I. Cameron, S. Ball, D. Wilson, and N. Hanley.

differentiation during early human pancreas development. Journal of Endocrinology, 181(1):11–23,

2004.

G. L. Pittenger, D. Taylor-Fishwick, and A. I. Vinik. The Role of Islet Neogeneis-Associated Protein

(INGAP) in Pancreatic Islet Neogenesis. Current Protein & Peptide Science, 10(1):37–45, 2009.

P. Proks, F. Reimann, N. Green, F. Gribble, and F. Ashcroft. Sulfonylurea Stimulation of Insulin

Secretion. Diabetes, 51(suppl 3):S368–S376, 2002.

57 S. Puri and M. Hebrok. Cellular Plasticity within the Pancreas - Lessons Learned from Development.

Developmental Cell, 18(3):342–356, 2010.

N. Quiskamp, J. E. Bruin, and T. J. Kieffer. Differentiation of human pluripotent stem cells into

β-cells: Potential and challenges. Best Practice & Research Clinical Endocrinology & Metabolism,

29(6):833–847, 2015.

M. M. Rankin, C. J. Wilbur, K. Rak, E. J. Shields, A. Granger, and J. A. Kushner. β-Cells Are Not

Generated in Pancreatic Duct Ligation-Induced Injury in Adult Mice. Diabetes, 62(5):1634–1645,

2013.

J. M. Redondo, L. Yu, M. Hawa, T. Mackenzie, A. D. Pyke, S. G. Eisenbarth, and G. R. D. Leslie.

Heterogeneity of Type I diabetes: analysis of monozygotic twins in Great Britain and the United

States. Diabetologia, 44(3):354–362, 2001.

D. A. Rees and J. C. Alcolado. Animal models of diabetes mellitus. Diabetic Medicine, 22(4):359–370,

2005.

A. Rezania, J. E. Bruin, P. Arora, A. Rubin, I. Batushansky, A. Asadi, S. O’Dwyer, N. Quiskamp,

M. Mojibian, T. Albrecht, Y. H. C. Yang, J. D. Johnson, and T. J. Kieffer. Reversal of diabetes

with insulin-producing cells derived in vitro from human pluripotent stem cells. Nat Biotech, 32

(11):1121–1133, 2014.

D. A. Robinton and G. Q. Daley. The promise of induced pluripotent stem cells in research and

therapy. 481(7381):295–305, 2012.

D. J. Rodda, J.-L. Chew, L.-H. Lim, Y.-H. Loh, B. Wang, H.-H. Ng, and P. Robson. Transcriptional

regulation of nanog by oct4 and sox2. Journal of Biological Chemistry, 280(26):24731–24737, 2005.

I. Rooman, J. Lardon, and L. Bouwens. Gastrin Stimulates β-Cell Neogenesis and Increases Islet

Mass From Transdifferentiated but Not From Normal Exocrine Pancreas Tissue. Diabetes, 51(3):

686–690, 2002.

F. Rouhani, N. Kumasaka, M. C. de Brito, A. Bradley, L. Vallier, and D. Gaffney. Genetic Back-

ground Drives Transcriptional Variation in Human Induced Pluripotent Stem Cells. PLoS Genet,

10(6):e1004432–, 2014.

J. M. Rukstalis and J. F. Habener. Snail2, a mediator of epithelial-mesenchymal transitions, ex-

pressed in progenitor cells of the developing endocrine pancreas. Gene Expression Patterns, 7(4):

471–479, 2007.

58 H. A. Russ, Y. Bar, P. Ravassard, and S. Efrat. In Vitro Proliferation of Cells Derived From Adult

Human β-Cells Revealed By Cell-Lineage Tracing. Diabetes, 57(6):1575–1583, 2008.

H. A. Russ, P. Ravassard, J. Kerr-Conte, F. Pattou, and S. Efrat. Epithelial-Mesenchymal Transition

in Cells Expanded In Vitro from Lineage-Traced Adult Human Pancreatic Beta Cells. PLoS ONE,

4(7):e6417, 2009.

W. Russell and R. Burch. The Principles of Humane Experimental Technique. Methuen, 1959.

E. A. Ryan, B. W. Paty, P. A. Senior, D. Bigam, E. Alfadhli, N. M. Kneteman, J. R. Lakey, and

A. J. Shapiro. Five-Year Follow-Up After Clinical Islet Transplantation. Diabetes, 54(7):2060–2069,

2005.

R. J. Salisbury, J. Blaylock, A. A. Berry, R. E. Jennings, R. De Krijger, K. Piper Hanley, and N. A.

Hanley. The window period of NEUROGENIN3 during human gestation. Islets, 6(3):e954436–,

2014.

R. J. Salisbury, B. Han, R. E. Jennings, A. A. Berry, A. Stevens, Z. Mohamed, S. A. Sugden,

R. De Krijger, S. E. Cross, P. P. Johnson, M. Newbould, K. E. Cosgrove, K. P. Hanley, I. Banerjee,

M. J. Dunne, and N. A. Hanley. Altered Phenotype of β-Cells and Other Pancreatic Cell Lineages

in Patients With Diffuse Congenital Hyperinsulinism in Infancy Caused by Mutations in the ATP-

Sensitive K-Channel. Diabetes, 64(9):3182–3188, 2015.

S. E. Samaras, M. A. Cissell, K. Gerrish, C. V. E. Wright, M. Gannon, and R. Stein. Conserved

Sequences in a Tissue-Specific Regulatory Region of the pdx-1 Gene Mediate Transcription in

Pancreatic β Cells: Role for Hepatocyte Nuclear Factor 3β and Pax6. Molecular and Cellular

Biology, 22(13):4702–4713, 2002.

S. E. Samaras, L. Zhao, A. Means, E. Henderson, T.-a. Matsuoka, and R. Stein. The Islet β Cell-

enriched RIPE3b1/Maf Transcription Factor Regulates pdx-1 Expression. Journal of Biological

Chemistry, 278(14):12263–12270, 2003.

M. Sander, A. Neubuser, J. Kalamaras, H. C. Ee, G. R. Martin, and M. S. German. Genetic analysis

reveals that PAX6 is required for normal transcription of pancreatic hormone genes and islet

development. Genes & Development, 11(13):1662–1673, 1997.

V. Schwitzgebel, D. Scheel, J. Conners, J. Kalamaras, J. Lee, D. Anderson, L. Sussel, J. Johnson, and

M. German. Expression of neurogenin3 reveals an islet cell precursor population in the pancreas.

Development, 127(16):3533–3542, 2000.

59 K. L. Seeberger, J. M. Dufour, A. M. J. Shapiro, J. R. T. Lakey, R. V. Rajotte, and G. S. Korbutt.

Expansion of mesenchymal stem cells from human pancreatic ductal epithelium. Lab Invest, 86

(2):141–153, 2006.

K. L. Seeberger, A. Eshpeter, R. V. Rajotte, and G. S. Korbutt. Epithelial cells within the human

pancreas do not coexpress mesenchymal antigens: epithelial-mesenchymal transition is an artifact

of cell culture. Lab Invest, 89(2):110–121, 2008.

T. Seki, S. Yuasa, M. Oda, T. Egashira, K. Yae, D. Kusumoto, H. Nakata, S. Tohyama, H. Hashimoto,

M. Kodaira, Y. Okada, H. Seimiya, N. Fusaki, M. Hasegawa, and K. Fukuda. Generation of induced

pluripotent stem cells from human terminally differentiated circulating t cells. 7(1):11–14, 2010.

P. A. Seymour. Sox9: A Master Regulator of the Pancreatic Program. The Review of Diabetic

Studies : RDS, 11(1):51–83, 2013.

P. A. Seymour, K. K. Freude, M. N. Tran, E. E. Mayes, J. Jensen, R. Kist, G. Scherer, and M. Sander.

SOX9 is required for maintenance of the pancreatic progenitor cell pool. Proceedings of the National

Academy of Sciences, 104(6):1865–1870, 2007.

A. J. Shapiro, J. R. Lakey, E. A. Ryan, G. S. Korbutt, E. Toth, G. L. Warnock, N. M. Kneteman,

and R. V. Rajotte. Islet Transplantation in Seven Patients with Type 1 Diabetes Mellitus Using

a Glucocorticoid-Free Immunosuppressive Regimen. New England Journal of Medicine, 343(4):

230–238, 2000.

A. A. Sharov, S. Masui, L. V. Sharova, Y. Piao, K. Aiba, R. Matoba, L. Xin, H. Niwa, and M. S. H.

Ko. Identification of pou5f1, sox2, and nanog downstream target genes with statistical confidence

by applying a novel algorithm to time course microarray and genome-wide chromatin immunopre-

cipitation data. BMC Genomics, 9(1):1–19, 2008.

K. Shimomura, M. Tusa, M. Iberl, M. F. Brereton, S. Kaizik, P. Proks, C. Lahmann, N. Yaluri,

S. Modi, H. Huopio, J. Ustinov, T. Otonkoski, M. Laakso, and F. M. Ashcroft. A Mouse Model of

Human Hyperinsulinism Produced by the E1506K Mutation in the Sulphonylurea Receptor SUR1.

Diabetes, 62(11):3797–3806, 2013.

J. Silva, J. Nichols, T. W. Theunissen, G. Guo, A. L. van Oosten, O. Barrandon, J. Wray, S. Ya-

manaka, I. Chambers, and A. Smith. Nanog is the gateway to the pluripotent ground state. 138

(4):722–737, 2009.

M. Skelin, M. Rupnik, and A. Cencic. Pancreatic beta cell lines and their applications in diabetes

mellitus research. ALTEX, 27(2):105–13, 2010.

60 V. Sordi, R. Melzi, A. Mercalli, R. Formicola, C. Doglioni, F. Tiboni, G. Ferrari, R. Nano,

K. Chwalek, E. Lammert, E. Bonifacio, and L. Piemonti. Mesenchymal Cells Appearing in Pan-

creatic Tissue Culture Are Bone Marrow-Derived Stem Cells With the Capacity to Improve Trans-

planted Islet Function. Stem Cells, 28(1):140–151, 2010.

R. L. Sorenson and T. C. Brelje. Adaptation of Islets of Langerhans to Pregnancy: β-Cell Growth,

Enhanced Insulin Secretion and the Role of Lactogenic Hormones. Horm Metab Res, 29(06):

301–307, 1997.

A. Stancakova, M. Javorsky, T. Kuulasmaa, S. M. Haffner, J. Kuusisto, and M. Laakso. Changes

in Insulin Sensitivity and Insulin Release in Relation to Glycemia and Glucose Tolerance in 6,414

Finnish Men. Diabetes, 58(5):1212–1221, 2009.

G. R. Stark and W. R. Taylor. Analyzing the G2/M Checkpoint. In Checkpoint Controls and Cancer,

volume 280 of Methods in Molecular Biology, pages 51–82. 2004.

J. C. Stendahl, D. B. Kaufman, and S. I. Stupp. Extracellular Matrix in Pancreatic Islets: Relevance

to Scaffold Design and Transplantation. Cell Transplantation, 18(1):1–12, 2009.

C. Stocking, U. Bergholz, J. Friel, K. Klingler, T. Wagener, C. Starke, T. Kitamura, A. Miyajima,

and W. Ostertag. Distinct classes of factor-independent mutants can be isolated after retroviral

mutagenesis of a human myeloid stem cell line. 8(3):197–209, 1993.

C. Street, Lakey, K. Seeberger, L. Helms, R. Rajotte, A. Shapiro, and G. Korbutt. Heterogenous

expression of nestin in human pancreatic tissue precludes its use as an islet precursor marker.

Journal of Endocrinology, 180(2):213–225, 2004.

M. Szabat, D. S. Luciani, J. M. Piret, and J. D. Johnson. Maturation of Adult β-Cells Revealed

Using a Pdx1/Insulin Dual-Reporter Lentivirus. Endocrinology, 150(4):1627–1635, 2009.

M. Szabat, F. C. Lynn, B. G. Hoffman, T. J. Kieffer, D. W. Allan, and J. D. Johnson. Maintenance of

β-Cell Maturity and Plasticity in the Adult Pancreas: Developmental Biology Concepts in Adult

Physiology. Diabetes, 61(6):1365–1371, 2012.

K. Takahashi, K. Tanabe, M. Ohnuki, M. Narita, T. Ichisaka, K. Tomoda, and S. Yamanaka. Induc-

tion of Pluripotent Stem Cells from Adult Human Fibroblasts by Defined Factors. Cell, 131(5):

861–872, 2007.

J. A. Thomson, J. Itskovitz-Eldor, S. S. Shapiro, M. A. Waknitz, J. J. Swiergiel, V. S. Marshall, and

J. M. Jones. Embryonic Stem Cell Lines Derived from Human Blastocysts. Science, 282(5391):

1145–1147, 1998.

61 F. Thorel, V. Nepote, I. Avril, K. Kohno, R. Desgraz, S. Chera, and P. L. Herrera. Conversion of

adult pancreatic α-cells to β-cells after extreme β-cell loss. Nature, 464(7292):1149–1154, 2010.

G. Touati, F. Poggi-Travert, H. Ogier de Baulny, J. Rahier, F. Brunelle, C. Nihoul-Fekete, P. Czerni-

chow, and J. M. Saudubray. Long-term treatment of persistent hyperinsulinaemic hypoglycaemia

of infancy with diazoxide: a retrospective review of 77 cases and analysis of efficacy-predicting

criteria. Eur J Pediatr., 157(8):628–33, 1998.

N. Tsubooka, T. Ichisaka, K. Okita, K. Takahashi, M. Nakagawa, and S. Yamanaka. Roles of sall4

in the generation of pluripotent stem cells from blastocysts and fibroblasts. Genes to Cells, 14(6):

683–694, 2009.

T. van der Meulen, R. Xie, O. G. Kelly, W. W. Vale, M. Sander, and M. O. Huising. Urocortin

3 Marks Mature Human Primary and Embryonic Stem Cell-Derived Pancreatic Alpha and Beta

Cells. PLoS ONE, 7(12):e52181–, 2012.

B. Viollet, B. Guigas, N. Sanz Garcia, J. Leclerc, M. Foretz, and F. Andreelli. Cellular and molecular

mechanisms of metformin: an overview. Clinical Science (London, England : 1979), 122(6):253–

270, 2012.

P. Wang, N. M. Fiaschi-Taesch, R. C. Vasavada, D. K. Scott, A. Garcia-Ocana, and A. F. Stewart.

Diabetes mellitus - advances and challenges in human β-cell proliferation. Nat Rev Endocrinol, 11

(4):201–212, 2015.

Q. Wang, L. Ye, H. Liu, X. Liu, S. Li, and Z. Chen. Reprogramming of bone marrow-derived

mesenchymal stem cells into functional insulin-producing cells by chemical regimen. American

Journal of Stem Cells, 1(2):128–137, 2012.

L. Warren, P. D. Manos, T. Ahfeldt, Y.-H. Loh, H. Li, F. Lau, W. Ebina, P. K. Mandal, Z. D. Smith,

A. Meissner, G. Q. Daley, A. S. Brack, J. J. Collins, C. Cowan, T. M. Schlaeger, and D. J. Rossi.

Highly efficient reprogramming to pluripotency and directed differentiation of human cells with

synthetic modified mrna. 7(5):618–630, 2010.

J. M. Wells and D. A. Melton. Vertebrate Endoderm Development. Annu. Rev. Cell Dev. Biol., 15

(1):393–410, 1999.

P. O. Wiebe, J. D. Kormish, V. T. Roper, Y. Fujitani, N. I. Alston, K. S. Zaret, C. V. E. Wright,

R. W. Stein, and M. Gannon. Ptf1a Binds to and Activates Area III, a Highly Conserved Region of

the Pdx1 Promoter That Mediates Early Pancreas-Wide Pdx1 Expression. Molecular and Cellular

Biology, 27(11):4093–4104, 2007.

62 M. E. Wilson, D. Scheel, and M. S. German. Gene expression cascades in pancreatic development.

Mechanisms of Development, 120(1):65–80, 2003.

S. Yamanaka. Induced Pluripotent Stem Cells: Past, Present, and Future. Cell Stem Cell, 10(6):

678–684, 2012.

P. Yi, J.-S. Park, and D. Melton. Betatrophin: A Hormone that Controls Pancreatic β Cell Prolif-

eration. Cell, 153(4):747–758, 2013.

J. Yu, M. A. Vodyanik, K. Smuga-Otto, J. Antosiewicz-Bourget, J. L. Frane, S. Tian, J. Nie, G. A.

Jonsdottir, V. Ruotti, R. Stewart, I. I. Slukvin, and J. A. Thomson. Induced pluripotent stem cell

lines derived from human somatic cells. Science, 318(5858):1917–1920, 2007.

C. Zanini, S. Bruno, G. Mandili, D. Baci, F. Cerutti, G. Cenacchi, L. Izzi, G. Camussi, and M. Forni.

Differentiation of Mesenchymal Stem Cells Derived from Pancreatic Islets and Bone Marrow into

Islet-Like Cell Phenotype. PLoS ONE, 6(12):e28175, 2011.

H. Zhang, W.-P. Wang, T. Guo, J.-C. Yang, P. Chen, K.-T. Ma, Y.-F. Guan, and C.-Y. Zhou. The

LIM-Homeodomain Protein ISL1 Activates Insulin Gene Promoter Directly through Synergy with

BETA2. Journal of Molecular Biology, 392(3):566–577, 2009.

H. Zhang, S. Gayen, J. Xiong, B. Zhou, A. Shanmugam, Y. Sun, H. Karatas, L. Liu, R. Rao, S. Wang,

A. Nesvizhskii, S. Kalantry, and Y. Dou. MLL1 Inhibition Reprograms Epiblast Stem Cells to

Naive Pluripotency. Cell Stem Cell, page ePub, 2016.

63 Chapter 2

A Novel Source of Mesenchymal

Stem Cells derived from the

Pancreas of Patients with

Congenital Hyperinsulinism.

Sophie G Kellaway, Karolina Mosinska, Zainaba Mohamed,

Indi Banerjee, Alexander Ryan, Karen E Cosgrove and Mark J Dunne

64 2.1 Abstract

Diabetes and congenital hyperinsulinism (CHI) are severe diseases affecting the pancreas. Current models for testing drugs to treat these diseases are in vivo in rodents or isolated rodent islets. Differ- ences between the human and rodent pancreas mean in vitro human models are needed. Developing islets from mesenchymal stem cells (MSCs) has shown promise but the final cells produced are not mature and MSCs show limited replicative capacity. In this study we have derived MSCs from the pancreas of patients with CHI. The pancreas of neonatal patients with CHI have greater prolifera- tion, plasticity and insulin secretion than from healthy counterparts. MSCs from these patients have the typical cell surface markers CD29, CD44, CD73, CD90 and CD105, mesenchymal cytoskeletal proteins Vimentin, Nestin and α-smooth muscle actin, and differentiation capacity to osteoblasts, chondrocytes and adipocytes. The MSCs also expressed a number of transcription factors associ- ated with pancreatic development, including ISLET1, a unique marker of pancreas-derived MSCs.

Pancreatic β-cell related genes that were not expressed, including PDX1 showed decreased promoter methylation compared to MSCs derived from bone marrow suggesting a possible ‘epigenetic mem- ory’. We have demonstrated here the feasibility of deriving pancreatic MSCs from patients with CHI which maintain some features of the pancreas. Re-differentiation of these cells to β-cells may provide a valuable new resource to study CHI in vitro.

65 2.2 Introduction

Diabetes is a chronic disease affecting nearly 10% of adults worldwide (Danaei et al., 2011) and cannot currently be cured. Cadaveric islet transplantation offers a long term treatment or potential cure by increasing β-cell mass (Ryan et al., 2005, Shapiro et al., 2000) but as a treatment is limited by lack of donors. Consequently, alternative sources of islets for cell therapies have been investi- gated, including derivation from human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs). HESCs and iPSCs do not typically have the epigenetic profile associated with mature

β-cells when differentiated, unless the iPSCs are derived from pancreatic tissue originally (Bar-Nur et al., 2011) and so we have considered stem cells from the pancreas.

Multiple putative stem cells have been isolated from the adult pancreas and differentiated to

β-like or islet-like cells; initially from rodents (Zulewski et al., 2001) and later humans. This includes proliferative ductal structures (Corritore et al., 2014), de-differentiated endocrine (Russ et al., 2009,

Joglekar et al., 2009) and exocrine tissue (Lima et al., 2013) which resemble mesenchymal stem cells (MSCs) and de-differentiated endocrine tissue which remains epithelial (Lima et al., 2013).

Additionally, MSCs from bone marrow and adipose tissue have also been differentiated to insulin secreting cells discussed by Bhonde et al. (2014). It can be hypothesised that if these cells can be re-differentiated in vitro then it can also take place in vivo - a process known as β-cell neogenesis.

Whilst hESCs and iPSCs may offer an exogenous source of islet cells for transplantation, somatic tissue stem cells can potentially provide a model to investigate neogenesis for allogeneic islet cell generation. However, pancreatic MSCs have proven challenging as a model due to slow growth, early senescence at around 30 population doublings (Hu et al., 2003, Carlotti et al., 2010) and limited insulin release by re-differentiated cells (Bhonde et al., 2014).

The exact origins of MSCs from the pancreas is unclear, whether they are tissue resident such as CD90/CD105 double positive cells (Carlotti et al., 2010) or de-differentiated cells. Numerous experiments have shown that they can de-differentiate from various cell types. When placed into culture, endocrine and non-endocrine cells produce similar MSCs, with lineage tracing being re- quired to reveal the origins of MSCs from mixed pancreas digests (Russ et al., 2009). Pancreatic

MSCs can be readily derived from exocrine pancreas, demonstrated with lineage tracing (Lima et al.,

2013). Exocrine cells are a useful source when only a small portion of tissue is available, such as from neonates, as the exocrine pancreas comprises over 95% of the pancreas, or when the islets are required for other in vitro studies. It was shown by Lima et al. (2016) that exocrine-derived MSCs can be readily differentiated to functional insulin secreting cells with a transcription factor regimen, suggesting plasticity or transdifferentiation from these cells is possible. Even after extended in vitro

66 culture these exocrine-derived MSCs were able to undergo mesenchymal-epithelial transition, high- lighting the flexibility of this in vitro cell phenomenon (Muir et al., 2015).

We hypothesised that MSCs could be derived from the exocrine pancreas of patients with con- genital hyperinsulinism (CHI). If these MSCs maintain the phenotype of CHI, this would be useful for modelling both CHI and other pancreatic disorders. There are three forms of CHI - focal, diffuse and atypical (also known as localised islet cell nuclear enlargement, LINE). Focal CHI is caused by a recessive mutation in the ABCC8 gene (which encodes the SUR1 subunit of the pancreatic

ATP-sensitive potassium (KATP) channel), where loss of heterozygosity leads to no functional allele and non-functional channels (Verkarre et al., 1998, Glaser et al., 1999, Fournet et al., 2001). Diffuse

CHI can be caused by a number of mutations (Glaser et al., 2000), including ABCC8 (Dunne et al.,

1997), but is homozygous and so affects the whole pancreas. Atypical CHI typically has a later onset than focal or diffuse CHI, is not caused by any known mutation (screening of the genes associated with focal and diffuse CHI excludes these, but some post-zygotic mutations have now been reported at meetings) and affects the whole pancreas (Arnoux et al., 2011). All three forms of CHI exhibit high levels of insulin secretion (Dunne et al., 2004) and hyperproliferation (Salisbury et al., 2015,

James et al., 2009). Increased proliferation would mean the cells are easier to work with than other

MSC types, high levels of insulin secretion would be beneficial for high-throughput screening of novel putative drug treatments for diabetes or CHI and as the cells are pancreatic they may maintain a pancreatic epigenetic profile. To explore these hypotheses, in this study we aimed to derive and characterise MSCs from the pancreas of patients with CHI.

67 2.3 Methods

All reagants were from Sigma-Aldrich or Fisher-Scientific.

2.3.1 Derivation of CHI pancreatic mesenchymal stem cell (CHI pMSC) lines

Patient tissue was received from surgeons with ethical permission and full consent from the patients’ parents or legal guardians. Tissue was rinsed in Krebs-Ringer-Hepes buffer (KRH buffer comprised:

129 mM sodium chloride, 5 mM sodium bicarbonate, 4.8 mM potassium chloride, 1.2 mM potassium orthophosphate, 1.2 mM magnesium sulphate, 10 mM HEPES, 2.5 mM calcium chloride, 5.6 mM

D-glucose, 0.1% bovine serum albumin (BSA) and the pH was adjusted to 7.4) to remove excess blood. The tissue was first digested with 0.75 mg/ml Liberase (Roche, Mannheim, Germany) in

KRH buffer with 1.5 mg/ml egg white trypsin inhibitor (Sigma-Aldrich, Irvine, UK) and 1.5 mg/ml soybean trypsin inhibitor (Gibco, Paisley, UK), for 3 minutes at 37°C with agitation, followed by

1 minute of vigorous shaking, and this was done three times. Ice cold KRH buffer was added and the digestion mixture centrifuged for 1 minute at 150 x g. The secondary digestion was with 0.5 mg/ml Liberase in KRH buffer with trypsin inhibitors, for 3 minutes at 37°C with agitation, followed by 1 minute of vigorous shaking and this was done twice. Ice cold KRH buffer was added and the digestion mixture centrifuged for 1 minute at 150 x g, this was done 4 times to ensure the preparation was as clean as possible. Where possible islets were removed from the mixture under a dissection microscope but due to the age and pathology of patients, islets were not clearly delineated. The resulting digestion mixture was transferred to 6-well tissue culture treated plate in RPMI 1640 5.5 mM glucose (0 mM glucose:RPMI 1640 11 mM glucose; 50:50; Gibco/Sigma-Aldrich) with 10% foetal bovine serum (FBS, Gibco), around 1 well per 20 mg wet weight tissue received. After 48 hours, cells and tissue which had not adhered were removed and media was changed thereon every 48 hours until 70% confluency was reached at which point the cells underwent the first passage.

2.3.2 Cell culture

Cells were routinely cultured on tissue culture plastic (plates or vented cap flasks, Corning, Schiphol-

Rijk, Netherlands) coated with human fibronectin (Chemicon, Merck Millipore, Nottingham, UK) to 0.5 µg/cm2 and vitronectin (Life Technologies, Paisley, UK) to 0.1 µg/cm2, the reasoning behind these growth conditions is shown and explained in Supplementary Information, Figure 2.8. Cells were cultured in RPMI-1640 5.5 mM glucose (mixed from 11 mM glucose RPMI, Sigma-Aldrich and

0 mM glucose RPMI, Gibco, Life Technologies to 5 mM) with 10% FBS. Cultured cells were kept in

68 a humidified incubator at 37°C, 5% CO2. Subculturing/passage was performed when cells reached approximately 70% confluency - cells were washed with Dulbecco’s phosphate buffered saline (dPBS), and detached from tissue culture plastic with 0.05% trypsin-EDTA (Sigma-Aldrich) for 3 minutes.

Trypsinisation was halted using 1 mg/ml soybean trypsin inhibitor (Gibco). Cells were pelleted at

50 x g and transferred to new vessels at a cell line and condition specific density, between 1:2 and

1:5. Medium was changed every 48 hours. Brightfield images were taken on an Olympus CKX41 microscope using 4x and 10x objectives and captured with QCapture Pro software. All experiments were performed on cells between passages 3 and 16. Total number of population doublings was estimated from number of passages and split ratios.

2.3.3 Karyotyping

Three days after seeding the cells, culture medium was changed and 20 ng/ml epidermal growth factor (Calbiochem, Merck Millipore), 20 ng/ml fibroblast growth factor (Peprotech, Rocky Hill,

USA) and 100 ng/ml Colcemid (Karyomax, Gibco) was added. At least 12 hours later cells were trypsinised as above and put into hypotonic solution, comprising 0.0375M potassium chloride (Fisher

Scientific, Loughborough, UK), for 5 minutes at 37°C. The cells were pelleted at 180 x g, then fixed using ice cold 3:1 methanol/acetic acid solution, added in a drop-wise fashion. The cells were pelleted again and more fixative added, this process was repeated one more time and the resulting solution frozen. G-banding and analysis was then performed by TDL genetics (The Doctor’s Laboratory,

London).

2.3.4 Short tandem repeat (STR) profiling

Cells were lysed and DNA amplified using a PowerPlex 16 HS system (Promega, Madison, USA), the samples were analysed on a 3730 DNA analyser (Applied Biosystems, California, USA) using

POP-7 polymer (Life Technologies). The number of repeats at each locus was then called using the

GeneMapper 4.0 software (Life Technologies). The PowerPlex 16 HS system is a multiplex PCR based assay which amplifies the 13 combined DNA index system loci (a set of nucleotides which have high copy-number variation, used in forensic applications for identifying individuals), two control low-stutter pentanucleotide markers and amelogenin for sex determination.

2.3.5 RT-PCR and qRT-PCR

Total RNA was extracted using an RNeasy Mini Kit (Qiagen, Crawley, UK) with on column DNase

1 (Qiagen) genomic DNA digestion according to the manufacturer’s instructions and quantified using a NanoDrop 1000 spectrophotometer. cDNA was synthesised from 500 ng RNA using a nanoScript

2 reverse transcription kit (Primer Design, Chandler’s Ford, UK) with oligo-dT primers and random

69 nonamer primers. RT-PCR was carried out using a Taq DNA Polymerase kit (Invitrogen, Life

Technologies), 1.5mM magnesium chloride and 2.5pmol of each primer with conditions as detailed in Table 2.1. PCR cycling conditions were 94°C for 5 minutes followed by 33 cycles (22 cycles for

18S rRNA, 38 cycles for INS) of 94°C for 30 seconds, annealing temperature as detailed in Table 2.1 for 30 seconds and 72°C for 90 seconds, with a final 72°C extension of 2 minutes. RT-PCR products were run on a 2% agarose gel stained with GelRed (Biotium, California, USA) and visualised on a Gel Doc XR system (Bio-Rad, Hemel Hempstead, UK). PCR images had the colour inverted in

IrfanView, and where necessary brightness and contrast were adjusted, to ensure that fainter bands visible on the transilluminator would be visible when printed. qRT-PCR was carried out using power

SYBR green PCR master mix (Life Technologies) and 2.5 pmol of each primer as detailed in Table

2.1 and run on a StepOnePlus Real-Time PCR System (Life Technologies). For qRT-PCR, the gene of interest CT values were normalised to the geometric mean of 2 reference genes, 18S rRNA and

GAPDH (∆CT), following which the sample comparisons were done (∆∆CT). Statistical significance was calculated by one way ANOVA with Bonferroni post-hoc test if appropriate. Bone marrow mesenchymal stem cell (bmMSC) mRNA was gratefully received from Dr Stephen Richardson, all procedures were the same. Primers were designed using Primer-BLAST (NCBI) and synthesised by Eurofins (Wolverhampton, UK). Specificity of the primers was assessed by BLAST (NCBI) and fragment size, optimal annealing temperatures were determined by temperature gradient PCR to ensure only one fragment was present and the most intense band present. QRT-PCR primers had an efficiency between 85 and 115% confirmed by serial dilution of standards.

2.3.6 Genotyping

Genomic DNA was extracted from cells using a PureLink genomic DNA kit (Invitrogen). PCR was carried out as Section 2.3.5, for the affected exon as determined in the patient by clinicians.

PCR products were purified using a QIAquick PCR purification kit (Qiagen). Products were then sequenced, using 15 ng of purified PCR product, 4 pmoles of each primer used previously (forward and reverse sequencing carried out separately) with the BigDye Terminator v3.1 cycle sequencing kit (Applied Biosystems) on a 3730 DNA analyser (Applied Biosystems) using POP-7 polymer (Life

Technologies).

Gene Sequence (5’ - 3’) Annealing tem- Product

perature (°C) size (base

pairs)

18S rRNA F - GTAACCCGTTGAACCCCATT 60 150

R - CCATCCAATCGGTAGTAGCG

70 Gene Sequence (5’ - 3’) Annealing tem- Product

perature (°C) size (base

pairs)

ABCC8 exon F - CCTATTTGCAGAGGATGTGAC 60 454

6* R - GATACTGCTATGGGCTTTGC

ABCC8 exon F - CCATGCACACATTTTCCAAC 60 325

37* R - ATCCCACTAAACCCTTTCCAA

ARX F - CTCCAACATCCACTCTCTCT 59 257

R - CTATCTTACAGGCTCGCAT

CDH1 F - CACCAGCCTCCTTTTTATTT 58 479

R - CTTTTCATAGTTCCGCTCTG

CK19 F - CGATGTGCGAGCTGATAGTG 58 258

R - TTGGCAGGTCAGGAGAAGA

COL2A1 F - CTGCATGAGGGCGCGGTA 62 384

R - CAGTGGCGAGGTCAGTTGG

FOXA2 F - CCGTTCTCCATCAACAACCT 59 325

R - GACCCCCACTTGCTCTCTC

GAPDH F - ATTGCCCTCAACGACCACTT 60 95

R - GGTCCACCACCCTGTTGC

GCG F - TTCTGAGGCCACATTGCTTT 59 289

R - CTGTGGCTACCAGTTCTTC-

TATTCT

INS F - ACCAGCATCTGCTCCCTCTA 60 114

R - GGTTCAAGGGCTTTATTCCA

ISL1 F - TATTTTGCCACAAGCGTCTC 62 282

R - TTCAAAGACCACCGTACAACC

ISL1 (qRT- F - GCAGCCCAATGACAAAACTAA 60 78

PCR) R - CCGTCGTGTCTCTCTGGACT

MAFB F - CGAGAGAGACGCCTACAAGG 59 237

R - CCCTCTCGCTCAAGTCAAAC

MYT1 F - AAGTGCGTTTTCTGTTTTA 58 419

R - TTTACCCTCTGACCTTGTG

NES F - AAGGTGAAAAGGGGTGTGG 62 95/103

R - ATGAGATGGAGCAGGCAAG

71 Gene Sequence (5’ - 3’) Annealing tem- Product

perature (°C) size (base

pairs)

NEUROD1 F - ACCAGCCCTTCCTTTGATGG 60 131

R - AGTGTCGCTGCAGGATAGTG

NGN3 F - CGAATGCACGACCTCAAC 59 125

R - AGTCAGCGCCCAGATGTAGT

OCLN F - CTCTGAAGTGAAACTGCTTG 55 136

R - AAAAATAGGGAGGCTGGTAG

PAX6 F - AAAAATCCCCAAAAGCAAAT 59 245

R - AGGCTGACACTGAACAAAAC

PDX1 F - AGTGATACTGGATTGGCGTTG 59 138

R - TAGGGAGCCTTCCAATGTGT

PPARG2 F - GCAAACCCCTATTCCATGCTG 59 355

R - GCAGGCTCCACTTTGATTGC

SST F - AGGATGAAATGAGGCTTGA 55 191

R - GGAGGATTAGGGAAGAGAGA

SOX9 F - AGCGAAATCAACGAGAAACT 60 222

R - ATCCCCTCAAAATGGTAATG

SOX17 F - CGCACGGAATTTGAACAGTAT 59 182

R - GGATCAGGGACCTGTCACAC

SPP1 F - TCCTAGCCCCACAGACCCTT 59 391

R - AACTCCTCGCTTTCCATGTGT

VIM F - GTGAAATGGAAGA- 58 274

GAACTTTGC R - GTATCAACCA-

GAGGGAGTGAAT

Table 2.1: Primers used for RT-PCR, details on design and use are in Section 2.3.5. Primers indicated with * from Ellard et al. (2007)

2.3.7 Adipogenesis, osteogenesis and chondrogenesis

For adipogenesis and osteogenesis, cells were seeded in duplicate at 1 x 104 cells/cm2 on uncoated plastic (six-well plate) and incubated for 4 days in standard growth medium (RPMI with 10% FBS).

On day 4, cells were switched to StemPro adipogenesis or osteogenesis complete media (Gibco) with one well being kept in standard growth media as a negative control. For chondrogenesis cells were seeded as a micromass culture - 8 x 104 cells in a 5 µl droplet in a six-well plate was allowed to

72 adhere for 2 hours, after which StemPro chondrogenesis complete medium (Gibco) or RPMI 1640 with 10% FBS was added. Medium was changed every three days. After 14 days for adipogenesis and chondrogenesis or 21 days for osteogenesis in these conditions, cells were harvested for RT-PCR as above or were were fixed for 30 minutes in 4% paraformaldehyde and washed in dPBS. To confirm adipogenesis, cells were stained for lipid droplets - a 5 minute incubation with 60% isopropanol was performed to reduce non-specific stains, followed by a 5 minute incubation in oil red O (0.3% w/v in

60% isopropanol; Sigma-Aldrich). This was washed off in distilled water and counterstained for 60 seconds with Mayer’s haematoxylin (Sigma-Aldrich), which was in turn washed off in distilled water.

To confirm osteogenesis, cells were stained for calcium deposits with a 3 minute incubation in Alizarin

Red S solution (2% w/v in distilled water, pH 4.2; Alfa Aesar, Heysham, UK) which was washed off in distilled water. Chondrogenesis was confirmed by staining for sulphated proteoglycans with

Alcian Blue solution (1% w/v in 0.1 N HCl; Alfa Aesar) for 30 minutes and rinsing in distilled water.

Lipid droplets or calcium deposits stained red and proteoglycans stained blue were observed on an

Olympus CKX41 microscope using 10x and 40x objectives and images captured using QCapture Pro.

2.3.8 Flow Cytometry

Cells were harvested by trypsinisation, spun and resuspended in dPBS. A cell count was performed, the cells were spun again and then resuspended to 1 x 106 cells/ml in PBS, 10% normal goat serum

(NGS, Sigma-Aldrich) and 0.1% sodium azide for a 15 minute incubation on ice to prevent non- specific binding. The cells were divided such that there were 1x105 cells per condition required and neat or diluted primary antibodies added to the final concentration listed in Table 2.2 for 30 minutes at room temperature. Cells were spun at 150 x g and antibody solution removed, 3 x 500 µl dPBS washes were performed, with spins to remove the solution from the cells. Cells were resuspended in 100 µl dPBS with 3% NGS and diluted secondary antibodies added to the final concentration in

Table 2.2 for 30 minutes at room temperature. Cells were spun at 150 x g and antibody solution removed, 3 x 500 µl dPBS washes were performed, with spins to remove the solution from the cells.

Cells were resuspended in 500 µl dPBS with 3% NGS and analysed on a Beckman Coulter Cyan

ADP with 635nm excitation. Gating was performed using Summit V4.3, based on forward scatter and side scatter of unlabelled control cells to exclude debris (low forward scatter), dead cells (low forward scatter, high side scatter) and doublets/cell clumps (high forward scatter). Markers were compared to the relevant isotype control.

2.3.9 Immunocytochemistry (ICC)

Cells were cultured on glass coverslips until of a suitable density and were fixed in 4% paraformalde- hyde or 4% paraformaldehyde/0.025% glutaraldehyde (for cytoskeleton proteins) for 20 minutes.

73 Cells were permeabilised in 0.3% Triton x-100 (Sigma-Aldrich) or in ice cold methanol (for cy- toskeleton proteins) and non-specific staining was prevented by incubation in 10% NGS in PBS

(with 0.3M glycine for cytoskeleton proteins). Primary antibodies were applied as Table 2.2 for 1 hour in 0.1% Triton x-100, 3% NGS in PBS and washed off in PBS/0.1% Tween-20 (Sigma-Aldrich).

Fluorophore-conjugated secondary antibody, specific to the species the primary antibody was raised in, was applied as Table 2.2 in 0.1% triton x-100, 3% NGS in PBS for 1 hour. Cells were washed in

PBS/0.1% Tween-20, then distilled water and allowed to dry. Coverslips were mounted using Pro

Long Gold antifade reagent (Life Technologies) with DAPI. Slides were viewed using an Olympus

BX51 upright microscope using 20x 0.50 UPlanFLN, 40x 0.75 UPlanFLN or 60x 1.25 UPlanFLN

(Ph 3) objectives and captured using a Coolsnap EZ camera (Photometrics, USA) through MetaVue

Software (Molecular Devices, USA). Filter sets for DAPI (31000v2), FITC (41001) and Cy3 (41007a) were used to prevent bleed through of colour channels whilst maximising signal. Images were pro- cessed and analysed using ImageJ (http://rsb.info.nih.gov/ij).

Antigen Species Concentration Manufacturer (µg/ml) α-smooth mus- Rabbit 0.5 Abcam (Cambridge, UK) cle actin CD29 Mouse 10 Abcam CD44 Mouse 1 Abcam CD45 Rabbit 2 Abcam CD73 Mouse 10 Miltenyi (Surrey, UK) CD90 Mouse 1 Abcam CD105 Mouse 10 Miltenyi E-Cadherin Mouse 1.66 BD Biosciences (Oxford, UK) IgG Rabbit Variable AbD Serotech (Oxford, UK) IgG1 Mouse Variable Cell Signalling Technology (Leiden, The Netherlands) IgG2a Mouse Variable Fisher Scientific Islet1 Rabbit 5 Abcam MafB Mouse 10 R&D (Abingdon, UK) Nestin Mouse 5 Millipore Occludin Mouse 10 R&D Pax6 Rabbit 5 Abcam Sox9 Rabbit 5 Millipore Vimentin Mouse 5 Abcam Cy2 anti- Goat 5 Jackson/Abcam rabbit Cy3 anti- Goat 5 Abcam mouse Cy5 anti- Goat 2.5 Eurogentec mouse Cy5 anti- Goat 2.5 Abcam rabbit

Table 2.2: Antibodies used in the study

74 2.3.10 Bisulfite sequencing

Genomic DNA was extracted from cells using a PureLink genomic DNA kit (Invitrogen). 1 µg of genomic DNA was bisulfite converted using the EpiTect fast DNA bisulfite kit (Qiagen) according to the manufacturer’s instructions. PCR was carried out as Section 2.3.5 with primers and temper- atures detailed in Table 2.3. PCR primers were designed with MethPrimer (Li and Dahiya, 2002).

BmMSC DNA was gratefully received from Dr Stephen Richardson, it had been previously extracted with the QIAamp DNA Mini Kit but all subsequent procedures were identical and carried out at the same time. PCR products purified with a Qiaquick PCR purification kit (Qiagen) according to the manufacturer’s instructions. Products were then sequenced, using 15 ng of purified PCR product, 4 pmoles of each primer used previously (forward and reverse sequencing carried out separately) with the BigDye Terminator v3.1 cycle sequencing kit (Applied Biosystems) on a 3730 DNA analyser

(Applied Biosystems) using POP-7 polymer (Life Technologies). Percentage methylation was deter- mined by measuring the peak heights of the cytosine or thymine for the forward trace, or adenine or guanine for the reverse trace on ChromasLite (Technelysium, Australia), with the proportion of cyto- sine or guanine (methylated) to total cytosine/guanine plus thymine/adenine being calculated. Data was visualised with Methylation plotter (Mallona et al., 2014) with statistical significance calculated within this program by the Kruskal-Wallis test.

Gene Sequence (5’ - 3’) Annealing Genomic loca- Product tempera- tion size ture (°C) (base pairs) PDX1 F - TTTGGTTTTTTGTGGTT- 64 27918743 to 226 TAAGTTTT R - AACCACTTTTTC- 27918969 TACACCCACTAAC AMY2A F - GTTTTTGGAAA- 61 103616149 to 292 GAAGTTGTTTTTGA R - CCC- 103616441 TACCCTATTCTATTTCTCCCTA CK19 F - TTTAATATTTTTGTG- 61 41529473 to 442 GTTGAATGAA R - TCCCTAT- 41529031 TAAAAACCCCTTACTTAC SPP1 F - GGTGGGTTGGGTAGTGGTA- 62 87975604 to 268 GAAAAT R - TTTTCCTTAAAATT- 87975872 TAAACTTCCCCATAAA INS F - TGTGAGTAGGGATAGGTTTG- 64 2161202 to 266 GTTAT R - AAAAACTAAAAACTAC- 2161468 TAAACCCCC

Table 2.3: Primers used for bisulfite sequencing

75 2.4 Results

2.4.1 MSCs were derived from three CHI patients

Cell lines were derived from three patients with CHI, the details of which are summarised in Table

2.4. The ages of the patients varied and they were from all three major disease backgrounds.

Cell line Age at Disease form Mutation Number of Number of surgery population passages doublings F-CHI 4 months Focal CHI ABCC8 exon 6 (het- 46 16 erozygous) A-CHI 17 months Atypical CHI Unknown 106 24 D-CHI 11 weeks Diffuse CHI ABCC8 exon 37 60 16

Table 2.4: Details of cell lines and patients

The cells discussed herein were initially identified by their morphology in culture. Between two and four days after the operation tissue had adhered to the plastic, following which cells were seen to be growing out around the tissue. Within two days of adherence, the remaining tissue was lost, either through the cells having moved into a monolayer form, or through the cells in the tissue having died and been removed with media changes.

The cells were maintained until they reached approximately 70% confluency. Some flexibility was required for the primary culture as there was not a uniform monolayer as seen at later stages, but rather cells were growing in islands based on where chunks of tissue had adhered. The different cultures also took different amounts of time to reach confluency - F-CHI took 12 days, A-CHI took

7 days and D-CHI took 14 days, this may reflect the starting number of cells which may therefore impact the number of population doublings we observed, which varied from approximately 50 to 100 across the three lines. At this stage cells were passaged ‘P1’ and the cell lines began to proliferate more uniformly until senescence which began after 16 passages in F-CHI and D-CHI and 24 passages in A-CHI. The cell lines had a similar and stable mesenchymal appearance throughout culture as shown in Figure 2.1, panel A.

The cells were initially identified as MSCs as detailed above by the characteristic morphology and plastic adherence but it was important to confirm that they were authentic MSCs. Authentic MSCs must express (or not express) a panel of specific cell surface markers and be able to differentiate to multiple mesodermal lineages (Dominici et al., 2006).

76 Figure 2.1: Derived cell lines showed characteristic features of MSCs (A) Brightfield images show a similar plastic adherent, mesenchymal morphology in each cell line, images shown are from 2 days after passaging (B) Flow cytometry showed the cells to be positive for CD29, CD44, CD73, CD90 and CD105 vs isotype control and negative for CD45. 1 x 104 cells were counted in each condition. Graphs are representative for between 1 and 4 experiments each. i is A-CHI, ii is D-CHI and iii is F-CHI. 77 We tested these characteristics in all three cell lines and found similar results. All three cell lines were negative for CD45 when tested by flow cytometry, a marker of haematopoeitic stem cells confirming no cross contamination with blood. This was important to exclude as the tissue post- surgery contains blood. Further, as shown in Figure 2.1, all three were positive for CD29, CD44,

CD73, CD90 and CD105. Each of these markers alone is insufficient to identify the cell type, but the combination is highly specific for MSCs giving further evidence that we had successfully isolated

MSCs from the patients’ pancreata. Authentic MSCs should be >95% positive for CD73, CD90 and

CD105 and <2% positive for CD45 (Dominici et al., 2006), which our cells were - the percentage of cells positive for each marker is shown in Figure 2.5.

Marker F-CHI A-CHI D-CHI CD29 84.3±4.4 93.7 77.2 CD44 95.1 88.06±10.6 95.2±4.0 CD45 2.21 1.9±0.7 1.9±1.0 CD73 97.6 96.1 99.7±0.04 CD90 81.0 96.7 94.6±3.1 CD105 98.7 99.9 99.8

Table 2.5: CD marker expression: Average percentage of cells expressing CD markers relative to isotype control is given, with standard deviation. Markers were assessed by flow cytometry at passages 9, 11 and 12 in F-CHI, 5, 11, 14 and 16 in D-CHI and 9, 12 and 14 in A-CHI

The average total cells expressing each CD marker is shown in Table 2.5. These are the results from between one and four experiments, in the case of CD29, CD73 and CD105 there are only one to two experiments for each cell line as the first antibodies used (all from Abcam) did not work well. For CD90, results were varied, with results showing anything from fully negative to >95% of cells expressing the antigen, despite cells being identical - this may have been due to the aliquots of antibody, further repeats using a new antibody may be helpful in this case.

One of the defining characteristics of MSCs is multipotency, which defines their ‘stemness’. This is assessed by the ability of the MSCs to differentiate to three mesoderm derived mature cell types

- adipocytes, osteoblasts and chondrocytes. The differentiated cells are normally assessed by a phenotypic assay that can be seen using histological stains (Dominici et al., 2006, Marion and Mao,

2006). Mature osteoblasts deposit calcium as in bone, mature adipocytes swell and contain lipid droplets as their function is to store fat, mature chondrocytes secrete proteoglycans in order to form connective tissues (Marion and Mao, 2006). Again, similar results were seen for all three cell lines. There was widespread positive staining seen for sulfated proteoglycans in all micromass chondrogenesis cultures and for calcium in the osteogenesis cultures. Only rare positive stains were

78 Figure 2.2: Derived cell lines showed multipotency, characteristic of MSCs (A) Histo- logical stains were done for differentiated osteoblasts (alizarin red), chondrocytes (alcian blue) and adipocytes (oil red) in all three cell lines after differentiation at passage 14 in D-CHI, 16 in A-CHI and 7 in F-CHI, and in control cells cultured in standard media, for the undifferentiated A-CHI only is shown, representative for all three. (B) shows further validation of the differentiation, where RT-PCR for a key marker gene was performed for each lineage in F-CHI P7 only.

79 seen in the adipogenesis cultures, <1% of cells contained lipid droplets, but those which did were packed full of lipid droplets. No positive stains were seen in the controls for calcium or lipids, slight staining was seen throughout the controls for the proteoglycan staining, but this was less intense than in the differentiated cells. Figure 2.2 shows images of each of the stains, with the positive cells being shown for adipogenesis rather than a representative field of the mostly negative stains. To confirm that what we were seeing was differentiation to the lineage, RT-PCR was performed for a marker of each lineage. We felt this was necessary, as proprietary kits were used so there was a chance that they were inducing formation of calcium deposits, lipid droplets or proteoglycan deposition without the cells being actually differentiated. The markers used were seen in F-CHI and so only histological stains were used for the other two cell lines. The three CHI derived cell lines showed plastic adherence, specific cell surface markers and the ability to differentiate to two of three cell types, and potentially the third (adipocytes). Together with the flow cytometry, these data confirm that the cells we had derived were MSCs.

One of the defining characteristics of MSCs, as suggested by the name is that they are mesenchy- mal, not epithelial. Most mature cells in the body, including islet cells, are epithelial. Mesenchymal cells are seen primarily in connective tissue and during development, and are characterised by an ab- sence of junction proteins and the presence of certain cytoskeletal components. We therefore wanted to investigate the mesenchymal or epithelial phenotype of the cells. The cell lines were assessed for gene and protein expression of a panel of markers of epithelial and mesenchymal cells as shown in

Figure 2.3.

Strong expression was seen for all three mesenchymal genes, and also for OCLN which is part of tight junctions, and at a much lower level, CDH1 which encodes E-cadherin, part of adherens junc- tions. The immunostaining showed similar results, clear expression was seen of the mesenchymal cytoskeleton proteins, with the characteristic localisation of the proteins. Similarly tight junction proteins were not clearly seen in the cells showing that they are not epithelial, although there was some signal for Occludin. The fact that expression of the genes encoding junction proteins E-Cadherin and Occludin is seen was unexpected but may suggest that the cells are able to switch to an epithelial phenotype given the necessary signals.

80 Figure 2.3: Mesenchymal proteins were expressed in CHI pMSCs Markers were assessed between passages 9 and 14 in all three lines to confirm if cells are epithelial or mesenchymal by (A) RT-PCR for 3 mesenchymal markers and 2 epithelial markers, mesenchymal genes NES, VIM and ACTA2 were present in all lines, epithelial markers OCLN and CDH1 were also present. (B) The mesenchymal markers were also present as protein in the characteristic cytoskeleton form Nestin and Vimentin shown in D-CHI and α-smooth muscle actin shown in A-CHI; epithelial marker E-Cadherin was not present at all as protein shown in F-CHI. Immunostaining images are representative for all three lines, all scale bars are 50 µm. Rabbit and mouse panels show secondary antibody only stains. (C) shows positive controls for the antibodies, wherein western blot was used to confirm that the antibodies bind at the correct size to cells known to express the target (HeLa, A549 and MCF7s were used).

81 Figure 2.4: CHI pMSCs are genetically stable and unique(A) G-band karyotypes of each cell line, chromosomes from 1 cell are shown, representative of 19-20 metaphase spreads from passage 10 in D-CHI, 12 in A-CHI and 9 in F-CHI. (B) DNA sequencing of the ABCC8 gene showed F-CHI to have both adenine and guanine at the indicated base, showing heterozygosity, and D-CHI to have just an adenine indicating homozygosity (C) STR profiles were unique for each cell line, the number of repeats at each locus in each cell line is given, assessed at passage 3 and 16 in D-CHI, 4 in A-CHI and 4 in F-CHI.

82 2.4.2 Derived cells were karyotypically stable, unique and contained the CHI causing mutations

To confirm that the cells were unique and specific to the patients, STR profiling was carried out.

This gave the unique fingerprint at 16 loci for each patient and therefore each cell line. Each STR profile was compared to the other cell lines and the ATCC catalogue, which includes STR profiles for all banked cell lines. No matches over 80% were seen to lines from the ATCC catalogue across the 8 loci which are free to compare, where 80% signifies a match (Capes-Davis et al., 2013). The number of repeats at each locus is shown in Figure 2.4 panel C.

Additional verification was through sequencing of the ABCC8 gene. As part of the clinical diag- nosis of CHI the full ABCC8 gene (among other genes known to cause CHI) is sequenced to identify the mutation responsible for the disease. We amplified and sequenced the exon reported by the hos- pital and compared this to the information given to verify the cells’ origins. The affected nucleotide and surrounding sequence are shown in Figure 2.4B, these were compared to the wild type sequence from NCBI to identify any mutations.

In F-CHI mutation was therefore identified as ABCC8 exon6 heterozygous d. 16894G>A; c.1016G>A ; p. Gly316Arg, matching the details provided by the clinicians. The sequencing con-

firmed the origins of the tissue from this patient, and also highlighted the fact that the cells grew out of non-lesion tissue as the DNA present is heterozygous. Focal CHI is caused by the secondary loss of the chromosome 11 p-arm so affected lesion cells would not have the wild type allele present.

In D-CHI the mutation was identified as ABCC8 exon37 homozygous c.4481G>A ; p.Arg1494Gln, again matching the details provided by the clinicians and confirming the origins of the tissue from this patient. The affected nucleotide is shown in Figure 2.4, panel B, the only adenine shown would be a guanine in healthy individuals.

A-CHI was not genotyped by us, as the clinicians did not detect a mutation in any known CHI- associated gene, which as discussed above includes ABCC8, KCNJ11, GCK and HNF4A.

The three cell lines were also confirmed as karyotypically normal, as shown in Figure 2.4A. This allowed confirmation that the cell lines were stable in culture and could therefore be used for further studies. For each cell line, 19-20 cells were analysed by G-banding and all cells were identical, except for 1 cell in the A-CHI line which had a reciprocal translocation. Low levels of random mutations are known to occur in proliferative cell lines in vitro, the A-CHI line was most proliferative meaning there was more chance of random mutations occurring. As this was only seen in one cell we determined

83 the cell line was still sufficiently normal to be used.

2.4.3 CHI derived MSCs express pancreatic markers

Figure 2.5: Pancreatic genes are expressed in CHI pMSCs RT-PCR for genetic markers of pancreatic development in A-CHI at passage 2, 16 days after the start of culture (labelled early) and for all three CHI lines at passage 9 (labelled late). 18S rRNA is shown as a housekeeping reference gene for all other genes. The developmental stages at which the genes are expressed is shown in the center.

Having confirmed that the CHI derived cells were MSCs we examined numerous pancreatic lineage markers in comparison to bmMSCs, which are the usual MSCs used in research. Having pancreatic

84 genes already expressed would be a benefit for differentiation in pancreatic research.

A panel of transcription factors from various stages throughout the classical pancreatic devel- opmental pathways was assessed (Pan and Wright, 2011). Gene expression was extremely stable from around passage 5 until senescence occurred when cells were grown on tissue culture plastic in standard culture media. Prior to passage 5, additional genes were expressed which were lost by passage 5, this is shown in Figure 2.5. This included PDX1, NEUROD, INS, GCG, SST and the transcription factors expressed after passage 5. The transcription factors expressed were the same across the three cell lines later, this is shown in Figure 2.5. Several of these transcription factors are expressed at multiple stages during development, for clarity these are shown at the earliest stage at which they are expressed only.

CHI derived MSCS expressed three of five early stage markers, covering early endoderm and multipotent progenitors, as shown in Figure 2.5. SOX9 and SOX17 were clearly expressed, FOXA2 was not expressed at all, PDX1 was possibly expressed, but the band was not clearly visible and further quantitation would be needed to confirm this. The SOX genes, or SRY-related HMG box genes, are involved in multiple developmental pathways and alone are not specific for pancreatic development. HES1 is a downstream target of the Notch pathway and is co-expressed with SOX9 in pancreatic progenitors during development - they were also co-expressed in our CHI-derived MSCs.

The cells also expressed two transcription factors associated with endocrine progenitors and en- docrine precursors. NEUROG3 is expressed transiently during development but was not seen at any point in the cell lines as shown in Figure 2.5. PAX6 is induced as the cells mature towards differentiated endocrine cells and was seen throughout culture in our cell lines. However as with the

SOX genes, PAX6 is a member of the paired box family of transcription factors which are involved in development in many lineages. ISL1 was expressed in our three cell lines, this gene as the name indicates is highly specific for pancreatic endocrine lineages.

There were few markers of immature or mature final endocrine cells present. MAFB is involved with differentiation of β-cells and also involved in developmental pathways throughout the body and is expressed in the cell lines. ARX is a transcription factor involved with α-cell differentiation but was not expressed in our cells. GCG, INS and SST encode the hormones expressed by α-, β- and

δ-cells respectively and all seemed to be expressed, although this was not as clear after passage 5 as before so quantification would be required. CK19 is a marker of duct cells within the pancreas and was clearly expressed.

85 Culturing cells on uncoated plasticware led to the loss of some gene expression after several pas-

Figure 2.6: Growing cells on vitronectin and fibronectin increased cell number and insulin gene expression (A) Increased cells were counted after 5 days growth on vitronectin and fibronectin, compared to no coating. Bars are mean of 2 experiments in A-CHI at passages 10 and 11, where 2 x 104 cells were seeded, error bars are standard deviation. (B) INS gene expression was maintained for additional passages in A-CHI when cultured on vitronectin and fibronectin but not when they were cultured on uncoated plasticware. Image shown is from passage 6. sages, which was shown in Figure 2.5. However, when cells were cultured routinely on vitronectin and fibronectin, early results suggested that this allowed maintenance of INS expression, as well as increased cell numbers, shown in Figure 2.6. Further comparisons were carried out of other ECMs, which was inconclusive . This was primarily because differences in gene expression from one passage to another were small, so longer experiments on each separate ECM would need to be carried out.

Attempts to ascertain for how long the gene expression was maintained were also inconclusive, as the qRT-PCR primers for INS did not have good sensitivity at the low expression end, with no true negatives being seen. Vitronectin and fibronectin were initially selected based on their presence in developing islets (Stendahl et al., 2009) and due to the initial results were used as standard culture conditions.

To summarise, the three CHI-derived pMSC lines strongly expressed a number of transcription factors associated with pancreatic development - SOX17, SOX9, ISL1, PAX6 and MAFB. They also seemed to express PDX1 and the mature hormones GCG, INS and SST. The genes expressed are not specific to any stage but range from early endoderm through to mature hormone expressing cells.

The presence of these genes seems indicative that the cells are of pancreatic lineage, have maintained

86 Figure 2.7: Pancreatic proteins are expressed in CHI pMSCs Immunocytochemistry showed SOX9, ISL1, PAX6 and MAFB mRNA was translated and expressed as proteins, but was not necessarily correctly located. Images shown are from D-CHI, representative for all three cell lines, from passages between 7 and 12. All scale bars are 50 µm. White arrow on Islet1 indicates the only nucleus stained positive with this antibody. Rabbit and mouse panels show secondary antibody only stains.

87 some aspects of the lineage and may therefore be able to be re-differentiated to mature pancreatic cells. The presence of factors from multiple stages of development, endocrine and ductal cells may be due to a mixed population of cells being present.

We assessed whether the markers, SOX9, MAFB, PAX6 and ISL1 were translated to proteins.

As these are transcription factors expression should be primarily nuclear. This was the case for

MAFB, but ISL1 was only seen to be nuclear in a minority of cells, shown in Figure 2.7. PAX6 and

SOX9 appeared to be solely located to the cytoplasm. This indicated that the proteins were likely not functional and causing downstream transcription of pancreatic genes, conditions to cause nuclear translocation of these proteins may help re-differentiation of these progenitor cells.

2.4.4 CHI pMSCs have altered gene expression and methylation patterns compared to bmMSCs

Figure 2.8: Comparison of gene expression between CHI pMSCs and bmMSCs (A) RT- PCR images for transcription factors involved in pancreatic differentiation that are expressed in our cells show they are also expressed in bmMSCs except for ISL1, A-CHI is shown in the first column as a reference (B) qRT-PCR fold changes for ISL1 in CHI lines vs bmMSCs show increased ISL1 expression in CHI lines, n=3 individual lines for the bmMSCs, passages 3 and 4, and n=3 passages for each of our CHI lines, passages 3 and 4, error bars show standard deviation.* p<0.05, *** p<0.005.

SOX9, ISL1, PAX6, MAFB and CK19 are implicated in differentiation in the pancreas, but are also seen in other roles and tissues, and so their gene expression was assessed in three independent bmMSC lines shown in Figure 2.8.

88 All of these genes were also expressed in bmMSCs except for ISL1 which did not appear to be expressed. This difference was quantified in our three cell lines across three passages and the three independent bmMSCs which confirmed that the bmMSCs did not express ISL1 but our cells did. In the bmMSCs there was no expression detected at all i.e. the CT value was called at 40, the maximum in the assay used, but this could in fact be much higher (infinite) giving an artificial fold change.

ISL1 is a gene specifically involved in the development of the endocrine pancreas, in particular the islets of langerhans for which it is named. The fact it is expressed in the pancreas derived lines but not the bmMSCs, highlights the fact that the cells have a remnant of their tissue of origin and may therefore be more suited to redifferentiating to this lineage than bmMSCs. Based on this we hypothesised that there may be epigenetic memory of the pancreatic lineage in these cells.

Of the genes which were not clearly expressed, we tested the methylation in the 5’ CpG islands in CHI derived pMSCs against the bmMSCs, to see if there was epigenetic memory in the cells.

Both PDX1 and CK19 showed overall less methylation in the CHI derived cells than the bmMSCs, with significantly less methylation at two loci in each, shown in Figure 2.9. In the PDX1 promoter, the differentially methylated loci align to the region closest to the coding sequence, just after the enhancer sequences (area II/III). The loci both show differences of 20% - locus 7: 36% and 56% in pMSCs and bmMSCs respectively, locus 9: 22% and 42%. CK19 had been used for comparison, as it appeared to be expressed in both CHI and bmMSCs, but showed differences in methylation at the 2 loci of over 10% (locus 2: 69% and 87%, locus 6: 61% and 73% in pMSCs and bmMSCs respectively). The differential degree of methylation may indicate alternate regulation in the different tissue types, such as for enhancer binding. INS is the gene which encodes insulin, the protein most essential when producing β-cells. There was no difference in methylation levels between the CHI derived and bmMSCs for INS. INS does not have a CpG island upstream and primers were therefore designed to amplify the most dense region of CpGs proximal to the gene – they may not be involved in regulation of the gene as this has not been studied. AMY2A, the gene encoding pancreatic amylase in exocrine cells showed different methylation in the CHI derived cells compared to the bmMSCs, with significantly less methylation at one locus (24% methylation in pMSCs and 49% methylation in bmMSCs, p=0.0122) and somewhat increased at another locus with the rest being identical. It is unclear whether this would mean the cells were able to preferentially differentiate to an exocrine lineage but exocrine cell markers may need to be excluded when attempting to differentiate the cells down the endocrine lineage.

To confirm that the decreased methylation seen was not global, we checked the 5’ region of SPP1

89 90 Figure 2.9: Comparison of gene methylation between CHI pMSCs and bmMSCs The percentage methylation at each CpG in the 5’ regions of PDX1, INS, AMY2A and CK19 was assessed by bisulfite sequencing. The top panel for each gene shows the percentage methylation (shade of grey) at each locus (individual circle), with the averages for the three CHI and three bmMSCs shown below to aid clarity. * p<0.05 assessed by Kruskal-Wallis test within MethPlotter. The boxed regions on the top panels indicate the locus at which significance was seen. Methylation was assessed in all samples at passage 3 or 4.

91 (osteopontin) - a marker of a non-pancreatic lineage that MSCs differentiate to i.e. osteoblasts.

Analysis was not fully possible as the three CHI pMSC lines did not have a wild type sequence, with an extra CpG being present at the -65 position of the promoter (Hijiya et al., 1994) as shown in Fig- ure 2.10, the blue and black peaks representing cytosine and guanine respectively within the boxed regions.It is unclear why this mutation would be present in only the CHI derived cells, although

SPP1 is a downstream target of SOX9 (Pritchett et al., 2012) and a putative pancreatic ductal marker (Roberts, 2011). A mutation affecting regulation of SPP1 expression in the pancreas might therefore have implications in the aetiology of CHI. We checked this mutation with normal DNA sequencing on the non-bisulfite converted gDNA. The sequencing revealed that the gDNA did not have the extra CpG, shown in Figure 2.10B - within the box where the sequence reads ”CTCCG”, the T would become G. This suggests that an artifact of the bisulfite conversion is responsible for the altered bisulfite sequencing result, potentially due to the degree of methylation at the true CpG locus.

92 Figure 2.10: Bisulfite sequencing and gene sequencing of SPP1 (A) Bisulfite sequencing traces of each of the three bmMSC samples and the three CHI pMSC samples used previously for the promoter region of SPP1. Boxed regions highlight the region of the trace where differences between CHI and bmMSCs were seen. (B) Sequencing of the same region on non bisulfite converted gDNA of the F-CHI line, boxed region corresponds to the boxed regions in panel A.

93 2.5 Discussion

In this study we report the reproducible derivation of cell lines from the pancreatic tissue of three patients with CHI. Each patient had a different form of the disease which had no impact on the ability to derive a cell line. The derived cells were not clonal, as the origins of pMSCs or the best cell type to use are not fully understood (Bhonde et al., 2014).

The cell lines have the defining characteristics of MSCs as set down by The International Society for Cellular Therapy (Dominici et al., 2006), shown in Figures 2.1 and 2.2. The derived cells show plastic adherence (which is how they are initially selected for, from the tissue), they are able to differentiate to osteoblasts, chondrocytes and partially to adipocytes and express cell surface mark- ers CD29, CD44, CD73, CD90 and CD105, whilst being negative for CD45. The low percentage of adipocytes arising from the differentiation is in concordance with previous data from pancreas derived MSCs (Russ et al., 2009). The authors of Russ et al. (2009) suggested that this may mean pMSCs are not authentic MSCs, however we believe this may be due to tissue specific biases as discussed below. An opposing study showed robust differentiation to adipocytes in pMSCs which had been lineage traced and shown to not be derived from INS/PDX1 positive cells (Chase et al.,

2007). This could suggest that poor adipocyte differentiation is a feature of MSCs derived from the pancreatic lineages and that in Chase et al. (2007), the derived cells were in fact contaminating bmMSCs as has happened in other studies (Sordi et al., 2010).

CHI pMSCs also express mesenchymal cytoskeletal proteins vimentin, nestin and α-smooth mus- cle actin and are negative for epithelial proteins E-cadherin and occludin as shown in Figure 2.3. This was in agreement with other studies of pMSCs (Davani et al., 2007, Gallo et al., 2007). E-cadherin is quickly lost in MSCs (Russ et al., 2008) with networks of epithelial-mesenchymal transition markers being seen (Kutlu et al., 2009) suggesting pMSCs are derived from epithelial cells within the pan- creas. Nestin has been discussed as a putative stem cell marker, but is co-expressed with vimentin,

α-smooth muscle actin, CK19 and amylase (Street et al., 2004) or with MSC markers CD90 and

CD105 within the pancreas (Carlotti et al., 2010). All of these (bar amylase) were expressed in the

CHI pMSCs, providing evidence of their similarity to other pMSCs. However this provides little information on whether they are derived from mesenchymal or ductal cells within the pancreas, or de-differentiated epithelial endocrine cells.

The source of MSCs within the body has been shown to be important, with differences seen be- tween proliferation and differentiation ability. BmMSCs undergo senescence before other types and only periosteum derived MSCs could be differentiated to β-cells sufficiently for glucose stimulated

94 insulin secretion, of four cell types tested (Kim et al., 2012). BmMSCs and pMSCs have been shown to be different at a basal level, and after differentiation to insulin secreting cells (Kutlu et al., 2009,

Zanini et al., 2011). We therefore investigated pancreatic markers within the CHI derived pMSCs compared to bmMSCs, the most common MSC source shown in Figures 2.4-8. We observed that the derived cells express a number of genes associated with pancreatic development – SOX17, SOX9,

PAX6, MAFB, HES1 and ISL1. Some of these markers were similar to those reported before in pMSCs (Eberhardt et al., 2006) although PAX6 and ISL1 have been shown to be downregulated in pMSCs compared to islets in one study (Kutlu et al., 2009), although another showed pMSCs to be

ISL1 negative (Gallo et al., 2007). HES1 has been reported before to be involved in adaptation of

β-cells to in vitro proliferation (Bar et al., 2008), which is likely the role of its expression in our cells.

Similarly, SOX9 is known to be associated with maintenance of progenitors in an undifferentiated state (Seymour et al., 2007). The genes we saw expressed are not solely associated with the pancreas, so we compared the expression of them in our cell lines to non-pancreatic MSCs, derived from bone marrow. All the genes were expressed in the bmMSCs except for ISL1. This suggests that there is some tissue specific genetic memory in the CHI derived MSCs, which may be an advantage for redifferentiating the cells to pancreatic lineages.

The cells also expressed CK19, a ductal marker (Sahin et al., 2005, Bouwens et al., 1994). How- ever, CK19 was also expressed in bmMSCs, it is not clear the role that CK19 may play in MSCs, and it has not been reported in the bone marrow in vivo. CK19 has been reported previously in pMSCs which were also derived from exocrine enriched pancreatic digest, however expression of CK19 was lost by P3 in this report (Lima et al., 2013). The bmMSCs used in this study which expressed CK19 were only available at P3, as in Lima et al. (2013), and so later gene expression was not studied; the

CHI derived cells expressed CK19 after P3 though. The prolonged CK19 expression in the CHI- derived pMSCs may be indicative of a ductal origin, which has been hypothesised as the source of

MSCs within the exocrine compartment (Fanjul et al., 2010). The ducts are frequently hypothesised to be the primary source of pancreatic stem cells lending weight to this theory.

Some genes essential for endocrine differentiation to β-cells, such as PDX1 and INS, were not expressed in the CHI-derived pMSCs. It has been shown that pMSCs have more active histone acetylation for INS and GCG than bmMSCs, although INS promoter accessibility was inhibited in both MSC types (Wilson et al., 2009). In line with this, we saw no difference in 5’ methylation of

CpGs upstream of INS, but there was significantly less methylation in the CpG islands upstream of

PDX1, CK19 and AMY2A. The location and magnitude of the differential methylation of the PDX1 promoter makes it most likely that this gene, of the genes tested, would have transcription affected

95 by methylation. This suggests that despite CHI pMSCs not expressing PDX1, re-induction may be easier than in bmMSCs. However, as several of the markers the MSCs expressed were from before endocrine commitment, there may also be the possibility of re-induction of amylase and differentia- tion to an exocrine lineage which would need to be excluded. De-methylation of INS may occur with expression of PDX1, but the use of DNA methyltransferase inhibitors may be required for successful differentiation.

In conclusion, pMSCs were reproducibly derived from three patients with CHI, with each form of the disease. All three lines showed a similar phenotype, characteristic of MSCs with a bias against adipose differentiation which may be characteristic of MSCs from the pancreas. Further contributing to the idea that the CHI pMSCs were unique for the pancreas, they expressed ISL1 which was not seen in bmMSCs and show differential promoter methylation to bmMSCs as we showed in Figures

2.8 and 2.9. This may mean CHI pMSCs are able to preferentially redifferentiate to β-cells over other MSC types. We therefore present the first derivation of MSCs from CHI patients and propose their use for pancreatic research by differentiation to β-cells.

96 2.6 References

J.-B. Arnoux, V. Verkarre, C. Saint-Martin, F. Montravers, A. Brassier, V. Valayannopoulos,

F. Brunelle, J.-C. Fournet, J.-J. Robert, Y. Aigrain, C. Bellanne-Chantelot, and P. de Lonlay.

Congenital hyperinsulinism: current trends in diagnosis and therapy. Orphanet Journal of Rare

Diseases, 6(1):1750–1172, 2011.

Y. Bar, H. A. Russ, S. Knoller, L. Ouziel-Yahalom, and S. Efrat. HES-1 Is Involved in Adaptation

of Adult Human β-Cells to Proliferation In Vitro. Diabetes, 57(9):2413–2420, 2008.

O. Bar-Nur, H. Russ, S. Efrat, and N. Benvenisty. Epigenetic Memory and Preferential Lineage-

Specific Differentiation in Induced Pluripotent Stem Cells Derived from Human Pancreatic Islet

Beta Cells. Cell Stem Cell, 9(1):17–23, 2011.

R. R. Bhonde, P. Sheshadri, S. Sharma, and A. Kumar. Making surrogate β-cells from mesenchymal

stromal cells: Perspectives and future endeavors. The International Journal of Biochemistry &

Cell Biology, 46:90 – 102, 2014.

L. Bouwens, R.-N. Wang, E. D. Blay, D. G. Pipeleers, and G. Kloppel. Cytokeratins as Markers of

Ductal Cell Differentiation and Islet Neogenesis in the Neonatal Rat Pancreas. Diabetes, 43(11):

1279–1283, 1994.

A. Capes-Davis, Y. A. Reid, M. C. Kline, D. R. Storts, E. Strauss, W. G. Dirks, H. G. Drexler,

R. A. MacLeod, G. Sykes, A. Kohara, Y. Nakamura, E. Elmore, R. W. Nims, C. Alston-Roberts,

R. Barallon, G. V. Los, R. M. Nardone, P. J. Price, A. Steuer, J. Thomson, J. R. Masters, and

L. Kerrigan. Match criteria for human cell line authentication: Where do we draw the line? Int.

J. Cancer, 132(11):2510–2519, 2013.

F. Carlotti, A. Zaldumbide, C. J. Loomans, E. van Rossenberg, M. Engelse, E. J. de Koning, and

R. C. Hoeben. Isolated human islets contain a distinct population of mesenchymal stem cells.

Islets, 2(3):164–173, 2010.

L. G. Chase, F. Ulloa-Montoya, B. L. Kidder, and C. M. Verfaillie. Islet-Derived Fibroblast-Like

Cells Are Not Derived via Epithelial-Mesenchymal Transition From Pdx-1 or Insulin-Positive Cells.

Diabetes, 56(1):3–7, 2007.

E. Corritore, E. Dugnani, V. Pasquale, R. Misawa, P. Witkowski, J. Lei, J. Markmann, L. Piemonti,

E. M. Sokal, S. Bonner-Weir, and P. A. Lysy. β-Cell Differentiation of Human Pancreatic Duct-

Derived Cells After In Vitro Expansion. Cellular Reprogramming, 16(6):456–466, 2014.

G. Danaei, M. M. Finucane, Y. Lu, G. M. Singh, M. J. Cowan, C. J. Paciorek, J. K. Lin, F. Farzadfar,

Y.-H. Khang, G. A. Stevens, M. Rao, M. K. Ali, L. M. Riley, C. A. Robinson, and M. Ezzati.

97 National, regional, and global trends in fasting plasma glucose and diabetes prevalence since 1980:

systematic analysis of health examination surveys and epidemiological studies with 370 country-

years and 2.7 million participants. The Lancet, 378(9785):31–40, 2011.

B. Davani, L. Ikonomou, B. M. Raaka, E. Geras-Raaka, R. A. Morton, B. Marcus-Samuels, and

M. C. Gershengorn. Human Islet-Derived Precursor Cells Are Mesenchymal Stromal Cells That

Differentiate and Mature to Hormone-Expressing Cells In Vivo. Stem Cells, 25(12):3215–3222,

2007.

M. Dominici, K. Le Blanc, I. Mueller, I. Slaper-Cortenbach, F. Marini, D. Krause, R. Deans, A. Keat-

ing, D. Prockop, and E. Horwitz. Minimal criteria for defining multipotent mesenchymal stromal

cells. The International Society for Cellular Therapy position statement. Cytotherapy, 8(4):315–7,

2006.

M. J. Dunne, C. Kane, R. M. Shepherd, J. A. Sanchez, R. F. James, P. R. Johnson, A. Aynsley-

Green, S. Lu, J. P. Clement, K. J. Lindley, S. Seino, L. Aguilar-Bryan, G. Gonzalez, and P. J. Milla.

Familial Persistent Hyperinsulinemic Hypoglycemia of Infancy and Mutations in the Sulfonylurea

Receptor. New England Journal of Medicine, 336(10):703–706, 1997.

M. J. Dunne, K. E. Cosgrove, R. M. Shepherd, A. Aynsley-Green, and K. J. Lindley. Hyperinsulinism

in Infancy: From Basic Science to Clinical Disease. Physiological Reviews, 84(1):239–275, 2004.

M. Eberhardt, P. Salmon, M.-A. von Mach, J. G. Hengstler, M. Brulport, P. Linscheid, D. Seboek,

J. Oberholzer, A. Barbero, I. Martin, B. Muller, D. Trono, and H. Zulewski. Multipotential nestin

and Isl-1 positive mesenchymal stem cells isolated from human pancreatic islets. Biochemical and

Biophysical Research Communications, 345(3):1167–1176, 2006.

S. Ellard, S. E. Flanagan, C. A. Girard, A.-M. Patch, L. W. Harries, A. Parrish, E. L. Edghill, D. J.

Mackay, P. Proks, K. Shimomura, H. Haberland, D. J. Carson, J. P. Shield, A. T. Hattersley,

and F. M. Ashcroft. Permanent Neonatal Diabetes Caused by Dominant, Recessive, or Compound

Heterozygous SUR1 Mutations with Opposite Functional Effects. The American Journal of Human

Genetics, 81(2):375–382, 2007.

M. Fanjul, V. Gmyr, C. Sengenes, G. Ratovo, M. Dufresne, B. Lefebvre, J. Kerr-Conte, and E. Hol-

lande. Evidence for Epithelial-Mesenchymal Transition in Adult Human Pancreatic Exocrine Cells.

Journal of Histochemistry & Cytochemistry, 58(9):807–823, 2010.

J.-C. Fournet, C. Mayaud, P. de Lonlay, M.-S. Gross-Morand, V. Verkarre, M. Castanet, M. Devillers,

J. Rahier, F. Brunelle, J.-J. Robert, C. Nihoul-Fekete, J.-M. Saudubray, and C. Junien. Unbalanced

Expression of 11p15 Imprinted Genes in Focal Forms of Congenital Hyperinsulinism. The American

Journal of Pathology, 158(6):2177–2184, 2001.

98 R. Gallo, F. Gambelli, B. Gava, F. Sasdelli, V. Tellone, M. Masini, P. Marchetti, F. Dotta, and

V. Sorrentino. Generation and expansion of multipotent mesenchymal progenitor cells from cul-

tured human pancreatic islets. Cell Death Differ, 14(11):1860–1871, 2007.

B. Glaser, F. Ryan, M. Donath, H. Landau, C. A. Stanley, L. Baker, D. E. Barton, and P. S.

Thornton. Hyperinsulinism caused by paternal-specific inheritance of a recessive mutation in the

sulfonylurea-receptor gene. Diabetes, 48(8):1652–1657, 1999.

B. Glaser, P. Thornton, T. Otonkoski, and C. Junien. Genetics of neonatal hyperinsulinism. Archives

of Disease in Childhood - Fetal and Neonatal Edition, 82(2):F79–F86, 2000.

N. Hijiya, M. Setoguchi, K. Matsuura, Y. Higuchi, S. Akizuki, and S. Yamamoto. Cloning and

characterization of the human osteopontin gene and its promoter. Biochemical Journal, 303(1):

255–262, 1994.

Y. Hu, L. Liao, Q. Wang, L. Ma, G. Ma, X. Jiang, and R. C. Zhao. Isolation and identification of

mesenchymal stem cells from human fetal pancreas. Journal of Laboratory and Clinical Medicine,

141(5):342 – 349, 2003.

C. James, R. R. Kapoor, D. Ismail, and K. Hussain. The genetic basis of congenital hyperinsulinism.

Journal of Medical Genetics, 46(5):289–299, 2009.

M. V. Joglekar, V. M. Joglekar, S. V. Joglekar, and A. A. Hardikar. Human fetal pancreatic insulin-

producing cells proliferate in vitro. Journal of Endocrinology, 201(1):27–36, 2009.

S.-J. Kim, Y.-S. Choi, E.-S. Ko, S.-M. Lim, C.-W. Lee, and D.-I. Kim. Glucose-stimulated insulin

secretion of various mesenchymal stem cells after insulin-producing cell differentiation. Journal of

Bioscience and Bioengineering, 113(6):771 – 777, 2012.

B. Kutlu, A. G. Kayali, S. Jung, G. Parnaud, D. Baxter, G. Glusman, N. Goodman, L. A. Behie,

A. Hayek, and L. Hood. Meta-analysis of gene expression in human pancreatic islets after in vitro

expansion. Physiological Genomics, 39(1):72–81, 2009.

L.-C. Li and R. Dahiya. MethPrimer: designing primers for methylation PCRs. Bioinformatics, 18

(11):1427–1431, 2002.

M. Lima, K. Muir, H. Docherty, N. McGowan, S. Forbes, Y. Heremans, H. Heimberg, J. Casey, and

K. Docherty. Generation of Functional Beta-Like Cells from Human Exocrine Pancreas. PLoS

One, 5(11):e0156204, 2016.

M. J. Lima, K. R. Muir, H. M. Docherty, R. Drummond, N. W. McGowan, S. Forbes, Y. Heremans,

I. Houbracken, J. A. Ross, S. J. Forbes, P. Ravassard, H. Heimberg, J. Casey, and K. Docherty.

99 Suppression of Epithelial-to-Mesenchymal Transitioning Enhances Ex Vivo Reprogramming of

Human Exocrine Pancreatic Tissue Toward Functional Insulin-Producing β-Like Cells. Diabetes,

62(8):2821–2833, 2013.

I. Mallona, A. Diez-Villanueva, and M. A. Peinado. Methylation plotter: a web tool for dynamic

visualization of DNA methylation data. Source Code for Biology and Medicine, 9(1):1–5, 2014.

N. W. Marion and J. J. Mao. Mesenchymal Stem Cells and Tissue Engineering. Methods in enzy-

mology, 420:339–361, 2006.

K. Muir, M. Lima, H. Docherty, N. McGowan, S. Forbes, Y. Heremans, S. Forbes, H. Heimberg,

J. Casey, and K. Docherty. Kruppel-Like Factor 4 Overexpression Initiates a Mesenchymal-to-

Epithelial Transition and Redifferentiation of Human Pancreatic Cells following Expansion in

Long Term Adherent Culture. PLoS One, 10(10):e0140352, 2015.

F. C. Pan and C. Wright. Pancreas organogenesis: From bud to plexus to gland. Dev. Dyn., 240(3):

530–565, 2011.

J. Pritchett, E. Harvey, V. Athwal, A. Berry, C. Rowe, F. Oakley, A. Moles, D. A. Mann, N. Bobola,

A. D. Sharrocks, B. J. Thomson, A. M. Zaitoun, W. L. Irving, I. N. Guha, N. A. Hanley, and

K. P. Hanley. Osteopontin is a novel downstream target of SOX9 with diagnostic implications for

progression of liver fibrosis in humans. Hepatology, 56(3):1108–1116, 2012.

N. A. Roberts. The role of Sox9 during human pancreatic development. Phd thesis, University of

Manchester, School of Medicine, 2011.

H. A. Russ, Y. Bar, P. Ravassard, and S. Efrat. In Vitro Proliferation of Cells Derived From Adult

Human β-Cells Revealed By Cell-Lineage Tracing. Diabetes, 57(6):1575–1583, 2008.

H. A. Russ, P. Ravassard, J. Kerr-Conte, F. Pattou, and S. Efrat. Epithelial-Mesenchymal Transition

in Cells Expanded In Vitro from Lineage-Traced Adult Human Pancreatic Beta Cells. PLoS ONE,

4(7):e6417, 2009.

E. A. Ryan, B. W. Paty, P. A. Senior, D. Bigam, E. Alfadhli, N. M. Kneteman, J. R. Lakey, and

A. J. Shapiro. Five-Year Follow-Up After Clinical Islet Transplantation. Diabetes, 54(7):2060–2069,

2005.

F. Sahin, W. Qiu, R. E. Wilentz, C. A. Iacobuzio-Donahue, A. Grosmark, and G. H. Su. RPL38,

FOSL1, and UPP1 Are Predominantly Expressed in the Pancreatic Ductal Epithelium. Pancreas,

30(2):158–167, 2005.

100 R. J. Salisbury, B. Han, R. E. Jennings, A. A. Berry, A. Stevens, Z. Mohamed, S. A. Sugden,

R. De Krijger, S. E. Cross, P. P. Johnson, M. Newbould, K. E. Cosgrove, K. P. Hanley, I. Banerjee,

M. J. Dunne, and N. A. Hanley. Altered Phenotype of β-Cells and Other Pancreatic Cell Lineages

in Patients With Diffuse Congenital Hyperinsulinism in Infancy Caused by Mutations in the ATP-

Sensitive K-Channel. Diabetes, 64(9):3182–3188, 2015.

P. A. Seymour, K. K. Freude, M. N. Tran, E. E. Mayes, J. Jensen, R. Kist, G. Scherer, and M. Sander.

SOX9 is required for maintenance of the pancreatic progenitor cell pool. Proceedings of the National

Academy of Sciences, 104(6):1865–1870, 2007.

A. J. Shapiro, J. R. Lakey, E. A. Ryan, G. S. Korbutt, E. Toth, G. L. Warnock, N. M. Kneteman,

and R. V. Rajotte. Islet Transplantation in Seven Patients with Type 1 Diabetes Mellitus Using

a Glucocorticoid-Free Immunosuppressive Regimen. New England Journal of Medicine, 343(4):

230–238, 2000.

V. Sordi, R. Melzi, A. Mercalli, R. Formicola, C. Doglioni, F. Tiboni, G. Ferrari, R. Nano,

K. Chwalek, E. Lammert, E. Bonifacio, and L. Piemonti. Mesenchymal Cells Appearing in Pan-

creatic Tissue Culture Are Bone Marrow-Derived Stem Cells With the Capacity to Improve Trans-

planted Islet Function. Stem Cells, 28(1):140–151, 2010.

J. C. Stendahl, D. B. Kaufman, and S. I. Stupp. Extracellular Matrix in Pancreatic Islets: Relevance

to Scaffold Design and Transplantation. Cell Transplantation, 18(1):1–12, 2009.

C. Street, Lakey, K. Seeberger, L. Helms, R. Rajotte, A. Shapiro, and G. Korbutt. Heterogenous

expression of nestin in human pancreatic tissue precludes its use as an islet precursor marker.

Journal of Endocrinology, 180(2):213–225, 2004.

V. Verkarre, J. C. Fournet, P. de Lonlay, M. S. Gross-Morand, M. Devillers, J. Rahier, F. Brunelle,

J. J. Robert, C. Nihoul-Fekete, J. M. Saudubray, and C. Junien. Paternal mutation of the sul-

fonylurea receptor (SUR1) gene and maternal loss of 11p15 imprinted genes lead to persistent

hyperinsulinism in focal adenomatous hyperplasia. The Journal of Clinical Investigation, 102(7):

1286–1291, 1998.

L. M. Wilson, S. H. Wong, N. Yu, E. Geras-Raaka, B. M. Raaka, and M. C. Gershengorn. Insulin

but Not Glucagon Gene is Silenced in Human Pancreas-Derived Mesenchymal Stem Cells. Stem

Cells, 27(11):2703–2711, 2009.

C. Zanini, S. Bruno, G. Mandili, D. Baci, F. Cerutti, G. Cenacchi, L. Izzi, G. Camussi, and M. Forni.

Differentiation of Mesenchymal Stem Cells Derived from Pancreatic Islets and Bone Marrow into

Islet-Like Cell Phenotype. PLoS ONE, 6(12):e28175, 2011.

101 H. Zulewski, E. J. Abraham, M. J. Gerlach, P. B. Daniel, W. Moritz, B. Muller, M. Vallejo, M. K.

Thomas, and J. F. Habener. Multipotential Nestin-Positive Stem Cells Isolated From Adult Pan-

creatic Islets Differentiate Ex Vivo Into Pancreatic Endocrine, Exocrine, and Hepatic Phenotypes.

Diabetes, 50(3):521–533, 2001.

102 103 Chapter 3

Increased Proliferation and Altered

Cell Cycle Regulation in Pancreatic

Stem Cells derived from Patients with Congenital Hyperinsulinism

Sophie G Kellaway, Bing Han, Rachel Salisbury, Zainaba Mohamed,

Indi Banerjee, Alexander Ryan, Karen E Cosgrove and Mark J Dunne

104 3.1 Abstract

Congenital hyperinsulinism (CHI) is a disease associated with profound hypoglycaemia and constant high levels of insulin secretion. Diffuse CHI has also been shown to be associated with increased rates of proliferation throughout the pancreas, along with altered localisation of CDK6 and p27Kip1. We have shown that increased proliferation is also seen in focal and atypical CHI. Increased proliferation was seen in stem cells grown from pancreas explants of patients with CHI. We also saw alterations to the cell cycle, increased CDK1 levels and decreased p27Kip1 nuclear localisation in these cells when compared to healthy adult controls. Alongside the basal differences seen within CHI stem cells, manipulating the glucose and insulin in culture showed that total p27Kip1 is decreased in low glucose and nuclear localisation of p27Kip1 is further decreased with high insulin. In conclusion, pancreatic stem cells from patients with CHI show increased proliferation rates in vitro similar to the CHI pancreas tissue in vivo. The CHI cells show altered cell cycle regulators which are further affected by glucose and insulin levels. Pancreas stem cells from CHI patients may be a model to further understand the cell cycle alterations leading to increased proliferation in CHI.

105 3.2 Introduction

Congenital hyperinsulinism (CHI) is a disease associated with profound hypoglycaemia due to high levels of unregulated insulin secretion (Dunne et al., 2004). There are three forms of CHI - focal, diffuse and atypical. Focal CHI is most commonly due to a recessive mutation in the ABCC8 gene, where loss of heterozygosity leading to no functional allele and non-functional ATP-sensitive potas- sium (KATP) channels (Verkarre et al., 1998, Glaser et al., 1999, Fournet et al., 2001). This form is named for the fact it only affects a focal lesion within the pancreas which is almost exclusively

β-cells. The loss of heterozygosity also affects the cyclin-dependent kinase inhibitor (CKI) – p57Kip2

– a likely cause of the β-cell hyperplasia seen (Fournet et al., 2001, Kassem et al., 2001). Diffuse

CHI is due to a homozygous recessive mutation in a number of genes (Glaser et al., 2000), including

ABCC8 (Dunne et al., 1997) and every β-cell within the pancreas is affected. Atypical CHI typically has a later onset than focal or diffuse CHI, is not caused by any known mutation (screening of the genes associated with focal and diffuse CHI excludes these) and affects the whole pancreas (Arnoux et al., 2011). It has also been shown that atypical CHI is associated with altered hexokinase expres- sion (Henquin et al., 2013).

It was recently shown that there are increased levels of proliferation in diffuse CHI patients com- pared to age-matched controls, as documented by the number of Ki67 positive cells, which may be a factor in the disease pathology. This was in turn found to be associated with a high number of

β-cell nuclei containing CDK6 and p27Kip1 (Salisbury et al., 2015).

Kip1 CDK6 and p27 are cell cycle regulators involved in the G1/S transition. Quiescent cells are said to be in G0, but all cells which are mitotically active can be allocated to a stage of the cell cycle. These cell cycle stages are Gap 1 or G1-phase which resembles G0 where cells are resting or growing, Synthesis or S-phase where DNA is duplicated, Gap 2 or G2-phase where the cell prepares to divide and Mitosis or M-phase where cell division occurs and two daughter cells are produced, reviewed by Harashima et al. (2013). The progression through the G1/S checkpoint commits a cell to division (Bertoli et al., 2013), the G2/M checkpoint controls both DNA integrity and cell size at the point of division (Stark and Taylor, 2004). Alterations to cell cycle regulators at either of these stages affects the proliferation rates of cells.

The progression of cells through the cell cycle is controlled by a multitude of both positive and negative regulators. Positive regulation is by cyclins and their binding partners: cyclin-dependent kinases (CDKs). These are further regulated by phosphorylation, localisation within the cell and interaction with CKIs, some of the complexities of this system are reviewed in (Morgan, 1995).

106 Cyclin-CDK complexes are only functional when localised to the nucleus. Cyclin-CDKs appear to be primarily nuclear in vivo, but they do shuttle between the cytoplasm and nucleus with their ap- parent nuclear localisation being due to faster import than export (Jackman et al., 2002). There are several CDKs and cyclins, which stimulate different portions of the cell cycle and show redundancy due to sequence and binding similarity. CDK1 is the least redundant CDK, with knock out mice suffering early embryonic lethality (Santamaria et al., 2007). It appears that the sequence similarities and ability to bind to both D and B-type Cyclins means CDK1 can be sufficient to drive the cell cycle alone when required as discussed by Satyanarayana and Kaldis (2009). However, normally,

CyclinD-CDK4/6 are responsible for entry to G1. CDK4 and CDK6 can compensate for each other and are not both required in every cell in the body (Malumbres et al., 2004) although CDK6 appears to be essential in β-cells (Rane et al., 1999). Therefore, whilst CDK6 has been shown to be important in β-cell proliferation in CHI, it may not be the factor primarily responsible for proliferation in the other cells of the pancreas.

CKIs as the name suggest are generally negative regulators of the cell cycle by inhibiting CDK action, but can also be associated with positive cell cycle progression (Sherr and Roberts, 1999).

If the activity of the CKI – p27Kip1 – is reduced, stem cell expansion is observed (Besson et al.,

2007), whilst increased nuclear and cytoplasmic p27Kip1 is seen in healthy tissue when compared to cancerous cells (Claudio et al., 2002). In endocrine cells of the pancreas p27Kip1 has been shown to be of particular importance in regulating proliferation in p27Kip1 knockout mice, both α- and

β- cells proliferate during late embryogenesis when they should be quiescent (Georgia and Bhushan,

2006). Conversely, it has been shown that p27Kip1, alongside other CKIs such as p21Cip1 and p57Kip2 help the cyclin-CDK complexes to form in the nucleus (LaBaer et al., 1997), in the knockout mice detectable levels of these complexes are not formed although interestingly Rb, the main target of

G1/S CDKs, still gets phosphorylated (Cheng et al., 1999). This may be related to a threshold level of the proteins required as a 1:1:1 ratio of CyclinD-CDK4/6-p27Kip1 can still phosphorylate

Rb, whereas a 1:1:1 ratio of CyclinA-CDK2-p27Kip1 is unable to (Blain et al., 1997). CDK1 and

CDK2 act in the absence of p27Kip1 (Cerqueira et al., 2014), so it may be that the CyclinD-CDK4/6 complexes act as a sink for excess p27Kip1 thereby allowing CDK1 or 2 to drive proliferation. This is further backed up by the observation that there is no free p27Kip1 found in vivo, it is bound to and therefore either activating or inhibiting cyclinD-CDK4 complexes based on its phosphorylation status (James et al., 2008).

The cell cycle is intimately linked with the endocrine cells of the pancreas in a variable manner.

We also know that there are differences between the cell types of the pancreas in terms of prolif-

107 eration. This includes altered regulation of p27Kip1, of which expression is decreased in response to exendin-4 in β-cells but not in α-cells (Cai et al., 2014). Rb, when decreased causes prolifer- ation in β-cells and cell cycle arrest in α-cells (Cai et al., 2014) and also has differing effects on the transcription factors controlling β- and α-cell fate (Cai et al., 2013). However, the cell cycle regulators also have effects outside of the cell cycle. The inhibitor p16Ink4A can impact on insulin secretion in an immortalised β-cell line, where the immortalisation method means that the cell cycle cannot be involved in the observation (Pal et al., 2015). Insulin itself though causes translocation of cyclinD1-CDK4 complexes to the nucleus in mice, with no increase seen in proliferation suggesting a metabolic function (Lee et al., 2014). These data together may point to some form of feedback loop between the cell cycle and insulin.

Insulin is known to function as a growth factor alongside its functions related to glucose home- ostasis. It is therefore probable that the hyper-physiological levels of insulin within the CHI pancreas are driving increased rates of proliferation. At very high concentrations, insulin binds the insulin- like growth factor receptors (IGFRs) which as the name suggests triggers growth (Denley et al.,

2004, Belfiore and Malaguarnera, 2011). Insulin has a much greater affinity for the insulin recep- tor (IR) than IGFRs, and downstream signalling due to dimerisation or adjacence of receptors has been excluded (De Meyts et al., 1995). However, insulin does have mitogenic effects through its own receptor, as shown by experiments in cells with functional IR but without IGFRs which exhibit dose- dependent growth in media with insulin (Ish-Shalom et al., 1997). Downstream of the IR, mitogenic effects are via PI3K/Akt and mTORC signalling. In mouse β-cells Akt induces proliferation through cyclins and CDK4 (Fatrai et al., 2006) and in mTORC knockout mice Akt is decreased resulting in an increase in total p27Kip1 protein and a resulting decrease in β-cell numbers (Gu et al., 2011).

There is evidence that the mitogenic effects of high levels of insulin in humans are not just theoretical

- increased rates of cancer have been observed in type 2 diabetics (Belfiore and Malaguarnera, 2011,

Giovannucci et al., 2010, Rose and Vona-Davis, 2012) and insulin receptor signalling is important for

β-cell compensatory growth response in pre-diabetes (Okada et al., 2007).

One difficulty with studying a disease such as CHI is that studies are primarily on fixed post- operative tissue. This means that observations are a static snapshot and cannot be manipulated.

Whilst rodent models of CHI have been generated (Shimomura et al., 2013), there is proliferation and cell cycle features in the rodent pancreas which are not present in the human pancreas meaning this element of the disease cannot be modelled (Kulkarni et al., 2012, Fiaschi-Taesch et al., 2009).

There are also differences in the cell cycle regulators used by rodents and humans, the cell cycle is well studied in rodents but there are several elements which are unknown in humans (Kulkarni

108 et al., 2012). CDK6 which has been shown to be important in CHI is not present in rodent islets and CDK4 alone is used, whilst cyclinD2 is important in rodent islets but can be compensated for in humans (Fiaschi-Taesch et al., 2009). Further, CHI patients are undergoing treatment to normalise their glucose and insulin levels as far as possible prior to the operations. This must therefore be considered when analysing the tissue although the exact conditions within the pancreas at the time the tissue is fixed may not be explicitly clear.

109 3.3 Methods

All reagants were from Sigma-Aldrich or Fisher-Scientific.

3.3.1 Cell culture

Cell line Age at Disease form Mutation Number of popu- surgery lation doublings F-CHI 4 months Focal CHI ABCC8 exon 6 (het- 46 erozygous) A-CHI 17 months Atypical CHI Unknown 106 D-CHI 11 weeks Diffuse CHI ABCC8 exon 37 60

Table 3.1: Details of cell lines and patients

Cell lines were derived as detailed in Chapter 2, information regarding the age of the patient and mutation is shown in Table 3.1. Cells were routinely cultured on tissue culture plastic (plates or vented cap flasks, Corning, Schiphol-Rijk, Netherlands) coated with human fibronectin (Chemicon,

Merck Millipore, Nottingham, UK) to 0.5 µg/cm2 and vitronectin (Life Technologies, Paisley, UK) to 0.1 µg/cm2, in RPMI-1640 5.5 mM glucose (mixed from 11 mM glucose RPMI, Sigma-Aldrich and

0 mM glucose RPMI, Gibco, Life Technologies to 5 mM) with 10% FBS. Cultured cells were kept in a humidified incubator at 37°C, 5% CO2. Subculturing/passage was performed when cells reached approximately 70% confluency - cells were washed with Dulbecco’s phosphate buffered saline (dPBS), and detached from tissue culture plastic with 0.05% trypsin-EDTA (Sigma-Aldrich) for 3 minutes.

Trypsinisation was halted using 1 mg/ml soybean trypsin inhibitor (Gibco). Cells were pelleted at

50 x g and transferred to new vessels at a cell line and condition specific density, between 1:2 and

1:5. Medium was changed every 48 hours. Brightfield images were taken on an Olympus CKX41 microscope using 4x and 10x objectives and captured with QCapture Pro software. All experiments were performed on CHI cells between passages 5 and 14, and on adult cells between 4 and 9; at least one passage prior to senescence.

For growth curves, 2 x 102 cells were seeded for each well, in uncoated 24-well plates and 3 individual wells were counted at each time point to minimise variation. Cell counts were performed using trypan blue exclusion on a haemocytometer. Low glucose culture was in 2 mM glucose RPMI with 10% FBS (mixed as above to 1.5 mM glucose, 0.5 mM glucose from FBS); high insulin culture was in RPMI with 10% FBS (38 nM insulin determined by ELISA) supplemented with 200 nM insulin (Life Technologies).

110 3.3.2 Derivation of adult pancreatic mesenchymal stem cell (A pMSC) lines

Purified islets from adult organ donors were received from DRWF Human Islet Isolation Facility,

University of Oxford, when preparations were of insufficient purity or viability for transplantation.

The islets were directly put into 6-well tissue culture-treated plate, around 100 islets per well, and cultured as per the CHI pMSC lines

3.3.3 Flow Cytometry (propidium iodide)

Cells were harvested by trypsinisation, spun and resuspended in dPBS. A cell count was performed, the cells were spun again and then resuspended to 2.5 x 102 cells/ml in 70% ethanol whilst vortexing.

Cells were stored in ethanol at 4°C for between 4 hours and 2 weeks. The cell suspension was centrifuged and cells washed in dPBS. They were then resuspended in PBS with 2.5 g/ml RNase

(Invitrogen, Life Technologies) and 40 g/ml propidium iodide (Invitrogen). Cells were analysed on a Beckman Coulter Cyan ADP with 635nm excitation. The percentage of cells in each phase was analysed on Modfit LT after gating by forward and side scatter, and by pulse height and area (doublet discrimination - show in supplementary Figure 3.8B). Statistical analysis was by one way ANOVA with Bonferroni post-hoc test separately for each phase.

3.3.4 Immunocytochemistry (ICC)

Cells were cultured on glass coverslips until of a suitable density and were fixed in 4% paraformalde- hyde for 20 minutes. Cells were permeabilised in 0.3% Triton x-100 (Sigma-Aldrich) and non-specific staining was prevented by incubation in 10% NGS in PBS. Primary antibodies were applied as Ta- ble 3.2 for 1 hour in 0.1% Triton x-100, 3% NGS in PBS and washed off in PBS/0.1% Tween-20

(Sigma-Aldrich). Fluorophore-conjugated secondary antibody, specific to the species the primary antibody was raised in, was applied as Table 3.2 in 0.1% triton x-100, 3% NGS in PBS for 1 hour.

Cells were washed in PBS/0.1% Tween-20, then distilled water and allowed to dry. Coverslips were mounted using Pro Long Gold antifade reagent (Life Technologies) with DAPI. Slides were viewed using an Olympus BX51 upright microscope using 20x 0.50 UPlanFLN, 40x 0.75 UPlanFLN or

60x 1.25 UPlanFLN (Ph 3) objectives and captured using a Coolsnap EZ camera (Photometrics) through MetaVue Software (Molecular Devices). Filter sets for DAPI (31000v2), FITC (41001) and

Cy3 (41007a) were used to prevent bleed through of colour channels whilst maximising signal. Images were processed and analysed using ImageJ(http://rsb.info.nih.gov/ij). To quantify nuclear stains of p27Kip1, 5 fields were imaged, which were selected without first looking down the microscope, from both central and peripheral regions of the coverslips to minimise bias, and the colour channels were

111 separated. DAPI could be quantified by the analyse particles function within ImageJ, but p27Kip1 could not be due to the difference between nuclear and cytoplasmic intensity not being consistent across the fields, so this was counted by manually marking nuclei and expressing these as a per- centage of the total nuclei determined by DAPI. Statistical analysis was by one way ANOVA with

Bonferroni post-hoc test.

3.3.5 Western Blotting

Cells were lysed in RIPA buffer (comprising 150 mM sodium chloride, 1% triton x-100, 0.5% sodium deoxycholate, 0.1% sodium dodecyl sulphate (SDS), 50 mM Tris and the pH was adjusted to 8.0) with protease inhibitors (Promega, Madison, USA) on ice with agitation, cells were centrifuged at 16000 x g to pellet cell debris leaving protein in the supernatant. Protein concentration was assessed using

Bradford reagant (Sigma-Aldrich) and 20 g of total protein was added to laemmli buffer (comprising

4% SDS, 10% 2-mercaptoethanol, 20% glycerol, 0.004% bromophenol blue, 0.125 M Tris, with the pH adjusted to 6.8) and separated on a denuaturing poly-acrylamide stacking gel (BioRad, Hemel

Hempstead, UK) by SDS electrophoresis in running buffer (comprising 25 mM Tris, 190 mM glycine,

0.1% SDS). Protein from the gel was transferred to a methanol activated polyvinylidene fluoride

(PVDF) membrane (BioRad) by electroblotting. The membrane was blocked in 5% (w/v) milk powder (Sigma), or 5% (w/v) bovine serum album (Sigma) for CDK1/p-CDK1 only, in Tris buffered saline with TBS/0.1% Tween-20. The primary antibody was added as Table 3.2 (dilutions are given where the manufacturer does not provide a concentration), diluted in blocking solution for an overnight incubation at 4°C. Membrane was washed 3 times in TBS/0.1% Tween-20, then incubated in the HRP conjugated secondary antibody (specific for the animal the primary antibody was raised in), diluted in blocking solution as Table 3.2 for one hour at room temperature. Membrane was washed three times in TBS/0.1% Tween-20 and bound antibodies visualised using the enhanced chemiluminescent (ECL) system (HRP Substrate, Millipore). Membrane was exposed to Biomax

XAR film (Kodak) and developed in a Mini Med Film Processor (ImageWorks). Densitometry analysis was performed on ImageJ, with normalisation to the housekeeping protein β-actin, followed by either the adult control or to standard glucose/standard insulin as appropriate. Statistical analysis was by one way ANOVA with Bonferroni post-hoc test.

3.3.6 Ki67 stain and count

Ki67 staining and counting on patient tissue was performed as previously described (Salisbury et al.,

2015). Briefly, the tissue was fixed in 4% paraformaldehyde and embedded in paraffin wax; 5 µm sections were used for staining. Tissue was de-waxed with xylene and rehydrated with an ethanol gradient, antigen retrieval was by a 20 minute boil in 10mM citric acid buffer (pH 6.0). Permeabili-

112 sation was with 0.1% Triton X-100 and non-specific staining was prevented by incubation with 10% horse serum and 3% hydrogen peroxide. Primary antibody, as Table 3.2, was incubated overnight at

4°C in PBS and washed off in PBS/0.1% Tween-20. Biotinylated secondary antibody was applied as

Table 3.2 in 0.1% triton x-100, 3% NGS in PBS for 1 hour. The slide was washed and streptavidin- conjugated horse radish peroxidase (Vector, CA, USA) applied for 1 hour. Finally, the stain was developed using 3,3-Diaminobenzidine (Sigma-Aldrich) and the slide dehydrated through ethanol and xylene and mounted. Images were acquired and digitized by a 20x/0.80 Plan Apo objective using the 3DHISTECH Pannoramic 250 Flash II slide scanner. Pannoramic Viewer and HistoQuant software packages (both 3DHISTECH) were used for data analysis and high-content cell counting.

Antigen Species ICC Con- Blot Con- Manufacturer centration centration (µg/ml) (µg/ml) β-actin Rabbit N/A 1:5000 Cell Signalling (Leiden, The Netherlands)) CyclinB1 Mouse N/A 1.33 Santa Cruz (CA, USA) CDK1 Mouse N/A 1:1000 Cell Signalling CDK6 Rabbit 0.33 N/A Santa Cruz Ki67 Mouse 1:100 N/A Novocastra (Wetzlar, Ger- many) p-CDK1 Rabbit N/A 1:1000 Cell Signalling p27Kip1 Rabbit 1 0.5 Santa Cruz Biotin Horse 3.75 N/A Vector anti- mouse Cy2 Goat 5 N/A Jackson/Abcam (Cambridge, anti- UK) rabbit Cy5 Goat 2.5 N/A Eurogentec anti- mouse Cy5 Goat 2.5 N/A Abcam anti- rabbit HRP Goat N/A 1:10000 Cell Signalling anti- mouse HRP Goat N/A 1:10000 Cell Signalling anti- rabbit

Table 3.2: Antibodies used in the study

113 3.4 Results

3.4.1 Increased proliferation in CHI tissue and CHI-derived pMSCs

We had previously derived mesenchymal stem cell (MSC) lines from neonatal patients with CHI, see chapter 2. This diffuse form of the disease features high rates of proliferation throughout all parts of the pancreas, including in the normally non-proliferative endocrine cells. We hypothesised that this feature may be observed in the derived cell lines as we needed to passage the cell lines more frequently or at higher split ratios than the adult islet derived MSCs.

6 s l l e c

e 4 v i t i s o p

7 2 6 i K % 0 l I l I o H o H tr C tr -C n - n o F o A c c th th n n o o m m 0 4 1

Figure 3.1: Increased proliferation in CHI tissue compared to age-matched controls The A-CHI and F-CHI patient tissue showed an increased percentage of Ki67 positive nuclei compared to the closest age-matched control, percentages shown are for the average of all fields counted by high content analysis so n=1 for each patient/control.

We first wanted to confirm that the high rates of proliferation are also seen in the non-diffuse forms of CHI. Ki67 stains were done in the patient tissue from which the F-CHI and A-CHI lines were derived, and the nearest available aged control, shown in Figure 3.1. These showed a fold change of 7.4 and 1.5 times increased Ki67 for A-CHI and F-CHI, respectively. This therefore suggests increased proliferation in the focal and atypical CHI tissue compared to age matched control, further samples would be needed to confirm this trend. Proliferation was not documented for D-CHI as the

fixed tissue was of insufficient quality, but it is likely that it would also be high as seen in other diffuse CHI patients (Salisbury et al., 2015).

As shown in Figure 3.2 we saw higher rates of proliferation in all 3 CHI derived MSC lines than in

114 the adult derived MSCs. The A-CHI MSCs had the fastest proliferation rates by far, with a doubling time of 38 hours, to the nearest hour, but higher rates were also seen in D-CHI (doubling time 68 hours) and F-CHI (doubling time 68 hours). The adult-derived cells had a doubling time of 78 hours making them the slowest to proliferate1.

Figure 3.2: Increased proliferation in CHI-derived MSCs compared to adult controls CHI-derived MSCs showed higher proliferation than adult MSCs: cells were counted across 5 days (A) shows an average of 3 individual growth curves for each of the CHI cell lines at passages 10, 11 and 12 in F-CHI, 8, 10 and 15 in A-CHI, 5, 7 and 11 in D-CHI; and an average of 1 growth curve for 3 unique adult cell lines as passages 3, 5 and 6. Error bars show the standard deviation of the 3 replicates. * represents p<0.05, ** p<0.01 (B) shows the calculations used for estimating doubling times, with the exponential curves generated in Microsoft Excel.

Whilst the doubling times of D-CHI and F-CHI were identical, slightly more cells were seen at

1Doubling times were calculated from days 1 - 3 of the growth curves to allow direct comparison, as this is the only period in which exponential growth was seen across all cell lines. However, if the doubling time is calculated from days 1 - 4 for the adult cell lines which is the total exponential period for these, the doubling time is 89 hours which may therefore be more accurate.

115 each time point in D-CHI. This difference was not significant at any time point, including day 5 where proliferation appeared to have ceased in F-CHI. Over the course of growing this cell line, occasional higher split ratios were required though suggesting the difference, whilst small may be real. This is likely accounted for by better survival and attachment after passage in D-CHI as the mean number of cells observed at day 1 was 17680 in D-CHI (88.4% of the total cells seeded) and 15360 in F-CHI

(76.8% of the cells seeded).

The difference in proliferation rates between CHI-derived and adult cells may be partially ac- counted for by the age difference, as neonatal tissue would normally be more proliferative than adult tissue. However, within the 3 CHI patients there was no correlation between age at the point of surgery and proliferation rate - A-CHI 17 months, D-CHI 7 weeks, F-CHI 4 months.

3.4.2 Cell cycle regulation in CHI MSCs

The differences in proliferation rates between adult-derived and CHI-derived cells could be related to either the length of the cell cycle or the proportion of cells in the cell cycle (rather than G0). To investigate which we analysed the cell cycle stages in each cell line using propidium iodide and flow cytometric analysis, the averaged results of which, as well as the full plots, are shown in Figure 3.3.

This, and subsequent experiments, were performed four days after seeding. Whilst day 3 would have been optimal (significantly higher numbers of cells in all three CHI lines compared to adults) in order to minimise variability, the adult lines had such poor survival and growth that there was insufficient cell density for analysis at day three.

The cell cycle analysis showed significantly more cells in G2- or M-phase in the F-CHI cells com- pared to the adult cells (mean in F-CHI, 13.28% vs. adult, 10.55%). No other significant differences were seen, but the increase in the number of F-CHI cells in G2- or M-phase may be related to a slight decrease in G0- or G1-phase, though this difference was not significant. The cell cycle phases are calculated as percentages of the total cells, so where one phase increases there must also be a decrease in at least one of the other phases. In both D-CHI and A-CHI lines there appeared to be a slight decrease in the number of cells in G0- or G1-phase, and G2- or M-phase with this being balanced by an increase in S-phase. Again these changes were not significant as there was too much variability, further samples may reveal if there are indeed any changes to the cell cycle.

Despite the variability, of interest was the trend of a slight decrease seen in G0/G1 across each of

116 Figure 3.3: Alterations to cell cycle control in CHI-derived MSCs compared to adult controls CHI-derived and adult MSCs had a similar but slight differences in the proportion of cells in each cell cycle phase: (A) the proportion of cells in each cell cycle phase was assessed 4 days after seeding by propidium iodide flow cytometry analysis, with the mean of three experiments shown from passages 4, 10 and 12 in F-CHI, 10, 12 and 13 in A-CHI, 7, 11 and 12 in D-CHI and 6, 7 and 9 in adult pMSCs. Error bars show the standard deviation of the 3 replicates, * p<0.05. (B) shows the individual graphs for each replicate with the curves that were assigned by ModfitLT. (C) Gating was applied as shown to ensure only single cells were analysed.

117 the three CHI-derived lines compared to the adults (mean in adult 83.80%, F-CHI 78.81%, D-CHI

78.97%, A-CHI 80.47%). This could partially explain the differences in proliferation rates if more adult cells were in G0 or G1-phase rather than actively proliferating, but could not be confirmed.

The G1/S transition is the point at which commitment to division occurs. The networks of cell cycle proteins are complex so to probe the observed differences further we first looked at the proteins

Kip1 known to be altered in CHI - CDK6 and p27 - regulators involved in G1/S transition.

The average results of the cell cycle analysis are shown in Figure 3.3. To get these averages, curves were fit to the traces from the flow cytometer by ModfitLT, which is shown in Figure 3.3B.

The curves were generally a good fit, but particularly in D-CHI there is a shoulder on the G1- peak of cells which were not included as the software could not determine if these fit with G1- or S-phase. It was suggested by the software that this could be due to additional genetic material, as occurs in cancerous cells but the cell lines were karyotypically normal, as discussed in Section 2.4.2. Panel C of Figure 3.3 shows the gating used for the analysis prior to curve fitting, which was to ensure only single cells were counted, so this also does not explain the uncounted cells. Analysis of the cell cycle with BRdU would likely be necessary to confirm or refute the results seen and to understand what this shoulder on the peak represents.

Kip1 3.4.3 Distribution of G1/S molecules - decreased nuclear p27 in CHI- derived pMSCs

To investigate the increased proliferation seen in CHI MSCs, we investigated cell cycle regulators.

We initially started with the regulators CDK6 and p27Kip1 which have been previously reported to be altered in CHI (Salisbury et al., 2015), in order to correlate the cell lines with the tissue.

CDK6 is located in increased numbers of nuclei in CHI β-cells, was expressed predominately in the cytoplasm of the CHI-derived pMSCs, shown in Figure 3.4. Some nuclear staining was seen in obviously mitotic cells (where condensed chromosomes could be seen with DAPI or where two smaller side by side nuclei were seen) but this is seen artifactually for many antibodies so it is un- clear whether this was a true stain. This suggested that CDK6 was likely not a key regulator of the cell cycle in these cells and so was not investigated further.

On the other hand, p27Kip1, a cell cycle inhibitor also shown to be in increased numbers of nuclei in CHI β-cells, as mentioned above, was found to be expressed in both the nucleus and the cytoplasm in the pMSCs. We found that there were more cells with nuclear p27Kip1 in adult pMSC cultures

118 Kip1 Figure 3.4: Altered levels of nuclear p27 in CHI MSCs The distribution of G1/S cell cycle regulators altered in CHI, in CHI-derived pMSCs and adult pMSCs: (A) Immunostaining of CDK6 showed it was expressed but appeared to be localised to the cytoplasm. Image shown is from D-CHI, four days after passage and the staining pattern was similar in all CHI and adult lines. (B) Immunostaining four days after passage showed the expression of p27Kip1 to be variable, with staining seen in the nucleus and cytoplasm, F-CHI image is representative for all three CHI lines. The white arrows indicate nuclei where positive stains of p27Kip1 can be seen, the red arrows where no clear nuclear stains can be seen. Rabbit panel shows the secondary antibody only stain, this is the same antibody used for panel A and B, pseudocoloured red in A and green in B. (C) Quantification of the number of cells with clear nuclear staining of p27Kip1 showed less positive nuclei in CHI cells compared to adult cells. Graph shows the mean of 3 passages for each CHI line and 3 unique adult lines, with standard deviations; * p<0.05, ** p<0.01. (D) The total levels of p27Kip1 by western blot were not significantly different between adult and CHI cells, a representative blot is shown and densitometry analysis of three experiments confirmed there was no difference. A-D are from passages 9, 10 and 11 in F-CHI, 10, 11 and 12 in A-CHI, 9, 10 and 12 in D-CHI and 3, 6 and 7 in adult pMSCs.

119 than in the CHI derived cell lines. However, the total amount of p27Kip1 protein was not significantly different and was potentially slightly increased, although this may just be due to high variability.

This therefore suggests there is either a lower amount of p27Kip1 per nuclei in the adult cells (which did not appear to be the case from the immunostaining intensity) or that there is a greater amount of cytoplasmic p27Kip1 in the CHI cells. The latter appears to be more likely based on these data, but subcellular fractionation would be needed to confirm this.

As p27Kip1 is known to act as a cell cycle inhibitor when it is located in the nucleus, the de- creased number of p27Kip1 positive nuclei in the CHI cells broadly correlates with their increased

Kip1 proliferation rates. The regulation by p27 is at the G1/S restriction point, but we saw an in- crease in the number of cells in G2/M in the F-CHI line, and a trend of an increase in the number of A-CHI and D-CHI cells in S-phase which was not solely accounted for by a decrease in G1 (i.e. the G1 to S phase transition). We therefore wanted to see if G2/M regulators could be playing a role in the increased proliferation seen in CHI cells; this had not previously been studied in CHI tissue.

3.4.4 Distribution of G2/M molecules - possible increase of CDK1 in CHI- derived pMSCs

We found that the G2/M regulators were quite variable in the cells, making analysis more challeng- ing. The most consistent factor was CyclinB1 - which showed no change across the CHI-derived and adult pMSCs, across three experiments.

CDK1 (also known as Cdc2) was more variable. The overall trend was that the levels of total

CDK1 were increased in the three CHI-derived, and this was the case in two of the three experiments, with averages of all three shown in Figure 3.5. In the third experiment (i.e. with the third adult control cell line) the levels of CDK1 were similar between the adult control cells and the CHI-derived cells, which is the main source of variation. The levels in CHI cells relative to each other were stable.

All adult cell lines were derived in the same way, from cadaveric donors aged above 40, and all three experiments were carried out on adult cells between passages 4 and 10 (as with the proliferation and cell cycle experiments) and so it is unclear why this adult line was different to the other two.

If this cell line was excluded from analysis, CDK1 was 14-fold higher in both F-CHI and D-CHI compared to the adult lines, and 24-fold higher in A-CHI compared to the adults, which is shown in

Figure 3.5E. This correlates closely with the relative proliferation rates seen in the three cell lines - where F-CHI and D-CHI are similar and A-CHI is almost twice as fast. More experiments with addi- tional adult lines would therefore be beneficial to find out whether CDK1 levels are increased and if so

120 Figure 3.5: Levels of G2/M regulators in CHI and adult MSCs G2/M regulators were not significantly different in CHI or adult MSCs: A-C show mean densitometry analysis of three experiments at passages 9, 10 and 11 in F-CHI, 10, 11 and 12 in A-CHI, 9, 10 and 12 in D-CHI and 3, 6 and 7 in adult pMSCs, error bars show standard deviation. (A) CDK1 showed a ten to fifteen fold increase on average in all CHI derived lines, but there was some variability so the difference was not significant. (B) Levels of inactive phosphorylated CDK1 were on average slightly increased in F-CHI and A-CHI, but high levels of variability were seen between experiments. The higher levels in CHI cells may be partially artefactual due to the very low levels of total CDK1 seen in the adult cells. (C) No differences were seen between the levels of CyclinB1 (D) An example of the blots from one experiment is shown - this pattern was similar in 2 of 3 experiments, in the third the adult cells had levels of CDK1 similar to the CHI cells. (E) shows the two blots using the other two adult control cell lines, for CDK1 and β-actin, to demonstrate the variability that was present in CDK1 is due to the adult sample.

121 are involved with increased proliferation in CHI, unfortunately only three donors were available to us.

CDK1 activity is regulated by phosphorylation of Tyr15, the phosphorylated form is inactive, and the de-phosphorylated form is active and able to bind CyclinB1. We therefore wanted to find out whether there were merely increased levels of inactive CDK1 or whether the active form was increased and could therefore be driving G2/M transition. As western blotting is semi-quantitative, levels must be normalised to a control. This made analysis of phospho-CDK1 levels challenging as there were minimal levels of total CDK1 seen in two of three experiments, so phosphorylated CDK1 was virtually undetectable. The proportions of phosphorylated to total CDK1 in CHI-derived cells compared to adults are therefore very likely to be over-estimations. So whilst it appears that there is an increased proportion of inactive CDK1 in both F-CHI and A-CHI, this should be considered with this caveat.

3.4.5 Glucose and insulin affect p27Kip1 protein levels and localisation

Following the above experiments, we wanted to understand how the basal alterations to the cell cycle seen in the CHI-derived cells related to the actual situation in the CHI tissue. In particular, we wanted to understand why there was increased nuclear p27Kip1 in CHI tissue, but decreased in the CHI cells. One explanation for the increased nuclear p27Kip1 in CHI tissue is that it is acting as a chaperone for CDK6 and so helping to promote proliferation. However, whilst CDK6 was seen to co-stain with Ki67 in the CHI in line with it promoting proliferation, p27Kip1 was not observed to co-stain with Ki67 (Salisbury, 2014). An alternative explanation is therefore that the increased insulin levels are driving proliferation and the increase in p27Kip1 is a mechanism for the tissue to

‘put the brakes on’, as unlike cancer the regulatory pathways should still be functioning.

To investigate these hypotheses we cultured the cells with the standard conditions, i.e. 5.5 mM glucose and 38 nM insulin (from serum only), supplemented with 200 nM insulin to isolate the effect of the insulin, and supplemented with 200 nM insulin and with lowered (2 mM) glucose to simulate

CHI. The culture conditions were started when the cells were passaged, and the cells assayed at day four as above; the experiments were carried out on cells already in culture that were potentially adapted to the standard glucose and standard insulin media.

As expected we found insulin to have a mitogenic effect on the CHI cell lines, as shown in Figure

3.6A. When glucose levels were lowered, insulin was still able to drive higher rates of proliferation although not as strongly as with physiological levels of glucose. This may suggest that the increased

122 insulin levels in the CHI pancreas are able to cause increased proliferation despite lowered glucose levels (or in the setting of more normal glucose levels at the point of surgery).

We found that p27Kip1 was affected by both the altered glucose and insulin levels. Interestingly total levels of p27Kip1 were unchanged by increasing the insulin levels shown in Figure 3.6B, so this is not the method by which the increased proliferation is driven. Further, there was a dramatic decrease in the total levels of p27Kip1 in response to decreasing the glucose levels in all three cell lines. Looking at the distribution patterns of p27Kip1 in these conditions, there was a trend of decreased nuclear p27Kip1 when insulin levels were increased, but slightly more nuclei stained in the lower glucose condition than standard glucose. This trend is shown in Figure 3.6 panel C, each cell line is shown individually as the pattern was present in each line although the changes were of different magnitudes.

Together these data suggest that increasing insulin causes p27Kip1 to be sequestered in the cy- toplasm thereby allowing cell cycle progression, either by translocation from the nucleus to the cytoplasm or inhibition of shuttling to the nucleus. It was interesting that the trend of nuclear staining of p27Kip1 was the inverse pattern of the proliferation rates, but the total levels were not.

The role of p27Kip1 in the cytoplasm of the CHI cells is not known but may help us to understand the response to CHI conditions.

123 Figure 3.6: Glucose and insulin concentrations determine p27Kip1 localisation Culturing CHI pMSCs in variable glucose and insulin concentrations to simulate CHI resulted in increased growth and altered p27Kip1 expression after four days, experiments were from each of the three lines at passage 12 in F-CHI, 13 in A-CHI and 10 in D-CHI: (A) Culturing cells in low glucose media with high insulin resulted in a significant increase in growth of around 15% after four days, culturing cells in standard glucose media with high insulin resulted in around a 30% increase in growth which was significantly increased over both the standard glucose/standard insulin and the low glucose/high insulin condition. Graphs show the mean of the three unique CHI lines, with cell counts having been normalised to the standard glucose/standard insulin condition to account for the variability in growth rates between the three lines, error bars show standard deviation, * p<0.05, *** p<0.005. (B) Western blotting with densitometry analysis of p27Kip1 showed significantly decreased levels of p27Kip1 in the low glucose/high insulin conditions compared to both standard glucose/standard insulin and standard glucose/high insulin in all three CHI lines, bars shown the mean of the densit- ometry of one experiment in each of the three lines, with the blot of the three lines shown above. (C) Analysis of the percentage of cells with clear nuclear p27Kip1 as in Figure 3.4, showed a trend of fewer positive nuclei in high insulin, with the fewest number of positive nuclei in cells cultured in standard glucose. (A)-(C) shown the mean of one experiment in the three CHI cell lines.

124 3.5 Discussion

CHI is a disease which features increased rates of proliferation, along with increased nuclear localisa- tion of two G1/S-phase regulators (Salisbury et al., 2015). The precise mechanism of this increased proliferation has not yet been elucidated, but may be related to the increased levels of insulin acting as a mitogen. Insulin can signal via the mTORC pathway which is known to cause β-cell prolifera- tion (Gu et al., 2011). Sirolimus, an mTORC inhibitor has been used as a treatment for CHI with some success (Senniappan et al., 2014, Minute et al., 2015). The mechanism of action of sirolimus in

CHI is unknown and could be related to insulin induced proliferation being reversed or due to inhition of aberrant mTORC expression seen in CHI (Alexandrescu et al., 2010). Research into mechanisms of CHI have so far been limited to short term ex vivo cultures or fixed tissue with unknown physio- logical conditions. Rodent models for CHI have limited suitability given the differences seen both in islet structure (Cabrera et al., 2006) and cell cycle regulation between the human and rodent pan- creas (Fiaschi-Taesch et al., 2009). To allow us to investigate the alterations in the cell cycle in the

CHI pancreas we developed a stem cell model which retained features of the pancreas (see chapter 1).

In line with the increased proliferation seen in the pancreas of CHI patients, we observed in- creased proliferation rates in stem cells derived from the CHI patients’ pancreas when compared to adult control cells. This difference was despite identical culture conditions showing there to be underlying differences in the cells. This may have been partially due to the age of the patients, unfixed neonatal control tissue was not available. The rates of proliferation between CHI cells was not correlated to the individual patients’ ages with the A-CHI line showing the fastest proliferation despite the patient being over 1 year older than the other two. The only published proliferation rates for MSCs derived from non-adults, are for foetal pMSCs which had an average 30 hour doubling time (Hu et al., 2003), however these cells were cultured in low serum but with increased insulin, epi- dermal growth factor and platelet-derived growth factor making the proliferation rates incomparable.

The cell cycle distribution was also studied in the foetal pMSCs, over 90% of cells were in

G0/G1 (Hu et al., 2003), suggesting a lower proportion of cells were actively cycling than in either the CHI cells (<80% in G1) or the adult cells (around 85% in G1). If this is the case then the faster doubling time in the foetal cells would be indicative of a shorter cell cycle time rather than increased proliferation. In contrast the CHI cells had both a faster doubling time and a higher proportion of cells proliferating compared to the adult cells, suggesting that the increased proliferation may be due to increased entry to the cell cycle. The disadvantage of using propidium iodide is that it provides a static picture at this particular time point, rather than dynamic information, such as cell cycle length. It can therefore only be considered a snapshot of growth status in the culture at that time

125 point. It also does not allow discrimination between G0- and G1-, and G2- and M-phase. Further evidence for increased cell cycle entry, comes from the decreased nuclear expression of p27Kip1 in the CHI cells compared to the adult cells if we consider it to be fulfilling its primary role of a G1/S inhibitor rather than as a cyclin-CDK assembly factor. As a CKI p27Kip1 inhibits the action of cyclinD-CDK4/6 complexes. CDK6 did not seem to be active (located to the nucleus) in the CHI cells suggesting that p27Kip1 was not acting on this, to find out whether CDK4 is instead fulfilling the role, or if this was due to a problem with the antibody used, would require further studies.

G2/M regulators have not been studied within the CHI pancreas previously. The differences in the proportion of cells in each phase of the cell cycle could be accounted for by increased pro- gression through M-phase so we investigated the primary promoters of entry to M-phase - CDK1 and CyclinB1 which act together as a complex. Both of these factors are reported to be expressed stably throughout the cell cycle with the majority of regulation post-translational - by sub-cellular localisation and phosphorylation of CDK1. CyclinB1 was expressed at a consistent level across all the CHI and adult cell lines, but its partner, CDK1 was expressed at a much higher level in the CHI cells compared to two of three adult lines. It was unclear why there were higher levels of CDK1 in one adult line compared to the other two, more adult controls would need to be used to confirm if the increased levels of CDK1 in the CHI lines were consistent and significant. Going forward, it would be beneficial to find out if there is increased CDK1 seen in fixed CHI tissue and in which cells, to confirm if the phenomenon seen in the cell lines holds true in vivo; unfortunately the antibody used in this study was not suitable for immunohistochemistry. This would clarify whether CDK1 is involved with hyperproliferation in CHI. Investigation into why CDK1 is increase but not its binding partner CyclinB1 and what effect this is having in CHI would then be warranted using these stem cell lines.

As we saw differences in p27Kip1 in the CHI cells, which may account for the basal differences in proliferation, we need to correlate this to what has been observed in patients. The expression levels seemed to be opposite in vivo and in vitro, with the increased levels of nuclear localisation in vivo perhaps indicating its use as an assembly factor but the decreased levels in vitro showing a predominate use as an inhibitor. However, p27Kip1 did not co-localise with Ki67 in vivo, which one would expect it to if it were assembling the G1/S promoting molecules. We therefore hypoth- esised that levels of nuclear p27Kip1 were increased due to the cells accumulating it in response to the over-proliferation occurring. By stimulating the cells with insulin, as occurs in CHI we aimed to recapitulate this but in fact saw a trend of further decreased in nuclear p27Kip1, mirroring the increased proliferation. This was still seen despite a low glucose concentration, which is also seen in

126 CHI, although there was a significant decrease in total p27Kip1 in response to low glucose which did not affect its localisation. This suggests that the nuclear-cytoplasmic distribution is most important in regulating proliferation but does invite the question of how the decreased total levels of p27Kip1 with low glucose are affecting the cells, what regulatory role this plays and how the cytoplasmic- nuclear distribution of the protein is regulated in response to the insulin stimulus. This mirrors the

findings of Zhang et al. (2014), who found that increasing glucose cause an increase in total p27Kip1, although they found this to cause a decrease in proliferation, which was also what we saw with low glucose despite the apparent decrease in the inhibitor protein. This still did not match the situation seen in the CHI tissue, so prolonged culture in these conditions may be required in order for the cells to accumulate p27Kip1. Alternatively we may see this increase when cells are cultured until con- tact inhibition occurs which may be similar to the high cellular density of an over-proliferative organ.

In conclusion we have developed a pancreatic stem cell model from patients with CHI which demonstrates increased rates of proliferation, similar to what has been observed in patients. Increased proliferation seems to be due in part to an increased progression of cells through the cell cycle with a decrease in nuclear p27Kip1 and an increase in total CDK1. The cells respond dynamically to glucose and insulin as expected, with proliferation changes similar to β-cells in vivo (Paris et al.,

2003, Annicotte et al., 2009). Both glucose and insulin affected p27Kip1 but in different ways, the interplay between these two stimuli and the resulting effects on p27Kip1 may help to explain some of the pathology of CHI. We therefore propose CHI pMSCs as a novel model which can be manipulated to understand the increased proliferation seen in CHI and test potential treatment options. Where possible, observations from CHI pMSCs should be confirmed in fixed tissue though as some variation was seen and there are differences between the different cell types of the pancreas in terms of cell cycle (Cai et al., 2014) and response to glucose (Bonner-Weir et al., 1989).

127 3.6 References

S. Alexandrescu, N. Tatevian, O. Olutoye, and R. E. Brown. Persistent hyperinsulinemic hypo-

glycemia of infancy: constitutive activation of the mtor pathway with associated exocrine-islet

transdifferentiation and therapeutic implications. International Journal of Clinical and Experi-

mental Pathology, 3(7):691–705, 2010.

J.-S. Annicotte, E. Blanchet, C. Chavey, I. Iankova, S. Costes, S. Assou, J. Teyssier, S. Dalle,

C. Sardet, and L. Fajas. The CDK4-pRB-E2F1 pathway controls insulin secretion. Nat Cell Biol,

11(8):1017–1023, 2009.

J.-B. Arnoux, V. Verkarre, C. Saint-Martin, F. Montravers, A. Brassier, V. Valayannopoulos,

F. Brunelle, J.-C. Fournet, J.-J. Robert, Y. Aigrain, C. Bellanne-Chantelot, and P. de Lonlay.

Congenital hyperinsulinism: current trends in diagnosis and therapy. Orphanet Journal of Rare

Diseases, 6(1):1750–1172, 2011.

A. Belfiore and R. Malaguarnera. Insulin receptor and cancer. Endocrine-Related Cancer, 18(4):

R125–R147, 2011.

C. Bertoli, J. M. Skotheim, and R. A. M. de Bruin. Control of cell cycle transcription during G1

and S phases. Nat Rev Mol Cell Biol, 14(8):518 – 528, 2013.

A. Besson, H. C. Hwang, S. Cicero, S. L. Donovan, M. Gurian-West, D. Johnson, B. E. Clurman,

M. A. Dyer, and J. M. Roberts. Discovery of an oncogenic activity in p27Kip1 that causes stem

cell expansion and a multiple tumor phenotype. Genes & Development, 21(14):1731–1746, 2007.

S. W. Blain, E. Montalvo, and J. Massagu. Differential Interaction of the Cyclin-dependent Ki-

nase (Cdk) Inhibitor p27Kip1 with Cyclin A-Cdk2 and Cyclin D2-Cdk4 . Journal of Biological

Chemistry, 272(41):25863–25872, 1997.

S. Bonner-Weir, D. Deery, J. L. Leahy, and G. C. Weir. Compensatory Growth of Pancreatic β-Cells

in Adult Rats After Short-Term Glucose Infusion. Diabetes, 38(1):49–53, 1989.

O. Cabrera, D. M. Berman, N. S. Kenyon, C. Ricordi, P.-O. Berggren, and A. Caicedo. The unique

cytoarchitecture of human pancreatic islets has implications for islet cell function. Proceedings of

the National Academy of Sciences of the United States of America, 103(7):2334–2339, 2006.

E. Cai, C. Luk, X. Wu, S. Schroer, S. Shi, T. Sivasubramaniyam, J. Brunt, E. Zacksenhaus, and

M. Woo. Rb and p107 are required for alpha cell survival, beta cell cycle control and glucagon-like

peptide-1 action. Diabetologia, 57(12):2555–2565, 2014.

128 E. P. Cai, X. Wu, S. A. Schroer, A. J. Elia, M. C. Nostro, E. Zacksenhaus, and M. Woo. Retinoblas-

toma tumor suppressor protein in pancreatic progenitors controls α- and β-cell fate. Proceedings

of the National Academy of Sciences, 110(36):14723–14728, 2013.

A. Cerqueira, A. Martn, C. E. Symonds, J. Odajima, P. Dubus, M. Barbacid, and D. Santamara.

Genetic Characterization of the Role of the Cip/Kip Family of Proteins as Cyclin-Dependent

Kinase Inhibitors and Assembly Factors. Molecular and Cellular Biology, 34(8):1452–1459, 2014.

M. Cheng, P. Olivier, J. Diehl, M. Fero, M. F. Roussel, J. M. Roberts, and C. J. Sherr. The p21Cip1

and p27Kip1 CDK ‘inhibitors’ are essential activators of cyclin D-dependent kinases in murine

fibroblasts. The EMBO Journal, 18(6):1571–1583, 1999.

P. P. Claudio, A. Zamparelli, F. U. Garcia, L. Claudio, G. Ammirati, A. Farina, A. Bovicelli,

G. Russo, G. G. Giordano, D. E. McGinnis, A. Giordano, and G. Cardi. Expression of Cell-

Cycle-regulated Proteins pRb2/p130, p107, p27kip1, p53, mdm-2, and Ki-67 (MIB-1) in Prostatic

Gland Adenocarcinoma. Clinical Cancer Research, 8(6):1808–1815, 2002.

P. De Meyts, C. Christoffersen, B. Ursa, B. Wallach, K. Grant W. Bookernskov, F. Yakushiji, and

R. Shymko. Role of the time factor in signaling specificity: application to mitogenic and metabolic

signaling by the insulin and insulin-like growth factor-I receptor tyrosine kinases. Metabolism:

clinical and experimental, 44(10 Suppl 4):2–11, 1995.

A. Denley, E. R. Bonython, G. W. Booker, L. J. Cosgrove, B. E. Forbes, C. W. Ward, and J. C.

Wallace. Structural Determinants for High-Affinity Binding of Insulin-Like Growth Factor II to

Insulin Receptor (IR)-A, the Exon 11 Minus Isoform of the IR. Molecular Endocrinology, 18(10):

2502–2512, 2004.

M. J. Dunne, C. Kane, R. M. Shepherd, J. A. Sanchez, R. F. James, P. R. Johnson, A. Aynsley-

Green, S. Lu, J. P. Clement, K. J. Lindley, S. Seino, L. Aguilar-Bryan, G. Gonzalez, and P. J. Milla.

Familial Persistent Hyperinsulinemic Hypoglycemia of Infancy and Mutations in the Sulfonylurea

Receptor. New England Journal of Medicine, 336(10):703–706, 1997.

M. J. Dunne, K. E. Cosgrove, R. M. Shepherd, A. Aynsley-Green, and K. J. Lindley. Hyperinsulinism

in Infancy: From Basic Science to Clinical Disease. Physiological Reviews, 84(1):239–275, 2004.

S. Fatrai, L. Elghazi, N. Balcazar, C. Cras-Mneur, I. Krits, H. Kiyokawa, and E. Bernal-Mizrachi.

Akt Induces β-Cell Proliferation by Regulating Cyclin D1, Cyclin D2, and p21 Levels and Cyclin-

Dependent Kinase-4 Activity. Diabetes, 55(2):318–325, 2006.

N. Fiaschi-Taesch, T. A. Bigatel, B. Sicari, K. K. Takane, F. Salim, S. Velazquez-Garcia, G. Harb,

K. Selk, I. Cozar-Castellano, and A. F. Stewart. Survey of the Human Pancreatic β-Cell G1/S

129 Proteome Reveals a Potential Therapeutic Role for Cdk-6 and Cyclin D1 in Enhancing Human

β-Cell Replication and Function In Vivo. Diabetes, 58(4):882–893, 2009.

J.-C. Fournet, C. Mayaud, P. de Lonlay, M.-S. Gross-Morand, V. Verkarre, M. Castanet, M. Devillers,

J. Rahier, F. Brunelle, J.-J. Robert, C. Nihoul-Fekete, J.-M. Saudubray, and C. Junien. Unbalanced

Expression of 11p15 Imprinted Genes in Focal Forms of Congenital Hyperinsulinism. The American

Journal of Pathology, 158(6):2177–2184, 2001.

S. Georgia and A. Bhushan. p27 Regulates the Transition of β-Cells From Quiescence to Proliferation.

Diabetes, 55(11):2950–2956, 2006.

E. Giovannucci, D. M. Harlan, M. C. Archer, R. M. Bergenstal, S. M. Gapstur, L. A. Habel, M. Pollak,

J. G. Regensteiner, and D. Yee. Diabetes and Cancer: A consensus report. Diabetes Care, 33(7):

1674–1685, 2010.

B. Glaser, F. Ryan, M. Donath, H. Landau, C. A. Stanley, L. Baker, D. E. Barton, and P. S.

Thornton. Hyperinsulinism caused by paternal-specific inheritance of a recessive mutation in the

sulfonylurea-receptor gene. Diabetes, 48(8):1652–1657, 1999.

B. Glaser, P. Thornton, T. Otonkoski, and C. Junien. Genetics of neonatal hyperinsulinism. Archives

of Disease in Childhood - Fetal and Neonatal Edition, 82(2):F79–F86, 2000.

Y. Gu, J. Lindner, A. Kumar, W. Yuan, and M. A. Magnuson. Rictor/mTORC2 Is Essential for

Maintaining a Balance Between β-Cell Proliferation and Cell Size. Diabetes, 60(3):827–837, 2011.

H. Harashima, N. Dissmeyer, and A. Schnittger. Cell cycle control across the eukaryotic kingdom.

Trends in Cell Biology, 23(7):345 – 356, 2013.

J.-C. Henquin, C. Sempoux, J. Marchandise, S. Godecharles, Y. Guiot, M. Nenquin, and J. Rahier.

Congenital Hyperinsulinism Caused by Hexokinase I Expression or Glucokinase-Activating Muta-

tion in a Subset of β-Cells. Diabetes, 62(5):1689–1696, 2013.

Y. Hu, L. Liao, Q. Wang, L. Ma, G. Ma, X. Jiang, and R. C. Zhao. Isolation and identification of

mesenchymal stem cells from human fetal pancreas. Journal of Laboratory and Clinical Medicine,

141(5):342 – 349, 2003.

D. Ish-Shalom, C. Christoffersen, P. Vorwerk, N. Sacerdoti-Sierra, R. Shymko, D. Naor, and

P. De Meyts. Mitogenic properties of insulin and insulin analogues mediated by the insulin recep-

tor. Diabetologia, 40(S2):S25–31, 1997.

M. Jackman, Y. Kubota, N. den Elzen, A. Hagting, and J. Pines. Cyclin A- and Cyclin E-Cdk

Complexes Shuttle between the Nucleus and the Cytoplasm. Molecular Biology of the Cell, 13(3):

1030–1045, 2002.

130 M. K. James, A. Ray, D. Leznova, and S. W. Blain. Differential Modification of p27Kip1 Controls

Its Cyclin D-cdk4 Inhibitory Activity . Molecular and Cellular Biology, 28(1):498–510, 2008.

S. A. Kassem, I. Ariel, P. S. Thornton, K. Hussain, V. Smith, K. J. Lindley, A. Aynsley-Green, and

B. Glaser. p57KIP2 Expression in Normal Islet Cells and in Hyperinsulinism of Infancy. Diabetes,

50(12):2763–2769, 2001.

R. N. Kulkarni, E.-B. Mizrachi, A. G. Ocana, and A. F. Stewart. Human β-Cell Proliferation and

Intracellular Signaling: Driving in the Dark Without a Road Map. Diabetes, 61(9):2205–2213,

2012.

J. LaBaer, M. D. Garrett, L. F. Stevenson, J. M. Slingerland, C. Sandhu, H. S. Chou, A. Fattaey, and

E. Harlow. New functional activities for the p21 family of CDK inhibitors. Genes & Development,

11(7):847–862, 1997.

Y. Lee, J. E. Dominy, Y. J. Choi, M. Jurczak, N. Tolliday, J. P. Camporez, H. Chim, J.-H. Lim,

H.-B. Ruan, X. Yang, F. Vazquez, P. Sicinski, G. I. Shulman, and P. Puigserver. Cyclin D1-Cdk4

controls glucose metabolism independently of cell cycle progression. Nature, 510(7506):547–551,

2014.

M. Malumbres, R. Sotillo, D. Santamaria, J. Galan, A. Cerezo, S. Ortega, P. Dubus, and M. Barbacid.

Mammalian Cells Cycle without the D-Type Cyclin-Dependent Kinases Cdk4 and Cdk6. Cell, 118

(4):493–504, 2004.

M. Minute, G. Patti, G. Tornese, E. Faleschini, C. Zuiani, and A. Ventura. Sirolimus Therapy in

Congenital Hyperinsulinism: A Successful Experience Beyond Infancy. Pediatrics, 136(5):e1373–6,

2015.

D. O. Morgan. Principles of CDK regulation. Nature, 374(6518):131–134, 1995.

T. Okada, C. W. Liew, J. Hu, C. Hinault, M. D. Michael, J. Krtzfeldt, C. Yin, M. Holzenberger,

M. Stoffel, and R. N. Kulkarni. Insulin receptors in β-cells are critical for islet compensatory

growth response to insulin resistance. Proceedings of the National Academy of Sciences, 104(21):

8977–8982, 2007.

A. Pal, T. P. Potjer, S. K. Thomsen, H. J. Ng, A. Barrett, R. Scharfmann, T. J. James, D. T.

Bishop, F. Karpe, I. F. Godsland, H. F. Vasen, J. Newton-Bishop, H. Pijl, M. I. McCarthy, and

A. L. Gloyn. Loss-of-Function Mutations in the Cell-Cycle Control Gene CDKN2A Impact on

Glucose Homeostasis in Humans. Diabetes, 2015.

131 M. Paris, C. Bernard-Kargar, M.-F. Berthault, L. Bouwens, and A. Ktorza. Specific and Com-

bined Effects of Insulin and Glucose on Functional Pancreatic β-Cell Mass in Vivo in Adult Rats.

Endocrinology, 144(6):2717–2727, 2003.

S. G. Rane, P. Dubus, R. V. Mettus, E. J. Galbreath, G. Boden, E. P. Reddy, and M. Barbacid.

Loss of Cdk4 expression causes insulin-deficient diabetes and Cdk4 activation results in β-islet cell

hyperplasia. Nat Genet, 22(1):44–52, 1999.

D. P. Rose and L. Vona-Davis. The cellular and molecular mechanisms by which insulin influences

breast cancer risk and progression. Endocrine-Related Cancer, 19(6):R225–R241, 2012.

R. J. Salisbury. Gene expression and cell cycle regulation in human pancreas development and

congenital hyperinsulinism. PhD thesis, University of Manchester, School of Medicine, 2014.

R. J. Salisbury, B. Han, R. E. Jennings, A. A. Berry, A. Stevens, Z. Mohamed, S. A. Sugden,

R. De Krijger, S. E. Cross, P. P. Johnson, M. Newbould, K. E. Cosgrove, K. P. Hanley, I. Banerjee,

M. J. Dunne, and N. A. Hanley. Altered Phenotype of β-Cells and Other Pancreatic Cell Lineages

in Patients With Diffuse Congenital Hyperinsulinism in Infancy Caused by Mutations in the ATP-

Sensitive K-Channel. Diabetes, 64(9):3182–3188, 2015.

D. Santamaria, C. Barriere, A. Cerqueira, S. Hunt, C. Tardy, K. Newton, J. F. Caceres, P. Dubus,

M. Malumbres, and M. Barbacid. Cdk1 is sufficient to drive the mammalian cell cycle. Nature,

448(7155):811–815, 2007.

A. Satyanarayana and P. Kaldis. Mammalian cell-cycle regulation: several Cdks, numerous cyclins

and diverse compensatory mechanisms. Oncogene, 28(33):2925–2939, 2009.

S. Senniappan, S. Alexandrescu, N. Tatevian, P. Shah, V. Arya, S. Flanagan, S. Ellard, D. Ram-

pling, M. Ashworth, R. E. Brown, and K. Hussain. Sirolimus Therapy in Infants with Severe

Hyperinsulinemic Hypoglycemia. New England Journal of Medicine, 370(12):1131–1137, 2014.

C. J. Sherr and J. M. Roberts. CDK inhibitors: positive and negative regulators of G1-phase

progression. Genes & Development, 13(12):1501–1512, 1999.

K. Shimomura, M. Tusa, M. Iberl, M. F. Brereton, S. Kaizik, P. Proks, C. Lahmann, N. Yaluri,

S. Modi, H. Huopio, J. Ustinov, T. Otonkoski, M. Laakso, and F. M. Ashcroft. A Mouse Model of

Human Hyperinsulinism Produced by the E1506K Mutation in the Sulphonylurea Receptor SUR1.

Diabetes, 62(11):3797–3806, 2013.

G. R. Stark and W. R. Taylor. Analyzing the G2/M Checkpoint. In Checkpoint Controls and Cancer,

volume 280 of Methods in Molecular Biology, pages 51–82. 2004.

132 V. Verkarre, J. C. Fournet, P. de Lonlay, M. S. Gross-Morand, M. Devillers, J. Rahier, F. Brunelle,

J. J. Robert, C. Nihoul-Fekete, J. M. Saudubray, and C. Junien. Paternal mutation of the sul-

fonylurea receptor (SUR1) gene and maternal loss of 11p15 imprinted genes lead to persistent

hyperinsulinism in focal adenomatous hyperplasia. The Journal of Clinical Investigation, 102(7):

1286–1291, 1998.

Z. Zhang, J. Li, X. Jiang, L. Yang, L. Lei, D. Cai, H. Zhang, and H. Chen. GLP-1 ameliorates the

proliferation activity of INS-1 cells inhibited by intermittent high glucose concentrations through

the regulation of cyclins. Molecular Medicine Reports, 10(2), 2014.

133 Chapter 4

Stem Cell Derived β-cells: a new resource for Congenital

Hyperinsulinism?

Sophie G Kellaway, Karolina Mosinska, Magdalena Mazur, Zainaba Mohamed,

Indi Banerjee, Alexander Ryan, Karen E Cosgrove and Mark J Dunne

134 4.1 Abstract

Diabetes is a chronic disease which cannot be cured and so requires novel long term treatments or methods for β-cell replacement. Current in vitro models show inadequate insulin secretion making the models challenging to work with, whilst rodent islets have multiple differences compared to hu- mans. We had previously derived mesenchymal stem cells (MSCs) from patients with congenital hyperinsulinism (CHI), a disease associated with excessive insulin secretion. To use the cells for disease modelling they must be differentiated to insulin-secreting cells. Following a survey of pro- tocols from the literature, we were able to differentiated the CHI MSCs to insulin expressing cells.

The addition of DKK1 to the protocol results in increased PDX1 and INS due to early induction of neuronal genes. The resulting cells secreted insulin but not in a glucose responsive manner. Further investigation will reveal if this is associated with CHI, or due to immaturity of the resulting β-cells.

The efficiency of differentiation of induced pluripotent stem cells (iPSCs) to β-cells has recently advanced and so MSC-derived iPSCs were explored as an alternative source for modelling CHI. We report the first derivation of an iPSC line from the pancreas of a CHI patient. The CHI iPSC line was phenotypically normal, with three germ layer differentiation and nuclear OCT4, SOX2 and NANOG.

Unlike a previous report, we did not find that deriving the iPSCs from the pancreas was epigeneti- cally favourable. Following published protocols, the cells were differentiated with high efficiency to definitive endoderm, and to PDX1 positive cells. As with the MSCs, mature insulin secreting β-cells were not obtained, though resulting cells did express glucagon and ABCC8, suggesting they were terminal islet cells. We anticipate that differentiated CHI MSCs and iPSCs may be used for drug testing with further work.

135 4.2 Introduction

CHI is an inherited disorder which causes profound hypoglycaemia due to unregulated insulin secre- tion (Dunne et al., 2004). There are three forms of CHI - focal, diffuse and atypical. All three forms share the phenotype of inappropriately high levels of insulin secretion and high proliferation however, with many patients undergoing a partial pancreatectomy in order to cure them of the disease at an early age. The inappropriate insulin secretion is typically due to constitutive cellular depolarisation and calcium influx due to non-functional ATP-sensitive potassium (KATP) channels (Dunne et al., 2004). Focal CHI is caused by a recessive mutation in the ABCC8 gene (which encodes the SUR1 subunit of the pancreatic KATP channel), where loss of heterozygosity leading to no functional allele and non-functional channels (Verkarre et al., 1998, Glaser et al., 1999, Fournet et al., 2001). This focal form is named for the fact it only affects a focal lesion within the pancreas which is almost exclusively β-cells. Diffuse CHI can be caued by a number of mutations (Glaser et al., 2000), includ- ing ABCC8 (Dunne et al., 1997), but is homozygous and so affects the whole pancreas. Atypical

CHI typically has a later onset than focal or diffuse CHI, is not caused by any known mutation

(screening of the genes associated with focal and diffuse CHI excludes these) and affects the whole pancreas (Arnoux et al., 2011). It has also been shown that atypical CHI is associated with altered hexokinase expression (Henquin et al., 2013).

In order to develop novel treatments for CHI, models are needed. Rodent models have been generated such as an ABCC8 mutant knock in mouse (Shimomura et al., 2013), but rodent models cannot fully capture all aspects of the disease. This is because the rodent pancreas is able to re- generate in ways that the human pancreas may not, and there are differences between human and rodent islet architecture (Cabrera et al., 2006) and function. When CHI patient pancreas tissue has to be surgically removed, it can be used for ex vivo studies. However, this is not the most practical research tool due to the infrequency of CHI surgeries (Arnoux et al., 2010, Meissner et al., 2003). The

final in vitro option is transfecting cells which do not normally express ABCC8, such as HEK293, with constitutively active ABCC8 containing a known CHI-causing mutation (Arya et al., 2014).

This method allows the study of the channel functions, but does not allow study of the effects on the insulin secretion pathways themselves. Stem cell sources of islets for cell therapies and modelling have been investigated, including derivation from adult somatic stem cells and pluripotent stem cells.

The potential of mesenchymal stem cells (MSCs) to differentiate to β-cells has been seen in MSCs derived from multiple sources in the body, including bone marrow, adipose, pancreas, periosteum and dental pulp (Bhonde et al., 2014). Efficiency and method of differentiation of MSCs towards

β-cells appears to be varied, with better insulin response seen in cells differentiated from pancreatic

MSCs (pMSCS) than bone marrow MSCs (bmMSCs) (Zanini et al., 2011).

136 Multiple putative stem cells have been isolated from the adult pancreas in both humans and ro- dents and differentiated to β-like or islet-like cells. This includes proliferative ductal structures (Cor- ritore et al., 2014), de-differentiated endocrine (Russ et al., 2009, Joglekar et al., 2009) and exocrine tissue (Lima et al., 2013) which resemble MSCs and de-differentiated endocrine tissue which remains epithelial (Lima et al., 2013). When placed into culture, all pancreatic cell types seemingly produce similar MSCs. These pMSCs have proven challenging as a model due to limited insulin release by re-differentiated cells (Bhonde et al., 2014).

Pancreatic development is a complex process, and it is unclear where pMSCs may fit into this pro- cess. A process similar to mesenchymal-epithelial transition (MET) does occur during de-lamination and formation of islets (Greiner et al., 2009, Gouzi et al., 2011) but the transcription factors ex- pressed show differences. The ultimate aim of differentiating MSCS in pancreatic research is to achieve functional insulin secreting cells, transfection of Pdx1 into rat MSCs is sufficient for Ins and

Glut2 (Yuan et al., 2010), and transfection of Pdx1, NeuroD1 and MafA into mouse MSCs was suf-

ficient for insulin expression, with Pdx1 inducing NeuroD1 (Qing-Song et al., 2012). This suggests that PDX1 expression is most essential for converting MSCs to insulin secreting cells, which matches to the fact PDX1 is essential for β-cell production during embryogenesis (Ahlgren et al., 1996).

Early experiments with pMSCs showed that they would spontaneously aggregate to form hor- mone (insulin and glucagon) positive cell spheroids with serum withdrawal, although the aggregates were unable to graft into animals (Dalvi et al., 2009, Davani et al., 2009, Carlotti et al., 2010). Upon addition of serum the cell spheroids return to resembling MSCs, positive for vimentin and nestin which, alongside the migration of MSCs within the body, accounts for the failure of engraftment of hormone positive spheroids (Dalvi et al., 2009). When the pMSCs differentiate or undergo this process resembling MET, β-catenin relocates to the cell surface from the nucleus. As passage num- ber increases nuclear β-catenin, which is not seen in islets increases (Ikonomou et al., 2008). The reformation of hormone positive spheroids was reported at the earliest passages of the pMSCs only, so high levels of nuclear β-catenin may be inhibiting this simple re-differentiation. As a result, a number of groups have used additional growth factors and small molecules to improve upon the differentiation process.

The factors commonly used to induce differentiation to the pancreatic lineages including GLP-1 or its analogue Exendin-4, betacellulin, activin A and nicotinamide (Seeberger et al., 2006, Bhandari et al., 2011, Zanini et al., 2011, Wang et al., 2012, Kim et al., 2012, Li et al., 2012, Gabr et al., 2014).

137 Histone de-acetylase (HDAC) inhibitors are also used (Jafarian et al., 2014, Gabr et al., 2014), as

MSCs have been shown to have repressive histone acetylation marks in the insulin gene compared to islets (Wilson et al., 2009). Bhandari et al. (2011) showed differentiation of a variety of MSCs to

C-peptide secreting cells with a combination between the HDAC inhibitor sodium butyrate, activin

A and a fibroblast growth factor, although the levels of C-peptide detected were less than 3% that of adult human islets. Similarly, Gabr et al. (2014) showed insulin secretion at around 3% that of human islets following differentiation with a HDAC inhibitor trichostatin A, and GLP-1, with a protocol including culture of the cells as free floating spheroids of cells similar to the spontaneous aggregates. A further improvement on this by Zanini et al. (2011) used no HDAC inhibitors but a combination of multiple factors including activin A and GLP-1, together with culture as free floating aggregates. In this protocol glucose-stimulated insulin secretion (GSIS) was also detected, and in- sulin granules were observed with electron microscopy. Whilst multiple protocols have been reported for differentiating MSCs to insulin secreting cells, they typically are based on similar factors, and involve culture of the cells in floating spheroids (see Chapter 1, Tables 1.1-2).

Aside from the use of MSCs, pluripotent stem cells can be used as a source to differentiate to

β-cells. Historically, obtaining functional β-cells from induced pluripotent stem cells (iPSCs) or hu- man embryonic stem cells (hESCs) has been challenging, as discussed by Hanley (2014), Mfopou and

Bouwens (2015). Issues have included lack of insulin secretion, lack of glucose responsiveness, poly- hormonal cells being produced and low efficiency of differentiation (D’Amour et al., 2006, Kunisada et al., 2012, Rezania et al., 2012). This is one reason why iPSCs have not previously been used in

CHI research.

Recently two groups have reported improvements in the process of differentiating iPSCs to β- cells (Rezania et al., 2014, Pagliuca et al., 2014). The resulting cells reported were monohormonal

(contained insulin and not glucagon as well) and secreted insulin in response to glucose multiple times. Importantly for CHI research, the iPSC-derived β-cells expressed ABCC8 (SUR1). Healthy donor (commercial) iPSCs and hESCs are not usable for CHI research, due to not possessing the disease causing mutations. This leaves the options of using gene editing such as CRISPR/Cas9 to insert the mutation, or to generate iPSCs from CHI patient cells. In the case of atypical CHI, where the disease causing mutation(s) are not usually known and so gene editing is not an option; deriving iPSCs from atypical CHI patients is therefore an ideal method to further investigate this disease form.

During reprogramming of somatic cells to iPSCs the epigenome is largely reset. There are how- ever, both tissue-specific (Kim et al., 2011) and donor-specific (Shao et al., 2013) CpG methylations

138 seen in iPSCs. By deriving iPSCs from CHI patient pancreas, the iPSCs would be most likely to have an epigenome associated with pancreatic gene expression (Bar-Nur et al., 2011), and have any disease-specific methylations (Robertson, 2005). Furthermore this would provide a proof-of-principle for personalised medicine for CHI, which may become an option as technology moves forward.

We had previously derived MSCs from the pancreas of patients with congenital hyperinsulinism

(CHI), see Chapter 2. These CHI pMSCs showed pancreas specific gene expression and promoter methylation (Chapter 2, Figures 2.8 and 2.9), as well as one of the hallmarks of CHI - increased proliferation (Chapter 3, Figure 3.2). Due to the inappropriate insulin secretion seen in CHI, we hypothesised that re-differentiated CHI pMSCs may show high levels of insulin secretion as the patients’ own β-cells do. The published protocols provided a point to start at to differentiate our

CHI pMSCs to insulin secreting cells. We aimed to repeat and improve upon these protocols in our cells, to assess the use of CHI pMSCs as an in vitro model of CHI. Further, in order to assess the potential for using patient-specific iPSCs for CHI research, we generated an iPSC line from ex vivo expanded pancreas tissue from a patient with focal CHI. We have further assessed the potential of these CHI iPSCs to differentiate to pancreatic β-cells using the recently published protocols.

139 4.3 Methods

All reagants were from Sigma-Aldrich or Fisher-Scientific.

4.3.1 MSC culture

Cell line Age at Disease form Mutation surgery F-CHI 4 months Focal CHI ABCC8 exon 6 (het- erozygous) A-CHI 17 months Atypical CHI Unknown D-CHI 11 weeks Diffuse CHI ABCC8 exon 37

Table 4.1: Details of cell lines and patients

Cell lines were derived as detailed in Chapter 2, information regarding the age of the patients and mutation is shown in Table 4.1. Cells were routinely cultured on tissue culture plastic (plates or vented cap flasks, Corning, Schiphol-Rijk, Netherlands) coated with human fibronectin (Chemicon,

Merck Millipore, Nottingham, UK) to 0.5 µg/cm2 and vitronectin (Life Technologies, Paisley, UK) to 0.1 µg/cm2, in RPMI-1640 5.5 mM glucose (mixed from 11 mM glucose RPMI, Sigma-Aldrich

(Irvine, UK) and 0 mM glucose RPMI, Gibco, Life Technologies to 5 mM) with 10% FBS. Cultured cells were kept in a humidified incubator at 37°C, 5% CO2. Subculturing/passage was performed when cells reached approximately 70% confluency - cells were washed with Dulbecco’s phosphate buffered saline (dPBS), and detached from tissue culture plastic with 0.05% trypsin-EDTA (Sigma-

Aldrich) for 3 minutes. Trypsinisation was halted using 1 mg/ml soybean trypsin inhibitor (Gibco).

Cells were pelleted at 50 x g and transferred to new vessels at a cell line and condition specific density, between 1:2 and 1:5. Medium was changed every 48 hours. Monolayer brightfield images were taken on an Olympus CKX41 microscope using 4x and 10x objectives and captured with QCapture Pro software, 3-dimensional spheroid images were taken on a M205FA (Leica, Wetzlar, Germany), with a 1.0X PlanApo objective and optical zoom, with images captured on LASAF software (Leica). All experiments were performed on cells between passages 3 and 16.

4.3.2 Feeder dependent iPSC culture iPSCs were cultured in 6-well plates (Corning) in iPSC medium comprising: DMEM/F12 (Gibco),

20% knock-out serum replacement (Gibco), 1% insulin-transferrin-selenium supplement (Gibco), 100

M 2-mercaptoethanol (Gibco), 1X non-essential amino acids (Gibco) and 4 ng/ml bFGF (Peprotech,

Rocky Hill, USA). Medium was changed daily. Approximately 24 hours prior to passage, irradiated mouse embryonic fibroblasts (iMEFs; Life Technologies) were seeded on 0.1% gelatin (Sigma-Aldrich) at a density of 2.2 x 105 per well of a 6-well plate. Colonies were passaged by microdissection - a

140 glass pipette was pulled over a flame and snapped to create a cutting edge, this was used to cut the colony with a grid. The small chunks of iPSCs were rinsed off and removed with a pipette and re-plated at a roughly 1:3 ratio. Medium was not changed until 48 hours after passage. Cultured cells were kept in a humidified incubator at 37°C, 5% CO2. Characterisation was between passages 1 and 10, differentiation experiments were carried out between passages 8 and 25.

4.3.3 Feeder free iPSC culture

For culture without feeder cells (iMEFs), iPSCs were cultured in 6-well plates (Corning) coated with vitronectin (Gibco) at 0.5 µg/cm2, in Essential 8 medium (Gibco), according to the method developed by Chen et al. (2011). To passage, cells were rinsed with dPBS and 0.48 mM EDTA was added until holes were visible in the colonies, at which point fresh medium was added and detached colonies were removed and replated at around a 1:5 ratio (with the addition of 10 µM Y-27632 hydrochloride (Sigma-Aldrich) for 24 hours of the first feeder-free passage). Medium was replaced every 24 hours. Cultured cells were kept in a humidified incubator at 37°C, 5% CO2.

4.3.4 Derivation of iPSCs

F-CHI pMSCs were cultured to passage 5, as described in Chapter 1, such that there were approx- imately 2 x 105 cells per well of a 6-well plate after 48 hours. The pMSCs were then transduced with Cytotune 2.0 Sendai reprogramming virus (SeV; Life Technologies) following the manufac- turer’s instructions, based on the method developed by Takahashi et al. (2007) at an MOI of 5:5:3;

Klf4/Oct4/Sox2:c-Myc:Klf4. Culture medium was replaced 24 hours after transduction, then every

48 hours until day 7. Transduced cells were replated iMEFs on day 7 at a density of 5 x 104 per 60 mm dish, still in pMSC medium. After 24 hours, culture medium was exchanged for iPSC medium which was then replaced daily. 21 days after replating, emerging colonies were identified by their characteristic morphology. To confirm reprogramming, colonies were stained by adding Tra-1-81 antibody (Pierce, Fisher-Scientific, Loughborough, UK) to culture medium for 1 hour, followed by

Cy3 conjugated goat anti-mouse antibody (Abcam, Cambridge, UK). Live colonies were imaged and those to which the antibodies had bound were noted. These colonies were cut out using a pulled glass pipette and the clumps replated onto fresh iMEFs. Each colony was transferred to a separate well such that the clones were cultured separately.

4.3.5 Three germ layer iPSC differentiation

Cells were dissociated with accutase to small clumps and transferred to ultra low adherence plates

(Corning), cells were cultured in DMEM/F12, 20% knock-out serum replacement, 1% insulin-transferrin- selenium supplement, 100 M 2-mercaptoethanol and 1X non-essential amino acids for 7 days to form

141 embryoid bodies. Following this the embryoid bodies were transferred to 0.1% gelatin coated cover- slips. After 48 hours the cells on the coverslips were fixed.

4.3.6 MSC differentiation

Differentiation protocols were commenced when cells reached around 70% confluency to ensure there were sufficient cells to analyse at the end of the protocol. The primary protocol was adapted from

Zanini et al. (2011) as follows. Cells were cultured on 0.5 µg/cm2 laminin (Sigma-Aldrich) as a monolayer on tissue culture plastic (Corning) for the first seven days in 5 mM glucose DMEM/F12

(comprising 0 mM glucose DMEM, Gibco and 10 mM glucose F12, Sigma-Aldrich) with 1% Insulin- transferrin-selenium supplement (Gibco) and 100 µg/ml ascorbic acid (Sigma-Aldrich), with the differentiation cocktail of 10 mM nicotinamide (Sigma-Aldrich), 10 nM exendin-4 (Sigma-Aldrich),

10 ng/ml betacellulin (Peprotech) and EGF 20 ng/ml (Calbiochem, Merck Millipore). For the

first 24 hours this was further supplemented with 10 µM retinoic acid (Sigma), 100 ng/ml DKK1

(Peprotech) and 100 ng/ml activin A (Peprotech). Activin A and DKK1 alone were supplemented for a further 48 hours without retinoic acid. After the seven days as a monolayer, cells were trypsinised and transferred to ultra-low adherence plates (Corning), 0.5 - 1 x 106 cells per well and cultured in 17.5 mM glucose DMEM/F12 (Gibco) with 1% Insulin-transferrin-selenium supplement (Gibco) and 100 µg/ml ascorbic acid (Sigma) with the differentiation cocktail for fourteen days. Controls were cultured in the basal media (DMEM/F12 with insulin-transferrin-selenium and ascorbic acid) without the differentiation cocktail, or with the time-specific factors (Figure 4.3) as appropriate.

4.3.7 Differentiation: Melton/Kieffer

Adapted from Pagliuca et al. (2014), hereon referred to as ‘Melton’ protocol, and Rezania et al.

(2014), hereon referred to as ‘Kieffer’ protocol. Details for the protocols are shown in Table (Figure)

4.1 for clarity and to allow comparison between the protocols. All stages for both protocols were in

MDCB 131 (Gibco) with 2% BSA (Sigma), 10 mM glucose (Fisher) and 1.5 g/L sodium bicarbonate

(Sigma). Controls were in the basal media alone, with DMSO added as appropriate for factors which had been dissolved in DMSO according to the manufacturer’s instructions.

Briefly the Melton protocol: cells were plated onto matrigel in E8, 48 hours later 100 ng/ml activin A (Peprotech) and 3 µm Chir99021 (Tocris) in basal medium were added. After 24 hours

Chir99021 was removed, 48 hours later media was replaced with 50 ng/ml keratinocyte growth factor

(KGF, R&D, Abingdon, UK), or cells were harvested (definitive endoderm). After 48 hours in KGF alone, 0.25 µm Sant1 (Sigma-Aldrich), 2 µm retinoic acid (Sigma-Aldrich), 200 nM LDN-193189

(Generon, Berkshire, UK) and 500 nM phorbol 12,13-dibutyrate (PdBU) (Sigma) were added, along

142 Figure 4.1: Melton/Kieffer differentiation protocols

143 with KGF. After 24 hours, LDN-193189 was removed, and a further 24 hours later media was re- placed with just 50 ng/ml KGF, 0.25 µm Sant1 and 100 nM retinoic acid and after 48 hours PDX1 samples were harvested.

Briefly the Kieffer protocol: cells were plated onto matrigel in E8, 48 hours later 100 ng/ml myo- statin (Peprotech) and 3 µm Chir99021 in basal medium were added. After 24 hours Chir99021 was exchanged to 0.3 µM and removed another 24 hours later; 24 hours later media was re- placed with 50 ng/ml KGF, or cells were harvested (definitive endoderm). After 24 hours in KGF alone, 0.25 µm Sant1, 1 µm retinoic acid, 100 nM LDN-193189 and 200 nM ((2S,5S)-(E,E)-8-(5-

(4-(trifluoromethyl)phenyl)-2,4-pentadienoylamino)benzolactam (TPB, Sigma-Aldrich) were added, along with KGF. After 48 hours media was replaced to contain 2 ng/ml KGF, 0.25 µM Sant1,

0.1 µM retinoic acid, 200 nM LDN-193189 and 100 nM TPB, and a further 48 hours later media was refreshed with the same factors. After 24 hours, samples were harvested for ‘PDX1’ cells or were removed from the plate by accutase and transferred to ultra low adherence plates (Corning) in DMEM, 10 µM ALK5iII (Cayman, Cambridge, UK), 100 nM LDN-193189, 1 µM T3 (Sigma-

Aldrich), 10 µg/ml heparin (Sigma-Aldrich), 1% B27 (Gibco). Media was exchanged every 48 hours for 7 days at which point cells were assayed.

Differentiation: D’amour

Adapted from D’Amour et al. (2006). Cells were cultured until 70-90% confluency and then medium was changed as follows. Day 1: RPMI 1640 (Sigma), 100 ng/ml activin A (Peprotech), 25 ng/ml

Wnt3a (Peprotech). Day 2: RPMI 1640, 0.2% knock-out serum replacement, 100 ng/ml activin A.

Cells were fixed or harvested for analysis at day 3.

4.3.8 RT-PCR and qRT-PCR

Total RNA was extracted using an RNeasy Mini Kit (Qiagen, Crawley, UK) with on column DNase

1 (Qiagen) genomic DNA digestion according to the manufacturer’s instructions and quantified using a NanoDrop 1000 spectrophotometer. cDNA was synthesised from 500 ng RNA using a nanoScript

2 reverse transcription kit (Primer Design, Chandler’s Ford, UK) with oligo-dT primers and random nonamer primers. RT-PCR was carried out using a Taq DNA Polymerase kit (Invitrogen, Life

Technologies), 1.5mM magnesium chloride and 2.5pmol of each primer with conditions as detailed in Table 4.2. PCR cycling conditions were 94°C for 5 minutes followed by 33 cycles (22 cycles for

18S rRNA, 38 cycles for INS) of 94°C for 30 seconds, annealing temperature as detailed in Table 4.2 for 30 seconds and 72°C for 90 seconds, with a final 72°C extension of 2 minutes. RT-PCR products were run on a 2% agarose gel stained with GelRed (Biotium, California, USA) and visualised on

144 a Gel Doc XR system (Bio-Rad, Hemel Hempstead, UK). PCR images had the colour inverted in

IrfanView, and where necessary brightness and contrast were adjusted, to ensure that fainter bands visible on the transilluminator would be visible when printed - this is particularly the case for Figure

4.4A PDX1 where the band was the same size as the loading dye and so difficult to see when printed. qRT-PCR was carried out using power SYBR green PCR master mix (Life Technologies) and 2.5 pmol of each primer as detailed in Table 4.2 and run on a StepOnePlus Real-Time PCR System

(Life Technologies). For qRT-PCR, the gene of interest CT values were normalised to the geometric mean of 2 reference genes, 18S rRNA and GAPDH (∆CT), following which the sample comparisons were done (∆∆CT). Statistical significance was calculated by one way ANOVA with Bonferroni post-hoc test if appropriate. Primers were designed using Primer-BLAST (NCBI) and synthesised by Eurofins (Wolverhampton, UK). Specificity of the primers was assessed by BLAST (NCBI) and fragment size, optimal annealing temperatures were determined by temperature gradient PCR to ensure only one fragment was present and the most intense band present. QRT-PCR primers had an efficiency between 85 and 115% confirmed by serial dilution of standards.

Gene Sequence (5’ - 3’) Annealing tem- Product

perature (°C) size (base

pairs)

18S rRNA F - GTAACCCGTTGAACCCCATT 60 150

R - CCATCCAATCGGTAGTAGCG

ABCC8 F - GCCTTCCTGACAGCGAGATA 66 85

R - CGGGGCCTCTCTTCCTGAT

FOXA2 F - ATGTCCTCGGAGCAGCAG 60 68

R - GCCTTGAGGTCCATTTTGTG

GAPDH F - ATTGCCCTCAACGACCACTT 60 95

R - GGTCCACCACCCTGTTGC

GCG F - TTCTGAGGCCACATTGCTTT 59 289

R - CTGTGGCTACCAGTTCTTC-

TATTCT

INS F - ACCAGCATCTGCTCCCTCTA 60 114

R - GGTTCAAGGGCTTTATTCCA

NANOG F - GATTTGTGGGCCTGAAGAAA 60 152

R - AAGTGGGTTTGCCTTTG

NEUROD1 F - ACCAGCCCTTCCTTTGATGG 60 131

R - AGTGTCGCTGCAGGATAGTG

145 Gene Sequence (5’ - 3’) Annealing tem- Product

perature (°C) size (base

pairs)

NGN3 F - CGAATGCACGACCTCAAC 59 125

R - AGTCAGCGCCCAGATGTAGT

PDX1 F - AGTGATACTGGATTGGCGTTG 59 138

R - TAGGGAGCCTTCCAATGTGT

POU5F1 F - CGAAAGAGAAAGCGAACCAG 60 157

R - GCCGGTTACAGAACCACACT

SeV genome F - GGATCACTAGGTGATATCGAGC 60 181

R - ACCAGACAAGAGTTTAAGA-

GATATGTATC

SOX17 F - CGCACGGAATTTGAACAGTAT 59 182

R - GGATCAGGGACCTGTCACAC

SOX9 F - AGCGAAATCAACGAGAAACT 60 222

R - ATCCCCTCAAAATGGTAATG

TUB3 F - ATCGGGGCCAAGTTC 60 110

R - TTGTAGTAGACGCTGATCC

Table 4.2: Primers used for RT-PCR

4.3.9 Bisulfite sequencing

Genomic DNA was extracted from cells using a PureLink genomic DNA kit (Invitrogen). 1 µg of genomic DNA was bisulfite converted using the EpiTect fast DNA bisulfite kit (Qiagen) according to the manufacturer’s instructions. PCR was carried out as Section 4.3.8 with primers and temper- atures detailed in Table 4.3. PCR primers were designed with MethPrimer (Li and Dahiya, 2002).

PCR products purified with a Qiaquick PCR purification kit (Qiagen) according to the manufac- turer’s instructions. Products were then sequenced, using 15 ng of purified PCR product, 4 pmoles of each primer used previously (forward and reverse sequencing carried out separately) with the

BigDye Terminator v3.1 cycle sequencing kit (Applied Biosystems) on a 3730 DNA analyser (Ap- plied Biosystems) using POP-7 polymer (Life Technologies). Percentage methylation was determined by measuring the peak heights of the cytosine or thymine for the forward trace, or adenine or gua- nine for the reverse trace on ChromasLite (Technelysium), with the proportion of cytosine or guanine

(methylated) to total cytosine/guanine plus thymine/adenine being calculated. Data was visualised with Methylation plotter (Mallona et al., 2014).

146 Gene Sequence (5’ - 3’) Annealing Genomic loca- Product tempera- tion size ture (°C) (base pairs) AMY2A F - GTTTTTGGAAA- 61 103616149 to 292 GAAGTTGTTTTTGA R - CC- 103616441 CTACCCTATTCTATTTCTCCCTA CK19 F - TTTAATATTTTTGTG- 61 41529473 to 442 GTTGAATGAA R - TCCCTAT- 41529031 TAAAAACCCCTTACTTAC INS F - TGTGAGTAGGGATAGGTTTG- 64 2161202 to 266 GTTAT R - AAAAACTAAAAAC- 2161468 TACTAAACCCCC

Table 4.3: Primers used for bisulfite sequencing

4.3.10 Immunocytochemistry (ICC)

For 2D culture, cells were cultured on glass coverslips, for 3D culture the individual spheroids were gathered into a tube and were fixed in 4% paraformaldehyde for 20 minutes. Cells were perme- abilised in 0.3% Triton x-100 (Sigma-Aldrich) and non-specific staining was prevented by incubation in 10% NGS in PBS. Primary antibodies were applied as Table 4.4 (dilutions are given where the manufacturer does not provide a concentration) for 1 hour for 2D culture, or overnight at 4°C, in 0.1% Triton x-100, 3% NGS in PBS and washed off in PBS/0.1% Tween-20 (Sigma-Aldrich).

Fluorophore-conjugated secondary antibody, specific to the species the primary antibody was raised in, was applied as Table 4.4 in 0.1% triton x-100, 3% NGS in PBS for 1 hour. Cells were washed in

PBS/0.1% Tween-20, then distilled water and allowed to dry. For 2D cells, coverslips were mounted onto microscope slides; for 3D cells the spheroids were suspended in minimal distilled water and pipetted onto a microscope slide and a coverslip mounted, in both cases using Pro Long Gold an- tifade reagent with DAPI (Life Technologies). Slides of 2D cells were viewed using an Olympus BX51 upright microscope using 20x 0.50 UPlanFLN, 40x 0.75 UPlanFLN or 60x 1.25 UPlanFLN (Ph 3) objectives and captured using a Coolsnap EZ camera (Photometrics) through MetaVue Software

(Molecular Devices). Filter sets for DAPI (31000v2), FITC (41001) and Cy3 (41007a) were used to prevent bleed through of colour channels whilst maximising signal. Slides of 3D spheroids were imaged on a Leica TCS SP5 AOBS inverted confocal using a 60x/ 0.50 Plan Fluotar objective. The confocal settings were as follows: pinhole 1 airy unit, scan speed 512Hz bidirectional, format 1024 x 1024. Images were collected using the following detection mirror settings: DAPI 400-500nm, Cy2

494-530nm, Cy5 640-690nm using the 405nm blue diode, 488nm (20%) argon and 633nm (100%) helium-neon laser lines respectively. Each colour channel was collected sequentially. When acquiring

3D optical stacks the confocal software was used to determine the optimal number of Z sections.

Only the maximum intensity projections of these 3D stacks are shown in the results. Images were

147 processed and analysed using ImageJ(http://rsb.info.nih.gov/ij).

4.3.11 Western blot

Cells were lysed in RIPA buffer (comprising 150 mM sodium chloride, 1% triton x-100, 0.5% sodium deoxycholate, 0.1% sodium dodecyl sulphate (SDS), 50 mM Tris and the pH was adjusted to 8.0) with protease inhibitors (Promega, Madison, USA) on ice with agitation, cells were centrifuged at 16000 x g to pellet cell debris leaving protein in the supernatant. Protein concentration was assessed using

Bradford reagant (Sigma-Aldrich) and 20 µg of total protein was added to laemmli buffer (comprising

4% SDS, 10% 2-mercaptoethanol, 20% glycerol, 0.004% bromophenol blue, 0.125 M Tris, with the pH adjusted to 6.8) and separated on a denuaturing poly-acrylamide stacking gel (BioRad) by SDS electrophoresis in running buffer (comprising 25 mM Tris, 190 mM glycine, 0.1% SDS). Protein from the gel was transferred to a methanol activated polyvinylidene fluoride (PVDF) membrane (BioRad) by electroblotting. The membrane was blocked in 5% (w/v) milk powder (Sigma-Aldrich), in Tris buffered saline with TBS/0.1% Tween-20. The primary antibody was added as Table 4.4 (dilutions are given where the manufacturer does not provide a concentration), diluted in blocking solution for an overnight incubation at 4°C. The membrane was washed 3 times in TBS/0.1% Tween-20, then incubated in the HRP conjugated secondary antibody (specific for the animal the primary antibody was raised in), diluted in blocking solution as Table 4.4 for one hour at room temperature. The membrane was washed three times in TBS/0.1% Tween-20 and bound antibodies visualised using the enhanced chemiluminescent (ECL) system (HRP Substrate, Millipore). The membrane was exposed to Biomax XAR film (Kodak) and developed in a Mini Med Film Processor (ImageWorks).

Densitometry analysis was performed on ImageJ, with normalisation to the housekeeping protein

GAPDH.

4.3.12 Glucose-stimulated insulin secretion (GSIS) assay

Around 24 hours prior to assaying, cell spheroids were transferred to culture media with 5 mM glucose and without insulin (all other components were the same as detailed above). On the day of the assay spheroids were rinsed in Krebs-Ringer-HEPES (KRH) buffer (comprising 129 mM sodium chloride,

5 mM sodium bicarbonate, 4.8 mM potassium chloride, 1.2 mM potassium orthophosphate, 1.2 mM magnesium sulphate, 10 mM HEPES, 2.5 mM calcium chloride, 5.6 mM D-glucose, 0.1% bovine serum albumin (BSA) and the pH was adjusted to 7.4) with 0 mM glucose three times. Roughly

20 spheroids were transferred to a 0.5 ml tube and 100 µl of KRH with 2 mM glucose was added.

The tube was incubated at 37°C for one hour, then the KRH was removed, spun at 2000 x g to remove any cells and stored at -80°C until needed. Following the removal of the first buffer, 100 µl of

KRH with 20 mM glucose was added and incubated at 37°C for one hour. KRH was then removed,

148 spun and stored as previous and KRH with 1% triton X-100 was added to lyse cells (on ice, with vortexing). To assay the supernatants and lysate, an enzyme-linked immunosorbent assay (ELISA) was performed, with an ultra-sensitive human insulin ELISA kit (Alpco, Salem, USA), according to the manufacturers instructions. Insulin content was calculated by comparison with the standard curve, on undiluted samples. DNA content of the lysed cells was assessed using a NanoDrop 1000 spectrophotometer, to provide normalisation for the number of cells used for each assay.

149 Antigen Species ICC Con- WB Con- Manufacturer centration centration (µg/ml) (µg/ml) α- Rabbit 0.04 Abcam smooth muscle actin Brachyury Goat 1:10 R&D NL557- conjugated C- Rabbit 1:100 Cell Signalling Technologies peptide (Leiden, The Netherlands) Foxa2 Goat 4 R&D GAPDH Rabbit 1:1000 Cell Signalling Technologies Gata-4 Goat 1:10 R&D NL493- conjugated Glucagon Mouse 1:200 Abcam Hand1 Goat 1:10 R&D NL637- conjugated Insulin Rabbit 2.5 Abcam Nanog Rabbit 4 Abcam Nestin Mouse 1 Millipore NeuroD1 Rabbit 5 Abcam Oct4 Rabbit 1 Abcam Otx-2 Goat 1:10 R&D NL557- conjugated Pdx1 Goat 4 Santa Cruz (CA, USA) Sox1 Goat 1:10 R&D NL493- conjugated Sox17 Rabbit 1:3000 Cell Signalling Technology Sox17 Goat 1:10 R&D NL637- conjugated Sox2 Rabbit 2 Abcam Sox9 Rabbit 0.5 Millipore Vimentin Mouse 0.2 Abcam Cy2 Goat 5 Jackson/Abcam anti- rabbit Cy2 Donkey 7.5 Jackson anti-goat Cy5 Goat 2.5 Eurogentec anti- mouse Cy5 Goat 2.5 Abcam anti- rabbit

Table 4.4: Antibodies used in the study

150 4.4 Results

4.4.1 Serum withdrawal was insufficient to induce differentiation of CHI MSCs

Figure 4.2: Serum withdrawal enhanced spheroid formation but was not sufficient alone Cells were cultured with or without serum, as monolayer or in non-adherent culture for fourteen days (A-B) Representative brightfield images from 1 cell line for all 3 CHI pMSC lines between passages 7 and 12. (A) Serum withdrawal did not strongly induce spheroid formation (images shown from fourteen days with or without serum after cells reached 70% confluency). (B) Spheroids readily formed in low adherence plates with or without serum, without serum spheroids were smaller and more spherical. (C) Densitometry analysis of western blots showed that 3D culture reduced expression of α-smooth muscle actin, expression of other mesenchymal markers were variable and the epithelial marker E-cadherin was not induced. Densitometry analysis was from the mean of 1 experiment on each of 3 CHI pMSC lines, normalised to GAPDH, error bars represent standard deviation.

We had previously derived MSCs from the pancreas of patients with CHI, which retained some pancreas specific genetic features but did not express insulin. To use the cells for modelling CHI, functional β-cells would need to be generated from the MSCs. It has previously been reported that

MSCs will undergo MET when transferred to low adherence conditions or serum-free media. Mature endocrine cells are epithelial and so we wanted to see if the CHI-MSCs would also undergo MET under these conditions. As can be seen in Figure 4.2 panel A, the cells continued to proliferate with a mesenchymal morphology when deprived of serum, with ever so slight clumping. When put into ultra-low adherence cell culture dishes, the cells spontaneously formed floating spheroids of cells which morphologically resembled islets. The spheroids formed whether grown with or without serum in the media, but as can be seen in panels B and C, the cells put into serum-containing media in low

151 adherence formed one very large spheroid of cells by seven days of culture whereas the cells in serum- free media formed many small spheroids. The large spheroid of cells seen in the serum-containing media is almost certainly apoptotic in the centre due to deprivation of oxygen and nutrients.

We could not tell by morphology alone whether the cells in the spheroids were epithelial or mesenchymal so further studies were required. Assessment of the epithelial and mesenchymal pro- teins showed a non-significant reduction of α-smooth muscle actin when the cells were grown in

3D spheroids, but VIM and NES seemed very variable. SOX9, an MSC marker also seemed to be reduced with serum withdrawal which could be related to previous observations regarding MET and induction of hormone expression with serum withdrawal (Dalvi et al., 2009, Davani et al., 2009).

E-cadherin, a key epithelial marker was not clearly induced in any condition, faint bands may have been visible but it was not clear with the low signal:background. The lack of E-cadherin confirmed to us that neither 3D culture nor serum withdrawal could cause a complete MET, but possibly just a down-regulation of some mesenchymal markers without the relevant increase in epithelial markers.

Protein yields from 3D samples were low compared to the monolayers, so despite an attempt at normalisation of the mass of protein used in the assays, some variability seen was probably due to variation in the concentration of protein loaded.

As well as not seeing MET in either 3D culture or with serum withdrawal, we also failed to see any induction of hormones or genes associated with β-cells (see controls for Figures 4.6 and 4.8); this is in contrast with what has previously been reported. Based on this we attempted to differentiate the cells towards the β-cell lineage using additional factors from published protocols, which also combined with serum withdrawal to reduced MSC/ductal SOX9 expression and 3D culture to reduce mesenchymal/activated stellate α-smooth muscle actin expression.

4.4.2 A step-wise differentiation protocol shows stage specific gene induc- tion, enhanced by the addition of DKK1

Several protocols failed to show any hormones or genes associated with β-cells 1. The protocol used by Zanini et al. (2011) showed insulin gene expression and slightly positive dithizone staining in pilot experiments, and so this protocol was further investigated, optimised with the CHI pMSCs and discussed from here on. The protocol was step-wise, with changes of the culture conditions after one,

1Protocols investigated which showed no positive results were Seeberger et al. (2006), Battula et al. (2007), Kim et al. (2012) and exendin-4/betacellulin/nicotinamide alone or in combination, 2D or 3D for 1 week based on previous internal work. Results for these are not shown as all that was seen were fully blank gel images based on PCR for INS, PDX1 and NEUROD1.

152 Figure 4.3: Step-wise differentiation induces and reduces gene expression All three cell lines were differentiated between passages 7 and 12 by a stepwise protocol (panel E), with the effect of each factor assessed by its use compared to relevant control (water or DMSO) at each stage seen by RT-PCR as (A) A possible reduction in HES1 expression at day 1, to be confirmed by qRT-PCR, (B) Induction of NEUROD1, with retinoic acid, activin A and DKK1 only and TUB3 expression at day 3 and (C) Induction of PDX1 and loss of TUB3 by day 7. (D) Addition of DKK1 to the protocol resulted in an increase in PDX1 (qRT-PCR) and an apparent INS (estimated from RT-PCR densitometry and qRT-PCR of housekeepers) expression after 21 days of differentiation and a more intense dithizone stain. Images shown are from one experiment in one cell line and representative for one experiment, each in two CHI pMSC lines. (E) shows a schematic of the differentiation protocol and the timing of the factors used, for each experiment within this figure only one stage was assessed at a time to add or remove the factors, previous stages were constant (with all previous factors).

153 three and seven days, with the end of the protocol after 21 days. The gene expression was assessed at each of these time points in an attempt to understand and optimise the factors being used, this is shown in Figure 4.3. After the first day, there was an apparent reduction in expression of the gene HES1 in cells treated with retinoic acid versus controls treated with the vehicle (DMSO) alone, this is shown in panel A but was not confirmed by quantification. HES1 is a downstream target of

NOTCH signalling which is highly expressed in CHI pMSCs, which when inhibited allows NGN3 and

NEUROD1 to be induced during development (Jensen et al., 2000, Murtaugh et al., 2003). Despite the potential change in HES1 expression, we did not see a concordant induction of either NGN3 or

NEUROD1 so it may be an artifact of the non-quantitative RT-PCR. SOX9 which is also highly expressed in the CHI pMSCs was unaffected by this treatment. Therefore no clear changes in gene expression were seen by the use of retinoic acid.

Initially, following the Zanini et al. (2011) protocol of using retinoic acid and activin A alone, no changes in gene expression were seen after three days. When the Wnt inhibitor, DKK1, was added to the protocol for these first three days, we saw induction of NEUROD1 which is associated with the β-cell lineage as shown in Figure 4.2 panel B. This gene induction was only seen with the com- bination of all three factors. NEUROD1 is also expressed in neuronal lineages, and so we assessed expression of a specific neuronal marker - β-tubulin (TUB3 ). Surprisingly TUB3 appeared to be induced by treatment with any of the three factors (retinoic acid, activin A, DKK1), separately or in combination. Assessment of the cells at the end of the differentiation protocol, after 21 days, showed that the addition of DKK1 to the published protocol enhanced INS and PDX1 gene expression and the intensity of dithizone staining, shown in Figure 4.3 panel D. Dithizone stains zinc which is present in insulin granules and so can be used for rapid, but potentially inaccurate, screening for insulin containing cells.

The remaining treatment of the cells as a monolayer was with a cocktail of growth factors com- monly used in the β-cell field. The use of this cocktail induced expression of the PDX1 and turned off expression of TUB3, by day seven of treatment, shown in Figure 4.3 panel D. Where the cocktail was not used TUB3 expression remained following its induction by retinoic acid/activin A/DKK1, and PDX1 was not induced. This suggests the cocktail of growth factors is essential for switching the cells from differentiating towards a neuronal fate, to a pancreatic fate, as a result of the overlapping transcription factors.

154 Figure 4.4: Spheroids treated with differentiation inducers were of a similar size to human pancreatic islets After 21 days of either differentiation according to the protocol shown in Figure 4.3E (treated) or growth in serum free media (control) as detailed in Section 4.3.6 (A) Treated spheroids were smaller and more rounded than control spheroids and did not merge into large atypical spheroids. (B) Histograms of the diameter of spheroids showed a size distribution for all three cell lines resembling that of human islets, with an average diameter around 100 µm. At least 100 spheroids were measured for two experiments for each cell line between passages 7 and 12. Each bin is 25 µm.

155 4.4.3 Phenotypic analysis of differentiated CHI pMSCs

Following the seven days of treatment as a monolayer, the cells were transferred to low adherence plates, which as above allowed formation of cell spheroids. As with the experiments shown in Figure

4.4, the spheroids were generally small and rounded as serum was not used in the culture media.

The spheroids treated according to the differentiation protocol were even more rounded than the serum-free only controls, further details of this are shown in - Figure 4.5. The distribution of sizes of the treated spheroids was similar to isolated human islets (Lehmann et al., 2007) as shown in Figure

4.4, with an average diameter of around 100 µm which also matches islets. There was a greater spread in spheroid size for D-CHI compared to A-CHI and F-CHI, with spheroids over 400 µm being present only in D-CHI cells. It is unclear what the reason for this would be, as it is unlikely to be related cell number or growth rate as A-CHI proliferates faster than D-CHI (see Chapter 3, Figure

3.2). The treated spheroids therefore phenotypically resemble human pancreatic islets at the gross level.

Figure 4.5: Some untreated spheroids had a capsule which was (A) visible on brightfield images, indicated by arrows, (B) stained positive for insulin, indicated by arrows and (C) stained with alcian blue, indicative of acidic polysaccharides. Images are from after 21 days as the control for differentiation, i.e. in basal media (DMEM/F12) with insulin/transferrin/selenium only at passage 10 in A-CHI.

We observed that the control spheroids had a more defined border than the spheroids treated with growth factors, with a capsule structure seen on brightfield images, this is shown in Figure 4.5.

156 This capsule stained positive with the insulin antibody, but this appeared to be non-specific as it was in a thin band rather than being associated with whole cells. This could possibly be due to whatever the capsule was formed from absorbing and binding insulin from the media, as the control cells were kept in serum free media with insulin/transferrin/selenium supplement to maintain viability, or due to non-specific binding of the antibody. Given the lack of insulin in control spheroid lysate by ELISA, non-specific binding may be more likely, although it could also be the case that the capsule was not released by the lysis buffer used.

We hypothesised that this capsule was most likely comprised of extracellular matrix proteins secreted by the cells. Further investigation showed that the spheroids stained positive for alcian blue which recognises acidic polysaccharides including glycosaminoglycans and sulfated proteoglycans.

This includes versican and aggrecan which are secreted by undifferentiated MSCs and at increased levels in chondrocytes. More specific studies would be required to find out if this capsule is as a result of a build up from undifferentiated MSCs across the fourteen days, or due to chondrogenic differentiation because of the dense culture without TGF-β inhibition (Bosnakovski et al., 2004).

Either condition is undesirable though so the absence of this in our treated spheroids was suggestive of a more targeted differentiation. Supplementing proteoglycans, such as in a hydrogel, may help the formation of the spheroids into stronger structures when they are not secreting their own matrix.

The fact that the capsule was only seen in untreated spheroids may give an indication of differenti- ation efficiency for the treated spheroids.

To assess how much the spheroids resembled pancreatic islets, various markers were assessed.

PDX1 gene expression was maintained, from its induction at day seven (Figure 4.3C). Expression of the hormone encoding genes, INS and GCG was induced after the 14 days of culture as spheroids

(21 days of treatment), but ABCC8 encoding the SUR1 subunit of the KATP channels, which are essential for GSIS was not induced, nor was NEUROD1 which would normally be expressed in β- cells. Regardless of treatment, SOX9 gene expression was lost after 14 days of low adherence culture in serum-free conditions, by day 21 of the experiment. Based on the results shown in Figure 4.2 this could be expected, and suggests that there are little or no basal MSCs remaining. This is supported by the fact that if spheroids were re-plated at the end of the experiment, MSCs took several days to reappear as with islets (observation).

PDX1 and insulin proteins were also seen in treated spheroids, more clearly than in untreated spheroids. C-peptide protein was seen at a very low level in treated spheroids only, the stain was not as intense as the insulin alone suggesting some of the insulin may have been absorbed from the

157 Figure 4.6: Differentiated CHI pMSCs express INS and PDX1 After 21 days of differenti- ation according to the protocol shown in Figure 4.3E, the CHI pMSCs showed (A) Gene expression of INS and PDX1 by RT-PCR, where ‘Treatment’ includes all or no factors detailed in Section 4.3.6, (B) Insulin and PDX1 protein, with faint staining for C-peptide by immunofluorescence. Rabbit and mouse panel show secondary antibody only dual stain.(A-B) Images shown are from one experiment in one cell line and representative for at least one experiment in three CHI pMSC lines between passages 7 and 12.

158 culture media. To understand this further, an insulin ELISA was performed to assess both content and secretion of insulin by the cells, this is shown in Figure 4.7. Glucagon was not seen to be ex- pressed at all in either condition so the spheroids did not resemble pancreatic islets but seemed to be made predominantly of β-like cells. All stains were confined to the cytoplasm of the cells, this is to be expected for insulin and c-peptide. PDX1 is known to shuttle between the cytoplasm and nucleus, but is not active in the cytoplasm, suggesting it may not actually be active in these spheroids.

Figure 4.7: Differentiated spheroids contained and secreted more insulin than control spheroids After 21 days of differentiation according to the protocol shown in Figure 4.3E, more insulin protein was seen in the lysate and supernatant of spheroids which had been treated according to the differentiation protocol, but this was not glucose responsive, (A) shows the lysates and (B) shows the supernatants in response to 2 mM and 20 mM glucose, this is the results of one experiment in D-CHI only, at passage 11.

We wanted to see if the spheroids were functional at all, and so assessed if the insulin seen by immunofluorescence could be secreted. We therefore performed a GSIS assay on both treated and untreated spheroids. No secretion or insulin content was seen for untreated spheroids, shown in

Figure 4.7. Both insulin content and secretion were seen in treated spheroids although the secretion was not glucose-dependent. This was to be expected based on the absence of ABCC8 and suggests secretion may be via a non-standard pathway. This was only documented from one experiment using the D-CHI cell line which would not be expected to be glucose responsive anyway due to non-functional KATP channels. Further experiments with additional cell lines would be needed to

159 confirm the functionality of the differentiated cells, with a C-peptide ELISA to confirm whether the insulin was produced by the cells rather than being absorbed from the media and leached back out.

4.4.4 Spheroid formation was essential for differentiation

Figure 4.8: Cells differentiated as monolayer do not have the same expression profile as spheroids After 21 days of differentiation without low adherence culture, the CHI pMSCs showed (A) gene expression of INS alone by RT-PCR, where ‘Treatment’ includes all or no factors detailed in Section 4.3.2, (B) no expression of insulin or C-peptide protein, with apparent PDX1 protein expression. Images shown are from one experiment in one cell line and representative for at least one experiment in three CHI pMSC lines, between passages 7 and 12. Secondary antibody only stain is not shown here as staining was carried out alongside the samples shown in Figure 4.6.

Analysis of the cell spheroids was challenging, and minimal differences were seen between mes- enchymal proteins for 2D or 3D culture, so we wondered whether differentiation as spheroids was essential. Therefore the differentiation protocol was carried out in an identical fashion but without trypsinisation and transfer to 3D culture at day seven. As with the spheroids, SOX9 gene expression was completely lost in both treated and untreated cells, showing that this is as a result of the pro- longed serum withdrawal and unrelated to 3D culture. INS gene expression was induced, as with the

3D culture, but PDX1 did not appear to be induced in any of the three cell lines, shown in Figure

4.8. At the protein level, there was no staining for insulin or C-peptide seen although cytoplasmic staining for PDX1 was observed in the treated cells only. This was unexpected given that PDX1 gene expression was not observed - it may have been that gene expression was very low and that additional PCR cycles would be required but there was insufficient mRNA (cDNA) to check this, or

160 that the PDX1 transcript was unstable (Abreu et al., 2009). Given that there was no insulin protein seen in these experiments it suggests that 3D culture was essential for differentiation towards the

β-cell phenotype but for reasons unrelated to MET.

4.4.5 Derivation of iPSCs

Differentiation of CHI MSCs proved extremely challenging, despite numerous published reports of producing functional β-cells from both pancreatic and non-pancreatic MSCs. Due to the low density

(high surface area of cells) at which MSCs grow, culturing sufficient cells for various analyses was time consuming and expensive, limiting optimisation. We were able to induce expression of insulin, but preliminary experiments suggested that any insulin secretion was not in response to glucose stimulation. During the course of this work, novel protocols with a high reported efficiency were published for iPSCs (Pagliuca et al., 2014, Rezania et al., 2014). Compared to MSCs, iPSCs prolif- erate quickly and at a higher density allowing more experiments to be carried out and so we derived an iPSC line from the F-CHI MSC line.

Successful reprogramming of the cells by the SeV was confirmed. MSCs were transduced at passage 5 with SeV containing Oct4, Sox2, Klf4 and c-Myc and colonies were clonally selected for further expansion. Initial selection merely confirmed uptake of the SeV by the MSCs, for long term use the cells have to be expressing the factors themselves. Three clones were selected from the ini- tial transduction plate by morphology and Tra-1-81 staining. However, two of these did not show the characteristic pluripotent morphology after four to five passages, with the colonies appearing to differentiate. This is indicative that they were not fully reprogrammed, so when the SeV was cleared from the cells they no longer contained all of the necessary factors. The third colony was confirmed to have a stable morphology across multiple passages past P5, as shown in Figure 4.9A this is the clone which was used for all experiments detailed in the main body of the text. The cells grew as dense colonies between the feeder layers, with a high nuclear:cytoplasmic ratio and a defined clear edge. The MSCs that the cells were transduced from, in comparison, had an elongated and spread morphology, with the cells growing individually.

Clearance of SeV was confirmed by qRT-PCR - SeV is a single-stranded RNA virus so its pres- ence or absence can be confirmed by RT-PCR similar to gene expression. SeV was consistently not seen to be present, with a CT value of 40. In comparison to the positive control this worked out to a fold change of nearing 1.5 x 106 confirming that the SeV had been successfully cleared from the cells.

In the non-transduced MSCs, POU5F1 (encoding OCT4) and NANOG were expressed at a very

161 Figure 4.9: MSCs were transduced and cleared the SeV The clone used was assessed by (A) morphology on brightfield after five passages, where the white arrow indicates the colony with a defined border and the black arrow indicates the iMEFs, (B) qRT-PCR to confirm loss of SeV, in passage 5, 8 and 14 cells (the mean is shown), compared to positive control from d7 of transduction and (C) qRT-PCR to confirm expression of POU5F1 (Oct4) and NANOG, at passages 5, 8 and 14 compared to the pre-transduction cells at passages 3, 7 and 9; bars indicate the mean of the three passages, with standard deviation.

162 low level and not expressed, respectively. POU5F1 is normally considered a marker of pluripotent cells only so it was somewhat surprising to see it expressed in the non-transduced cells, but it may be involved in the multipotency of MSCs too. In comparison, after transduction and confirmation of clearance of SeV (by using the same mRNA) high levels of both POU5F1 and NANOG were seen, shown in Figure 4.9.

Following confirmation of successful transduction and clearance of SeV (see Figure 4.9), cells were switched to feeder free conditions (see Figure 4.11) and pluripotency was assessed. It was confirmed that the essential pluripotency factors, OCT4, SOX2 and NANOG, were expressed as proteins and correctly located to the nucleus, as shown in Figure 4.10. These three proteins are the core tran- scription factors responsible for pluripotency (Takahashi et al., 2007).

Secondly three cell surface markers were studied by flow cytometry on un-permeabilised cells and expression of all three of these was seen in 85% of cells. Expression of SSEA4 and Tra-1-81 was seen in over 95% of cells. Lower expression of Tra-1-60 was seen, however this may have been due to the antibody, as normal distribution is still seen but with low fluorescence levels which overlap with isotype control.

Alongside these markers it was important to confirm pluripotency by a functional assay. To confirm that the cells are indeed pluripotent they must be able to differentiate into cells of all three germ layers - endoderm, ectoderm and mesoderm. This is historically done by teratoma formation in mice (Thomson et al., 1998) but can also be done by in vitro differentiation which is more in keeping with the ethics of the sponsor of this project (NC3Rs). Random differentiation via embryoid bodies yielded endoderm, ectoderm and mesoderm with staining for two nuclear transcription factors confirmed for each, the markers used were from a commercial kit designed for this purpose based on published lineage markers. EBs were cultured for one week, and allowed to adhere to coverslips for 48 hours, one week culture should allow EBs to develop all three germ layers. Efficiency seemed low for mesoderm with few cells seen to be positive for HAND1, large regions of both endoderm (the germ layer from which the β-cells important for this work are derived) and ectoderm were seen however.

Together these data confirm that the transduction was successful and the MSCs were reprogrammed to induced pluripotent stem cells (iPSCs).

The cells were switched from growth on MEFs to feeder-free conditions. Growth in defined condi- tions without a feeder layer is important to confirm that effects of growth factors and small molecules is specific to the iPSCs and not the feeder cells. Feeder-free conditions are also essential for flow

163 Figure 4.10: Derived iPSCs were phenotypically and functionally pluripotent Pluripo- tency was confirmed by (A) nuclear immunostaining of the transcription factors OCT4, NANOG and SOX2, images shown are from one experiment and are representative of passages 7, 9 and 10, rabbit panel shows secondary antibody only stain (B) flow cytometry for the cell surface markers SSEA4, Tra-1-60 and Tra-1-81, histograms are from one experiment and representative of passages 7, 9 and 10 and (C) differentiation to the three germ layers via embryoid body formation at passage 16, embryoid bodies were grown for 7 days, then allowed to adhere to coverslips for 48 hours, images shown are from one experiment only. All scale bars (panels A and C) represent 50 µm

164 Figure 4.11: Transition of iPSCs to feeder-free culture (A) IPSCs maintained colony mor- phology (i) rounded after the first manual passage from feeders or (ii) as linked networks following EDTA passage. (B) Vitronectin and Matrigel were compared to see which supported growth better but showed similar colonies after two days.

165 cytometry studies and techniques such as RT-PCR and western blot where whole cell populations are measured to ensure only the stem cells are being assessed.

Growth was in the commercial essential 8 media (Chen et al., 2011), which has been optimised for various pluripotent stem cells. This media requires the use of an extracellular matrix for attachment of the cells, either vitronectin or matrigel. For the first few passages vitronectin was used as this is what is recommended by the group who developed the media. In the first passage cells were cut manually from the feeders into sections and transferred, this caused them to grow in large round colonies as they do on the MEF layers, this can be seen in Figure 4.15 panel Ai. The only adaptation required was 50:50 of the media used with feeders and essential 8 media for the first two days, and the use of a ROCK inhibitor for the 24 hours post passage . Following this, cells were not passaged manually but by using EDTA. EDTA passage led to colonies not being rounded, but being linked across the dish, still with compact cells and defined edges as can be seen in Figure 4.15 panel Aii, which is normal for this method of culture.

After 3 stable passages in feeder-free conditions a comparison was carried out, on whether vit- ronectin or matrigel supported growth better for the cells. Cells were seeded at the same density and a rock inhibitor used in both cases. After 24 hours of culture the cells on matrigel covered more of the plate, but were more spread out, whereas the cells on vitronectin had the dense clusters of cells normally associated with pluripotent colonies. However, after 48 hours of culture cells in both conditions resembled normal dense colonies with little difference between the two. Given the lack of any clear advantage of either extracellular matrix, we continued to use vitronectin as this is more defined than matrigel, which is a mixture of various proteins secreted by mouse tumour cells.

The iPSCs were derived from pancreas-derived MSCs, which maintained an epigenetic profile of the pancreas, with significant differences seen in the 5’ regions of AMY2A, CK19, PDX1, although not for INS, see Chapter 2. Some of these differences were maintained after transduction to iPSC, as shown in Figure 4.7. For AMY2A, an exocrine marker, at the locus which was significantly differ- ent (p=0.012) between the pMSCs (24% methylation) and bmMSCs (49% methylation), the iPSCs showed even further decreased methylation (22%). However for CK19, a ductal marker, at the two loci which were different between pMSCs and bmMSCs (locus 2: 69% and 87%, locus 6: 61% and

73% respectively), there was increased methylation in the iPSCs compared to the untransduced cells, similar to the levels seen in non-pancreatic cells (locus 2: 87%, locus 6: 81%). It should be noted though, that CK19 is expressed in the MSCs, with an unknown function, but is not expressed at a detectable level in the iPSCs (data not shown). There was also increased methylation of the INS

166 promoter, encoding the β-cell specific hormone, despite high levels of methylation in pMSCs and the non-pancreatic bmMSCs. This may suggest a bias against differentiation to ductal cells or β-cells, and towards exocrine cells instead. Due to experimental issues PDX1 was not able to be assessed.

4.4.6 Differentiation of CHI-iPSCs to definitive endoderm

When differentiating CHI MSCs towards the β-cell lineage, the protocol was step-wise, but the steps do not clearly map to embryonic development. On the other hand, when differentiating iPSCs, the stages are based on development. The first step in differentiation of pluripotent cells towards β- cells is therefore definitive endoderm. The higher the efficiency at early stages of differentiation the greater numbers of the desired cell type will be present at the end of the process. Whilst there are a number of markers of definitive endoderm including CXCR4 and GATA4/6, we chose to compare the efficiency of differentiation to definitive endoderm using the two most commonly used markers,

SOX17 and FOXA2 (GATA4 was used previously in Figure 4.6 as this was more suitable for simul- taneous dual staining but as discussed in Section 1.2.4, GATA4 can be selective for the exocrine pancreas (Ketola et al., 2004)).

Efficient differentiation to definitive endoderm was achieved with the CHI iPSCs using two proto- cols. Using both the ‘Melton’ and ‘Kieffer’ protocols, expression of the gene SOX17 was upregulated by 200-300-fold, and expression of FOXA2 by 40-fold compared to the controls, shown in Figure

4.13. In both cases the gene expression was translated to protein, with strong nuclear staining of both transcription factors observed in the differentiated cells but not controls (except in rare cells which had undergone spontaneous differentiation).

We quantified the percentage of cells which were positive for SOX17 and FOXA2. By both pro- tocols, over 85% of cells were positive for SOX17 or FOXA2. Dual staining was not performed but clearly a large proportion of cells were also positive for both markers. The difference in percentage positive cells for each protocol was small and not significant. This means that both protocols were equally effective to differentiate CHI iPSCs to definitive endoderm, with a high efficiency and so the only advantage of either protocol at this stage is that the ‘Melton’ protocol to this stage is cheaper.

The first major report of producing β-cells from pluripotent stem cells in the literature was by

D’Amour et al. (2006) and this protocol was also assessed. Strikingly, when our iPSCs were treated with this protocol it was found that almost all cells died by the third day of treatment. Indeed, there were insufficient cells at the end of the 3 day protocol to analyse.

167 We hypothesised that this may be due to the fact that our cells were cultured in feeder-free conditions, whereas the original report used stem cells grown on MEFs. We therefore switched the cells back to growing on MEFs and repeated the protocol. In this case, the cells survived and as shown in Figure 4.14 stained positive for SOX17. It is unclear why MEFs are required to prevent apoptosis when using this protocol, but as we wanted to be able to use the feeder free conditions, the later stages of this protocol were not investigated.

4.4.7 Differentiation of CHI-iPSCs to pancreatic endoderm

The next major stage after definitive endoderm, is pancreatic endoderm. This is the point at which the pancreas is specified, as opposed to other endodermal lineages such as the liver. Pancreatic endoderm is primarily characterised by the presence of PDX1, with NEUROD1 and NGN3 following shortly after. As the ‘Kieffer’ protocol called for cells to be non-adherent at the stage where NEU-

ROD1 is to be assessed, and due to the technical challenges associated with this, PDX1 alone was measured.

PDX1 expression was significantly and highly induced in response to both differentiation proto- cols, compared to the controls. With the ‘Kieffer’ protocol there was around a 1000-fold increase in

PDX1, with the ‘Melton’ protocol a 10,000-fold increase, shown in Figure 4.15. There was therefore also a 10-fold difference in PDX1 expression between the two protocols, with significantly higher expression using the ‘Melton’ protocol.

Using both protocols, PDX1 protein was also detected in the differentiated cells and not the controls. PDX1 was not confined solely to the nucleus, with diffuse staining observed throughout the cells. This meant that quantification of numbers of PDX1 positive cells was not readily achiev- able, this could possibly be done using flow cytometry instead. From observation of the slides, the generation of PDX1 positive cells also appeared to be highly efficient with both protocols. We could not assess by numbers of cells if the difference in PDX1 gene expression between the two protocols is translated. We could also not assess levels of PDX1 protein by staining intensity, due to variation between experiments. The total amount of PDX1 protein could therefore be assessed by western blot to confirm or refute whether the ‘Melton’ protocol is more efficient at generating PDX1 positive cells.

168 4.4.8 Differentiation of CHI-iPSCs to hormone positive islet cells

The final stage of the differentiation process, is to hormone positive (insulin and/or glucagon) islet cells. At this stage only the ‘Kieffer’ protocol was assessed as the ‘Melton’ protocol calls for spinner

flasks which were not available within our lab, the results of which are shown in Figure 4.16.

We observed the formation of islet-like clusters of cells following the further differentiation, similar to those seen when differentiating the CHI MSCs (Figures 4.4-6). These clusters of cells expressed

NEUROD1 and ABCC8, genes associated with β-cells and GCG, the gene encoding the α-cell specific hormone. PDX1 was also detected, although this was not clear. The β-cell specific hormone INS did not appear to be expressed at the gene level. We also saw an apparent reduction in POU5F1 and

NANOG, the pluripotency genes, this would need to be confirmed and the magnitude investigated by qRT-PCR. Assessment of the pluripotency markers demonstrates how efficient the protocol is, as in order to use iPSC-derived cells for transplantations they need to no longer have any pluripotent cells present.

The cell clusters were stained only for insulin, glucagon and C-peptide; NEUROD1 and ABCC8

(SUR1) proteins were not assessed. They appeared to be positive for glucagon, and potentially slightly positive for C-peptide, although the C-peptide stain was not convincing as some signal was seen not associated with nuclei and by inference, cells. Very strong staining was seen for insulin.

Given the negative insulin expression and negligible C-peptide levels, this stain is almost certainly artefactual (Hansson et al., 2004), due to the extremely high levels of insulin the cells were cultured in

(4 mg/L). Insulin secretion was assessed by ELISA but no insulin was detected at all in supernatant, even in response to 20 mM glucose (data not shown). This data therefore suggests that the iPSCs have preferentially differentiated towards an α-cell fate rather than β-cell. However, compared to the

MSCs, expression was seen of more β-cell specific genes and so further optimisation of the protocol may yield insulin secreting cells.

169 Figure 4.12: CHI-iPSCs did not maintain hypomethylation seen in CHI-pMSCs Methy- lation analysis of the AMY2A, CK19 and INS 5 regions revealed differences between the transduced (iPS) at passage 20 and untransduced MSCs (F-CHI) at passage 4. The loci which were significantly different between pMSCs and bone marrow MSCs (bmMSCs) are boxed.

170 Figure 4.13: CHI iPSCs efficiently differentiate to definitive endoderm: ‘Melton’ and ‘Kieffer’ protocols for obtaining definitive endoderm were compared (A) qRT-PCR showed SOX17 and FOXA2 gene expression were both significantly increased after treatment with both protocols, and were not significantly different between the two protocols. (B) Immunocytochemistry showed nuclear staining of SOX17 and FOXA2 (C) The number of SOX17 and FOXA2 positive nuclei compared to total nuclei (stained with DAPI) showed no significant differences between the two protocols. (A) and (C) show the mean of three experiments for each protocol at passages 14, 18 and 24, with standard deviation, (B) shows an image from one experiment, representative for the three. Secondary antibody only images were not available at the time of writing; these would be needed to confirm that the stains were specific. 171 Figure 4.14: CHI iPSCs differentiate to definitive endoderm by D’amour protocol only when cultured on feeders Cells differentiated according to the D’amour protocol stained positive for SOX17, controls did not.

Figure 4.15: CHI iPSCs efficiently differentiate to pancreatic endoderm: Protocols from ‘Melton And ‘Kieffer for obtaining pancreatic endoderm were compared (A) qRT-PCR showed PDX1 gene expression was significantly increased after treatment with both protocols, and was significantly higher following treatment with the ‘Melton protocol. (B) Immunocytochemistry showed nuclear and cytoplasmic staining of PDX1, images and staining were not taken at the same time for each protocol so staining intensity cannot be compared. (A) shows the mean of three experiments for each protocol at passages 18, 21 and 24, with standard deviation, (B) shows an image from one experiment, representative for the three. Secondary antibody only images were not available at the time of writing; these would be needed to confirm that the stains were specific.

172 Figure 4.16: CHI iPSCs differentiated to cells with characteristics of immature α and β- cells Following 17 days of differentiation with the ‘Kieffer protocol cell clusters showed (A) decreased NANOG and POU5F1 gene expression with induction of NEUROD1, PDX1, GCG and ABCC8 by RT-PCR and (B) positive insulin and glucagon staining by immunofluorescence. Rabbit and mouse panel show secondary antibody only dual stain.(A)-(B) show results of one experiment only at passage 22.

173 4.5 Discussion

In this study we report the first derivation of an iPSC line from a CHI patient, and we assessed the capacity of both CHI pMSCs and iPSCs to differentiate to insulin secreting cells. Neither the use of pancreatic MSCs nor CHI tissue as a source for iPSCs has previously been reported. IPSCs have been derived from bone marrow and adipose MSCs (Sugii et al., 2010, Lister et al., 2011, Ohnishi et al.,

2012, Shao et al., 2013) and human β-cells (Bar-Nur et al., 2011). In a disease such as focal CHI where very limited pancreatic tissue is available, initially generating MSCs allows an easy method for expanding the pancreatic tissue, whilst potentially maintaining pancreas-specific epigenetic marks

(see Chapter 1). We have found some promise for using the MSCs themselves for CHI research (see

Chapter 3), but iPSCs may allow further opportunities which we aimed to explore, in comparison to the MSCs. Differentiating iPSCs will allow the study of developmental differences seen in CHI

(Salisbury et al., 2015), but both MSCs and iPSCs could be used to produce β-cells for drug testing for CHI.

Studies with pMSCs by other groups documented that upon withdrawal of serum, the MSCs would spontaneously aggregate to form hormone positive cell spheroids with a down-regulation of the defining mesenchymal proteins. We found that on standard tissue culture plastic, serum withdrawal did not cause aggregation of CHI pMSCs or downregulation of mesenchymal proteins, although a possible downregulation of the stem cell marker SOX9 (Seymour et al., 2007, Furuyama et al., 2011,

Seymour, 2013, Kawaguchi, 2013) was observed, shown in Figure 4.2. We did find that the pMSCs would spontaneously aggregate when placed into ultra low adherence culture, with or without serum.

This spontaneous aggregation in serum-free conditions led to the formation of cell spheroids resem- bling human islets with a trend of down-regulation of α-smooth muscle actin. α-smooth muscle actin is a protein associated with active hepatic stellate cells (Carpino et al., 2005) and so may be related to invasiveness and extracellular matrix deposition. No other mesenchymal proteins were affected and hormones (insulin and glucagon) were not detected as mRNA or protein.

Several protocols for differentiating MSCs to β-like cells were trialled on the CHI pMSCs with poor results, however, we were partially able to replicate the protocol developed by Zanini et al.

(2011), in that insulin was detected as both gene and protein expression. The initial stage of this protocol, treatment with retinoic acid appeared to have no effect apart from an unconfirmed decrease in HES1 expression. Retinoic acid is a known NOTCH pathway inhibitor and HES1 is a downstream

NOTCH target. During development when the NOTCH pathway is suppressed, NGN3 is upregu- lated but we did not observe this. We found that addition of DKK1, a Wnt inhibitor, for the first

72 hours of this protocol enhanced differentiation, with greater PDX1 and INS gene expression and

174 greater intensity of zinc staining which is shown in Figure 4.2. The combination of retinoic acid, activin A and DKK1 induced NEUROD1, which has not been reported previously. The combination of activin A and Wnt3a, followed by DKK1 and retinoic acid induced PDX1 expression in adipose derived MSCs (Li et al., 2012), so similar signals may be inducing NEUROD1 in CHI pMSCs. Ac- tivin A is known to be expressed in islet cells (Yasuda et al., 1993), and function as an activator of stellate cells (Ohnishi et al., 2003), which are similar to MSCs. DKK1 is involved in the sig- nalling when cells undergo MET, to down-regulate SOX9 and upregulate NGN3 (Larsen et al., 2015) and which may be why DKK1 enhanced differentiation of mesenchymal cells towards the endocrine cell types. Several pancreatic transcription factors are also used to signal for neuronal pathways

(Wilson et al., 2003, Martens et al., 2011), including NEUROD1. We therefore assessed whether the cells were being induced towards a neuronal lineage by examining TUB3. TUB3 was induced alongside NEUROD1, but was also induced by each of the factors alone or in combination (except where the combination caused severe growth inhibition, for DKK1 alone, or cell death for activin A and retinoic acid where gene expression could therefore not be assessed). Following the induction of these neuronal markers, the use of the cocktail of growth factors down-regulated the neuronal markers but instead upregulated the pancreas specific PDX1. PDX1 is essential for differentiating MSCs to insulin secreting cells (Yuan et al., 2010) highlighting the importance of growth factors, including be- tacellulin and exendin-4, in switching the CHI pMSCs from a neuronal fate towards a pancreatic fate.

Following the induction of PDX1 in CHI pMSCs, we have shown that they will differentiate fur- ther towards insulin containing cells. We observed intense insulin staining by immunofluorescence, shown in Figure 4.4. It has been reported that insulin stains can be artefactual in culture due to apoptotic cells absorbing insulin from cell culture media (Hansson et al., 2004). The insulin staining we saw was of greater intensity in differentiated cells compared to the control cells despite both be- ing cultured in the same concentration of insulin. Further, we saw INS mRNA and faint C-peptide staining, again of greater intensity in the differentiated cells compared to control cells, both of which support the fact that the insulin staining is, at least in part, as a result of production of insulin by the cells rather than absorption. Further, in the differentiated cells only, we saw some insulin secretion and content by ELISA. The cells were not glucose responsive suggesting immaturity, likely due to the absence of KATP channels and potentially other factors as we did not assess glucokinase or glut transporters.

With only immature β-like cells being produced from the CHI MSCs, we wanted to assess if iP-

SCs would be a superior stem cell source. The iPSC line was derived from the F-CHI MSCs, which have a healthy allele of the ABCC8 gene (see Table 4.1) and so can be used for screening protocols,

175 although a healthy control iPSC line could also have been used, with a CHI causing mutation later engineered in. The cells were characterised for pluripotency, with consistent expression of the nuclear factors OCT4, NANOG and SOX2 seen after clearance of the transfection virus, as shown in Figures

4.9 and 4.10. Differentiation to the three germ layers was also observed via generation of embryoid bodies, with low efficiency of differentiation to mesoderm seen. Low numbers of HAND1 cells may have been due to assessing the EBs too early, expression peaks at day 8 in mouse ESC differentiation

(Smart et al., 2010), where mouse cells exhibit a shorter differentiation timescale; human EBs show increased expression of HAND1 after 14 days of differentiation (Cai et al., 2006). Bias of iPSCs to certain germ layers or lineages is often seen in iPSCs, with variation between clones (Bock et al.,

2011), patients/donors (Shao et al., 2013) and source cell type seen (Kim et al., 2011). Culturing the EBs for longer may help confirm if our iPSCs had full differentiation potential to the mesoderm lineage, or if this is just variation; it may be expected that the cells would not be biased against the mesoderm lineage being derived from MSCs, unless the MSCs were not truly mesodermal due to being derived from the pancreas. As we were most interested in differentiation towards endodermal lineages, the bias seen in the clone we selected was acceptable for our use.

The source cell type when deriving an iPSC line influences its epigenome and differentiation potential in some cases (Kim et al., 2011). Deriving iPSCs from β-cells yields β-cell specific hy- pomethylation and a strong bias towards differentiation to β-cells when compared to iPSCs from foreskin fibroblasts (Bar-Nur et al., 2011). The protocol used to show this differentiation bias is based on that of (D’Amour et al., 2006), which as discussed in Sections 4.4.6 and 4.7.5 was lethal to the cells without feeder layers. We had previously found that MSCs derived from the pancreas also showed regions of demethylation upstream of pancreas-specific genes, including PDX1 and AMY2A.

Comparing these marks in the MSCs and iPSCs (i.e the same cells before and after transduction), we found a trend of increased methylation in the iPSCs as can be seen in Figure 4.12, including at the loci that were significantly different between pancreas and non-pancreas in the MSCs. The only exception to this was the locus in AMY2A which was significantly less methylated in pancreatic

MSCs, this was even further de-methylated in the iPSCs. This may be just an aberrant result of the reprogramming process, further samples would be needed to clarify this. This increased methylation is most likely due to the fact that we re-programmed our cells using the ‘Yamanaka factors’ (Taka- hashi et al., 2007), failure to de-methylate genes has been seen with this combination of factors before i.e. when KLF4 and cMYC are used, rather than NANOG and LIN28 (Planello et al., 2014). We did not see hypomethylation upstream of INS as was reported with β-cell derived iPSCs (Bar-Nur et al., 2011), but as the iPSCs were derived from MSCs from mixed pancreatic cells, differential methylation may be seen for other genes such as PDX1 which we unfortunately could not assess.

176 One further consideration is that iPSCs reportedly become more ES-like, epigenetically, as they are passaged (Nishino et al., 2011). We assessed methylation after around 20 passages, Bar-Nur et al.

(2011) do not mention at what point they assessed methylation status. It may therefore be the case that lineage-specific bias is only seen for the first few passages rendering the tissue source largely unimportant (Krijger et al., 2016). The concept of lineage specific bias in iPSCs is controversial, despite the previous studies, others have shown that person to person variability in methylation is greater than tissue source (Kyttala et al., 2016) and that in some cases this bias can be a result of incompletely reprogrammed cells (Muller et al., 2011). Further iPSC lines derived from pMSCs would be required to confirm if the methylation status was common to all pancreas-derived iPSCs, as no differences could be confirmed just using one line, healthy aged matched controls would also be useful in case there is disease specific methylation differences.

Despite unclear results on whether the iPSCs had a pancreatic ‘memory’, we nevertheless wanted to assess their ability to differentiate to β-cells. In order for the iPSCs to be a tool for research into CHI they need to be differentiated to functional β-cells. We assessed two published protocols

(referred to as ‘Melton’ and ‘Kieffer’ protocols), in order to test whether they are able to differ- entiate the cells as reported and to compare which is more effective for our cells which is shown in Figures 4.13 - 4.16. The third protocol (D’Amour et al., 2006) that was investigated was lethal to the cells without feeder layers, seemingly related to the Wnt3a and so was not assessed further.

This adds further discrepancy when trying to assess whether our cells showed a β-cell bias as in the

Bar-Nur et al. (2011) paper but this was judged to be less important than feeder-free culture with the variability that use of feeder cells can add. Both the ‘Melton’ and ‘Kieffer’ protocols yielded definitive endoderm from the CHI iPSCs with good efficiency (around 90% of cells were positive for

SOX17 and/or FOXA2) and we found no significant difference between the two protocols. The only difference is that activin A, used in the ‘Melton’ protocol is less expensive than GDF8/myostatin used in the ‘Kieffer’ protocol. At the next stage, pancreatic endoderm we again saw high efficiency, with the majority of cells being PDX1 positive. Due to the amount of cytoplasmic staining seen, and the morphology of the cells at this stage we were unable to quantify the number of PDX1 positive cells to calculate or compare the efficiency. There was however, 10-fold higher expression of PDX1 when using the ‘Melton’ protocol. Comparison of the total amount of PDX1 protein by western blot at this stage of both protocols would clarify if the difference in gene expression was translated.

For the final stage, from PDX1 positive to hormone-positive cells, only the ‘Kieffer’ protocol was assessed, due to the ‘Melton’ protocol calling for equipment not available within many labs including our own, and following inconclusive differences between the two protocols. For this final stage, the

177 Rezania et al. (2014) paper provided two alternate methods - culturing the cells in ultra low adher- ence plates, or at the air-liquid interface on trans-well filter inserts. The authors suggest that the latter is more effective, but we were unable to achieve viable cells from this protocol2. With culture in ultra low adherence the cells readily formed islet-like clusters providing the cells were not dispersed to single cells prior to transfer. We found these clusters were positive for genes associated with α- and β-cells including PDX1, NEUROD1, GCG and ABCC8 but we did not see INS expressed. Ex- amining the proteins expressed in these clusters showed that they expressed glucagon, as per the gene expression. They also appeared to strongly express insulin, but not C-peptide. This may suggest that the insulin has been absorbed from the media as has been previously reported (Hansson et al.,

2004) or that there were issues with the assays. Further experiments would clarify which is the case, as due to time constraints the full protocol was only performed twice, once to assess gene expression and once to assess the proteins. Regardless, this data suggests that this protocol has generated α-like cells. Even if β-cells are also generated, the presence of glucagon-expressing cells would mean that the protocol requires further optimisation. Control healthy iPSCs would be useful to optimise this protocol in our lab, as it has been reported that there are cells which are both glucagon and insulin positive in CHI. A longer timeframe for the final stages of differentiation may be needed to develop mature insulin secreting cells, the timing used in these experiments was the minimum suggested in the original papers, but the authors presented data from much later differentiation points too.

Formation of cell spheroids was essential for differentiation of both CHI pMSCs and iPSCs to- wards insulin containing cells following induction of PDX1. MSCs differentiated by our protocol, but as a monolayer did not stain positive for insulin. Cell-cell interactions are essential in islets

(Bavamian et al., 2007, Benninger et al., 2011), with islets maintained in gels showing superior in- sulin secretion (Beattie et al., 2002), and so this is presumably the reason why 3D culture aided differentiation of the MSCs towards an insulin secreting phenotype. Some of the undifferentiated

MSC spheroids had a capsule which appeared to be comprised of extracellular matrix, which can be secreted by MSCs (Amable et al., 2014). This capsule would support the structure and cell- cell interactions of spheroids, but was absent from differentiated cells due to the concomitant loss of matrix-secretion undifferentiated MSCs. Culture in hydrogels (Beattie et al., 2002) or in colla- gen (Mason et al., 2009) may therefore likely support differentiation further in both MSCs and iPSCs.

In conclusion, we have successfully derived iPSCs from the pancreas tissue of a patient with focal

CHI, following expansion of the pancreas ex vivo as MSCs. We chose to derive iPSCs from the

2Following the cessation of my lab work, whilst I was writing this thesis KM was able to obtain viable cells at the air-liquid interface, these were positive for NGN3 but not MAFA and so this method may also not be the ‘silver bullet’.

178 pancreas rather than blood cells despite the very limited amount of tissue obtained, in an attempt to maintain epigenetic memory as previously reported, but we did not see strong evidence of this occurring. In this study, we present a step-wise method to differentiate CHI pMSCs towards an immature insulin secreting cell, and an assessment of protocols for differentiating iPSCs to the same fate, via PDX1 positive pancreatic endoderm. Based on a published protocol, we found that the addition of DKK1 enhanced differentiation of the MSCs towards a β-cell phenotype. Whilst serum withdrawal alone did not cause differentiation of MSCs to islet-like spheroids as has been reported previously (Davani et al., 2009, Carlotti et al., 2010), culture of both types of stem cells in float- ing spheroids proved essential. We were able to efficiently differentiate the cells to PDX1 positive progenitors using published protocols, via definitive endoderm. The final cells produced resembled

α-cells more than β-cells though, suggesting further optimisation of the differentiation process is required, possibly using the first stages of the ‘Melton’ protocol to induce maximal PDX1 with the

final stage of the ‘Kieffer’ protocol to avoid the use of spinner flasks and investigate the efficiency of air-liquid interface culture. One advantage to using the iPSCs over MSCs for CHI research was that with the iPSCs we induced expression of ABCC8, although we did not assess the protein so it is possible that functional KATP channels were not present. As ABCC8 is the gene responsible for causing CHI in most cases (Dunne et al., 1997, Glaser et al., 2000), including this one, expression of this would allow examination of the channel function in response to drugs currently used, or being developed, to treat CHI. As this cell line is heterozygous for an ABCC8 mutation, a healthy allele should be produced and correctly trafficked, allowing optimisation before developing lines which may not correctly localise the channel proteins. The disadvantage of using the iPSCs however, is that the

CHI iPSCs did not clearly contain or secrete insulin, whereas the MSCs appeared to. With further work to produce authentic functional β-cells both MSC and iPSC derived β-cells could be used for modelling and drug testing in CHI; the use of two stem cell sources will likely yield more information than just a single source based on our gene expression and epigenetic data. Once the model has been further developed, control cell lines (preferably) age matched would be needed, alongside human islets as a positive control, to confirm any putative disease specific results.

179 4.6 References

R. d. S. Abreu, L. O. Penalva, E. M. Marcotte, and C. Vogel. Global signatures of protein and

mRNA expression levels. Molecular bioSystems, 5(12):1512–1526, 2009.

U. Ahlgren, J. Jonsson, and H. Edlund. The morphogenesis of the pancreatic mesenchyme is uncou-

pled from that of the pancreatic epithelium in IPF1/PDX1-deficient mice. Development, 122(5):

1409–1416, 1996.

P. R. Amable, M. V. T. Teixeira, R. B. V. Carias, J. M. Granjeiro, and R. Borojevic. Protein

synthesis and secretion in human mesenchymal cells derived from bone marrow, adipose tissue

and Wharton’s jelly. Stem Cell Research & Therapy, 5(2):53–53, 2014.

J.-B. Arnoux, P. de Lonlay, M.-J. Ribeiro, K. Hussain, O. Blankenstein, K. Mohnike,

V. Valayannopoulos, J.-J. Robert, J. Rahier, C. Sempoux, C. Bellanne, V. Verkarre, Y. Aigrain,

F. Jaubert, F. Brunelle, and C. Nihoul-Fekete. Congenital hyperinsulinism. Early Human Devel-

opment, 86(5):287–294, 2010.

J.-B. Arnoux, V. Verkarre, C. Saint-Martin, F. Montravers, A. Brassier, V. Valayannopoulos,

F. Brunelle, J.-C. Fournet, J.-J. Robert, Y. Aigrain, C. Bellanne-Chantelot, and P. de Lonlay.

Congenital hyperinsulinism: current trends in diagnosis and therapy. Orphanet Journal of Rare

Diseases, 6(1):1750–1172, 2011.

V. B. Arya, Q. Aziz, A. Nessa, A. Tinker, and K. Hussain. Congenital hyperinsulinism: clinical and

molecular characterisation of compound heterozygous ABCC8 mutation responsive to Diazoxide

therapy. International Journal of Pediatric Endocrinology, 2014(1):1–5, 2014.

O. Bar-Nur, H. Russ, S. Efrat, and N. Benvenisty. Epigenetic Memory and Preferential Lineage-

Specific Differentiation in Induced Pluripotent Stem Cells Derived from Human Pancreatic Islet

Beta Cells. Cell Stem Cell, 9(1):17–23, 2011.

V. L. Battula, P. M. Bareiss, S. Treml, S. Conrad, I. Albert, S. Hojak, H. Abele, B. Schewe, L. Just,

T. Skutella, and H.-J. Bahring. Human placenta and bone marrow derived MSC cultured in

serum-free, b-FGF-containing medium express cell surface frizzled-9 and SSEA-4 and give rise to

multilineage differentiation. Differentiation, 75(4):279 – 291, 2007.

S. Bavamian, P. Klee, A. Britan, C. Populaire, D. Caille, J. Cancela, A. Charollais, and P. Meda. Islet-

cell-to-cell communication as basis for normal insulin secretion. Diabetes, Obesity and Metabolism,

9:118–132, 2007.

180 G. M. Beattie, A. M. Montgomery, A. D. Lopez, E. Hao, B. Perez, M. L. Just, J. R. Lakey, M. E.

Hart, and A. Hayek. A Novel Approach to Increase Human Islet Cell Mass While Preserving β-Cell

Function. Diabetes, 51(12):3435–3439, 2002.

R. K. P. Benninger, W. S. Head, M. Zhang, L. S. Satin, and D. W. Piston. Gap junctions and other

mechanisms of cell-cell communication regulate basal insulin secretion in the pancreatic islet. The

Journal of Physiology, 589(22):5453–5466, 2011.

D. R. Bhandari, K.-W. Seo, B. Sun, M.-S. Seo, H.-S. Kim, Y.-J. Seo, J. Marcin, N. Forraz, H. L.

Roy, D. Larry, M. Colin, and K.-S. Kang. The simplest method for in vitro β-cell production from

human adult stem cells. Differentiation, 82(3):144 – 152, 2011.

R. R. Bhonde, P. Sheshadri, S. Sharma, and A. Kumar. Making surrogate β-cells from mesenchymal

stromal cells: Perspectives and future endeavors. The International Journal of Biochemistry &

Cell Biology, 46:90 – 102, 2014.

C. Bock, E. Kiskinis, G. Verstappen, H. Gu, G. Boulting, Z. D. Smith, M. Ziller, G. F. Croft, M. W.

Amoroso, D. H. Oakley, A. Gnirke, K. Eggan, and A. Meissner. Reference Maps of Human ES

and iPS Cell Variation Enable High-Throughput Characterization of Pluripotent Cell Lines. Cell,

144(3):439–452, 2011.

D. Bosnakovski, M. Mizuno, G. Kim, T. Ishiguro, M. Okumura, T. Iwanaga, T. Kadosawa, and

T. Fujinaga. Chondrogenic differentiation of bovine bone marrow mesenchymal stem cells in pellet

cultural system. Experimental Hematology, 32(5):502–509, 2004.

O. Cabrera, D. M. Berman, N. S. Kenyon, C. Ricordi, P.-O. Berggren, and A. Caicedo. The unique

cytoarchitecture of human pancreatic islets has implications for islet cell function. Proceedings of

the National Academy of Sciences of the United States of America, 103(7):2334–2339, 2006.

J. Cai, J. Chen, Y. Liu, T. Miura, Y. Luo, J. F. Loring, W. J. Freed, M. S. Rao, and X. Zeng.

Assessing self-renewal and differentiation in human embryonic stem cell lines. 24(3):516–530,

2006. ISSN 1549-4918.

F. Carlotti, A. Zaldumbide, C. J. Loomans, E. van Rossenberg, M. Engelse, E. J. de Koning, and

R. C. Hoeben. Isolated human islets contain a distinct population of mesenchymal stem cells.

Islets, 2(3):164–173, 2010.

G. Carpino, S. Morini, S. Ginanni Corradini, A. Franchitto, M. Merli, M. Siciliano, F. Gentili,

A. Onetti Muda, P. Berloco, M. Rossi, A. Attili, and E. Gaudio. Alpha-SMA expression in hepatic

stellate cells and quantitative analysis of hepatic fibrosis in cirrhosis and in recurrent chronic

hepatitis after liver transplantation. Digestive and Liver Disease, 37(5):349–356, 2005.

181 G. Chen, D. R. Gulbranson, Z. Hou, J. M. Bolin, V. Ruotti, M. D. Probasco, K. Smuga-Otto, S. E.

Howden, N. R. Diol, N. E. Propson, R. Wagner, G. O. Lee, J. Antosiewicz-Bourget, J. M. C. Teng,

and J. A. Thomson. Chemically defined conditions for human iPS cell derivation and culture.

Nature methods, 8(5):424–429, 2011.

E. Corritore, E. Dugnani, V. Pasquale, R. Misawa, P. Witkowski, J. Lei, J. Markmann, L. Piemonti,

E. M. Sokal, S. Bonner-Weir, and P. A. Lysy. β-Cell Differentiation of Human Pancreatic Duct-

Derived Cells After In Vitro Expansion. Cellular Reprogramming, 16(6):456–466, 2014.

M. P. Dalvi, M. R. Umrani, M. V. Joglekar, and A. A. Hardikar. Human pancreatic islet progenitor

cells demonstrate phenotypic plasticity in vitro. Journal of Biosciences, 34(4):523–528, 2009.

K. A. D’Amour, A. G. Bang, S. Eliazer, O. G. Kelly, A. D. Agulnick, N. G. Smart, M. A. Moorman,

E. Kroon, M. K. Carpenter, and E. E. Baetge. Production of pancreatic hormone-expressing

endocrine cells from human embryonic stem cells. Nat Biotech, 24(11):1392–1401, 2006.

B. Davani, S. Ariely, L. Ikonomou, Y. Oron, and M. C. Gershengorn. Human islet-derived precursor

cells can cycle between epithelial clusters and mesenchymal phenotypes. Journal of Cellular and

Molecular Medicine, 13:2570–2581, 2009.

M. J. Dunne, C. Kane, R. M. Shepherd, J. A. Sanchez, R. F. James, P. R. Johnson, A. Aynsley-

Green, S. Lu, J. P. Clement, K. J. Lindley, S. Seino, L. Aguilar-Bryan, G. Gonzalez, and P. J. Milla.

Familial Persistent Hyperinsulinemic Hypoglycemia of Infancy and Mutations in the Sulfonylurea

Receptor. New England Journal of Medicine, 336(10):703–706, 1997.

M. J. Dunne, K. E. Cosgrove, R. M. Shepherd, A. Aynsley-Green, and K. J. Lindley. Hyperinsulinism

in Infancy: From Basic Science to Clinical Disease. Physiological Reviews, 84(1):239–275, 2004.

J.-C. Fournet, C. Mayaud, P. de Lonlay, M.-S. Gross-Morand, V. Verkarre, M. Castanet, M. Devillers,

J. Rahier, F. Brunelle, J.-J. Robert, C. Nihoul-Fekete, J.-M. Saudubray, and C. Junien. Unbalanced

Expression of 11p15 Imprinted Genes in Focal Forms of Congenital Hyperinsulinism. The American

Journal of Pathology, 158(6):2177–2184, 2001.

K. Furuyama, Y. Kawaguchi, H. Akiyama, M. Horiguchi, S. Kodama, T. Kuhara, S. Hosokawa,

A. Elbahrawy, T. Soeda, M. Koizumi, T. Masui, M. Kawaguchi, K. Takaori, R. Doi, E. Nishi,

R. Kakinoki, J. M. Deng, R. R. Behringer, T. Nakamura, and S. Uemoto. Continuous cell supply

from a Sox9-expressing progenitor zone in adult liver, exocrine pancreas and intestine. Nat Genet,

43(1):34–41, 2011.

M. M. Gabr, M. M. Zakaria, A. F. Refaie, S. M. Khater, S. A. Ashamallah, A. M. Ismail, N. El-Badri,

and M. A. Ghoneim. Generation of Insulin-Producing Cells from Human Bone Marrow-Derived

182 Mesenchymal Stem Cells: Comparison of Three Differentiation Protocols. BioMed Research In-

ternational, 832736:9, 2014.

B. Glaser, F. Ryan, M. Donath, H. Landau, C. A. Stanley, L. Baker, D. E. Barton, and P. S.

Thornton. Hyperinsulinism caused by paternal-specific inheritance of a recessive mutation in the

sulfonylurea-receptor gene. Diabetes, 48(8):1652–1657, 1999.

B. Glaser, P. Thornton, T. Otonkoski, and C. Junien. Genetics of neonatal hyperinsulinism. Archives

of Disease in Childhood - Fetal and Neonatal Edition, 82(2):F79–F86, 2000.

M. Gouzi, Y. H. Kim, K. Katsumoto, K. Johansson, and A. Grapin-Botton. Neurogenin3 initiates

stepwise delamination of differentiating endocrine cells during pancreas development. Dev. Dyn.,

240(3):589–604, 2011.

T. U. Greiner, G. Kesavan, A. Stahlberg, and H. Semb. Rac1 regulates pancreatic islet morphogenesis.

BMC Developmental Biology, 9(1):1–13, 2009.

N. Hanley. Closing in on pancreatic beta cells. Nat Biotech, 32(11):1100–1102, Nov. 2014.

M. Hansson, A. Tonning, U. Frandsen, A. Petri, J. Rajagopal, M. C. Englund, R. S. Heller, J. Hakans-

son, J. Fleckner, H. N. Skold, D. Melton, H. Semb, and P. Serup. Artifactual Insulin Release From

Differentiated Embryonic Stem Cells. Diabetes, 53(10):2603–2609, 2004.

J.-C. Henquin, C. Sempoux, J. Marchandise, S. Godecharles, Y. Guiot, M. Nenquin, and J. Rahier.

Congenital Hyperinsulinism Caused by Hexokinase I Expression or Glucokinase-Activating Muta-

tion in a Subset of β-Cells. Diabetes, 62(5):1689–1696, 2013.

L. Ikonomou, E. Geras-Raaka, B. M. Raaka, and M. C. Gershengorn. β-catenin signalling in mes-

enchymal islet-derived precursor cells. Cell Proliferation, 41(3):474–491, 2008.

A. Jafarian, M. Taghikhani, S. Abroun, Z. Pourpak, A. Allahverdi, and M. Soleimani. Generation

of high-yield insulin producing cells from human bone marrow mesenchymal stem cells. Molecular

Biology Reports, 41(7):4783–4794, 2014.

J. Jensen, R. S. Heller, T. Funder-Nielsen, E. E. Pedersen, C. Lindsell, G. Weinmaster, O. D. Madsen,

and P. Serup. Independent development of pancreatic alpha- and beta-cells from neurogenin3-

expressing precursors: a role for the notch pathway in repression of premature differentiation.

Diabetes, 49(2):163–176, 2000.

M. V. Joglekar, V. M. Joglekar, S. V. Joglekar, and A. A. Hardikar. Human fetal pancreatic insulin-

producing cells proliferate in vitro. Journal of Endocrinology, 201(1):27–36, 2009.

183 Y. Kawaguchi. Sox9 and programming of liver and pancreatic progenitors. J Clin Invest, 123(5):

1881–1886, 2013.

I. Ketola, T. Otonkoski, M.-A. Pulkkinen, H. Niemi, J. Palgi, C. M. Jacobsen, D. B. Wilson, and

M. Heikinheimo. Transcription factor GATA-6 is expressed in the endocrine and GATA-4 in the

exocrine pancreas. Molecular and Cellular Endocrinology, 226(1–2):51–57, 2004.

K. Kim, R. Zhao, A. Doi, K. Ng, J. Unternaehrer, P. Cahan, H. Hongguang, Y.-H. Loh, M. J. Aryee,

M. W. Lensch, H. Li, J. J. Collins, A. P. Feinberg, and G. Q. Daley. Donor cell type can influence

the epigenome and differentiation potential of human induced pluripotent stem cells. Nat Biotech,

29(12):1117–1119, 2011.

S.-J. Kim, Y.-S. Choi, E.-S. Ko, S.-M. Lim, C.-W. Lee, and D.-I. Kim. Glucose-stimulated insulin

secretion of various mesenchymal stem cells after insulin-producing cell differentiation. Journal of

Bioscience and Bioengineering, 113(6):771 – 777, 2012.

P. Krijger, B. DiStefano, E. deWit, F. Limone, C. vanOevelen, W. deLaat, and T. Graf. Cell-of-

origin-specific 3d genome structure acquired during somatic cell reprogramming. Cell Stem Cell,

18(5):597–610, 2016.

Y. Kunisada, N. Tsubooka-Yamazoe, M. Shoji, and M. Hosoya. Small molecules induce efficient

differentiation into insulin-producing cells from human induced pluripotent stem cells. Stem Cell

Research, 8(2):274–284, 2012.

A. Kyttala, R. Moraghebi, C. Valensisi, J. Kettunen, C. Andrus, K. Pasumarthy, M. Nakanishi,

K. Nishimura, M. Ohtaka, J. Weltner, B. VanHandel, O. Parkkonen, J. Sinisalo, A. Jalanko,

R. Hawkins, N.-B. Woods, T. Otonkoski, and R. Trokovic. Genetic variability overrides the impact

of parental cell type and determines ipsc differentiation potential. 6(2):200–212, 2016.

B. M. Larsen, S. M. Hrycaj, M. Newman, Y. Li, and D. M. Wellik. Mesenchymal Hox6 function

is required for mouse pancreatic endocrine cell differentiation. Development, 142(22):3859–3868,

2015.

R. Lehmann, R. A. Zuellig, P. Kugelmeier, P. B. Baenninger, W. Moritz, A. Perren, P.-A. Clavien,

M. Weber, and G. A. Spinas. Superiority of Small Islets in Human Islet Transplantation. Diabetes,

56(3):594–603, 2007.

J. Li, L. Zhu, X. Qu, J. Li, R. Lin, L. Liao, J. Wang, S. Wang, Q. Xu, and R. C. Zhao. Stepwise

Differentiation of Human Adipose-Derived Mesenchymal Stem Cells Toward Definitive Endoderm

and Pancreatic Progenitor Cells by Mimicking Pancreatic Development In Vivo. Stem Cells and

Development, 22(10):1576–1587, 2012.

184 L.-C. Li and R. Dahiya. MethPrimer: designing primers for methylation PCRs. Bioinformatics, 18

(11):1427–1431, 2002.

M. J. Lima, K. R. Muir, H. M. Docherty, R. Drummond, N. W. McGowan, S. Forbes, Y. Heremans,

I. Houbracken, J. A. Ross, S. J. Forbes, P. Ravassard, H. Heimberg, J. Casey, and K. Docherty.

Suppression of Epithelial-to-Mesenchymal Transitioning Enhances Ex Vivo Reprogramming of

Human Exocrine Pancreatic Tissue Toward Functional Insulin-Producing β-Like Cells. Diabetes,

62(8):2821–2833, 2013.

R. Lister, M. Pelizzola, Y. S. Kida, R. D. Hawkins, J. R. Nery, G. Hon, J. Antosiewicz-Bourget,

R. O/’Malley, R. Castanon, S. Klugman, M. Downes, R. Yu, R. Stewart, B. Ren, J. A. Thomson,

R. M. Evans, and J. R. Ecker. Hotspots of aberrant epigenomic reprogramming in human induced

pluripotent stem cells. Nature, 471(7336):68–73, 2011.

I. Mallona, A. Diez-Villanueva, and M. A. Peinado. Methylation plotter: a web tool for dynamic

visualization of DNA methylation data. Source Code for Biology and Medicine, 9(1):1–5, 2014.

G. A. Martens, L. Jiang, K. H. Hellemans, G. Stang, H. Heimberg, F. C. Nielsen, O. Sand,

J. Van Helden, F. K. Gorus, and D. G. Pipeleers. Clusters of Conserved Beta Cell Marker Genes

for Assessment of Beta Cell Phenotype. PLoS ONE, 6(9):e24134–, 2011.

M. N. Mason, C. A. Arnold, and M. J. Mahoney. Entrapped Collagen Type 1 Promotes Differentiation

of Embryonic Pancreatic Precursor Cells into Glucose-Responsive β-Cells When Cultured in Three-

Dimensional PEG Hydrogels. Tissue Engineering Part A, 15(12):3799–3808, 2009.

T. Meissner, U. Wendel, P. Burgard, S. Schaetzle, and E. Mayatepek. Long-term follow-up of 114

patients with congenital hyperinsulinism. European Journal of Endocrinology, 149(1):43–51, 2003.

J. K. Mfopou and L. Bouwens. Diabetes: β cells at last. Nat Rev Endocrinol, 11(1):5–6, Jan. 2015.

F.-J. Muller, B. M. Schuldt, R. Williams, D. Mason, G. Altun, E. P. Papapetrou, S. Danner, J. E.

Goldmann, A. Herbst, N. O. Schmidt, J. B. Aldenhoff, L. C. Laurent, and J. F. Loring. A

bioinformatic assay for pluripotency in human cells. 8(4):315–317, 2011.

L. C. Murtaugh, B. Z. Stanger, K. M. Kwan, and D. A. Melton. Notch signaling controls multiple

steps of pancreatic differentiation. Proceedings of the National Academy of Sciences, 100(25):

14920–14925, 2003.

K. Nishino, M. Toyoda, M. Yamazaki-Inoue, Y. Fukawatase, E. Chikazawa, H. Sakaguchi, H. Akutsu,

and A. Umezawa. DNA Methylation Dynamics in Human Induced Pluripotent Stem Cells over

Time. PLoS Genet, 7(5):e1002085–, May 2011.

185 H. Ohnishi, Y. Oda, T. Aoki, M. Tadokoro, Y. Katsube, H. Ohgushi, K. Hattori, and S. Yuba. A com-

parative study of induced pluripotent stem cells generated from frozen, stocked bone marrow- and

adipose tissue-derived mesenchymal stem cells. Journal of Tissue Engineering and Regenerative

Medicine, 6(4):261–271, 2012.

N. Ohnishi, T. Miyata, H. Ohnishi, H. Yasuda, K. Tamada, N. Ueda, H. Mashima, and K. Sugano.

Activin A is an autocrine activator of rat pancreatic stellate cells: potential therapeutic role of

follistatin for pancreatic fibrosis. Gut, 52(10):1487–1493, 2003.

F. Pagliuca, J. Millman, M. Gurtler, M. Segel, A. VanDervort, J. Ryu, Q. Peterson, D. Greiner, and

D. Melton. Generation of Functional Human Pancreatic β Cells In Vitro. Cell, 159(2):428–439,

2014.

A. C. Planello, J. Ji, V. Sharma, R. Singhania, F. Mbabaali, F. Muller, J. A. Alfaro, C. Bock,

D. D. De Carvalho, and N. N. Batada. Aberrant DNA methylation reprogramming during in-

duced pluripotent stem cell generation is dependent on the choice of reprogramming factors. Cell

Regeneration, 3(1):1–14, 2014.

G. Qing-Song, Z. Ming-Yan, W. Lei, F. Xiang-Jun, L. Yu-Hua, W. Zhi-Wei, Z. Sha-Jun, W. Yao,

and H. Yan. Combined Transfection of the Three Transcriptional Factors, PDX-1, NeuroD1, and

MafA, Causes Differentiation of Bone Marrow Mesenchymal Stem Cells into Insulin-Producing

Cells. Experimental Diabetes Research, page 10, 2012.

A. Rezania, J. E. Bruin, M. J. Riedel, M. Mojibian, A. Asadi, J. Xu, R. Gauvin, K. Narayan,

F. Karanu, J. J. O’Neil, Z. Ao, G. L. Warnock, and T. J. Kieffer. Maturation of Human Embryonic

Stem Cell-Derived Pancreatic Progenitors Into Functional Islets Capable of Treating Pre-existing

Diabetes in Mice. Diabetes, 61(8):2016–2029, 2012.

A. Rezania, J. E. Bruin, P. Arora, A. Rubin, I. Batushansky, A. Asadi, S. O’Dwyer, N. Quiskamp,

M. Mojibian, T. Albrecht, Y. H. C. Yang, J. D. Johnson, and T. J. Kieffer. Reversal of diabetes

with insulin-producing cells derived in vitro from human pluripotent stem cells. Nat Biotech, 32

(11):1121–1133, 2014.

K. D. Robertson. Dna methylation and human disease. Nat Rev Genet, 6(8):597–610, 2005.

H. A. Russ, P. Ravassard, J. Kerr-Conte, F. Pattou, and S. Efrat. Epithelial-Mesenchymal Transition

in Cells Expanded In Vitro from Lineage-Traced Adult Human Pancreatic Beta Cells. PLoS ONE,

4(7):e6417, 2009.

R. J. Salisbury, B. Han, R. E. Jennings, A. A. Berry, A. Stevens, Z. Mohamed, S. A. Sugden,

R. De Krijger, S. E. Cross, P. P. Johnson, M. Newbould, K. E. Cosgrove, K. P. Hanley, I. Banerjee,

186 M. J. Dunne, and N. A. Hanley. Altered Phenotype of β-Cells and Other Pancreatic Cell Lineages

in Patients With Diffuse Congenital Hyperinsulinism in Infancy Caused by Mutations in the ATP-

Sensitive K-Channel. Diabetes, 64(9):3182–3188, 2015.

K. L. Seeberger, J. M. Dufour, A. M. J. Shapiro, J. R. T. Lakey, R. V. Rajotte, and G. S. Korbutt.

Expansion of mesenchymal stem cells from human pancreatic ductal epithelium. Lab Invest, 86

(2):141–153, 2006.

P. A. Seymour. Sox9: A Master Regulator of the Pancreatic Program. The Review of Diabetic

Studies : RDS, 11(1):51–83, 2013.

P. A. Seymour, K. K. Freude, M. N. Tran, E. E. Mayes, J. Jensen, R. Kist, G. Scherer, and M. Sander.

SOX9 is required for maintenance of the pancreatic progenitor cell pool. Proceedings of the National

Academy of Sciences, 104(6):1865–1870, 2007.

K. Shao, C. Koch, M. K. Gupta, Q. Lin, M. Lenz, S. Laufs, B. Denecke, M. Schmidt, M. Linke,

H. C. Hennies, J. Hescheler, M. Zenke, U. Zechner, T. Saric, and W. Wagner. Induced Pluripotent

Mesenchymal Stromal Cell Clones Retain Donor-derived Differences in DNA Methylation Profiles.

Mol Ther, 21(1):240–250, 2013.

K. Shimomura, M. Tusa, M. Iberl, M. F. Brereton, S. Kaizik, P. Proks, C. Lahmann, N. Yaluri,

S. Modi, H. Huopio, J. Ustinov, T. Otonkoski, M. Laakso, and F. M. Ashcroft. A Mouse Model of

Human Hyperinsulinism Produced by the E1506K Mutation in the Sulphonylurea Receptor SUR1.

Diabetes, 62(11):3797–3806, 2013.

N. Smart, K. N. Dub, and P. R. Riley. Identification of thymosin β4 as an effector of hand1-mediated

vascular development. Nature Communications, 1:46, 2010.

S. Sugii, Y. Kida, T. Kawamura, J. Suzuki, R. Vassena, Y.-Q. Yin, M. K. Lutz, W. T. Berggren,

J. C. Izpisa Belmonte, and R. M. Evans. Human and mouse adipose-derived cells support feeder-

independent induction of pluripotent stem cells. Proceedings of the National Academy of Sciences,

107(8):3558–3563, 2010.

K. Takahashi, K. Tanabe, M. Ohnuki, M. Narita, T. Ichisaka, K. Tomoda, and S. Yamanaka. Induc-

tion of Pluripotent Stem Cells from Adult Human Fibroblasts by Defined Factors. Cell, 131(5):

861–872, 2007.

J. A. Thomson, J. Itskovitz-Eldor, S. S. Shapiro, M. A. Waknitz, J. J. Swiergiel, V. S. Marshall, and

J. M. Jones. Embryonic Stem Cell Lines Derived from Human Blastocysts. Science, 282(5391):

1145–1147, 1998.

187 V. Verkarre, J. C. Fournet, P. de Lonlay, M. S. Gross-Morand, M. Devillers, J. Rahier, F. Brunelle,

J. J. Robert, C. Nihoul-Fekete, J. M. Saudubray, and C. Junien. Paternal mutation of the sul-

fonylurea receptor (SUR1) gene and maternal loss of 11p15 imprinted genes lead to persistent

hyperinsulinism in focal adenomatous hyperplasia. The Journal of Clinical Investigation, 102(7):

1286–1291, 1998.

Q. Wang, L. Ye, H. Liu, X. Liu, S. Li, and Z. Chen. Reprogramming of bone marrow-derived

mesenchymal stem cells into functional insulin-producing cells by chemical regimen. American

Journal of Stem Cells, 1(2):128–137, 2012.

L. M. Wilson, S. H. Wong, N. Yu, E. Geras-Raaka, B. M. Raaka, and M. C. Gershengorn. Insulin

but Not Glucagon Gene is Silenced in Human Pancreas-Derived Mesenchymal Stem Cells. Stem

Cells, 27(11):2703–2711, 2009.

M. E. Wilson, D. Scheel, and M. S. German. Gene expression cascades in pancreatic development.

Mechanisms of Development, 120(1):65–80, 2003.

H. Yasuda, K. Inoue, H. Shibata, T. Takeuchi, Y. Eto, Y. Hasegawa, N. Sekine, Y. Totsuka, T. Mine,

and E. Ogata. Existence of activin-A in A- and D-cells of rat pancreatic islet. Endocrinology, 133

(2):624–630, 1993.

H. Yuan, J. Li, N. Xin, Z. Zhao, and G. Qin. Expression of Pdx1 mediates differentiation from

mesenchymal stem cells into insulin-producing cells. Molecular Biology Reports, 37(8):4023–4031,

2010.

C. Zanini, S. Bruno, G. Mandili, D. Baci, F. Cerutti, G. Cenacchi, L. Izzi, G. Camussi, and M. Forni.

Differentiation of Mesenchymal Stem Cells Derived from Pancreatic Islets and Bone Marrow into

Islet-Like Cell Phenotype. PLoS ONE, 6(12):e28175, 2011.

188 Chapter 5

Conclusions

In this thesis, I have described the derivation, characterisation and attempted differentiation of novel stem cell lines, from patients with congenital hyperinsulinism (CHI). Whilst both mesenchymal stem cells (MSCs) and induced pluripotent stem cells (iPSCs) have previously been derived from pancre- atic tissue (Gallo et al., 2007, Bar-Nur et al., 2011), they have not been reported from CHI tissue. I therefore needed to characterise the CHI-derived stem cells and assess their capacity to be used for in vitro research for either diabetes or CHI.

5.1 CHI pMSCs were derived and have a pancreatic pheno-

type

MSCs were derived from CHI tissue, from three patients, one with each form of the disease (focal, diffuse and atypical). The MSCs were derived by plastic adherence. Deriving cells from CHI tissue by plastic adherence has been previously reported (MacFarlane et al., 1999), but the cell line - called

NES2Y - was not an MSC line. They are described as storing insulin and having a β-cell like (i.e. epithelial) phenotype. Having worked with cells identified as NES2Y within our lab (no data is shown), we found a 92% match by STR profiling to a carcinoma line A549. Further the original paper in which they are described (MacFarlane et al., 1999) mentions that they were aneupoid. This may explain some discrepancies including why NES2Y was not described originally as MSC if it were cross-contaminated or fused early on - the capacity for NES2Y to undergo cell-cell fusion has been demonstrated (Adams et al., 2009). Conversely, all cell lines I derived showed unique STR profiles.

The lines were karyotypically stable and for the two patients with known mutations, the mutations were also detected in the cell lines. The cell lines were also confirmed as MSCs by multiple methods, shown in 2.4.1 (Dominici et al., 2006).

189 As MSCs have not been derived from CHI tissue before, I wanted to assess whether the cells maintained characteristics of CHI pancreas. It has been previously mentioned that pancreatic MSCs

(pMSCs) express ISL1 (Eberhardt et al., 2006), a pancreatic gene (Ahlgren et al., 1997). I found that

CHI pMSCs express ISL1 as expected, along with other pancreas associated markers: SOX9, PAX6,

MAFB and CK19. It was not clear if any of these markers were specific to pMSCs. All of these genes were expressed in bone marrow MSCs (bmMSCs) as well, except for ISL1, suggesting this is a unique marker for MSCs from the pancreas as opposed to other bodily locations. Other pancreatic genes such as PDX1 were not expressed. Altered histone acetylation regulating the pancreatic genes

INS and GCG has been found in pMSCs and not bmMSCs (Wilson et al., 2009). Similarly, I found the CHI pMSCS showed reduced methylation upstream of PDX1, AMY2A and CK19. Reduced methylation of upstream areas of genes can be associated with higher transcription of the gene, or easier induction of gene expression (Lim and Maher, 2010). We could not conclude that the reduced methylation would affect gene expression. Inducing PDX1 expression in either the CHI pMSCs or bmMSCs would allow the efficiency of induction to be measured in both to confirm any effect of dif- ferential methylation. This could be done either by quantifying gene expression or transfecting the cells to produce luciferase under the control of the PDX1 promoter. After confirming if hypomethy- lation of PDX1 affects gene expression, methylation of other pancreatic gene promoters, including

NEUROD1, GCG and ABCC8, should also be assessed. We attempted to assess both NEUROD1 and GCG, but the primers were unsuitable for direct bisulfite sequencing. Further primer sets or clonal sequencing should be used for these going forward.

5.2 CHI pMSCs were differentiated to immature β-like cells

In order to use CHI pMSCs for in vitro research, either for CHI or diabetes, they need to be differenti- ated to functional β-cells. Based on expression of ISL1 and hypomethylation of PDX1 I hypothesised that pMSCs may differentiate to β-cells more efficiently than non-pancreatic MSCs. Initial papers about pMSCs reported that serum withdrawal induced formation of hormone positive cell clusters

(Dalvi et al., 2009, Davani et al., 2009, Carlotti et al., 2010). We found no evidence of this occurring in CHI pMSCs, it is unclear why, although may be related to passage number. There are numerous protocols for differentiating bmMSCs to β-cells, which also did not work as documented when ap- plied to the CHI pMSCs. A protocol reported by Zanini et al. (2011) to produce insulin-secreting cells, showing superior differentiation of pMSCs than bmMSCs, yielded promising results with CHI pMSCs. Initial screening of multiple protocols showed the Zanini protocol produced cell clusters,

190 which were positive for insulin gene, protein and stained positive with the zinc chelator dithizone; controls of the protocol were not positive for these.

Based on its use in another protocol (Li et al., 2012), and the reported involvement in mesenchymal- epithelial transition (MET) during development (Larsen et al., 2015), I assessed the addition of the

Wnt inhibitor DKK1 to differentiation of the CHI pMSCs. I found DKK1 markedly enhanced differ- entiation of CHI pMSCs by increasing expression of INS and PDX1 genes in the final cells. DKK1, when combined with retinoic acid and activin A in the initial stages of the protocol, induced NEU-

ROD1. No other pancreatic genes were induced, although TUB3 was, so DKK1 may have been functioning to activate neuronal pathways (Cajanek et al., 2009). As this has not been investigated previously, further work to understand the function of DKK1 on pMSCs would be helpful. NEU-

ROD1 was not present in the final cells, but should be expressed in β-cells. Therefore, prolonged or later use of DKK1, or another Wnt inhibitor such as niclosamide (Osada et al., 2011), may be able to induce NEUROD1 in the final β-like cells. We did not assess DKK1 at any other points in this protocol due to the high cost.

Using an adaptation of the Zanini protocol, with the addition of DKK1, INS and PDX1 positive cells were produced. Initial results suggested that the resulting cells were able to secrete insulin, but were not glucose sensitive and did not express ATP-sensitive potassium (KATP) channels. This therefore limits the use of CHI pMSCs for modelling either diabetes or CHI. As the cells resembled immature β-cells, further maturation may be possible in order to make the cells functional for drug testing. To assess whether the cells could be matured, they were transplanted into nude mice by a collaborator. Transplantations were performed using single undifferentiated cells, control cell clus- ters, and clusters differentiated according to my protocol. The single cells were not detectable in the kidney capsule 12 weeks after transplantation. Transplanted clusters, both differentiated and control, were detected after 12 weeks, but did not clearly express hormones. Testing the blood glucose of the mice for human C-peptide suggested that there may be low levels present, but the results were inconclusive due to high background in the untransplanted controls. We were therefore unable to ascertain if CHI pMSCs can be fully matured.

191 5.3 A CHI iPSC line could be differentiated to pancreatic

cells

Given the inconclusive results from differentiating MSCs, and recent advancements in the pluripotent stem cell field (Rezania et al., 2014, Pagliuca et al., 2014), I also wanted to assess iPSCs from CHI patients. As with MSCs, iPSCs have not been reported from CHI patients. I derived an iPSC line from the pMSCs, from the focal CHI patient. As the MSCs were heterozygous for the CHI causing mutation, any differentiated cells should secrete insulin normally in response to glucose. This ini- tially allows assessment and optimisation of differentiation protocols. In order to use the cells for

CHI research, the healthy allele can be mutated - as a point mutation to simulate diffuse CHI (Glaser et al., 2000), or megabase deletion to simulate focal CHI (Giurgea et al., 2006). This can be done using the CRISPR/Cas9 system (Sander and Joung, 2014); guide RNAs and transfection protocols have been designed in order to engineer the mutation to be homozygous.

The derived cells were a clonal pluripotent line, able to differentiate to all three germ layers. It was unclear if there was any advantage to deriving them from pMSCs. The reduced methylation seen in pancreatic genes in the pMSCs was not maintained after reprogramming. Sequencing of PDX1, arguably the most important gene as discussed in 1.2.4, failed and so methylation was not measured.

We did not assess histone acetylation, the other level of epigenetic regulation. If further iPSCs were to be derived from CHI patients, this would be important to confirm as blood cells are more readily available.

Differentiation of iPSCs through early stages of pancreatic development was very efficient and achievable. Yield of definitive endoderm was over 90% of cells by two protocols. Similarly, both protocols allowed production of PDX1 positive cells. Following the protocol from Rezania et al.

(2014) to completion yielded cells which were positive for glucagon and ABCC8, but were not clearly positive for insulin (although results were mixed). Optimising this protocol was not possible due to time limitations, but it should feasibly be possible to obtain insulin-secreting cells as was reported in the paper. Most likely, this would be by using the air-liquid interface protocol described, if the technical challenges of this culture method can be overcome.

Deriving β-cells from CHI stem cells has so far proved challenging, but shown some progress.

From the pMSCs the resulting cells most resembled immature β-cell, from the iPSCs they were more

α-like, or possibly poly-hormonal. The protocols used for the two stem cell types were very different, based as they were, on published protocols. The growth factors and small molecules used targeted

192 different pathways, and so the only similarity was the 3D culture as islet-like spheroids. It is possible that we could learn from both systems. IPSC differentiation does not include an MET, resembling de-lamination of islets, as MSCs may. On the other hand, PDX1 is induced to a much higher degree in iPSCs. Further, as Rezania et al. (2014) use the air-liquid interface for optimal differentiation of iPSCs, this may also offer a way to mature MSC-derived β-cells. Until mature insulin-secreting cells can be developed from either stem cell type, we cannot use stem cells from CHI for an in vitro pancreas model.

5.4 Increased proliferation in CHI

Aside from the increased insulin secretion, the CHI pancreas also exhibits increased proliferation in all three forms of the disease as shown in 3.4.1 and Salisbury et al. (2015). This is an aspect of the disease which cannot be modelled effectively in rodents (Fiaschi-Taesch et al., 2009, Kulkarni et al., 2012). We also could not use iPSCs for modelling this, as they were transduced using c-Myc, but the cell cycle in MSCs has not been exogenously altered. Interestingly, the CHI pMSCs had increased rates of proliferation, compared to adult controls. Whilst I could not exclude that this was age-related, the highest proliferation was seen in the eldest patient. This patient was 18 months old, in a healthy individual proliferation would have essentially stopped by this age (<0.5% proliferative

β cells) (Meier et al., 2008), making it more likely to be disease than age related.

Based on the innately increased proliferation, I assessed cell-cycle progression and regulation in the more proliferative CHI pMSCs compared to healthy adult MSCs. I found an inverse correlation of nuclear p27Kip1 and proliferation, which was further altered by the insulin/glucose levels seen in

CHI. The cells showed reduced nuclear p27Kip1 with increased proliferation, suggesting a canonical

CKI role (Sherr and Roberts, 1999). CHI tissue, on the other hand, shows increased nuclear p27Kip1

(Salisbury et al., 2015), although not in Ki67 positive proliferative cells (Salisbury, 2014) where the role is therefore less clear. There was therefore some evidence that CHI pMSCs could be used to model increased proliferation in CHI but there were also differences. I hypothesised that the differ- ences seen in nuclear p27Kip1 levels, may be due to the sustained high insulin levels in the CHI tissue.

This could be causing the pancreas cells to increase CKI expression in an attempt to compensate the proliferation rates and prevent tumour formation. Longer experiments using sustained high insulin in culture may reveal if this is true. I also attempted to use the MSCs to understand if other cell cycle regulators could play a role in the aetiology of CHI. Assessing G2/M-phase regulators in the CHI cells showed increased CDK1, although very stable levels of the CDK1 binding partner, CyclinB1.

193 I therefore propose a role for CDK1 in CHI. Observing CDK1 in archived fixed tissue would allow confirmation if CDK1 has a role in CHI, and if CHI pMSCs are a suitable in vitro model for increased proliferation and altered cell cycle in CHI.

5.5 Conclusion

In conclusion, I have described the first derivation of MSCs and an iPSC line from patients with CHI.

We hypothesised that they could be used as an in vitro model for CHI; high rates of insulin secretion and increased proliferation may also mean CHI stem cells could be an easier to use model for testing diabetes drugs. Based on results of testing these hypotheses, I suggest that CHI pMSCs may be a good model for altered cell cycling in CHI, a process which is not well understood. However, neither cell type is a good model for β-cells in CHI or diabetes. Whilst progress was made to differentiate the cells to β-cells, no functional β-cells were produced. The MSC-derived cells had insulin but not ABCC8 ; iPSC-derived cells had glucagon and ABCC8, but not INS. Combining aspects of differentiation from each cell type, together with culture in hydrogel or using the air-liquid interface, as discussed above, may enable the cells to be differentiated to functional β-cells. With further work to produce functional β-cells, the derived MSCs and iPSCs may become a useful model for CHI or diabetes, and so further work is needed.

5.6 Future work

From the studies presented here, the most promising results for developing β-cells from CHI patient stem cells were seen from iPSCs. I therefore feel that going forward it would be most sensible to concentrate on iPSCs rather than MSCs. However, it would be beneficial to conclude the work regarding DKK1, to investigate how it has enhanced differentiation for example by a comparative microarray to see which signalling pathways and transcription factors are being activated. Alongside this, experiments into using DKK1 for longer within the differentiation protocol may demonstrate that MSC differentiation to β-cells is more possible than we observed, so this should be tested before fully ruling out the use of MSCs for differentiation to a β-cell model.

The data regarding any tissue specific methylation or bias in iPSCs was inconclusive, more patient samples along with relevant control iPSCs (healthy non pancreas derived, from adults if age-matched is not available) would be needed to confirm if there is any advantage to deriving iPSCs from CHI pancreatic tissue. If not, future iPSC lines could be created from blood samples taken during di- agnostics, allowing access to greater numbers of both patient and matched control samples. Some

194 drugs involved in the treatment of CHI such as rapamycin, as mentioned previously, can affect cell cycle. Reprogramming further iPSCs without the use of c-Myc would allow the maximum amount of data to be gathered regarding the mechanism of drugs being tested.

Further optimisation of the differentiation of iPSCs to β-cells is also needed before they can be used for assays. Whilst differentiation to pancreatic progenitors (PDX1 positive) was efficient, the later stages showed a poor yield of insulin positive cells, with no insulin secretion. Comparison of total PDX1 protein levels by western blot between the two protocols would confirm which was the best protocol to use up to that stage, as there was a significant difference between the mRNA ex- pression from each. Culture of the cells on trans-well inserts at the air-liquid interface, as was done in the ‘Kieffer’ paper may help to improve the efficiency of the final stages. Extended time in culture may also enhance the proportion of β-cells present and would likely increase their maturity, the time points selected in this thesis were from the early end of the time bracket given in the paper. Once the technical barriers to obtaining insulin-secreting cells from the CHI iPSCs have been negated, the usefulness of the cells for modelling CHI and assaying drugs can be investigated. This would minimally involve checking the effects of known drugs such as octreotide on insulin secretion, calcium

flux and viability. Additional information such as the effects on other proteins, signalling pathways, and proliferation could then be obtained which could be used to inform further drug discovery. With the relative ease of manipulating iPSCs now, a reporter line could be generated from this model with, for example, GFP tagged insulin, for the use in larger scale screening if early studies showed the cells to be a useful model for CHI.

To follow up on the finding that MSCs from CHI patients show hyperproliferation, similar to hyperproliferation in CHI tissue, further samples would be beneficial. Age-matched controls would be ideal, to control for the fact that tissue from young patients may be expected to be more prolifer- ative than adult pancreas tissue. Further, additional MSC lines from all forms of CHI would clarify whether the variation seen, particularly in proliferation rate and cell cycle distribution is due to the differing disease state, or is due to natural variability between people. Finally, at least one additional adult control line is needed to explore the differences seen in CDK1 expression, to understand if there is a difference between the CHI and adult samples, and if the one adult sample which was different to the other two is an outlier. Extending the glucose and insulin experiments for a longer period of time may allow us to match the cell and tissue p27Kip1 data more closely. Alongside this p27Kip1 phosphorylation status would be indicative of AKT activity, which would give a more detailed molec- ular explanation for the proliferation seen in CHI. These findings should be corroborated with fixed archived tissue where possible, particularly for CDK1 until the cell line model has been fully validated.

195 196 5.7 References

G. G. Adams, A. Uddin, M. Vives-Pi, and R. Pujol-Borrell. Characterisation of the NES2Y cell line

and its use in the production of human glucose-responsive insulin producing (hGRIP) cell lines by

cell-cell fusion. Islets, 1(2):117–123, 2009.

U. Ahlgren, S. L. Pfaff, T. M. Jessell, T. Edlund, and H. Edlund. Independent requirement for ISL1

in formation of pancreatic mesenchyme and islet cells. Nature, 385(6613):257–260, 1997.

O. Bar-Nur, H. Russ, S. Efrat, and N. Benvenisty. Epigenetic Memory and Preferential Lineage-

Specific Differentiation in Induced Pluripotent Stem Cells Derived from Human Pancreatic Islet

Beta Cells. Cell Stem Cell, 9(1):17–23, 2011.

L. Cajanek, D. Ribeiro, I. Liste, C. L. Parish, V. Bryja, and E. Arenas. Wnt/β-Catenin Signal-

ing Blockade Promotes Neuronal Induction and Dopaminergic Differentiation in Embryonic Stem

Cells. STEM CELLS, 27(12):2917–2927, 2009.

F. Carlotti, A. Zaldumbide, C. J. Loomans, E. van Rossenberg, M. Engelse, E. J. de Koning, and

R. C. Hoeben. Isolated human islets contain a distinct population of mesenchymal stem cells.

Islets, 2(3):164–173, 2010.

M. P. Dalvi, M. R. Umrani, M. V. Joglekar, and A. A. Hardikar. Human pancreatic islet progenitor

cells demonstrate phenotypic plasticity in vitro. Journal of Biosciences, 34(4):523–528, 2009.

B. Davani, S. Ariely, L. Ikonomou, Y. Oron, and M. C. Gershengorn. Human islet-derived precursor

cells can cycle between epithelial clusters and mesenchymal phenotypes. Journal of Cellular and

Molecular Medicine, 13:2570–2581, 2009.

M. Dominici, K. Le Blanc, I. Mueller, I. Slaper-Cortenbach, F. Marini, D. Krause, R. Deans, A. Keat-

ing, D. Prockop, and E. Horwitz. Minimal criteria for defining multipotent mesenchymal stromal

cells. The International Society for Cellular Therapy position statement. Cytotherapy, 8(4):315–7,

2006.

M. Eberhardt, P. Salmon, M.-A. von Mach, J. G. Hengstler, M. Brulport, P. Linscheid, D. Seboek,

J. Oberholzer, A. Barbero, I. Martin, B. Muller, D. Trono, and H. Zulewski. Multipotential nestin

and Isl-1 positive mesenchymal stem cells isolated from human pancreatic islets. Biochemical and

Biophysical Research Communications, 345(3):1167–1176, 2006.

N. Fiaschi-Taesch, T. A. Bigatel, B. Sicari, K. K. Takane, F. Salim, S. Velazquez-Garcia, G. Harb,

K. Selk, I. Cozar-Castellano, and A. F. Stewart. Survey of the Human Pancreatic β-Cell G1/S

Proteome Reveals a Potential Therapeutic Role for Cdk-6 and Cyclin D1 in Enhancing Human

β-Cell Replication and Function In Vivo. Diabetes, 58(4):882–893, 2009.

197 R. Gallo, F. Gambelli, B. Gava, F. Sasdelli, V. Tellone, M. Masini, P. Marchetti, F. Dotta, and

V. Sorrentino. Generation and expansion of multipotent mesenchymal progenitor cells from cul-

tured human pancreatic islets. Cell Death Differ, 14(11):1860–1871, 2007.

I. Giurgea, C. Bellanna-Chantelot, M. Ribeiro, L. Hubert, C. Sempoux, J.-J. Robert, O. Blankenstein,

K. Hussain, F. Brunelle, C. Nihoul-Fekete, J. Rahier, F. Jaubert, and P. de Lonlay. Molecular

Mechanisms of Neonatal Hyperinsulinism. Horm Res Paediatr, 66(6):289–296, 2006.

B. Glaser, P. Thornton, T. Otonkoski, and C. Junien. Genetics of neonatal hyperinsulinism. Archives

of Disease in Childhood - Fetal and Neonatal Edition, 82(2):F79–F86, 2000.

R. N. Kulkarni, E.-B. Mizrachi, A. G. Ocana, and A. F. Stewart. Human β-Cell Proliferation and

Intracellular Signaling: Driving in the Dark Without a Road Map. Diabetes, 61(9):2205–2213,

2012.

B. M. Larsen, S. M. Hrycaj, M. Newman, Y. Li, and D. M. Wellik. Mesenchymal Hox6 function

is required for mouse pancreatic endocrine cell differentiation. Development, 142(22):3859–3868,

2015.

J. Li, L. Zhu, X. Qu, J. Li, R. Lin, L. Liao, J. Wang, S. Wang, Q. Xu, and R. C. Zhao. Stepwise

Differentiation of Human Adipose-Derived Mesenchymal Stem Cells Toward Definitive Endoderm

and Pancreatic Progenitor Cells by Mimicking Pancreatic Development In Vivo. Stem Cells and

Development, 22(10):1576–1587, 2012.

D. H. K. Lim and E. R. Maher. Dna methylation: a form of epigenetic control of gene expression.

The Obstetrician & Gynaecologist, 12(1):37–42, 2010.

W. M. MacFarlane, J. C. Chapman, R. M. Shepherd, M. N. Hashmi, N. Kamimura, K. E. Cosgrove,

R. E. O’Brien, P. D. Barnes, A. W. Hart, H. M. Docherty, K. J. Lindley, A. Aynsley-Green, R. F. L.

James, K. Docherty, and M. J. Dunne. Engineering a Glucose-responsive Human Insulin-secreting

Cell Line from Islets of Langerhans Isolated from a Patient with Persistent Hyperinsulinemic

Hypoglycemia of Infancy. Journal of Biological Chemistry, 274(48):34059–34066, 1999.

J. J. Meier, A. E. Butler, Y. Saisho, T. Monchamp, R. Galasso, A. Bhushan, R. A. Rizza, and

P. C. Butler. β-Cell Replication Is the Primary Mechanism Subserving the Postnatal Expansion

of β-Cell Mass in Humans. Diabetes, 57(6):1584–1594, 2008.

T. Osada, M. Chen, X. Y. Yang, I. Spasojevic, J. B. Vandeusen, D. Hsu, B. M. Clary, T. M. Clay,

W. Chen, M. A. Morse, and H. K. Lyerly. Antihelminth Compound Niclosamide Downregulates

Wnt Signaling and Elicits Antitumor Responses in Tumors with Activating APC Mutations. Can-

cer Research, 71(12):4172–4182, 2011.

198 F. Pagliuca, J. Millman, M. Gurtler, M. Segel, A. VanDervort, J. Ryu, Q. Peterson, D. Greiner, and

D. Melton. Generation of Functional Human Pancreatic β Cells In Vitro. Cell, 159(2):428–439,

2014.

A. Rezania, J. E. Bruin, P. Arora, A. Rubin, I. Batushansky, A. Asadi, S. O’Dwyer, N. Quiskamp,

M. Mojibian, T. Albrecht, Y. H. C. Yang, J. D. Johnson, and T. J. Kieffer. Reversal of diabetes

with insulin-producing cells derived in vitro from human pluripotent stem cells. Nat Biotech, 32

(11):1121–1133, 2014.

R. J. Salisbury. Gene expression and cell cycle regulation in human pancreas development and

congenital hyperinsulinism. PhD thesis, University of Manchester, School of Medicine, 2014.

R. J. Salisbury, B. Han, R. E. Jennings, A. A. Berry, A. Stevens, Z. Mohamed, S. A. Sugden,

R. De Krijger, S. E. Cross, P. P. Johnson, M. Newbould, K. E. Cosgrove, K. P. Hanley, I. Banerjee,

M. J. Dunne, and N. A. Hanley. Altered Phenotype of β-Cells and Other Pancreatic Cell Lineages

in Patients With Diffuse Congenital Hyperinsulinism in Infancy Caused by Mutations in the ATP-

Sensitive K-Channel. Diabetes, 64(9):3182–3188, 2015.

J. D. Sander and J. K. Joung. CRISPR-Cas systems for editing, regulating and targeting genomes.

Nat Biotech, 32(4):347–355, 2014.

C. J. Sherr and J. M. Roberts. CDK inhibitors: positive and negative regulators of G1-phase

progression. Genes & Development, 13(12):1501–1512, 1999.

L. M. Wilson, S. H. Wong, N. Yu, E. Geras-Raaka, B. M. Raaka, and M. C. Gershengorn. Insulin

but Not Glucagon Gene is Silenced in Human Pancreas-Derived Mesenchymal Stem Cells. Stem

Cells, 27(11):2703–2711, 2009.

C. Zanini, S. Bruno, G. Mandili, D. Baci, F. Cerutti, G. Cenacchi, L. Izzi, G. Camussi, and M. Forni.

Differentiation of Mesenchymal Stem Cells Derived from Pancreatic Islets and Bone Marrow into

Islet-Like Cell Phenotype. PLoS ONE, 6(12):e28175, 2011.

199