152
1 PDX1 PROTEIN THERAPY: A NOVEL APPROACH FOR TREATMENT OF DIABETES By VIJAY KOYA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF FLORIDA IN PARTIAL FULFILLMENT OF THE REQUIREMENTS FOR THE DEGREE OF DOCTOR OF PHILOSOPHY UNIVERSITY OF FLORIDA 2009

© 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

  • Upload
    others

  • View
    6

  • Download
    0

Embed Size (px)

Citation preview

Page 1: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

1

PDX1 PROTEIN THERAPY: A NOVEL APPROACH FOR TREATMENT OF DIABETES

By

VIJAY KOYA

A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL

OF THE UNIVERSITY OF FLORIDA IN PARTIAL FULFILLMENT OF THE REQUIREMENTS FOR THE DEGREE OF

DOCTOR OF PHILOSOPHY

UNIVERSITY OF FLORIDA

2009

Page 2: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

2

© 2009 Vijay Koya

Page 3: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

3

To my brother and parents, for their support and appreciation for higher education, and to my wife for her support all through my graduate school.

Page 4: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

4

ACKNOWLEDGMENTS

I have spent last 4 years at the University of Florida working towards my doctoral degree.

In the process I had opportunity to learn, explore, and teach. I have had unique experiences that

helped me to develop as an individual and helped me plan my career for the future. There have

many people who have influenced my thought process, my behavior and my interaction. During

this 4 year long journey, I have been helped by many people to successfully reach my research

goals.

Firstly, I would like to thank my mentor, Dr. Lijun Yang for giving me an opportunity to

conduct research in her lab. She has been exceptionally helpful in achieving my research goals.

She has supported me in every possible way to have an enjoyable experience in her lab. Her

unwavering support has helped me to achieve my goal to obtain admission into residency

program. Her enthusiasm and perseverance has motivated me to work extra hard.

Next, I would like to thank my supervisory committee Dr. Daniell Purich, Dr. Lung-ji

Chang, Dr. Chen Liu, and Dr. Sally litherland. Their insight has helped me to accelerate my

research progress and made sure I am progressing in right direction. I should thank Dr. Purich for

his personal advice. Conversation with him has been inspiring and motivates me to achieve

greater heights in science. I should thank Dr. Lung-ji Chang for providing with reagents

necessary for my experiments.

Next, I would like to thank Dr. Shiwu Li (expert in Molecular biology) for his constant

support technically and intellectually. Working with him has been a memorable experience. He

has been a constant backbone for my research achievements. He has optimized to generate high

quality Pdx1 protein, the most valuable reagent that shaped my research projects. He is very

approachable person, has a great knowledge in various fields of biomedical sciences, technically

sound, and has a great sense of humor. He has been a constant source to clarify my doubts.

Page 5: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

5

I would like to thank Dr. Shun Lu for his help with the Balb/C mice project. I would like to thank

Dr. Chang Qing and Dr. Dongqi Tang for teaching me techniques related to animal surgery and

dissection. I would like to thank my lab colleagues Cathy and Hong Fong for their technical

assistance with the experiments. I would like to thank William Donlean for his assistance with

lab ordering, lab maintenance and equipment handling. He is a great person to work and having

him around is always fun.

I should thank Michael lagoe and Adam Kenney, undergraduates in the lab who have

been a helping hand in conducting some of my experiments. Mike is a fun guy to work with. He

has shown extreme level of dedication towards the experiments that I have assigned to him. I

would like to thank Fred Hutchinson for his help in the mouse colony. He has been exceptionally

helpful in maintaining my mouse colonies. I would like to thank Sushama (my wife), Dr. Satish

Medisetty, Suntiha, Karthik, Rushi and Will for proof-reading my dissertation. I would like to

thank all my friends and colleagues in the PhD program for their support and encouragement

through all these years. I would like to thank my family for their constant support and

encouragement all these years. I would like to thank my wife for her exceptional support with

both academic and no-academic affairs. I would like to thank the IDP and MCB program

directors for making it an enjoyable learning experience.

.

Page 6: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

6

TABLE OF CONTENTS page

ACKNOWLEDGMENTS ...............................................................................................................4

LIST OF TABLES .........................................................................................................................10

LIST OF FIGURES .......................................................................................................................11

LIST OF ABBREVIATIONS ........................................................................................................14

ABSTRACT ...................................................................................................................................16

CHAPTER

1 INTRODUCTION ..................................................................................................................18

Diabetes ..................................................................................................................................18 Type 1 Diabetes ...............................................................................................................18 Type 2 Diabetes ...............................................................................................................19 Current Treatment Options ..............................................................................................20 Novel Approaches for Treatment of Diabetes .................................................................21

Pancreatic Duodenal Homeobox 1 (Pdx1) .............................................................................21 Function of Pdx1 in Pancreatic Beta Cells ......................................................................22 Protein Transduction Domain (PTD) ..............................................................................23 Beta Cell Regeneration ....................................................................................................24 Liver Cell Transdifferentiation into insulin-producing cells (IPCs) ...............................24

Significance ............................................................................................................................26

2 GENERAL METHODS .........................................................................................................27

Recombinant DNA Technology .............................................................................................27 Plasmid Isolation .............................................................................................................27 Preparation of LB Broth ..................................................................................................27 Agar Gel Electrophoresis ................................................................................................27 Polymerase Chain Reaction .............................................................................................27 Restriction Digestion .......................................................................................................28 DNA Ligation ..................................................................................................................28 Transformation of Competent E.coli Cells ......................................................................28

Protein Expression, Purification and Analysis .......................................................................29 Protein Expression in Bacteria ........................................................................................29 Protein Purification ..........................................................................................................29 Dialysis of the Purified Protein .......................................................................................30 Sodium Dodecylsulphate - Polyacrylamide Gel Electrophoresis (SDS-PAGE) .............31 Western Blotting ..............................................................................................................31

Mammalian Cell Culture Protocols ........................................................................................32

Page 7: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

7

Cell Culture .....................................................................................................................32 Transfection of Plasmid in Mammalian Cells .................................................................32 RNA Isolation from Cells/Tissue ....................................................................................33 cDNA Synthesis ..............................................................................................................33 Measurement of Luciferase Activity ...............................................................................34

Extraction and Measurement of Insulin ..................................................................................34 Serum Sample Collection for Measurement of Insulin by ELISA ..................................34 Tissue Sample Collection for Measurement of Insulin by ELISA ..................................34 ELISA for Measurement of Insulin .................................................................................35

Procedures for Imaging ...........................................................................................................35 Haematoxylin and Eosin (H &E) Staining ......................................................................35 Immunofluroscence .........................................................................................................36

General Animal Procedures ....................................................................................................36 Animal Housing ...............................................................................................................36 Collection of Blood .........................................................................................................37 Tissue Sample Collection for Histology ..........................................................................37 Measurement of Blood Glucose ......................................................................................37 Tissue Sample Collection for RNA Isolation ..................................................................37 Anesthesia ........................................................................................................................38 Pancreatectomy ................................................................................................................38 Splenectomy ....................................................................................................................38

3 REVERSAL OF STREPTOZOTOCIN-INDUCED DIABETES IN MICE BY CELLULAR TRANSDUCTION WITH RECOMBINANT PANCREATIC TRANSCRIPTION FACTOR PANCREATIC DUODENAL HOMEOBOX-1 ...................40

Introduction .............................................................................................................................40 Materials and Methods ...........................................................................................................41

Construction and Production of rPdx1, PTD–Green Fluorescent Protein, and Mutant Pdx1 Fusion Proteins .......................................................................................41

Cell Entry and Immunoblotting .......................................................................................42 rPdx1 Functional Analyses Using NeuroD-Luciferase Reporter Construct ....................43 Animal Studies ................................................................................................................43 Low-Dose Streptozotocin-Induced Diabetes ...................................................................43 Intraperitoneal Glucose Tolerance Test ...........................................................................44 Pdx1 Protein in vivo Kinetics and Tissue Distribution ....................................................44 RT-PCR ...........................................................................................................................44 Quantitative Real-Time RT-PCR Analysis .....................................................................45 Immunohistochemistry and Immunofluorescence ..........................................................46 Tissue and Serum Insulin Measurements by Enzyme-Linked Immunosorbent Assay ...47 Statistical Analysis ..........................................................................................................47

Results .....................................................................................................................................47 Generation of Recombinant Fusion Proteins ...................................................................47 Characterization of Recombinant Fusion Proteins ..........................................................48 In vivo Kinetics and Tissue Distribution .........................................................................49 In vivo Effects of rPdx1 on Blood Glucose Levels in Diabetic Mice .............................50

Page 8: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

8

Pdx1 Treatment Promoting Endogenous beta cell Regeneration ....................................51 Pdx1 Treatment Promoting Liver Cell Transdifferentiation into IPCs ...........................52 Relationship between Pancreas and Liver at Tissue Insulin Levels ................................53

Discussion ...............................................................................................................................54

4 PDX1 PROTEIN THERAPY PREVENTS THE ONSET OF TYPE 1 DIABETES IN NOD MICE .............................................................................................................................79

Introduction .............................................................................................................................79 Materials and Methods ...........................................................................................................80

Animal Studies ................................................................................................................80 Intraperitoneal Glucose Tolerance Test (IPGTT) ............................................................80 Serum Insulin Measurements ..........................................................................................81 Construction of Truncated Pdx1 Proteins ........................................................................81 Extraction of Splenocytes from the Spleen .....................................................................82 Lymphocyte Proliferation Assay .....................................................................................82 BDC 2.5 Mice Splenocyte Transfer ................................................................................83

Results .....................................................................................................................................83 Administration of rPdx1 to NOD Mice Prevents the Onset of Diabetes .........................83 Beta cell Functional Analysis and Determination of Tissue Insulin Levels ...................84 Pdx1 Treatment Preserves Islet Mass Possibly by Beta cell regeneration and

Promotes Liver Cell Transdifferentiation into Insulin-Producing Cells. .....................84 Detection of Pdx1 Antibodies in Serum Samples of rPdx1/Saline-treated Mice ............85 Determination of Antigenic Epitope of Pdx1 Protein .....................................................85 PAA emerge before the onset of diabetes. ......................................................................86 Demonstration of Autoreactive Lymphocytes Against Pdx1 Protein in NOD Mice ......87 Decreased Proliferation of Donor Derived BDC2.5 CD4+ Lymphocytes in

Pancreatic Lymph Node ...............................................................................................87 Effect of Mutant-rPdx1 On the Onset of Diabetes in NOD mice ....................................88

Discussion ...............................................................................................................................88

5 MOLECULAR MECHANISM OF THE ROLE OF PDX1 IN BETA CELL PROLIFERATION ...............................................................................................................112

Introduction ...........................................................................................................................112 Materials and Methods .........................................................................................................113

Cell Culture ...................................................................................................................113 Design of siRNA for the Pdx1 Transcript .....................................................................113 siRNA Mediated Knockdown of Pdx1 Transcript in INS-1 Cells ................................113 Western Blotting with Anti-Pdx1 and Anti-tubulin Antibody ......................................114 BrdU Proliferation Assay ..............................................................................................114 Real Time PCR ..............................................................................................................115 Construction of pGL4.10-Cyclin B1 Promoter-Luciferase Vector ...............................116 Analysis of Promoter Constructs for Activation by Pdx1 or Nkx6.1 using

Luciferase Reporter Assays .......................................................................................116 Results ...................................................................................................................................117

Page 9: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

9

Phenotype of INS-1 Cells Treated with Pdx1 siRNA ...................................................117 siRNA-mediated Knockdown of Pdx1 Transcripts in INS-1 Cells ...............................117 Decrease in Cell Proliferation of Pdx1 Knockdown Cells ............................................117 Analysis of the Gene Expression of Beta Cell Specific transcription factors and

Cell Cycle Genes........................................................................................................118 Analysis of Cyclin B1 Promoter using CMV-Pdx1 .......................................................118 Nkx6.1 Activation of Cyclin B1 Promoter ....................................................................118 Pdx1 Activation of Nkx6.1 Promoter ............................................................................119

Discussion .............................................................................................................................119

6 FUTURE STUDIES .............................................................................................................132

Reversal of Streptozotocin-induced Diabetes in Mice by Cellular Transduction with Recombinant Pancreatic Transcription Factor, Pancreatic Duodenal Homeobox-1 .........132

Future Studies ................................................................................................................132 Pdx1 Protein Therapy for the Treatment of Type 1 Diabetes in NOD Mice ........................134

Future Studies ................................................................................................................134 Molecular Mechanism of the Role of Pdx1 in Beta Cell Proliferation .................................135

Future Studies ................................................................................................................135

APPENDIX

A NOD MICE TRIAL 1 ...........................................................................................................136

B NOD MICE TRIAL 2 ...........................................................................................................137

REFERENCES ............................................................................................................................138

BIOGRAPHICAL SKETCH .......................................................................................................152

Page 10: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

10

LIST OF TABLES

Table page Table 3-1. Primer name, sequences, size, GenBank #, and PCR condition ...................................59

Table 3-2. Real time PCR Primer name, sequences, size, GenBank #, and PCR condition ..........60

Table 5-1. Real time PCR Primer name, sequences, size, GenBank #, and PCR condition ........122

Table A-1. Mice database of trial 1 .............................................................................................136

Table B-1. Mice database of trial 2 ..............................................................................................137

Page 11: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

11

LIST OF FIGURES

Figure page 3-1 Cloning, expression, purification, and characterization of rat Pdx1, PTD-GFP, and

rPdx1-mut fusion proteins. ...............................................................................................61

3-2 Time course of cell entry of rPdx1 and rPdx1-mut proteins. .............................................62

3-3 Functional analysis of rPdx1 protein. ................................................................................63

3-4 In vivo kinetics and tissue distribution of rPdx1 following intra-peritoneal injection. .....64

3-5 In vivo tissue distribution of rPdx1 following intraperitoneal injection. ...........................65

3-6 Experimental timeline for rPdx1 treatment........................................................................66

3-7 In vivo effects of rPdx1 protein on blood glucose levels. ..................................................67

3-8 Intraperitoneal glucose tolerance test. ................................................................................68

3-9 Comparison of average blood glucose and Insulin levels between rPdx1 treated and untreated control mice. .......................................................................................................69

3-10 Histology of pancreatic islets. ............................................................................................70

3-11 Immunostaining with anti-KI67 antibody and anti-insulin antibody. ................................71

3-12 Quantitative Realtime-PCR analyses of pancreatic tissue between rPdx1 treated and control mice. ......................................................................................................................72

3-13 Insulin immunohistochemical staining of liver tissue. ......................................................73

3-14 RT-PCR analysis of the expression of pancreatic genes in the liver.. ...............................74

3-15 Quantitative Real Time-PCR analyses of pancreatic gene expression in liver.. ................75

3-16 Expression of pancreatic genes in other organs.. ...............................................................76

3-17 Pancreatic tissue insulin measurements as determined by ELISA. ...................................77

3-18 Liver tissue insulin measurements as determined by ELISA. ...........................................78

4-1 Time line for the experimental plan for treatment of NOD mice. .....................................92

4-2 Effect of rPdx1 (12-weeks) on the onset of diabetes in NOD mice. ..................................93

4-3 Blood glucose levels of rPdx1 (12-weeks) treated NOD mice. .........................................94

Page 12: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

12

4-4 Body weights of rPdx1 (12-weeks) treated NOD mice. ....................................................95

4-5 Effect of short term rPdx1treatment (4-weeks) on the onset of diabetes in NOD mice. ...96

4-6 Blood glucose levels of rPdx1 (4-weeks) treated NOD mice. ...........................................97

4-7 Effect of short term rPdx1treatment (8-weeks) on the onset of diabetes in NOD mice. ...98

4-8 Blood glucose levels of rPdx1 (8-weeks) treated NOD mice. ...........................................99

4-9 Intraperitoneal glucose tolerance test. IPGTT (n=4/group). ...........................................100

4-10 Serum insulin measurements as determined by ELISA. ................................................101

4-11 Haematoxylin & Eosin staining and immunohistochemistry of pancreatic tissue sections. ............................................................................................................................102

4-12 Pancreas and liver tissue insulin levels as determined by ELISA. ................................103

4-13 Detection of Pdx1 antibodies by ELISA in the NOD mice. ............................................104

4-14 Confirmation of anti-Pdx1 antibody in the NOD mice by western blotting. ...................105

4-15 Determination of antigenic epitope by western blot. .......................................................106

4-16 Relationship between Pdx1 autoantibody levels and onset of diabetes. ..........................107

4-17 Antigen-stimulated lymphocyte proliferation. .................................................................108

4-18 Transfer of splenocytes from donor BDC2.5 mice to the recipient NOD mice. .............109

4-19 Illustration of Pdx1 and mutant Pdx1proteins. ..............................................................110

4-20 Effects of mutant-Pdx1 on the onset of diabetes. . .........................................................111

5-1 Location of 21 mer siRNAs on Pdx1 mRNA transcript. .................................................123

5-2 Phenotype of INS1 cells treated with Pdx1 siRNA.. .......................................................124

5-3 Confirmation of Pdx1 knockdown using western blotting. .............................................125

5-4 Measurement of INS-1 cell proliferation using BrdU incorporation. ..............................126

5-5 Analysis of relative change in the gene expression. ........................................................127

5-6 Analysis of cyclin B1 promoter using CMV-Pdx1. .........................................................128

5-7 Nkx6.1 activation of Cyclin B1promoter .........................................................................129

5-8 Pdx1 activation of Nkx6.1 promoter. ...............................................................................130

Page 13: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

13

5-9 The chain of events triggered by Pdx1 towards β-cell cycle progression.. .....................131

Page 14: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

14

LIST OF ABBREVIATIONS

BrdU Bromo-deoxy-uridine

CFSE Carboxyfluorescein diacetate, succinimidyl ester

CMV Cauliflower mosaic virus

ELISA Enzyme-linked immunosorbance assay

GAD Glutamic acid decarboxylase

GFP Green fluorescent protein

GLP-1 Glucagon-like peptide-1

GST Glutathione transferase

H&E Hematoxylin & eosin

HRP Horse radish peroxidase

Hsp Heat shock protein

IAA Insulin autoantibody

IACUC Institutional animal care & use committee

IAPP Islet amyloid polypeptide

ICA Islet cell autoantibody

IDDM Insulin dependent diabetes mellitus

IgG Immunoglobulin G

INGAP Islet neogenesis associated protein

INS-1 Insulinoma cell line

IP Intra peritoneal

IPCs Insulin-producing cells

IPGTT Intraperitoneal glucose tolerance test

NOD Non-obese diabetic

Page 15: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

15

NIDDM Non-insulin dependent diabetes mellitus

PAGE Poly acrylamide gel electrophoresis

PBS Phosphate-buffered saline

Pdx1 Pancreatic duodenal homeobox-1

PTD Protein transduction domain

rPdx1 Recombinant Pancreatic duodenal homeobox-1

RT-PCR Reverse transcriptase polymerase chain reaction

SDS Sodium dodecyl-sulphate

STZ Streptozotocin

Page 16: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

16

Abstract of Dissertation Presented to the Graduate School of the University of Florida in Partial Fulfillment of the Requirements for the Degree of Doctor of Philosophy

PDX1 PROTEIN THERAPY: A NOVEL APPROACH FOR TREATMENT OF DIABETES

By

Vijay Koya

August 2009

Chair: Lijun Yang Major: Medical Sciences—Molecular Cell Biology

Diabetes is a metabolic disorder characterized by hyperglycemic condition that occurs due

to insulin deficiency (autoimmune type 1 diabetes) or insulin resistance (type 2 diabetes).

Pdx1 is widely regarded as a master transcriptional regulator of the pancreas and is critical for

development, regeneration, and maintenance of beta-cell function. Pdx1 protein possesses a

protein transduction domain (PTD) sequence that facilitates its entry into cells. This study,

therefore, sought to evaluate the capacity of in vivo administered recombinant Pdx1 (rPdx1) in an

effort to ameliorate hyperglycemia in mice with streptozotocin (STZ)-induced diabetes.

Administration of rPdx1 into STZ-induced diabetic mice led to restoration of euglycemia.

Insulin, glucagon, and Ki67 immunostaining revealed increased islet cell number and

proliferation in islets of pancreata of rPdx1-treated mice. Analysis of liver tissue by RT-PCR and

insulin immunostaining demonstrated that the rPdx1 treatment promoted liver cell

reprogramming into insulin-producing cells. This novel PTD-based protein therapy offers a

promising way to treat patients with diabetes while circumventing the potential side effects

associated with the use of viral vectors.

We further investigated the therapeutic effects of rPdx1 in NOD mice (type 1 diabetic

mouse model). Administration of rPdx1 to pre-diabetic NOD mice prevented the onset of

Page 17: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

17

diabetes. The characterization of NOD mice revealed the presence of anti-Pdx1 autoantibodies in

pre-diabetic mice indicating the possible role of Pdx1 as an autoantigen. The role of Pdx1 as an

autoantigen was confirmed by demonstration of Pdx1 dependent activation of lymphocytes and

mapping of the epitope region responsible for autoantibody production. Decreased proliferation

of CD4+ T lymphocytes in the pancreatic lymph nodes of rPdx1-treated mice, confirmed the role

of rPdx1 in immunomodulation. In conclusion, Pdx1 is a novel autoantigen and rPdx1 antigen-

based immunotherapy could serve as an effective strategy for prevention of the onset of diabetes.

In order to understand the role of Pdx1 in beta cell regeneration, we explored to study the

siRNA mediated knockdown of Pdx1 in rat insulinoma cells (INS-1cells). The BrdU

proliferation assay demonstrated a decrease in the proliferation of cells treated with Pdx1 siRNA.

The knockdown studies suggested that beta cell specific transcription factors and cell cycle

regulators may be involved in regulation of beta cell proliferation. Molecular studies indicated

that Pdx1 promotes beta cell cycle progression by activation of cyclin B1 expression via Nkx6.1.

In conclusion, these studies investigated the role of Pdx1 in promoting beta cell

regeneration, liver cell transdifferentiation, and immunomodulation towards the treatment of

diabetes.

Page 18: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

18

CHAPTER 1 INTRODUCTION

Diabetes

Diabetes is a metabolic disorder characterized by hyperglycemic condition. The symptoms

include frequent urination (polyuria), excessive thirst (polydipsia), extreme hunger (polyphagia),

increased fatigue and unusual weight loss. According to the estimates by the American Diabetic

Association, there are 20.8 million children and adults in the United States, or 7% of the

population suffering with diabetes. The exact cause of diabetes continues to be a mystery, it has

been demonstrated that both genetic and environmental factors play a role in causing the

disease(1-5). Diabetes is classified into two types, type 1 (insulin dependent diabetes mellitus or

IDDM) and type 2 (non-insulin dependent diabetes mellitus or NIDDM).

Type 1 Diabetes

IDDM occurs due to an absolute deficiency of the insulin hormone. It occurs mostly in

children and young adults, hence, termed as juvenile diabetes. The disease is characterized by the

autoimmune destruction of pancreatic beta cells(6). Predisposition to a given autoimmune

response requires the requisite allele(s) that controls antigen presentation by antigen-presenting

cells for T-cell recognition. The main susceptibility genes code for polymorphic HLA molecules

and in particular alleles of class II MHC genes (DR, DQ and DP). Polymorphisms of individual

genes outside the MHC also contribute to diabetes risk but recent evidence suggests that there

are additional non-HLA genes determining susceptibility linked to the MHC (7). Some

autoimmune responses emerge following infection by a pathogen, whose protein(s) possess

structural similarities in some of its epitopes to regions on proteins of the host(2). Thus,

antibodies evoked against a pathogen might cross-react with a self-protein and act as

autoantibodies, and the involved autoantigen then provides a source for persistent stimulation.

Page 19: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

19

Proteins to which the immune system is ordinarily self-tolerant might, if altered, elicit auto-

immune responses. Environmental factors appear to play an important role in the pathogenesis of

childhood-onset type 1 diabetes(2). The most important factors are thought to be infectious,

dietary, perinatal, and psychosocial. Enteroviruses (especially Coxsackie B virus), breastfeeding,

the early presence or lack of certain foods, birth weight, childhood over-nutrition, maternal islet

autoimmunity, and negative stress events have been shown to be related to the prevalence of type

1 diabetes. However, clear conclusions to date are limited (8).

It has been reported that approximately 50% of the genetic risk for type 1 diabetes can be

attributed to the HLA region. The highest risk HLA-DR3/4 DQ8 genotype has been shown to be

highly associated with beta-cell autoimmunity. The first antibodies described in association with

the development of type 1 diabetes were islet cell autoantibodies (ICA) (9). Subsequently,

antibodies to insulin (IAA)(10), glutamic acid decarboxylase (GAD)(11) and protein tyrosine

phosphatase (IA2 or ICA512) have all been characterized(12). Several studies have been

conducted in non-obese diabetic (NOD) mice to understand the antigen based

immunotherapeutic strategies to suppress beta cell auto immunity. Much success has been

achieved in NOD mice using administration of autoantigens such as insulin(13;14),

GAD65(11;15-19), and Hsp60(20;21). Few of the antigen molecules have been tested in human

patients with limited success. The Insulin antigen based clinical trials were conducted in the pre-

diabetic human patients in Diabetes Prevention Trail-1 (DPT-1), however the treatment had

limited or no significant effects(22;23). Similarly inconclusive results were obtained with

clinical trials using Hsp, P27-peptide in human subjects(24).

Type 2 Diabetes

Type 2 or non-insulin dependent diabetes mellitus (NIDDM), accounts for 90% of cases

and is characterized by a triad of (1) resistance to insulin action on glucose uptake in peripheral

Page 20: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

20

tissues, especially skeletal muscle and adipocytes, (2) impaired insulin action to inhibit hepatic

glucose production, and (3) dysregulated insulin secretion (25). In most cases, Type 2 diabetes is

a polygenic disease with complex inheritance patterns (4;25). Environmental factors, especially

diet, physical activity, and age, interact with genetic predisposition to affect disease prevalence.

(26). Type 2 diabetes is often accompanied by other conditions, including hypertension, high

serum low-density-lipoprotein (LDL) cholesterol concentrations, and low serum high-density-

lipoprotein (HDL) cholesterol concentrations. Hyperinsulinemia occurring in response to insulin

resistance may play an important role in the genesis of these abnormalities. Increased free fatty

acid levels, inflammatory cytokines from fat, and oxidative factors, have all been implicated in

the pathogenesis of type 2 diabetes. Insulin insensitivity is an early phenomenon partly related to

obesity and pancreas beta-cell function declines gradually over time already before the onset of

clinical hyperglycemia. Several mechanisms have been proposed, including increased non-

esterified fatty acids, inflammatory cytokines, adipokines, and mitochondrial dysfunction for

insulin resistance, glucotoxicity, lipotoxicity, and amyloid formation for beta-cell dysfunction

(27).

Current Treatment Options

The most common method of treatment of both type 1 and type 2 diabetes is Insulin

replacement therapy. Insulin replacement therapy was first initiated during 1920’s and continues

to be the most common approach to overcome adverse effects of the diabetic condition.

However, insulin replacement therapy remains to be a management drug that can ameliorate the

symptoms but is not considered a true remedy (28). Although it is the most common approach

for the management of diabetes, it has several complications. Acute complications include

dosage issue where in patients may be at a risk of overdose leading to hypoglycemia. Chronic

Page 21: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

21

complications are associated with aberrant glucose metabolism that include retinopathy,

nephropathy and cardiovascular problems (29).

With the development of Edmonton protocol for islet transplantation, it gained importance

as an alternative long term therapy for type1 diabetes patients. The treatment regimen also

included long term use of immunosuppressive drugs sirolimus and tacrolimus. Patients did

achieve insulin independence only for a short period of time (1 year) (30). The long term usage

of immunosuppressive drugs and insufficient number of donor cadavers for transplantation

procedures has put the brakes on this approach.

Novel Approaches for Treatment of Diabetes

With the advancements in stem cell technologies, the cell based therapy has become a

promising avenue for treatment of diabetes. Transdifferentiation of bone marrow derived cells

into beta-like cells has been achieved with limited success in mice models(31). Autologous

hematopoietic stem cell transplantation approach is currently under clinical trial in Brazil. The

newly onset diabetic patients (15 patients) were identified and administered with immune-

modulatory drugs and GM-CSF to mobilize bone marrow cells(32). The patients could achieve

insulin independence for over a period of 3 years. Efforts have been made to engineer cord blood

stem cells into insulin producing beta like cells, however the complexity of this approach has

significantly slowed down the progress in this area(33). A major mile stone has been achieved

recently in cell based therapy with the engineering of glucose responsive insulin secreting cell

from embryonic cells(34)

Pancreatic Duodenal Homeobox 1 (Pdx1)

Pdx1 is a transcriptional factor, plays crucial role in pancreas development and beta cell

differentiation during embryogenesis(35;36). Its role in post natal stage is maintaining mature

beta cell function and regulating gene expression of several important genes in the

Page 22: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

22

pancreas(35;37;38). During embryogenesis, Pdx1 expression domain appears in the duodenum

early in development along with Hlxb9 and demarcates a population of cells that represent the

earliest pancreatic precursors. Later it is expressed in all cells differentiating toward the exocrine

and endocrine components of the pancreas where it triggers the genetic cascade, necessary for

differentiation of precursor cells into different cell types in the pancreas. In the adult pancreas,

Pdx1 expression is predominantly restricted to the insulin-producing islet beta cells and a subset

of somatostatin producing delta cells(39-41). Pdx1 null mutant resulted in agenesis of pancreas

with hyperglycemia and did not survive after birth(35). Pdx1 has been found to play key role in

beta cell neogenesis and insulin production (38).

Function of Pdx1 in Pancreatic Beta Cells

In mature beta cells, Pdx1 transactivates the insulin gene and islet amyloid polypeptide

(IAPP) and other genes involved in glucose sensing and metabolism, such as GLUT2(35;42) and

glucokinase(35;43). Pdx1 regulates the pancreas-specific expression of other transcriptional

factors such as Pax4, Nkx6.1 and Pdx1 itself. Pdx1 co-operates with Hnf-3b to auto-activate its

own transcription (44). During regeneration of the islets, either from duct cells or by replication,

Pdx1 is plays a major role by triggering the genetic cascade of differentiation(45). Pdx1 protein

possesses N-terminal region transactivation domain, in the middle region, an antennapedia-like

homeodomain consisting of 3 helices. The homeodomain of the Pdx1 protein plays a critical role

in DNA binding and protein-protein interactions as a transcriptional activation mechanism.

Furthermore, it contains a protein transduction domain within the helix 3 region consisting of 7

amino acids (RRMKWKK) that is conserved in mice, rats, and humans(46). This sequence plays

a major role in nuclear entry of the Pdx1 protein to perform its major transcriptional events

occurring in the pancreas.

Page 23: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

23

Protein Transduction Domain (PTD)

The protein transduction domains are short peptides with rich basic amino acids such as

arginine and lysine, help facilitate binding to negatively charged heparin sulphate proteoglycans

on the lipids membrane followed by internalization via receptor-independent macropinocytosis

into the cytoplasmic and nuclear compartments(47-49). This phenomemon was first reported in

HIV Tat transactivator (50;51). Later such transduction domains were discovered in

homeodomain region of the antennapedia, a transcriptional factor of the Drosophila

melanogaster(47;52). The protein transduction domain has been proved to successfully carry

protein into the cells and be functional. The artificial engineered protein transduction domains

were covalently linked the macromolecules to transduce the macromolecule into the cell. Site-

specific recombination in human embryonic stem cells was demonstrated using the PTD

mediated Cre recombinase(53). The protein transduction domain has been exploited successfully

to deliver wide variety of molecules such as antigenic epitopes(54) and large protein

molecules(55;56). Most recently recombinant proteins with protein transduction domains were

used for generation of induced pluripotent stem cells(57). The first in vivo studies of PTDs were

demonstrated by engineering beta-gal molecule to the PTD(58). Protein transduction domains

were engineered with therapeutic molecules demonstrated their biological activity in disease

models of asthma (59), Inflammation(60), cancer(61), Stroke(61;62). Pdx1 protein has its own

inbuilt antennapedia like protein transduction domain(63;64) that will help the protein to enter

the cells in a receptor independent manner by lipid raft dependent macropinocytosis(65;66) . The

Pdx1 protein has been used to transduce into pancreatic duct cells and differentiate them into

insulin-producing cells in vitro(63). Despite several studies in vitro showing functional

significance of PTD in Pdx1, in vivo functional studies are yet to be explored.

Page 24: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

24

Beta Cell Regeneration

Pdx1 is regarded as a master transcriptional regulator of pancreas development and is

critically needed for the development, regeneration, and function of islet β-cells. Beta cell

regeneration has been known to occur during pregnancy, initial stages of insulin resistance to

meet the insulin demands, early stages of beta cell destruction, partial pancreatectomy. During

post natal condition, the primary mechanism by which new beta cells are formed is by self

duplication or replication of the existing cells(67;68) and/or differentiation of beta cell from duct

cell progenitors (neogenesis). Pancreatic duodenal homeobox (Pdx1) is the key transcriptional

factor controlling the beta cell regeneration(37;39;68). However, the molecular mechanism

associated with the role of Pdx1 in beta cell regeneration is yet to be investigated. In vivo

regeneration of residual islet β-cells appear to be linked to increased biosynthesis of the

pancreatic duodenal homeobox-1 (Pdx1) transcription factor(45). However, Pdx1 mediated

regeneration mechanism is severely hindered during diabetic condition due to the oxidative

stress(45;69-71). Previous studies in HIT-T15 cells and zucker diabetic rats have shown that

oxidative stress causes post transcriptional defect in Pdx1 mRNA processing(69;72) . One

possible approach to overcome the diabetic condition could be restoring and/or expediting the

beta cell replication mechanism by augmenting the Pdx1 expression. The mechanism of beta cell

replication in hyperglycemic state could be enhanced by hyper-expression of key transcription

factors such as factor pancreatic duodenal homeobox (Pdx1) necessary for beta cell regeneration.

Liver Cell Transdifferentiation into insulin-producing cells (IPCs)

Liver and pancreas are closely related both developmentally and phylogenetically. Liver

and pancreas are derived from appendages of the upper primitive foregut endoderm. It has been

suggested that the late separation of liver and pancreas during organogenesis in primitive ventral

Page 25: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

25

endoderm might have left both tissues with pluripotent cells that are capable of giving rise to

both hepatic and pancreatic lineages. In lower organisms, such as worms and fish, the two organs

never gets separated during organogenesis and the ‘hepato–pancreas’ functions as both liver and

pancreas (73-76). Tissues of both the organs have some metabolical similarities such as

responsiveness to glucose, and both express a large group of specific transcription factors such

as hepatic nuclear factor 1α and 3β that are known to control the Pdx1 expression in pancreatic

cells and could cooperate other wise silent Pdx1 expression during transdifferentiation

(39;44;74). Under the experimental and pathological conditions such as cancer, transconversion

of pancreatic acinar cells and hepatocytes in both rodents and humans has been reported(77).

Mounting evidence indicates that liver stem cells(78;79) and adult hepatocytes(80) can be

reprogrammed into IPCs by ectopic overexpression of Pdx1 that restore euglycemia in diabetic

mice(81). Pdx1 induces developmental redirection from hepatic to pancreatic lineage by

repressing the expression of key hepatic transcription factor CCAAT/enhancer -binding protein β

(C/EBPβ)(82). Liver cell transdifferentiation into IPCs was achieved in both in vitro and in vivo

conditions by hyper expressing Pdx1 via gene transfer approach using lentivirus or

adenovirus(78;83-85). Transient over expression of Pdx1 using adenovirus gene therapy in the

liver in vivo has led to transdifferentiaton of liver cells into insulin-producing cells. This

remarkable approach has several advantages such as IPCs derived from liver cell do have

resistance against the autoimmune attack unlike the beta cells in islets(86). However, studies

from other groups using gene therapy resulted in hepatitis(86;87) and development of tumors

which limited the feasibility to apply for human subjects. The alternative approach using protein

therapy using protein transduction domain could be a safe and efficient approach for the

production of insulin-producing cells. A safe and efficient method of delivering Pdx1 to

Page 26: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

26

reprogram the target cells would eventually make this approach more feasible for the clinical

setup. Investigations are underway to characterize the cell types within liver that are susceptible

to transdifferentiation. It was demonstrated that hepatocyte proliferation induced by partial

hepatectomy and enforced ectopic Pdx1 expression using gene therapy accelerate

transdifferentiation process in liver(88). Recent studies have used DDC mouse model for

demonstrating the transdifferentiation of oval cells into IPCs(89).

Significance

Given its key role in pancreatic development, islet β-cell regeneration, liver-to-endocrine

pancreas transdifferentiation as well as its ability to autoregulate itself, Pdx1 protein is an ideal

candidate for protein therapy, especially because of its antennapedia-like PTD, permitting its

rapid entry into cells. We therefore aim to test the hypothesis that effective in vivo delivery of

recombinant Pdx1 protein (rPdx1) will allow its entry into pancreatic or liver progenitor/adult

cells, where it can exert transcriptional control on its target genes, leading to restoration of

normoglycemia in Streptozotocin-induced diabetic mouse model and delay in the onset of

diabetes in Non-obese diabetic mouse model. We believe that the proposed study will serve as a

model system to design a rational protocol for pre-clinical investigation using protein based

approach for the treatment of diabetes. This approach will eliminate the need for gene therapy

that is facing severe challenges in clinical trials.

Page 27: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

27

CHAPTER 2 GENERAL METHODS

This chapter discusses general methodologies and materials that are common used in the

laboratory. However, specific experimental procedures in relation to the specific aims are

described in detail in respective chapters.

Recombinant DNA Technology

Plasmid Isolation

The DNA plasmid was isolated and purified using Qiagen plasmid miniprep kit or

Qiagen plasmid midi kit.

Preparation of LB Broth

Ten LB medium capsules (Lennox) in 500 mL dd water were used in 2 L flask to make

500 mL of media. The flask was autoclaved and cooled to room temperature before use.

Agar Gel Electrophoresis

For 1% agarose gel, 1 gram of electrophoresis grade agarose (Biorad) was mixed in 1X

TAE buffer (Tris-acetic acid-EDTA). The mix was boiled for 1 min 30 sec in a microwave. The

solution was allowed to cool at room temperature. 5 µL of ethidium bromide (10,000X) was

added to the solution and mixed well. The mix was poured onto the gel casting apparatus and it

was allowed it to solidify. The gel was placed in an electrophoresis tank with 1X TAE buffer.

After loading the samples, the gel was run at 100 V.

Polymerase Chain Reaction

All reactions were performed using a standard procedure with variation in annealing

temperature. 95°C denaturing temperature 5 min, followed by 30 cycles of 94°C denaturing

temperature for 30 sec, 55-58°C of annealing temperature for 30 sec and 72°C extension

temperature for 30 sec. The reaction ended with final extension temperature of 72°C for 7 min.

Page 28: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

28

Taq polymerase (Eppendorf) was used for all the PCR reactions.

Restriction Digestion

Approximately 0.5-1 µg of DNA was used for digestion in a 20 µL reaction. For each 20

µL reaction, 2 μL of 10X buffer (New England Biolabs, NEB), 1 μL of restriction enzyme

(NEB) and sterile dH2O to make up the volume to 20 μL was used in the reaction. The reaction

was incubated for 3 hours at 37°C. 0.2 μL 100X BSA was used for certain enzyme digestions

recommended by NEB catalog. For double digestions, compatible buffer was used based on

NEB catalog recommendations.

DNA Ligation

Approximately 100 ng of digested DNA was used for a 20 µL ligation reaction. For each

20 µL reaction, 2 µL of T4 DNA ligase buffer, 10X (Invitrogen) and 1 μL of ligase enzyme and

sterile dH2O to make up the volume to 20 μL was used in the reaction. The reaction was

incubated at 16°C overnight.

Transformation of Competent E.coli Cells

One Shot MAX Efficiency DH5α-T1 chemically competent cells (Invitrogen, CA) were

used for the transformation of bacteria. The competent cells were stored at -80°C. The cells were

thawed on ice (50 µL total). 1-50 ng DNA was added to the tube containing cells, gently swirled

and incubated on ice for 30 min. The cells were treated with heat shock by placing the tube in

water bath at 42°C for 1 min. The cells were then immediately placed on ice for 2 min. 250-450

µL S.O.C. medium (Invitrogen, CA) was added to the tube and incubated at 37°C for 1 hour on a

shaker at 225-250 rpm. Finally, 20 µL and 100 µL of tube mix were plated on separate LB agar

plates containing ampicillin. The plates were incubated overnight at 37°C. The plates were stored

at 4°C in plastic wrap to prevent dehydration.

Page 29: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

29

Protein Expression, Purification and Analysis

Protein Expression in Bacteria

A single isolated clone was obtained and grown in 20 mL LB broth with 20 µL of 0.1%

kanamycin (100 µg/mL) (Sigma). The culture was allowed to grow overnight on a shaker at 220

rpm and 37°C. The entire culture was transferred to a large flask containing 500 mL of LB broth.

After growth at 37°C to an optical density (OD600) of 0.8, culture was incubated at room

temperature for another 18 hours in the presence of 0.5 mmol/L (final concentration) isopropyl-

D-1-thiogalactopyranoside (Fisher Scientific Stock: IPTG 1 g/ 4.2 mL ddH2O). The bacteria

culture was pelleted by centrifugation at 6000 rpm for 30 min. The supernatant was discarded

and pellet was stored at -80°C.

Protein Purification

The purification protocol was optimized for 3 L bacterial culture. The bacterial pellet was

stored at -80°C. After thawing at room temperature, 50 mL of 5 mM imidazole (Sigma) buffer

with 1 tablet of protease inhibitor cocktail (Roche, CA) and 0.2 g lysozyme (Sigma) was added.

The lysozyme was first dissolved in water before the imidazole was added. This allowed the

lysozyme to dissolve completely. 500 mL of Buffer A (10 mL of 1 M Tri/HCL pH.8, 50 mL of

5 M NaCl, 0.5 mL of Triton X-114, 800 µL of 3 M Imidazole , added ddH2O to bring the

volume to 500 mL) solution was prepared and stored at 4°C. 100 mL of Buffer A was added to

the bacterial pellet and left on ice for 30 min, this was followed by vigorous vortexing for 30

min. At this point the highly viscous sticky mix was formed. Later, the mix was transferred to

two centrifuge tubes and the solution was sonicated for 15 sec X 4 times each on ice. After

sonication, the solution became less sticky. The tubes were balanced and centrifuged at 12000

rpm for 30 min 4°C. The supernatant was saved. (If the supernatant wasn't clear, the

centrifugation step was repeated). While centrifugation, Ni-NTA agarose bead (Invitrogen) was

Page 30: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

30

washed with ddH2O first and then equilibrated with 5 mM imidazole solution. The clear

supernatant obtained by centrifugation in the earlier step was mixed with ready to use Ni-NTA

before leaving it on the spinner at 4°C for 30 min. The column with NI-NTA and supernatant

mix was placed on the stand and allowed to drip down. 150 mL of 40 mM imidazole washing

solution (1.74 mL of 3 M imidazole into 150 mL of buffer A) was added to the column for

washing unwanted proteins. This was followed by another wash step with 150 mL of 40 mM

imidazole solution (1.74 mL of 3 M imidazole into 150 mL of 5 mM imidazole buffer). This step

was incorporated to get rid of the triton X-114 used in an earlier step. At the last wash, the

protein content was checked by mixing 25 µL sample with 500 µL Bradford solution (Bio-Rad

Protein Assay (5X) 10 mL + 40 mL ddH2O). If the color was brown, the washing step was

discontinued. The target His tagged protein was eluted by adding 60 mL of 300 mM imidazole

elution buffer (Elution buffer: 3 mL of 3 M imidazole + 27 mL of 5 mM imidazole. The samples

were collected in eppendorf tubes and checked the target protein by mixing 25 µL sample with

500 µL Bradford solution. In order to check the purity, SDS (Sodium Dodecylsulphate) gel was

run with 25 µL sample + 25 µL 2X sample buffer (Bio-Rad Laemmli Sample Buffer). Each lane

was loaded with 30 µL and the gel was run at 80 V until the dye front migrated through the gel.

The gel was later separated from the cast and washed with water on a shaker for 1 hour. The gel

was finally stained with Coomassie blue (Bio-Rad Bio-safe Coomassie).

Dialysis of the Purified Protein

In order to eliminate the imidazole from the purified protein, the elute was dialyzed against

the PBS. First, the dialysis tubing was boiled at 95°C in ddH2O for 5 min to make it flexible. 2 L

of PBS was prepared using ddH2O and chilled by placing it in the refrigerator. The sample was

placed into the dialysis tubing and dialyzed against 1 L of 1X PBS while stirring overnight. The

Page 31: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

31

next morning, PBS was changed and dialyzed for 3-4 hours. Finally, the protein concentration

was measured and stored at -80°C by adding 10% (v/v) of glycerol.

Sodium Dodecylsulphate - Polyacrylamide Gel Electrophoresis (SDS-PAGE)

The SDS-PAGE was performed using a mini gel electrophoresis apparatus (Bio- Rad). The

pre-cast polyacrylamide gels (Bio-Rad) were used to run the samples. The samples were diluted

in 2X sample loading buffer (Laemmli loading dye, Bio-Rad). Composition: 62.5 mM Tris-HCl,

pH6.8, 2% SDS, 25% Glycerol, 0.01% Bromophenol Blue. 50 µL of β-mercaptothanol was

added to 950 µL of 2X sample loding buffer. The gel was run using 1x TGS buffer, 25 mM Tris,

192 mM Glycine, 0.1 % SDS, pH 8.3, (Bio-Rad). Equal volumes of the sample and 2x sample

loading buffer were mixed and boiled at 95°C for 5 min. The electrodes of the apparatus were

connected to the power pack (Bio-Rad) and run at 80 V for 1-2 hours.

Western Blotting

Transfer buffer with 15% methanol (100 mL 1x TGS buffer, 150 mL methanol, and 750

mL ddH2O was used to transfer the proteins onto the nitrocellulose membrane.

A transfer sandwich was prepared by encasing the gel and nitrocellulose membrane

between the filter paper and sponge (sponge - filter paper - gel - PVDF membrane - filter paper -

sponge). Then the transfer sandwich was placed in the electrophoresis tank. The apparatus was

run at 80 volts for 1.5 hours. The membrane was washed in 1x TBST by combining 100 mL 10x

TBS, 1 mL Tween 20, and 899 mL ddH2O. The membrane was then blocked with blocking

buffer using TBST with 5% non-fat dry milk (50 mL TBST + 2.5g dry milk) for 1 hour at room

temperature or at 4°C overnight with gentle rotation. The primary antibody was diluted (as per

company’s recommendation) in 10 mL of blocking buffer with 1% non-fat dry milk and

incubated at room temperature for 1 hour or at 4°C overnight with gentle rotation. The

membrane was washed with approximately 30 mL 1x TBST buffer for 15 min followed by 2

Page 32: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

32

washes for 10 min each. The membrane was then incubated with the secondary antibody with

HRP conjugation (diluted as per company’s recommendation) in blocking buffer with 1% non-

fat dry milk and incubated at room temperature for 0.5-1 hour with gentle rotation. The

membrane was finally washed with 30 mL 1x TBST buffer for 15 min, followed by 2 washes for

10 min each. The membrane was dried by blotting the corner on a paper towel for 5 seconds.

One mL each of western blotting detection ECL reagents 1 and 2 (Amersham Biosciences) was

mixed. The membrane was placed on parafilm or polyvinyl wrap and covered evenly with

western blotting reagent mixture and incubated at room temperature for 1-2 min. The x-ray film

was developed in the dark room by exposing it for appropriate amount of time depending on the

signal.

Mammalian Cell Culture Protocols

Cell Culture

The INS-1 cells were grown in RPMI 1640 (Sigma) (430 mL) with 10% fetal bovine

serum supplemented with 5 mL of HEPES (1M), 5 mL of L-glutamine (200mM), 5 mL of

Sodium pyruvate (100 mM), 5 mL of Pen/Strep, 454 µL of 2-Mercaptoethanol (1000x) and 2 mL

Kanamycin (Stock: 50 mg/mL) per bottle of media. 3T3 cells were grown in Dulbecco's

Modified Eagle Medium (Sigma) 500 mL bottle supplemented with 10% FBS (55 mL) and 2 mL

Kanamycin (50 mg/mL).

Transfection of Plasmid in Mammalian Cells

Approximately 2 μg of DNA was added to 100 μL of serum free medium (Gibco) in a

microfuge tube for each well. In a separate tube, 4 μl of lipofectamine (Invitrogen) was added to

100 μL of serum free medium for each well. The tubes were vortexed and incubated for 5 min at

room temperature. The lipofectamine and plasmid preparations were mixed together, vortexed,

and incubated for 20 min at room temperature. The cell medium was changed immediately

Page 33: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

33

before adding transfection medium. One preparation transfection medium (200 μL) was added

directly to the normal medium (1 mL) for each well and incubated for 4-6 hours at 37°C. The

transfection medium was aspirated and 1 mL of normal medium was added to each well. The

cells were incubated for 24 hours hours at 37°C.

RNA Isolation from Cells/Tissue

The cells or tissue were homogenized and mixed with trizol reagent (Invitrogen, CA). For

1 mL of trizol mix, 0.2 mL of chloroform (Fisher Scientific) was added and mixed vigorously

and allowed to incubate at room temperature for 15 min. The mix was then centrifuged at 12,000

rpm for 15 min at 4°C. The supernatant aqueous phase was transferred to a clean tube and RNA

was precipitated by adding 0.5 mL of isoproponal (Sigma) and mixing well. The solution was

allowed to incubate for 10 min at room temperature. This was followed by centrifugation at

12,000 rpm for 8 min at 4°C. The supernatant was discarded and the pellet was washed with 1

mL of 75% ethanol and centrifuged at 7,500 rpm for 15 min. The supernatant was discarded and

the pellet was allowed to dry for 10 min under the fume hood. The pellet was resuspended in 50

μL of RNAse free DEPC water (Invitrogen), the concentration was measured and stored at -

80°C.

cDNA Synthesis

Total of 2 μg of RNA was used to synthesize the cDNA. Random hexamers (2 μl)

(Invitrogen) was used and the volume was brought up to 11 μL with RNAse free DEPC water.

The mix was incubated at 70°C for 5 min followed by 5 min on ice. A cocktail mix was prepared

with 5x-first strand buffer (4 μl/sample), DTT (2 μl/sample), dNTP (2 μL/sample) and 1 unit of

superscript II reverse transcriptase (Invitrogen). 9 μl of mix per sample was added to the RNA

mix previously incubated on ice. The final mix was placed in PCR machine and run at 25°C for

Page 34: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

34

10 min followed by 42°C for 50 min. The enzyme was finally inactivated by heating at 65°C for

10 min.

Measurement of Luciferase Activity

Dual-Luciferase Reporter Assay System (Promega) was used for the assay. Luciferase

assays were performed, using a luminometer tube for each sample and 100 µL of the Luciferase

Assay Reagent was loaded into each tube. The luminometer was programmed with a 2 second

measurement delay followed by a 10 second measurement read for luciferase activity. Next, 20

µL of cell lysate was added to the luminometer tube containing the Luciferase Assay Reagent

and it was mixed by pipetting three times. The tube was placed in the luminometer and the

reading was initiated and designated as M1. Next the tube was reloaded with 100 µL of Stop and

Glo buffer and placed in the luminometer and the reading was initiated and designated as M2.

The ratio of M1/M2 determined the ratio of luciferase activity (promoter) vs renilla activity

(internal control).

Extraction and Measurement of Insulin

Serum Sample Collection for Measurement of Insulin by ELISA

The mice were starved for 6 hours before blood collection. The mice were weighed and 2

mg/g body weight glucose was injected intraperitoneally. After 15 min of delivering the glucose

bolus, blood was collected from the retro-orbital sinus using a pasture pipette. The blood was

stored on ice for 30 min. The serum was extracted by centrifuging at 10,000 rpm for 10 min. The

supernatant was collected and stored at -80°C.

Tissue Sample Collection for Measurement of Insulin by ELISA

All the organs with major focus on the pancreas and liver were collected and weighed.

The organs were collected in ice cold Acid-ethanol solution (180 mM HCL in 70% ethanol (500

mL) 350 mL ethanol + 7.76 mL HCL + 142.24 mL H2O). The mechanical tissue homogenizer

Page 35: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

35

was used to mince the tissue into tiny particles. The suspension was allowed to rotate on a

mechanical rotor overnight for the release of tissue extract into the solution. The suspension was

centrifuged at 6000 rpm for 15 min. The supernatant was collected and stored at -80°C.

ELISA for Measurement of Insulin

ELISA was performed to measure the insulin levels using Mouse ultra sensitive enzyme

immunoassay (Alpco Diagnostics) according to the manufacturer's protocol. The protocol was

modified slightly to suit our experimental objectives. 5 μL of tissue extracts were used for the

assay. High-concentration samples such as pancreas extract were diluted 50~500 times with

Calibrator '0'. 25 µL of Calibrator 0 was added to each well of the coated plate, then 5 µL of

Calibrators 3-7 or unknown samples were added to each well (duplicate for each calibrator or

sample). Then 50 µL of enzyme conjugate solution was added to each well. The plate was sealed

with plastic film and incubated on shaker at 800 rpm for 2 hours. Microplate washer was used to

wash the plate 6 times using an inbuilt program. After washing, 200 μl of substrate solution was

added to each well and incubated for 30 min. The plate was covered with aluminum foil to avoid

light. The reaction was stopped using 50 μl stop solution. The plate was then read on microplate

reader (Bio-rad) at 450 nm wavelength. The standard curve and unknown samples concentrations

were analyzed using software (Microplate manager).

Procedures for Imaging

Haematoxylin and Eosin (H &E) Staining

For the H & E staining, the following steps were followed. 3 x 5 min Xylene, 2 x 2 min

100% ethanol, 1 x 2 min 95% ethanol, 1 x 2 min deionized H2O, Hematoxalin staining, 1 x 2.25

min Hematoxalin (Vector Laboratories, CA), 1 x 15 sec deionized water, 1 x 1 min deionized

water, 1 x 1 min Blueing, 1 x 1 min Deionized water , 1x 1.5 min Eosin (Fisher Scientific) , 95%

Page 36: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

36

ethanol 2 dips, 1 x 30s 95% ethanol , 1 x 1 min 100% ethanol, 3 x 1 min Xylene. Finally the

slides were covered with coverslip slides using permount medium (Fisher Scientific).

Immunofluroscence

The paraffin sections were first de-paraffinized and rehydrated using the following steps.

(Xylene: 5 min, Xylene: 5 min, 100% alcohol: 3 min, 100% alcohol: 3 min, 95% alcohol: 3 min,

70% alcohol: 15 dips. These steps were followed by rinsing for 5 min in tap water. For staining

for nuclear antigens, the slides were incubated in 95°C Preheated Trilogy (fresh) solution for 30

min. Later the slides were washed with distilled water for 5 min. Chilled 0.5% Triton-X100

(triton-X100 250 µL+50 mL TBS-0.5% Triton) in TBS was added on the slides and incubated

for 5 min and rinsed for 5 minutes in tap water, followed by wash step with TBS for 5 minutes.

This was followed by Block step for 20 min with serum (15 μL serum in 1 mL TBS). Serum

must be from the same species as the one in secondary antibody. This was followed by

incubation with primary antibody (diluted appropriately according to manufacturer's

recommendations) for 1 hour at room temperature or 4°C overnight. This was followed by wash

step with TBS for 5 min. Next, the tissue sections were incubated with secondary antibody

(diluted appropriately according to manufacturer's recommendations) for 45 min. A final wash

with TBS was continued for 5 min. The slides were mounted with Vectashield with Dapi (Vector

Laboratories, Burlingame, CA) and the coverslips were applied.

General Animal Procedures

Animal Housing

Balb/c mice were bred in pathology mouse colony, University of Florida), and housed in

SPF conditions. NOD female mice, 8 weeks old age were ordered from Jackson labs (Bar Harbor,

ME, USA) and housed in SPF conditions in pathology mouse colony. All procedures were carried

out as described in approved Institutional Animal Care & Use Committee (IACUC) protocols.

Page 37: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

37

Collection of Blood

Blood was collected from the mice through retro orbital bleeding. The mice were

anesthetized using isoflurane. The mice was held with the left hand and a glass capillary tube

was placed on the medial aspect of the right eye and gently rolled until the blood started flowing

through the tube. A maximum of 100 µL blood was collected for mice under experimentation,

while a maximum of 400 µL was collected from mice designated for terminal bleeding.

Tissue Sample Collection for Histology

The mice were sacrificed according to the University of Florida, IACUC guidelines. The

mice were sprayed with 70% ethanol. A mid-ventral incision was made using scissors, followed

by collection of organ samples such as pancreas, liver, small intestine, kidneys, spleen, heart,

lung, and brain. The organ samples were preserved in 10% formalin for 24 hours and later

transferred to PBS. The organs were cut into small pieces and placed in cassettes for paraffin

embedding at the pathology core facility. The paraffin blocks were cut into 5 µm sections.

Measurement of Blood Glucose

The mice were either fasted or non-fasted before measuring the blood glucose. Before

measuring IPGTT, the mice were fasted for 6 hours. For measurement of fasting blood glucose

levels, the BALB/C mice were fasted for 8-10 hours. The mice were pricked with a 22 guage

needle on the tip of the tail. A drop of blood was squeezed from the tip of the tail. The

glucometer (Accu-Chek) was used to measure the blood glucose. The glucose strip was inserted

into the glucometer and a drop of blood was collected in a slot on the strip. The glucometer

displayed readings with units of mg/dL.

Tissue Sample Collection for RNA Isolation

The mice were sacrificed according to the University of Florida, IACUC guidelines. For

the large organs such as liver, the tissue samples were cut into small pieces. Several pieces were

Page 38: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

38

collected from various liver lobes. For smaller organs such as kidney, spleen, the organs were cut

into 2-3 pieces. The tissue samples were wrapped in aluminum foil and snap frozen by dropping

them in liquid nitrogen. The samples were later stored at -80°C.

Anesthesia

The mice were given general anesthesia using isoflurane placed under isoflurane assembly

unit with nose cone apparatus as per IACUC guidelines.

Pancreatectomy

The mice were anaesthetized using an isoflurane assembly unit with nose cone apparatus. Toe

pinch method was used to verify if the mice was under anesthetic condition. The anesthesia was

maintained until the end of procedure. The anesthetized mouse was laid on its right lateral side. The

abdominal area of the left lateral side was wiped with 70% ethanol. An incision was made on left

lateral abdominal area where pancreas is located. Following incision, the peritoneal cavity was

exposed, pancreas was located and pulled out using forceps. The head of the pancreas is attached to

the duodenum and the tail is attached to the spleen. A nylon suture material was used to tie a knot on

the blood vessel running between spleen and tail of pancreas. Another knot was tied at the head of

the pancreas. Knot was tied deep in order to gain access to as much pancreas as possible. The tail of

the pancreas was then cut. The knot tied at the head of the pancreas is pulled gently in order to gain

access closer to the head. Another deep knot was tied closer to the head of the pancreas. A cut was

made between two knots that are tied close to the head of pancreas. A major chunk of pancreas was

isolated. The mouse incision was closed using staples. The mouse was monitored until it recovered

its consciousness. The staples were removed after 7 days.

Splenectomy

The mice were anaesthetized using an isoflurane assembly unit with nose cone apparatus. Toe

pinch method was used to verify if the mice was under anesthetic condition. The anesthetized mouse

Page 39: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

39

was laid on their right lateral side. An incision was made on left lateral abdominal area where spleen

is located. Following incision, the peritoneal cavity was exposed, spleen was located and pulled out

using forceps. A knot was tied at both the ends of the spleen in order to prevent internal bleeding.

The spleen was slowly separated by cutting at both the ends of spleen and the mesenteric

connections. The associated organs such as pancreas are pushed back gently into the abdominal

cavity. The mouse incision was closed using staples. The mouse was monitored until it recovered its

consciousness. The staples were removed after 7 days.

Page 40: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

40

CHAPTER 3 REVERSAL OF STREPTOZOTOCIN-INDUCED DIABETES IN MICE BY CELLULAR TRANSDUCTION WITH RECOMBINANT PANCREATIC TRANSCRIPTION FACTOR

PANCREATIC DUODENAL HOMEOBOX-1

Introduction

Type 1 diabetes is a metabolic disorder resulting from the autoimmune destruction of

pancreatic beta-cells. An intense research effort has been directed at identifying a means for

restoring beta-cell mass as well as glucose-regulated insulin production through islet cell

transplantation (90).Though progress has been made, the scarcity of donor islets and the potential

need for lifelong immunosuppression will, in theory, greatly limit its potential for widespread

application (91). Many alternate strategies have been pursued including vector-mediated delivery

of pancreatic transcription factors that allow for conversion of adult cells into insulin-producing

cells (IPCs) and for use of cocktails of beta-cell growth factors to promote differentiation of

embryonic stem cells into tissues capable of producing insulin(92). In vivo, regeneration of

residual islet beta-cells (90) has been noted to occur with a variety of beta-cell growth factors

including glucagon-like peptide-1 (93;94), exendin-4 (95;96), and the islet neogenesis–

associated protein (INGAP)(97). These outcomes appear linked to an increased biosynthesis of

the pancreatic duodenal homeobox-1 (Pdx1) transcription factor (45;93;94;96;98). Pdx1 is

widely regarded as a master transcriptional regulator of the pancreas and is critical for

development (35;99;100), regeneration (37;45), and maintenance of beta-cell function(37).

Liver stem cells (101) and adult hepatocytes (84;86) reportedly have been reprogrammed by

ectopic overexpression of Pdx1 into insulin-producing cells (IPCs) that are also capable of

restoring euglycemia in diabetic mice. However, the strategies involving the use of viruses as a

means for gene delivery in these studies raise safety concerns. An alternative delivery strategy

has recently been identified wherein short, highly basic peptide sequences called protein

Page 41: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

41

transduction domains (PTD) act as molecular passports for facile penetration of cellular

membranes by proteins (49). Indeed, studies seeking to understand the cell internalization of

antennapedia-like homeodomain peptides, a form of PTD, support the view that transcription

factors can be transferred from cell to cell, possibly with direct paracrine activity (47;102). The

likely mechanism of cell entry by PTD-containing proteins relies on strong electrostatic

interactions of the cationic PTD with phospholipid elements residing in the plasma membrane,

followed by macropinocytosis and eventual release into the cytoplasm (49;103). Once

internalized, the protein is then free to exert its biological activity on susceptible targets. Given

its key role in pancreatic development, islet beta-cell regeneration, and liver-to-endocrine

pancreas transdifferentiation, Pdx1 represents an ideal candidate for protein therapy, especially

since its antennapedia-like domain is a PTD, mediating its rapid entry into cells (63;65;66).

Previous in vitro studies involving pancreatic duct and islet cells (63), as well as embryonic stem

cells (104), have demonstrated that Pdx1 protein cell entry induces insulin gene expression and

positive autoregulation of Pdx1 gene expression (63;104). Therefore, the ability of Pdx1 to

stimulate insulin gene transcription in vivo renders it a highly attractive approach as a potential

treatment for diabetes. This study investigated the therapeutic effects of rPdx1 in the Stz-induced

diabetic mouse model.

Materials and Methods

Construction and Production of rPdx1, PTD–Green Fluorescent Protein, and Mutant Pdx1 Fusion Proteins

To express recombinant rat Pdx1-His6 (hereafter designated rPdx1), full-length rat Pdx1

cDNA was amplified by PCR and subcloned using NdeI and XhoI sites in pET28b (Novagen,

Madison, WI). To express PTD–green fluorescent protein (PTD-GFP)-His6 (hereafter designated

PTD-GFP), the PTD-GFP plasmid containing the coding sequence for the 11-residue

Page 42: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

42

(YGRKKRRQRRR) PTD of HIV-1 TAT positioned at the NH2-terminus of green fluorescent

protein was constructed by PCR and cloned into a pT7/CT-TOPO expression plasmid

(Invitrogen, Carlsbad, CA). To prepare the 16-residue PTD-deletion mutant of rPdx1-His6

(rPdx1-mut), a mutant rat Pdx1 expression plasmid missing PTD residues 188–203

(RHIKIWFQNRRMKWKK) within the rPdx1 homeodomain was constructed using PCR

amplification with appropriate primers containing sequences before and after the PTD of Pdx1

cDNA. The two PCR products were subsequently ligated to generate the PTD-deletion mutant.

After confirmation of the cDNA sequence, the resulting mutant Pdx1 cDNA subcloned into the

NdeI and XhoI sites of pET28b (Novagen). After growth at 37°C to an optical density (OD600) of

0.8, plasmid-containing BL21 (DE3) cells were incubated at room temperature for another 18

hours in the presence of 0.5 mmol/l (final concentration) isopropyl-D-1-thiogalactopyranoside.

Bacteria were lysed by pulse sonication in buffer A (20 mmol/l Tris/HCI pH 8.0, 500 mmol/l

NaCl, and 0.1% Triton X-100) containing 5 mmol/l imidazole and proteinase inhibitors (Roche

Diagnostics, Basel, Switzerland). After centrifugation, the cell-free supernatant was applied to a

column of Ni-nitrilotriacetate agarose (Invitrogen) and washed with several volumes of buffer A

containing 25 mmol/l imidazole. The protein was eluted by buffer A containing 250 mmol/l

imidazole. The purity of the eluted rPdx1, PTD-GFP, and rPdx1-mut fusion proteins were

characterized by SDS-PAGE/Coomassie Blue staining following dialysis against PBS.

Cell Entry and Immunoblotting

Rat liver epithelial stem cells (WB cells) (105;106) at 70% confluence were treated with

purified rPdx1 or rPdx1-mut at a 1 µm concentration for various time points, and the cells were

washed three times with PBS, harvested in lysis buffer (150 mmol/l NaCl, 50 mmol/l Tris-HCl,

pH 7.5, 500 µmol/l EDTA, 1.0% Triton X-100, and 1% sodium deoxycholate) containing a

protease inhibitor cocktail (Roche Diagnostics). For Western blotting (27,31), antibodies against

Page 43: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

43

rPdx1 (rabbit serum, 1:1,000 dilution), his-tag (1:2000; Invitrogen), or actin (1:2000; DAKO,

Carpinteria, CA) were used to detect rPdx1, rPdx1-mut, actin, or PTD-GFP fusion protein in cell

lysates (50 μg total protein/lane) after SDS/PAGE separation.

rPdx1 Functional Analyses Using NeuroD-Luciferase Reporter Construct

3T3 cells were co-transfected with NeuroD-luciferase reporter construct (1µg) and Tk-

renilla reporter construct (0.5 µg), followed by either transfection with Pdx1 cDNA (0.5 µg) or

transduction with rPDx1 protein (10 µm). 50 μm Choloroquine, lysosomotropic agent was added

to the all the wells for the release of protein from the lysosomes. The luminescence was

measured using luminometer after 24 hours incubation.

Animal Studies

BALB/c mice (8–10 weeks-old, University of Florida, pathology mouse colony) were

injected with 50 mg/kg body wt streptozotocin (Sigma-Aldrich, St. Louis, MO) for 5 consecutive

days to induce diabetes (27,31). Animals with fasting blood glucose levels for two consecutive

readings of above 250-300 mg/dL received protein treatment. The diabetic mice were

intraperitoneally injected with either rPdx1 or PTD-GFP protein (0.1 mg /day/ mouse) for 10

consecutive days. Fasting blood glucose levels were measured regularly using a glucometer after

the mice were fasted for 6 hours. The sequential experimental events of the animal studies are

summarized in Fig. 3-6.

Low-Dose Streptozotocin-Induced Diabetes

1.47 grams of sodium citrate (Fisher Scientific) was dissolved in 50 mL of distilled water.

The pH was adjusted to 4.5 and stored at room temperature. Appropriate amount of

streptozotocin (STZ), depending on the number of animals, was weighed and dissolved in right

amount of sodium citrate solution so that final concentration is 7.5 mg/mL.

Page 44: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

44

Appropriate amount of STZ- Na citrate was intra-peritoneally injected so that the final

concentration is 50 mg/kg.bdwt of mouse. The mice were injected daily with STZ and blood

glucose was measured before the injection. The dosage was continued until the blood glucose

levels reach more than 250 mg/dL. The maximum number of doses was limited to five. Once the

hyperglycemia was confirmed, the treatment with rPdx1 was begun immediately.

Intraperitoneal Glucose Tolerance Test

The normal, rPdx1, or PTD-GFP treated mice (n > 4 for each group) were fasted for 6

hours following intraperitoneal injection with glucose (1 mg/g body weight), and blood glucose

levels were measured at 5, 15, 30, 60, and 120 min postinjection. Subtotal pancreatectomy (~80-

90%) (n >4) was performed at day 30 post-treatment under general anesthesia. The mice were

killed at two time points (days 14 or 40 post-treatment), and organs and blood were collected for

evaluation of histology, gene expression, serum, and tissue insulin levels.

Pdx1 Protein in vivo Kinetics and Tissue Distribution

Mice were injected intraperitoneally with rPdx1 (0.1 or 1 mg). Blood samples were drawn

at 0.25, 0.5, 1, 2, 6, and 24 hours. Tissues from normal and rPdx1-treated mice were harvested at

1 or 24 hours, fixed in 10% formalin, and embedded in paraffin for Pdx1 immunohistochemistry

using rabbit anti-Pdx1 antibody (1:3,000; a gift of Dr. Christopher V. Wright, Vanderbilt

University).

RT-PCR

Total RNA was prepared from mouse tissues of the pancreas and liver using TRIZOL

reagent and cDNA synthesized using Superscript II reverse transcriptase (Invitrogen) with

random hexamer primers. Liver gene expression was determined by RT-PCR as previously

described (31). The forward and reverse PCR primers were designed to be intron spanning, and

their sequences and conditions are listed in Table 3-1. To eliminate false positives, no reverse

Page 45: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

45

transcriptase, positive, or blank controls were included. All data represent at least three

measurements from at least four mice.

Quantitative Real-Time RT-PCR Analysis

cDNA from pancreatic and liver tissues was subjected to three independent PCR

reactions. Each reaction was performed in duplicate or triplicate in a Thermocycler Sequence

detection system (DNA engine opticon 2; MJ Research, Springfield, MO) using SYBR green

(Qiagen, Valencia, CA). Total pancreatic and liver RNA was used as templates for preparing

cDNA. Tissues were snap-frozen in liquid nitrogen and RNA was extracted in RNase free tubes

using Trizol reagent (Invitrogen). The integrity and stability of the RNAs from both pancreas and

liver were confirmed by demonstrating the intact 28s and 18s bands on gel electrophoresis.

cDNA was synthesized from 5 µg total RNA using Superscript reverse transcriptase enzyme

(Invitrogen) with random hexamer primers according to the manufacturer’s protocol.

Amplication of the correct product was confirmed by gel electrophoresis. Conditions for real-

time PCR were as follows: after initial denaturation at 95°C for 15 min to activate the enzyme,

38 cycles of PCR (denaturation 0.5 min at 94°C, annealing 0.5 min at 61°C [for liver] or 56°C

[for pancreas], and elongation 0.5 min at 72°C with a final extension 5 min at 72°C) were carried

out. Each gene was tested three times (3–4 mice/group). Relative gene expression in mouse

pancreas treated by rPdx-1 and PTD-GFP on day 14 was calculated by 2ΔΔCt method. We used

mouse β-actin as internal control and PTD-GFP treated day-14 pancreas cDNA as calibrator.

First, both threshold cycle (CT) values of target gene from rPdx1-treated day-14 pancreas cDNA

(n =3) and PTD-GFP–treated day-14 pancreas cDNA (n =4) were normalized by CT value of the

internal control. Then, the former was subtracted by the latter, namely ΔΔCT rPdx1/GFP _ (CT,Target

_ CT,Actin)rPdx1 _ (CT,Target _ CT, Actin)GFP. Similarly, ΔΔCT between rPdx1-treated day-14 liver

cDNA (n =3) and rPDX1-treated day-40 liver cDNA (n =4,) was also calculated by ΔΔCT day

Page 46: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

46

14/day 40 _ (CT, Target _ CT,Actin) day 14 _ (CT,Target _ CT, Actin) day 40. The value of 2ΔΔCT, the

fold change of gene expression, was plotted into the figure. The primers were designed in

accordance to the real-time PCR conditions, and the sequences are listed in Table 3-2.

Immunohistochemistry and Immunofluorescence

Paraffin blocks containing the pancreas and liver were obtained from at least 5

mice/group. Antigen retrieval from paraffin sections was done in trilogy solution (Cell Marque,

Rocklin, CA) at 95°C for 30 min for unmasking nuclear antigens such as Pdx1 and Ki-67.

Paraffin sections (5 µm) were incubated with anti-swine insulin (1:1,000), rabbit anti-Pdx1

(1:5,000), rabbit anti-human Ki67 (1:100; Novus Biologicals, Littleton, CO), and goat anti-

glucagon (1:200) antibodies, followed by incubation with anti-mouse or rabbit IgG (1:5,000)

conjugated with HRP and detection with DAB substrate kit (Vector Laboratories, Burlingame,

CA) as previously described (31). For double immunofluorescence, tissue sections were

incubated with anti-swine insulin (1:200; Dako, CA) and goat anti-glucagon (1:150; Santa Cruz

Biotechnology, Santa Cruz, CA) primary antibody overnight at 4°C, followed by donkey anti–

guinea pig IgG conjugated with fluorescein isothiocyanate (1:1,000; RDI Research Diagnostics,

Concord, MA) and donkey anti-goat IgG with Alexa flour 594 flourochrome (1:500; Invitrogen).

For Ki67/insulin sequential immunostaining, the paraffin sections from pancreas were first

immunostained for Ki67 nuclear antigen and developed in brown color using the HRP/DAB

system. After being counterstained with hematoxylin to highlight cell nuclei (blue), the slides

were immunostained for insulin using guinea pig anti-porcine insulin (cross-react with mouse

insulin) antibody (Dako) at a dilution of 1:200 for 16 min. Presence of insulin in pancreatic islet

beta cells was visualized in red color with the Ventana Ultra View red detection kit (Ventana

Medical System, Tucson, AR).

Page 47: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

47

Tissue and Serum Insulin Measurements by Enzyme-Linked Immunosorbent Assay

Whole pancreas and liver organs were obtained from at least five mice/group. They were

harvested, weighed, and immediately placed in acidethanol solution (180 mmol/l HCl in 70%

ethanol) on ice with a corresponding tissue volume (1 mL buffer/0.1 g liver or 0.05 g pancreas)

in accordance with a previously published procedure, with minor modifications (32). Tissue

insulin levels were measured using an ultrasensitive mouse insulin enzyme linked

immunosorbent assay (ELISA) kit (ALPCO, Diagnostics, Salem NH). Absorbance was

measured using a BIO-RAD 3550-UV microplate reader, with final results converted to

nanograms insulin/milligrams pancreas tissue or nanograms insulin/grams liver. For

measurement of serum insulin, both normal and treated mice were first fasted for 6 hours, and

blood samples were collected at 15-min intervals following intraperitoneal glucose (1 mg/g body

wt) stimulation. Serum insulin levels were determined by ELISA.

Statistical Analysis

Statistical significance was analyzed using an independent sample t test, requiring a P

value <0.05 for the data to be considered statistically significant.

Results

Generation of Recombinant Fusion Proteins

The expression plasmids containing rat Pdx1, rat Pdx1-mut, or PTD-GFP cDNA were

constructed, each containing an additional nucleotide sequence coding for a His6 tag for rapid

purification using Ni2+-nitrilotriacetate columns. To obtain nearly homogeneous proteins in

sufficient amounts for the in vitro and in vivo animal studies, bacterial expression conditions

were optimised to yield 10 mg highly pure rPdx1 per liter of growth medium. Figure 3-1A shows

the structural organization of rPdx1 (a), PTD-GFP (b), and rPdx1-mut (c) and a Coomassie blue-

stained SDS gel for rPdx1, PTD-GFP, and rPdx1-mut fusion proteins. These proteins consistently

Page 48: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

48

had purity >90–95%, based on densitometry. The rPdx1, rPdx1-mut, and PTD-GFP proteins was

confirmed by Western blotting with anti-Pdx1 or anti–histidine-tag antibody.

Characterization of Recombinant Fusion Proteins

To confirm that rPdx1 protein possessed the ability to penetrate cells due to antennapedia-

like PTD within the homeodomain of Pdx1, WB cells were incubated with rPdx1 or rPdx1-mut

protein (1 µmol/l) for specified times, after which the cells were washed three times with PBS.

Cell lysates were separated by SDS-PAGE and blotted with anti-Pdx1 and anti-actin antibodies.

The relative amount of rPdx1 in the cell blots was quantified by densitometry and normalized

relative to actin. As shown in Fig. 3-2a, rPdx1 protein entry commenced within 5 min. As rPdx1

incorporation proceeded, cellular rPdx1 protein level reached peak values at 1–2 hours and began

to fall by 6 hours. In contrast, rPdx1-mut with deletion of the 16aa antennapedia-like PTD failed

to enter the cells, even though the cell culture medium still contained high levels of rPdx1-mut

protein (Fig. 3-2b). Since this assay cannot distinguish genuine rPdx1 protein entry versus

nonspecific binding to cell membrane surface, it was determined whether internalized rPdx1

protein was biologically active. To test the transcriptional function of the rPdx1 protein, 3T3 cells

were transfected with pNeuroD-luciferase reporter plasmid with or without rPdx1. NeuroD is

direct downstream target gene of Pdx1 protein. The cells were incubated in the presence or

absence of rPdx1 (10 µm). 3T3 cells co-transfection with CMV-Pdx1 plasmid for 24hours served

as a positive control. A comparable transcription efficacy for CMV-Pdx1–treated cells and rPdx1

protein–treated cells after 24 hrs was observed (Fig. 3-3). Both treatments showed statistically

significant differences when compared with control cells containing pNeuroD-luciferase plasmid

alone. These results clearly demonstrated that rPdx1 rapidly entered cells and efficiently

activated its downstream Neuro-D target gene.

Page 49: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

49

In vivo Kinetics and Tissue Distribution

While the antennapedia-like PTD allows in vitro–administered rPdx1 to enter into the cells,

the in vivo tissue distribution and kinetics of rPdx1 were unknown. To examine rPdx1

distribution and kinetics, BALB/c mice were intraperitoneally injected with 0.1 or 1 mg rPdx1

protein, blood samples were collected at various times, and rPdx1 was detected in sera by anti-

Pdx1 immunoblotting. The rPdx1 became evident in sera as early as 1 hour postinjection,

reaching peak values at 2 h and then markedly falling by 6 hours. No rPdx1 protein was

detectable in the 24-hour serum samples (Fig. 3-4). Major organs were harvested at 1 or 24 hour

after intraperitoneal injection and probed with anti-Pdx1 antibody. As shown in the representative

images of liver, pancreas, and kidney tissues (Fig. 3-5), the rPdx1 was concentrated mainly in the

nuclei of hepatocytes, and the greatest intensity of Pdx1-positive cells was found nearest the

hepatic terminal veins. This distribution pattern is consistent with the predicted pathway for

rapidly internalized rPdx1 via the portal vein system. The rPdx1 was also detected in peripheral

acinar cells of the pancreas, possibly as a result of direct uptake or through local circulation along

pancreatic terminal capillaries. In kidneys, rPdx1 was mostly concentrated in brush borders of

proximal tubular cells. Little protein was seen in the cytoplasm and none in the nuclei. At 24

hours postinjection, only faint rPdx1 protein was focally detected in liver, pancreas, and kidney

tissue sections. A low level of rPdx1 was also detected in the tissues of the spleen, heart, lung,

and brain at 1 hour postinjection, but it became undetectable at 24 hours postinjection (data not

shown). Similar findings were also noted in mice treated with 1 mg rPdx1 protein. Control mice

showed no detectable rPdx1 protein in any tissue section except for pancreatic islet beta cells. On

the basis of these findings, the 0.1-mg rPdx1 at 24 hours intervals was chosen for in vivo

condition for assessing in vivo effects of rPdx1 on mice with streptozotocin-induced diabetes.

Page 50: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

50

In vivo Effects of rPdx1 on Blood Glucose Levels in Diabetic Mice

With this, an experimental timeline was designed for rPdx1 protein administration (Fig. 3-

6) in mice with streptozotocin-induced diabetes. Diabetic BALB/c mice (body weights 20 g)

were intraperitoneally administered with 0.1 mg rPdx1 or nontherapeutic PTD-GFP protein

(negative control) over 10 consecutive days. Fasting blood glucose levels were monitored as

indicated. Mice receiving rPdx1 injections achieved near normoglycemia within 2 weeks of first

injection (Fig.3-7); however, no amelioration of hyperglycemia was observed in control mice

receiving the PTD-GFP.

At days 14 and 40 postinjection, intraperitoneal glucose tolerance test (IPGTT) (Fig. 3-8)

showed that the mice receiving rPdx1 injection, in contrast to those diabetic mice receiving PTD-

GFP, exhibited a much improved, nearly normal IPGTT curve. To further assess the ability of

glucose-stimulated insulin release in the rPdx1-treated diabetic mice, healthy normal and treated

mice were challenged at days 14 and 40 post-rPdx1 or–PTD-GFP injection with intraperitoneal

bolus of glucose. Sera collected from normal, rPdx1-treated, or PTD-GFP–treated mice at 15 min

postglucose injection were assayed for insulin and glucose (Fig. 3-9). Serum insulin levels in

rPdx1-treated mice were 6.9 times higher at day 14 and 11.3 times higher at day 40 than those in

the PTD-GFP–treated group (Fig. 3-9), indicating a significant improvement in the ability of the

rPdx1-treated mice to handle glucose challenge. Overall, the serum insulin levels were inversely

related to the blood glucose levels on days 14 and 40 postinjection in mice treated with rPdx1 or

PTD-GFP protein. Although near euglycemia was achieved at day 40 in rPdx1-treated mice, the

released insulin (2.6 µg/l) following 15 min of glucose stimulation was still much lower than that

in normal nondiabetic mice (5.7 µg/l), suggesting either functional immaturity of newly formed

islet beta cells or suboptimal beta cell mass in the pancreas or IPCs in nonpancreatic tissues for

glucose homeostasis.

Page 51: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

51

Pdx1 Treatment Promoting Endogenous beta cell Regeneration

To explore whether rPdx1-mediated normoglycemia resulted from islet beta cell

regeneration, rPdx1-treated mice were subjected to subtotal (>90%) pancreatectomy at day 30 (n

= 4) postinjection. Blood glucose levels rose sharply following subtotal pancreatectomy (Fig. 3-

7), indicating that pancreatic cells (presumably the islet beta cells) played a dominant role in

restoring normoglycemia at 30 days post–rPdx1 injection. To further determine the role of the

pancreas at earlier stages of blood glucose normalization, subtotal pancreatectomy was

performed at day 12 postinjection in rPdx1-treated mice (n = 4) and resulted in elevated blood

glucose levels: from an average of 170 to 350 mg/dL within 48 hours postoperation. These results

raise the distinct possibility that in vivo delivery of rPdx1 promoted endogenous beta cell

regeneration.

Immunohistochemical examination confirmed vigorous islet beta cell regeneration, with

larger and more abundant islets evident in rPdx1-treated mouse pancreata, in contrast to rare and

scattered small islets in PTD-GFP–treated mice (Fig. 3-10A). Such findings indicate that when

administered in vivo in multiple doses, rPdx1 appears to promote endogenous beta cell

regeneration via an as yet undefined molecular/cellular mechanism. To further characterize the

regenerated islets, double immunofluorescence studies were conducted with anti-glucagon and

anti-insulin antibodies. Significantly, a change in the alpha cell–to–beta cell ratio and distribution

patterns was noted (Fig. 3-10B) consistent with a dynamic process of islet cell regeneration and

maturation. To determine the proliferation rate in the newly regenerated islets following rPdx1

treatment, Ki67 staining was sequentially performed, followed by insulin immunostaining, on

pancreas sections from various groups of animals. As shown in Fig. 3-11, representative

micrographs indicate that Ki67-positive pancreatic islet cells from normal mice were rarely seen

(0–1 Ki67 positive cell/islet (Fig. 3-11). Although few proliferating pancreatic islet cells were

Page 52: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

52

observed in the PTD-GFP–treated mice, it was difficult to identify normal-sized islets except for

scant scattered small islets (Fig. 3-11). However, Ki67-positive islet cells were markedly

increased in the newly regenerated islets in the pancreata of the rPdx1-treated mice compared

with those of control mice. (Fig. 3-11). Sequential double Ki67/insulin immunostaining (Fig. 3-

11) showed that both insulin-positive islet beta-cells and non–beta-cells were proliferating in

islets from tissues obtained at days 14 and 40 post–rPdx1 treatment.

To determine the molecular events underlying rPdx1-mediated islet beta cell regeneration,

real-time RT-PCR was used to examine the expression of key genes relevant to pancreas

regeneration. rPdx1 treatment of diabetic mice (Fig. 3-12) at day 14 resulted in markedly

upregulated levels of INS-I (21.3 times), Pdx1 (3.8 times), INGAPrP (14.5 times), Reg3 (6.8

times), and PAP (34.3 times) relative to their corresponding control values (PTD-GFP–treated

pancreas). A similar pattern of upregulation of the aforementioned genes was observed at day 40

post-treatment. Interestingly, expression of PAP, although upregulated at day 14 when compared

with that in PTD-GFP–treated mice, was noticeably reduced at day 40 posttreatment. These

results indicate that rPdx1 protein can promote islet beta cell regeneration, possibly by

upregulating genes involved in pancreatic cell regeneration.

Pdx1 Treatment Promoting Liver Cell Transdifferentiation into IPCs

To access, whether in vivo intraperiteonal rPdx1 delivery affects the liver, presence of IPCs

were detected in treated mice at days 14 and 40 postinjection. Representative liver images from

mice treated with PTD-GFP or rPdx1 at day 14 posttreatment (Fig. 3-13) indicate that most of the

scattered insulin-staining–positive liver cells were distributed along the edges of hepatic terminal

veins (H.T.V.), a pattern consistent with rPdx1 tissue distribution in the liver (Fig. 3-13). There

were scattered individual insulin-positive hepatocytes (arrows) having small bilobed nuclei and

Page 53: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

53

dark condensed chromatin, hinting at a more mature cell pattern. No IPCs were observed in the

control GFP-treated mouse liver.

The expression profiles of pancreatic genes were investigated for rPdx1 and PTD-GFP–

treated livers (Fig. 3-14). As expected, the rPdx1-treated livers at day 14 posttreatment (lane 6)

expressed many Pdx1-target pancreatic genes, including Pdx1, INS-I, GLUC, ELAS, and IAPP.

Upregulation of other pancreatic endocrine genes (INS-II, SOM, NeuroD, and ISL-I) and

pancreatic exocrine genes (p48 and AMY), was noted in the livers of the rPdx1-treated mice

compared with that in the PTD-GFP–treated mice. However, Ngn3 gene expression was not

detectable at either time point. Interestingly, by day 40, INS-I, GLUC, ELAS, and IAPP gene

expression became undetectable, whereas expression of the aforementioned pancreatic genes

continued, albeit at reduced levels. To quantitatively compare the changes of the pancreatic genes

in the livers between days 14 and 40, real-time RT-PCR was performed for selected mRNAs

including Ins-i, Gluc, Pdx1, p48, Amy, and Elas. As indicated in Fig. 3-15, a several-fold increase

was noted at day 14 vs. day 40 mice post–rPdx1 treatment.

Given the intrinsic ability of rPdx1 protein to penetrate cells indiscriminately, the tissue

specificity of rPdx1's effect on expression of pancreatic Pdx1, Ins-I, Gluc, and Amy genes was

examined by RT-PCR. Little or no pancreatic gene activation was detected at day 14 post–rPdx1

treatment in kidney, brain, heart, lung, small bowel, or spleen (Fig. 3-16). These results suggest

that rPdx1 selectively promotes liver expression of pancreatic genes and pancreatic beta cell

regeneration without detectable evidence of undesired expression in other tissues.

Relationship Between Pancreas and Liver at Tissue Insulin Levels

To determine the relative contribution of pancreas and liver tissue–derived insulin to

ameliorating blood glucose levels following rPdx1 treatment, pancreas and liver tissue insulin

content of both normal and treated mice was measured at day 14 or 40 by ELISA. Pancreatic

Page 54: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

54

insulin content in rPdx1-treated diabetic mice at days 14 and 40 postinjection was 44 and 68%,

respectively, of the normal pancreatic levels (Fig. 3-17). These levels were also 6.7 times higher

at day 14 (P < 0.01, Student's t-test) and 15.8 times higher at day 40 (P < 0.001) post-treatment

compared with those in the PTD-GFP–treated diabetic mice.

There is a marked increase ( 16 times) in the liver tissue insulin content at day 14 post-

treatment in the rPdx1-treated mice over that of the PTD-GFP–treated mice (P < 0.001) and

nearly a nine fold increase over that of normal liver (Fig. 3-18). Interestingly, there was sharply

reduced liver insulin content at day 40 post–rPdx1 treatment, although it was still 7.3 times

higher than that in PTD-GFP–treated mice (P < 0.01) and 2 times higher than that in normal

liver. These findings confirmed that in vivo rPdx1 treatment promoted pancreatic islet beta cell

regeneration and transient liver cell transdifferentiation into IPCs, suggesting that liver and

pancreas both contributed to achieving glucose homeostasis in a compensatory fashion.

Discussion

Given that absolute and relative insulin deficiencies, respectively, form the basis of type 1

and 2 diabetes, the identification of a means for restoring functional beta cell mass would hold

immense promise as a means for curing these disorders. In this study, several novel findings

were observed: 1) in vivo administration of rPdx1 ameliorates hyperglycemia in diabetic mice, 2)

amelioration of hyperglycemia is attended by both pancreatic beta cell regeneration and liver cell

transdifferentiation, and 3) the observed therapeutic effect is likely to require Pdx1 to have an

intact protein transduction domain. These experiments therefore constitute a proof-of-principle

demonstration that protein therapy in the form of in vivo Pdx1 delivery to animals can be a

highly effective therapeutic strategy, one that exploits intrinsic properties of a naturally occurring

pancreatic transcription factor and that avoids undesirable effects typically associated with viral

vector-mediated gene therapies.

Page 55: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

55

Specifically, these results indicate that in vivo delivery of rPdx1 can promote both beta cell

regeneration as well as liver cell transdifferentiation into IPCs. Pdx1-mediated pancreatic islet

beta cell regeneration appears to be the dominant effect on glucose homeostasis, since marked

hyperglycemia was observed in mice receiving nearly total pancreatectomy. Although the exact

cellular and molecular events responsible for rPdx1-mediated beta cell regeneration and liver cell

transdifferentiation remain to be defined, based on the findings, rPdx1 protein enters the

circulation via terminal veins and capillaries and penetrates cellular membranes to gain nucleus

entry into target cells in the liver and pancreas (Fig. 3-5), resulting in activation of rPdx1-

dependent transcription factor cascade. The notion that rPdx1 promotes pancreatic beta cell

regeneration is supported by the presence of numerous pancreatic large islets (Fig. 3-10), an

increasing number of proliferating islet cells (Fig. 3-11), an ensuing increase in the level of

pancreatic tissue insulin (Fig. 3-17), and the significant upregulation of several key genes related

to pancreatic cell regeneration in rPdx1-treated mice (Fig. 3-12). The fact that rPdx1 vigorously

promoted pancreatic islet cell proliferation and regeneration raises intriguing questions about the

type of pancreatic cells that are the targets of rPdx1 (e.g., what is the cell origin for the newly

regenerated islets?). Possible mechanisms include residual islet cell proliferation, exocrine acinar

cell transdifferentiation, and pancreatic ductal/stem cell neogenesis. Presently, this approach

limits the tracking of the cells of origin into/within the target tissue, resulting in the newly

formed islets.

Several genes (INGAPrP, Reg-3 , and PAP) upregulated by rPdx1 treatment are members

of the pancreatic regenerating (Reg) gene family originally identified in animal models of beta

cell regeneration (107). Their gene products play important roles in the maintenance of

progenitors in the process of pancreas regeneration. Despite variation between individual

Page 56: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

56

samples, expression levels of the Reg genes determined by RT-PCR correlated well with an

earlier study involving conditional expression of Pdx1 in a transgenic mouse model (37).

The observed functional effects of rPdx1 on the liver are consistent with the published

results of ectopic expression of Pdx1 gene via adenovirus vectors resulting in liver cell

transdifferentiation (83;84). In the current experimental model, liver and pancreas appear to work

in a sequential, compensatory manner to ameliorate hyperglycemia and ultimately to restore

euglycemia in rPdx1-treated mice. The kinship between the liver and pancreas in controlling

glucose homeostasis is also supported by a recent study using liver and pancreas double-injury

animal models (89). The early phase of rPdx1-induced hepatic insulin production is supported by

an intense expression of the insulin I gene (Fig. 3-14) and a nearly 18-fold increase in liver tissue

insulin in comparison with that of control liver (Fig. 3-15). While the precise mechanism

underlying the rPdx1-mediated surge in hepatic insulin during early-stage glucose homeostasis

remains to be elucidated, possible explanations include the following: 1) action of hepatic insulin

on pancreatic progenitor cells via the insulin signaling pathway to promote beta cell regeneration

via IRS2-Akt-Pdx1–mediated signal transduction (108); 2) insulin-mediated facilitation of beta

cell neogenesis, involving amelioration of hyperglycemic toxic effects on residual beta cell

regeneration (109); and 3) rPdx1-mediated hepatic insulin production, resulting in an increased

rate of glucose clearance by the liver, perhaps by promoting glucokinase expression and/or by

insulin's stimulatory action on glycogen synthase, thereby lowering blood glucose levels (110).

Pdx1 gene expression has also been reported to be associated with beta cell neogenesis in

rodent pancreas injury models (37;45;108;111). Although this study showed roughly a 2 to 4x

increase of Pdx1 gene expression in the pancreas of rPdx1- over PTD-GFP–treated mice,

pancreatic beta cell function appears to be exquisitely sensitive to small changes in Pdx1 gene

Page 57: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

57

expression levels in both humans and mice (111;112). The rPdx1 protein can positively regulate

Pdx1 gene expression, as evidenced by upregulation of endogenous Pdx1 gene expression in the

livers of rPdx1-treated mice. These observations are consistent with the findings of others

indicating that Pdx1 protein binds to its own promoter and positively regulates its own gene

expression (44). These results suggest that rPdx1-based protein therapy may not require a large-

dose or long-term treatment and, thus, may reduce or eliminate potential dosage-related systemic

toxicity.

Although in this study it was shown that rPdx1 effectively reverses hyperglycemia in

diabetic mice, there are potential obstacles to clinical translation. One concern is that rPdx1 could

be partially degraded by serum proteases. To assess this possibility, further studies on rPdx1

stability in whole blood and plasma would be helpful. Moreover, detailed pharmacokinetic

studies are needed to optimize dosages, routes of delivery, and the interval between treatments.

The polyclonal anti-Pdx1 antiserum used in this study precludes distinguishing between intact

and any partially degraded rPdx1 protein. Significantly, the ability of rPdx1 to ameliorate

hyperglycemia in diabetic mice, along with its nuclear localization in liver and pancreatic acinar

cells (Fig. 3-5), indicates in vivo availability of a sufficient amount of intact rPdx1 protein or

biologically active degradation products capable of translocation into cells. Another concern is

the potential toxicity of rPdx1 to off-target organs via its PTD, since the rPdx1 protein has the

potential to enter almost any tissue or cell type. We can, however, exclude the activation of rPdx1

target genes in tissues other than liver and pancreas at day 14 posttreatment. Moreover, the

animals appeared normal, without evidence of weight loss or abnormal organ morphology. In

fact, the diabetic mice treated with rPdx1 gained body weight, showed an improved IPGTT, and

exhibited markedly reduced blood glucose levels. Nonetheless, a full toxicity profile of rPdx1,

Page 58: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

58

especially at earlier time points, is required to address this question. These studies are beyond the

scope of the present manuscript and will be pursued in the future.

Interestingly, the distribution of rPdx1 in the kidney is quite different from that in the liver

and pancreas at the early 1-hour time point (Fig. 3-5). Instead of being present in the cell nuclei,

rPdx1 was localized near the brush border of the proximal tubular cells. Such a distribution was

not observed at 24 hours. As the rPdx1 protein rapidly enters the bloodstream (Fig. 3-4) after

intraperitoneal injection, it may be filtered through glomerular capillaries into the urinary spaces

via the fenestrated capillary endothelial cells and glomerular basement membrane. Alternatively,

rPdx1 may gain entry via cells by virtue of its on-board PTD. Once in the urinary spaces, it

would not be surprising if the cationic rPdx1 protein interacts electrostatically with polyanions

(e.g., sialic acid and phopholipids) present on the apical surface of proximal tubular epithelial

cells.

In conclusion, this demonstration that in vivo rPdx1 delivery into diabetic mice rapidly

restores euglycemia exploits the intrinsic properties of this key pancreatic transcription factor

(e.g., its built-in antennapedia-like PTD, its positive autoregulation,(113), its vital role in

pancreatic cell development and regeneration (37;45), and its role in maintaining pancreatic beta

cell function) (37;114;115). Indeed, it is possible that that Pdx1-based protein therapy should

allow for a redirection (or reactivation) of pancreatic stem/precursor cell differentiation and

transdifferentiation (or reprogramming) from non-pancreatic cells along pancreatic beta cell

developmental pathways, a feature that could prove beneficial in treating patients with diabetes.

Page 59: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

59

Table 3-1. Primer name, sequences, size, GenBank #, and PCR condition Genes Forward primer Reverse primer PCR

size GenBank Tm Cycle

(bp) Acc. No. (°C) No. HPRT CTCGAAGTGTTGGATACAGG TGGCCTATAGGCTCATAGTG 350 NM_01355

6 56 40

INS-I TAGTGACCAGCTATAATCAGAG CAGTAGTTCTCCAGCTGGTA 372 NM_008386

56 40

INS-II GCTCTTCCTCTGGGAGTCCCAC CAGTAGTTCTCCAGCTGGTA 288 NM_008387

56 40

GLUC TGAAGACCATTTACTTTGTGGCT TGGTGGCAAGATTGTCCAGAAT 492 NM_008100

57 40

SOM CTCTGCATCGTCCTGGCTTTG GGCTCCAGGGCATCATTCTCT 173 NM_009215

56 40

IAPP TGAACCACTTGAGAGCTACAC TCACCAGAGCATTTACACATA 282 NM_010491

55 40

Glut-2 CGGTGGGACTTGTGCTGCTGG GAAGACGCCAGGAATTCCAT 412 NM_031197

56 40

P48 CCCAGAAGGTTATCATCTGCC CGTACAATATGCACAAAGACG 245 NM_018809

57 40

ELAS AATGACGGCACCGAGCAGTATGT CCATCTCCACCAGCGCACAC 344 NM_033612

57 40

AMY TGGGTGGTGAGGCAATTAAAG TGGTCCAATCCAGTCATTCTG 371 NM_009669

56 40

PDX1 ACCGCGTCCAGCTCCCTTTC CCGAGGTCACCGCACAATCT 357 NM_008814

57 40

NeuroD1

CATCAATGGCAACTTCTCTTT TGAAACTGACGTGCCTCTAAT 257 NM_010894

56 40

ISL-I AGACCACGATGTGGTGGAGAG GAAACCACACTCGGATGACTC 296 NM_021459

56 40

NGN3 TGGCACTCAGCAAACAGCGA AGATGCTTGAGAGCCTCCAC 516 NM_009719

56 40

INS-I = Insulin I, INS-II = insulin-II, GLUC = glucagon, SOM = somatostatin, ELAS = elastase, AMY = amylase. NGN3 = neurogenin 3, IAPP = islet amyloid polypeptide, Glut-2 = glucose transporter-2, ISL-I = islet-1.

Page 60: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

60

Table 3-2. Real time PCR Primer name, sequences, size, GenBank #, and PCR condition Genes Forward primer Reverse primer PCR

size GenBank Acc. No

Tm (°C)

Cycle No.

Actin ACCACACCTTCTACAATGAGC GGTACGACCAGAGGCATACA 185 NM_007393 56 38 INS-I GCCCTTAGTGACCAGCTAT GGA CCA CAA AGA TGC TGT TT 167 NM_008386.2 56 38 PDX1 ATGAAATCCACCAAAGCTCAC AGTTCAACATCACTGCCAGCT 190 NM_008814.2 56 38 INGAPrP GCTCTTATCTCAGGTTCAAGG AGATACGAGGTGTCCTCCAGG 178 NM_013893.1 56 38 Reg3γ CATGACCCGACACTGGGCTATG GCAGACATAGGGTAACTCTAAG 190 NM_011260.1 56 38 PAP AATACACTTGGATTGGGCTCC CCTCACATGTCATATCTCTCC 195 NM_011036.1 56 38

INS-I+ insulin, INGAPrP= Islet neogenesis associated protein-related protein, Reg3γ = regenerating islet-derived 3 gamma, PAP = pancreatitis-associated protein.

Page 61: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

61

Figure 3-1. Cloning, expression, purification, and characterization of rat Pdx1, PTD-GFP, and

rPdx1-mut fusion proteins. The Pannel (A-C) represents the schematic structures of fusion proteins of rPdx1, PTD-GFP, and rPdx1-mut. The cDNAs coding rPdx1, mutant Pdx1, or PTD-GFP were cloned into the expression plasmid. Proteins were expressed and purified by an Ni-column. The purified proteins were run in a 10% SDS-PAGE gel stained with Coomassie Blue and confirmed by Western blotting using anti-Pdx1 antibody and anti–his-tag antibody.

Page 62: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

62

Figure 3-2. Time course of cell entry of rPdx1 and rPdx1-mut proteins. WB cells were incubated

with rPdx1 or rPdx1-mut at a final concentration of 1 µmol/l for indicated times. Proteins were detected by Western blotting with rabbit anti-Pdx1 (1:1,000) or anti-actin (1:5,000) antibodies. The relative amount of cellular rPdx1 protein was quantified by densitometry and the values normalized to actin. The cells treated with rPdx1-mut shows the protein levels in the culture medium by the end of the treatment.

Page 63: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

63

Figure 3-3. Functional analysis of rPdx1 protein. 3T3 cells were co-transfected with NeuroD- promoter with luciferase and Tk-renilla plasmid constructs. The cells were either treated with CMV-Pdx1 or transduced with 10 µm of rPdx1. A comparable transcription efficacy for CMV-Pdx1–treated cells and rPdx1 protein–treated cells was observed after 24 hours compared to the untreated cells.

Page 64: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

64

Figure 3-4. In vivo kinetics and tissue distribution of rPdx1 following intra-peritoneal injection. Normal BALB/c mice were injected intraperitoneally with rPdx1 protein (0.1 mg/mouse). Blood samples were collected at indicated times, and 20 µL serum/lane was loaded in SDS-PAGE gels. The rPdx1 was detected by Western blotting with anti-Pdx1 antibody.

Page 65: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

65

Figure 3-5. In vivo tissue distribution of rPdx1 following intraperitoneal injection. Liver,

pancreas, and kidney tissues were harvested at 1 or 24 h after rpdx1 intraperitoneal injection and fixed in 10% formalin. Paraffin sections were immunostained with anti-Pdx1 antibody (1:1,000). Typical distribution patterns of rpdx1 protein in liver (1, 4, and 7), pancreas (2, 5, and 8), and kidney (3, 6, and 9) were visualized by light microscopy at 1 h and 24 h posttreatment. Pdx1 immunostaining of the liver, pancreas, and kidney tissue sections from normal mice is indicated in the bottom row (10–12). The arrow in panel 2 indicates a small islet in the pancreas with strong nuclear Pdx1 immunostaining.

Page 66: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

66

Figure 3-6. Experimental timeline for rPdx1 treatment. Timing of streptozotocin (Stz) treatment followed by treatment with rPdx1 or PTD-GFP proteins and selective pancreatectomy are indicated. The timing of blood glucose determinations and the measurement of blood insulin levels, IPGTT, measurement of tissue insulin, and the determination of gene expression by RT-PCR were shown.

Page 67: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

67

Figure 3-7. In vivo effects of rPdx1 protein on blood glucose levels. Diabetic BALB/c mice were

treated with daily intraperitoneal injections of 0.1 mg rPdx1 or PTD-GFP for 10 consecutive days (long arrow), and blood glucose levels were determined by glucometer. The blood glucose levels of rPdx1 treated mice demonstrated amelioration in hyperglycemic condition compared to the PTD-GFP treated that remained hyperglycemic. Nearly total pancreatectomy was performed in selected control and rPdx1-treated mice at day 30 (short arrow).

Page 68: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

68

Figure 3-8. Intraperitoneal glucose tolerance test. The IPGTT was performed by i.p. injected

with 1mg/g bd.wt of glucose, and blood glucose was measured at 0, 15, 30, 60, and 120 min in normal, rPdx1-, or PTD-GFP–treated mice. The mice treated with rPdx1 had better glucose handling ability than PTD-GFP treated mice. rPdx1 treated mice had better glucose handling ability at day 40 compared to day 14.

Page 69: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

69

Figure 3-9. Comparison of average blood glucose and Insulin levels between rPdx1 treated and

untreated control mice. Glucose and insulin levels were measured in rPdx1- and PTD-GFP–treated mice 15 min after IPGTT on days 14 and 40 post-treatment (n = 5 mice per group). **P < 0.05; ***P < 0.001 (Student's t test).

Page 70: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

70

Figure 3-10. Histology of pancreatic islets. A. Insulin immunohistochemistry of pancreatic

tissue. Representative picture of immunohistochemsitry of Paraffin-embedded pancreas tissues from mice treated with either PTD-GFP or rPdx1 were sectioned and immunostained with anti-insulin antibody (1:1,000). The Islets in rPdx1 treated mice are relatively larger in size with more insulin-producing beta cells compared to the islets in PTD-GFP treated mice. B. Paraffin sections from PTD-GFP–and rPdx1-treated mouse pancreas tissues were immunostained with both rabbit anti-glucagon/phycoethrin (red) and Guinea pig anti-insulin/FITC (green) and visualized under fluorescence microscopy. There is change in the ratio of alpha-cell–to–beta-cell ratio and distribution patterns.

Page 71: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

71

Figure 3-11. Immunostaining with anti-KI67 antibody and anti-insulin antibody. The islets were stained with Ki-67(brown colour) from tissues of normal mice (1), PTD-GFP treated (2), rPdx1 treated (5, 6). The islets of rPdx1 treated mice were also stained for insulin (Red colour) (7) and double stained for Ki-67 and Insulin (7a)

Page 72: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

72

Figure 3-12. Quantitative Realtime-PCR analyses of pancreatic tissue between rPdx1 treated and

control mice. Total RNA from diabetic mouse pancreas (days 14 and 40 post –rPdx1 or –PTD-GFP treatment) was used for real-time PCR analysis of INS-I, Pdx1, INGArP, Reg3 , and Pap gene expression. Expression levels are normalized to actin gene expression. Data are from 3 mice/group. INGAPrP, islet neogenesis-associated protein related protein; PAP, pancreatitis-associated protein; Reg3 , regenerating islet-derived 3 .

Page 73: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

73

Figure 3-13. Insulin immunohistochemical staining of liver tissue. Paraffin sections from liver

were stained with anti-insulin antibody (1:250). Representative images were taken at 40x or 100x magnification. Insulin-positive cells are seen in the rPdx1-treated mouse liver section at day 14 post-treatment. Arrow, condensed nuclear chromatin of a bilobed-nucleated insulin-expressing liver cell. HTV, hepatic terminal vein.

Page 74: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

74

Figure 3-14. RT-PCR analysis of the expression of pancreatic genes in the liver. Expression of

pancreatic genes in the liver. RT-PCR amplification of RNA extracted from livers of normal, PTD-GFP, or rPdx1-treated mice were analyzed by agarose gel electrophoresis. RNA from mouse pancreas was used as a positive control. For Ngn3 RT-PCR analysis, Ngn3 cDNA plasmid (*) was used as positive control because adult pancreas does not express this gene. No RT, no reverse transcription.

Page 75: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

75

Figure 3-15. Quantitative Real Time-PCR analyses of pancreatic gene expression in liver. Total RNA from diabetic mouse liver (days 14 and 40 post–rPdx1 treatment) was analyzed by real-time PCR for the expression of five Pdx1 target genes (INS-I, Gluc, Pdx1, p48, Amy, Elas). Expression levels are normalized to actin gene expression. Fold changes (D14 over D40) are representative of data from 3 mice/group.

Page 76: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

76

Figure 3-16. Expression of pancreatic genes in other organs. Total RNA from other organs of rPdx1-treated diabetic mice at day 14 post-treatment and expression of four key pancreatic genes (Ins-I, Gluc, Amy, and Pdx1) were examined by RT-PCR. Pancreatic gene expression was observed only in Liver and not in other organs. Data are from 3 mice/group and are representative of three independent experiments.

Page 77: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

77

Figure 3-17. Pancreatic tissue insulin measurements as determined by ELISA. Pancreatic insulin content in rPdx1-treated diabetic mice at days 14 postinjection was 6.7 times higher at day 14 and 15.8 times higher at day 40 post-treatment compared with those in the PTD-GFP–treated diabetic mice. ** = p<0.05. *** = p<0.001.

Page 78: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

78

Figure 3-18. Liver tissue insulin measurements as determined by ELISA. There is insulin

expression of day 14 in significant levels (16 times) compared to normal and PTD-GFP group. However, the insulin levels have reduced by day 40. ** = p<0.05. *** = p<0.001.

Page 79: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

79

CHAPTER 4 PDX1 PROTEIN THERAPY PREVENTS THE ONSET OF TYPE 1 DIABETES IN NOD

MICE

Introduction

Type 1 Diabetes is an autoimmune disorder characterized by the lymphocyte infiltration

into the pancreatic islets resulting in decreased beta cell mass. The treatment of type 1 diabetes

has been challenging due to lack of complete understanding of disease pathogenesis. The key

elements that need to be addressed in order to cure type 1 diabetes include 1) successful

manipulation of immune system(116), and 2) restoration of beta cell mass(117).

It has been reported that approximately 50% of the genetic risk for type 1 diabetes can be

attributed to the HLA region. The highest risk HLA-DR3/4 DQ8 genotype has been shown to be

frequently associated with beta cell autoimmunity(3;7). The first antibodies described in

association with the development of type 1 diabetes were islet cell autoantibodies (ICA) (9).

Subsequently, antibodies to insulin (IAA)(10), glutamic acid decarboxylase (GAA or GAD) and

protein tyrosine phosphatase (IA2 or ICA512) have been characterized (12).

Non-obese diabetes (NOD) mouse is an animal model for investigating the human

juvenile type 1 diabetes. Female NOD mouse (Jackson labs) spontaneously becomes

hyperglycemic accompanied by polyuria, polydipsia, glucosuria, hypercholesteremia and rapid

weight loss. The incidence of diabetes onset is 80% by 30 weeks of age. Histological

examination of pancreas reveals lymphocyte infiltration around and/or into the islets. This

pathological nature can be observed as early as five weeks of age. The number and size of the

islets are markedly reduced in overt diabetic mice. Although the mechanism of pathogenesis is

not clear yet, the NOD mouse has been extensively used as an animal model for investigating the

human juvenile type 1 diabetes(118).

Page 80: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

80

Previous studies have shown that GLP-1R analog (Extendin4) delays the onset of diabetes

in NOD mice(119;120). Studies by other groups in NOD mice have shown that GLP-1 improves

glucose sensitivity in beta cells (121), and protects beta cells from cytokine-induced apoptosis

and necrosis (122). GLP-1 is not only an incretin hormone but also a modulator of cellular

immune system. The multiple pleiotropic effects such as glucoregulatory, proliferative, and

cytoprotective actions of GLP-1 on beta cells are essentially dependent on beta cell Pdx1 gene

expression (122).

The studies conducted in STZ-induced diabetic mice demonstrated that rPdx1 via its

protein transduction domain can enter the targets cells such as pancreatic islets and liver

hepatocytes to promote beta cell regeneration and liver cell transdifferentiation into insulin-

producing cells (IPC’s) towards treatment of diabetes(123). This study is aimed at investigating

the therapeutic effects of rPdx1 treatment in NOD type 1 diabetes mouse model.

Materials and Methods

Animal Studies

Ten-week old female NOD mice (Jackson labs, USA) were injected i.p. daily with 0.1 mg

of rPdx1 (n=20) for 12 weeks or with saline (n=20). Blood glucose (mg/dL) and body weight

(grams) were monitored weekly. Blood glucose was measured using glucometer (Accu-Chek).

IPGTT, serum insulin levels, morphology, and tissue insulin content were examined in NOD

mice between 22-25 weeks of age.

Intraperitoneal Glucose Tolerance Test (IPGTT)

Both the rPdx1-treated and saline treated (n=4 per group) were fasted for six hours before

injecting glucose. To prepare glucose solution, 1 gram of dextrose (Fisher Scientific) was

dissolved in 10 mL of distilled water to make 100 mg/mL solution. After 6 hours of fasting, a

fasting glucose level was obtained using glucometer. Tail tip was pricked with needle to draw

Page 81: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

81

drop of blood. The mice were weighed. 2 mg/g body weight glucose was injected

intraperitoneally. Blood glucose values were obtained at 5, 15 30, 60, and 120 min.

Serum Insulin Measurements

The mice were injected with 2 mg/g bd.wt of glucose (Stock-100 mg/mL). Serum

samples were collected after 15 min of glucose injection from rPdx1-treated NOD mice (22

weeks), diabetic control NOD mice (22 weeks), and normal NOD mice (9 weeks); n=4 per

group. The insulin levels were measured using ultra sensitive mouse ELISA kit (Alpco

Diagnostics) according the manufacturer’s instructions as described earlier.

Construction of Truncated Pdx1 Proteins

Site-directed mutagenesis was performed by use of a QuikChange site-directed

mutagenesis kit (Stratagene). The introduction of a stop codon (TAG) mutations into Pdx1

cDNA at the position of the 120th, , 160th , or 200th amino acid, residue , respectively, was

achieved by the use of pET28-rPdx-1 expression vector as template DNA together with the

following pairs of mutagenic primers: for Pdx1-120, fwd( 5'-gcgttcatct ccctttctag aggatgaaat

ccaccaaagctcac) and rev (5'- ttggtggatttcatcctctagaaagggagatgaacgcggc); for Pdx1-160, fwd (5'-

cgggccca gctgctctagatggagaagg aattcttatttaac) and rev (aataagaattccttctccatctagagcagctggg-

cccgagtgtagg); for Pdx1-200, fwd (5'-tccaaaaccg tcgcatctagaggaagaaag aggaagataagaaacgtag)

and rev (5'-ttatcttcctctttcttcctctagatgcgacggttttggaaccag); (the nucleotide changes giving the

appropriate mutations are shown in bold). After initial denaturation at 95°C for 3 min, the

cycling parameters were 0.5 min at 95°C followed by 1.0 min at 55°C and 7 min

at 72°C (30 cycles).The parental, supercoiled double-stranded DNA in the PCR reaction

mixture was digested with DpnI at 37°C for 40 min before being transformed into competent E.

coli BL21(DE3) cells. The cDNA with Mutations were verified by DNA sequencing. The

Page 82: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

82

expression and purification of the each pdx-1 truncated protein was done as our previously

reported.

Extraction of Splenocytes from the Spleen

The mice were sacrificed and spleen was extracted immediately. The spleen was placed in

a Petri dish with ice cold PBS. Spleen was then minced into fine pieces by placing it between the

tissue section slides. The entire procedure was conducted in sterile conditions under the laminar

flow hood. The fine particles in saline solution were filtered through the tissue strainer (Fisher

Scientific). The solution collected under the strainer was centrifuged at 1000 rpm for 5 min at

4°C. Supernatant was discarded and the cells were resuspended in 2 mL RBS lysis buffer (BD

pharmingen) and followed by incubation at room temperature for 10 min. The cells were

centrifuged again at 1000 rpm for 5 min at 4°C.The supernatant was discarded and the pellet was

resuspended in DMEM with 10% fetal bovine serum.

Lymphocyte Proliferation Assay

Splenocytes were harvested from the normoglycemic rPdx1-treated and control mice.

Splenocytes were resuspended in 10 mL of PBS. 10 µL of suspension was taken and added to 90

µL trypan blue (10 fold dilution). The cells were counted on hemocytometer in the four

quadrants. The average of the four quadrants was multiplied by dilution factor (10) and total

volume 10 mL * 1000. [No. of Cells = Avg. * Dil. Fac* vol* 1000.] One million cells were

seeded into a 96-well plate in triplicates using DMEM with 10% FBS. The cells were treated

with Glutathione transferase (GST), anti-CD3, insulin, or rPdx1 (40 μg each) for 48 hours

followed by incubation with [3H] thymidine (1microcurie per well) for an additional 24 hours.

After 72 hours, the plate was washed and the radioactivity was measured using scintillation

counter (Beckman Coulter).

Page 83: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

83

BDC 2.5 Mice Splenocyte Transfer

Splenocytes were collected from BDC2.5 TCR donor mice(124). The cells were labeled

with carboxyfluorescein diacetate, succinimidyl ester (CFSE) label. 1 million cells were injected

into the age matched rPdx1-treated and control mice (non-diabetic). After 3 days, the

lymphocytes were collected from the pancreatic, cervical and inguinal lymph nodes of rPdx1

treated mice (4 weeks) and control mice. Lymphocytes were analyzed with flow cytometry for

the donor derived CD4+ population.

Results

Administration of rPdx1 to NOD Mice Prevents the Onset of Diabetes

An experimental timeline was designed for rPdx1 protein administration (Fig. 4-1) in pre-

diabetic NOD mice of 10 weeks age group. Mice were intraperitoneally administered with 0.1

mg rPdx1/mouse/day or saline (negative control) for 12 weeks. Random blood glucose levels

(Fig. 4-3) and body weights (Fig. 4-4) were monitored weekly for 40 weeks. 70% of the NOD

mice receiving rPdx1 injections remained normoglycemic until the end of the 40 weeks study

(Fig. 4-2); however, 95% of the NOD mice that received saline became hyperglycemic by 25

weeks of age. These results demonstrated that rPdx1 can significantly prevent the onset of NOD

mice. In order to determine, if short course of treatment can achieve results similar to 12-week

long treatment, NOD mice (n=10) of 10 weeks age were administered with rPdx1 i.p for 4 and 8

weeks. Random blood glucose levels were monitored until the end of 30 weeks of age (Fig. 4-6,

4-8). 70% of the mice that received rPdx1 for 4 and 8 weeks maintained normoglycemia (Fig. 4-

5 and 4-7), while 80% of the saline-treated control mice became hyperglycemic by the end of 30

week study.

Page 84: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

84

Beta cell Functional Analysis and Determination of Tissue Insulin Levels

Intraperitoneal glucose tolerance test (IPGTT) (Fig. 4-9) at 25 weeks of age showed that

the mice receiving rPdx1 injection, in contrast to mice that received saline and became diabetic,

exhibited a better, nearly normal IPGTT curve. To further assess the ability of glucose-stimulated

insulin release in the rPdx1-treated and saline-treated mice, they were challenged with

intraperitoneal bolus of glucose. Sera collected from rPdx1-treated, or saline-treated mice at 15

min post glucose injection were assayed for insulin (Fig. 4-9). Serum insulin levels in rPdx1-

treated mice were 7.5 times higher compared to the saline-treated group (Fig. 4-10), indicating a

significant difference in the ability of the rPdx1-treated mice to handle glucose challenge.

Pdx1 Treatment Preserves Islet Mass Possibly by Beta cell regeneration and Promotes Liver Cell Transdifferentiation into Insulin-Producing Cells.

To explore whether treatment of NOD mice with rPdx1 has resulted in preserved islet cell

mass, pancreatic histology and insulin levels were analyzed at 22 weeks age. H&E staining and

immunohistochemical examination confirmed the presence of more number of larger size islets

evident in the pancreata of rPdx1-treated mice, in contrast to rare and scattered small islets in

saline-treated mice (Fig. 4-11). To further characterize the islets, immunohisto-chemical studies

were conducted with anti-insulin and anti-glucagon antibodies. More insulin positive cells and

organized glucagon positive cells were observed in islets of rPdx1-treated mice compared to the

islets from control mice (Fig. 4-11). In the H& E stained tissues, there was evidence of peri-

insulitis (T-cell infiltration along the perimeter of the islet) in the pancreatic sections of rPdx1-

treated mice (Fig. 4-11) while the pancreatic sections of the saline-treated mice showed extreme

insulitis (invasion and destruction of islet by T-cell infiltration). The insulin levels were

determined in the extracts of both pancreas and liver by ELISA and the levels were significantly

higher in rPdx1-treated mice compared to saline-treated mice (Fig. 4-12). These results indicate

Page 85: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

85

that rPdx1 treatment preserves beta cell mass and promotes liver cell transdifferentiation into

IPCs.

Detection of Pdx1 Antibodies in Serum Samples of rPdx1/Saline-treated Mice

Administration of rPdx1 to the mice could result in production of anti-Pdx1 antibodies.

ELISA was performed in order to determine the presence of anti-Pdx1 antibodies in the serum.

As expected there were detectable levels of antibodies against rPdx1 protein in the rPdx1-treated

mice, Surprisingly there were detectable levels of anti-Pdx1 antibodies in the saline-treated mice

(Fig. 4-13, panel A). In order to determine if this phenomenon was specific for NOD mice,

ELISA was performed using several sera samples from various mouse strains such as BALB/C,

C57/B6, NOD-SCID and NOD mice. The results indeed confirmed the presence of anti-Pdx1

antibodies specifically in NOD mice (Fig. 4-13, panel B). To further validate the observations,

western blot was performed to determine the specificity of the assay. rPdx1 protein was run on

SDS-polyacrylamide gel. Following transfer onto the nitrocellulose membrane, the membranes

were probed with either serum from BALB/C (negative control), or NOD mouse sera that tested

positive or negative for Pdx1 antibody by ELISA. The 41 kDa band was detectable only in the

serum samples of NOD mice that were determined positive by ELISA (Fig. 4-14). These results

validated the presence of anti-Pdx1 antibodies in the sera of NOD mice.

Determination of Antigenic Epitope of Pdx1 Protein

To determine if anti-Pdx1 antibody is generated as a result of autoimmunity against Pdx1

protein or due to secondary immune as a result of immune cell infiltration into the already

injured beta cells. The autoantibodies that are generated as a result of autoimmunity usually have

a specific antigenic determinant and have a monoclonal antibody production while the antibodies

that are formed as a result of secondary immune response are usually polyclonal in nature. To

determine the position of the antigenic epitope, recombinant truncated proteins were generated

Page 86: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

86

by introducing a stop codon at the end of 119, 159, or 199 amino acids. The full-length and

truncated proteins were run on SDS-polyacrylamide gel. Following transfer, the membrane was

incubated with anti-Pdx1 polyclonal rabbit sera or sera from NOD mice. As expected the

polyclonal rabbit sera recognized full-length protein as well as truncated proteins. However, the

serum from NOD mice could recognize only the full-length protein (Fig. 4-15, panel A). This

result indicates that the antigenic epitope is located in the C-terminal portion of the Pdx1 protein

between 200-283 amino acids. In order to demonstrate that the sera from NOD mice can

recognize the 83 amino acids at the C-terminus, a GST fusion protein with C-terminal 83 amino

acid portion (P83) was generated. Western blot using NOD mouse serum was able to recognize

the C-terminal 83 amino acid fragment of the Pdx1 protein (Fig.4-15, panel B). These results

also indicate that sera from NOD mice could be monoclonal which strongly suggests the

autoimmune origin of the antibody.

PAA emerge before the onset of diabetes.

To determine whether the Pdx1 autoantibody can predict disease onset, the relationship

between Pdx1 autoantibody and hyperglycemia was explored (Fig.4-16). Five-week-old female

NOD mice (n = 20) were studied longitudinally and blood samples were taken biweekly until the

onset of diabetes. Blood glucose levels were monitored weekly beginning at 10 weeks of age.

Pdx1 autoantibodies were first detected at ages ranging from 5 to15 weeks, and their levels

gradually increased, peaked, and then declined over the next 2-3 months. Pdx1 autoantibody

often decreased to lower positive levels or disappeared completely after the onset of diabetes and

the peak levels often preceded disease onset. In general, there was an inverse correlation over

time between peak time of Pdx1 autoantibody and blood glucose levels in individual mice.

However, some mice maintained high levels of Pdx1 autoantibody after the onset of diabetes.

Figure 4-16 illustrates the relationship between levels of Pdx1 autoantibody and blood glucose in

Page 87: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

87

three mice. As noted, in two mice Pdx1 autoantibody levels peaked prior to the onset of diabetes.

In the third mouse (m19L), low levels were observed consistently without an apparent peak; this

mouse remained normoglycemia at 25 weeks. These results suggest that Pdx1 autoantibody

could be used as diagnostic marker for prediction of the onset of type 1 diabetes in NOD mice.

Demonstration of Autoreactive Lymphocytes Against Pdx1 Protein in NOD Mice

The presence of autoreactive T-lymphocytes is the hallmark of an autoimmune disease.

Immunotherapy with the soluble autoantigen promotes immunomodulation as observed in

previously known autoantigens such as insulin (125) and GAD (11). Lymphocyte proliferation

assay was performed to demonstrate the presence of Pdx1 autoreactive T-lymphocytes in the

NOD mice and to observe if rPdx1 therapy promoted immunomodulation. The splenocytes

extracted from both rPdx1 and saline-treated mice were treated with CD3, gluthathione-

transferase (GST), insulin, rPdx1, or P83. The splenocytes derived from saline- treated control

mice showed increased antigen-dependent proliferation when treated with insulin, rPdx1, and

P83 suggesting the lymphocyte response to autoantigen. There was decreased proliferation in

splenocytes derived from rPdx1-treated mice when treated with insulin, rPdx1 and P83

suggesting the role of rPdx1 treatment in immunomodulation (Fig. 4-17). These results confirm

the presence of pathogenic reactive lymphocytes that get activated in response to the Pdx1

antigen.

Decreased Proliferation of Donor Derived BDC2.5 CD4+ Lymphocytes in Pancreatic Lymph node

To determine if rPdx1 treatment in the NOD mice has promoted immunomodulation, the

donor derived CD4+ lymphocytes specific to an islet antigen were analyzed for the proliferation

in the pancreatic lymph node. The BDC2.5 donor mice splenocytes labeled with

carboxyfluorescein diacetate, succinimidyl ester dye (CFSE) were transplanted into the recipient

Page 88: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

88

NOD mice pre-treated with rPdx1 or saline for 4 weeks. Flowcytometric analysis of the donor

derived CD4+ lymphocytes in the pancreatic lymph node demonstrated 39% CD4+ donor cell

population, while the rPdx1 mice showed a 15.1% CD4+ donor cell population. (Fig. 4-18). This

result indicates that the immunomodulatory effect of rPdx1 treatment is due to decrease in

proliferation of islet antigen specific CD4+ T lymphocytes within the pancreatic lymph node.

There was no significant difference in the CD4+ T cell population in cervical and inguinal

lymph nodes indicating the immunomodulatory effect was restricted to islet antigen reactive

CD4+ T-cells in the pancreatic lymph node.

Effect of Mutant-rPdx1 On the Onset of Diabetes in NOD mice

To confirm the immunomodulatory role of rPdx1 protein, the mutant protein (rPdx1-mut)

was generated by deletion of 188-203 amino acids. Deletion of these amino acids eliminated the

DNA binding domain (191-196 amino acids) and antennapedia-like protein transduction domain

(188-203 amino acids), thus making the protein biologically inactive. However, the antigenic

determinant region (203- 283 amino acids) remained intact, thus preserving the immunologically

active component of the rPdx1 protein (Fig. 4-19). To determine the efficacy of the rPdx1-mut

protein, NOD mice were treated daily with 0.1 mg of either full-length rPdx1 or rPdx1-mut

protein for 8 weeks. After 30 weeks of follow up, 70% of mice remained normoglycemic with

no significant difference in the onset of diabetes between both the groups (Full-length rPdx1 and

rPdx1-mut) by 30 weeks of age (Fig. 4-20), thus indicating the predominant immunomodulatory

role of rPdx1 in the prevention of diabetes in the NOD mice.

Discussion

This study demonstrates that daily administration of rPdx1 in NOD mice prevents the onset

of diabetes. There is slight difference in onset of diabetes between long-term (12 weeks) and

short-term (4 and 8 weeks) treatment by 30 weeks of age. The long term treatment maintained

Page 89: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

89

normoglycemia in 90% of mice (Fig. 4-2), however short term treatment maintained

normoglycemia in 70% of the mice at 30 weeks of age (Fig. 4-5, 4-7. In the process of

understanding the therapeutic effects of rPdx1 in NOD mice, this study has directed us towards

discovering of a novel Pdx1 autoantigen. Detection of Pdx1 autoantibodies in the control

untreated NOD mice (Fig. 4-13, panel A) has indicated the possible role of Pdx1 as an

autoantigen. There have been extensive studies on the role of autoantibodies such as islet cell

autoantibodies (9), insulin autoantibodies (10), and GAD autoantibodies in the pathogenesis of

type 1 diabetes. In this study, we discovered the presence of Pdx1 autoantibodies in NOD mice

and correlated its presence with the onset of diabetes (Fig. 4-16). It is important to differentiate

between the pathogenic autoantibody and the antibodies that are formed as a secondary immune

response due to infiltration of antigen presenting cells into the already injured beta cells. In case

of autoimmunity, a small peptide epitope region usually evokes a monoclonal antibody response

while the antibodies developed due to immune cell infiltration into injured beta cells are

polyclonal in nature. The fact that NOD serum antibody can recognize only the c-terminal

epitope region indicates the monoclonal and autoimmune nature of the antibody and the possible

role of B-cells in pathogenesis of type 1 diabetes (126).

The in vitro lymphocyte proliferation assay demonstrated a decrease in the proliferation of

lymphocytes derived from rPdx1-treated mice, upon stimulation with Pdx1 autoantigen. This

reduced proliferation was not only limited to stimulation with Pdx1 autoantigen but also spread

to other known beta cell autoantigens such as insulin (Fig. 4-17). This phenomenon of bystander

suppression could possibly be due to production of regulatory cytokines as a result of

administration of rPdx1 soluble antigen (127). These regulatory cytokines produced within the

target tissue (islets) or local lymph node (pancreatic lymph node) are directed against the other

Page 90: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

90

locally expressed autoantigens(128). Pancreatic lymph node serves as the hub for autoimmunity

directed against islet antigens with CD4+ T lymphocytes as mediators. The splenocyte transfer

study from donor BDC2.5 TCR mice (124;129) into recipient NOD mice demonstrated a

decreased proliferation of CD4+ lymphocytes in rPdx1-treated mice (Fig. 4-18), thus

demonstrating specifically the immunomodulatory role of rPdx1 on the CD4+ lymphocytes

residing within the pancreatic lymph node.

In this study we employed three distinct approaches to demonstrate that that Pdx1 is a

novel autoantigen. Firstly, demonstration of anti-Pdx1 antibodies specifically in NOD mice

indicates the involvement of B-lymphocytes (Fig. 4-13, panel B). Secondly, mapping out the

epitope region responsible for the antibody production (Fig. 4-15). Thirdly, demonstration of

antigen mediated lymphocyte proliferation in response to Pdx1 antigen (Fig. 4-17). Besides

these, the immunomodulatory role of rPdx1 in the NOD mice corroborates the above findings

(Fig. 4-2, 4-20).

The previously known autoantigens such as Insulin(125), GAD(11), Hsp90(12), and Znt80

(130) have guided in designing effective therapeutic strategies using soluble antigens

(11;14;20;131). On similar lines, treatment of NOD mice with soluble rPdx1 serves as an

effective immunotherapeutic strategy to prevent the onset of diabetes. The treatment with rPdx1

over a period of time could possibly induce immune tolerance as observed in immunotherapeutic

strategies using other soluble autoantigens. The induction of tolerance could be due to the

stimulation of specialized subpopulation of T cells (CD4+, CD25+, FoxP3+ regulatory T cells)

(28;132) resulting in a shift in the balance from Th1 pro-inflammatory to Th2 anti-inflammatory

cytokine release (133). Previous studies have shown that the administration of soluble

autoantigens such as GAD (16) and Hsp60 (20) leads to shift in immune response towards Th2-

Page 91: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

91

dependent T-cell cytokine release with preferential production of IL-4, IL-5, and IL-10

production (127;128). We expect a similar phenomenon with the rPdx1 immunotherapy.

The other mechanism by which immune tolerance may be achieved is likely by clonal

deletion or anergy, where frequent administration of the soluble antigen will familiarize the

immune system to recognize Pdx1 as self antigen, thus resulting in peripheral clonal deletion.

(131;134). The antigen induced tolerance was previously studied using evidence in TCR

transgenic mice models (127) hence future studies with Pdx1 antigen specific TCR would reveal

the role of Pdx1 antigen in the pathogenesis of type 1 diabetes.

The treatment of NOD mice with rPdx-mut protein (with no biological function but intact

immunologically active component (Fig. 4-19) has demonstrated incidence of diabetes similar to

the wild type rPdx1 (Fig. 4-20). This suggests the predominant immunomodulatory role of rPdx1

to prevent the onset of type 1 diabetes. However, the contribution of rPdx1 in promoting beta cell

regeneration should not be ignored as in case of prevention trials, there is minimal requirement

for regeneration component for the maintenance of islet mass. IPCs derived by transdifferen-

tiation of liver hepatocytes should play a minor role in contributing towards glucose sensitive

insulin secretion.

In conclusion, this study shows that Pdx1 is a novel auto antigen that could have an active

role in the pathogenesis of type 1 diabetes. Pdx1 autoantibodies could serve as an effective

biomarker for prediction of the disease and Pdx1 antigen-based immunotherapy could serve as

an effective strategy for prevention of the onset of diabetes.

Page 92: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

92

1

0 5 10 15 20 25 30 35 40

23

PDx1 treatment (100ug/mouse/day.

1

2

3

Pre-diabetic stage.

IPGTT. Insulin levels, Histology, Lymphocyte assay

Age in weeks

1

0 5 10 15 20 25 30 35 40

23

PDx1 treatment (100ug/mouse/day.

1

2

3

Pre-diabetic stage.

IPGTT. Insulin levels, Histology, Lymphocyte assay

1

0 5 10 15 20 25 30 35 40

23

PDx1 treatment (100ug/mouse/day.

1

2

3

Pre-diabetic stage.

IPGTT. Insulin levels, Histology, Lymphocyte assay

0 5 10 15 20 25 30 35 40

23

PDx1 treatment (100ug/mouse/day.

1

2

3

Pre-diabetic stage.

IPGTT. Insulin levels, Histology, Lymphocyte assay

Age in weeks

Figure 4-1. Time line for the experimental plan for treatment of NOD mice. Ten-week-old

female NOD mice were injected i.p. daily with 0.1 mg of rPdx1 (n=20) for 12 weeks or with saline (n=20). Blood glucose and body weight were monitored regularly. IPGTT, serum insulin levels, morphology and tissue insulin content were examined in NOD mice between 20-25 weeks of age.

Page 93: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

93

Incidence of Diabetes

0

20

40

60

80

100

1 3 5 7 9 11 13 15 17 19 21 23 25 27 29 31Time in weeks

Perc

enta

ge D

iabe

tic

Pdx1 group Ctrl group

10 15 20 25 30 35 40

Pdx1 treatment

Incidence of Diabetes

0

20

40

60

80

100

1 3 5 7 9 11 13 15 17 19 21 23 25 27 29 31Time in weeks

Perc

enta

ge D

iabe

tic

Pdx1 group Ctrl group

10 15 20 25 30 35 40

Incidence of Diabetes

0

20

40

60

80

100

1 3 5 7 9 11 13 15 17 19 21 23 25 27 29 31Time in weeks

Perc

enta

ge D

iabe

tic

Pdx1 group Ctrl group

10 15 20 25 30 35 40

Incidence of Diabetes

0

20

40

60

80

100

1 3 5 7 9 11 13 15 17 19 21 23 25 27 29 31Time in weeks

Perc

enta

ge D

iabe

tic

Pdx1 group Ctrl group

Incidence of Diabetes

0

20

40

60

80

100

1 3 5 7 9 11 13 15 17 19 21 23 25 27 29 31Time in weeks

Perc

enta

ge D

iabe

tic

Pdx1 group Ctrl group

10 15 20 25 30 35 40

Pdx1 treatment

Figure 4-2. Effect of rPdx1 (12-weeks) on the onset of diabetes in NOD mice. Daily treatment of pre-diabetic female NOD mice with rPdx1 for 12 weeks resulted in significant protection of the mice from the onset of diabetes (90% at 33 weeks, and 80% at 39 weeks, and 70% at 40 weeks). The saline control mice became 95% hyperglycemic (diabetic) at age of 25 week.

Page 94: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

94

Figure 4-3. Blood glucose levels of rPdx1 (12-weeks) treated NOD mice. Daily treatment of pre-diabetic female NOD mice with rPdx1 for 12 weeks resulted in maintenance of normoglycemia in 70% of mice at 40 weeks. 3 out of 10 mice became hyperglycemic (>250 mg/dL) at age 17, 35 and 40 weeks (Panel A). The saline- treated control mice became 95% hyperglycemic (diabetic) at the age of 25 weeks (Panel B)

A

B

Page 95: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

95

Figure. 4-4. Body weights of rPdx1 (12-weeks) treated NOD mice. Daily treatment of pre-diabetic female NOD mice with rPdx1 for 12 weeks resulted in increase in body weight (Panel A). The saline-treated control mice progressively lost body weights or could not maintain their body weights (Panel B ).

0

5

10

15

20

25

30

10 12 14 16 18 20 22 24 26 28 30 32 34 36 38 40

1L 1R

1B 1N

2L 2R

2B 2N

3L 3R

0

5

10

15

20

25

30

10 11 12 13 14 15 16 17 18 19 20 21 22 23 24

6L

6R

6B

6N

7L

7R

7B

7N

8L

8R

8B

8N

9L

9R

9B

9N

10L

10R

10B

10N

Time points in weeks

Time points in weeks

Bod

y w

eigh

tsB

ody

wei

ghts

0

5

10

15

20

25

30

10 12 14 16 18 20 22 24 26 28 30 32 34 36 38 40

1L 1R

1B 1N

2L 2R

2B 2N

3L 3R

0

5

10

15

20

25

30

10 11 12 13 14 15 16 17 18 19 20 21 22 23 24

6L

6R

6B

6N

7L

7R

7B

7N

8L

8R

8B

8N

9L

9R

9B

9N

10L

10R

10B

10N

0

5

10

15

20

25

30

10 12 14 16 18 20 22 24 26 28 30 32 34 36 38 40

1L 1R

1B 1N

2L 2R

2B 2N

3L 3R

0

5

10

15

20

25

30

10 11 12 13 14 15 16 17 18 19 20 21 22 23 24

6L

6R

6B

6N

7L

7R

7B

7N

8L

8R

8B

8N

9L

9R

9B

9N

10L

10R

10B

10N

Time points in weeks

Time points in weeks

Bod

y w

eigh

tsB

ody

wei

ghts

A

B

Page 96: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

96

Pdx1 treatment (4 wks)

0

20

40

60

80

100

1 2 3 4 5 6 7 8 9 101112131415161718192021

Perc

enta

ge D

iabet

ic

Pdx1 (4 wks) Ctrl

Pdx1 treatment

10 15 20 25 30

Pdx1 treatment (4 wks)

0

20

40

60

80

100

1 2 3 4 5 6 7 8 9 101112131415161718192021

Perc

enta

ge D

iabet

ic

Pdx1 (4 wks) Ctrl

Pdx1 treatment

Pdx1 treatment (4 wks)

0

20

40

60

80

100

1 2 3 4 5 6 7 8 9 101112131415161718192021

Perc

enta

ge D

iabet

ic

Pdx1 (4 wks) Ctrl

Pdx1 treatment (4 wks)

0

20

40

60

80

100

1 2 3 4 5 6 7 8 9 101112131415161718192021

Perc

enta

ge D

iabet

ic

Pdx1 (4 wks) Ctrl

Pdx1 treatmentPdx1 treatment

10 15 20 25 30

Figure 4-5. Effect of short term rPdx1treatment (4-weeks) on the onset of diabetes in NOD mice. To determine if short-term treatment with rPdx1 could prevent the onset of diabetes, ten-week-old female NOD mice were administered 0.1 mg rPdx1 i.p. for 4 weeks (n=10). Daily treatment of pre-diabetic female NOD mice with rPdx1 for 4 weeks resulted in significant protection of the mice from the onset of diabetes (70% at 30 weeks).

Page 97: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

97

Figure 4-6. Blood glucose levels of rPdx1 (4-weeks) treated NOD mice. Daily treatment of pre-diabetic female NOD mice with rPdx1 for 4 weeks resulted in maintenance of normoglycemia in 70% of mice at 30 weeks.

Page 98: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

98

Pdx1 treatment (8 wks)

0

20

40

60

80

100

1 2 3 4 5 6 7 8 9 101112131415161718192021

Perc

entag

e Diab

etic

Pdx1 (8wks) Ctrl

10 15 20 25 30

Pdx1 treatment

Pdx1 treatment (8 wks)

0

20

40

60

80

100

1 2 3 4 5 6 7 8 9 101112131415161718192021

Perc

entag

e Diab

etic

Pdx1 (8wks) Ctrl

10 15 20 25 30

Pdx1 treatment (8 wks)

0

20

40

60

80

100

1 2 3 4 5 6 7 8 9 101112131415161718192021

Perc

entag

e Diab

etic

Pdx1 (8wks) Ctrl

10 15 20 25 30

Pdx1 treatment (8 wks)

0

20

40

60

80

100

1 2 3 4 5 6 7 8 9 101112131415161718192021

Perc

entag

e Diab

etic

Pdx1 (8wks) Ctrl

10 15 20 25 30

Pdx1 treatment (8 wks)

0

20

40

60

80

100

1 2 3 4 5 6 7 8 9 101112131415161718192021

Perc

entag

e Diab

etic

Pdx1 (8wks) Ctrl

10 15 20 25 30

Pdx1 treatment

Figure 4-7. Effect of short term rPdx1treatment (8-weeks) on the onset of diabetes in NOD mice. To determine if short-term treatment with rPdx1 could prevent the onset of diabetes, ten-week-old female NOD mice were administered 0.1 mg rPdx1 i.p. for 8 weeks (n=10). 70% of the rPdx1-treated mice maintained normoglycemia upto 30 weeks.

Page 99: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

99

Figure 4-8. Blood glucose levels of rPdx1 (8-weeks) treated NOD mice. Daily treatment of pre-diabetic female NOD mice with rPdx1 for 8 weeks resulted in maintenance of normoglycemia in 70% of mice at 30 weeks. (Panel A). The saline- treated control mice became 90% hyperglycemic (diabetic) at the age of 30 weeks (Panel B).

A

B

A

B

Page 100: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

100

IPGTT

050

100150200250300350400450

0 5 15 30 60 120

Time in minutes

Blo

od

Glu

co

se (

mg

/dl)

Pdx1Ctrl

Figure 4-9. Intraperitoneal glucose tolerance test. IPGTT (n=4/group). Mice were injected with 2 mg/g of glucose i.p. Blood glucose was monitored at 0, 5, 15, 30, 60, 120 min. The rPdx1 treated mice at 22 weeks demonstrated better glucose handling ability than the age-matched diabetic control mice.

Page 101: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

101

Serum Insulin levels

0123456

Normal Pdx1 Ctrl

Ser

um

In

suli

n (

ug

/L)

* *

Serum Insulin levels

0123456

Normal Pdx1 Ctrl

Ser

um

In

suli

n (

ug

/L)

* ** *

Figure 4-10. Serum insulin measurements as determined by ELISA. Glucose-stimulated (15 min) serum samples were collected from rPdx1 treated NOD mice (22 wks), diabetic control NOD mice (22 wks), and normal NOD mice (9 wks). rPdx1-treated mice have an 8-fold higher serum insulin level compared with the diabetic control mice. (n=4/group) ** = P<0.05; Student’s t-test.

Page 102: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

102

Figure 4-11. Haematoxylin & Eosin staining and immunohistochemistry of pancreatic tissue sections. H & E staining of pancreatic tissue sections from rPdx1- treated mice showed many large islets with numerous insulin-stained beta cells. On the contrary, control mice had much fewer and smaller islets and most of the islet cells contained glucagon-positive cells.

Page 103: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

103

Pancreatic insulin content

0

500

1000

1500

2000

2500

In

su

lin

co

nte

nt

(n

g)

Normal mice

Pdx1 group

Ctrl group

Liver insulin content

0

100

200

300

400

500

600

700

800

Ins

ulin

co

nte

nt

(ng

/g)

Ctrl

Pdx1

* *

Liver insulin content

0

100

200

300

400

500

600

700

800

Ins

ulin

co

nte

nt

(ng

/g)

Ctrl

Pdx1

* ** *

Figure 4-12. Pancreas and liver tissue insulin levels as determined by ELISA. Tissue insulin was extracted from whole organs of pancreases and livers, measured by ELISA, and normalized with wet tissue weight. Insulin measurements in the pancreas (Left) and liver (right) extracts of rPdx1- treated mice (n=4) are significantly higher than the control diabetic mice. ** = P<0.05; Student’s t-test.

A B

Page 104: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

104

Figure 4-13. Detection of Pdx1 antibodies by ELISA in the NOD mice. Serum samples were collected from both female control NOD and pre- and post-Pdx1-treated NOD mice at indicated ages. Presence of anti-Pdx1 antibodies was determined by ELISA. High levels of Pdx1 antibodies were detected in Pdx1-treated mice. However, control mice also had various levels of anti-Pdx1 autoantibodies (A) (panel A). To determine if the antibodies are specific to NOD autoimmune diabetic mouse model, serum samples were collected from other mouse strains such as NOD-SCID, C57/B6 and Balb/C (Panel B). The anti-Pdx1 autoantibodies were highly specific to NOD strain as determined by ELISA.

A

B

Page 105: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

105

rPdx1

Balb/c

Western blottingCoomassieblue gel

Pdx1 AA( -)Pdx1 AA(+) ELISAPdx1 AA(-)

NOD mouse sera

1 2 3

rPdx1

rPdx1

Balb/c

Western blottingCoomassieblue gel

Pdx1 AA( -)Pdx1 AA(+) ELISAPdx1 AA(-)

NOD mouse sera

1 2 3

rPdx1

Figure 4-14. Confirmation of anti-Pdx1 antibody in the NOD mice by western blotting. Anti-Pdx1 antibody was confirmed by western blot using purified Pdx1 protein as antigen. The membrane was probed with control serum with high-titer for Pdx1 antibody (lane 2) and serum with basal titer (Lane 3). A mouse serum from BALB/c mice was used as negative control for Pdx1 antibody.

Page 106: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

106

Figure 4-15. Determination of antigenic epitope by western blot. Illustration of full-length Pdx1 and truncated proteins. A mix of full-length and truncated proteins was run on SDS-gel. The membrane was probed with either rabbit polyclonal serum, or sera from NOD mice (6L, 4L). The polyclonal sera detected all the proteins bands while the NOD sera detected only the full-length indicating that the epitope is in the C-terminal 83 amino acids (panel A). The C-terminal 83 amino acid portion was expressed as GST fusion protein. Panel B is Coomassie blue stained gel and the results of blotting that were using two autoantibody A(+) mouse sera. Arrows indicate the positions of Pdx1, P83-GST or GST.

A

B

Page 107: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

107

Figure.4-16. Relationship between Pdx1 autoantibody levels and onset of diabetes. 5-week old female NOD mice were purchased (n=20) and serum samples were collected biweekly. Pd1 autoantibodies were detected by ELISA and expressed in OD values at 450 nM. Blood glucose levels were monitored weekly via tail vein snipping. Three representative mice data is shown here.

Page 108: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

108

T-cell proliferation assay

0

2000

4000

6000

8000

10000

12000

14000

16000

18000

None CD3 GST Ins Pdx1 P83

cpm Pdx1

Ctrl

Figure 4-17. Antigen-stimulated lymphocyte proliferation. Splenocytes were harvested from the rPdx1-treated and control mice. One million cells were seeded into a 96-well plate in triplicates. The cells were treated with anti-CD3, insulin, and Pdx1 for 48 hours followed by incubation with [3H] thymidine for an additional 24 hours. Splenic T-cell lymphocyte proliferation was stimulated in control mice by rPdx1 and insulin, whereas antigen-stimulated splenic T-cell lymphocyte proliferation was significantly lower in 4-week-rPdx1-treated mice compared with control mice.

Page 109: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

109

Figure 4-18. Transfer of splenocytes from donor BDC2.5 mice to the recipient NOD mice. The transfer of CFSE labeled splenocytes from donor BDC2.5 mice into the recipient NOD mice pretreated with rPdx1 or saline (4 weeks) demonstrated a decrease in donor-derived CD4-T-cell proliferation in the draining pancreatic lymph nodes of rPdx1 treated mice.

Page 110: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

110

Pdx11 283

188 203

1 79

transactivation

146 206homeobox

191 196

DNA Binding

Antennapedia domain

Pdx11 283

Pdx1Pdx11 283

188 203188 203

1 79

transactivation

1 79

transactivation

146 206homeobox

191 196

DNA Binding

Antennapedia domain

146 206homeobox

146 206homeobox

191 196

DNA Binding

191 196

DNA Binding

Antennapedia domain

Figure 4-19. Illustration of Pdx1 and mutant Pdx1proteins. A short 16 amino acid sequence was deleted between amino acids 188-203 of the Pdx1 protein, which eliminated both DNA binding and the antennapedia-like protein transduction domain. Thus a biologically inactive mutant Pdx1 protein was produced.

Page 111: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

111

mut-Pdx1 treatment (8 wks)

0

20

40

60

80

100

1 2 3 4 5 6 7 8 9 101112131415161718192021

Perc

enta

ge D

iabe

tic

mut-Pdx1 (8wks) Ctrl Pdx1(8 Wks)

10 15 20 25 30

mut-Pdx1 treatment (8 wks)

0

20

40

60

80

100

1 2 3 4 5 6 7 8 9 101112131415161718192021

Perc

enta

ge D

iabe

tic

mut-Pdx1 (8wks) Ctrl Pdx1(8 Wks)

10 15 20 25 30

Figure 4-20. Effects of mutant-Pdx1 on the onset of diabetes. Ten-week-old female NOD mice were treated with full-length Pdx1 (n=10), mutant Pdx1 (n=10), or saline (n=10). Incidence of diabetes was monitored until 30 weeks. By 30 wks, both full-length Pdx1- and mut-Pdx1 treatments prevented/delayed the onset of diabetes in 70% of NOD mice whereas 80% of the control mice became diabetic.

Page 112: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

112

CHAPTER 5 MOLECULAR MECHANISM OF THE ROLE OF PDX1 IN BETA CELL PROLIFERATION

Introduction

Pancreatic and duodenal homeobox 1(Pdx1) is a transcriptional factor that plays a crucial

role in pancreas development and beta cell differentiation during embryogenesis(35;36). It plays

a major role in beta cell maturation, maintenance, and regulation of gene expression in the beta

cell (35;37;38). During embryogenesis, Pdx1 gene expression appears in the duodenum early in

development while in the adult pancreas, Pdx1 gene expression is predominantly restricted to the

insulin-producing islet beta cells and a subset of somatostatin producing delta cells(39-41). Mice

with a null mutant of Pdx1 resulted in agenesis of pancreas with hyperglycemia and could not

survive after birth(35). Pdx1 heterozygous mutant mice failed to increase beta-cell mass in

response to insulin resistance (112). Pdx1 has been found to play key role in beta cell neogenesis

and insulin production(38). Pdx1 is a homeodomain transcription factor that binds to specific

sequences of its target genes and activates them by recruiting other co-transcriptional factors

such as NeuroD, MafA, and Pax4. Conditional expression of Pdx1 in transgenic mice

demonstrated its role in beta cell regeneration(37). Previous studies using growth factors such as

GLP-1, extendin-4, and INGAP have shown that beta cell regeneration is accompanied by

increase in levels of Pdx1 protein (98;135). Our previous studies using the STZ-induced diabetic

mouse model demonstrated that rPdx1 treatment promoted beta cell proliferation(123). Studies

on partial pancreatectomy have demonstrated an increased Pdx1 expression is associated with

beta cell proliferation(45;108). However the molecular mechanisms of the role of Pdx1 in beta

cell regeneration is not known yet.

Several studies were conducted to understand the functional role of cell cycle proteins of

the beta cells which include cyclins, cyclin-dependent kinases, and cyclin-dependent kinase

Page 113: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

113

inhibitors(136;137). There is an age dependent decline in the beta cell self-renewal capacity

(138) due to the activation of Ink inhibitors with increasing age (139). Previous studies have

shown that cyclin A2 (135), cyclin B1 (140), cyclin E1 (141), and cyclin D2 (142) are involved

in beta cell proliferation. However, the association between Pdx1 protein and cyclins in beta cell

regeneration is not established yet.

Materials and Methods

Cell Culture

INS-1 cells were grown in RPMI 1640 (Sigma) (430 mL) with 10% fetal bovine serum

supplemented with 5 mL of HEPES (1M), 5 mL of L-glutamine (200 mM), 5 mL of Sodium

pyruvate (100 mM), 5 mL of Pen/Strep, 454 µL of 2-Mercaptoethanol (1000x) and 2 mL

Kanamycin (50 mg/mL) per each bottle (430 mL) of media. 3T3 cells were grown in Dulbecco's

Modified Eagle Medium (Sigma) supplemented with 10% FBS (55 mL) and 2 mL Kanamycin

(50 mg/mL).

Design of siRNA for the Pdx1 Transcript

21-mer siRNA was designed for specific binding to mRNA at three different locations

(Fig. 5-1). Sense Strand (A): GGAAGAUAAGAAACGUAGUUU mRNA loc. 712

Sense Strand (B): GGAUCAUGAGGCUUAACCUUU mRNA loc. 1033

Sense Strand (C): CGAGCAAUCUAAGGUUGAGUU mRNA loc. 1336

siRNA Mediated Knockdown of Pdx1 Transcript in INS-1 Cells

The INS1 cells (2 x 105 cells) were grown upto 60% confluence in 6 well plates at 37° C

in a 5% CO2 incubator. For each transfection, 8 μl of siRNA (Santa cruz Biotechnology, CA)

was diluted (i.e. 1 μg or 80 pmols siRNA) into 100 μl serum free minimal medium (Gibco).

For each transfection, 2 μl of Lipofectamine reagent (Invitrogen) was diluted in 100 μl serum

free minimal medium in a separate tube. Next the siRNA solution was added directly to the

Page 114: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

114

diluted transfection reagent. The solution was mixed gently by pipetting the solution up and

down and incubated for 30 minutes at room temperature. The cells were washed once with 2 mL

of serum free minimal medium. The medium was aspirated before proceeding to the next step.

For each transfection, 0.8 mL of siRNA transfection mix was added to each well. The cells were

incubated for 5-7 hours at 37° C in a 5% CO2 incubator. Later, the transfection mix was removed

and replaced with normal growth medium. The cells were harvested at the end of 48, 72, and 96

hours.

Western Blotting with anti-Pdx1 and anti-tubulin Antibody

The cellular protein lysates from 6 well plates were collected using cell lysis RIPA buffer

after 72 and 96 hours of treatment with siRNA. The cellular total protein content was quantified

using spectrophotometer. 50 µg of protein (from cells treated with Pdx1 siRNA and Ctrl siRNA)

was loaded onto SDS-gel. The gel was run at 80V until the dye front migrated through the gel.

After transfer and overnight blocking (1% milk powder in PBST), the membrane was incubated

with rabbit polyclonal anti-Pdx1 antibody (1:1000) for 1hr at room temperature. After washing

the membrane with PBS for 15 min, it was incubated with anti-rabbit IgG conjugated to HRP

(1:20,000) (Amersham Biosciences) for 1 hour at room temperature. After development, the

membrane was stripped-off with strip buffer (2% SDS, 100 mM beta-mercaptoethanol, 50 mM

Tris, pH 6.8) by incubating at 70°C for 30 min. The membrane was then blocked over night and

incubated with anti-tubulin antibody (1:5000; Abcam) followed by anti-mouse-IgG conjugated

with HRP (1:5000; Cayman Chemicals).

BrdU Proliferation Assay

Four thousand cells were plated in 96 well plates in triplicates and allowed to grow until

they became 50% confluent. 0.75 µg of siRNA was added to each well in triplicates and the

cells were incubated for 48 or 72 hours depending on the preferred harvesting time. 50 µM of

Page 115: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

115

BrdU was added before the last 18 hours of incubation. The cells were fixed with 4%

paraformaldehyde in vitro for 30 min at 4ºC. Next, the samples were incubated in 0.1 M PBS

(pH 7.4) + 1% Triton X-100 + 1M Glycine + 5% goat serum for 1 hour prior to overnight

incubation at 4°C with mouse anti-BrdU (1:100; BD biosciences). The plate was washed gently

thrice with PBS, followed by incubation with secondary anti-mouse IgG conjugated with HRP

(1:4000; Cayman Chemical Laboratories). The wells were then incubated with TMB solution

(Cell signaling) for 10 min. The reaction was stopped with stop solution (Cell signaling). The

plate was read at 450 nm using 650 nm as reference wavelength.

Real Time PCR

cDNA from INS-1 cells (both Pdx1 siRNA and Ctrl siRNA treated) was subjected to three

independent PCR reactions. Each reaction was performed in duplicate in a thermocycler

sequence detection system (DNA engine opticon 2; MJ Research, Springfield, MO) using SYBR

green (Qiagen, Valencia, CA). RNA was extracted in RNase free tubes using Trizol reagent

(Invitrogen). cDNA was synthesized from 2 µg of total RNA using Superscript reverse

transcriptase enzyme (Invitrogen) with random hexamer primers according to the manufacturer’s

protocol. The expected amplified product using specific primers listed in Table 5-1, was

confirmed by agarose gel electrophoresis. Conditions for real-time PCR were as follows: after

initial denaturation at 95°C for 15 min to activate the enzyme, 35 cycles of PCR (denaturation

0.5 min at 94°C, annealing 0.5 min at 58°C, and elongation 0.5 min at 72°C with a final

extension for 5 min at 72°C) were carried out. Each gene was tested three times. The single band

amplification was verified by observing the melting curve and by agarose gel electrophoresis.

Relative gene expression in INS-1 cells treated by Pdx1 siRNA and Ctrl siRNA was calculated

by the 2ΔΔCt method. More details about calculations were discussed in chapter 3.

Page 116: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

116

Construction of pGL4.10-Cyclin B1 Promoter-Luciferase Vector

The -991 bp sequence of the human cyclin B1 promoter was amplified using following

primers Forward- CTCGAGGCTGGTGTGTTTTGAGGAGTA; Reverse-

AGATCTCCAAGGACCTACACC-CAGCAG. Human genomic DNA was used as a template for

PCR amplification. Restriction sites Xho1 and BglII were introduced using forward and reverse

primers respectively. Annealing temperature was programmed at 56°C for 30 sec and the

amplification for 30 cycles. The PCR product (~1000 bp) was cloned into the XhoI and BglII

sites of the pGL4.10 luciferase reporter vector. The shorter version (-551 bp cyclin B1 cloned

into pGL2 vector) was a kind gift from Dr. Karen Katula, Univ. of North Carolina, Greensboro,

NC. The Nkx6.1 promoter with luciferase reporter and CMV-human Nkx6.1 constructs were

obtained as gift from Dr. Raghavendra Mirmira’s lab and CMV-human Pdx1 was purchased

from Open biosystems, AL.

Analysis of Promoter Constructs for Activation by Pdx1 or Nkx6.1 using Luciferase Reporter Assays

NIH 3T3 cells were grown in 6 well plates until they became 50% confluent. For each

well, the cells were co-transfected with 0.5 µg of Tk-renilla plasmid (as internal control) and 1

µg of Nkx6.1/ cyclin B1 promoter driven luciferase plasmid. The cells were treated with

0.5 µg of test plasmids (CMV-Pdx1, CMV-Nkx6.1) or pcDNA3 vector control. The cells were

harvested after 24 hrs in passive lysis buffer (Promega). The lysates were analyzed using dual

luciferase assay kit (Promega). Details of the transfection protocol and luciferase activity

measurements were described in chapter 2.

Page 117: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

117

Results

Phenotype of INS-1 Cells Treated with Pdx1 siRNA

INS-1 cells were treated with 1 µg of either Ctrl siRNA or Pdx1 siRNA. A phenotype

change was observed in cells treated with Pdx1 siRNA from flat and polygonal to round and

smaller cells as shown in Fig. 5.2. The cells treated with Ctrl siRNA remained flat and

polygonal. The phenotypes become more pronounced from 24 hours to 72 hours. This indicates

the possibility of cell cycle arrest mediated by Pdx1 siRNA.

siRNA-mediated Knockdown of Pdx1 Transcripts in INS-1 Cells

To study the effects of Pdx1 protein on cell cycle regulation, Pdx1 gene expression was

depleted in INS-1 cells using siRNA against Pdx1 transcript. The decrease in gene expression

was of Pdx1 was observed at both the transcript and protein level using real time PCR (Fig. 5.5)

and western blotting (Fig. 5.3) respectively. Both real time PCR and western blotting confirmed

more than 70% decrease in the gene expression.

Decrease in Cell Proliferation of Pdx1 Knockdown Cells

In order to study if knockdown of Pdx1 has any significant effect on cell proliferation, an

in vitro proliferation assay was conducted using the BrdU incorporation method. INS-1 cells

were treated with 1 µg of Pdx1 siRNA or Ctrl siRNA for a period of 48 or 72 hours. The final 18

hours were incubated with 50 μM BrdU. The BrdU proliferation assay demonstrated that the

Pdx1 siRNA treated cells showed decrease in the proliferation to 59.84% and 70% at 48 and 72

hours respectively as shown in Fig. 5.4. This validates that the knockdown of Pdx1 gene

expression indeed reduced proliferation of INS-cells, thus demonstrating that Pdx1 is essential

for the beta cell proliferation.

Page 118: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

118

Analysis of the Gene Expression of Beta Cell Specific transcription factors and Cell Cycle Genes

The decrease in the cell proliferation of Pdx1 knockdown cells could be attributed to

decreased activity of cell cycle proteins and other beta cell specific transcription factors involved

in cell cycle progression, perhaps at the gene expression level. In order to study the effects of

Pdx1 siRNA, specific primers were designed to observe the downstream effects on beta cell

specific transcription factors and cyclin genes. In order to study the association of Pdx1 protein

with cell cycle regulators, cDNA was synthesized from the RNA extracted from the cells treated

with either Pdx1 siRNA or ctrl siRNA. The real time PCR assay demonstrated a significant

decrease in the gene expression of beta cell transcription factors such as Nkx6.1 (50%) and

cyclin genes such as cyclin B1 (40%) and cyclin E1 (65%) (Fig. 5-5).

Analysis of Cyclin B1 Promoter using CMV-Pdx1

Bioinformatic analysis of the human cyclin B1 promoter has revealed multiple potential

Pdx1 protein binding sites. In order to verify this observation experimentally, NIH-3T3 mouse

fibroblast cells were transfected with the 1 µg of cyclin B1 promoter-luciferase reporter vector.

These cells were simultaneously co-transfected with pcDNA3 null vector or CMV-Pdx1 plasmid.

We observed no change in the activity of cyclin B1 promoter when co-transfected with CMV-

Pdx1 (Fig. 5-6) suggesting that Pdx1 protein may not interact directly with cyclin B1 promoter.

Nkx6.1 Activation of Cyclin B1 Promoter

Bioinformatic analysis of the human cyclin B1 promoter has revealed potential binding

sites for Nkx6.1. We also observed a decrease in the transcript levels of Nkx6.1 in Pdx1

knockdown cells (Fig.5-5). We therefore sought to determine the association between Nkx6.1

and cyclin B1 promoter. Cyclin B1 promoter (-991bp) luciferase reporter vector was co-

transfected with either pcDNA3 null vector or CMV-Nkx6.1 plasmid. A 5 fold increase in cyclin

Page 119: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

119

B1 promoter activity was observed when co-transfected with CMV-Nkx6.1 plasmid (Fig. 5-7)

demonstrating that Nkx6.1 protein could bind to the cyclin B1 promoter. A truncated version of

the cyclin B1 promoter (-551bp) with no putative binding site for Nkx6.1, when co-transfected

with either pcDNA3 or with CMV-Nkx6.1 plasmid, did not demonstrate any increase in activity

(Fig. 5-7).

Pdx1 Activation of Nkx6.1 Promoter

We determined that Pdx1 did not activate cyclin B1 directly, however, Nkx6.1 could

directly activate cyclin B1 promoter. In order to determine the association between Pdx1 and

Nkx6.1, the Nkx 6.1 promoter was studied using luciferase assay in NIH 3T3 fibroblast cells.

Nkx6.1 promoter-luciferase reporter vector was co-transfected with either pcDNA3 null vector

or CMV-Pdx1 plasmid. There was a 2.5 fold increase in the Nkx6.1 promoter activity, when co-

transfected with Pdx1 plasmid demonstrating that Pdx1 protein activates Nkx6.1 promoter (Fig.

5-8).

Discussion

Pdx1 is a beta cell specific transcriptional factor required for the regeneration and

maintenance of beta cell in the adult stage. During the post natal stage, beta cells within a normal

islet have a finite life span, with slow turn over and with majority of the cells in the resting phase

of cell cycle. However, the dynamics of beta cell proliferation dramatically change upon injury

or mitogenic stimulation(143). Proliferation of beta cells is triggered by mitogens that in turn

initiate an intracellular signaling between set of beta cell specific transcriptional factors and cell

cycle machinery. One such important signaling pathway is the insulin signaling pathway where

insulin binds to the insulin receptor that leads to phosphorylation of tyrosine residues. This

further leads to the activation of the PI-3 kinase-Akt signaling pathway which in turn

phosphorylates FoxO protein and results in the activation of Pdx1 gene expression.

Page 120: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

120

Previous studies on partial pancreatectomy have demonstrated that an increased Pdx1

expression is associated with beta cell proliferation (45). Treatment of diabetic animal models

with mitogenic agents such as GLP-1 (121;122), Extendin4 (120), INGAP (98) or rPdx1(123),

results in upregulation of Pdx1. All these studies suggest that local upregulation of Pdx1 above a

threshold level is necessary to activate downstream events that regulate beta cell proliferation.

The question that remains to be investigated is how Pdx1 interacts with beta cell cycle

machinery to induce proliferation. Pdx1 is a homeodomain transcription factor that interacts

with other transcription factors and cofactors such as E47/Pan1, BETA2/NeuroD, p300, hnf1,

and Pax6 (144-148) to perform specific functions within beta cell. The real time PCR has

demonstrated a significant decrease in the levels of Nkx6.1, cyclin B1, and cyclin E1 in the Pdx1

knockdown cells (Fig. 5-5). However, the other cyclins such as cyclin A2 and cyclin D1 may be

playing a role in beta cell regeneration through multiple signaling pathways involving other beta

cell specific transcription factors. Cyclin B1 is the regulatory subunit of serine/threonine kinase

Cdc2 (cdk1) and is essential for the entry into mitosis. Our studies have shown that Pdx1 does

not have a direct interaction with cyclin B1 (Fig. 5-6), however, Nkx6.1 can activate cyclin B1

promoter (Fig. 5-7). At the upstream of Nkx6.1, Pdx1 can activate Nkx6.1 promoter activity

(Fig-5-8). These observations may be one among several other cell cycle events that could be

occurring within the islet beta cells. This does not however, rule out the possibility of

involvement of other signaling pathways and transcriptional factors in beta cell proliferation.

We conclude that beta cell proliferation is induced by local upregulation of Pdx1 protein

by upstream mitogenic signals. Pdx1 protein further autoregulates its own gene expression which

results in amplified levels of Pdx1 protein (113;123). Once the levels reach above the threshold

level, Pdx1 activates Nkx6.1 gene expression that results in increased levels of Nkx6.1 protein

Page 121: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

121

which in turn interacts with cyclin B1 promoter to trigger cell entry into the mitotic phase (Fig.

5-9).

Page 122: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

122

Table 5-1. Real time PCR Primer name, sequences, size, GenBank #, and PCR condition Genes Forward primer Reverse primer PCR

size GenBank Acc. No

Tm (°C)

Cycle No.

Actin ATTGAACACGGCATTGTCACCA CTGGATGGCTACGTACATGGCT 200 NM_031144.2

58 35

Nkx6.1 AAACGAAGTACTTGGCAGGACC TAATCGTCGTCGTCCTCCTCGTT 167 NM_031737.1

58 35

PDX1 TCCACCAAAGCTCACGCGTGG GAATTCCTTCTCCAGCTCCAGCAG 190 NM_022852.3

58 35

Cyclin A2 TGAAGAGGCAACCAGACATCAC AGCCAAATGCAGGGTCTCAT 178 NM_053702.2

58 35

Cyclin B1 TGAGCCTGAGCCTGAACCTG CTGCATCTACATCACTCACTGC 200 NM_171991.2

58 35

Cyclin D2 CTGCAGAACCTGTTGACTATCGAGC GGTAATTCATGGCCAGAGGAAAG 194 NM_022267.1

58 35

Cyclin E1 GCAATAGAGAAGAGGTCTGGAGGAT GTCCTGTGCCAAGTAGAATGTCTCT 182 NC_005100 58 35

Page 123: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

123

Figures

Figure 5-1. Location of 21 mer siRNAs on Pdx1 mRNA transcript.

(A): GGAAGAUAAGAAACGUAGUUU mRNA loc. 712

(B): GGAUCAUGAGGCUUAACCUUU mRNA loc. 1033

(C): CGAGCAAUCUAAGGUUGAGUU mRNA loc. 1336

Page 124: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

124

Figure 5-2. Phenotype of INS1 cells treated with Pdx1 siRNA. The INS1 cells grown in serum free minimal medium were treated with Ctrl siRNA and Pdx1 siRNA for 48 hours. The cells treated with Pdx1 siRNA morphologically appeared round and small while the Ctrl siRNA treated cells appeared flat and polygonal (normal).

Page 125: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

125

Figure 5-3. Confirmation of Pdx1 knockdown using western blotting. INS-1 cells treated with ctrl siRNA and Pdx1 siRNA for 72 and 96 hours. The cell lysates were analyzed for Pdx1 protein levels using anti-Pdx1 antibody and anti-tubulin antibody as loading control.

1 2 3 4

Page 126: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

126

Brdu proliferation assay

100 100

59.8470

020

406080

100120

48 72

Time points (hrs)

Pe

rce

nta

ge

pro

life

rati

on

Ctrl siRNA

Pdx1 siRNA

Figure 5-4. Measurement of INS-1 cell proliferation using BrdU incorporation. INS-1 cells were treated with Ctrl or Pdx1 siRNA for 48 and 72hrs. The final 18 hours were incubated with BrdU. The cells were measured for BrdU incorporation as measured by ELISA using anti-BrdU antibody.

Page 127: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

127

Change in Gene expression of cells treated with Pdx1 siRNA

0

0.2

0.4

0.6

0.8

1

1.2

Pdx1 Nkx6.1 Cyc A2 Cyc B1 Cyc D2 Cyc E1

Rel

ativ

e ch

ange

Ctrl siRNA

PDx1 siRNA

Figure 5-5. Analysis of relative change in the gene expression. Real time PCR was performed using specific primers to analyze the change in gene expression in cells treated with Pdx1 siRNA for 48 hours compared to the cells treated with Ctrl siRNA. β-actin primers were used as internal control.

Page 128: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

128

Figure 5-6. Analysis of cyclin B1 promoter using CMV-Pdx1. The 3T3 cells were transfected with cyclin B1 promoter (-991 bp) driving luciferase gene construct. The cells were co-transfected with either CMV-Pdx1 plasmid. No change in the activity levels was observed with increased concentration of CMV-Pdx1 plasmid.

0

0.2

0.4

0.6

0.8

1

1.2

1.4

0 0.1 0.5 1 2 5CMV-Pdx1 Concentration (ug)

Rel

ativ

e Lu

cife

rase

Val

ue

Page 129: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

129

pcDNA3 + -521CycB1-Luc

Nkx6.1+ -521CycB1-Luc

pcDNA3 + -521CycB1-Luc

Nkx6.1+ -521CycB1-Luc

pcDNA3 + -521CycB1-Luc

Nkx6.1+ -521CycB1-Luc

pcDNA3 + -521CycB1-Luc

Nkx6.1+ -521CycB1-Luc

Figure 5-7. Nkx6.1 activation of Cyclin B1promoter. (Panel A) The 3T3 cells were transfected with cyclin B1 promoter (-991 bp) driving luciferase gene construct. The cells were co-transfected with either pcDNA3 null vector or Nkx6.1 cDNA plasmid. Increased activation was observed in cells treated with Pdx1 cDNA plasmid). (Panel B) The 3T3 cells were transfected with cyclin B1 short promoter (-551 bp) driving luciferase gene construct. The cells were co-transfected with either pcDNA3 null vector or Nkx6.1 cDNA plasmid. No change in the activity levels was observed between the two treatments

pcDNA3 + -921CycB1-Luc Nkx6.1 (0.5ug) + -921CycB1-Luc

pcDNA3 + -921CycB1-Luc Nkx6.1 (0.5ug) + -921CycB1-Luc pcDNA3 + -921CycB1-Luc Nkx6.1 (0.5ug) + -921CycB1-Luc

A

B

Page 130: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

130

Figure 5-8. Pdx1 activation of Nkx6.1 promoter. The 3T3 cells were transfected with Nkx6.1 promoter driving luciferase reporter construct. The cells were co-transfected with either (1) pcDNA3 null vector or (2) Pdx1 cDNA plasmid. Increased activation was observed in cells treated with Pdx1 cDNA plasmid.

Page 131: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

131

Figure 5-9. Chain of events triggered by Pdx1 towards beta cell cycle progression. Pdx1 protein activates its own promoter to increase Pdx1 levels. The increased pdx1 levels in turn activate Nkx6.1 promoter which further binds to cyclin B1 promoter to promote cell cycle progression into mitotic phase.

Pdx1 promoter

Page 132: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

132

CHAPTER 6 FUTURE STUDIES

Reversal of Streptozotocin-induced Diabetes in Mice by Cellular Transduction with Recombinant Pancreatic Transcription Factor, Pancreatic Duodenal Homeobox-1

Future Studies

This study explored a novel approach for treatment of diabetes in STZ-induced diabetes

model. This approach has exploited three properties of Pdx1 protein 1) It is a transcription factor

that promotes beta cell regeneration and liver cell transdifferentiation into IPCs , 2) It has a 16

amino acid inbuilt protein transduction domain, and 3) It has ability to autoregulate its own gene

expression. All the above properties make Pdx1 protein an ideal candidate molecule to promote

beta cell regeneration and liver cell transdifferentiation into IPCs.

Beta cell regeneration is an intriguing phenomenon that dramatically shifts the ability of

the body to maintain glucose homeostasis. There are multiple theories regarding the pattern of

beta cell regeneration. Of these, self duplication/replication of existing beta cells(67;149) and

neogenesis from pancreatic ducts(150-152) are believed to be the most common ways by which

beta cell mass increases. The other theories include recapitulation of embryonic

development(153;154), extra pancreatic stem cell source such as bone marrow(31), and exocrine

cell transdifferentation into beta cell(155). It would be interesting to dissect the pattern of beta

cell regeneration induced by rPdx1 protein therapy. The Cre-Lox and /or Pulse-Chase

approaches in transgenic mice will provide an unambiguous conclusion of the pattern of beta cell

regeneration.

In a clinical setup, Pdx1 therapy would be ideal for type 2 diabetes patients. However, the

STZ-induced diabetic model does not entirely simulate the type 2 diabetic condition. The future

study should determine the therapeutic effects of Pdx1 protein in type 2 diabetic mouse models

such as BB rat, Ob/Ob mice, and db/db mice. These are genetic models of type 2 diabetes with

Page 133: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

133

leptin or leptin receptor deficiency. The homozygotes of the ob/ob strains are leptin deficient

mice. They become obese and severely diabetic consequencing in the regression of islets and die

prematurely. They also exhibit hyperphagia, glucose intolerance, elevated plasma insulin,

subfertility, and impaired wound healing. Injection of recombinant leptin into obese

homozygotes sharply reduces their body weight, decreases food intake, increases energy

expenditure, and restores fertility in male mice.

db/db homozygous mice become diabetic due to spontaneous mutation in the leptin

receptor around three to four weeks of age. The mice become hyperglycemic at 4-8 weeks of

age. Homozygous mutant mice exhibit polyphagia, polydipsia, and polyuria. The other

pathological findings exhibited include obesity, uncontrolled rise in blood sugar, and severe

depletion of the insulin-producing beta-cells of the pancreatic islets with death occurring by 10

months of age. However, studies in genetic models of type 2 diabetes may be challenging due to

the complexity of the disease pathogenesis. There are several factors that need to be considered

such as insulin resistance, obesity, and beta cell stress. In order to characterize the therapeutic

effects, a detailed metabolic profile including body weight, plasma insulin, blood glucose,

cholesterol, tri-glycerides, and basic metabolic rate should be evaluated.

Transdifferentiation of cells from one cell type to another(155) and induction of

pluripotent stem cells from adult cells(156-159) have opened a new era for cell based therapy.

However, extensive studies on the epigenetic modifications are yet to be conducted before we

come out with therapeutic implications(160;161). Similarly, it will be interesting to understand

the epigenetic changes that occur within the chromatin of the hepatocytes that transdifferentiated

into IPCs. This could be initiated by isolating transdifferentiated cells by laser capture

microdissection. Epigenetic modifications that occur on Pdx1 as well as the insulin promoter and

Page 134: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

134

the role of histone acetyl transferases, deacetylases, and methyltransferases will provide insight

into the mechanism of transdifferentiation.

Pdx1 Protein Therapy for the Treatment of Type 1 Diabetes in NOD Mice

Future Studies

This study has lead to the discovery of a novel autoantigen that could play a major role in

the pathogenesis of type 1 diabetes. Immunotherapy with rPdx1 provides us with clues that Pdx1

autoantigen has a role in the pathogenesis of type 1 diabetes. We need to determine, if Pdx1 is a

primary autoantigen. The future studies should focus on extraction of T-cell clones with

receptors for the Pdx1 peptide. Adoptive transfer studies using such homogenous T- cell clones

would lead to better understanding of the pathogenesis of type 1 diabetes.

The discovery of a novel autoantigen in NOD mice will prove useful if we observe

similar findings in human patients. A screening method needs to be designed with hundreds of

human serum samples. Based on the interpretations from NOD mice, it is important to obtain the

serum samples from high risk patients even before the onset of diabetes. The current screening

methods include evaluation of insulin, GAD and islet cell autoantibodies(162). Multiple antibody

screening approaches are engaged in patients for better specificity, sensitivity and positive-

predictive value(163). However, the current screening methods can be enhanced with the

discovery of new autoantibodies. It is therefore worthwhile to evaluate if anti-Pdx1 antibodies

can be used as a potential biomarker marker for screening the onset of diabetes in population at

risk.

A mechanism of induction of tolerance with rPdx1 immunotherapy needs to be established.

Previous studies using Insulin (125) and GAD (11) immunotherapy have demonstrated the role

of CD4+CD25+, Foxp3+ regulatory T-cells. Induction of tolerance is also mediated by the shift

in balance from the Th1 pro-inflammatory immune response to Th2 anti-inflammatory immune

Page 135: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

135

response. A detailed cytokine analysis of both pro-and anti-inflammatory mediators using the

serum obtained from peripheral blood or in vitro cytokine stimulation from the splenocytes or

lymph nodes should be determined.

Molecular Mechanism of the Role of Pdx1 in Beta Cell Proliferation

Future Studies

The experiments to understand the role of Pdx1 in beta cell proliferation were conducted

using Rat INS-1 beta cell line. It is a transformed cell line(164) where variations in the activity of

cell cycle machinery compared to the pancreatic islet cells are possible. The future studies should

include Pdx1 knockdown studies in human pancreatic islets. In order to knockdown Pdx1 in

pancreatic islets, a more efficient siRNA approach using viral vectors as opposed to use of

oligomers must be designed to attain better knockdowns. Similarly, it is well known that the rate

of beta cell replication decreases with increasing age (138;143). It is important to study how the

cell cycle events differ between islets from young mice and older mice with respect to Pdx1

interaction with cell cycle machinery.

These in vitro studies must be further continued to understand the role of Pdx1 in beta cell

replication in vivo. The Pdx1 gene conditional knockout mouse would be an ideal model that will

have synergistic advantages to providing insights into the role of Pdx1 in beta cell regeneration.

Page 136: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

136

APPENDIX A NOD MICE TRIAL 1

Table A-1. Mice database of trial 1 Animal Number Trial 1

Rx Duration of Rx

Blood glucose before Sac

Time of Sac

Serum Formalin Fix

Frozen Liver

Insulin Ext

Splenocytes

1L Pdx1 12 wks 96 40 wks Yes Yes Yes 1R Pdx1 12 wks 147 32 wks Yes Yes Yes 1B Pdx1 12 wks 600 35 wks 1N Pdx1 12 wks 600 20 wks Yes Yes Yes Yes 2L Pdx1 12 wks 117 33 w/die 2R Pdx1 12 wks 127 40 wks Yes Yes Yes 2B Pdx1 12 wks 122 22 wks Yes Yes 2N Pdx1 12 wks 318 40 wks Yes Yes Yes 3L Pdx1 12 wks 122 40 wks Yes Yes Yes 3R Pdx1 12 wks 134 40 wks Yes Yes Yes 3B Pdx1 16 wks 103 34 w/ die 3N Pdx1 16 wks 110 32 wks 4L Pdx1 16 wks 123 32 wks Yes Yes Yes 4R Pdx1 16 wks 412 35 w/ die 4B Pdx1 16 wks 600 40 wks Yes Yes Yes 4N Pdx1 16 wks 174 38 wks Yes 5L Pdx1 16 wks 136 32 wks Yes Yes Yes 5R Pdx1 16 wks 530 17 wks Yes Yes Yes 5B Pdx1 16 wks 97 25 w/die 5N Pdx1 16 wks 121 32 wks Yes 6L Saline 10 wks 600 20 wks Yes Yes Yes 6R Saline 12 wks 600 22 wks Yes Yes Yes 6B Saline 12 wks 327 23 wks Yes 6N Saline 12 wks 316 23 wks Yes 7L Saline 12 wks 600 22 wks Yes Yes Yes 7R Saline 12 wks 435 22 wks Yes Yes Yes 7B Saline 7 wks 528 17 wks Yes Yes Yes 7N Saline 11 wks 600 21 wks Yes Yes Yes 8L Saline 12 wks 94 24 wks Yes Yes 8R Saline 7 wks 600 17 wks Yes Yes Yes 8B Saline 8 wks 600 18 wks Yes Yes Yes 8N Saline 11 wks 600 21 wks Yes Yes Yes 9L Saline 12 wks 330 22 wks Yes Yes Yes 9R Saline 7 wks 600 17 wks Yes Yes Yes 9B Saline 8 wks 600 18 w/die 9N Saline 8 wks 600 18 w/die 10L Saline 8 wks 600 18 wks Yes Yes Yes 10R Saline 12 wks 600 22 wks Yes Yes Yes 10B Saline 12 wks 241 22 wks Yes Yes Yes 10N Saline 12 wks 600 22 wks Yes Yes Yes

Page 137: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

137

APPENDIX B

NOD MICE TRIAL 2

Table B-1. Mice database of trial 2 Animal Number Trial 2

Rx Duration of Rx

Blood glucose before Sac

Time of Sac

Serum Formalin Fix

Frozen Liver

Insulin Ext

Splenocytes

11L Pdx1 4 wks 118 27 wks Yes Yes Yes 11R Pdx1 4 wks 112 28 wks Yes Yes Yes 11B Pdx1 4 wks 600 27 wks Yes Yes 11N Pdx1 4 wks 131 30 wks Yes Yes Yes 12L Pdx1 4 wks 124 20 wks Yes Yes Yes, LNs 12R Pdx1 4 wks 108 20 wks Yes Yes Yes, LNs 12B Pdx1 4 wks 600 23 w/die 12N Pdx1 4 wks 102 20 wks Yes Yes Yes, LNs 13L Pdx1 4 wks 122 13R Pdx1 4 wks 415 23 wks 13B Pdx1 4 wks 103 13N Pdx1 8wks 110 14L Pdx1 8wks 146 32 wks Yes Yes Yes, LNs 14R Pdx1 8wks 118 20 wks Yes Yes Yes, LNs 14B Pdx1 8wks 252 20 wks Yes 14N Pdx1 8wks 109 20 wks Yes Yes Yes, LNs 15L Pdx1 8wks 124 25w/die 15R Pdx1 8wks 600 26 wks Yes 15B Pdx1 8wks 203 30 wks Yes Yes Yes 15N Pdx1 8wks 121 30 wks Yes Yes Yes 16L Mutant 8wks 154 30 wks Yes Yes Yes 16R Mutant 8wks 136 34 wks Yes Yes Yes 16B Mutant 8wks 600 26 wks Yes Yes 16N Mutant 8wks 199 30 wks Yes Yes Yes 17L Mutant 8wks 121 30 wks 17R Mutant 8wks 106 30 wks Yes Yes Yes 17B Mutant 8wks 600 24 wks 17N Mutant 8wks 600 27 wks 18L Mutant 8wks 134 34 wks Yes Yes Yes 18R Mutant 8wks 131 34 wks Yes Yes Yes 18B Mutant 8wks 116 34wks Yes Yes Yes 18N Saline 11 wks 540 24 wks 19L Saline 12 wks 113 28 wks Yes Yes Yes 19R Saline 7 wks 336 17 wks 19B Saline 8 wks 546 16 wks 19N Saline 8 wks 248 28 wks Yes Yes Yes 20L Saline 8 wks 262 22 wks 20R Saline 12 wks 600 23 wks 20B Saline 12 wks 560 17 wks 20N Saline 12 wks 600 16 wks Yes

Page 138: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

138

REFERENCES

1. Atkinson,M.A. and Eisenbarth,G.S. 2001. Type 1 diabetes: New perspectives on disease pathogenesis and treatment. Lancet 358:221-229.

2. Wilson,K. and Eisenbarth,G.S. 1990. Immunopathogenesis and immunotherapy of type 1 diabetes. Annu.Rev.Med. 41:497-508.

3. Jahromi,M.M. and Eisenbarth,G.S. 2006. Genetic determinants of type 1 diabetes across populations. Ann.N.Y.Acad.Sci. 1079:289-299.

4. Kahn,C.R., Vicent,D., and Doria,A. 1996. Genetics of non-insulin-dependent (type-II) diabetes mellitus. Annu.Rev.Med. 47:509-531.

5. Doria,A., Patti,M.E., and Kahn,C.R. 2008. The emerging genetic architecture of type 2 diabetes. Cell Metab 8:186-200.

6. Yoon,J.W. and Jun,H.S. 2001. Cellular and molecular pathogenic mechanisms of insulin-dependent diabetes mellitus. Ann.N.Y.Acad.Sci. 928:200-211.

7. Jahromi,M.M. and Eisenbarth,G.S. 2007. Cellular and molecular pathogenesis of type 1A diabetes. Cell Mol.Life Sci. 64:865-872.

8. Peng,H. and Hagopian,W. 2006. Environmental factors in the development of Type 1 diabetes. Rev.Endocr.Metab Disord. 7:149-162.

9. Bottazzo,G.F., Florin-Christensen,A., and Doniach,D. 1974. Islet-cell antibodies in diabetes mellitus with autoimmune polyendocrine deficiencies. Lancet 2:1279-1283.

10. Miao,D., Yu,L., and Eisenbarth,G.S. 2007. Role of autoantibodies in type 1 diabetes. Front Biosci. 12:1889-1898.

11. Kaufman,D.L., Clare-Salzler,M., Tian,J., Forsthuber,T., Ting,G.S., Robinson,P., Atkinson,M.A., Sercarz,E.E., Tobin,A.J., and Lehmann,P.V. 1993. Spontaneous loss of T-cell tolerance to glutamic acid decarboxylase in murine insulin-dependent diabetes. Nature 366:69-72.

12. Taplin,C.E. and Barker,J.M. 2008. Autoantibodies in type 1 diabetes. Autoimmunity 41:11-18.

13. Daniel,D. and Wegmann,D.R. 1996. Intranasal administration of insulin peptide B: 9-23 protects NOD mice from diabetes. Ann.N.Y.Acad.Sci. 778:371-372.

14. Daniel,D. and Wegmann,D.R. 1996. Protection of nonobese diabetic mice from diabetes by intranasal or subcutaneous administration of insulin peptide B-(9-23). Proc.Natl.Acad.Sci.U.S.A 93:956-960.

Page 139: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

139

15. Tian,J., Clare-Salzler,M., Herschenfeld,A., Middleton,B., Newman,D., Mueller,R., Arita,S., Evans,C., Atkinson,M.A., Mullen,Y. et al. 1996. Modulating autoimmune responses to GAD inhibits disease progression and prolongs islet graft survival in diabetes-prone mice. Nat.Med. 2:1348-1353.

16. Tian,J., Atkinson,M.A., Clare-Salzler,M., Herschenfeld,A., Forsthuber,T., Lehmann,P.V., and Kaufman,D.L. 1996. Nasal administration of glutamate decarboxylase (GAD65) peptides induces Th2 responses and prevents murine insulin-dependent diabetes. J.Exp.Med. 183:1561-1567.

17. Tisch,R., Yang,X.D., Singer,S.M., Liblau,R.S., Fugger,L., and McDevitt,H.O. 1993. Immune response to glutamic acid decarboxylase correlates with insulitis in non-obese diabetic mice. Nature 366:72-75.

18. Tisch,R., Wang,B., Atkinson,M.A., Serreze,D.V., and Friedline,R. 2001. A glutamic acid decarboxylase 65-specific Th2 cell clone immunoregulates autoimmune diabetes in nonobese diabetic mice. J.Immunol. 166:6925-6936.

19. Tisch,R., Liblau,R.S., Yang,X.D., Liblau,P., and McDevitt,H.O. 1998. Induction of GAD65-specific regulatory T-cells inhibits ongoing autoimmune diabetes in nonobese diabetic mice. Diabetes 47:894-899.

20. Elias,D., Reshef,T., Birk,O.S., van der,Z.R., Walker,M.D., and Cohen,I.R. 1991. Vaccination against autoimmune mouse diabetes with a T-cell epitope of the human 65-kDa heat shock protein. Proc.Natl.Acad.Sci.U.S.A 88:3088-3091.

21. Bowman,M. and Atkinson,M.A. 2002. Heat shock protein therapy fails to prevent diabetes in NOD mice. Diabetologia 45:1350-1351.

22. Skyler,J.S., Krischer,J.P., Wolfsdorf,J., Cowie,C., Palmer,J.P., Greenbaum,C., Cuthbertson,D., Rafkin-Mervis,L.E., Chase,H.P., and Leschek,E. 2005. Effects of oral insulin in relatives of patients with type 1 diabetes: The Diabetes Prevention Trial--Type 1. Diabetes Care 28:1068-1076.

23. 2002. Effects of insulin in relatives of patients with type 1 diabetes mellitus. N.Engl.J.Med. 346:1685-1691.

24. Raz,I., Avron,A., Tamir,M., Metzger,M., Symer,L., Eldor,R., Cohen,I.R., and Elias,D. 2007. Treatment of new-onset type 1 diabetes with peptide DiaPep277 is safe and associated with preserved beta-cell function: extension of a randomized, double-blind, phase II trial. Diabetes Metab Res.Rev. 23:292-298.

25. DeFronzo,R.A. 1997. Insulin resistance: a multifaceted syndrome responsible for NIDDM, obesity, hypertension, dyslipidaemia and atherosclerosis. Neth.J.Med. 50:191-197.

26. Kahn,B.B. and Rossetti,L. 1998. Type 2 diabetes--who is conducting the orchestra? Nat.Genet. 20:223-225.

Page 140: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

140

27. Stumvoll,M., Goldstein,B.J., and van Haeften,T.W. 2005. Type 2 diabetes: Principles of pathogenesis and therapy. Lancet 365:1333-1346.

28. Li,L., Yi,Z., Tisch,R., and Wang,B. 2008. Immunotherapy of type 1 diabetes. Arch.Immunol.Ther.Exp.(Warsz.) 56:227-236.

29. Sari,R. and Balci,M.K. 2005. Relationship between C peptide and chronic complications in type-2 diabetes mellitus. J.Natl.Med.Assoc. 97:1113-1118.

30. Shapiro,A.M., Ricordi,C., Hering,B.J., Auchincloss,H., Lindblad,R., Robertson,R.P., Secchi,A., Brendel,M.D., Berney,T., Brennan,D.C. et al. 2006. International trial of the Edmonton protocol for islet transplantation. N.Engl.J.Med. 355:1318-1330.

31. Ianus,A., Holz,G.G., Theise,N.D., and Hussain,M.A. 2003. In vivo derivation of glucose-competent pancreatic endocrine cells from bone marrow without evidence of cell fusion

250. J.Clin.Invest 111:843-850. 32. Voltarelli,J.C., Couri,C.E., Stracieri,A.B., Oliveira,M.C., Moraes,D.A., Pieroni,F.,

Coutinho,M., Malmegrim,K.C., Foss-Freitas,M.C., Simoes,B.P. et al. 2007. Autologous nonmyeloablative hematopoietic stem cell transplantation in newly diagnosed type 1 diabetes mellitus. JAMA 297:1568-1576.

33. Denner,L. and Urban,R.J. 2008. Critical issues for engineering cord blood stem cells to produce insulin. Expert.Opin.Biol.Ther. 8:1251-1254.

34. Kroon,E., Martinson,L.A., Kadoya,K., Bang,A.G., Kelly,O.G., Eliazer,S., Young,H., Richardson,M., Smart,N.G., Cunningham,J. et al. 2008. Pancreatic endoderm derived from human embryonic stem cells generates glucose-responsive insulin-secreting cells in vivo. Nat.Biotechnol. 26:443-452.

35. Jonsson,J., Carlsson,L., Edlund,T., and Edlund,H. 1994. Insulin-promoter-factor 1 is required for pancreas development in mice. Nature 371:606-609.

36. Offield,M.F., Jetton,T.L., Labosky,P.A., Ray,M., Stein,R.W., Magnuson,M.A., Hogan,B.L., and Wright,C.V. 1996. PDX-1 is required for pancreatic outgrowth and differentiation of the rostral duodenum. Development 122:983-995.

37. Holland,A.M., Gonez,L.J., Naselli,G., MacDonald,R.J., and Harrison,L.C. 2005. Conditional expression demonstrates the role of the homeodomain transcription factor Pdx1 in maintenance and regeneration of beta-cells in the adult pancreas. Diabetes 54:2586-2595.

38. Sharma, A. and Stein, R. Glucose-induced transcription of the insulin gene is mediated by factors required for beta-cell-type-specific expression. Mol.Cell Biol. 14(2), 871-879. 1994.

39. Gerrish,K., Gannon,M., Shih,D., Henderson,E., Stoffel,M., Wright,C.V., and Stein,R.

2000. Pancreatic beta cell-specific transcription of the pdx-1 gene. The role of conserved

Page 141: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

141

upstream control regions and their hepatic nuclear factor 3beta sites. J.Biol.Chem. 275:3485-3492.

40. Guz,Y., Montminy,M.R., Stein,R., Leonard,J., Gamer,L.W., Wright,C.V., and Teitelman,G. 1995. Expression of murine STF-1, a putative insulin gene transcription factor, in beta cells of pancreas, duodenal epithelium and pancreatic exocrine and endocrine progenitors during ontogeny. Development 121:11-18.

41. Habener, J. F., Kemp, D. M., and Thomas, M. K. Minireview: transcriptional regulation in pancreatic development. Endocrinology 146(3), 1025-1034. 2005.

42. Waeber, G., Thompson, N., Nicod, P., and Bonny, C. Transcriptional activation of the

GLUT2 gene by the IPF-1/STF-1/IDX-1 homeobox factor. Mol.Endocrinol. 10(11), 1327-1334. 1996.

43. Watada, H., Kajimoto, Y., Miyagawa, J., Hanafusa, T., Hamaguchi, K., Matsuoka, T.,

Yamamoto, K., Matsuzawa, Y., Kawamori, R., and Yamasaki, Y. PDX-1 induces insulin and glucokinase gene expressions in alphaTC1 clone 6 cells in the presence of betacellulin. Diabetes 45(12), 1826-1831. 1996.

44. Gerrish,K., Cissell,M.A., and Stein,R. 2001. The role of hepatic nuclear factor 1 alpha

and PDX-1 in transcriptional regulation of the pdx-1 gene. J.Biol.Chem. 276:47775-47784.

45. Sharma,A., Zangen,D.H., Reitz,P., Taneja,M., Lissauer,M.E., Miller,C.P., Weir,G.C., Habener,J.F., and Bonner-Weir,S. 1999. The homeodomain protein IDX-1 increases after an early burst of proliferation during pancreatic regeneration. Diabetes 48:507-513.

46. Ashizawa, S., Brunicardi, F. C., and Wang, X. P. PDX-1 and the pancreas. Pancreas 28(2), 109-120. 2004.

47. Joliot,A. and Prochiantz,A. 2004. Transduction peptides: from technology to physiology.

Nat.Cell Biol. 6:189-196.

48. Sandgren, S., Cheng, F., and Belting, M. Nuclear targeting of macromolecular polyanions by an HIV-Tat derived peptide. Role for cell-surface proteoglycans. J.Biol.Chem. 277(41), 38877-38883. 10-11-2002.

49. Wadia,J.S., Stan,R.V., and Dowdy,S.F. 2004. Transducible TAT-HA fusogenic peptide

enhances escape of TAT-fusion proteins after lipid raft macropinocytosis. Nat.Med. 10:310-315.

50. Frankel, A. D. and Pabo, C. O. Cellular uptake of the tat protein from human immunodeficiency virus. Cell 55(6), 1189-1193. 12-23-1988.

Page 142: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

142

51. Green,M. and Loewenstein,P.M. 1988. Autonomous functional domains of chemically synthesized human immunodeficiency virus tat trans-activator protein. Cell 55:1179-1188.

52. Joliot, A. Transduction peptides within naturally occurring proteins. Sci.STKE. 2005(313), e54. 12-6-2005.

53. Nolden,L., Edenhofer,F., Haupt,S., Koch,P., Wunderlich,F.T., Siemen,H., and Brustle,O.

2006. Site-specific recombination in human embryonic stem cells induced by cell-permeant Cre recombinase. Nat.Methods 3:461-467.

54. Shibagaki, N. and Udey, M. C. Dendritic cells transduced with protein antigens induce cytotoxic lymphocytes and elicit antitumor immunity. J.Immunol. 168(5), 2393-2401. 3-1-2002.

55. Krosl,J., Austin,P., Beslu,N., Kroon,E., Humphries,R.K., and Sauvageau,G. 2003. In

vitro expansion of hematopoietic stem cells by recombinant TAT-HOXB4 protein. Nat.Med. 9:1428-1432.

56. Nagahara,H., Vocero-Akbani,A.M., Snyder,E.L., Ho,A., Latham,D.G., Lissy,N.A., Becker-Hapak,M., Ezhevsky,S.A., and Dowdy,S.F. 1998. Transduction of full-length TAT fusion proteins into mammalian cells: TAT-p27Kip1 induces cell migration. Nat.Med. 4:1449-1452.

57. Zhou,H., Wu,S., Joo,J.Y., Zhu,S., Han,D.W., Lin,T., Trauger,S., Bien,G., Yao,S., Zhu,Y. et al. 2009. Generation of Induced Pluripotent Stem Cells Using Recombinant Proteins. Cell Stem Cell.

58. Schwarze, S. R. and Dowdy, S. F. In vivo protein transduction: intracellular delivery of biologically active proteins, compounds and DNA. Trends Pharmacol.Sci. 21(2), 45-48. 2000.

59. Myou, S., Zhu, X., Myo, S., Boetticher, E., Meliton, A. Y., Liu, J., Munoz, N. M., and

Leff, A. R. Blockade of airway inflammation and hyperresponsiveness by HIV-TAT-dominant negative Ras. J.Immunol. 171(8), 4379-4384. 10-15-2003.

60. Bucci, M., Gratton, J. P., Rudic, R. D., Acevedo, L., Roviezzo, F., Cirino, G., and Sessa,

W. C. In vivo delivery of the caveolin-1 scaffolding domain inhibits nitric oxide synthesis and reduces inflammation. Nat.Med. 6(12), 1362-1367. 2000.

61. Shibagaki, N. and Udey, M. C. Dendritic cells transduced with TAT protein transduction

domain-containing tyrosinase-related protein 2 vaccinate against murine melanoma. Eur.J.Immunol. 33(4), 850-860. 2003.

62. Asoh, S. and Ohta, S. [Protein transduction and protection of cells against apoptosis]. Seikagaku 76(7), 654-658. 2004.

Page 143: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

143

63. Noguchi,H., Kaneto,H., Weir,G.C., and Bonner-Weir,S. 2003. PDX-1 protein containing its own antennapedia-like protein transduction domain can transduce pancreatic duct and islet cells. Diabetes 52:1732-1737.

64. Noguchi,H. and Matsumoto,S. 2006. Protein transduction technology: a novel therapeutic perspective. Acta Med.Okayama 60:1-11.

65. Noguchi,H., Matsushita,M., Matsumoto,S., Lu,Y.F., Matsui,H., and Bonner-Weir,S. 2005. Mechanism of PDX-1 protein transduction. Biochem.Biophys.Res.Commun. 332:68-74.

66. Noguchi,H., Matsumoto,S., Okitsu,T., Iwanaga,Y., Yonekawa,Y., Nagata,H., Matsushita,M., Wei,F.Y., Matsui,H., Minami,K. et al. 2005. PDX-1 protein is internalized by lipid raft-dependent macropinocytosis. Cell Transplant. 14:637-645.

67. Dor,Y., Brown,J., Martinez,O.I., and Melton,D.A. 2004. Adult pancreatic beta-cells are formed by self-duplication rather than stem-cell differentiation. Nature 429:41-46.

68. Nir, T., Melton, D. A., and Dor, Y. Recovery from diabetes in mice by beta cell regeneration. J.Clin.Invest 117(9), 2553-2561. 9-4-2007.

69. Harmon,J.S., Gleason,C.E., Tanaka,Y., Oseid,E.A., Hunter-Berger,K.K., and

Robertson,R.P. 1999. In vivo prevention of hyperglycemia also prevents glucotoxic effects on PDX-1 and insulin gene expression. Diabetes 48:1995-2000.

70. Robertson,R., Zhou,H., Zhang,T., and Harmon,J.S. 2007. Chronic oxidative stress as a mechanism for glucose toxicity of the beta cell in Type 2 diabetes. Cell Biochem.Biophys. 48:139-146.

71. Robertson,R.P. and Harmon,J.S. 2006. Diabetes, glucose toxicity, and oxidative stress: A case of double jeopardy for the pancreatic islet beta cell. Free Radic.Biol.Med. 41:177-184.

72. Sharma,A., Olson,L.K., Robertson,R.P., and Stein,R. 1995. The reduction of insulin gene transcription in HIT-T15 beta cells chronically exposed to high glucose concentration is associated with the loss of RIPE3b1 and STF-1 transcription factor expression. Mol.Endocrinol. 9:1127-1134.

73. Kito, H., Ose, Y., Mizuhira, V., Sato, T., Ishikawa, T., and Tazawa, T. Separation and purification of (Cd, Cu, Zn)-metallothionein in carp hepato-pancreas. Comp Biochem.Physiol C. 73(1), 121-127. 1982.

74. Meivar-Levy, I. and Ferber, S. New organs from our own tissues: liver-to-pancreas

transdifferentiation. Trends Endocrinol.Metab 14(10), 460-466. 2003. 75. Michalopoulos,G.K. and DeFrances,M.C. 1997. Liver regeneration. Science 276:60-66.

Page 144: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

144

76. Yang, H., Morrison, C. M., Conlon, J. M., Laybolt, K., and Wright, J. R., Jr. Immunocytochemical characterization of the pancreatic islet cells of the Nile Tilapia (Oreochromis niloticus). Gen.Comp Endocrinol. 114(1), 47-56. 1999.

77. Wolf,H.K., Burchette,J.L., Jr., Garcia,J.A., and Michalopoulos,G. 1990. Exocrine

pancreatic tissue in human liver: a metaplastic process? Am.J.Surg.Pathol. 14:590-595.

78. Tang,D.Q., Lu,S., Sun,Y.P., Rodrigue,E., Chou,W., Yang,C., Chang,L.J., and Yang,L.J. 2006. Reprogramming Liver-Stem WB Cells into Functional Insulin-Producing Cells by Persistent Expression of Pdx1- and Pdx1-VP16 Mediated by Lentiviral Vectors. Lab Invest 86:83-93.

79. Tang,D.Q., Cao,L.Z., Chou,W., Shun,L., Farag,C., Atkinson,M.A., Li,S.W., Chang,L.J., and Yang,L.J. 2006. Role of Pax4 in Pdx1-VP16-mediated liver-to-endocrine pancreas transdifferentiation. Lab Invest 86:829-841.

80. Fodor, A., Harel, C., Fodor, L., Armoni, M., Salmon, P., Trono, D., and Karnieli, E. Adult rat liver cells transdifferentiated with lentiviral IPF1 vectors reverse diabetes in mice: an ex vivo gene therapy approach. Diabetologia 50(1), 121-130. 2007.

81. Yechoor,V., Liu,V., Espiritu,C., Paul,A., Oka,K., Kojima,H., and Chan,L. 2009.

Neurogenin3 is sufficient for transdetermination of hepatic progenitor cells into neo-islets in vivo but not transdifferentiation of hepatocytes. Dev.Cell 16:358-373.

82. Meivar-Levy,I., Sapir,T., Gefen-Halevi,S., Aviv,V., Barshack,I., Onaca,N., Mor,E., and Ferber,S. 2007. Pancreatic and duodenal homeobox gene 1 induces hepatic dedifferentiation by suppressing the expression of CCAAT/enhancer-binding protein beta. Hepatology 46:898-905.

83. Ber,I., Shternhall,K., Perl,S., Ohanuna,Z., Goldberg,I., Barshack,I., Benvenisti-Zarum,L., Meivar-Levy,I., and Ferber,S. 2003. Functional, persistent, and extended liver to pancreas transdifferentiation. J.Biol.Chem. 278:31950-31957.

84. Ferber,S., Halkin,A., Cohen,H., Ber,I., Einav,Y., Goldberg,I., Barshack,I., Seijffers,R., Kopolovic,J., Kaiser,N. et al. 2000. Pancreatic and duodenal homeobox gene 1 induces expression of insulin genes in liver and ameliorates streptozotocin-induced hyperglycemia. Nat.Med. 6:568-572.

85. Sapir,T., Shternhall,K., Meivar-Levy,I., Blumenfeld,T., Cohen,H., Skutelsky,E.,

Eventov-Friedman,S., Barshack,I., Goldberg,I., Pri-Chen,S. et al. 2005. Cell-replacement therapy for diabetes: Generating functional insulin-producing tissue from adult human liver cells. Proc.Natl.Acad.Sci.U.S.A 102:7964-7969.

86. Shternhall-Ron,K., Quintana,F.J., Perl,S., Meivar-Levy,I., Barshack,I., Cohen,I.R., and Ferber,S. 2007. Ectopic PDX-1 expression in liver ameliorates type 1 diabetes. J.Autoimmun.

Page 145: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

145

87. Kojima,H., Fujimiya,M., Matsumura,K., Younan,P., Imaeda,H., Maeda,M., and Chan,L. 2003. NeuroD-betacellulin gene therapy induces islet neogenesis in the liver and reverses diabetes in mice. Nat.Med. 9:596-603.

88. Koizumi,M., Doi,R., Toyoda,E., Tulachan,S.S., Kami,K., Mori,T., Ito,D., Kawaguchi,Y.,

Fujimoto,K., Gittes,G.K. et al. 2004. Hepatic regeneration and enforced PDX-1 expression accelerate transdifferentiation in liver. Surgery 136:449-457.

89. Kim,S., Shin,J.S., Fisher,R.C., Lee,M.J., and Kim,K.W. 2007. Streptozotocin-induced diabetes can be reversed by hepatic oval cell activation through hepatic transdifferentiation and pancreatic islet regeneration. Lab Invest In press.

90. Bonner-Weir,S. and Weir,G.C. 2005. New sources of pancreatic beta-cells. Nat.Biotechnol. 23:857-861.

91. Meier,J.J., Bhushan,A., and Butler,P.C. 2006. The potential for stem cell therapy in diabetes. Pediatr.Res. 59:65R-73R.

92. Trucco,M. 2006. Is facilitating pancreatic beta cell regeneration a valid option for clinical therapy? Cell Transplant. 15 Suppl 1:S75-S84.

93. Perfetti,R., Zhou,J., Doyle,M.E., and Egan,J.M. 2000. Glucagon-like peptide-1 induces cell proliferation and pancreatic- duodenum homeobox-1 expression and increases endocrine cell mass in the pancreas of old, glucose-intolerant rats

31. Endocrinology 141:4600-4605.

94. Stoffers,D.A., Kieffer,T.J., Hussain,M.A., Drucker,D.J., Bonner-Weir,S., Habener,J.F., and Egan,J.M. 2000. Insulinotropic glucagon-like peptide 1 agonists stimulate expression of homeodomain protein IDX-1 and increase islet size in mouse pancreas. Diabetes 49:741-748.

95. Xu,G., Stoffers,D.A., Habener,J.F., and Bonner-Weir,S. 1999. Exendin-4 stimulates both beta-cell replication and neogenesis, resulting in increased beta-cell mass and improved glucose tolerance in diabetic rats

1. Diabetes 48:2270-2276.

96. Zhou,J., Pineyro,M.A., Wang,X., Doyle,M.E., and Egan,J.M. 2002. Exendin-4 differentiation of a human pancreatic duct cell line into endocrine cells: Involvement of PDX-1 and HNF3beta transcription factors. J.Cell Physiol 192:304-314.

97. Pittenger,G.L., Taylor-Fishwick,D.A., Johns,R.H., Burcus,N., Kosuri,S., and Vinik,A.I. 2007. Intramuscular injection of islet neogenesis-associated protein peptide stimulates pancreatic islet neogenesis in healthy dogs. Pancreas 34:103-111.

98. Gagliardino,J.J., Del Zotto,H., Massa,L., Flores,L.E., and Borelli,M.I. 2003. Pancreatic duodenal homeobox-1 and islet neogenesis-associated protein: a possible combined marker of activateable pancreatic cell precursors. J.Endocrinol. 177:249-259.

Page 146: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

146

99. Holland,A.M., Hale,M.A., Kagami,H., Hammer,R.E., and MacDonald,R.J. 2002. Experimental control of pancreatic development and maintenance. Proc.Natl.Acad.Sci.U.S.A 99:12236-12241.

100. Stoffers,D.A., Zinkin,N.T., Stanojevic,V., Clarke,W.L., and Habener,J.F. 1997. Pancreatic agenesis attributable to a single nucleotide deletion in the human IPF1 gene coding sequence. Nat.Genet. 15:106-110.

101. Yang,L.J. 2006. Liver stem cell-derived beta-cell surrogates for treatment of type 1 diabetes. Autoimmun.Rev. 5:409-413.

102. Prochiantz,A. and Joliot,A. 2003. Can transcription factors function as cell-cell signalling molecules? Nat.Rev.Mol.Cell Biol. 4:814-819.

103. Noguchi, H. and Matsumoto, S. Protein transduction technology offers a novel therapeutic approach for diabetes. J.Hepatobiliary.Pancreat.Surg. 13(4), 306-313. 2006.

104. Jin, L. H., Bahn, J. H., Eum, W. S., Kwon, H. Y., Jang, S. H., Han, K. H., Kang, T. C.,

Won, M. H., Kang, J. H., Cho, S. W., Park, J., and Choi, S. Y. Transduction of human catalase mediated by an HIV-1 TAT protein basic domain and arginine-rich peptides into mammalian cells. Free Radic.Biol.Med. 31(11), 1509-1519. 12-1-2001.

105. Coleman,W.B., McCullough,K.D., Esch,G.L., Faris,R.A., Hixson,D.C., Smith,G.J., and

Grisham,J.W. 1997. Evaluation of the differentiation potential of WB-F344 rat liver epithelial stem-like cells in vivo. Differentiation to hepatocytes after transplantation into dipeptidylpeptidase-IV-deficient rat liver. Am.J.Pathol. 151:353-359.

106. Couchie,D., Holic,N., Chobert,M.N., Corlu,A., and Laperche,Y. 2002. In vitro differentiation of WB-F344 rat liver epithelial cells into the biliary lineage. Differentiation 69:209-215.

107. Narushima,Y., Unno,M., Nakagawara,K., Mori,M., Miyashita,H., Suzuki,Y., Noguchi,N., Takasawa,S., Kumagai,T., Yonekura,H. et al. 1997. Structure, chromosomal localization and expression of mouse genes encoding type III Reg, RegIII alpha, RegIII beta, RegIII gamma. Gene 185:159-168.

108. Peshavaria,M., Larmie,B.L., Lausier,J., Satish,B., Habibovic,A., Roskens,V., Larock,K., Everill,B., Leahy,J.L., and Jetton,T.L. 2006. Regulation of pancreatic beta-cell regeneration in the normoglycemic 60% partial-pancreatectomy mouse. Diabetes 55:3289-3298.

109. Guz,Y., Nasir,I., and Teitelman,G. 2001. Regeneration of pancreatic beta cells from intra-islet precursor cells in an experimental model of diabetes. Endocrinology 142:4956-4968.

110. O'Doherty,R.M., Lehman,D.L., Telemaque-Potts,S., and Newgard,C.B. 1999. Metabolic impact of glucokinase overexpression in liver: lowering of blood glucose in fed rats is accompanied by hyperlipidemia. Diabetes 48:2022-2027.

Page 147: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

147

111. Waguri,M., Yamamoto,K., Miyagawa,J.I., Tochino,Y., Yamamori,K., Kajimoto,Y., Nakajima,H., Watada,H., Yoshiuchi,I., Itoh,N. et al. 1997. Demonstration of two different processes of beta-cell regeneration in a new diabetic mouse model induced by selective perfusion of alloxan. Diabetes 46:1281-1290.

112. Kulkarni,R.N., Jhala,U.S., Winnay,J.N., Krajewski,S., Montminy,M., and Kahn,C.R. 2004. PDX-1 haploinsufficiency limits the compensatory islet hyperplasia that occurs in response to insulin resistance. J.Clin.Invest 114:828-836.

113. Marshak,S., Benshushan,E., Shoshkes,M., Havin,L., Cerasi,E., and Melloul,D. 2000. Functional conservation of regulatory elements in the pdx-1 gene: PDX-1 and hepatocyte nuclear factor 3beta transcription factors mediate beta- cell-specific expression

37. Mol.Cell Biol. 20:7583-7590.

114. Kushner,J.A., Ye,J., Schubert,M., Burks,D.J., Dow,M.A., Flint,C.L., Dutta,S., Wright,C.V., Montminy,M.R., and White,M.F. 2002. Pdx1 restores beta cell function in Irs2 knockout mice. J.Clin.Invest 109:1193-1201.

115. Hui,H. and Perfetti,R. 2002. Pancreas duodenum homeobox-1 regulates pancreas development during embryogenesis and islet cell function in adulthood. Eur.J.Endocrinol. 146:129-141.

116. Sherry,N.A., Kushner,J.A., Glandt,M., Kitamura,T., Brillantes,A.M., and Herold,K.C. 2006. Effects of autoimmunity and immune therapy on beta-cell turnover in type 1 diabetes. Diabetes 55:3238-3245.

117. Ablamunits,V., Sherry,N.A., Kushner,J.A., and Herold,K.C. 2007. Autoimmunity and Beta cell regeneration in mouse and human type 1 diabetes: the peace is not enough. Ann.N.Y.Acad.Sci. 1103:19-32.

118. Makino,S., Kunimoto,K., Muraoka,Y., Mizushima,Y., Katagiri,K., and Tochino,Y. 1980. Breeding of a non-obese, diabetic strain of mice. Jikken Dobutsu 29:1-13.

119. Zhang,J., Tokui,Y., Yamagata,K., Kozawa,J., Sayama,K., Iwahashi,H., Okita,K., Miuchi,M., Konya,H., Hamaguchi,T. et al. 2007. Continuous stimulation of human glucagon-like peptide-1 (7-36) amide in a mouse model (NOD) delays onset of autoimmune type 1 diabetes. Diabetologia 50:1900-1909.

120. Hadjiyanni,I., Baggio,L.L., Poussier,P., and Drucker,D.J. 2008. Exendin-4 modulates diabetes onset in nonobese diabetic mice. Endocrinology 149:1338-1349.

121. Linn,T., Schneider,K., Goke,B., and Federlin,K. 1996. Glucagon-like-peptide-1 (7-36) amide improves glucose sensitivity in beta-cells of NOD mice. Acta Diabetol. 33:19-24.

122. Li,Y., Cao,X., Li,L.X., Brubaker,P.L., Edlund,H., and Drucker,D.J. 2005. beta-Cell Pdx1 expression is essential for the glucoregulatory, proliferative, and cytoprotective actions of glucagon-like peptide-1. Diabetes 54:482-491.

Page 148: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

148

123. Koya,V., Lu,S., Sun,Y.P., Purich,D.L., Atkinson,M.A., Li,S.W., and Yang,L.J. 2008. Reversal of streptozotocin-induced diabetes in mice by cellular transduction with recombinant pancreatic transcription factor pancreatic duodenal homeobox-1: a novel protein transduction domain-based therapy. Diabetes 57:757-769.

124. Katz,J.D., Wang,B., Haskins,K., Benoist,C., and Mathis,D. 1993. Following a diabetogenic T cell from genesis through pathogenesis. Cell 74:1089-1100.

125. Zhang,L., Nakayama,M., and Eisenbarth,G.S. 2008. Insulin as an autoantigen in NOD/human diabetes. Curr.Opin.Immunol. 20:111-118.

126. Wong,F.S., Wen,L., Tang,M., Ramanathan,M., Visintin,I., Daugherty,J., Hannum,L.G., Janeway,C.A., Jr., and Shlomchik,M.J. 2004. Investigation of the role of B-cells in type 1 diabetes in the NOD mouse. Diabetes 53:2581-2587.

127. Bach,J.F. and Chatenoud,L. 2001. Tolerance to islet autoantigens in type 1 diabetes. Annu.Rev.Immunol. 19:131-161.

128. Bach,J.F. 2001. Immunotherapy of insulin-dependent diabetes mellitus. Curr.Opin.Immunol. 13:601-605.

129. Hoglund,P., Mintern,J., Waltzinger,C., Heath,W., Benoist,C., and Mathis,D. 1999. Initiation of autoimmune diabetes by developmentally regulated presentation of islet cell antigens in the pancreatic lymph nodes. J.Exp.Med. 189:331-339.

130. Wenzlau,J.M., Juhl,K., Yu,L., Moua,O., Sarkar,S.A., Gottlieb,P., Rewers,M., Eisenbarth,G.S., Jensen,J., Davidson,H.W. et al. 2007. The cation efflux transporter ZnT8 (Slc30A8) is a major autoantigen in human type 1 diabetes. Proc.Natl.Acad.Sci.U.S.A 104:17040-17045.

131. Tisch,R. and McDevitt,H.O. 1994. Antigen-specific immunotherapy: is it a real possibility to combat T-cell-mediated autoimmunity? Proc.Natl.Acad.Sci.U.S.A 91:437-438.

132. Pasquali,L., Giannoukakis,N., and Trucco,M. 2008. Induction of immune tolerance to facilitate beta cell regeneration in type 1 diabetes. Adv.Drug Deliv.Rev. 60:106-113.

133. Raz,I., Eldor,R., and Naparstek,Y. 2005. Immune modulation for prevention of type 1 diabetes mellitus. Trends Biotechnol. 23:128-134.

134. Liblau,R.S., Pearson,C.I., Shokat,K., Tisch,R., Yang,X.D., and McDevitt,H.O. 1994. High-dose soluble antigen: peripheral T-cell proliferation or apoptosis. Immunol.Rev. 142:193-208.

135. Song,W.J., Schreiber,W.E., Zhong,E., Liu,F.F., Kornfeld,B.D., Wondisford,F.E., and Hussain,M.A. 2008. Exendin-4 stimulation of cyclin A2 in beta-cell proliferation. Diabetes 57:2371-2381.

Page 149: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

149

136. Lee,Y.C. and Nielsen,J.H. 2009. Regulation of beta cell replication. Mol.Cell Endocrinol. 297:18-27.

137. Heit,J.J., Karnik,S.K., and Kim,S.K. 2006. Intrinsic regulators of pancreatic beta-cell proliferation. Annu.Rev.Cell Dev.Biol. 22:311-338.

138. Rankin,M.M. and Kushner,J.A. 2009. ADAPTIVE BETA CELL PROLIFERATION IS SEVERELY RESTRICTED WITH ADVANCED AGE. Diabetes.

139. Krishnamurthy, J., Ramsey, M. R., Ligon, K. L., Torrice, C., Koh, A., Bonner-Weir, S., and Sharpless, N. E. p16INK4a induces an age-dependent decline in islet regenerative potential. Nature 443(7110), 453-457. 9-28-2006.

140. Schisler,J.C., Fueger,P.T., Babu,D.A., Hohmeier,H.E., Tessem,J.S., Lu,D., Becker,T.C.,

Naziruddin,B., Levy,M., Mirmira,R.G. et al. 2008. Stimulation of human and rat islet beta-cell proliferation with retention of function by the homeodomain transcription factor Nkx6.1. Mol.Cell Biol. 28:3465-3476.

141. Cozar-Castellano,I., Harb,G., Selk,K., Takane,K., Vasavada,R., Sicari,B., Law,B., Zhang,P., Scott,D.K., Fiaschi-Taesch,N. et al. 2008. Lessons from the first comprehensive molecular characterization of cell cycle control in rodent insulinoma cell lines. Diabetes 57:3056-3068.

142. Kushner,J.A., Ciemerych,M.A., Sicinska,E., Wartschow,L.M., Teta,M., Long,S.Y., Sicinski,P., and White,M.F. 2005. Cyclins D2 and D1 are essential for postnatal pancreatic beta-cell growth. Mol.Cell Biol. 25:3752-3762.

143. Teta,M., Long,S.Y., Wartschow,L.M., Rankin,M.M., and Kushner,J.A. 2005. Very slow turnover of beta-cells in aged adult mice. Diabetes 54:2557-2567.

144. Andersen, F. G., Jensen, J., Heller, R. S., Petersen, H. V., Larsson, L. I., Madsen, O. D., and Serup, P. Pax6 and Pdx1 form a functional complex on the rat somatostatin gene upstream enhancer. FEBS Lett. 445(2-3), 315-320. 2-26-1999.

145. Goudet,G., Delhalle,S., Biemar,F., Martial,J.A., and Peers,B. 1999. Functional and

cooperative interactions between the homeodomain PDX1, Pbx, and Prep1 factors on the somatostatin promoter. J.Biol.Chem. 274:4067-4073.

146. Peers, B., Sharma, S., Johnson, T., Kamps, M., and Montminy, M. The pancreatic islet factor STF-1 binds cooperatively with Pbx to a regulatory element in the somatostatin promoter: importance of the FPWMK motif and of the homeodomain. Mol.Cell Biol. 15(12), 7091-7097. 1995.

147. Qiu,Y., Sharma,A., and Stein,R. 1998. p300 mediates transcriptional stimulation by the

basic helix-loop-helix activators of the insulin gene. Mol.Cell Biol. 18:2957-2964.

Page 150: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

150

148. Qiu,Y., Guo,M., Huang,S., and Stein,R. 2002. Insulin gene transcription is mediated by interactions between the p300 coactivator and PDX-1, BETA2, and E47. Mol.Cell Biol. 22:412-420.

149. Teta,M., Rankin,M.M., Long,S.Y., Stein,G.M., and Kushner,J.A. 2007. Growth and regeneration of adult beta cells does not involve specialized progenitors. Dev.Cell 12:817-826.

150. Bonner-Weir, S., Toschi, E., Inada, A., Reitz, P., Fonseca, S. Y., Aye, T., and Sharma, A. The pancreatic ductal epithelium serves as a potential pool of progenitor cells. Pediatr.Diabetes 5 Suppl 2, 16-22. 2004.

151. Bonner-Weir,S. and Sharma,A. 2002. Pancreatic stem cells. J.Pathol. 197:519-526. 152. Dor,Y. and Melton,D.A. 2008. Facultative endocrine progenitor cells in the adult

pancreas. Cell 132:183-184.

153. Bonner-Weir,S., Baxter,L.A., Schuppin,G.T., and Smith,F.E. 1993. A second pathway for regeneration of adult exocrine and endocrine pancreas. A possible recapitulation of embryonic development. Diabetes 42:1715-1720.

154. Gu,G., Dubauskaite,J., and Melton,D.A. 2002. Direct evidence for the pancreatic lineage: NGN3+ cells are islet progenitors and are distinct from duct progenitors

3. Development 129:2447-2457.

155. Zhou,Q., Brown,J., Kanarek,A., Rajagopal,J., and Melton,D.A. 2008. In vivo reprogramming of adult pancreatic exocrine cells to beta-cells. Nature 455:627-632.

156. Yamanaka,S. and Takahashi,K. 2006. [Induction of pluripotent stem cells from mouse fibroblast cultures]. Tanpakushitsu Kakusan Koso 51:2346-2351.

157. Takahashi,K. and Yamanaka,S. 2006. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126:663-676.

158. Lewitzky,M. and Yamanaka,S. 2007. Reprogramming somatic cells towards pluripotency by defined factors. Curr.Opin.Biotechnol. 18:467-473.

159. Wernig,M., Meissner,A., Foreman,R., Brambrink,T., Ku,M., Hochedlinger,K., Bernstein,B.E., and Jaenisch,R. 2007. In vitro reprogramming of fibroblasts into a pluripotent ES-cell-like state. Nature 448:318-324.

160. Maherali,N., Sridharan,R., Xie,W., Utikal,J., Eminli,S., Arnold,K., Stadtfeld,M., Yachechko,R., Tchieu,J., Jaenisch,R. et al. 2007. Directly reprogrammed fibroblasts show global epigenetic remodeling and widespread tissue contribution. Cell Stem Cell 1:55-70.

Page 151: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

151

161. Mikkelsen,T.S., Ku,M., Jaffe,D.B., Issac,B., Lieberman,E., Giannoukos,G., Alvarez,P., Brockman,W., Kim,T.K., Koche,R.P. et al. 2007. Genome-wide maps of chromatin state in pluripotent and lineage-committed cells. Nature 448:553-560.

162. Korhonen,S., Knip,M.M., Kulmala,P., Savola,K., Akerblom,H.K., and Knip,M. 2002. Autoantibodies to GAD, IA-2 and insulin in ICA-positive first-degree relatives of children with type 1 diabetes: a comparison between parents and siblings. Diabetes Metab Res.Rev. 18:43-48.

163. Knip,M. 2002. Can we predict type 1 diabetes in the general population? Diabetes Care 25:623-625.

164. Hohmeier,H.E. and Newgard,C.B. 2004. Cell lines derived from pancreatic islets. Mol.Cell Endocrinol. 228:121-128.

Page 152: © 2009 Vijay Koya - ufdcimages.uflib.ufl.edu · 1 pdx1 protein therapy: a novel approach for treatment of diabetes by vijay koya a dissertation presented to the graduate school of

152

BIOGRAPHICAL SKETCH

Vijay Koya was born in India, in 1978. He graduated from Jubilee Hills Public School in

1993. Following high school, he enrolled into two year college program in biological sciences. In

1996, he was admitted into Acharya N.G. Ranga Agriculture University. He completed his

bachelor’s degree in veterinary medicine in 2001. Due to his interest in basic sciences, he moved

on to pursue graduate studies in United States. In the spring of 2002, he was admitted into

master’s program in the Department of Molecular Biology & Microbiology at the University of

Central Florida, Orlando. In the summer of 2002, Vijay joined Dr. Henry Daniell’s laboratory to

conduct research on the development of new generation anthrax vaccine in transgenic plants.

Besides research, he has actively participated in teaching human anatomy for pre-medical

students. During his master’s program, he received “University Merit Scholar Award” for

academic excellence. Vijay graduated with MS degree in molecular biology& microbiology in

fall 2004 and continued to work in Dr. Henry Daniell’s lab until the end of summer 2005. During

this period, he worked on developing a prototype model for oral delivery of plant expressed

therapeutic proteins. In the fall of 2005, he was admitted into the Interdisciplinary Program (IDP)

in Biomedical Sciences at the College of Medicine at the University of Florida. Vijay joined Dr.

Lijun Yang’s lab in the Department of Pathology to conduct doctoral research. He investigated

on the Pdx1 protein therapy; a novel approach for the treatment of diabetes. He has received

Scientific Achievement Award for the best abstract, at the Annual Rachmiel Levine Symposium,

for two consecutive years 2008 and 2009. He received Ph.D from the University of Florida in the

summer of 2009.