Abstract

Antiinflammatory Effects of Withaferin A in Islet Transplantation and Pancreatitis

Mazhar A. Kanak, Ph.D.

Mentor: Bashoo Naziruddin, Ph.D.

Islet transplantation is a promising treatment for patients severely affected by type

1 diabetes and chronic pancreatitis. Islets are transplanted into the portal vein of the liver, and direct contact of islets with blood initiates a strong innate immune response called instant blood-mediated response (IBMIR). Approximately 50% to 60% of islets may be lost during the peritransplant period due to IBMIR. A known as nuclear factor kappa B (NFκB) primarily mediates the proinflammatory response in islets. Preliminary studies using withaferin A (WA), a plant-derived inhibitor of NFκB, showed protection of islets from proinflammatory cytokine-mediated damage.

An in vitro tube model for IBMIR was developed to determine the inhibitory effect of

WA. After transplantation, islet damage was evaluated using C-peptide and proinsulin levels in serum, which are influenced by metabolic stimuli. Identification of a biomarker specific for islet damage uninfluenced by

physiological changes is a critical need. MicroRNA 375 is highly expressed in β cells of

islets, and damage of islets results in release of this miRNA into the bloodstream.

Analysis of serum samples during and after islet infusion revealed high levels of

microRNA 375, indicating damage by IBMIR, and the estimated islet loss was

comparable to that in previous reports. Serum microRNA 375 was also analyzed to

validate the inhibitory effect of WA on IBMIR in vitro.

Chronic pancreatitis is an inflammatory disease affecting the exocrine and

subsequently endocrine function of the pancreas. Currently, there is no therapeutic

approach to delay progression of the disease. The underlying mechanism of pathogenesis

of chronic pancreatitis is not clearly understood. The activation of NFκB in acinar cells

has been reported to enhance the severity of chronic pancreatitis. NFκB inhibition by WA

was tested in a cerulein-induced pancreatitis mouse model. The sustained inflammation

that leads to chronic pancreatitis is potentiated by endoplasmic reticulum stress and inflammasome activation within the pancreas. Treatment with WA prevented activation of these mechanisms, as evidenced by molecular analysis and histological assessment.

In summary, inhibition of NFκB by WA could be a promising strategy to treat chronic pancreatic diseases. Antiinflammatory Effects of Withaferin A in Islet Transplantation and Pancreatitis

by

Mazhar A. Kanak B. Tech., M.S.

A Dissertation

Approved by the Institute of Biomedical Studies

Robert Kane, Ph.D., Chairperson

Submitted to the Graduate Faculty of Baylor University in Partial Fulfillment of the Requirements for the Degree of Doctor of Philosophy

Approved by the Dissertation Committee

Bashoo Naziruddin, Ph.D., Chairperson

Robert Kane, Ph.D.

Sangkon Oh, Ph.D.

Christopher Kearney, Ph.D.

Michael Trakselis, Ph.D.

Michael C. Lawrence, Ph.D.

Accepted by the Graduate School August 2015

J. Larry Lyon, Ph.D., Dean

Page bearing signatures is kept on file in the Graduate School. Copyright © 2015 by Mazhar A. Kanak

All rights reserved TABLE OF CONTENTS

LIST OF FIGURES……………………………………………………………………………..viii

LIST OF TABLES………………………………………………………………………………...x

LIST OF ABBREVIATIONS………………………………………………………………….....xi

ACKNOWLEDGMENTS……………………………………………………………………....xvi

DEDICATION…………………………………………………………………………………xviii

CHAPTER ONE…………………………………………………………………………………..1

Introduction………………………………………………………………………………..1

Diseases of the Pancreas…………………………………………………………...2

Islet Transplantation……………………………………………………………….4

Causes of the Inflammatory Response in Islet Transplantation……………………4

Pancreas Donors…………………………………………………………………...6

Ischemic Time……………………………………………………………………..6

Islet Isolation Process……………………………………………………………...7

Pretransplant Islet Culture…………………………………………………………8

Peritransplant Inflammation: IBMIR……………………………………………...9

Posttransplant Inflammation……………………………………………………..19

Inflammatory Response from Islets……………………………………………...21

Antiinflammatory Strategies to Improve Islet Transplantation…………………..22

Withaferin A……………………………………………………………………..26

Future Directions…………………………………………………………………29

v CHAPTER TWO………………………………………………………………………………...31

Alleviation of Instant Blood-Mediated Inflammatory Reaction in Autologous Conditions through Treatment of Human Islets with Nuclear Factor-Kappa B Inhibitors…………………………………………………………………………………31

Introduction……………………………………………………………………....31

Materials and Methods…………………………………………………………...33

Results……………………………………………………………………………36

Discussion………………………………………………………………………..41

CHAPTER THREE……………………………………………………………………………...47

Evaluation of MicroRNA 375 as a Novel Biomarker for Graft Damage in Clinical Islet Transplantation…………………………………………………………….47

Introduction………………………………………………………………………47

Materials and Methods…………………………………………………………...49

Results……………………………………………………………………………51

Discussion………………………………………………………………………..58

CHAPTER FOUR………………………………………………………………………………..63

Delineating the Molecular Pathogenesis of Chronic Pancreatitis and Use of NFκB Inhibitor to Prevent its Progression……………………………………………….63

Introduction……………………………………………………………………....63

Materials and Methods…………………………………………………………...65

Results……………………………………………………………………………70

Discussion………………………………………………………………………..85

CHAPTER FIVE………………………………………………………………………………...92

Summary and Conclusions………………………………………………………………92

Summary…………………………………………………………………………92

vi Alleviation of Instant Blood Mediated Inflammatory Reaction in Autologous Conditions through Treatment of Human Islets with NFκB inhibitors………………………………………………………………….93

Evaluation of miRNA 375 as a Novel Biomarker for Graft Damage In Clinical Islet Transplantation………………………………………………….95

Delineating the Molecular Pathogenesis of Chronic Pancreatitis and Use of NFκB Inhibitor to Prevent its Progression…………………………….....97

Conclusions………………………………………………………………………99

Bibliography……………………………………………………………………………100

vii LIST OF FIGURES

Figure Page

1.1 Factors that induce an inflammatory reaction to islet grafts………………………………5

1.2 Flow diagram for method of miniaturized tube model……………………………………11

1.3 Autologous IBMIR activation in vitro and validation parameters……………………….11

1.4 Elevation of pro-inflammatory cytokines/chemokines in serum after IBMIR……………12

1.5 Activation of IBMIR in clinical TPIAT patient samples………………………………....13

1.6 Hypothetical Model of the mechanism of IBMIR………………………………………..14

1.7 WA improved syngeneic mice islet transplant outcome and reduced cytokine induced ………………………………………………………………………...27

1.8 Western blot showing inhibition of NFκB activation by WA…………………………….28

1.9 WA inhibits proinflammatory cytokine/chemokine production by islet treated with cytokines……………………………………………………………………………28

2.1 Viability of islets after culture in autologous blood……………………………………....36

2.2 Autologous islet damage and the impact of NFκB inhibitors…………………………….37

2.3 Inhibition of proinflammatory cytokine secretion by NFκB inhibitors…………………..39

2.4 Neutrophil infiltration in islet clots blocked by NFκB inhibitors………………………...40

2.5 Tissue factor (TF) expression in islets after exposure to autologous blood………………41

3.1 miR375 expression correlates with islet mass and increases similarly to C-peptide in response to islet damage in a miniaturized in vitro tube model……………………….53

3.2 WA blocked islet damage by IBMIR: miRNA 375, c-peptide and proinsulin…………..55

3.3 miRNA levels in vivo indicate immediate islet damage after infusion…………………..56

3.4 Standard curve plot for the quantification of miRNA375……………………………….57

viii 3.5 Correlation of miRNA 375 levels and IEQ transplanted………………………………...57

4.1 Protocol for development and treatment of chronic pancreatitis………………………...67

4.2 WA reduces severity of AP and abolishes immune cell infiltration……………………..72

4.3 WA can prevent development of Chronic Pancreatitis (CP)….…………………………76

4.4 Withaferin A (WA) can abrogate progression of severe chronic pancreatitis (CP)- therapeutic model………………………………………………………………………...77

4.5 WA reduces cerulein induced acinar damage and blocks neutrophil infiltration………..78

4.6 WA inhibits cerulein induced NFκB in acinar cells and downregulates expression of proinflammatory and pro-apoptotic molecules………………………………………..80

4.7 Withaferin A (WA) downregulates cerulein-induced expression of proinflammatory and proapoptotic molecules in therapeutic CP model…………………………………....81

4.8 WA reduces endoplasmic reticulum (ER) stress……………...…………………………83

4.9 WA reduces expression of inflammasome related genes……………….…………….....84

4.10 Real-time PCR analysis of ER stress and inflammasome related genes in therapeutic model………………………………………………………………….……..86

4.11 Heat map of proinflammatory and proapoptotic genes in healthy and CP patients..…….87

4.12 Schematic representation of the signaling pathways operative in the incidence of CP and the blockade by WA……………………………………………………………...... 91

ix LIST OF TABLES

Table Page

1.1 Antiinflammatory agents in clinical allogeneic islet transplantation…………………….25

3.1 Patient and transplant characteristics in autologous recipients for sub-analysis………...54

x LIST OF ABBREVIATIONS

ANOVA Analysis of variance

AP Acute Pancreatitis

AP-1 activator protein 1

APC Antigen Presenting Cells

ASC Apoptosis associated Speck-like protein

ATF Activating transcription factor

ATP

BD Brain dead

BSA Bovine serum albumin

CAPS Cryopyrin associated periodic syndromes

CARD activation and recruit domains

Casp

CAY CAY 10512 (potent analog of reservatrol)

CCL CC chemokine ligand

CD cluster of differentiation cDNA complementary deoxyribonucleic acid

CER Cerulein

CFTR Cystic Fibrosis transmembrane conductance regulator

CHOP C/EBP homologous protein

CIT Consortium of islet transplantation

xi CMRL Connaught Medical Research Laboratories

CO2 Carbon dioxide

CP Chronic pancreatitis

C-peptide connecting peptide

CXCL Chemokine (C-X-C motif) ligand

CXCR Chemokine(C-X-C motif) receptor

CYT Cytokine cocktail

DAB 3,3'-Diaminobenzidine

DAMPs Damage associated molecular patterns

DAPI 4',6-diamidino-2-phenylindole

DC Dendritic cell

dsDNA double stranded DNA

Egr Early growth response protein

Eif2a Eukaryotic translation initiation factor 2a

ELISA enzyme linked immunosorbent assay

EP3 Prostaglandin E receptor 3

ER endoplasmic reticulum

ERK Extracellular signal-regulated kinases

FIH Factor inhibiting HIF

FITC Fluorescein isothiocyanate

G-CSF Granulocyte-colony stimulating factor

H&E Hematoxylin and Eosin

HbA1c Hemoglobin A1c

xii

HFD High Fat diet

HIF Hypoxia inducible factors

HLA human leukocyte antigen

HMGB1 High mobility group box-1

HO-1 Heme oxygenase 1

hrs hours

HSP Heat shock proteins

i.p. intraperitoneal

IACUC Institutional Animal Care and Use Committee

IBMIR Instant Blood Mediated Inflammatory Reaction

IEQ islet equivalents

IFN Interferon

IgG Immunoglobulin G

IgM Immunoglobulin M

IHC immunohistochemistry

IKKβ IκB kinase β

IL Interleukin iNOS inducible Nitric oxide synthases

IP-10 Interferon gamma inducible protein 10

IκB inhibitor of Kappa B

JNK c-Jun N-terminal kinases

LMWDS low molecular weight dextran sulphate

LPS lipopolysaccharide

xiii MAPK mitogen-activated protein kinase

MCP1 monocyte chemotactic protein 1

MIF Macrophage migration inhibitory factor mins minutes

MIP1α Macrophage inflammatory protein 1α miR miRNA miRNA microRNA

MKK MAPK kinase mmHg millimeter mercury mRNA messenger RNA

NALP3 NACHT, LRR and PYD domains-containing protein 3

NEMO NFκB essential modulator

NFAT Nuclear Factor of activated T cells

NFκB Nuclear Factor Kappa B

NKT natural killer T cells

NLR NOD-like receptors

NLRP3 NOD-like receptor family, pyrin domain containing 3

NOD non-obese diabetic

PAMPs Pathogen associated molecular patterns

PCR polymerase chain reaction

P-ERK phosphorylated ERK

PERK protein kinase R (PKR)-like endoplasmic reticulum kinase

PHD Prolyl hydroxylase

xiv

PMN Polymorphonuclear cells

pO2 partial pressure of oxygen

PP Pancreatic polypeptide

qRT quantitative realtime

RNA ribonucleic acid

ROS

rRNA ribosomal RNA

SPINK1 serine inhibitor Kazal-type 1

T1D Type 1 diabetes

TAT Thrombin-antithrombin

TBS Tris Buffered Saline

T-ERK total ERK

TF Tissue Factor

TLR toll like receptors

TNF

TP total pancreatectomy alone

TPIAT total pancreatectomy auto islet transplantation

TRITC Tetramethylrhodamine

TUNEL Terminal deoxynucleotidyl transferase dUTP nick end labeling

UPR Unfolded protein response

WA Withaferin A

XBP1 X-box binding protein 1

xv ACKNOWLEDGMENTS

First and foremost, I would like to thank Almighty GOD for giving me the strength and determination to complete my dissertation successfully. I would like to thank my family members for their considerable support and encouragement, without which this may not have been possible.

I am truly grateful to my mentor Dr. Bashoo Naziruddin for believing in me and guiding

me through the course of my research. I have learned a lot under his guidance and I wholeheartedly

appreciate his unceasing motivation that kept me moving forward. He has not only been a mentor

to me but also a great friend to share my ups and downs in research. He has always been optimistic

and taught me the same approach.

I would like to thank Baylor University and the graduate program for giving me the

opportunity to become a doctoral candidate and providing me with the support for doing research.

I am grateful to Dr. Robert Kane and the department of biomedical studies for making me a part

of their graduate team. I am sincerely thankful Dr. Kane for being there to answer any of my

questions both scientific and department related.

I would like to extend my humble gratitude to my committee members Drs. Christopher

Kearney, Sangkon Oh, and Michael C. Lawrence for being there to strengthen my dissertation to

the fullest. I would especially like to acknowledge Dr. Lawrence for his time and ideas that helped

me develop my research.

I have been blessed with the best lab members and colleagues who have supported me, and

helped me in every step of my research. There are so many people I would like to acknowledge

my gratitude but I may not be able to thank everyone specifically. Although, I would like to

xvi acknowledge some key members who have helped me get through this difficult path very easily. I would like to thank Drs. Levy, Itoh, Shahbazov, Faisal, Gumpei and many others. I would also like to acknowledge my colleagues Jeff Sorelle, Omar Khan, Ana Rahman, Yoshiko Tamura, Nofit

Borenstein, Regina Whitaker and last but not the least Dr. Carson for helping me with the completion of my course requirements for PhD. Finally, I would like to thank each and every member who has been a part of life and has helped me through my career path.

xvii DEDICATION

To my loving family

xviii CHAPTER ONE

Introduction

Part of this chapter published as: Kanak MA, Takita M, Kunnathodi F, Lawrence MC, Levy MF, Naziruddin B. Inflammatory response in islet transplantation. Int J Endocrinol 2014;2014:451035.

The pancreas is a vital organ of the gastrointestinal tract responsible for exocrine and endocrine secretions required to digest food and maintain glucose homeostasis.

Approximately 97% to 99% of the adult pancreas is the exocrine compartment (acinar cells), and about 1% to 3% is the source of endocrine cells (islets of Langerhans)1. The acinar cells produce a number of enzymes that are required for the digestion of food. The role of islets of Langerhans is quite different from that of acinar cells; their major function is to maintain glucose homeostasis. Islets of Langerhans are a cluster made up of at least 4 different types of endocrine cells. Most cells in the islet are β cells, which are responsible for production of insulin. The next most abundant cell type is α cells, which produce glucagon, which is also required for the maintenance of glucose homeostasis.

Other cell types that are present in an islet are δ cells and PP cells, which synthesize somatostatin and pancreatic polypeptide, which are required for absorption of food and appetite, respectively2. Both the exocrine and the endocrine compartments are prone to inflammation, which could result in diseases such as pancreatitis and/or diabetes.

This chapter reviews islet transplantation as treatment for two diseases of the pancreas: type 1 diabetes (TID) and chronic pancreatitis (CP). It highlights the inflammatory response in islet transplantation in preparation for Chapter Two, which

1 presents a study of alleviation of the instant blood-mediated inflammatory reaction

(IBMIR) through treatment of islets with nuclear factor-kappa B (NFκB) inhibitor.

Chapter Three continues the focus on clinical islet transplantation, evaluating

microRNA375 as a novel biomarker for graft damage. Chapter Four moves to the disease

of CP and presents research on use of NFκB inhibitor to prevent disease progression. The

dissertation closes with a summary and conclusions.

Diseases of the Pancreas

Type 1 Diabetes

T1D is an autoimmune disease that leads to destruction of the β cells of the

pancreas3. Due to the lack of β cells, patients with T1D suffer from hyperglycemia and other long-term complications related to it. T1D has afflicted more than 135 million people worldwide and in the United States, and an incidence of 15 to 18 per 100,000 people has been recorded per year4. The major focus of research in this field is on understanding triggers that lead to the onset of T1D, which is critical to strategize treatment options. Currently, exogenous administration of insulin is the prescribed therapy to maintain normoglycemia5. T1D has been known to cause chronic

complications, which are linked to morbidity and mortality6.

A major symptom of the disease is the unpredictable changes in glycemic levels, with sudden hyper- and hypoglycemic conditions that may result in unconsciousness, coma, and even death7. This condition, called “brittle” diabetes, can only be treated by

replacing the patient’s β cells with functioning β cells by whole-organ pancreas

transplantation or islet cell transplantation8. Although pancreas transplantation is a

2 promising therapeutic option, the surgery is complicated with an increased risk of

mortality9. Islet cell transplantation is growing in recognition as a potential treatment option for T1D patients because it restores functional β-cell mass, reduces the risk of long-term complications of diabetes, abolishes hypoglycemic episodes, and most importantly is a minimally invasive procedure10.

Chronic Pancreatitis

CP is a progressive inflammatory condition that leads to pancreatic exocrine insufficiency and irreversible damage of the pancreatic parenchyma11. In its early stages,

the disease affects pancreatic exocrine function, which could be followed by impaired

endocrine function that results in the onset of diabetes mellitus, also referred to as type 3c

diabetes12, 13. The mechanisms underlying the development of CP are not clearly defined.

Inflammation, heavy alcohol consumption, pancreatic ductal obstructions, calcification,

sphincter of Oddi dysfunction, certain genetic mutations, and periampullary tumors are

considered the major causes of CP. In addition to gradual loss of exocrine and endocrine

function, complications of CP include biliary or duodenal stenosis and intractable pain.

Total or partial pancreatectomy followed by intrahepatic transplantation of

autologous islets has emerged as a promising approach to treat CP, due to its ability to

reduce or eliminate pain while retaining endocrine function. The first pancreatectomy

followed by autologous islet transplantation was performed at the University of

Minnesota in 197714. Since then, more than 500 CP patients have successfully undergone

this procedure at several centers in North America and worldwide15. At 3 years

posttransplant, 30% of patients were insulin independent, 33% had partial islet function,

and 94% showed improved pain control16.

3 Islet Transplantation

Transplantation of pancreatic islets, a minimally invasive procedure involving

infusion of islet cells into the portal vein of the liver, was first demonstrated in an

experimental diabetic model by Lacy et al. in 197317. Results from the initial clinical

trials for T1D were only partially successful, with only 10% of the patients achieving

insulin independence at 1 year posttransplantation18. A quantum leap in islet transplantation occurred when the Edmonton group introduced a steroid-free immunosuppression and showed insulin independence in all 7 of their patients19. With

this advancement in immunosuppression and the continuous improvement in islet

isolation techniques, islet transplantation has entered a new era of heightened success.

Currently, two types of clinical islet transplantation are performed: allogenic and

autologous. Allogenic islet transplantation is typically performed on patients with severe

T1D, while autologous islet transplantation is performed on patients suffering from

severe CP and undergoing partial or total pancreatectomy. Compared with similar doses

of allogeneic islets, survival of autologous islets is far better over time16. In contrast to

allogeneic islet transplants, autologous islet transplants are devoid of an autoimmune

response and alloimmune rejection, and recipients do not require β-cell–toxic immuno- suppressive drugs. Similar to intrahepatic allogeneic islet transplants, however, autologous islet transplants are subject to ischemic, hypoxic, and innate inflammatory damage.

Causes of the Inflammatory Response in Islet Transplantation

Despite recent progress in clinical outcomes, several obstacles for allogenic and

autologous islet transplantation still need to be addressed. These include improvement in

the technical aspects of the isolation procedure; improvement in the quality of isolated

4 islets; reduction of inflammation during the peritransplant period triggered by incompatibility between islets and the blood interface, known as IBMIR; and improvement in the long-term survival of transplanted islets. One common underlying cause in each of the obstacles to clinical success is the production and secretion of inflammatory mediators, which induce apoptosis and impair function of transplanted islets. This section addresses the causes of inflammation in islets, including inflammation related to donor factors, islet isolation, pretransplant islet culture, the transplant itself, and posttransplant factors (Figure 1.1). Special attention is given to IBMIR and the center’s study of IBMIR in autologous islet transplantation. The sections that follow highlight strategies to reduce inflammation and discuss future directions.

Donor-induced inflammation Post-transplant hypoxia Cytokine storm Ischemia injury

Islet isolation Complement activation Pancreas digestion Hypoxia Mechanical stress Coagulation

Peri-transplant inflammation Activated innate immunity Cytokine induced

Figure 1.1. Factors that induce an inflammatory reaction to islet grafts.

5 Pancreas Donors

Damage to the islet cells begins with the donor. Immune cell infiltrates and

inflammatory mediators are produced in the donor pancreas upon brain death. Most islet

transplants use organs from heart-beating, brain-dead cadaveric donors. Organs from

brain-dead donors have much better outcomes than organs from non–heart-beating

donors. However, acute physiological changes after brain death in brain-dead donors may

still result in significant damage to the islets from inflammatory events. Indeed, several

reports have documented poor transplantation outcomes due to organ damage caused by

extremely high levels of inflammatory cytokines in the circulation20-23. This systemic

elevation of inflammatory cytokines, the so-called “cytokine storm” in brain-dead donors,

is due in part to hemodynamic and metabolic changes occurring after brain death24.

Inflammatory cytokines are effective mediators of β-cell dysfunction and death25.

Recent studies have indicated methods to improve islet graft function by

inhibiting inflammatory mediators in brain-dead donors upon procurement. One study

showed that maintaining normoglycemia after brain death by insulin reduces

inflammatory shock26. Transfusing sivelestat sodium, a selective neutrophil elastase

inhibitor, was shown to increase islet yield, protect islet function and quality, and inhibit

hypercytokinemia-mediated β-cell death in a brain-death rat model27.

Ischemic Time

Islets are subjected to hypoxic conditions from the onset of ischemia until graft revascularization. Hence, ischemic time is a major concern that needs to be addressed to

improve islet quality for transplantation. Since islets are physiologically adapted to

receiving an ample supply of oxygen, prolonged ischemic times increase damage to

6 islets. A previous study showed that isolations performed with a pancreas with a cold ischemic time of >8 hours produced significantly lower islet yields, with impaired islet function28. The quality of the islets can be improved by reducing organ ischemic time.

Another strategy used to reduce islet ischemic damage has been to maintain the procured pancreas in a high-oxygen-content solution. A mixture of University of Wisconsin solution along with perfluorocarbon, which has high affinity for oxygen, has been used to reduce islet damage by hypoxia29.

The viability of islets is significantly reduced due to depletion of ATP and apoptosis induced by hypoxia30. Hypoxia in islets activates NFκB signaling, induces transcription of inducible nitric oxide synthase (iNOS), and increases expression of monocyte chemotactic protein 1 (MCP-1), leading to infiltration of macrophages and destruction of islets31. During hypoxia, islets try to compensate for the low oxygen availability by activating the transcription factor hypoxia-induced factor (HIF)32. When activated, the HIF-1α and HIF-1β subunits translocate to the nucleus and bind to the hypoxia response element. This induces transcription of several genes, including toll-like receptors. Under normal conditions, HIF activity is rendered inactive by prolyl hydroxylases and factor-inhibiting HIF, both of which prevent the activation of inhibitor of kappa B (IκB) kinase B and hence inhibit NFκB activation33-37.

Islet Isolation Process

Isolation of high-quality islets in sufficient quantity remains a major impediment for the success of this procedure, due to chronic inflammatory conditions affecting the pancreas. The enzymes used for islet isolation could be a factor in the lack of satisfactory results in islet transplantation. It is well documented that enzymatic and mechanical stress

7 can induce inflammatory mediators in islets. Gene array studies have shown upregulation of genes related to inflammation, apoptosis, cell growth, and immediately after isolation, with a further increase after 72-hour culture. The most highly upregulated genes have been the ones for inflammation and apoptosis. Isolation and culture of islets have also been shown to downregulate expression of interleukin (IL)-10, which is a negative regulator of cytokine expression and also induces development of regulatory T cells. Conversely, isolation and culture of islets have upregulated the expression of IL-8, which is a potent inflammatory and angiogenic factor, and the expression increased several fold after 3 days in culture38. IL-8 has been shown to play a negative role in the longevity of the graft in kidney, liver, and lung transplants; overexpression of IL-8 has been associated with poor graft life39-41. Gene array analysis of cultured islets has also shown that the top 15 upregulated genes were all induced by the NFκB pathway38.

Therefore, inhibiting the NFκB pathway during isolation and/or culture might improve islet quality for transplantation.

Pretransplant Islet Culture

There has been a continuous debate as to which islets are superior: freshly isolated or cultured islets. Several reports have shown that freshly isolated islets are better than islets cultured for at least 24 hours42-44. A recent report from our center confirmed that freshly isolated islets outperformed cultured islets to address concerns over the rapid deterioration of islet counts after culture. The study showed a decrease in the islet counts by 13%, 24%, and 35% on days 1, 2, and 3, respectively. When transplanted into diabetic nude mice, the achievement of normoglycemia was significantly improved with fresh islets compared to cultured islets45, 46. It has been reported that culturing islets can lead to

8 overexpression of inflammatory mediators caused directly by islets, by stimuli in the

culture medium, or by contamination of exocrine tissue in the culture47. During culture,

islets have to depend on oxygen through diffusion, but the core of the islet begins to

undergo hypoxia, causing upregulation of genes induced by and

apoptosis48. Islets in the culture have been shown to induce expression of proteins such as

tissue factor and MCP-1; this induction could be abrogated by the use of nicotinamide in

the culture media49. Still, there remains the argument that culturing islets before transplantation will improve the purity of the islets, as acinar cells and damaged islets would die in the culture. It has also been suggested that providing a culturing period would allow islets to recover from the stress of the isolation process, although there is no

compelling data to support this hypothesis. Nonetheless, the Consortium of Islet

Transplantation has adopted pretransplant culture as a standard procedure for clinical

transplantation50.

Peritransplant Inflammation: IBMIR

One of the major causes of islet destruction during transplantation is IBMIR

(Figure 1.6). Bennet et al. first proposed that when freshly isolated “naked” islets come into direct contact with the ABO-compatible allogenic blood, activation of an innate

immune reaction ensues51. Tissue factor expressed on islets could be a major trigger for

such reactions. The islets, along with resident antigen-presenting cells (APCs), secrete cytokines and chemokines, which play a major role in the inflammatory process.

Infiltration of leukocytes and macrophages initiates destruction of the islet cells before engraftment51, 52. Blocking tissue factor using antibodies significantly reduced the clotting

reaction52.

9 IBMIR and Autologous Islet Transplantation

Although previous studies showed induction of IBMIR under allogenic and

xenogeneic conditions51, 52, our group showed for the first time that IBMIR was also an

issue during autologous islet transplantation. Blood collected from clinical transplant

patients at the time of transplantation was used to analyze markers of coagulation,

complement activation, and inflammatory cytokine secretion53. To demonstrate the

mechanism of IBMIR in vitro, I developed a miniaturized tube model. The model was

modified from the in vitro blood loop model developed by Korsgren et al54. Islets come in direct contact with the blood of the portal vein and flow with the bloodstream until they lodge in the liver. To mimic the same scenario of islets in the intraportal milieu, isolated human islets were added to autologous human blood in a heparinized Eppendorf tube and then the cells were placed in a 37°C shaker incubator and the plasma samples were collected at various time points (Figure 1.2). The in vitro miniaturized tube model was validated by measuring parameters such as partial pressure of oxygen, pH, and the glucose content of blood for up to 6 hours in the presence or absence of islets. The data suggested that the in vitro model was suitable for investigating IBMIR for up to 6 hours

(Figure 1.3). Results obtained from the in vitro experiment corroborated results of blood

samples from clinical autologous islet transplant patients, suggesting that IBMIR

occurred in autologous islet transplantation (Figures 1.3 and 1.4).

10 Figure 1.2. Flow diagram of the method for in vitro IBMIR using a miniaturized tube model.

Figure 1.3. Data demonstrating autologous IBMIR activation in vitro and validation parameters. The mixed blood and islet group (solid line) had significantly increased TAT (A) and C-peptide (B) levels (*p with repeated measured ANOVA < 0.05) although the blood-only (dotted line with squares) and islet-only samples (dotted line with triangles) did not. C3a levels were elevated in both the mixed blood-islet and the blood-only samples, but not in the islet-only samples (C). Culture conditions on pH (B), pO2 (D) and glucose level (F) were determined. The mixed blood and islet group showed similar trend of pH and glucose level to blood only group. Although pO2 levels were significantly lower in mixed group than blood only group (*p<0.05, D), pO2 level over of 60 mmHg was maintained for 6 hours in the mixed group. Data are expressed as mean ± SE.

11 Figure 1.4. Elevation of proinflammatory cytokines and chemokines in serum after IBMIR. The mixed blood and islet group had significantly higher proinflammatory cytokine levels of MCP-1 (A) and TNF-α (B) in culture for 6 hours when compared to blood only group (*p with repeated measurement ANOVA <0.05). Data are expressed as mean ±SE.

Serum samples collected from patients undergoing total pancreatectomy with islet

autotransplantation were collected at various time points during and after islet infusion.

Thrombin-antithrombin levels increased in these samples immediately after infusion of

islets, suggesting activation of the coagulation pathway; a decrease in platelet number

also suggested the same. C-peptide has been regarded as a marker for islet damage during

infusion. Elevation of C-peptide was seen immediately after infusion, suggesting that

islets were being destroyed in the mechanism. Interestingly, in contrast to allogeneic and

xenogeneic transplants, autologous islet transplantation did not cause complement

activation (Figure 1.5). This may suggest that the mechanism of IBMIR may be less

severe in the case of autologous islet transplantation; nonetheless, its effects are drastic.

Mechanisms Underlying IBMIR and Strategies to Block IBMIR

The IBMIR effect has been characterized by an immediate increase in the

thrombin-antithrombin levels after islet infusion and in the levels of C-peptide in the

12 blood. A recent clinical study utilized labeled islets combined with positron emission tomography and computed tomography and demonstrated that about 25% of the islets are lost immediately after transplantation55. IBMIR is also characterized by coagulation, complement activation, secretion of chemokines that attract innate immune cells, and release of proinflammatory cytokines leading to islet damage by apoptosis56. The mechanisms underlying IBMIR and methodologies to block this phenomenon are currently being investigated, as described below.

Figure 1.5. Activation of IBMIR in samples from patients undergoing total pancreatectomy with islet autotransplantation. Significant differences in TAT levels were observed for the middle of infusion to 1 hour after infusion when compared to the pre-infusion level (*p with repeated ANOVA < 0.05) (A). C- peptide levels for middle of infusion to day 7 were significantly higher than prior to infusion (*p <0.05, C); however, no significant differences were found in C3a and C4d levels (B and D). sC5b-9 levels had two peaks at islet infusion and day 5; however, these changes were within normal range and no significant differences were observed during peritransplant period up to day 7 when compared to pre-infusion level (F). Platelet counts significantly decreased at 1 hour to 3 days after infusion (*p <0.05) (E). Shadow area shows normal range. Data are expressed as mean ± SE for 10 patients.

13 Figure 1.6. Hypothetical model showing the mechanism of instant blood-mediated inflammatory reaction. IgM indicates immunoglobulin M; MCP-1, monocyte chemotactic protein 1; PMN, polymorphonuclear leukocytes; TF, tissue factor.

Coagulation: Coagulation is activated as soon as the islets come in direct contact with the portal blood during transplantation. The coagulation occurs through the extrinsic pathway, which requires tissue factor for activation. Tissue factor expressed on the surface of isolated islets interacts with factor VIIa and initiates the coagulation cascade by formation of thrombin and generation of fibrin clots52, 57, 58. The production of thrombin at high concentrations can exacerbate the destruction process because at high concentrations thrombin is proinflammatory and an inducer of apoptosis59, 60.

14 Nilsson et al. showed that the use of anticoagulants such as melagatran could

significantly reduce IBMIR in vitro in a blood loop model54. Johansson et al. previously

showed that low-molecular-weight dextran sulfate (LMWDS), a potent inhibitor of

coagulation and complement activation, could efficiently suppress IBMIR. They used an

in vitro tubing loop model to investigate the effect of LMWDS on islets when exposed to

human blood, showing effects on activated partial thromboplastin time, platelet function,

and complement activation61. Other strategies to block IBMIR that have been intensely

investigated have included encapsulating islets or coating the islet surface with inhibitory

molecules to promote successful islet engraftment. The islet surface has been coated with

heparin complex by exploiting biotin-avidin chemistry to inhibit coagulation62.

Endothelial cells have also been used to coat the islets; transplantation of these coated

islets has reduced coagulation, complement activation, and leukocyte infiltration63, 64.

Alternatively, the use of mesenchymal stem cell–coated islets has been shown to promote endothelial cell proliferation, revascularization, islet neogenesis, and immune modulation65. Another study showed that IBMIR could be targeted by overexpressing

CD39, which is an ectonucleotidase that degrades ATP required for platelet activation in the islets. In vivo and in vitro experiments showed less coagulation activity after transplantation using islets from CD39 overexpressing transgenic mice66.

Complement activation: Another major component of IBMIR is the activation of

the complement cascade. Complement activation is shown by the increase in the

concentration of complement proteins in the serum of islet transplant recipients56, 67.

Activation of complement protein C3a and C5a leads to recruitment and accumulation of

leukocytes, upregulation of adhesion molecules on the and platelets, and

15 production of reactive oxygen species (ROS) and cytokines67. Also, the accumulation of

IgG, IgM, and complement proteins C3, C4, and C9 on the surface of human islets

immediately after treatment with ABO-compatible blood indicates that immunoglobulin

is involved in the activation of the classical complement pathway67.

Complement protein C5a has been proposed as a critical molecule responsible for

activation of coagulation and inflammation by mediating tissue factor expression in

neutrophils68, 69. A recent short report from Tokodai et al. showed that the combined

effect of anticoagulant gabexate mesilate and complement protein C5a inhibitory peptide

successfully improved islet transplantation compared to individual use of the drugs in a

syngeneic rat transplant model70. Thrombin-antithrombin levels were also reduced with

the combined treatment. Tissue factor was downregulated by complement protein C5a

inhibitory peptide treatment, but only on the liver granulocytes and not on the islet

grafts71.

The use of a complement inhibitor such as compstatin has been shown to

significantly improve graft survival in in vivo models72. In a recent study, it has been

reported that immobilization of soluble complement receptor on the islet surface using

polyethylene glycol inhibited complement activation significantly and protected islets

from damage due to xenoreactive antibodies73. In clinical autologous islet transplantation,

complement proteins have no influence during IBMIR53.

Cytokine secretion, leukocyte infiltration, and β-cell damage: Syngeneic transplant models have shown that the damage to islet grafts is primarily due to a nonspecific inflammatory response74, 75. Similarly, clinical data on autologous islet

transplantation on patients with CP showed elevated levels of proinflammatory cytokines

16 immediately posttransplantation53. The islet resident macrophages, Kupffer cells, and

neutrophils secrete IL-1β, which impairs insulin secretion and induces islet cell

apoptosis76-78. IL-1β activates NFκB signaling and induces expression of several other

inflammatory mediators, including IL-1β, IL-8, IP-10, IL-6, tumor necrosis factor (TNF)-

α, MCP-1, intercellular adhesion molecule-1, vascular cell adhesion molecule-1, endothelial leukocyte adhesion molecule-1, iNOS, prostaglandin E2, and prostaglandin

EP379-82. These mediators further induce a cascade of inflammatory events that have deleterious effects on islet graft survival.

In addition to several proinflammatory cytokines, islets are known to express chemokines after isolation and during culture, of which CXCL10 (IP-10), CXCL8 (IL-8),

and CCL2 (MCP-1) are the major ones38, 56. IP-10 is widely recognized as a chemokine

that attracts leukocytes to the site of infection or inflammation. On the other hand, IL-8

and MCP-1 are known to recruit neutrophils and macrophages, respectively. The role of

chemokine receptors and ligands in various transplant rejections has been reviewed

elsewhere83.

In the context of islet transplantation, it has been shown that allograft function is

prolonged under anti-CXCL10 antibody therapy due to the lack of infiltration of

leukocytes to the graft site84. Using CCL2–/– knockout mice, it was demonstrated that

reducing CCL2 expression in recipients is more important than knocking out CCL2 in

donors. Knocking out CCL2 in the recipient also resulted in abrogation of other

chemokines (CXCL1, CCL4, CCL6, and CXCL9) that were upregulated in the liver

posttransplantation85. A recent approach using reparixin, an inhibitor of CXCR1/2, has

shown an improved islet transplant outcome in allogeneic mouse models. Furthermore, a

17 pilot study on human islet allotransplant patients also showed promising results.

Comparison of reparixin-treated group with nontreated controls showed a reduced insulin requirement, lower hemoglobin A1c levels after transplantation, and improved fasting C- peptide levels. The proposed mechanism is the lack of recruitment of polymorphonuclear leukocytes and natural killer T cells to the liver86. Based on these reports, it is conceivable that reducing secretion of proinflammatory cytokines and chemokines during the initial stages of transplantation could improve islet transplant outcomes.

Several reports have shown that allogeneic islet transplant recipients develop antibodies to both human leukocyte antigen (HLA) class I and II antigens of donor origin87-89. While it is evident that islets constitutively express HLA class I antigens, the expression of HLA class II on isolated human islets has not been clearly demonstrated.

Upon stimulation with inflammatory cytokines of TNFα and interferon (IFN)-γ, islet- enriched pancreatic cultures and MIN6 cell lines have been shown to express HLA class

II antigens90, 91.

Recently, our group has demonstrated that under inflammatory conditions, there is induction of HLA class II expression on the islet cell surface leading to the development of anti-donor class II antibodies in the recipient92. Islets isolated from deceased donor pancreases were used to demonstrate induction of HLA class II molecules through real- time polymerase chain reaction analysis, immunofluorescent staining, and flow cytometry. One of the transplant recipients developed antibodies to a mismatched donor

HLA class II allele (DR7). Posttransplant serum from this patient showed significant binding to cytokine-stimulated islet cells from DR7-positive donors only. These results clearly demonstrate that human islets express HLA class II under inflammatory

18

conditions. This could lead to an anti-class II response, which in turn may play a critical

role in rejection of islet allografts.

Posttransplant Inflammation

Hypoxia

Like other transplantable organs, islets are prone to ischemic reperfusion injury.

When isolated islets are infused into the blood circulation, several cytotoxic products are

produced due to oxygen metabolism, which is catalyzed by xanthine oxidase93.

Activation of this enzyme results in the formation of superoxide and hydrogen peroxide,

which can interact with free iron and copper radicals to produce cytotoxic ROS94. ROS can induce cell death by damaging and degrading proteins, lipids, and nucleic acids and activating NFκB inflammatory pathways95. Βeta cells of the islets are more vulnerable to

attack to ROS because of their low expression of , including glutathione

peroxidase, catalase, and superoxide dismutase96-98. Indeed, increasing the expression of

cellular antioxidants and treatment with exogenous antioxidants have both been shown to

protect islets in a diabetes models99, 100.

Following intraportal infusion, there is a low oxygen supply at the graft site in the liver, creating a hypoxic environment for the islets101, 102. Islet cells have counter

mechanisms to restore the oxygen supply32. Eventually, islet grafts begin to vascularize, but the amount of oxygen supply even after hepatic vascularization is significantly lower than in the pancreas (5 mm Hg as opposed to 40 mm Hg)103. Although the liver is

currently the preferential site for islet transplantation, it is still not considered the optimal

host site because of the lack of oxygen; therefore, other sites that might improve oxygen

19 availability to the islets are still under consideration. Cantarelli et al., for example, have provided evidence that bone marrow may be a more suitable graft site for islet transplantation because of the increased vasculature and abundant supply of oxygen104.

Furthermore, pilot studies on clinical autologous islet transplants have been performed in bone marrow which outperformed islet grafts in the liver105.

Innate Immunity

Pancreatic islet inflammation after transplantation is induced by innate immune cells such as neutrophils, islet resident macrophages, and Kupffer cells. Kupffer cells can induce damage to other cells by releasing free radicals and secreting proinflammatory cytokines106. The amount of proinflammatory cytokines IL-1β and TNFα secreted by

Kupffer cells is increased after islet transplantation107. This inflammatory cytokine elevation is blocked in the case of a macrophage-depleted recipient. Several factors have been postulated to explain the activation of Kupffer cells in the liver after islet transplantation. One report suggested that the collagenase used for islet isolation contains endotoxin, which could activate production of proinflammatory cytokines by islet- resident macrophages, and in turn activate Kupffer cells108. The presence of acinar tissue in the islet transplant preparation has also been investigated as the cause for activation of

Kupffer cells. It has been demonstrated that the amount of exocrine contamination is directly correlated to posttransplant necrosis and lower viability of pancreatic islet cells.

It has been postulated that when these acinar cells die, they release enzymes that trigger an inflammatory response leading to islet cell death109.

Another hypothesis is that the professional APCs residing inside the islets are responsible for inducing the inflammatory response. A recent study on the intraislet

20 immune cell population showed that 67% of the immune cells in the islets are APCs, and

50% of these APCs are B cells110. Use of anti-inflammatory agents that block the

production and secretion of these proinflammatory cytokines and chemokines would be a

useful strategy to protect islets from innate immune cell-mediated damage. Recently we

have shown that use of withaferin A (WA), which is an NFκB inhibitor, protected islet

viability, reduced secretion of proinflammatory chemokines and cytokines, and reduced

infiltration of neutrophils in an in vitro tube model111.

Inflammatory Response from Islets

Islets can also produce inflammatory mediators in response to metabolic, inflammatory, physical, and chemical stress. For example, IL-1β induces production of

cytokines, chemokines, and iNOS in pancreatic β cells112-114. In the context of islet

transplantation, multiple islet stress events may contribute to cascading cytokine-induced

cytokine production in islets. Thus, cytokines produced and released by β cells

throughout the islet transplant process may exacerbate islet inflammation.

MAP kinases ERK1/2, p38, and JNK have roles in islet dysfunction and apoptosis

in response to stress-specific signaling pathways115, 116. Calcineurin-dependent ERK1/2

activation has short-term stimulatory effects on insulin production and islet function in

response to glucose and secretagogues; however, sustained activation due to metabolic

and inflammatory stress may have deleterious effects117-120. Upon exposure to cytokines,

JNK and p38 are activated by MAP kinase kinase (MKK) 4 and MKK3/6,

respectively116. Additionally, exposure of islets to stress from the islet isolation process

activates JNK and p38 via MKK7 signaling. One study showed that an increased

p38/JNK activation relative to ERK1/2 activation after islet isolation correlates with

21 decreased islet survival121. Hence, identifying methods to selectively block p38 and JNK

pathways, while preserving acute ERK1/2 signaling, may enhance islet graft function and

survival in islet transplantation122-126.

Acute exposure of islets to a low dose of IL-1β has been shown to stimulate

proliferation and enhance β-cell function, whereas exposure to high-dose IL-1β impairs

secretion and induces apoptosis127-129. Induction of TNFα expression in β cells by IL-1β

requires activation of calcineurin and downstream transcription factor nuclear factor of

activated T cells (NFAT)112. Extended exposure of β cells to IL-1β results in sustained

MAP kinase signaling, which activates NFκB-dependent genes that are proapoptotic112,

130, 131. Although a number of reports suggest that NFκB is responsible for apoptosis of transplanted islet cells132, other reports have presented conflicting evidence suggesting

that NFκB prevents islet cell death133. This is also likely attributed to the strength and

duration of exposure as well as the type of stimulus, as both NFAT and NFκB have

overlapping roles in cytokine gene expression and β-cell proliferation and function112, 133-

136. Thus, minimizing sustained activation of MAP kinase and NFAT/

NFκB signaling during the transplantation process is likely key to reducing islet

inflammation and improving islet transplant outcomes.

Antiinflammatory Strategies to Improve Islet Transplantation

Antiinflammatory agents have been incorporated into immunosuppressive

regimens in recent clinical allogeneic islet transplant protocols137, 138. A recent review has

summarized progress related to this approach24. In the era of 1999 to 2002, TNFα

inhibitors were utilized in a small proportion (11.8%) of islet transplants, while in the

2007 to 2010 period, they were used in 33.8% of transplants139. The first report of islet

22

transplantation involving an antiinflammatory agent came from the University of

Minnesota, where etanercept was incorporated in peritransplant immunosuppression

therapy140. All the islet recipients achieved insulin independence after a single islet

infusion. The University of Miami group implemented an immunosuppression protocol

containing infliximab, which resulted in a high success rate of insulin independence

(87.5%)141. Recently, a meta-analysis showed significant improvement in the insulin

independence rate when a TNFα inhibitor was coupled with a T-cell depletion regimen,

compared with T-cell depletion only142. The success rate with T-cell depletion

immunosuppression plus a TNFα inhibitor is now likely comparable to that of whole-

organ pancreas transplantation alone, offering approximately 50% success at 5 years

posttransplant139, 142, 143.

No significant difference in the success rate, however, was seen between a T-cell–

depleting islet protocol with and without infliximab in a clinical study141. In an

immunodeficient rodent model transplanted with a marginal mass of human islets, no

significant benefits in islet engraftment were observed when only etanercept was given,

whereas significant improvement in diabetes reversal was demonstrated when both

etanercept and anakinra, an IL-1β receptor antagonist, were administered144. Thus,

administration of a TNFα inhibitor alone might be insufficient to fully control

peritransplant inflammation. Interestingly, our group has implemented combined therapy

of etanercept and anakinra in clinical allogenic islet transplantation and has shown that all

patients achieved insulin independence after a single infusion, although the number of

islet recipients was limited145. A large, well-designed clinical study is required to evaluate the impact of these antiinflammatory agents in islet engraftment. The use of

23 antiinflammatory agents in clinical allogeneic islet cell transplantation is summarized in

Table 1.1.

A major requirement in clinical islet transplantation is to identify a biomarker that can be used to evaluate islet damage during and after transplantation. Currently, the tools that are used as islet damage markers are serum levels of C-peptide and proinsulin that can easily be altered depending on the metabolic state of the patient. C-peptide and proinsulin are both functional proteins that can be released upon glucose stimulation, and these can also yield false positive results if used as damage markers for islet transplantation.

Several antiinflammatory compounds are currently being investigated and more are emerging to improve the outcomes of islet transplantation. An exhaustive review on the strategies and approaches to improve clinical islet transplantation has been mentioned24. Pileggi et al. have shown that expression of heme oxygenase-1 (HO-1) in islet cells protected the islets from apoptotic death. To induce HO-1 in islets, the cells were cultured in ferriprotoporphyrin IX chloride and cobaltic protoporphyrin IX chloride before transplantation146. The HO-1 gene plays an important role in iron homeostasis, but also behaves as a potent antiinflammatory and antiapoptotic gene147, 148. A similar study has shown the antiapoptotic effect when islets are treated with carbon monoxide. This treatment causes islets to produce cytoprotective cyclic guanosine monophosphate and its downstream kinases149. Another strategy that has shown promise is the expression of the

A20 gene in islets by ex vivo gene transfer, which protects these islets from cytokine- induced damage. A20 is an inhibitor of NFκB, which is a transcriptional regulator of several proinflammatory cytokines150. A different study involving the use of diannexin,

24 which is a homodimer of annexin 5, has demonstrated a significant decrease in β-cell apoptosis and improved graft function after in vivo transplantation151. The use of JNK inhibitor SP600125 in combination with nicotinamide and simvastatin protected porcine islets from peritransplant inflammation and apoptosis. Intraductal administration of JNK inhibitor protected islets by inhibiting the production of proinflammatory cytokines IL-

1β, TNFα, IFN-γ, IL-6, IL-8, and MCP-1 in vivo152.

Table 1.1. Antiinflammatory agents in clinical allogenic islet transplantation.

Pub. Pt Induction Maintenance Insulin year no. Anti-inflammatory agents therapy therapy independent Ref 2005 8 Etanercept: 50 mg i.v. rATG Sirolimus 8/8 after 140 pretransplant and 25 mg s.c. on Daclizumab Tacrolimus single days 3, 7, and 10 MMF infusion 2005 8 Infliximab: 5 mg/kg i.v. Daclizumab Sirolimus 7/8 141 pretransplant Tacrolimus 2008 7 Infliximab or etanercept: 5 Daclizumab Sirolimus 6/7 153 mg/kg i.v. pretransplant or 50 Tacrolimus mg i.v. pretransplant and 25 mg (Low-dose s.c. twice weekly for 2 weeks steroids for 3 pts) 2008 3 Etanercept: 50 mg i.v. Alemtuzumab Sirolimus 2/3 154 Tacrolimus pretransplant and 25 mg s.c. MMF twice weekly for 2 weeks 2008 6 Etanercept: 50 mg i.v. rATG (for first Cyclosporine 5/6 155 pretransplant and 24 mg s.c. on transplant) Everolimus days 3, 7, and 10 Daclizumab (for 2nd transplant) 2008 6 Etanercept: 50 mg i.v. Daclizumab Sirolimus 6/6 156 pretransplant and 24 mg s.c. on Tacrolimus days 3, 7 and 10 2011 3 Etanercept and anakinra: 50 mg rATG Tacrolimus 3/3 145 i.v. pretransplant and 25 mg s.c. MMF on days 3, 6, and 10 and 100 mg i.v. pretransplant and 100 mg s.c. for 7 days after transplant 2012* 22 Anti-TNFα T-cell depletion NA 11/22 for 5 142 protocol years i.v. indicates intravenous; II, insulin independence; MMF, mycophenolate mofetil; rATG, rabbit antithymocyte globulin; s.c., subcutaneous; TNF, tumor necrosis factor.

25 Withaferin A

To counter inflammation, one approach we explored involved WA, a naturally

occurring plant-derived molecule. WA has been known for its multifunctional abilities,

including its strong antiinflammatory, , and antiangiogenic properties157. WA

is originally derived from a plant called , which has been used for

centuries as traditional medicine with proven clinical safety158, 159. Several compounds have been extracted from the leaves of Withania somnifera, but a study by Kaileh et al. identified WA as the most potent inhibitor of the transcription factor NFκB based on IκB kinase β (IKKβ) hyperphosphorylation preventing degradation of IκB160. Another group

of researchers suggested that WA prevents activation of NFκB by intercepting the

NEMO/IKKβ complex formation161. WA has been widely investigated as an

antiinflammatory compound in cystic fibrosis and also to treat other chronic

inflammatory diseases such as rheumatoid arthritis, asthma, and inflammatory bowel

disease162-164.

Proinflammatory cytokines such as IL-1β, TNFα, and IFN-γ have been shown to

cause apoptosis of islets by activation of the NFκB pathway165,166. Isolated islets express

tissue factor and chemokines MCP-1 and IL-8, which are regulated mostly by NFκB38.

Therefore, blocking NFκB activation in islets is a potential strategy to improve islet

transplant outcomes. To test this hypothesis, WA was used because of its strong

inhibition of NFκB164.

Using a syngeneic mouse islet transplant model, the effectiveness of WA to

protect transplanted islets from cytokine-mediated damage was demonstrated. Mouse

syngeneic islets were transplanted intraportally and normoglycemia was achieved after

26 WA treatment, but control mice could not reverse diabetes. Islets cultured in cytokines experienced a significant increase in apoptotic cells, which was effectively prevented by

WA pretreatment (Figure 1.7).

a 40 d 30

20

10

0

Blood glucose (mmol/l) 0 10 20 30 Days post transplant b 40 30 Control CYT CYT+0.5WA CYT+1.0WA 20 10 40 0 Blood glucose (mmol/l) 0 10 20 30 + Days post transplant 30 c 40 20 30

20

10 10 Percent TUNELPercent 0 Blood glucose (mmol/l) 0 10 20 30 0 Treatment Days Post Transplant Period Control Cyt Cyt+0.5WA Cyt+1.0WA

Figure 1.7. WA improved outcomes in syngeneic mice and reduced cytokine-induced apoptosis. (a) Intraportal islet transplantation into C57BL/6 mice had a success rate of 100% (3/3) with 400 islets, but (b) using 200 syngeneic islets with injection of 0.5mL PBS (vehicle) resulted in 0% (0/5) of recipients achieving normoglycemia. (c) Daily administration of WA (1.25mg/kg body weight) starting immediately after transplantation and lasting 7 days yielded a much higher success rate of 83% (5/6), (d) Cytokine mediated beta-cell apoptosis was confirmed by TUNEL staining. Human islets were stained for insulin (red) and apoptotic nuclei (green). Manual counting of more than 250 nuclei revealed higher levels of apoptosis when islets were exposed to cytokines, which was significantly reduced by the addition of WA, which returned apoptosis levels to control levels (1.0 μg/ml WA).

NFκB activation induced by cytokines was efficiently blocked by WA pretreatment of islets (Figure 1.8). Pretreatment of islets with WA significantly reduced the production of proinflammatory cytokines and chemokines regulated by NFκB (Figure

1.9).

27 A Control CYT CYT + WA B 6 Control MIN6 5 CYT CYT + WA INS-1 IκB 4 *

Human Islet

IkB:ERK 3

MIN6 Ratio 2

ERK INS-1 1 * * Human Islet 0 MIN6 INS-1 Human Islet

Figure 1.8. Western blot showing IκB degradation upon cytokine treatment but WA prevents it in β cell line and human islets. Immunoblot analysis of IκB protein in human islets and beta-cell lines treated with a cytokine cocktail (CYT) for 20 min in the presence or absence of WA. Experiments were repeated three times and performed in duplicate. (b) Graphical representations are expressed as means ± SD (black- control, grey-cytokine treated, white-cytokine+1μg/ml WA).

a d 8000 8000 IL-8 IP-10 6000 6000 *

4000 * 4000 * pg/Islet pg/ Islet * 2000 2000

0 0

b e 2500 60 MCP-1 MIP-1α 2000 50 40 1500 * 30 / Islet * 1000 * pg

pg/ Islet 20 * 500 10 0 0

c f 800 500 G-CSF IL-6 400 600 * 300 * / Islet 400

pg * 200 * pg/ Islet 200 100 0 0

Figure 1.9. Withaferin A inhibits proinflammatory cytokine/chemokine production by islet treated with cytokines. Quintuplets of fifty handpicked, human islets were cultured for 96h in cytokines or cytokines supplemented with WA at 37oC. Supernatant was collected and analyzed in duplicate with Millipore’s human cytokine detection kit in the Luminex 200®. Results were standardized to the number of islets. Results shown are representative of three independent human islet preparations. Graphical representations are expressed as means ± SD. Significant differences compared to CYT as determined by Turkey-Kramer post-hoc tests are indicated by asterisks (p<0.05).

28

Based on these results, WA was chosen for further investigation of the role of

NFκB inhibition in the alleviation of IBMIR and also to delineate the mechanism that leads to the pathogenesis of CP and to prevent its occurrence.

Future Directions

Inflammation during islet transplantation is multifaceted, and the underlying mechanisms are quite diverse. Several factors could play critical roles in the induction or exacerbation of inflammation. Therefore, it is challenging to identify one particular approach or strategy to eliminate all inflammatory events in islets. A combinatorial approach based on the current understanding of inflammatory mechanisms will likely be required to improve islet transplant outcomes. Overall, isolation of islets from the pancreas with less ischemic time is an important aspect. Furthermore, culturing islets in conditions that are suitable for islet survival and use of antioxidants and antiinflammatory compounds including potent inhibitors of NFκB, which are known to regulate several proinflammatory genes, is important. Since a significant mass of islets are immediately lost to IBMIR, another major focus should be to reduce this response. Although the use of anticoagulants or complement inhibitors shows benefits in in vitro and in vivo settings, they fail to perform effectively in clinical settings. Moreover, in allogenic transplants, the inflammation is severe, and therefore a combined antiinflammatory regimen to reduce ensuing immune response would be required. For instance, LMWDS has shown potential benefits to prevent clotting. This could be combined with antiinflammatory compounds such as anakinra and etanercept and with complement inhibitors to form a complete regimen to block IBMIR. Some of the recent clinical studies, such as use of reparixin to block CXCR1/2 have shown promising outcomes. Several inflammatory mechanisms that

29 cause dysfunction of islets that have been discussed here should be addressed at the clinical level. Future studies should focus on these mechanisms to develop strategies for successful islet transplantation.

30 CHAPTER TWO

Alleviation of Instant Blood-Mediated Inflammatory Reaction in Autologous Conditions through Treatment of Human Islets with Nuclear Factor-Kappa B Inhibitors

This chapter published as: Kanak MA, Takita M, Itoh T, SoRelle JA, Murali S, Kunnathodi F, Shahbazov R, Lawrence MC, Levy MF, Naziruddin B. Alleviation of instant blood-mediated inflammatory reaction in autologous conditions through treatment of human islets with NF-κB inhibitors. Transplantation. 2014;98(5):578-84.

Introduction

Two types of clinical pancreatic islet transplants are currently being performed:

allogeneic islet transplants for those with type 1 diabetes and autologous islet transplants

for patients undergoing total or partial pancreatectomy for refractory chronic

pancreatitis16. After isolation, the islets are generally transfused intraportally, as the liver is considered the preferred site167. A significant number of patients become insulin

independent after transplantation, but this result is transient, and most of these patients

return to using exogenous insulin168. This is a clear indication of poor engraftment and/or

gradual decay of islet function, which could be due to several factors, including poor islet

quality, hypoxia, damage due to instant blood-mediated inflammatory response (IBMIR),

and in the case of allogenic islets, autoimmune and alloimmune responses138, 169.

Previous reports have shown that IBMIR is a significant factor in the damage to

allogenic and xenogenic islet transplants during the peritransplant period52, 54, 72. IBMIR

is a nonspecific response mediated by the innate immune system characterized by

coagulation, complement activation, platelet adhesion, and leukocyte infiltration into the

islets. Tissue factor (TF) expressed by the islets is considered a major trigger for IBMIR

31 leading to platelet activation and release of downstream inflammatory mediators such as

interleukin (IL)-8, monocyte chemotactic protein (MCP)-1, and macrophage migration

inhibitory factor (MIF)52, 170. We recently demonstrated the occurrence of IBMIR in clinical autologous islet transplantation53. IBMIR under autologous conditions was

characterized by strong coagulation and a rise in proinflammatory cytokines with a

minimal impact on complement activation. Analysis of blood samples drawn during

intraportal islet infusion and in the hours immediately afterwards showed significant islet

damage occurring within 3 hours of the beginning of infusion53.

Nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB) has long been known to be the major transcription factor for the regulation of various proinflammatory cytokines. IL-1β and tumor necrosis factor α (TNFα) are two major inflammatory cytokines that are released rampantly during IBMIR. These cytokines are regulated by NFκB and can induce transcription of many other downstream products, such as inducible nitric oxide synthases, prostaglandin E2, and interferon gamma- inducible protein 10 (IP-10). In a syngeneic islet transplant model, culturing islets with the NFκB inhibitor dehydroxymethylepoxyquinomicin resulted in improved islet engraftment171. We have also shown that treatment of islets with withaferin A (WA),

which is an NFκB inhibitor, resulted in protection of islets from proinflammatory

cytokine–induced damage and improved survival of syngeneic mouse islets transplanted

intraportally126. Previous reports have also shown that TF expression is also regulated by

NFκB signaling172.

WA is a steroidal isolated from the plant Withania somnifera that has

been reported to have antiinflammatory, antiangiogenic, and anticancer effects157. WA

32 has been used in Southeast Asia and the Middle East for a long time to cure various inflammatory disorders. A recent mechanistic investigation has shown that WA hyperphosphorylates IκB kinase beta and prevents phosphorylation of inhibitor of κB

(IκB) and thereby inhibits NFκB activation161. In the present study, we also employed

CAY10512 (CAY; a potent analog of resveratrol), another known NFκB inhibitor, for further confirmation173.

We used a miniature tube model174 comprising a mixture of pure human pancreatic islets mixed with autologous blood to evaluate the effects of NFκB inhibitors.

We hypothesized that specific inhibition of NFκB can alleviate the inflammatory component of IBMIR and improve the survival of transplanted islets during the crucial peritransplant phase.

Materials and Methods

Human Islet Isolation and Blood Collection

Research-grade human pancreases from brain-dead donors were provided from two local organ procurement organizations (Southwest Transplant Alliance, Dallas, TX, and LifeGift, Houston, TX). Pancreases were procured and preserved with the standard two-layer method. Islets were isolated using the modified Ricordi’s method175, 176. Blood samples from the donors were collected during pancreas procurement in heparinized tubes (BD Biosciences, Franklin Lakes, NJ) to prevent coagulation. Blood samples were maintained in a refrigerator until the start of the experiment (not more than 36 hours).

33

Miniaturized In Vitro Tube Model

Islets were cultured in CMRL 1066 islet culture medium (Mediatech, Manassas,

VA) overnight at 37°C with 5% CO2 to recover from the isolation stress. Prior to the

experiment, islets were washed and counted, 2000 islet equivalents (IEQ) were treated with CMRL medium containing 1.0 ug/mL WA (Enzo Life Sciences, Farmingdale, NY),

0.15 µM CAY (Cayman Chemicals, Ann Arbor, MI), or no drug for 1 hour, and blood samples were allowed to equilibrate at 37°C. The blood was aliquoted in heparinized

Eppendorf tubes (Cardinal Health, Dublin, OH) (0.5 mL in each tube), the islets (~200

IEQs/200uL conditioned media) were added to the blood, and the tubes with blood and

islets were placed in a 37°C shaker incubator. Tubes were removed from the incubator at

their respective time points.

Luminex Multiplex Assay, ELISA, and Viability

The procedures on Luminex multiplex assay and the C-peptide and proinsulin

enzyme-linked immunosorbent assay (ELISA) were performed on serum samples as per

the manufacturer’s protocol. Islets recovered after 6 hours of culture with blood were

washed several times with phosphate-buffered saline to remove red blood cells. Cells were then stained for 10 minutes with Hoechst 33342 and propidium iodide. ImageJ software was used to semiquantitatively determine the percentage viability of islets.

Neutrophil Infiltration in Purified Human Islets Mixed with Autologous Blood

Formalin-fixed paraffin-embedded islet clots were sectioned in 5-micron sections, and polymorphonuclear neutrophil staining was performed using Naphthol AS-D chloroacetate esterase (Sigma Aldrich, St. Louis, MO). At least 15 separate islets were

34

evaluated for neutrophil infiltration. Neutrophil infiltration was classified by counting

polymorphonuclear neutrophils in and around individual islets. Insulin staining was

performed on serial sections using rabbit anti-insulin (Cell Signal, Boston, MA) and anti-

rabbit HRP secondary antibody (Cell Signal, Boston, MA). Color was developed using a

DAB chromogen substrate kit (DAKO, Carpinteria, CA).

Fluorescence Staining for Tissue Factor

Islet tissue sections (5 μm) were obtained after fixing in 10% formalin and

embedding in paraffin. Tissue sections were deparaffinized, and heat-mediated antigen

retrieval was performed. TF was stained with mouse anti-TF antibody (Abcam,

Cambridge, England) following anti-mouse Alexa Flour 594 (Invitrogen, Grand Island,

NY), along with insulin staining with guinea pig anti-insulin antibody (Sigma, St. Louis,

MO) following anti-guinea pig Alexa Flour 488 (Invitrogen, Grand Island, NY) and 4',6- diamidino-2-phenylindole (Sigma-Aldrich, St. Louis, MO).

Statistical Analysis

Data were expressed as mean ± standard error (SE). Data were analyzed using

GraphPad Prism version 6.03 (GraphPad Software, Inc., San Diego, CA). The two-way repeated-measure (RM) analysis of variance (ANOVA) followed by Tukey's or Dunnett post hoc test was performed to evaluate statistical significance between time points or groups. Statistical significance was considered for P values <0.05.

35

Results

Islet Viability after Culture with Autologous Blood

Islets treated with and without WA or CAY were cultured with autologous blood

for 6 hours. In the control group, the viability of islets as analyzed by propidium iodide

and Hoechst 33342 staining was significantly reduced at 6 hours after culture initiation

compared to 1 hour after culture initiation (P < 0.01), but no significant differences in

viability between 1 and 6 hours were seen in the WA and CAY groups (Figure 2.1). In

addition, there were significant differences in 6-hour viability between the control, WA,

and CAY groups (56.4 ± 10.7, 83.8 ± 2.6, and 84.7 ± 4.1 % [average ± SE], respectively;

P < 0.01).

Figure 2.1. Viability of islets after culture in autologous blood. The viability was significantly decreased at 6 hours compared with 1 hour after the culture in the control group (**P < 0.01) but not in either NFκB inhibitor–treated group. When comparing viability at 6 hours, significant differences were found between the control and the withaferin A (WA) and CAY10512 (CAY) groups (**P < 0.01). Data express average ± SE based on 6 independent experiments. P values were calculated by two-way ANOVA with Tukey’s multiple comparison test.

36

Human Islet Damage and the Impact of NFκB Inhibitors

We investigated rapid C-peptide and proinsulin release from islets in the tube model to detect early islet damage. Only the control group showed a significant elevation of C-peptide level in the plasma at 3 and 6 hours compared to baseline (P < 0.05) as well as compared to the WA and CAY groups (P < 0.05, Figure 2.2a). Similarly, only the control group showed a significant increase of the proinsulin level at 1 and 3 hours (P <

0.01) compared to the baseline, although no significant differences between the

Figure 2.2. Autologous islet damage and the impact of NFκB inhibitors. The plasma levels of C-peptide (a), proinsulin (b), and thrombin-antithrombin (TAT) (c) in an autologous islet tube model are shown. Solid line (black circle), solid line (open circle), and dotted lines show the control, withaferin A (WA), and CAY10512 (CAY) groups, respectively. Symbols indicate the P value compared to baseline within each group of control*, WA‡ or CAY# and the significance levels: *P<0.05 and **<0.01. Also, the symbols indicate the P value when comparing the control and WA§ or CAY† groups. Data express average ± SE based on 3 independent experiments. Repeated-measurement two-way ANOVA with Dunnett’s multiple comparison test was performed for statistical evaluation.

37

control and NFκB inhibitor groups were found (Figure 2.2b). TAT levels were

significantly elevated, with a peak at 6 hours in all groups (P < 0.01 between control and

CAY groups; P < 0.05 between control and WA group; Figure 2.2c); there was no significant difference in TAT levels between the three groups. These results were expected because the NFκB signaling pathway is not involved in the formation of the

TAT complex.

Proinflammatory Cytokine Responses

Levels of proinflammatory cytokines in the plasma samples were determined using Millipore multiplex bead assay (Figure 2.3). In the control group, there was a significant elevation in the proinflammatory cytokines TNFα, MCP-1, IL-8, and IL-6 (P

<0.01, 0.01, 0.05, and 0.05, respectively), but not in IP-10, when compared to baseline

(Figure 2.3). Both WA and CAY blocked the significant elevation of cytokine levels up

to 6 hours, except for IL-6 in the WA group.

Neutrophil Infiltration in Autologous Islets in Tube Model

The clots of blood and islets at 3 hours were stained with Naphthol AS-D

chloroacetate esterase to evaluate neutrophil infiltration (Figure 2.4). The number of

infiltrated neutrophils was higher in the islets in the control group (Figure 2.4a). In

contrast, the islet clots obtained from the WA- and CAY-treated groups showed very few to no neutrophils (Figure 2.4b and c). The number of neutrophils in an islet was calculated from 20 different fields. The average of the number of infiltrated neutrophils was significantly lower in both the WA (P <0.0001) and CAY (P < 0.001) groups compared to the control (Figure 2.4d).

38

Figure 2.3. Inhibition of proinflammatory cytokine secretion by NFκB inhibitors. The plasma levels of TNFα (a), MCP-1 (b), IP-10 (c), IL-8 (d), and IL-6 (e) are shown. Solid line (black circle), solid line (open circle), and dotted lines show control, withaferin A (WA), and CAY10512 (CAY) groups, respectively. Symbols indicate the P value compared to baseline within each group of control* or WA‡ and the significance levels: *P<0.05 and **<0.01. Data expressed as average ± SE based on 3 independent experiments. Repeated-measurement two-way ANOVA with Dunnett’s multiple comparison test was performed for statistical evaluation.

39

Figure 2.4. Neutrophil infiltration in islet clots. Neutrophils were stained by Naphthol AS-D chloroacetate esterase reactions (purple), counterstaining with hematoxylin (blue) and insulin (brown). Staining was performed on islet clots obtained after 3 hours of exposure to autologous blood. Representative views of hematoxylin and eosin staining in control (a), insulin staining (b), and neutrophil staining (c) are shown. Neutrophil staining is also shown in control (d), withaferin A–treated (e), and CAY10512 (CAY)–treated (f) groups. Scale bar indicates 50 μm. The number of neutrophils within an islet was counted (g). Data express average ± SE based on 20 counts from each group. **P < 0.01 and ***P < 0.0001 evaluated with ANOVA followed by Tukey’s test.

Tissue Factor Expression in Islets

Islets can express TF, which could potentially trigger coagulation and induce inflammation52. We evaluated TF expression with fluorescence staining in conjunction with insulin using an autologous islet blood sample at 3 hours (Figure 2.5). Strong TF expression was observed in the control group, but not in the WA or CAY groups.

Staining for TF was more intense on insulin-positive cells.

40

Figure 2.5. Tissue factor (TF) expression in islets after exposure to autologous blood. The expression of TF was evaluated with fluorescence stain (red) where nuclei and insulin were stained as blue and green. The autologous islet-blood samples were taken at 3 hours after the culture initiation. The white bar indicates 50 μm. Representative sections are shown. WA indicates withaferin A; CAY, CAY10512.

Discussion

IBMIR is a critical problem in both allogeneic and autologous islet transplantation

that needs to be addressed to improve the outcome of islet transplantation. Nilsson’s

group was the first to describe the characteristics of IBMIR and to extensively investigate

different interventions to block IBMIR53. It has been well established that NFκB is a

major transcription factor involved in the modulation of the inflammatory response177. In

this study, we implemented NFκB blockade as a strategy to reduce the activation of

IBMIR using WA and CAY, which are NFκB inhibitors157, 173. A miniaturized in vitro

41

tube model was employed to investigate the effect of these two drugs on IBMIR174. Our data showed that NFκB inhibitors could be potent agents for pretreatment of islets to ameliorate IBMIR, supported by the significant reduction in proinflammatory cytokine levels in the mixture with autologous blood.

NFκB is one of the key transcription factors activated in response to stress177.

Activation of NFκB, depending on the type of stimuli, could be either protective or

harmful to cells. A gene array study showed that pancreatic islet isolation and the

following culture of islets led to an increase in the mRNA expression of proinflammatory

cytokines in islets178. This increased expression of proinflammatory cytokines might in

turn lead to stress and activation of NFκB in islet cells. Thus, we hypothesized that once

islets are mixed with autologous blood, the innate immune system could activate, and this

could result in secretion of chemokines by the islets, which recruits monocytes and

neutrophils.

After 6-hour culture with autologous blood, viability was significantly preserved

in islets treated with WA and CAY when compared with control (Figure 2.1), suggesting

that WA not only protects islets from IBMIR but also maintains islet viability by other

mechanisms, including prevention of apoptosis. A recent study from our laboratory has

shown that islets cultured in the presence of WA have much better viability compared to

control islets after 48 hours126. The study also showed that inflammatory cytokine–

mediated apoptosis was also reduced in the presence of WA.

Both clinical and in vitro studies have shown that, immediately after islet infusion

or mixing in autologous blood, C-peptide and proinsulin levels rise rapidly, and this

increase could be due to islet damage174. Using our in vitro model, we have also shown

42

that mixing of islets in the blood led to a rapid increase in C-peptide and proinsulin

levels, suggesting islet damage, but when islets were precultured with WA or CAY, the

C-peptide and proinsulin levels were significantly lower, suggesting less islet damage

(Figures 2.2a and 2.2b). It is crucial to prevent the initial damage that occurs to the islets,

because it could lead to a sequence of immunological events eventually affecting the

entire graft. A recent clinical study using positron emission tomography showed that 25%

of islets are lost immediately after transplantation55. Therefore, preventing islet damage

in the initial stages may be the key to successful long-term insulin independence.

With IBMIR, the levels of TAT in plasma increase immediately after the islets

come in contact with blood, indicating the activation of coagulation170. Consistent with

other reports, our data have also shown an immediate increase in TAT levels when islets

were mixed with autologous blood. No influence of WA or CAY on TAT levels was

noticed (Figure 2.2c); however, this was an expected result because the process of

coagulation is not NFκB dependent. On the other hand, it could be argued that

coagulation is triggered by the TF expressed and released by the islets. TF expression is

known to be partly regulated by NFκB172. Islets express high levels of TF, and this interacts with factor VIIa and activates the extrinsic coagulation cascade. Therefore, blocking the activation of NFκB represents a sound strategy to inhibit IBMIR. The TF gene is also regulated by several other transcription factors such as Egr, AP-1, and

NFAT179. Various signals lead to activation of different transcription factors that result in

the induction of the TF gene, and synergistic regulation by several transcription factors

would be required for TF expression. The regulation of the TF gene by NFκB is induced

mainly by cytokine stimulation such as IL-1β, TNFα, and by pathogen-associated

43

molecular patterns such as lipopolysaccharide180. In the case of IBMIR during islet

transplantation, the major factor that could induce TF expression in the islets would be

the proinflammatory cytokines. Previous studies on human monocytes have shown that

preventing IκBα degradation resulted in attenuation of TF gene expression181, and use of

salicylates such as aspirin has similar effects179. Therefore, blocking TF expression by

NFκB would be protective for the islets during early stages of transplantation. We have

shown that both WA and CAY significantly prevent TF expression in islets when

compared with the control. Since we cultured islets in WA or CAY only for an hour

before mixing with autologous blood, a prolonged treatment of NFκB might further

reduce TF expression in islets and prevent the activation of coagulation.

Our recent clinical study showed an increase in the levels of proinflammatory

cytokines and chemokines immediately after infusion of islets into the portal vein174. The

presence of high levels of these cytokines amplifies the inflammatory response, making

the islets more susceptible to cytokine-mediated apoptosis. In our in vitro study, we have demonstrated that the plasma levels of several proinflammatory cytokines were increased after mixing of islets with blood. TNFα is a proinflammatory cytokine known to induce apoptosis in β cells182. We found that TNFα levels increased gradually and reached a maximum at 3 and 6 hours. In both the WA and CAY groups, the level of TNFα did not increase and remained low at all time points (Figure 2.3a). IL-6, which can be either

proinflammatory or antiinflammatory depending on the stimuli, was also found to

increase in control samples, whereas the WA- and CAY-treated samples efficiently

suppressed the secretion of this cytokine (Figure 2.3e). IP-10 levels in the plasma were high to start with and remained high, but in the WA and CAY groups, the level of IP-10

44

was lower compared with the control (Figure 2.3c). IP-10 is considered one of the major

proinflammatory cytokines that can modulate islet graft survival84. MCP-1 is responsible

for attracting monocytes to the site of injury183. When islets are mixed with blood, MCP-

1 levels in the plasma begin to rise and remain high up to 6 hours. When WA- or CAY-

treated islets were mixed with blood, the MCP-1 level did not change (Figure 2.3b),

suggesting prevention of islet damage by macrophages. IL-8 is also a cytokine that

recruits polymorphonuclear cells to the site of inflammation. The level of IL-8 also

increased rapidly after mixing blood and islets, and this elevation was prevented in the

WA and CAY groups (Figure 2.3d). Other key proinflammatory cytokines, IL-1β and

interferon γ, were undetected in the samples.

Our in vitro tube model was validated for the study of islets mixed with blood

174 based on a comprehensive analysis of pH, pO2 and glucose levels . Since our present

study using the in vitro tube model showed that both WA and CAY can inhibit the

secretion of proinflammatory cytokines/chemokines, it is appropriate to show the

resulting outcome. Islet clots were sectioned and stained for neutrophils. The islets that

were mixed with blood showed massive infiltration of neutrophils even at 3 hours,

whereas the WA- and CAY-treated islets showed a very significant reduction in the

number of neutrophils infiltrated per islet (Figure 2.4). Thus, the histology results

correlate with proinflammatory cytokine or chemokine levels, especially with IL-8,

which is required for the maturation and infiltration of neutrophils.

The present study focused on the instant response occurring when purified islets

come into contact with blood. Long-term benefits of treatment with NFκB blockers on

islet graft outcome remain to be determined. IBMIR against allogenic and xenogenic

45 islets may be stronger than against autologous islets due to the involvement of humoral

immune components such as autoantibodies, alloantibodies, and xenoreactive antibodies.

Future studies using this miniature tube model could include analyze the effects of

inhibitors for various components of IBMIR. In summary, we have demonstrated that a

miniaturized in vitro tube model is an effective tool to study IBMIR. We have also shown that blocking NFκB is a promising strategy to alleviate the harmful effects of IBMIR on transplanted islets.

46

CHAPTER THREE

Evaluation of MicroRNA375 as a Novel Biomarker for Graft Damage in Clinical Islet Transplantation

This chapter published as: Kanak MA, Takita M, Shahbazov R, Lawrence MC, Chung WY, Dennison AR, et al. Evaluation of microRNA375 as a novel biomarker for graft damage in clinical islet transplantation. Transplantation. 2015 Mar 12 [Epub ahead of print].

Introduction

Transplantation of allogeneic islets from deceased donors into patients with type 1 diabetes has shown promising outcomes by restoring normoglycemia and eliminating the risk of hypoglycemic episodes139. In addition, for advanced cases of chronic pancreatitis and some benign pancreatic tumors, total pancreatectomy with islet autotransplantation

(TPIAT) offers relief from intractable pain and retention of endocrine function184, 185. In both forms of islet transplantation, a significant mass of islets are lost immediately after infusion into the portal vein of the liver due to an innate immune response called instant blood-mediated inflammatory reaction (IBMIR)51, 53, 56. The process of IBMIR encompasses coagulation, proinflammatory cytokine/chemokine production, complement activation, and infiltration of leukocytes56. In the case of type 1 diabetes, islets engrafted in the liver eventually undergo chronic rejection by the immune system186.

After transplantation, it has been challenging to investigate islet graft survival or chronic rejection in a patient. In clinical practice, islet graft function has been monitored by metabolic function via blood glucose, insulin, and C-peptide levels, as well as through imaging187. However, these tests are influenced by physiological changes, including islet

47 stimulation. It is necessary to identify a biomarker that can specifically detect islet

damage independent of metabolic stimuli.

One option for a biomarker is micro RNAs (miRNAs), a class of small noncoding

RNA (18–24 bp) known to regulate gene expression posttranscriptionally. According to

the miRBase registry (www.mirbase.org), about 2578 mature miRNA sequences have

been identified in the human genome188. Recently, miRNAs present in the circulation

have been found to serve as biomarkers for various and other diseases189, 190. An

islet-specific miR375 reported by Poy et al. is known to regulate insulin secretion by

regulating the protein myotrophin involved in intracellular trafficking191. A more

elaborate study using miR375 knockout mice demonstrated its importance in the

development of β cells192. At the same time, gene array analysis of miRNAs in β cells

showed no changes in the level of miR375 at different glucose concentrations193.

Recently, it was demonstrated that islet-specific miR375 is released in the serum of streptozotocin-injected mice and also in prediabetic NOD mice, both of which are models for β cell destruction; these results suggest that miR375 is released as a result of pancreatic β-cell death194.

Hence, we investigated whether miR375 could be a potential biomarker for islet

damage in clinical islet transplantation. We first used a miniaturized in vitro tube model

to determine if miR375 was released by damaged islets in response to autologous blood

and then measured miR375 using reverse-transcription polymerase chain reaction (RT-

PCR) in serum samples of clinical autologous and allogeneic islet transplant patients.

48 Materials and Methods

Patients

Levels of miR375 were tested in 29 patients: 15 autologous islet recipients (i.e., patients with chronic pancreatitis who underwent TPIAT), 8 allogeneic recipients with type 1 diabetes, and 6 control patients with total pancreatectomy alone; all patients were from Baylor University Medical Center (Dallas, TX, USA) or the University of Leicester

(Leicester, UK). This study was approved by the institutional review board, and written informed consent was obtained from all patients.

For both autologous and allogeneic transplants, pancreases were preserved and processed for islet isolation as described earlier176, 195. The final preparation of islets was assessed for count, purity, viability, and gram stain prior to infusion without any culture.

Isolated islets were infused into the portal vein via the mesenteric vein with heparin

(70 U/kg recipient body weight) over 15–60 min while the patients were under general anesthesia. Beginning 6 h after surgery, patients were continuously infused with 300 U/h heparin for 24 h. Subsequently patients were converted to enoxaparin (Lovenox®;

Sanofi-Aventis, Bridgewater, NJ) at 80 mg/day for 2 weeks. Among the 15 patients who underwent TPIAT, 3 received no antiinflammatory regimen, 6 received etanercept (TNFα blocker) alone, and 6 received a combination of etanercept and anakinra (IL-1β blocker) during islet infusion as described earlier145, 196.

Human research-grade pancreases were used for isolating islets using methods described previously195. Human islets were cultured in Prodo standard islet culture media

(Prodo Labs, Irvine, CA) supplemented with 5% human serum AB and 2% glutamine

(37°C, 5% CO2 up to 96 h).

49

Serum Collection and Storage

Blood samples were obtained for serum and plasma at admission, prior to islet

infusion, during islet infusion, at the completion of islet infusion, and 1 h, 3 h, 6 h, 24 h, 3

days, 5 days, and 7 days after islet infusion. After processing, sera or plasma were

aliquoted in cryogenic tubes and stored at –80°C until further analysis53. Blood drawn

from healthy individuals was used as a control to normalize the patients’ miR375 levels.

Miniaturized In Vitro Tube Model

A recently described in vitro model was used54. Briefly, islets were cultured

overnight to recover from the isolation stress. Before the experiment, islets were washed;

counted and 4000 IEQ were used. Blood samples were allowed to equilibrate at 37°C.

The blood was aliquoted in Eppendorf tubes (Cardinal Health, Dublin, OH) (0.5 mL in

each tube), the islets (~200 IEQ/100 µL media) were added to the blood, and the tubes

with blood and islets were placed in a 37°C shaker incubator. Tubes were removed from

the incubator at their respective time points and centrifuged at 1200 rpm for 5 min to

collect plasma. Another set of tubes where islets were treated with WA was used and

experiments were performed as previously described.

MiRNA Extraction, cDNA Synthesis, and Reverse-Transcription PCR

RNA extraction from 200 μL of samples of serum and/or plasma was performed

using the miRCURY RNA isolation kit for biofluids (Exiqon, Denmark) following the

manufacturer’s protocol. Extracted RNA was treated with rDNase to remove any DNA contamination. Complementary DNA strand synthesis was performed using the miRCURY Universal cDNA synthesis kit (Exiqon, Denmark) following the

50

manufacturer’s protocol. Complementary DNA was diluted 1:40 using nuclease-free

water, and RT-PCR was performed using SYBR green qPCR master mix (Exiqon,

Denmark) and locked nucleic acid–based miRNA primers (Exiqon, Denmark) following

the manufacturer’s protocol. The standard curve was prepared to quantify miRNA using

synthetic miRNA.

Statistical Analysis

All statistical analysis was performed with IBM SPSS Statistics 22 (IBM

Corporation, Armonk, NY) or Prism 6 for Windows (GraphPad Software, Inc., La Jolla,

CA). miR375 expression levels in the in vitro assay and patient serum samples were

normalized with miR16-1-3p and miR423-3p, respectively, using the 2ΔΔCt method197.

The statistical significance of circulating miR375 and serum C-peptide levels for the timeline were evaluated with RM two-way analysis of variance (ANOVA) with

Bonferroni correction followed by the post hoc Games-Howell test, which is designed for unequal variance. Three- or two-group comparison without the timeline was performed with ANOVA with Tukey’s post hoc test or Student t test. Categorical variables were assessed with Pearson chi-square test or Fisher’s exact test. Values are shown as average

± S.E.; P <0.05 was considered statistically significant.

Results

Expression of MiR375 in Human Islets and the Release by Autologous IBMIR

To determine the dose dependency of miR375 in islets, purified human islets were serially diluted. The amount of miR375 in cell lysate exhibited a statistically significant linear regression with islet mass (R2 = 0.94, P < 0.0001) (Figure 3.1a).

51

Using our previously established miniaturized in vitro tube model for investigating IBMIR53, 111. We confirmed the significant elevation of miR375 when islets were mixed with autologous blood, compared with an islet-alone or blood-alone group (P

<0.01 or <0.001, Figure 3.1b). Levels of miR375 peaked immediately after mixing blood with islets and remained high until completion of the experiment (6 hours). Similarly, we found a significant increase in C-peptide levels immediately after mixing islets with blood (Figure 3.1c). The islets alone group showed a short peak of C-peptide secretion at

15 minutes, while no elevation was observed in miR375 level, suggesting that C-peptide can be influenced by mechanical stress, unlike miR375. Of note, 4.46 fmol of miR375 was observed at 6-hour exposure of islets to autologous blood, which corresponds to

39.4% (78.8 out of 200 IEQ) islet damage.

To determine if WA protected islets from IBMIR-mediated damage, serum samples from previous in vitro miniaturized tube experiment were used. Human islets were pretreated with WA before mixing with autologous blood, and serum samples were collected at various time points. The miRNA 375 level in the serum of these samples revealed elevation in the nontreated group that continued to increase with time. WA treatment showed significantly reduced miRNA 375 levels at 1-hour, 3-hour, and 6-hour time points (Figure 3.2a). These data also correlated with the proinsulin and C-peptide levels of the samples (Figure 3.2b, 3.2c). Again, these results suggest that WA protects islets from IBMIR-mediated damage.

52

Figure 3.1. miR375 expression is dependent on islet mass and increases similarly to C-peptide in response to islet damage in a miniaturized in vitro tube model. miR375 amount had an excellent linear regression with islet mass (a) (equation: y = 0.05658x + 0.004165, R2 = 0.94, P < 0.0001). Solid and dotted lines show a regression line and the 95% confidence interval, respectively. Both miR375 expression (b) and C-peptide secretion (c) were higher with the blood and autologous islet combination compared with blood alone or islets alone. ** P with RM two-way ANOVA followed by post hoc Games-Howell test <0.01 and *** P <0.001. All data on miR375 were normalized with miR16-1-3. Results in parts b and c show average ± S.E. Data shown are from three independent experiments.

Circulating MiR375 in Clinical Allogeneic and Autologous Islet Transplantation

When comparing miR375 levels in the 15 autologous islet recipients, 8 allogeneic

recipients, and 6 control patients with total pancreatectomy alone, we found no

significant differences in patient characteristics, except for pretransplant hemoglobin A1c

levels, although transplanted islet yield and dose were significantly higher in patients

with allogeneic transplant than with autologous transplant (P = 0.008 and <0.001, respectively; Table 3.1).

Circulating miR375 levels were significantly elevated following autologous and allogeneic transplants compared with the control group (Figure 3.3a, P = 0.005 and

<0.001 with RM two-way ANOVA). In a post hoc comparison test, significant difference was seen between autologous and allogeneic patients at 3 hours after islet infusion (P <

0.05). The miR375 levels in autologous recipients peaked 1 hour after islet infusion, corresponding to an average of 1.31 × 104 fmol of miR375 released per patient. This 53 indicated that an average of 231,531 out of 441,731 IEQ transplanted (52.4%) were damaged after TPAIT, as calculated from 5.66 × 10-2 fmol/IEQ, shown in Figure 3.1a.

The autologous and allogeneic transplant groups also showed significant elevation in serum C-peptide level when compared with the control (Figure 3.3b, P = 0.003 and

0.047, respectively). Of note, no correlations with statistical significance were found between miR375 relative expression and transplanted islet mass in either autologous or allogeneic transplants (Figure 3.5).

Patients undergoing autologous islet transplants were further divided into three groups based on administration of 1) no antiinflammatory drug, 2) etanercept alone, or 3) a combination of etanercept and anakinra. No significant differences in patient and transplant characteristics were seen between the three groups (Table 3.1).

Table 3.1. Patient and transplant characteristics in autologous recipients for sub-analysis.

Control Single Double group blockage blockage P (n = 3) (n = 6) (n = 6) value a Patient characteristics Age 46.7 ± 5.2 39.0 ± 2.1 35.5 ± 4.9 0.27 Sex: female, n (%) 2 (66.7%) 4 (66.7%) 4 (66.7%) 1.0 Body weight (kg) 64.4 ± 6.0 77.9 ± 9.3 70.4 ± 3.4 0.49 Body mass index (kg/m2) 23.1 ± 1.6 27.4 ± 3.1 24.4 ± 1.2 0.47 Hemoglobin A1c (%) 5.4 ± 0.3 5.7 ± 0.2 5.4 ± 0.2 0.54 Transplantation Islet yield (×103 IEQ) 436 ± 107 392 ± 45 495 ± 86 0.59 Islet dose (×103 IEQ/kg of body 7.0 ± 1.9 5.1 ± 0.9 6.1 ± 0.8 0.49 weight) Tissue volume (mL) 26.0 ± 2.5 8.6 ± 1.2 9.7 ± 2.4 <0.001 Purity (%) 53.3 ± 6.9 58.3 ± 8.7 46.7 ± 15.0 0.76 Viability (%) 97.0 ± 1.2 96.3 ± 0.5 96.6 ± 0.5 0.81

54

a

b

c

Figure 3.2. Islet damage by IBMIR inhibited by WA (a) miRNA 375, (b) C-peptide and (c) proinsulin. All data on miR375 were normalized with miR 423-3p. Results in parts b and c show average ± S.E. Data shown are from three independent experiments. *P<0.05, **P<0.01, ***P<0.001, ****P<0.0001.

55

Figure 3.3. miRNA levels in vivo indicate immediate islet damage after infusion. Levels of circulating miR375 (a) and serum C-peptide (b) before and after autologous transplantation (n = 15, solid line with white circle), allogeneic transplantation (n = 8, bold dotted line with black circle), or total pancreatectomy alone (n = 6, dotted line with triangle) are shown. The trend of circulating miR375 levels in patients with TPIAT and TP alone (dotted line with triangle, n = 6) is shown in which the autologous islet recipients were divided into three groups: no blockage (solid line with white circle, n = 3), single blockage of TNFα (dotted line with block circle, n = 6) and double blockage of both TNFα and IL-1β (gray bold line with triangle, n = 6) (c and d). X axis indicates time post-islet infusion or surgery. Y axes in C and D are log scale. Data points show average ± S.E.. *P with RM two-way ANOVA < 0.05, **<0.01 and ***<0.001. a In post hoc analysis, a significant difference was observed between autologous and allogeneic recipients at 3 hours after islet infusion.

56

50

40

30

20 C t valu e

10

0 0 2 4 6 8 10 Log of Concentration

Figure 3.4. Standard curve plot for the quantification of miRNA375. Tenfold serial dilutions of synthetic miR375 were used to prepare standard curve. X and Y axes indicate log of miR375 (fmol/L) and threshold cycle (Ct) value corrected with UniSp6 Spike-in miRNA, respectively. The liner regression (y = -3.673x + 42.19) accounts for an excellent R square of 0.98.

a b

2,500 2,500

2,000 2,000

1,500 1,500

1,000 1,000

500 500 miR375 relative expression miR375 relative miR375 relative expression miR375 relative

0 0

200 400 600 800 200 400 600 800 1,000 1,000 3 3 IEQ transplanted (× 10 ) IEQ transplanted (× 10 )

Figure 3.5. miR375 levels posttransplant and transplanted islet mass dot plots show a relationship between miR375 relative expression levels at 1 hour post-islet infusion and transplanted islet mass (IEQ) in autologous (a) and allogeneic (b) islet transplantation. No correlations with statistical significance were found in either type of transplant.

57 Effect of Antiinflammatory Treatment in Circulating MiR375 in Autologous Islet Transplantation

The levels of miR375 in all three groups were significantly elevated immediately

after islet infusion up to 6 h (P = 0.04 with RM two-way ANOVA for time; Figure 3.3c),

although no significant difference was seen between the three groups. After 24 h,

significant differences were observed between the three groups: miR375 levels in the no- blockage control group remained significantly higher for 7 days postinfusion compared with either the single- or double-blockage groups (P = 0.002 and P = 0.001, respectively,

Figure 3.3d). The second peak at 24 h in the no-blockage control group could be due to islet damage induced by proinflammatory cytokines released after transplantation.

Interestingly, both the single- as well as double-blockage group showed significantly reduced levels in this second miR375 peak, suggesting a decrease in islet damage due to inflammation. The second peak of miR375 level in the double-blockage group was smaller than the peak in the single-blockage group, suggesting better protection of islets from dual blockage of TNFα and IL-1β, but statistical significance was not achieved. No significant difference in the insulin independence rate after surgery was seen between the three groups, although one out of three patients achieved insulin independence in the control group, compared with 4 of 6 in the single-blockage group and 5 of 6 in the

double-blockage group.

Discussion

In this study, we have analyzed miR375 as a biomarker to quantify human islets

and detect damage. We used our previously validated miniaturized in vitro tube model for

the study of IBMIR53, 111 and demonstrated a rapid increase in the miR375 levels in the

58

plasma immediately after mixing islets with blood, which was not seen in islet-only or

blood-only controls. The levels of C-peptide mirrored the levels of miR375. These data

indicate that this early damage is most likely related to the inflammatory response by the

innate immune system. Compared with patients undergoing total pancreatectomy only,

there was a significant increase (~1000-fold) in the serum miR375 level in patients

receiving autologous and allogenic islets, suggesting that miR375 is released from islets

undergoing damage due to inflammation.

The linear relationship between the amount of miR375 and the islet mass is a convincing result. It is estimated that 5.66 × 10-2 fmol of miR375 is present per human

IEQ. In an in vitro study with pure islets from a healthy donor, we found linear regression

that intersects at the origin of the graph (Figure 3.1a), suggesting that miR375 can be a

good biomarker to evaluate islet damage, and the slope of linear regression indicates the

amount of miR375 per islet.

Approximately 40% of autologous islets were damaged 6 hours after mixing with

autologous blood in an in vitro tube model. In our previous study, we showed that the

viability of islets was significantly decreased to 56.4% at 6 hours after mixture of

autologous blood and islet111. The amount of damaged islets, which was estimated by

miR375, might reflect the loss of viability. In a previous study, Eriksson et al. estimated

that approximately half of islets were lost within 1 hour after allogeneic islet

transplantation based on changes in radioactivity measured by positron emission

tomography combined with computed tomography (PET/CT)55. Similarly, our clinical

data showed that 52.4% of transplanted islets were damaged after TPIAT. Similarities in

the percentage of damaged islets in allogeneic and autologous transplant suggest that the

59 effects of IBMIR immediately after islet infusion might be equivalent regardless of the

type of transplant, although a validation study with a larger number of patients is

warranted for such a conclusion. The lack of correlation between miR375 levels at 1 hour

posttransplant and islet mass could be explained by differences in donor characteristics

and the degree of islet damage during isolation.

Of note, we found an inverse relationship in the elevation of circulating miR375

and serum C-peptide levels between autologous and allogeneic islet recipients; i.e.,

miR375 levels in autologous patients were higher than those in the allogeneic subjects,

while allogeneic patients showed a higher C-peptide response than autologous patients

(Figure 3.3). This result can be explained by the different biologic characteristics between the two biomarkers. Elevation of C-peptide levels represents both a physiologic response of pancreatic β cells to hyperglycemia and a mechanical release due to islet damage198,

199. In this study, the amount of transplanted islet mass was significantly higher in

allogeneic transplants than in autologous transplants, and thus, greater C-peptide

secretion or release was observed in allogeneic recipients than in autologous patients.

Circulating miR375 has been reported as a specific biomarker for β-cell death and is not

secreted in response to metabolic stimuli194. Autologous recipients undergo major surgery

and are on parenteral nutrition with no metabolic stimulation of transplanted islets. Also,

the autologous islets are isolated from inflamed pancreases and hence could be more

susceptible to immediate damage than allogenic islets184, and in turn, the levels of

circulating miR375 could be higher in autologous transplant than in allogeneic transplant.

Further investigations on biological conditions that modify circulating miR375 levels are

warranted.

60

We then tested the effect of antiinflammatory agents. More elaborate analysis of

miR375 peaks in autologous patients without any antiinflammatory agent demonstrated

an early peak at less than 1 h and a second peak at 24 h, which dropped gradually. On the

other hand, the patients who received antiinflammatory agents such as etanercept or a

combination of etanercept and anakinra had a smaller second peak, suggesting that the

islet damage observed after 24 h may be caused by proinflammatory cytokines. In a

recent preliminary study, Piemonti et al200 showed similar results for miR375 levels in

allogenic islet transplant recipients under three different immunosuppression regimens

and autologous islet recipients undergoing CXCR1/2 inhibitor therapy.

In addition to IBMIR, hypoxia in the liver could also cause early damage to

transplanted islets. A previous report from our group showed that the levels of high-

mobility group box 1 indicated islet damage during the peritransplant period199. The

damage to islets occurring due to inflammatory events after transplantation can be

reduced by using antiinflammatory agents, as we have shown here by the use of

etanercept and/or anakinra (Figure 3.3c).

Some of the major criteria to consider when evaluating a biomarker are

sensitivity, specificity, rapid detection, reproducibility, accuracy, and inexpensiveness.

New technologies such as locked nucleic acid primers and probes for miRNA could

significantly improve the sensitivity of the assay. One other characteristic of miR375 that

stands out is tissue specificity; miR375 is highly expressed only in the islets and

somewhat in neuronal cells, which makes it a very specific biomarker for islets191. Some

of the limitations of this study include (i) the small number of autologous and allogenic islet transplant patients analyzed; (ii) the lack of analysis of a broad repertoire of

61 miRNAs for the same objective; and (iii) analysis of a possible inverse correlation between miRNA levels and posttransplant islet graft function.

In summary, miR375 along with C-peptide and proinsulin in plasma/serum could be considered a stronger and more reliable marker for islet damage in clinical islet transplantation. Furthermore, measurement of miR375 should be performed to assess islet damage due to chronic rejection and may also be useful in detecting early islet damage during the development of type 1 diabetes.

62

CHAPTER FOUR

Delineating the Molecular Pathogenesis of Chronic Pancreatitis and Use of NFκB Inhibitor to Prevent its Progression

Introduction

Chronic pancreatitis (CP) is an inflammatory disease characterized by irreversible

structural alterations due to inflammation, fibrosis, and acinar atrophy. Patients with CP

experience debilitating abdominal pain, exocrine and endocrine pancreatic insufficiency,

and severely compromised quality of life201, 202. Factors known to cause CP are

categorized as toxic (e.g., alcohol, tobacco), genetic (e.g., CFTR mutation, SPINK1

mutation), autoimmune, recurrent and severe acute pancreatitis (AP), obstructive (e.g.,

pancreatic divisum, sphincter of Oddi dysfunction), and idiopathic203, 204. CP increases

the risk of pancreatic adenocarcinoma and type 3C diabetes mellitus and can sometimes

be lethal. CP is usually a result of repeated episodes of AP, which involves intra-acinar

trypsinogen activation, vacuole formation, necrosis, and apoptosis of acinar cells and

severe inflammation205. Currently there is no treatment for CP or a unified approach to

control the progression of the disease.

Several reports have identified the significant role played by nuclear factor kappa

B (NFκB) in the pathogenesis of CP206, but the downstream mechanisms in the

progression of the disease are still unclear. In transgenic mice, tamoxifen-induced

activation of NFκB and/or overexpression of active form of inhibitor of kappa B kinase 2

(IKK)-2 in the pancreatic acinar cell increased the severity of CP207, 208. A high-fat diet– induced CP showed increased NFκB activity to be required for the development of

63

disease209. A cerulein-induced experimental model of pancreatitis has been widely used

to investigate the mechanism of CP210, 211.

Endoplasmic reticulum (ER) stress or unfolded protein response (UPR) is a

cellular mechanism to overcome protein overload and regulate protein folding more

efficiently to regain homeostasis212. Sustained or uncontrolled ER stress leads to

apoptosis of the cell which is mainly regulated by transcription factor C/EBP homologous

protein (CHOP)213, 214. During the pathogenesis of pancreatitis, acinar cell death is prominent and this leads to overwhelming amount of protein production in the remaining acinar cells, which may result in ER stress. ER stress has been identified as an early event in pancreatic injury, and prolonged administration of cerulein to mice results in sustained activation of ER stress leading to CP215.

Cells of the innate immune system (neutrophils, macrophages, monocytes, and dendritic cells) use special receptors such as toll-like receptors (TLRs) and/or NOD-like receptors (NLRs) to sense damage or pathogen invasion. These cells express receptors that recognize ligands during tissue damage called damage-associated molecular patterns

(DAMPs) and during pathogen invasion called pathogen-associated molecular patterns.

NLRP3/NALP3 is an extensively investigated member of the NLR family that is responsible for the formation and activation of an inflammasome complex. NLRP3 oligomerizes and forms a complex with apoptosis associated Speck-like protein (ASC), the inflammasome complex, and then recruits pro-caspase 1 to the caspase recruitment domain (Pycard) present on ASC216. Active caspase-1 is released after cleavage of pro-

caspase-1 by the NLRP3 inflammasome. Subsequently, pro-IL-1β and pro-IL-18 are

processed by caspase-1 to yield active IL-1β and IL-18, which are then released in the

64

inflammatory microenvironment to amplify inflammation. NLRP3 inflammasome has

been reported to play a role in chronic inflammatory diseases such as cryopyrin

associated periodic syndromes, gout, atherosclerosis, and type 2 diabetes.

Withaferin A (WA) is a natural steroidal lactone that has been used in ancient

ayurvedic medicine as an anti-inflammatory drug. It has been shown to inhibit NFκB

activation by binding to I kappa kinase β (IKKβ) and preventing phosphorylation of

inhibitor of kappa B (IκB)217. In this study, we have investigated the role of ER stress and

inflammasome in a cerulein-induced CP model. Furthermore, we used WA to investigate

whether blocking NFκB would alleviate pancreatitis.

Materials and Methods

Mice

All mice were housed in filter-topped shoebox cages with autoclaved food and water. C57BL/6 mice purchased from Jackson Laboratories were used throughout the study and all experiments were conducted on age- and sex-matched littermates. Mice

were randomly assigned to control and experimental groups. All experiments were

performed in accordance to Baylor Research Institute’s animal care and use committee guidelines and regulations.

Reagents

Cerulein was purchased from Sigma Aldrich (St. Louis, MO), and WA from Enzo

Life Sciences (Farmingdale, NY). The amylase activity kit, Picrosirius red staining kit, and anti-neutrophil and anti-Ki-67 antibody were obtained from Abcam (Cambridge,

MA). Mouse CD45, CD11b, CD14, and Ly6g antibody were purchased from BioLegend

65

(San Diego, CA). Anti-NFκB antibody was bought from Santa-Cruz Biotechnology

(Dallas, TX). RNAlater and TRIzol were purchased from Life Technologies (Carlsbad,

CA). The high-capacity cDNA reverse transcription kit was obtained from Applied

Biosystems (Waltham, MA), and the qRT-PCR SYBR green/ROX master mix and custom-made human inflammatory gene array kit from Qiagen (Valencia, CA). IL-6 and

MCP-1 Luminex multiplex kit was obtained from Millipore (Darmstadt, Germany).

Acute Pancreatitis

Animals fasted overnight before induction of pancreatitis by cerulein; water was given ad libitum. Mice were injected with cerulein (50 μg/kg intraperitoneally) or saline every hour for a total of 7 injections and were sacrificed 1 hour after the last injection.

WA (1.25 mg/kg) was administered 1 hour before the first cerulein injection (n = 5 mice/group) (Figure 4.1a). Blood was collected from the inferior vena cava for analysis of serum markers of pancreatitis, and pancreas tissue was harvested immediately afterwards. The pancreas was divided for immunohistochemistry, RNA extraction, protein extraction, and immune cell infiltration analysis.

Chronic Pancreatitis

Animals were subjected to repeated episodes of AP by a supramaximal dose of cerulein. The mice fasted overnight before cerulein injection, with an ad libitum supply of water. In the preventive model, CP was induced when cerulein was injected intraperitoneally (50 μg/kg) every hour for a total of 7 injections. This injection was repeated once a week for 4 weeks. Control mice were injected with saline as a sham treatment. Another group of mice received WA (1.25 mg/kg) 1 hour before the first

66

cerulein injection every week (n = 6 mice/group) (Figure 4.1b). In the therapeutic model,

CP was induced when cerulein was injected intraperitoneally (50 μg/kg) every hour for a

total of 6 injections. This injection was repeated twice a week (Mondays and Thursdays)

for 8 weeks. WA (0.625 mg/kg) was injected 1 hour before the first cerulein injection, but

administration was initiated at week 5 and continued until the end of the experiment

(Figure 4.1c). In both models, mice were then sacrificed and tissue was harvested 1 day

after the last cerulein injection.

a. Acute Pancreatitis WA (1.25mg/kg) Weeks 1 Cerulein (50ug/kg) Once per week, 7 hourly shots i.p. b . Chronic Pancreatitis (Preventive) WA (1.25mg/kg) Weeks 1 2 3 4 Cerulein (50ug/kg) Once per week, 7 hourly shots i.p. c. Stringent Chronic Pancreatitis (Therapeutic) WA (0.625mg/kg) Weeks 1 2 3 4 5 6 7 8 Cerulein (50ug/kg) Once per week, 7 hourly shots i.p.

Figure 4.1. Protocol for development and treatment of pancreatitis: (a) acute pancreatitis, (b) chronic pancreatitis (preventive approach), (c) Stringent chronic pancreatitis (therapeutic approach).

Amylase Activity Assay

Serum samples obtained from mice after treatment were harvested and stored at –

80°C until further analysis. Serum was analyzed for amylase activity as per the

manufacturer’s protocol.

67 Histology and Immunohistochemistry

Pancreas tissue was placed in 10% formalin immediately after harvesting and

fixed for at least 48 hours at 4°C before processing. Tissue was embedded in paraffin,

and 5-um sections were made and mounted on slides for staining. Hematoxylin and eosin

staining was performed for histopathological evaluation, and a pathological observer

blinded to the study groups scored the tissue from 0 (normal) to 5 (severely inflamed).

Sirius red staining was performed according to the manufacturer’s protocol on the

sections to evaluate fibrotic change. The sections were deparaffinized in xylene and

rehydrated using ethanol. The slides were incubated in a citrate buffer of pH 6 at 97°C for

20 minutes. Following the antigen retrieval process, the slides were carefully washed 3

times for 5 minutes in Tris-buffered saline (TBS) containing 0.025% Triton X-100. The

slides were blocked in 1% bovine serum albumin (BSA) in TBS for 2 hours at room

temperature to block nonspecific binding of the antibodies. Then the slides were

incubated overnight with primary antibodies, which were diluted in TBS with 1% BSA.

The following day, the slides were gently rinsed 3 times for 5 minutes in TBS 0.025%

Triton X-100. Tetramethylrhodamine- or fluorescein isothiocyanate–labeled secondary

antibodies from Invitrogen were diluted in TBS with 1% BSA. The slides were

counterstained with 4',6-diamidino-2-phenylindole (DAPI) and mounted for microscopic analysis. ImageJ software was used to perform semiquantitative analysis of the staining.

Immune Cell Infiltration Assay

A cell suspension was prepared from the pancreas tissue after digestion with collagenase (2 mg/mL) at 37°C for 15 minutes. Tissue was further mechanically digested by pipetting several times. The suspension was then washed and filtered to obtain only

68

the single cell fraction. The cell suspension obtained was used for flow cytometric

analysis of infiltrated immune cells. 1 × 106 pancreatic cells were labeled with anti-

CD45, anti-CD11b, anti-CD14, and anti-Ly6g antibodies. Stained cells were detected

using FACScanto II, and data were analyzed using FlowJo software.

Luminex Assay

The mouse pancreas was homogenized, and the total lysate concentration was

measured. Lysate was analyzed directly according to the manufacturer’s protocol, and

results were normalized to total lysate concentration.

qRT-PCR

Pancreas tissue was stored in RNAlater at –80°C until the extraction procedure.

Total RNA from the pancreas tissue was extracted using TRIzol reagent. Tissue was

homogenized, and debris was removed by centrifugation. Chloroform was added to

separate the RNA from other biomolecules and was further precipitated using

isopropanol. The RNA pellet was washed with ethanol and quantified using NanoDrop

2000. cDNA synthesis was performed using a high-capacity cDNA reverse transcription

kit as per the manufacturer’s protocol. PCR primers for 18s rRNA, PERK, Atf6, Ern1,

Eif2a, Atf4, Xbp1, CHOP, HMGB1, Pycard, Nlrp3, IL-18, IL-1β, IL-6, TNFα, Nos2,

Casp3, Casp7, and Bax were purchased from Qiagen. qRT-PCR was performed using the

SYBR green/ROX master mix from Qiagen as per the manufacturer’s protocol.

Gene Array

Healthy human pancreas tissue was obtained from cadaveric donors. CP tissue was obtained from patients undergoing total pancreatectomy auto islet transplantation

69 who consented to provide samples for a research study. All protocols were performed according to regulations of Baylor Research Institute’s institutional review board.

Pancreas tissue was used for RNA extraction by TRIzol and cDNA synthesis, as previously described. A custom PCR array for inflammatory and NFκB regulated genes was obtained from Qiagen, with performance and analysis as per the manufacturer’s recommendations.

Statistical Analysis

Statistical analysis was performed using Graphpad Prism 6 software (La Jolla,

CA). Statistical significance for more than two groups was determined by one-way analysis of variance and Tukey’s multiple comparison test. Statistical analysis of two groups was determined using unpaired Student’s t test. Differences were considered significant when P values were less than 0.05.

Results

Effect of WA on Acute Pancreatitis Model

Administration of the cholecystokinin analog cerulein has been widely established as a model for induction of acute and CP218, 219. AP pancreatitis was induced in C57BL/6 mice by 7 hourly intraperitoneal injections of cerulein (50 μg/kg) (Figure 4.1a). The mice were euthanized 1 hour after the last injection to harvest plasma and pancreas tissue for analysis. The incidence of AP was determined by analysis of serum amylase levels and histopathological examination of the pancreatic sections. Cerulein treatment increased serum amylase levels by almost 4-fold compared with controls.

70

To test the ability of WA to inhibit cerulein-induced CP, WA was administered 1 hour prior to cerulein injection. WA treatment significantly reduced cerulein-induced serum amylase elevation (Figure 4.2e). Histopathological examination of tissue revealed significant necrosis of the pancreatic parenchyma and increased infiltration of inflammatory cells in cerulein-treated mice (Figure 4.2a, 4.2b). Administration of WA prevented damage to the pancreatic parenchyma and also reduced inflammatory cell infiltration significantly. Pancreas tissue was digested using collagenase, and single cells were stained for innate immune cell markers (CD45, CD11b, CD14, Ly-6g). Flow cytometric analysis revealed a significant increase in the percentage of leucocytes present in the pancreas 1 hour after the last cerulein injection compared with controls, but WA reduced the percentage of infiltrating leucocytes (Figure 4.2c, 4.2d). Myeloid cells, neutrophils, and macrophages were found to be increased in the mouse pancreas after cerulein administration, but this increase was abolished by WA pretreatment.

Several mechanisms of cerulein-induced pancreatitis have been described, but reactive oxygen species (ROS)–modulated secretion of enzymes by acinar cells has been widely investigated220. ROS activation by cerulein on the AR42J pancreatic acinar cell was not reduced by WA (data not shown). ROS activation is upstream of IKKβ activation220, 221, a known mechanism of inhibition of NFκB activation by WA.

71

Figure 4.2. Withaferin A (WA) reduces the severity of acute pancreatitis (AP) and abolishes immune cell infiltration. (a) Hematoxylin and eosin staining of pancreas sections from control, cerulein-treated, and cerulein-plus-WA–treated mice. Representative sections of pancreases from at least 5 mice per group. (b) A histological score of 0 to 5 was assigned by an individual blinded to the study groups, where 0 represents healthy pancreas morphology and 5 represents severe pancreatitis morphology, demonstrating immune cell infiltration, acinar atrophy, intraacinar vacuolization, and/or hemorrhage. At least 4 different loci per pancreas were visually inspected and scored; data are represented as mean ± SD of 5 independent experiments. (c) Flow cytometric analysis of pancreatic cells was performed to investigate immune cell infiltration. Pancreatic cells were stained with CD45, CD11b, Ly6G, and CD14. Data are representative of 5 independent experiments. (d) Representative bar graph showing percentage of CD45-positive leucocytes infiltrating the pancreas. Data are expressed as mean ± SD of 5 mice per group. (e) Elevated amylase level in the serum of mice is a hallmark of AP. Serum amylase was measured using ELISA. Data are expressed as mean ± SD of 5 independent experiments. (b, d, e) *P < 0.05, ***P < 0.001, ****P < 0.0001 (one-way ANOVA with multiple comparison).

72

Attenuation of CP Progression by WA

Two different models of CP were investigated, a preventive and a therapeutic

model. In the preventive model, CP was induced by 7 hourly injections of cerulein (50

μg/kg) injected once a week for 4 weeks. In the treatment group, WA was administered 1

hour prior to cerulein injections every week (Figure 4.1b). Vehicle-treated control mice

exhibited a normal pancreatic morphology and WA administration alone did not cause

any structural change in the pancreatic tissue compared to control (Figure 4.3a). The WA

dose (1.25 mg/kg) was very well tolerated by mice, and there was no significant change

in body weight. CP induced by repeated episodes of AP caused significant morphological

change in the pancreatic parenchyma, as evidenced by hematoxylin and eosin staining of

pancreas tissue. The acinar tissue was atrophic with significant infiltration of

inflammatory cells, ductal metaplasia, and hemorrhage.

Administration of WA 1 hour prior to cerulein injection significantly abolished

progression of CP. Histological examination confirmed that WA protected the pancreatic

parenchyma from cerulein-induced damage and maintained pancreas morphology.

Inflammatory cell infiltration, ductal metaplasia, and hemorrhage were reduced or completely absent in the WA-treated group (Figure 4.3b).

Intraacinar vacuolization is a histological feature in CP and manual blinded

counting revealed a higher number of cells showing intra-acinar vacuolization in the

cerulein-treated group than in the WA group (Figure 4.3c). Blood was collected from mice 1 hour after the last set of cerulein injection. Compared with the control and WA- only–treated group, there was significant elevation of amylase in the serum due to damaged acinar cells leaking enzymes in the blood stream. WA pretreatment reduced the

73

serum amylase levels suggesting a significant reduction in acinar damage due to cerulein

stimulation (Figure 4.3d).

Pancreas tissue collected at the time of harvesting was used to measure local

inflammation by measuring proinflammatory cytokine (IL-6) and chemokine (MCP-1)

from homogenate. Protein levels of both IL-6 and MCP-1 were increased by cerulein injection and WA treatment restored both proteins back to control levels (Figure 4.3e,

4.3f). Together these data suggest that WA has a preventive effect on CP progression.

Presently, there is no prognosis for CP unless it occurs due to heredity or genetic

predisposition11. CP is diagnosed only after several attacks or episodes of AP. Therefore,

to make the experiment clinically relevant, the therapeutic potential of WA was tested

using a therapeutic model. CP was induced by 6 hourly injections of cerulein (50 ug/kg)

injected twice a week for 8 weeks. Mice in the WA treatment group were injected with

cerulein for the first 4 weeks, and WA (0.625 mg/kg) administration was initiated from

week 5 until the end of the procedure (Figure 4.1c). The histopathological examination of

the tissue revealed a normal morphology of vehicle-treated mouse pancreases, while

cerulein administration based on the therapeutic protocol caused pronounced destruction

of pancreatic parenchyma, which was efficiently attenuated by WA administration

(Figure 4.4a). The acinar cell number dropped significantly in cerulein-treated mice

pancreases, and WA treatment protected cells from further damage by cerulein.

One of the hallmarks of advanced CP is the replacement of pancreatic tissue by

fibrosis. To visualize fibrotic changes, pancreas sections were stained for collagen fibers

with Sirius red. Mice injected with cerulein alone demonstrated distinct fibrotic change in

compared with both control and WA-administered mice (Figure 4.4a). The inflammation

74

in the tissue was scored histologically by a blinded observer, and the WA treatment group had a significantly lower score than the cerulein group (Figure 4.4b).

Another hallmark of advanced CP is the lack of enzyme production. Pancreatic amylase after cerulein treatment was undetectable in the serum (data not shown).

Homogenates from the pancreas were used to measure levels of proinflammatory molecules IL-6 and MCP-1. Cerulein treatment significantly increased levels of IL-6 and

MCP-1, which was diminished by WA administration (Figure 4.4c, 4.4d).

Cytoprotective and Antiinflammatory Actions of WA

Cerulein-induced necrotic damage to acinar cell triggers a regenerative response

and proliferation of cells to repair and replace dead cells. Ki67, a marker for proliferation,

was used to assess acinar cell damage. There were little to no Ki67-positive cells in the

pancreas of control mice, but repeated cerulein injections resulted in a significant

increase of Ki67-positive cells in the pancreas (Figure 4.5a). With WA administration,

the percentage of Ki67-positive cells was significantly lower (Figure 4.5b). Comparison

of the preventive and therapeutic CP model revealed more pronounced pancreas damage

in the therapeutic model, but WA was able to potently alleviate the damage caused even

in this model (Figure 4.5c-d).

Tissue sections of mice pancreas were stained for infiltration of neutrophils after

CP was induced. Cerulein-injected mice demonstrated a plethora of granulocytes

infiltrating the pancreas, but WA pretreatment significantly reduced infiltration of

neutrophils (Figure 4.5e, 4.5f).

75

Figure 4.3. Withaferin A (WA) can prevent development of Chronic Pancreatitis (CP). (a) Hematoxylin and eosin staining of pancreas from control, cerulein only, WA only, or cerulein plus WA–treated mice. Representative sections of pancreas from at least 6 mice per group. (b) Histological score was evaluated as previously mentioned from at least 5 different loci per pancreas, with a total of 6 independent experiments per group. (c) Number of intraacinar vacuolizations per frame was manually counted in at least 5 different frames per pancreas and in a total of 6 mice per group. (d) Serum amylase was measured by ELISA after 4 weeks of cerulein injection. Data are expressed as mean ± SD of six independent experiments. Luminex analysis of (e) IL-6 and (f) MCP-1 present in the pancreas homogenate was performed and data were normalized to total protein concentration in the lysate. Data are expressed as mean ± SD of 6 independent experiments. (b–f) *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001 (one-way ANOVA with multiple comparison).

76

Figure 4.4. Withaferin A (WA) can abrogate progression of severe chronic pancreatitis (CP)-therapeutic model. (a) Hematoxylin and eosin staining of pancreas from control, cerulein only, or cerulein plus WA– treated mice. Representative sections of pancreas from at least 6 mice per group. Fibrosis is a hallmark of severe CP, and Sirius red staining was used to visualize fibrotic change in the pancreatic parenchyma. Images are representative of 6 different experiments. (b) Histological score was evaluated as previously mentioned from at least 5 different loci per pancreas, with a total of 6 independent experiments per group. Luminex analysis of (c) IL-6 and (d)) MCP-1 present in the pancreas homogenate was performed and data were normalized to total protein concentration in the lysate. Data are expressed as mean ± SD of 6 independent experiments. (b–d) **P < 0.01, ****P < 0.0001 (one-way ANOVA with multiple comparison).

77

Figure 4.5. Withaferin A (WA) reduces cerulein-induced acinar damage and blocks neutrophil infiltration. (a, c) Immunohistochemical staining of mice pancreas tissue for Ki67, a marker for proliferation, demonstrates severity of damage. DAPI was used to stain the nucleus blue and anti-Ki67 antibody with anti-rabbit fluorescein isothiocyanate–labeled secondary antibody was used. (b, d) ImageJ software was used to determine the percentage of Ki67-positive cells per frame. Data are expressed as mean ± SD representative of 3 different loci in 3 independent experiments. (e) Neutrophil infiltration into the pancreas was visualized by staining for granulocyte marker Ly6G and TRITC-labeled secondary antibody; DAPI was used to stain the nucleus. (f) ImageJ software was used to determine the percentage of neutrophils per frame. Data are expressed as mean ± SD representative of 3 different loci in 3 independent experiments. **P < 0.01, ***P < 0.001 (one-way ANOVA with multiple comparison).

78

Neutrophils, one of the first responder cells to the site of injury, initiate the

inflammatory cascade207, 208, 222. Thus, lack of neutrophil infiltration in WA-pretreated mice suggests reduced inflammation. Furthermore, WA successfully reduced mRNA expression of proinflammatory cytokines (IL-6, TNFa) and proapoptotic molecules

(iNOS, Caspase 3, Caspase 7, and Bax) that were upregulated by cerulein administration

(Figure 4.6b–4.6g). In the therapeutic model, inhibition of IL-6, TNFα, Nos2, and Bax by

WA was observed in a similar fashion (Figure 4.7b–4.7e). Together, these data provide substantial evidence of the antiinflammatory and protective role of WA against cerulein-

induced CP.

Ability of WA to Block NFκB Activation and Inhibit ERK Activation

Overexpression or sustained activation of NFκB in the acinar cells increases the severity of cerulein induced pancreatitis207, 208, 222. WA is known to bind to IKKβ and

inhibit phosphorylation of IκBα, hence preventing activation and translocation of NFκB

into the nucleus. Cerulein administration to mice resulted in activation and translocation

of NFκB in the pancreatic acinar cells (Figure 4.6a). WA treatment not only reduced the

expression but also prevented translocation of the active subunit of NFκB into the

nucleus. Extracellular signaling–regulated kinase 1/2 (ERK1/2) has also been shown to

be activated upon cerulein treatment in acinar cells223. The actual signaling sequence is

still not well understood, but can be either upstream or downstream of NFκB activation

or even independent of NFκB signaling. We treated AR42J cell lines with cerulein 100

nM in the presence or absence of WA and found that ERK phosphorylation was

significantly increased by cerulein within 30 minutes. Strikingly, WA treatment

79 significantly inhibited ERK phosphorylation in the groups both with and without cerulein treatment (Figure 4.7a).

Figure 4.6. Withaferin A (WA) inhibits cerulein-induced nuclear factor kappa B (NFκB) in acinar cells and downregulates expression of proinflammatory and proapoptotic molecules. (a) NFκB translocation into the nucleus was visualized by staining the nucleus with DAPI (blue) and staining the P65 subunit of NFκB with anti-P65 and fluorescein isothiocyanate labeled secondary antibody (green). The black arrows show the translocation of NFκB into the nucleus visualized by merging of green and blue. (b–g) Real-time PCR analysis of proinflammatory cytokines and proapoptotic signals was performed on RNA extracted from the pancreases of CP-induced mice and control mice. (b) IL-6, (c) TNFα, (d) Nos2, (e) Casp3, (f) Casp7, (g) Bax. Data are expressed as box and whisker plot, where the top and bottom ends of the box indicate range, the whiskers indicate SD, and the line in the middle indicates median. Data are representative of at least 6 mice per group. *P < 0.05, **P < 0.01 (one-way ANOVA with multiple comparison).

80

Figure 4.7. Withaferin A (WA) downregulates cerulein-induced expression of proinflammatory and proapoptotic molecules in therapeutic CP model. (a) Western blot for total and phosphorylated extracellular signaling–regulated kinase (ERK) of the MAP kinase pathway from the lysates of AR42J cells treated with cerulein (10-7M) for 30 minutes in the presence or absence of WA (10-6M). Data are representative of 3 independent experiments. (b-e) Real-time PCR analysis of proinflammatory cytokines and proapoptotic signals was performed on RNA extracted from the pancreases of cerulein and cerulein+WA treated mice. (b) IL-6, (c) TNFα, (d) Nos2, (e) Bax. Data are expressed as box and whisker plot, where the top and bottom ends of the box indicate range, the whiskers indicate SD, and the line in the middle indicates median. Data are representative of at least 6 mice per group. *P < 0.05, **P < 0.01 (one- way ANOVA with multiple comparison).

81

Inhibition of ER Stress Pathway During Pathogenesis of CP

Previous reports have demonstrated the significance of sustained activation of ER stress in the pathogenesis of CP215. Due to the high ER volume and protein load on acinar cells, they are highly susceptible to ER stress224. ER stress leads to overexpression of proteins, such as PERK, ATF6 and IRE1α, in the ER lumen. Upon administration of cerulein, pancreas tissue showed significant upregulation of these ER stress proteins

(Figure 4.8a–4.8c). WA-treated mice pancreases had lower expression of these ER stress proteins, and the downregulation correlated with the reduced damage to the pancreas tissue. RNA expression of downstream signaling molecules in the ER stress pathway was also upregulated during CP induction, which was significantly abolished by WA administration (Figure 4.8d–4.8f). Similar results were observed in the therapeutic model of CP (Figure 4.10a–4.10f).

CHOP induction during ER stress in most cases leads to cell apoptosis225. Upon continuous injection of cerulein, CHOP expression increased significantly, which may result in apoptosis of the acinar cells. Cerulein-induced CHOP expression was reduced in mice pancreases by WA (Figure 4.8g, 4.6h).

Inflammasome Signaling in CP and Attenuation by WA

Inflammasome is a large multimeric protein complex produced and assembled by myeloid cells in many chronic inflammatory diseases226. The formation of inflammasome and its role in CP have not been investigated. Tissue damage results in release of several damage markers of DAMPs, such as high-mobility group box (HMGB-1), heat shock proteins (HSPs), dsDNA, and ATP. Cerulein injection caused a significant increase in the

82 expression of HMGB-1 in the acinar cells, but WA administration significantly reduced this expression (Figure 4.9a).

Figure 4.8. Withaferin A (WA) reduces endoplasmic reticulum (ER) stress. Real-time PCR analysis of (a–f) ER stress regulated genes was performed on RNA extracted from pancreases of CP-induced mice and control mice. Data are expressed as box and whisker plot, where the top and bottom ends of the box indicate range, the whiskers indicate SD, and the line in the middle indicates median. Data are representative of at least 6 mice per group. (a) PERK, (b) Atf6, (c) ERN1, (d) Atf4, (e) Xbp1, (f) CHOP. (g) Western blot analysis of lysates from pancreases of severe CP–induced mice and control mice. (h) Semiquantitative analysis of CHOP protein in the lysates. Data are expressed as mean ± SD of 3 independent experiments. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001 (one-way ANOVA with multiple comparison).

83

Figure 4.9. Withaferin A (WA) reduces expression of inflammasome related genes. Real-time PCR analysis inflammasome-related genes was performed on RNA extracted from pancreases of CP-induced mice and control mice. Data are expressed as box and whisker plot, where the top and bottom ends of the box indicate range, the whiskers indicate SD, and the line in the middle indicates median. Data are representative of at least 6 mice per group. (a) HMGB-1, (b) Pycard, (c) NLRP3, (d) IL-18, (e) IL-1β. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001 (one-way ANOVA with multiple comparison).

Infiltrating immune cells such as macrophages, neutrophils, or dendritic cells can

be primed by the DAMPs and initiate production and activation of inflammasome

molecules. CP induction by cerulein significantly increased the expression levels of

NLRP3, and this expression was blocked when WA was injected into the mice (Figure

4.9c). Another important protein required for activation of inflammasome complex is

ASC, and it has a caspase-binding domain called CARD. Expression of ASC was also

significantly increased when cerulein was injected to induce CP but was efficiently

inhibited by WA (Figure 4.9b). Upon activation of inflammasome, the inactive caspase-1

was cleaved to become active caspase-1, and the activated caspase-1 is required for

84

processing and release of IL-1β and IL-18. The expression level of these two cytokines

increased in cerulein-treated mice, but not in WA-pretreated mice (Figure 4.9d, 4.9e).

Similar results were obtained when WA was administered after CP was established in the

therapeutic model (Figure 4.10g–4.10i). In summary, inflammasome signaling took place

during CP, which had not been described previously, and WA administration effectively

inhibited this pathway.

To corroborate the above findings using the cerulein-induced CP model as

relevant to a clinical setting, we tested pancreatic tissue collected from healthy patients

(deceased organ donors) and CP patients (undergoing total pancreatectomy with islet

autotransplantation) for proinflammatory gene expression by RT-PCR array. As shown in

Figure 4.11, most of the proinflammatory genes were highly upregulated (5- to 60-fold

induction) in pancreases from CP patients compared with those from healthy donors.

Discussion

Pancreatitis is characterized by inflammation and parenchymal cell death. These

two key pathologic parameters determine the severity of the disease227. The mechanism of initial insult that leads to pancreatitis is still not clear, although the damage of acinar cells leads to release of several proinflammatory mediators that activate a cascade of molecular signaling and inflammatory events228, 229. In most cases, the inflammation

subsides and results in recovery of the pancreas; this is called AP. In other cases, there is

persistent chronic inflammation that causes continual damage of the pancreas, or there is

recurrent AP, and both situations lead to CP. One of the hallmarks of severe CP is the irreversible replacement of pancreatic acinar cells by fibrotic tissue.

85

Figure 4.10. Real-time PCR analysis of (a–f) ER stress-regulated genes and (g–i) inflammasome-related genes, performed on RNA extracted from pancreases of therapeutic CP-induced mice. Data are expressed as box and whisker plot, where the top and bottom ends of the box indicate range, the whiskers indicate SD, and the line in the middle indicates median. Percent reduction of gene expression by WA compared to cerulein only is shown. Data represent at least 4 mice per group. (a) PERK, (b) Atf6, (c) Ern1, (d) Atf4, (e) Xbp1, (f) CHOP, (g) HMGB1, (h) NLRP3, and (i) IL-1β. NS, not significant, *P<0.05, **P<0.01 (one-way ANOVA with multiple comparison test).

86

Healthy Chronic Pancreatitis

Figure 4.11. Proinflammatory and proapoptotic genes upregulated in the pancreas of human CP patients. (a) Heat map showing upregulation of proinflammatory and proapoptotic genes in the pancreases of patients with advanced-stage chronic pancreatitis (6 per group). Pancreas tissue from healthy individuals was used as a control to determine fold change. (b) The table shows genes that were upregulated 5-fold or more in the CP patient tissue compared to control and that were identified as statistically significant changes among the genes analyzed in the array. Analysis and statistics were performed by Sabiosciences-provided software for custom array plates.

In most severe cases, the CP progresses to diabetes or in some instances to

pancreatic adenocarcinoma. Therefore, it is crucial to identify the trigger and mechanism

that causes this life-threatening illness and to develop a cure for the disease. Many

different experimental models have been devised to understand the mechanism that leads

to different types of pancreatitis230. Cerulein-induced pancreatitis has been widely accepted as a clinically relevant model for recurrent acute necrotizing idiopathic pancreatitis. Several reports accrued over the past years have implicated the role of NFκB activation in the pathogenesis of CP206, 207, 210, but other reports have been

87 contradictory231, 232. Gene knockout studies revealed that the absence of NFκB activity

reduced the severity of AP upon cerulein injection210. When IKKβ was made

constitutively active in the acinar cells of mice, cerulein administration worsened the

disease. To further bolster this result, overexpression of dominant negative IKKβ

ameliorated CP208. Another report showed worsening of pancreatitis when RelA was knocked down in the acinar cells of mice231. Gukovsky et al. have reviewed the role of

NFκB in pancreatitis, providing possible theories behind such contradictory reports233.

Nevertheless, the cerulein model has been shown to activate NFκB in the acinar cells, with continuous stimulation leading to CP in mice211. Therefore, we hypothesized that inhibition of NFκB would protect the mice from progressing to CP.

Since AP is the early event that initiates an inflammatory response that may lead

to CP, it is crucial to block it early. In this study, using a cerulein-induced AP model, we

identified that using WA (an inhibitor of NFκB) reduced disease severity and

inflammation. We then tested if WA could protect the mice from recurrent episodes of

AP that would eventually lead to CP. In the first set of experiments, there were striking

histopathological differences between WA- and cerulein-treated mice pancreases

compared with pancreases of mice receiving cerulein injections alone. Serum amylase

levels, immune cell infiltration, Ki-67 staining, and inflammatory cytokine levels in the pancreas were close to control levels in the WA treatment group, in contrast to results of

the cerulein-treated mice.

Clinically, patients usually experience several episodes of AP before CP is

diagnosed. To make our study clinically relevant, we preestablished CP before WA

treatment was initiated using increased episodes of cerulein each week. We observed

88

striking differences in the morphology of the pancreas tissues. The mice receiving WA

showed significantly reduced inflammatory cell infiltration and fibrosis, but mice

receiving cerulein injections had progressed to severe CP, as demonstrated by fibrotic

change. The therapeutic CP model showed an overall increase in Ki67-positive cells

compared with the preventive CP model, albeit the WA-treated group had profoundly reduced damage. We believe that further cerulein-induced damage was reduced after WA treatment, which delayed the advancement of CP. Together, these data support the protective role of WA in the progression of CP.

Recently, ER stress has been recognized as a possible pathological mechanism in

CP progression215. Loss of pancreatic acinar cells by necroptosis due to indefinite

activation of trypsinogen enzymes leads to activation of many inflammatory signaling

pathways234. This loss may also result in ER stress in remaining cells due to the

overwhelming amount of protein synthesis and release requirements. Cerulein has been

shown to activate the ER stress pathway within 30 minutes of injection, although the

stress molecules subside when the mice pancreases recover. Continuous injection of

cerulein results in chronic ER stress that promotes CP215. Confirming this finding, we

also observed an increase in the expression levels of the ER stress markers upon cerulein

injection to induce CP. More importantly, WA was able to efficiently block the

upregulation of all the ER stress markers analyzed. It is still unclear if the mechanisms of

NFκB and ER stress activation are independent of each other in the pathogenesis of CP,

although activation of NFκB and ER stress signaling take place very early upon cerulein

administration215. Interestingly, inhibition of NFκB activation was able to significantly

89

inhibit the ER stress pathway that leads to CP. Figure 4.12 illustrates such of the

signaling pathways operative in CP and the mechanism of blockade by WA.

The role of inflammasome in the development of CP had previously not been

investigated. Inflammasome activation has been identified as a major innate

inflammatory response in several chronic diseases235. Knockdown of key molecules such

as NLRP3, ASC, and caspase 1 reduced the severity of AP236. We have shown for the

first time the role of inflammasome in the pathogenesis of CP and have shown that NFκB

inhibition significantly reduced inflammasome expression and reduced pancreatitis

severity. NFκB is required for the regulation of IL-1β, which gets processed by the

inflammasome complex. It is plausible that due to chronic inflammation, immune cells

infiltrating the pancreas get primed by DAMPs such as HMGB-1, HSPs, or ATP released by damaged acinar cells via TLRs.

This causes assembly and activation of the inflammasome complex that escalates inflammation during CP. Upon cerulein administration, we have observed significant upregulation of HMGB-1, which is a damage marker and initiator of inflammasome signaling. WA profoundly inhibited HMGB-1 expression. NLRP-3, ASC, IL-1β, and IL-

18 levels were also significantly upregulated, suggesting inflammasome activity in cerulein-induced CP. WA was able to strongly inhibit the expression levels of all the above molecules. Therefore, based on these results, it is likely that NLRP3 inflammasome may be partly responsible for enhancing the severity of CP and that WA can be used as a therapeutic intervention to ameliorate this pathway.

90

FIGURE 4.12: Schematic representation of the signaling pathways operative in the incidence of CP and the blockage by WA. During an initial insult to the acinar cells, intraacinar trypsinogen activation leads to ROS release and perturbation in the ER calcium concentrations. ROS can directly activate the kinases responsible for activation of NFκB leading to the production of proinflammatory cytokines/chemokines. Enzymes released from acinar cells cause further necrotic damage to the neighboring cells, while cytokines/chemokines cause local inflammation, recruitment of the innate immune cells. DAMPs initiate inflammasome activation which also requires NFκB activity for maturation. Loss of acinar cells results in UPR in the remaining acinar cells due to protein overload. Chronic ER stress and inflammasome potentiates CP. Withaferin A inhibits activation of NFκB, and prevents inflammatory response.

91

CHAPTER FIVE

Summary and Conclusions

Summary

The purpose of this study was to identify inflammatory events that occur in the pancreatic exocrine and endocrine tissue and to use the nuclear factor-kappa B (NFκB) inhibitor withaferin A (WA) to diminish the destruction caused by inflammation.

Inflammation in islets during transplantation is a major issue because it causes significant loss of islets within the first 24 hours after transplantation. The mechanism is termed instant blood-mediated inflammatory reaction (IBMIR). After the islets have been

transplanted, it is very difficult to monitor the number of islets that have been

successfully engrafted and the mass of islets that has been lost in the process. Therefore,

identification and validation of a potential biomarker was necessary to monitor graft loss.

Inflammation in the pancreatic acinar cell leads to acute pancreatitis (AP) and chronic

pancreatitis (CP). We hypothesized that blocking a key mediator of inflammmation,

namely NFκB, using WA could reduce inflammation and the severity of CP.

Therefore, this dissertation addressed three major pitfalls:

1) The significant loss of transplanted islets due to IBMIR

2) The lack of a biomarker to determine islet graft loss after transplantation

3) The unclear understanding of the mechanism of, and cure for, CP

92

Since inflammation is the root cause for these pitfalls, and it is well known that NFκB is a master regulator of inflammatory signaling pathways, I have used WA to block NFκB and overcome inflammatory damage.

Alleviation of Instant Blood-Mediated Inflammatory Reaction in Autologous Conditions through Treatment of Human Islets with NFκB Inhibitors

The successful outcome of islet transplantation has been hindered by several

factors. Inflammation begins when the islets are extracted from their natural environment

and continues for a long time after transplantation. NFκB is a major transcription factor

involved in the regulation of inflammatory signaling pathways177. Therefore, NFκB

blockade using WA and CAY10512 (CAY; a potent analog of resveratrol) was

implemented as a strategy to reduce the activation of IBMIR. A miniaturized in vitro tube

model was developed to investigate the effect of these two compounds on IBMIR174.

Previous studies have reported that isolation and culture of islets leads to production of

proinflammatory cytokines and chemokines that are regulated by NFκB178. Thus, we

hypothesized that once islets are mixed with blood, the innate immune system could be

activated, and this could result in secretion of chemokines by the islets, which recruit

monocytes and neutrophils.

Our data showed that NFκB inhibitors could be potent agents for pretreatment of

islets to ameliorate IBMIR. Both WA and CAY treatment preserved islet viability

compared to the control. Both clinical and in vitro studies have reported immediate islet

destruction after infusion or mixing in autologous blood, shown by elevated C-peptide

and proinsulin levels174. When islets were precultured with WA or CAY, the C-peptide

and proinsulin levels were significantly reduced in the serum, suggesting less islet

93

damage. Contact of islets with blood triggers coagulation, and an immediate increase in

thrombin-antithrombin levels was observed when islets were mixed with autologous

blood. WA or CAY did not affect thrombin-antithrombin levels; however, this was an

expected result because coagulation is an NFκB-independent process.

Islets express high levels of tissue factor (TF), and this interacts with factor VIIa

and activates the extrinsic coagulation cascade. The TF gene is regulated by several transcription factors such as NFκB, Egr, AP-1, and NFAT179. In the case of IBMIR, the

major factor that could induce TF expression in the islets would be the proinflammatory

cytokines. Therefore, blocking TF expression by NFκB would protect islets from IBMIR.

Both WA and CAY significantly reduced TF expression in islets when compared with the control. Since islets were cultured in WA or CAY only for an hour before mixing with autologous blood, a prolonged treatment of NFκB might further reduce TF expression in islets and prevent activation of coagulation.

In this study, the plasma levels of several proinflammatory cytokines were found to be increased after mixing of islets with blood. Proinflammatory cytokines TNFα and

IL-6 and chemokines IL-8, IP-10, MCP-1, and G-CSF increased with time in the control group, and both WA and CAY significantly reduced the level of all the cytokines and chemokines. Infiltration of innate inflammatory cells such as neutrophils into the islets was observed at early stages of transplantation. The islets that were mixed with blood showed an intense neutrophil presence even at 3 hours, whereas the WA- and CAY- treated islets showed significantly reduced infiltration of neutrophils. Future studies using this miniature tube model could analyze the effects of inhibitors for various components of IBMIR, such as different anticoagulants and blockers of the innate immune response.

94

In summary, we have demonstrated that a miniaturized in vitro tube model is an effective

tool to study IBMIR. We have also shown that blocking NFκB is a promising strategy to

alleviate the harmful effects of IBMIR on transplanted islets.

Evaluation of MicroRNA375 as a Novel Biomarker for Graft Damage in Clinical Islet Transplantation

In the present study, microRNA 375 (miR375) has been identified as a biomarker

to quantify human islets and detect damage. In the miniaturized in vitro tube model that

was developed to study IBMIR, a rapid increase in the miR375 levels in the plasma

immediately after mixing islets with blood was observed, which was not seen in islet-

only or blood-only controls. The levels of C-peptide correlated with the levels of miR375. This suggests that the early damage is most likely related to an inflammatory response by the innate immune system. There was a significant increase (~1000-fold) in the serum miR375 level in patients receiving autologous and allogenic islets compared with patients undergoing total pancreatectomy only, suggesting that miR375 is released from islets undergoing damage due to inflammation.

A linear relationship was observed between the amount of miR375 and the islet mass. Using this linearity, it was estimated for the first time that each human islet equivalent consists of about 5.66 × 10-2 fmol of miR375. This information is critical to

quantitatively estimate the number of islets damaged under in vitro and/or in vivo

conditions. This data qualified miR375 as a reliable biomarker to evaluate islet damage,

and the slope of linear regression indicated the amount of miR375 per islet.

Based on miRNA quantification, within 6 hours after mixing with autologous

blood in an in vitro tube model, 40% of islets were damaged. Previously, I showed that

95

the viability of islets was reduced to 56.4% at 6 hours after mixing autologous blood and

islets111. Thus, the amount of damaged islets correlates with the loss of viability data.

Clinical data revealed that 52.4% of transplanted islets were damaged after total

pancreatectomy with islet autotransplantation based on miR375 quantitation, matching

previous observations by other groups. The lack of correlation between miR375 levels 1

hour posttransplant and islet mass could be explained by differences in donor

characteristics and the degree of islet damage during isolation.

Of note, miR375 levels in autologous patients were higher than in allogeneic

subjects, while allogeneic patients showed a higher C-peptide response than autologous

patients. This result is interesting, and the difference in biologic characteristics between

the two biomarkers plays a role in the reverse effect. Elevation of C-peptide levels

represents both the physiologic response of pancreatic β cells to hyperglycemia and

mechanical release due to islet damage198, 199. Autologous recipients undergo major

surgery and are on parenteral nutrition with no metabolic stimulation of transplanted

islets. Also, the islets are isolated from inflamed CP pancreases and hence could be more susceptible to immediate damage than allogenic islets in autologous patients184. Further

investigations on biological conditions that modify circulating miR375 levels are

warranted.

The effects of antiinflammatory agents to improve islet transplantation in patients

were tested. Autologous patients not receiving any antiinflammatory compounds

demonstrated 2 peaks of miRNA elevation, one at 1 hour and the other at 24 hours. The

patients who received antiinflammatory agents such as etanercept or a combination of

etanercept and anakinra had a smaller second peak, suggesting that the islet damage

96

observed after 24 hours may be caused by proinflammatory cytokine–induced apoptosis.

Using the in vitro miniaturized model, it was shown that blocking NFκB using WA

significantly reduced miR375 in the serum compared with control, thus confirming the

role of WA in protection of islets from IBMIR-mediated damage. It is hypothesized that the second peak seen at 24 hours may also be significantly reduced by WA, but an in vitro model is not suitable for the longer time points.

Delineating the Molecular Pathogenesis of Chronic Pancreatitis and Use of NFκB Inhibitor to Prevent Its Progression

CP is an inflammatory disease that causes debilitating abdominal pain, exocrine and endocrine pancreatic insufficiency, and severely compromised quality of life. To date, there is no curative treatment for CP or a unified approach to control its progression.

Cerulein-induced pancreatitis is widely accepted as a clinically relevant model for recurrent acute necrotizing idiopathic pancreatitis. The role of NFκB in the development of CP has been widely documented. Therefore, it was hypothesized that inhibition of

NFκB using WA would protect the mice from progressing to CP.

Using a cerulein-induced AP model, it was observed that WA reduced disease severity and inflammation. The ability of WA to protect mice from recurrent episodes of

AP that eventually lead to CP was tested. WA-treated mice pancreases demonstrated reduced inflammation based on histopathological analysis compared with pancreases of mice receiving cerulein injection alone. Serum amylase levels, immune cell infiltration,

Ki-67 staining, and inflammatory cytokine levels in the pancreas were lower in the WA treatment group than in the cerulein-treated mice.

97 To make the experiment clinically relevant, CP was induced prior to WA

treatment. The morphology of the pancreas tissue in WA-treated mice had significantly less damage than that of the non-WA–treated mice. The mice receiving WA showed significantly reduced inflammatory cell infiltration and fibrosis compared to the cerulein-

only–treated mice. These data suggest that further cerulein-induced inflammatory damage was reduced after WA treatment, which delayed the advancement of CP.

CP may result in endoplasmic reticulum (ER) stress in remaining cells due to the overwhelming amount of protein synthesis and release requirements. An increase in the expression levels of ER stress markers was observed in CP mice upon cerulein injection.

WA was able to downregulate the expression of all ER stress markers analyzed.

Interestingly, inhibition of NFκB activation resulted in significant inhibition of the ER stress pathway that promotes the severity of CP.

The role of inflammasome in the development of CP had not been previously investigated. With the current study, the emergence of inflammasome in the pathogenesis of CP has been identified for the first time, and WA significantly reduced inflammasome expression and reduced pancreatitis severity. Cerulein administration resulted in significant upregulation of high-mobility group box-1 (HMGB-1), which is a damage- associated molecular pattern molecule and initiator of inflammasome signaling. WA profoundly inhibited HMGB-1 expression. WA was able to strongly inhibit the expression levels of inflammasome molecules that were upregulated by cerulein.

Therefore, based on these results, it is likely that NLRP3 inflammasome may be partly responsible for enhancing the severity of CP and that WA could be a therapeutic intervention to ameliorate CP.

98

Conclusion

In conclusion, the role of NFκB in causing inflammation to the exocrine (acinar) and endocrine (islet) compartments of the pancreas has been identified. I have demonstrated that WA, a potential inhibitor of NFκB, can resolve inflammation in the islets and acinar cells. WA can be used as a potential therapeutic treatment to improve islet transplant outcomes and also to ameliorate CP. Finally, I have also identified miR375 as a potential biomarker for islet damage during islet transplantation.

99

BIBLIOGRAPHY

1. Langerhans P, Morrison H. Contributions to the Microscopic Anatomy of the Pancreas: Johns Hopkins Press; 1869.

2. Bonner-Weir S. Anatomy of Islet of Langerhans. The Endocrine Pancreas. 1991;edited by E Samols(New York; Raven Press):15-27.

3. American Diabetes A. Diagnosis and classification of diabetes mellitus. Diabetes Care. 2013;36 Suppl 1:S67-74.

4. Soltesz G, Patterson CC, Dahlquist G, Group ES. Worldwide childhood type 1 diabetes incidence--what can we learn from epidemiology? Pediatr Diabetes. 2007;8 Suppl 6:6-14.

5. Pugliese A, Skyler JS. George S. Eisenbarth: insulin and type 1 diabetes. Diabetes Care. 2013;36(6):1437-42.

6. Kobayashi N. The current status of islet transplantation and its perspectives. Rev Diabet Stud. 2008;5(3):136-43.

7. Ryan EA, Shandro T, Green K, Paty BW, Senior PA, Bigam D, et al. Assessment of the severity of hypoglycemia and glycemic lability in type 1 diabetic subjects undergoing islet transplantation. Diabetes. 2004;53(4):955-62.

8. Leitao CB, Tharavanij T, Cure P, Pileggi A, Baidal DA, Ricordi C, et al. Restoration of hypoglycemia awareness after islet transplantation. Diabetes Care. 2008;31(11):2113-5.

9. Vardanyan M, Parkin E, Gruessner C, Rodriguez Rilo HL. Pancreas vs. islet transplantation: a call on the future. Curr Opin Organ Transplant. 2010;15(1):124-30.

10. Ichii H, Ricordi C. Current status of islet cell transplantation. J Hepatobiliary Pancreat Surg. 2009;16(2):101-12.

11. Braganza JM, Lee SH, McCloy RF, McMahon MJ. Chronic pancreatitis. Lancet. 2011;377(9772):1184-97.

12. Nair RJ, Lawler L, Miller MR. Chronic pancreatitis. Am Fam Physician. 2007;76(11):1679-88.

13. Behrman SW, Fowler ES. Pathophysiology of chronic pancreatitis. Surg Clin North Am. 2007;87(6):1309-24, vii.

100

14. Sutherland DE, Matas AJ, Najarian JS. Pancreatic islet cell transplantation. Surg Clin North Am. 1978;58(2):365-82.

15. Center CC. Seventh Annual Report of the Collaborative Islet Transplant Registry. 2011. Available from: http://www.citregistry.org/.

16. Sutherland DE, Gruessner AC, Carlson AM, Blondet JJ, Balamurugan AN, Reigstad KF, et al. Islet autotransplant outcomes after total pancreatectomy: a contrast to islet allograft outcomes. Transplantation. 2008;86(12):1799-802.

17. Kemp CB, Knight MJ, Scharp DW, Ballinger WF, Lacy PE. Effect of transplantation site on the results of pancreatic islet isografts in diabetic rats. Diabetologia. 1973;9(6):486-91.

18. BJ H, MD B, AD S, RG B. Insulin independence following islet transplantation in man: a comparison of different recipient categories. 1996. p. 5-19.

19. Shapiro AM, Lakey JR, Ryan EA, Korbutt GS, Toth E, Warnock GL, et al. Islet transplantation in seven patients with type 1 diabetes mellitus using a glucocorticoid-free immunosuppressive regimen. N Engl J Med. 2000;343(4):230-8.

20. Birks EJ, Owen VJ, Burton PB, Bishop AE, Banner NR, Khaghani A, et al. Tumor necrosis factor-alpha is expressed in donor heart and predicts right ventricular failure after human heart transplantation. Circulation. 2000;102(3):326-31.

21. Nijboer WN, Schuurs TA, van der Hoeven JA, Fekken S, Wiersema-Buist J, Leuvenink HG, et al. Effect of brain death on gene expression and tissue activation in human donor kidneys. Transplantation. 2004;78(7):978-86.

22. Fisher AJ, Donnelly SC, Hirani N, Haslett C, Strieter RM, Dark JH, et al. Elevated levels of interleukin-8 in donor lungs is associated with early graft failure after lung transplantation. Am J Respir Crit Care Med. 2001;163(1):259-65.

23. Murugan R, Venkataraman R, Wahed AS, Elder M, Hergenroeder G, Carter M, et al. Increased plasma interleukin-6 in donors is associated with lower recipient hospital-free survival after cadaveric organ transplantation. Crit Care Med. 2008;36(6):1810-6.

24. Chhabra P, Brayman KL. Current status of immunomodulatory and cellular therapies in preclinical and clinical islet transplantation. Journal of transplantation. 2011;2011:637692.

101

25. Kutlu B, Cardozo AK, Darville MI, Kruhøffer M, Magnusson N, Ørntoft T, et al. Discovery of gene networks regulating cytokine-induced dysfunction and apoptosis in insulin-producing INS-1 cells. Diabetes. 2003;52(11):2701- 19.

26. Barklin A, Larsson A, Vestergaard C, Kjaergaard A, Wogensen L, Schmitz O, et al. Insulin alters cytokine content in two pivotal organs after brain death: a porcine model. Acta Anaesthesiol Scand. 2008;52(5):628-34.

27. Shinichiro O, Kuroki T, Adachi T, Kosaka T, Okamoto T, Tanaka T, et al. The effect of selective neutrophil elastase inhibitor on pancreatic islet yields and functions in rat with hypercytokinemia. Ann Transplant. 2011;16(4):99-106.

28. Kühtreiber WM, Ho LT, Kamireddy A, Yacoub JA, Scharp DW. Islet isolation from human pancreas with extended cold ischemia time. Transplant Proc. 2010;42(6):2027-31.

29. Tsujimura T, Kuroda Y, Churchill TA, Avila JG, Kin T, Shapiro AM, et al. Short- term storage of the ischemically damaged human pancreas by the two- layer method prior to islet isolation. Cell Transplant. 2004;13(1):67-73.

30. De Vos P, Van Straaten JF, Nieuwenhuizen AG, de Groot M, Ploeg RJ, De Haan BJ, et al. Why do microencapsulated islet grafts fail in the absence of fibrotic overgrowth? Diabetes. 1999;48(7):1381-8.

31. de Groot M, Schuurs TA, Keizer PP, Fekken S, Leuvenink HG, van Schilfgaarde R. Response of encapsulated rat pancreatic islets to hypoxia. Cell Transplant. 2003;12(8):867-75.

32. Semenza GL. Life with oxygen. Science. 2007;318(5847):62-4.

33. Naugler WE, Karin M. NF-kappaB and -identifying targets and mechanisms. Curr Opin Genet Dev. 2008;18(1):19-26.

34. Vallabhapurapu S, Karin M. Regulation and function of NF-kappaB transcription factors in the immune system. Annu Rev Immunol. 2009;27:693-733.

35. Pasparakis M. IKK/NF-kappaB signaling in intestinal epithelial cells controls immune homeostasis in the gut. Mucosal Immunol. 2008;1 Suppl 1:S54-7.

36. Taylor CT. Interdependent roles for hypoxia inducible factor and nuclear factor- kappaB in hypoxic inflammation. J Physiol. 2008;586(Pt 17):4055-9.

37. Cummins EP, Berra E, Comerford KM, Ginouves A, Fitzgerald KT, Seeballuck F, et al. Prolyl hydroxylase-1 negatively regulates IkappaB kinase-beta, giving insight into hypoxia-induced NFkappaB activity. Proc Natl Acad Sci U S A. 2006;103(48):18154-9.

102

38. Negi S, Jetha A, Aikin R, Hasilo C, Sladek R, Paraskevas S. Analysis of beta-cell gene expression reveals inflammatory signaling and evidence of dedifferentiation following human islet isolation and culture. PLoS One. 2012;7(1):e30415.

39. Ilmakunnas M, Höckerstedt K, Mäkisalo H, Siitonen S, Repo H, Pesonen EJ. Hepatic IL-8 release during graft procurement is associated with impaired graft function after human liver transplantation. Clin Transplant. 2010;24(1):29-35.

40. Rao JN, Clark SC, Ali S, Kirby J, Flecknell PA, Dark JH. Improvements in lung compliance after pulmonary transplantation: correlation with interleukin 8 expression. Eur J Cardiothorac Surg. 2003;23(4):497-502.

41. De Perrot M, Sekine Y, Fischer S, Waddell TK, McRae K, Liu M, et al. Interleukin-8 release during early reperfusion predicts graft function in human lung transplantation. Am J Respir Crit Care Med. 2002;165(2):211-5.

42. Olsson R, Carlsson PO. Better vascular engraftment and function in pancreatic islets transplanted without prior culture. Diabetologia. 2005;48(3):469-76.

43. King A, Lock J, Xu G, Bonner-Weir S, Weir GC. Islet transplantation outcomes in mice are better with fresh islets and exendin-4 treatment. Diabetologia. 2005;48(10):2074-9.

44. Ihm SH, Matsumoto I, Zhang HJ, Ansite JD, Hering BJ. Effect of short-term culture on functional and stress-related parameters in isolated human islets. Transpl Int. 2009;22(2):207-16.

45. Noguchi H, Naziruddin B, Jackson A, Shimoda M, Ikemoto T, Fujita Y, et al. Fresh islets are more effective for islet transplantation than cultured islets. Cell Transplant. 2012;21(2-3):517-23.

46. Noguchi H, Naziruddin B, Shimoda M, Fujita Y, Chujo D, Takita M, et al. Comparison of fresh and cultured islets from human and porcine pancreata. Transplant Proc. 2010;42(6):2084-6.

47. Lakey JR, Helms LM, Kin T, Korbutt GS, Rajotte RV, Shapiro AM, et al. Serine- protease inhibition during islet isolation increases islet yield from human pancreases with prolonged ischemia. Transplantation. 2001;72(4):565-70.

48. Giuliani M, Moritz W, Bodmer E, Dindo D, Kugelmeier P, Lehmann R, et al. Central necrosis in isolated hypoxic human pancreatic islets: evidence for postisolation ischemia. Cell Transplant. 2005;14(1):67-76.

103 49. Moberg L, Olsson A, Berne C, Felldin M, Foss A, Källen R, et al. Nicotinamide inhibits tissue factor expression in isolated human pancreatic islets: implications for clinical islet transplantation. Transplantation. 2003;76(9):1285-8.

50. Rickels MR, Liu C, Shlansky-Goldberg RD, Soleimanpour SA, Vivek K, Kamoun M, et al. Improvement in beta-Cell Secretory Capacity After Human Islet Transplantation According to the CIT07 Protocol. Diabetes. 2013;62(8):2890-7.

51. Bennet W, Sundberg B, Groth CG, Brendel MD, Brandhorst D, Brandhorst H, et al. Incompatibility between human blood and isolated islets of Langerhans: a finding with implications for clinical intraportal islet transplantation? Diabetes. 1999;48(10):1907-14.

52. Moberg L, Johansson H, Lukinius A, Berne C, Foss A, Källen R, et al. Production of tissue factor by pancreatic islet cells as a trigger of detrimental thrombotic reactions in clinical islet transplantation. Lancet. 2002;360(9350):2039-45.

53. Naziruddin B, Iwahashi S, Kanak MA, Takita M, Itoh T, Levy MF. Evidence for instant blood-mediated inflammatory reaction in clinical autologous islet transplantation. Am J Transplant. 2014;14(2):428-37.

54. Ozmen L, Ekdahl KN, Elgue G, Larsson R, Korsgren O, Nilsson B. Inhibition of thrombin abrogates the instant blood-mediated inflammatory reaction triggered by isolated human islets: possible application of the thrombin inhibitor melagatran in clinical islet transplantation. Diabetes. 2002;51(6):1779-84.

55. Eriksson O, Eich T, Sundin A, Tibell A, Tufveson G, Andersson H, et al. Positron emission tomography in clinical islet transplantation. Am J Transplant. 2009;9(12):2816-24.

56. Nilsson B, Ekdahl KN, Korsgren O. Control of instant blood-mediated inflammatory reaction to improve islets of Langerhans engraftment. Curr Opin Organ Transplant. 2011;16(6):620-6.

57. Rao LV, Rapaport SI, Bajaj SP. Activation of human factor VII in the initiation of tissue factor-dependent coagulation. Blood. 1986;68(3):685-91.

58. Morrissey JH. Tissue factor: an enzyme cofactor and a true receptor. Thromb Haemost. 2001;86(1):66-74.

59. Donovan FM, Pike CJ, Cotman CW, Cunningham DD. Thrombin induces apoptosis in cultured neurons and astrocytes via a pathway requiring tyrosine kinase and RhoA activities. J Neurosci. 1997;17(14):5316-26.

104

60. Turgeon VL, Lloyd ED, Wang S, Festoff BW, Houenou LJ. Thrombin perturbs neurite outgrowth and induces apoptotic cell death in enriched chick spinal motoneuron cultures through caspase activation. J Neurosci. 1998;18(17):6882-91.

61. Johansson H, Goto M, Dufrane D, Siegbahn A, Elgue G, Gianello P, et al. Low molecular weight dextran sulfate: a strong candidate drug to block IBMIR in clinical islet transplantation. American journal of transplantation : official journal of the American Society of Transplantation and the American Society of Transplant Surgeons. 2006;6(2):305-12.

62. Cabric S, Sanchez J, Lundgren T, Foss A, Felldin M, Källen R, et al. Islet surface heparinization prevents the instant blood-mediated inflammatory reaction in islet transplantation. Diabetes. 2007;56(8):2008-15.

63. Johansson U, Elgue G, Nilsson B, Korsgren O. Composite islet-endothelial cell grafts: a novel approach to counteract innate immunity in islet transplantation. American journal of transplantation : official journal of the American Society of Transplantation and the American Society of Transplant Surgeons. 2005;5(11):2632-9.

64. Kim HI, Yu JE, Lee SY, Sul AY, Jang MS, Rashid MA, et al. The effect of composite pig islet-human endothelial cell grafts on the instant blood- mediated inflammatory reaction. Cell Transplant. 2009;18(1):31-7.

65. Johansson U, Rasmusson I, Niclou SP, Forslund N, Gustavsson L, Nilsson B, et al. Formation of composite endothelial cell-mesenchymal stem cell islets: a novel approach to promote islet revascularization. Diabetes. 2008;57(9):2393-401.

66. Dwyer KM, Mysore TB, Crikis S, Robson SC, Nandurkar H, Cowan PJ, et al. The transgenic expression of human CD39 on murine islets inhibits clotting of human blood. Transplantation. 2006;82(3):428-32.

67. Tjernberg J, Ekdahl KN, Lambris JD, Korsgren O, Nilsson B. Acute antibody- mediated complement activation mediates lysis of pancreatic islets cells and may cause tissue loss in clinical islet transplantation. Transplantation. 2008;85(8):1193-9.

68. Fujita E, Farkas I, Campbell W, Baranyi L, Okada H, Okada N. Inactivation of C5a anaphylatoxin by a peptide that is complementary to a region of C5a. J Immunol. 2004;172(10):6382-7.

69. Ritis K, Doumas M, Mastellos D, Micheli A, Giaglis S, Magotti P, et al. A novel C5a receptor-tissue factor cross-talk in neutrophils links innate immunity to coagulation pathways. J Immunol. 2006;177(7):4794-802.

105 70. Tokodai K, Goto M, Inagaki A, Nakanishi W, Okada N, Okada H, et al. C5a- inhibitory peptide combined with gabexate mesilate prevents the instant blood-mediated inflammatory reaction in a rat model of islet transplantation. Transplant Proc. 2010;42(6):2102-3.

71. Tokodai K, Goto M, Inagaki A, Nakanishi W, Ogawa N, Satoh K, et al. Attenuation of cross-talk between the complement and coagulation cascades by C5a blockade improves early outcomes after intraportal islet transplantation. Transplantation. 2010;90(12):1358-65.

72. Goto M, Tjernberg J, Dufrane D, Elgue G, Brandhorst D, Ekdahl KN, et al. Dissecting the instant blood-mediated inflammatory reaction in islet xenotransplantation. Xenotransplantation. 2008;15(4):225-34.

73. Luan NM, Teramura Y, Iwata H. Immobilization of the soluble domain of human complement receptor 1 on agarose-encapsulated islets for the prevention of complement activation. Biomaterials. 2010;31(34):8847-53.

74. Nagata M, Mullen Y, Matsuo S, Herrera M, Clare-Salzler M. Destruction of islet isografts by severe nonspecific inflammation. Transplant Proc. 1990;22(2):855-6.

75. Alejandro R, Cutfield RG, Shienvold FL, Polonsky KS, Noel J, Olson L, et al. Natural history of intrahepatic canine islet cell autografts. J Clin Invest. 1986;78(5):1339-48.

76. Nathan CF. Secretory products of macrophages. J Clin Invest. 1987;79(2):319-26.

77. Bendtzen K, Mandrup-Poulsen T, Nerup J, Nielsen JH, Dinarello CA, Svenson M. Cytotoxicity of human pI 7 interleukin-1 for pancreatic islets of Langerhans. Science. 1986;232(4757):1545-7.

78. Xenos ES, Stevens RB, Sutherland DE, Lokeh A, Ansite JD, Casanova D, et al. The role of nitric oxide in IL-1 beta-mediated dysfunction of rodent islets of Langerhans. Implications for the function of intrahepatic islet grafts. Transplantation. 1994;57(8):1208-12.

79. Montgomery KF, Osborn L, Hession C, Tizard R, Goff D, Vassallo C, et al. Activation of endothelial-leukocyte adhesion molecule 1 (ELAM-1) gene transcription. Proc Natl Acad Sci U S A. 1991;88(15):6523-7.

80. Grilli M, Chiu JJ, Lenardo MJ. NF-kappa B and Rel: participants in a multiform transcriptional regulatory system. Int Rev Cytol. 1993;143:1-62.

81. Degitz K, Li LJ, Caughman SW. Cloning and characterization of the 5'- transcriptional regulatory region of the human intercellular adhesion molecule 1 gene. J Biol Chem. 1991;266(21):14024-30.

106 82. Wachlin G, Augstein P, Schröder D, Kuttler B, Klöting I, Heinke P, et al. IL- 1beta, IFN-gamma and TNF-alpha increase vulnerability of pancreatic beta cells to autoimmune destruction. J Autoimmun. 2003;20(4):303-12.

83. Tan J, Zhou G. Chemokine receptors and transplantation. Cellular & molecular immunology. 2005;2(5):343-9.

84. Baker MS, Chen X, Rotramel AR, Nelson JJ, Lu B, Gerard C, et al. Genetic deletion of chemokine receptor CXCR3 or antibody blockade of its ligand IP-10 modulates posttransplantation graft-site lymphocytic infiltrates and prolongs functional graft survival in pancreatic islet allograft recipients. Surgery. 2003;134(2):126-33.

85. Melzi R, Mercalli A, Sordi V, Cantarelli E, Nano R, Maffi P, et al. Role of CCL2/MCP-1 in islet transplantation. Cell Transplant. 2010;19(8):1031- 46.

86. Citro A, Cantarelli E, Maffi P, Nano R, Melzi R, Mercalli A, et al. CXCR1/2 inhibition enhances pancreatic islet survival after transplantation. J Clin Invest. 2012;122(10):3647-51.

87. Naziruddin B, Wease S, Stablein D, Barton FB, Berney T, Rickels MR, et al. HLA Class I Sensitization in Islet Transplant Recipients - Report from the Collaborative Islet Transplant Registry. Cell Transplant. 2012;21(5):901- 8.

88. Roep BO, Stobbe I, Duinkerken G, van Rood JJ, Lernmark A, Keymeulen B, et al. Auto- and alloimmune reactivity to human islet allografts transplanted into type 1 diabetic patients. Diabetes. 1999;48(3):484-90.

89. van Kampen CA, van de Linde P, Duinkerken G, van Schip JJ, Roelen DL, Keymeulen B, et al. Alloreactivity against repeated HLA mismatches of sequential islet grafts transplanted in non-uremic type 1 diabetes patients. Transplantation. 2005;80(1):118-26.

90. Pujol-Borrell R, Todd I, Doshi M, Bottazzo GF, Sutton R, Gray D, et al. HLA class II induction in human islet cells by interferon-gamma plus tumour necrosis factor or lymphotoxin. Nature. 1987;326(6110):304-6.

91. Soldevila G, Buscema M, Doshi M, James RF, Bottazzo GF, Pujol-Borrell R. Cytotoxic effect of IFN-gamma plus TNF-alpha on human islet cells. J Autoimmun. 1991;4(2):291-306.

92. Jackson AM, Connolly JE, Matsumoto S, Noguchi H, Onaca N, Levy MF, et al. Evidence for Induced Expression of HLA Class II on Human Islets: Possible Mechanism for HLA Sensitization in Transplant Recipients. Transplantation. 2009;87(4):500-6.

107 93. Hearse DJ, Manning AS, Downey JM, Yellon DM. Xanthine oxidase: a critical mediator of myocardial injury during ischemia and reperfusion? Acta Physiol Scand Suppl. 1986;548:65-78.

94. Southorn PA, Powis G. Free radicals in medicine. I. Chemical nature and biologic reactions. Mayo Clin Proc. 1988;63(4):381-9.

95. Zikria BA, Mehmet O, Carlson RW. Reperfusion injuries and clinical capillary leak syndrome: Futura Pub.; 1994.

96. Malaisse WJ, Malaisse-Lagae F, Sener A, Pipeleers DG. Determinants of the selective toxicity of alloxan to the pancreatic . Proc Natl Acad Sci U S A. 1982;79(3):927-30.

97. Lenzen S, Drinkgern J, Tiedge M. Low antioxidant enzyme gene expression in pancreatic islets compared with various other mouse tissues. Free Radic Biol Med. 1996;20(3):463-6.

98. Hotta M, Tashiro F, Ikegami H, Niwa H, Ogihara T, Yodoi J, et al. Pancreatic beta cell-specific expression of thioredoxin, an antioxidative and antiapoptotic protein, prevents autoimmune and streptozotocin-induced diabetes. J Exp Med. 1998;188(8):1445-51.

99. Hohmeier HE, Thigpen A, Tran VV, Davis R, Newgard CB. Stable expression of manganese superoxide dismutase (MnSOD) in insulinoma cells prevents IL-1beta- induced cytotoxicity and reduces nitric oxide production. J Clin Invest. 1998;101(9):1811-20.

100. Benhamou PY, Moriscot C, Richard MJ, Beatrix O, Badet L, Pattou F, et al. Adenovirus-mediated catalase gene transfer reduces oxidant stress in human, porcine and rat pancreatic islets. Diabetologia. 1998;41(9):1093- 100.

101. Carlsson PO, Palm F, Andersson A, Liss P. Markedly decreased oxygen tension in transplanted rat pancreatic islets irrespective of the implantation site. Diabetes. 2001;50(3):489-95.

102. Arteel GE, Thurman RG, Yates JM, Raleigh JA. Evidence that hypoxia markers detect oxygen gradients in liver: pimonidazole and retrograde perfusion of rat liver. Br J Cancer. 1995;72(4):889-95.

103. Carlsson PO, Liss P, Andersson A, Jansson L. Measurements of oxygen tension in native and transplanted rat pancreatic islets. Diabetes. 1998;47(7):1027- 32.

104. Cantarelli E, Melzi R, Mercalli A, Sordi V, Ferrari G, Lederer CW, et al. Bone marrow as an alternative site for islet transplantation. Blood. 2009;114(20):4566-74.

108

105. Maffi P, Balzano G, Ponzoni M, Nano R, Sordi V, Melzi R, et al. Autologous Pancreatic Islet Transplantation in Human Bone Marrow. Diabetes. 2013.

106. Barshes NR, Wyllie S, Goss JA. Inflammation-mediated dysfunction and apoptosis in pancreatic islet transplantation: implications for intrahepatic grafts. J Leukoc Biol. 2005;77(5):587-97.

107. Bottino R, Fernandez LA, Ricordi C, Lehmann R, Tsan MF, Oliver R, et al. Transplantation of allogeneic islets of Langerhans in the rat liver: effects of macrophage depletion on graft survival and microenvironment activation. Diabetes. 1998;47(3):316-23.

108. Vargas F, Vives-Pi M, Somoza N, Armengol P, Alcalde L, Martí M, et al. Endotoxin contamination may be responsible for the unexplained failure of human pancreatic islet transplantation. Transplantation. 1998;65(5):722-7.

109. Gray DW, Sutton R, McShane P, Peters M, Morris PJ. Exocrine contamination impairs implantation of pancreatic islets transplanted beneath the kidney capsule. J Surg Res. 1988;45(5):432-42.

110. Coffey LC, Berman DM, Willman MA, Kenyon NS. Immune cell populations in nonhuman primate islets. Cell Transplant. 2009;18(10):1213-22.

111. Kanak MA, Takita M, Itoh T, Sorelle JA, Murali S, Kunnathodi F, et al. Alleviation of Instant Blood-Mediated Inflammatory Reaction in Autologous Conditions Through Treatment of Human Islets with NF- kappaB Inhibitors. Transplantation. 2014.

112. Lawrence MC, Naziruddin B, Levy MF, Jackson A, McGlynn K. Calcineurin/nuclear factor of activated T cells and MAPK signaling induce TNF-{alpha} gene expression in pancreatic islet endocrine cells. J Biol Chem. 2011;286(2):1025-36.

113. Corbett JA, McDaniel ML. Intraislet release of interleukin 1 inhibits beta cell function by inducing beta cell expression of inducible nitric oxide synthase. J Exp Med. 1995;181(2):559-68.

114. Cardozo AK, Kruhoffer M, Leeman R, Orntoft T, Eizirik DL. Identification of novel cytokine-induced genes in pancreatic beta-cells by high-density oligonucleotide arrays. Diabetes. 2001;50(5):909-20.

115. Paraskevas S, Aikin R, Maysinger D, Lakey JR, Cavanagh TJ, Agapitos D, et al. Modulation of JNK and p38 stress activated protein kinases in isolated islets of Langerhans: insulin as an autocrine survival signal. Annals of surgery. 2001;233(1):124-33.

109

116. Abdelli S, Ansite J, Roduit R, Borsello T, Matsumoto I, Sawada T, et al. Intracellular stress signaling pathways activated during human islet preparation and following acute cytokine exposure. Diabetes. 2004;53(11):2815-23.

117. Arnette D, Gibson TB, Lawrence MC, January B, Khoo S, McGlynn K, et al. Regulation of ERK1 and ERK2 by glucose and peptide hormones in pancreatic beta cells. J Biol Chem. 2003;278(35):32517-25.

118. Lawrence MC, McGlynn K, Park BH, Cobb MH. ERK1/2-dependent activation of transcription factors required for acute and chronic effects of glucose on the insulin gene promoter. J Biol Chem. 2005;280(29):26751-9.

119. Lawrence MC, McGlynn K, Shao C, Duan L, Naziruddin B, Levy MF, et al. Chromatin-bound mitogen-activated protein kinases transmit dynamic signals in transcription complexes in beta-cells. Proc Natl Acad Sci U S A. 2008;105(36):13315-20.

120. Fei H, Zhao B, Zhao S, Wang Q. Requirements of calcium fluxes and ERK kinase activation for glucose- and interleukin-1beta-induced beta-cell apoptosis. Molecular and cellular biochemistry. 2008;315(1-2):75-84.

121. Paraskevas S, Aikin R, Maysinger D, Lakey JR, Cavanagh TJ, Hering B, et al. Activation and expression of ERK, JNK, and p38 MAP-kinases in isolated islets of Langerhans: implications for cultured islet survival. FEBS letters. 1999;455(3):203-8.

122. Widenmaier SB, Ao Z, Kim SJ, Warnock G, McIntosh CH. Suppression of p38 MAPK and JNK via Akt-mediated inhibition of apoptosis signal- regulating kinase 1 constitutes a core component of the beta-cell pro- survival effects of glucose-dependent insulinotropic polypeptide. J Biol Chem. 2009;284(44):30372-82.

123. Omori K, Valiente L, Orr C, Rawson J, Ferreri K, Todorov I, et al. Improvement of human islet cryopreservation by a p38 MAPK inhibitor. American journal of transplantation : official journal of the American Society of Transplantation and the American Society of Transplant Surgeons. 2007;7(5):1224-32.

124. Matsuda T, Omori K, Vuong T, Pascual M, Valiente L, Ferreri K, et al. Inhibition of p38 pathway suppresses human islet production of pro-inflammatory cytokines and improves islet graft function. American journal of transplantation : official journal of the American Society of Transplantation and the American Society of Transplant Surgeons. 2005;5(3):484-93.

110

125. Noguchi H, Nakai Y, Ueda M, Masui Y, Futaki S, Kobayashi N, et al. Activation of c-Jun NH2-terminal kinase (JNK) pathway during islet transplantation and prevention of islet graft loss by intraportal injection of JNK inhibitor. Diabetologia. 2007;50(3):612-9.

126. SoRelle JA, Itoh T, Peng H, Kanak MA, Sugimoto K, Matsumoto S, et al. Withaferin A inhibits pro-inflammatory cytokine-induced damage to islets in culture and following transplantation. Diabetologia. 2013;56(4):814-24.

127. Spinas GA, Palmer JP, Mandrup-Poulsen T, Andersen H, Nielsen JH, Nerup J. The bimodal effect of interleukin 1 on rat pancreatic beta-cells-- stimulation followed by inhibition--depends upon dose, duration of exposure, and ambient glucose concentration. Acta endocrinologica. 1988;119(2):307-11.

128. Lawrence MC, Shao C, McGlynn K, Naziruddin B, Levy MF, Cobb MH. Multiple chromatin-bound protein kinases assemble factors that regulate insulin gene transcription. Proc Natl Acad Sci U S A. 2009;106(52):22181-6.

129. Corbett JA, Sweetland MA, Wang JL, Lancaster JR, Jr., McDaniel ML. Nitric oxide mediates cytokine-induced inhibition of insulin secretion by human islets of Langerhans. Proc Natl Acad Sci U S A. 1993;90(5):1731-5.

130. Shao C, Lawrence MC, Cobb MH. Regulation of CCAAT/enhancer-binding protein homologous protein (CHOP) expression by interleukin-1 beta in pancreatic beta cells. J Biol Chem. 2010;285(26):19710-9.

131. Oetjen E, Blume R, Cierny I, Schlag C, Kutschenko A, Kratzner R, et al. Inhibition of MafA transcriptional activity and human insulin gene transcription by interleukin-1beta and mitogen-activated protein kinase kinase kinase in pancreatic islet beta cells. Diabetologia. 2007;50(8):1678- 87.

132. Sekine N, Ishikawa T, Okazaki T, Hayashi M, Wollheim CB, Fujita T. Synergistic activation of NF-kappab and inducible isoform of nitric oxide synthase induction by interferon-gamma and tumor necrosis factor-alpha in INS-1 cells. J Cell Physiol. 2000;184(1):46-57.

133. Chang I, Kim S, Kim JY, Cho N, Kim YH, Kim HS, et al. Nuclear factor kappaB protects pancreatic beta-cells from tumor necrosis factor-alpha-mediated apoptosis. Diabetes. 2003;52(5):1169-75.

134. Heit JJ, Apelqvist AA, Gu X, Winslow MM, Neilson JR, Crabtree GR, et al. Calcineurin/NFAT signalling regulates pancreatic beta-cell growth and function. Nature. 2006;443(7109):345-9.

111

135. Goodyer WR, Gu X, Liu Y, Bottino R, Crabtree GR, Kim SK. Neonatal beta cell development in mice and humans is regulated by calcineurin/NFAT. Developmental cell. 2012;23(1):21-34.

136. Kim S, Millet I, Kim HS, Kim JY, Han MS, Lee MK, et al. NF-kappa B prevents beta cell death and autoimmune diabetes in NOD mice. Proc Natl Acad Sci U S A. 2007;104(6):1913-8.

137. Shapiro AM. State of the art of clinical islet transplantation and novel protocols of immunosuppression. Curr Diab Rep. 2011;11(5):345-54.

138. McCall M, James Shapiro AM. Update on islet transplantation. Cold Spring Harb Perspect Med. 2012;2(7):a007823.

139. Barton FB, Rickels MR, Alejandro R, Hering BJ, Wease S, Naziruddin B, et al. Improvement in outcomes of clinical islet transplantation: 1999-2010. Diabetes Care. 2012;35(7):1436-45.

140. Hering BJ, Kandaswamy R, Ansite JD, Eckman PM, Nakano M, Sawada T, et al. Single-donor, marginal-dose islet transplantation in patients with type 1 diabetes. JAMA. 2005;293(7):830-5.

141. Froud T, Ricordi C, Baidal DA, Hafiz MM, Ponte G, Cure P, et al. Islet transplantation in type 1 diabetes mellitus using cultured islets and steroid- free immunosuppression: Miami experience. American journal of transplantation : official journal of the American Society of Transplantation and the American Society of Transplant Surgeons. 2005;5(8):2037-46.

142. Bellin MD, Barton FB, Heitman A, Harmon JV, Kandaswamy R, Balamurugan AN, et al. Potent induction immunotherapy promotes long-term insulin independence after islet transplantation in type 1 diabetes. Am J Transplant. 2012;12(6):1576-83.

143. Kandaswamy R, Stock PG, Skeans MA, Gustafson SK, Sleeman EF, Wainright JL, et al. OPTN/SRTR 2011 Annual Data Report: pancreas. American journal of transplantation : official journal of the American Society of Transplantation and the American Society of Transplant Surgeons. 2013;13 Suppl 1:47-72.

144. McCall M, Pawlick R, Kin T, Shapiro AM. Anakinra potentiates the protective effects of etanercept in transplantation of marginal mass human islets in immunodeficient mice. American journal of transplantation : official journal of the American Society of Transplantation and the American Society of Transplant Surgeons. 2012;12(2):322-9.

112

145. Matsumoto S, Takita M, Chaussabel D, Noguchi H, Shimoda M, Sugimoto K, et al. Improving Efficacy of Clinical Islet Transplantation with Iodixanol Based Islet Purification, Thymoglobulin Induction and Blockage of IL-1- beta and TNF-alpha. Cell transplantation. 2011;20(10):1641-7.

146. Pileggi A, Molano RD, Berney T, Cattan P, Vizzardelli C, Oliver R, et al. Heme oxygenase-1 induction in islet cells results in protection from apoptosis and improved in vivo function after transplantation. Diabetes. 2001;50(9):1983-91.

147. Poss KD, Tonegawa S. Reduced stress defense in heme oxygenase 1-deficient cells. Proc Natl Acad Sci U S A. 1997;94(20):10925-30.

148. Poss KD, Tonegawa S. Heme oxygenase 1 is required for mammalian iron reutilization. Proc Natl Acad Sci U S A. 1997;94(20):10919-24.

149. Günther L, Berberat PO, Haga M, Brouard S, Smith RN, Soares MP, et al. Carbon monoxide protects pancreatic beta-cells from apoptosis and improves islet function/survival after transplantation. Diabetes. 2002;51(4):994-9.

150. Grey ST, Longo C, Shukri T, Patel VI, Csizmadia E, Daniel S, et al. Genetic engineering of a suboptimal islet graft with A20 preserves beta cell mass and function. J Immunol. 2003;170(12):6250-6.

151. Cheng EY, Sharma VK, Chang C, Ding R, Allison AC, Leeser DB, et al. Diannexin decreases inflammatory cell infiltration into the islet graft, reduces β-cell apoptosis, and improves early graft function. Transplantation. 2010;90(7):709-16.

152. Jin SM, Kim KS, Lee SY, Gong CH, Park SK, Shin JS, et al. The sequential combination of a JNK inhibitor and simvastatin protects porcine islets from peritransplant apoptosis and inflammation. Cell Transplant. 2011;20(7):1139-51.

153. Cure P, Pileggi A, Froud T, Messinger S, Faradji RN, Baidal DA, et al. Improved metabolic control and quality of life in seven patients with type 1 diabetes following islet after kidney transplantation. Transplantation. 2008;85(6):801-12.

154. Froud T, Baidal DA, Faradji R, Cure P, Mineo D, Selvaggi G, et al. Islet transplantation with alemtuzumab induction and calcineurin-free maintenance immunosuppression results in improved short- and long-term outcomes. Transplantation. 2008;86(12):1695-701.

155. Bellin MD, Kandaswamy R, Parkey J, Zhang HJ, Liu B, Ihm SH, et al. Prolonged insulin independence after islet allotransplants in recipients with type 1 diabetes. Am J Transplant. 2008;8(11):2463-70.

113

156. Gangemi A, Salehi P, Hatipoglu B, Martellotto J, Barbaro B, Kuechle JB, et al. Islet transplantation for brittle type 1 diabetes: the UIC protocol. Am J Transplant. 2008;8(6):1250-61.

157. Vanden Berghe W, Sabbe L, Kaileh M, Haegeman G, Heyninck K. Molecular insight in the multifunctional activities of Withaferin A. Biochem Pharmacol. 2012;84(10):1282-91.

158. Chopra A, Lavin P, Patwardhan B, Chitre D. A 32-week randomized, placebo- controlled clinical evaluation of RA-11, an Ayurvedic drug, on osteoarthritis of the knees. J Clin Rheumatol. 2004;10(5):236-45.

159. Cooley K, Szczurko O, Perri D, Mills EJ, Bernhardt B, Zhou Q, et al. Naturopathic care for anxiety: a randomized controlled trial ISRCTN78958974. PLoS One. 2009;4(8):e6628.

160. Kaileh M, Vanden Berghe W, Heyerick A, Horion J, Piette J, Libert C, et al. Withaferin a strongly elicits IkappaB kinase beta hyperphosphorylation concomitant with potent inhibition of its kinase activity. J Biol Chem. 2007;282(7):4253-64.

161. Grover A, Shandilya A, Punetha A, Bisaria VS, Sundar D. Inhibition of the NEMO/IKKbeta association complex formation, a novel mechanism associated with the NF-kappaB activation suppression by Withania somnifera's key metabolite withaferin A. BMC Genomics. 2010;11 Suppl 4:S25.

162. Barnes PJ. Nuclear factor-kappa B. Int J Biochem Cell Biol. 1997;29(6):867-70.

163. Barnes PJ, Karin M. Nuclear factor-kappaB: a pivotal transcription factor in chronic inflammatory diseases. N Engl J Med. 1997;336(15):1066-71.

164. Maitra R, Porter MA, Huang S, Gilmour BP. Inhibition of NFkappaB by the natural product Withaferin A in cellular models of Cystic Fibrosis inflammation. J Inflamm (Lond). 2009;6:15.

165. Flodstrom M, Welsh N, Eizirik DL. Cytokines activate the nuclear factor kappa B (NF-kappa B) and induce nitric oxide production in human pancreatic islets. FEBS letters. 1996;385(1-2):4-6.

166. Ortis F, Pirot P, Naamane N, Kreins AY, Rasschaert J, Moore F, et al. Induction of nuclear factor-kappaB and its downstream genes by TNF-alpha and IL- 1beta has a pro-apoptotic role in pancreatic beta cells. Diabetologia. 2008;51(7):1213-25.

114

167. Sutherland DE, Radosevich DM, Bellin MD, Hering BJ, Beilman GJ, Dunn TB, et al. Total pancreatectomy and islet autotransplantation for chronic pancreatitis. Journal of the American College of Surgeons. 2012;214(4):409-24; discussion 24-6.

168. Ryan EA, Paty BW, Senior PA, Bigam D, Alfadhli E, Kneteman NM, et al. Five- year follow-up after clinical islet transplantation. Diabetes. 2005;54(7):2060-9.

169. Robertson RP. Islet transplantation a decade later and strategies for filling a half- full glass. Diabetes. 2010;59(6):1285-91.

170. Johansson H, Lukinius A, Moberg L, Lundgren T, Berne C, Foss A, et al. Tissue factor produced by the endocrine cells of the islets of Langerhans is associated with a negative outcome of clinical islet transplantation. Diabetes. 2005;54(6):1755-62.

171. Takahashi T, Matsumoto S, Matsushita M, Kamachi H, Tsuruga Y, Kasai H, et al. Donor pretreatment with DHMEQ improves islet transplantation. J Surg Res. 2010;163(1):e23-34.

172. Kollander R, Solovey A, Milbauer LC, Abdulla F, Kelm RJ, Jr., Hebbel RP. Nuclear factor-kappa B (NFkappaB) component p50 in blood mononuclear cells regulates endothelial tissue factor expression in sickle transgenic mice: implications for the coagulopathy of sickle cell disease. Translational research : the journal of laboratory and clinical medicine. 2010;155(4):170-7.

173. Cucciolla V, Borriello A, Oliva A, Galletti P, Zappia V, Della Ragione F. Resveratrol: from basic science to the clinic. . 2007;6(20):2495- 510.

174. Naziruddin B, Iwahashi S, Kanak M, Takita M, Itoh T, Levy M. Evidence for instant blood-mediated inflammatory reaction in clinical autologous islet transplantation. American journal of transplantation : official journal of the American Society of Transplantation and the American Society of Transplant Surgeons. In Press.

175. Matsumoto S, Okitsu T, Iwanaga Y, Noguchi H, Nagata H, Yonekawa Y, et al. Successful islet transplantation from nonheartbeating donor pancreata using modified Ricordi islet isolation method. Transplantation. 2006;82(4):460-5.

176. Ricordi C, Lacy PE, Finke EH, Olack BJ, Scharp DW. Automated method for isolation of human pancreatic islets. Diabetes. 1988;37(4):413-20.

115

177. Pasparakis M. Regulation of tissue homeostasis by NF-kappaB signalling: implications for inflammatory diseases. Nature reviews Immunology. 2009;9(11):778-88.

178. Jackson A, McWilliams C, Kaizer E, Chaussabel D, Glaser C, Noguchi H, et al. Gene expression profiling of human pancreatic islets undergoing a simulated process of instant blood-mediated inflammatory reaction. Transplant Proc. 2008;40(2):430-2.

179. Mackman N. Regulation of the tissue factor gene. Thromb Haemost. 1997;78(1):747-54.

180. Ruf W, Riewald M. Regulation of Tissue Factor Expression. In: ten Cate H, Levi M, editors. Molecular mechanisms of disseminated intravascular coagulation. Georgetown, TX: Landes Bioscience; 2003. p. 61–80.

181. Parry GC, Mackman N. Transcriptional regulation of tissue factor expression in human endothelial cells. Arteriosclerosis, thrombosis, and vascular biology. 1995;15(5):612-21.

182. Ishizuka N, Yagui K, Tokuyama Y, Yamada K, Suzuki Y, Miyazaki J, et al. Tumor necrosis factor alpha signaling pathway and apoptosis in pancreatic beta cells. Metabolism: clinical and experimental. 1999;48(12):1485-92.

183. Ziraldo C, Vodovotz Y, Namas RA, Almahmoud K, Tapias V, Mi Q, et al. Central Role for MCP-1/CCL2 in Injury-Induced Inflammation Revealed by In Vitro, In Silico, and Clinical Studies. PLoS One. 2013;8(12):e79804.

184. Bellin MD, Beilman GJ, Dunn TB, Pruett TL, Chinnakotla S, Wilhelm JJ, et al. Islet autotransplantation to preserve beta cell mass in selected patients with chronic pancreatitis and diabetes mellitus undergoing total pancreatectomy. Pancreas. 2013;42(2):317-21.

185. Balzano G, Piemonti L. Autologous islet transplantation in patients requiring pancreatectomy for neoplasm. Curr Diab Rep. 2014;14(8):512.

186. Bruni A, Gala-Lopez B, Pepper AR, Abualhassan NS, Shapiro AJ. Islet cell transplantation for the treatment of type 1 diabetes: recent advances and future challenges. Diabetes Metab Syndr Obes. 2014;7:211-23.

187. Berney T, Toso C. Monitoring of the islet graft. Diabetes Metab. 2006;32(5 Pt 2):503-12.

188. Griffiths-Jones S, Saini HK, van Dongen S, Enright AJ. miRBase: tools for microRNA genomics. Nucleic acids research. 2008;36(Database issue):D154-8.

116

189. Gilad S, Meiri E, Yogev Y, Benjamin S, Lebanony D, Yerushalmi N, et al. Serum microRNAs are promising novel biomarkers. PLoS One. 2008;3(9):e3148.

190. Heneghan HM, Miller N, Lowery AJ, Sweeney KJ, Newell J, Kerin MJ. Circulating microRNAs as novel minimally invasive biomarkers for . Annals of surgery. 2010;251(3):499-505.

191. Poy MN, Eliasson L, Krutzfeldt J, Kuwajima S, Ma X, Macdonald PE, et al. A pancreatic islet-specific microRNA regulates insulin secretion. Nature. 2004;432(7014):226-30.

192. Poy MN, Hausser J, Trajkovski M, Braun M, Collins S, Rorsman P, et al. miR- 375 maintains normal pancreatic alpha- and beta-cell mass. Proc Natl Acad Sci U S A. 2009;106(14):5813-8.

193. Tang X, Muniappan L, Tang G, Ozcan S. Identification of glucose-regulated miRNAs from pancreatic {beta} cells reveals a role for miR-30d in insulin transcription. Rna. 2009;15(2):287-93.

194. Erener S, Mojibian M, Fox JK, Denroche HC, Kieffer TJ. Circulating miR-375 as a biomarker of beta-cell death and diabetes in mice. Endocrinology. 2013;154(2):603-8.

195. Matsumoto S, Noguchi H, Naziruddin B, Onaca N, Jackson A, Nobuyo H, et al. Improvement of pancreatic islet cell isolation for transplantation. Proc (Bayl Univ Med Cent). 2007;20(4):357-62.

196. Takita M, Matsumoto S, Shimoda M, Chujo D, Itoh T, Sorelle JA, et al. Safety and tolerability of the T-cell depletion protocol coupled with anakinra and etanercept for clinical islet cell transplantation. Clin Transplant. 2012;26(5):E471-84.

197. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real- time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 2001;25(4):402-8.

198. Jones AG, Hattersley AT. The clinical utility of C-peptide measurement in the care of patients with diabetes. Diabetic medicine : a journal of the British Diabetic Association. 2013;30(7):803-17.

199. Itoh T, Iwahashi S, Kanak MA, Shimoda M, Takita M, Chujo D, et al. Elevation of high-mobility group box 1 after clinical autologous islet transplantation and its inverse correlation with outcomes. Cell transplantation. 2014;23(2):153-65.

200. Piemonti L, Correa-Tapia M, Lampasona V, Ferrer J. Circulating miR-375 levels after islet transplantation in humans: A biomarker for ongoing beta cell injury. CellR4. 2013;1(1):e35.

117

201. Ammann RW, Muellhaupt B. The natural history of pain in alcoholic chronic pancreatitis. Gastroenterology. 1999;116(5):1132-40.

202. Mullady DK, Yadav D, Amann ST, O'Connell MR, Barmada MM, Elta GH, et al. Type of pain, pain-associated complications, quality of life, disability and resource utilisation in chronic pancreatitis: a prospective cohort study. Gut. 2011;60(1):77-84.

203. Muniraj T, Aslanian HR, Farrell J, Jamidar PA. Chronic pancreatitis, a comprehensive review and update. Part I: epidemiology, etiology, risk factors, genetics, pathophysiology, and clinical features. Dis Mon. 2014;60(12):530-50.

204. Muniraj T, Aslanian HR, Farrell J, Jamidar PA. Chronic pancreatitis, a comprehensive review and update. Part II: Diagnosis, complications, and management. Dis Mon. 2015;61(1):5-37.

205. Whitcomb DC. Genetic risk factors for pancreatic disorders. Gastroenterology. 2013;144(6):1292-302.

206. Chen X, Ji B, Han B, Ernst SA, Simeone D, Logsdon CD. NF-kappaB activation in pancreas induces pancreatic and systemic inflammatory response. Gastroenterology. 2002;122(2):448-57.

207. Aleksic T, Baumann B, Wagner M, Adler G, Wirth T, Weber CK. Cellular immune reaction in the pancreas is induced by constitutively active IkappaB kinase-2. Gut. 2007;56(2):227-36.

208. Baumann B, Wagner M, Aleksic T, von Wichert G, Weber CK, Adler G, et al. Constitutive IKK2 activation in acinar cells is sufficient to induce pancreatitis in vivo. J Clin Invest. 2007;117(6):1502-13.

209. Yan MX, Ren HB, Kou Y, Meng M, Li YQ. Involvement of nuclear factor kappa B in high-fat diet-related pancreatic fibrosis in rats. Gut Liver. 2012;6(3):381-7.

210. Altavilla D, Famulari C, Passaniti M, Galeano M, Macri A, Seminara P, et al. Attenuated cerulein-induced pancreatitis in nuclear factor-kappaB- deficient mice. Lab Invest. 2003;83(12):1723-32.

211. Steinle AU, Weidenbach H, Wagner M, Adler G, Schmid RM. NF-kappaB/Rel activation in cerulein pancreatitis. Gastroenterology. 1999;116(2):420-30.

212. Kaufman RJ. Stress signaling from the lumen of the endoplasmic reticulum: coordination of gene transcriptional and translational controls. Genes Dev. 1999;13(10):1211-33.

118

213. Ron D, Walter P. Signal integration in the endoplasmic reticulum unfolded protein response. Nat Rev Mol Cell Biol. 2007;8(7):519-29.

214. Schroder M, Kaufman RJ. The mammalian unfolded protein response. Annu Rev Biochem. 2005;74:739-89.

215. Sah RP, Garg SK, Dixit AK, Dudeja V, Dawra RK, Saluja AK. Endoplasmic reticulum stress is chronically activated in chronic pancreatitis. J Biol Chem. 2014;289(40):27551-61.

216. Schroder K, Tschopp J. The inflammasomes. Cell. 2010;140(6):821-32.

217. Heyninck K, Lahtela-Kakkonen M, Van der Veken P, Haegeman G, Vanden Berghe W. Withaferin A inhibits NF-kappaB activation by targeting 179 in IKKbeta. Biochem Pharmacol. 2014;91(4):501-9.

218. Lara LF, Levy MJ. Idiopathic recurrent acute pancreatitis. MedGenMed. 2004;6(4):10.

219. Neuschwander-Tetri BA, Burton FR, Presti ME, Britton RS, Janney CG, Garvin PR, et al. Repetitive self-limited acute pancreatitis induces pancreatic fibrogenesis in the mouse. Dig Dis Sci. 2000;45(4):665-74.

220. Yu JH, Lim JW, Namkung W, Kim H, Kim KH. Suppression of cerulein-induced cytokine expression by antioxidants in pancreatic acinar cells. Lab Invest. 2002;82(10):1359-68.

221. Gloire G, Legrand-Poels S, Piette J. NF-kappaB activation by reactive oxygen species: fifteen years later. Biochem Pharmacol. 2006;72(11):1493-505.

222. Huang H, Liu Y, Daniluk J, Gaiser S, Chu J, Wang H, et al. Activation of nuclear factor-kappaB in acinar cells increases the severity of pancreatitis in mice. Gastroenterology. 2013;144(1):202-10.

223. Ju KD, Yu JH, Kim H, Kim KH. Role of mitogen-activated protein kinases, NF- kappaB, and AP-1 on cerulein-induced IL-8 expression in pancreatic acinar cells. Ann N Y Acad Sci. 2006;1090:368-74.

224. Kim I, Xu W, Reed JC. Cell death and endoplasmic reticulum stress: disease relevance and therapeutic opportunities. Nat Rev Drug Discov. 2008;7(12):1013-30.

225. Walter P, Ron D. The unfolded protein response: from stress pathway to homeostatic regulation. Science. 2011;334(6059):1081-6.

226. Latz E, Xiao TS, Stutz A. Activation and regulation of the inflammasomes. Nature reviews Immunology. 2013;13(6):397-411.

119

227. Pandol SJ, Saluja AK, Imrie CW, Banks PA. Acute pancreatitis: bench to the bedside. Gastroenterology. 2007;132(3):1127-51.

228. Grady T, Liang P, Ernst SA, Logsdon CD. Chemokine gene expression in rat pancreatic acinar cells is an early event associated with acute pancreatitis. Gastroenterology. 1997;113(6):1966-75.

229. Gukovskaya AS, Gukovsky I, Zaninovic V, Song M, Sandoval D, Gukovsky S, et al. Pancreatic acinar cells produce, release, and respond to tumor necrosis factor-alpha. Role in regulating cell death and pancreatitis. J Clin Invest. 1997;100(7):1853-62.

230. Lerch MM, Gorelick FS. Models of Acute and Chronic Pancreatitis. Gastroenterology. 2013;144(6):1180-93.

231. Algul H, Treiber M, Lesina M, Nakhai H, Saur D, Geisler F, et al. Pancreas- specific RelA/p65 truncation increases susceptibility of acini to inflammation-associated cell death following cerulein pancreatitis. J Clin Invest. 2007;117(6):1490-501.

232. Neuhofer P, Liang S, Einwachter H, Schwerdtfeger C, Wartmann T, Treiber M, et al. Deletion of IkappaBalpha activates RelA to reduce acute pancreatitis in mice through up-regulation of Spi2A. Gastroenterology. 2013;144(1):192- 201.

233. Gukovsky I, Gukovskaya A. Nuclear factor-kappaB in pancreatitis: Jack-of-all- trades, but which one is more important? Gastroenterology. 2013;144(1):26-9.

234. He S, Wang L, Miao L, Wang T, Du F, Zhao L, et al. Receptor interacting protein kinase-3 determines cellular necrotic response to TNF-alpha. Cell. 2009;137(6):1100-11.

235. Ozaki E, Campbell M, Doyle SL. Targeting the NLRP3 inflammasome in chronic inflammatory diseases: current perspectives. J Inflamm Res. 2015;8:15-27.

236. Hoque R, Sohail M, Malik A, Sarwar S, Luo Y, Shah A, et al. TLR9 and the NLRP3 inflammasome link acinar cell death with inflammation in acute pancreatitis. Gastroenterology. 2011;141(1):358-69.

120