The ERdj4 is required for survival, glucose metabolism and B cell development

A dissertation submitted to the University of Cincinnati

in partial fulfillment of the requirements for the degree of

DOCTOR OF PHILOSOPHY

in the Immunobiology Graduate Program

of the College of Medicine

2012

by

Jill Marie Fritz

B.A., Miami University, Oxford, OH

Advisory Committee: Timothy Weaver, M.S., Ph.D., Chair George Deepe, M.D. Fred Finkelman, M.D. H. Leighton Grimes, Ph.D. Christopher Karp, M.D. Francis McCormack, M.D.

ABSTRACT

The ER-localized DnaJ homologue 4 (ERdj4) is a soluble ER chaperone induced by the unfolded response (UPR) to assist in the removal of unfolded/misfolded from the ER lumen for proteasomal degradation. To elucidate the function of ERdj4 in vivo, ERdj4 trap (ERdj4GT/GT) mice were generated from embryonic stem cells harboring a gene trap cassette inserted into the ERdj4 . ERdj4GT/GT mice expressed hypomorphic levels of ERdj4 with a 10-100 fold reduction in all tissues and cell types examined. Approximately 30-50% of ERdj4GT/GT mice died perinatally in association with growth retardation and hypoglycemia. ERdj4GT/GT neonates exhibited signs of delayed pancreatic development, including abnormal distribution of pancreatic

α- and β-cells and reduced insulin and glucagon in the pancreas. Surviving adult

ERdj4GT/GT mice were glucose intolerant, resulting from hypoinsulinemia rather than insulin resistance. Pancreatic β-cells exhibited increased ER stress, including ER dilation and upregulation of the UPR. Proinsulin accumulated in the ER of β-cells from

ERdj4GT/GT mice consistent with our previous finding that proinsulin is a substrate for

ERdj4. The insulin processing enzymes, including Pcsk1, Pcsk2 and CPE, also associated with ERdj4, contributing to defects in proinsulin biosynthesis in ERdj4GT/GT mice. ERdj4 deficiency also resulted in pancreatic α-cell hyperplasia in association with increased ER stress. Since previous studies had demonstrated that the UPR is required for both early and late B lymphopoiesis, we hypothesized that ERdj4 deficiency would affect B cell development. Pro-B, pre-B, immature and mature B cell populations were significantly reduced in the bone marrow of ERdj4GT/GT mice in association with increased pro-B cell death. Further, mature B cell populations were reduced in the spleen and peritoneal cavity. ERdj4GT/GT donor cells transplanted into ERdj4+/+ recipients rescued all stages of B cell development, confirming a defect in the microenvironment. Osteogenic cells support B cell development and the UPR is required for osteoblast differentiation and function. Bone-lining cells, which differentiate from osteoblasts, were reduced in ERdj4GT/GT mice in association with increased ER stress. Further, primary bone cells from ERdj4GT/GT mice had reduced expression of osteogenic-specific markers. These data suggest that the loss of ERdj4 affects the maturation/survival of osteogenic cells, which in turn leads to a defect in B lymphopoiesis. Collectively, this work reveals an important role for ERdj4 in survival, glucose metabolism and B cell development.

iii iv ACKNOWLEDGMENTS

First and foremost, I wish to express sincere gratitude and appreciation to my scientific thesis advisor, Dr. Timothy Weaver. Without his dedication, guidance and encouragement, this work would not be possible.

I also wholeheartedly thank my committee members Dr. George Deepe, Dr. Fred

Finkelman, Dr. Lee Grimes, Dr. Chris Karp and Dr. Frank McCormack for their insight and support during my scientific training. I sincerely appreciate their efforts and direction in completing this work.

I acknowledge all the members of the Weaver lab, both past and present, for their friendship, motivation and expertise. I would especially like to thank Dr. Henry Akinbi for his careful supervision and support, especially during the first few years of my thesis project.

Pursuing my Ph.D. would not have been possible without my undergraduate mentor, Dr.

John Stevenson, who introduced me to the exciting world of scientific research.

This thesis is dedicated to my mother, Barbara A. Fritz and my father, Gerald W. Fritz, as well as my grandma, Barbara H. Bridge and my grandpa Edward D. Bridge. They instilled within me their passion, persistence and inquisitive mind. Their unconditional love and support make it possible to achieve any goal.

v TABLE OF CONTENTS

CONTENT PAGE

List of tables and figures xiii

List of abbreviations xvi

CHAPTER I Molecular Chaperones and the UPR in 1

Development and Metabolism: Background,

Significance and Hypothesis

1. Molecular Chaperones in the Endoplasmic Reticulum 2

1a. BiP: an ER-localized chaperone 2

1b. DnaJ family of cochaperones 3

1bi. Structure and classification 5

1bii. Functionality 5

1biii. ER-localized DnaJ proteins 6

1biv. ERdj4 8

2. The Unfolded Protein Response 9

2a. Discovery 9

2b. Initial discovery of UPR signaling components in yeast 10

2c. The mammalian UPR 10

2ci. The ER stress sensors: ATF6, PERK and IRE1 11

2cii. ER stress-induced apoptosis 14

3. β-cell Stress, the UPR and Diabetes 14

3a. Inducers of ER stress in β-cells 15

3ai. Glucose 15

vi 3aii. Free fatty acids 16

3aiii. Proinflammatory cytokines 16

3aiv. Misfolded protein 18

3av. Islet amyloid peptide 18

3b. The UPR in β-cell dysfunction and diabetes 18

3bi. PERK/eIF2α 18

3bii. IRE1α/XBP1 19

3biii. ATF6α 21

3biv. ER chaperones 21

3c. The UPR and insulin resistance 22

4. B Cell Subsets and Effector Functions 22

4a. B1 cells 23

4b. B2 cells: Follicular and marginal zone B cells 24

4bi. Follicular B cells 24

4bii. Marginal zone B cells 24

5. B Cell Development 25

5a. B1 cell development 25

5b. B2 cell development 28

5bi. Early B2 cell differentiation in the bone marrow 28

5bii. Transcriptional regulation of early B2 cell development 30

5biii. Transitional B cells 31

5biv. Follicular verses marginal zone B cell fate 32

5c. The UPR in B cell development 34

vii 6. Growth Factors and Cellular Niches Required for B Cell Development 35

6a. B cell growth factors 36

6ai. CXCL12 36

6aii. IL-7 36

6aiii. FLT3L 38

6aiv. RANKL 38

6av. SCF 39

6b. Bone marrow niches that support B cell development 40

6bi. CAR niche: reticular cells expressing CXCL12 40

6bii. IL-7-expressing cells 40

6biii. Dendritic cells 41

6biv. Osteoblasts 41

7. The UPR in Osteoblast Differentiation 42

7a. PERK/eIF2α 42

7b. ATF4 44

7c. ATF6 45

7d. IRE1α/XBP1 45

8. Summary and Hypothesis 46

References 48

CHAPTER II Deficiency of ERdj4 is associated with perinatal 66

lethality, constitutive ER stress and glucose

intolerance

viii Abstract 68

Introduction 69

Results 72

Generation of ERdj4 gene trap mice 72

Elevated ER stress and increased susceptibility to cell death 72

in ERdj4GT/GT MEFs

Hypoglycemia and growth retardation in ERdj4GT/GT mice 73

Constitutive ER stress is associated with histological 74

abnormalities in pancreatic islets of ERdj4GT/GT mice

ERdj4 deficiency impairs insulin biosynthesis 75

Hypoinsulinemia causes glucose intolerance in ERdj4GT/GT 76

mice

Discussion 77

Materials and Methods 81

Acknowledgements 87

Figure Legends 88

Figures 93

References 105

CHAPTER III Deficiency of the ER stress chaperone ERdj4 is 110

associated with defects in osteogenesis and B cell

development

Abstract 112

ix Introduction 113

Results 117

Bone-lining cells are reduced in ERdj4GT/GT mice 117

ERdj4 deficiency affects early B lymphopoiesis 119

ERdj4 deficiency in the bone marrow microenvironment 119

impairs B cell development

Mature B cell populations are reduced in ERdj4GT/GT mice 120

Plasma cell differentiation and antibody secretion are normal 121

in ERdj4GT/GT mice

Discussion 122

Materials and Methods 126

Acknowledgements 130

Figure Legends 131

Figures 136

References 146

CHAPTER IV Discussion and Future Directions 151

Summary 152

1. Gene trapping ERdj4: a blessing in disguise? 154

2. What is the function of ERdj4 in perinatal survival? 155

3. ER stress and Type 2 Diabetes 157

3a. Do physiological stressors cause diabetes in ERdj4GT/GT mice? 158

3b. What causes β-cell failure in ERdj4GT/GT mice? 159

x 3c. Does rescue of ERdj4 expression in pancreatic β-cells result in 160

normal insulin secretion and glucose metabolism in ERdj4GT/GT mice?

3d. Is the chaperone activity of ERdj4 important in α-cells? 163

3di. Does loss of ERdj4 affect glucagon biosynthesis? 163

3dii. What causes α-cell hyperplasia in ERdj4GT/GT mice? 164

4. The role of ERdj4 in the bone marrow microenvironment 165

4a. Does the loss of ERdj4 in osteogenic cells impair B cell development 165

in ERdj4GT/GT mice?

4ai Do ERdj4GT/GT osteogenic cells support B cell maturation 165

in vitro?

4aii. Does administration of PTH rescue the defect in B cell 166

development in ERdj4GT/GT mice?

4aiii. Would rescue of ERdj4 expression in osteogenic cells result in 167

normal B cell development in ERdj4GT/GT mice?

4b. Which cell(s) of the osteogenic lineage affects B cell development? 168

4c. How does ERdj4 deficiency affect peripheral B cells? 169

4d. How does the loss of ERdj4 affect postosteoblasts and skeletal 170

development?

4di. Quantitation of osteogenic cells from ERdj4GT/GT mice 170

4dii. Why are postosteoblasts reduced in ERdj4GT/GT mice? 171

4diii. Does the loss of postosteoblasts affect skeletal development 171

in ERdj4GT/GT mice?

5. Molecular chaperones in the treatment of ER stress-related disease 172

xi 6. ERdj4 in cellular function and homeostasis 173

References 175

xii LIST OF TABLES AND FIGURES

CHAPER I PAGE

Figure 1 The BiP/HSP70 chaperone cycle 4

Figure 2 The mammalian ER-localized DnaJ proteins 7

Figure 3 The mammalian UPR 12

Figure 4 Mechanisms of ER stress-induced β-cell dysfunction 17

and apoptosis

Table 1 Diabetic phenotypes due to genetic alteration of ER 20

homeostasis

Figure 5 B1 and B2 development occur in distinct, overlapping waves 26

Figure 6 B1 cell development 27

Figure 7 B2 cell development in the bone marrow 29

Figure 8 Marginal and follicular B cell development 33

Table 2 B cell defects in mice that are deficient in microenvironmental 36

factors in the bone marrow

Figure 9 Microenvironmental niches for B cell development in the 37

bone marrow

Figure 10 The UPR in osteogenesis 43

CHAPER II

Figure 1 Generation of ERdj4GT/GT mice 93

Figure 2 Constitutive ER stress and decreased cell viability in 94

ERdj4GT/GT MEFs

xiii Figure 3 Hypoglycemia and growth retardation in ERdj4GT/GT mice 95

Figure 4 Elevated ER stress in pancreatic β-cells of ERdj4GT/GT mice 96

Figure 5 ERdj4 deficiency impairs insulin biosynthesis 97

Figure 6 Hypoinsulinemia causes glucose intolerance in ERdj4GT/GT mice 98

Suppl 1 Elevated ER stress in tissues of ERdj4GT/GT mice 99

Suppl 2 Ultrastructural evidence of ER stress in α-cells of ERdj4GT/GT mice 100

Suppl 3 ER stress in pancreatic β-cells of ERdj4GT/GT mice 101

Suppl 4 ER stress in pancreatic acinar cells of ERdj4GT/GT mice does 102

not impair function

Table 1. Genotype distribution of P21 progeny from heterozygous 103

intercrosses

Table 2. Genotype distribution of day 18.5 embryos from heterozygous 104

intercrosses

CHAPTER III

Figure 1 Bone-lining cells are reduced in ERdj4GT/GT mice 136

Figure 2 Primary osteogenic cells from ERdj4GT/GT mice have an 137

immature phenotype

Figure 3 B cell development is impaired in ERdj4GT/GT mice 138

Figure 4 ERdj4 deficiency in the bone marrow microenvironment 139

affects B cell development

Figure 5 Mature B cells are decreased in the spleen and peritoneal 140

cavity of ERdj4GT/GT mice

xiv Suppl 1 Skeletal development of ERdj4+/+ and ERdj4GT/GT embryos 141

Suppl 2 BMP2-treated ERdj4GT/GT MEFs have reduced osteoblast 142

differentiation potential

Suppl 3 B cells and erythrocytes are decreased in the bone marrow 143

of ERdj4GT/GT mice

Suppl 4 Bone marrow B cell proliferation and peripheral pro-B cells are 144

normal in ERdj4GT/GT mice

Suppl 5 Plasma cell differentiation and antibody production are normal 145

in ERdj4GT/GT mice

xv LIST OF ABBREVIATIONS

ALP Alkaline phosphatase

APRIL A proliferation-inducing ligand

ASK1 Apoptosis-signal-regulator 1

ATF Activating transcription factor

BAFF B cell activating factor

BAFFR B cell activating factor receptor

BCL10 B cell lymphoma 10

BCR B cell receptor

BiP Immunoglobulin binding protein

BMP2 Bone morphogenic protein 2

BTK Bruton’s tyrosine kinase bZIP basic leucine zipper

CHOP C/EBP-homologous protein

CLP Common lymphoid progenitor

CRE Cyclic AMP-responsive element

CTD Carboxy-terminal domain

CXCL12 CXC-chemokine ligand 12

CXCR4 CXC-chemokine receptor 4

DC Dendritic cell

EBF Early B cell factor eIF2α Eukaryotic initiation factor 2 alpha

ER Endoplasmic reticulum

xvi ERAD ER-associated degradation

ERSE ER stress response element

FACS Fluorescence activated cell sorting

FFA Free fatty acid

FLT3 Fms-related tyrosine kinase 3

GFP Green fluorescent protein

GLP Glucagon-like peptide

GRP Glucose-regulated protein

HPD His, Pro, Asp motif

HSC Hematopoietic stem cell

IAPP Islet amyloid polypeptide

IgH Immunoglobulin heavy chain

IgL Immunoglobulin light chain

IKKβ IκB kinase β

IRE1 Inositol-requiring enzyme 1

IRF Interferon regulatory factor

JNK c-Jun N-terminal kinase

MALT1 Mucosa-associated lymphoid tissue lymphoma translocation protein 1

MAML1 Mastermind-like 1

MIDY INS-gene-induced diabetes of youth

MIF1 Migration-inhibitory factor 1

MINT MSX2-interacting protein

MPP Multipotential progenitor

xvii MSC Mesenchymal stem cell

NBD Nucleotide binding domain

NEF Nucleotide exchange factor

Nemo NF-κb essential modulator

NF-κb Nuclear factor-kappa b

NIK NF-κb-inducing kinase

NKT Natural Killer T

NO Nitric oxide

OASIS Old astrocyte specifically induced substance

PAS Para-aortic splanchnopleura

PAX5 Paired box protein 5

PBA 4-phenyl butyric acid

PERK Protein kinase RNA (PKR)-like ER kinase

PI3K Phosphoinositide 3-kinase

PLCγ2 Phospholipase Cγ2

PTH Parathyroid hormone

RAG Recombination activating gene

RANKL Receptor activator of nuclear factor-κB ligand

RBP-Jκ Recombining binding protein suppressor of hairless

ROS Reactive oxygen species

SBD Substrate binding domain

SCF Stem cell factor

SHP Small heterodimer partner

xviii siRNA Small interfering RNA

SLC Surrogate light chain

SNP Single nucleotide pair

T1 Transitional 1

T2 Transitional 2

TI-2 T cell-independent type 2

TRAF2 TNF-receptor-associated factor 2

TRAP Tartrate-resistant acid phosphatase

TUDCA Tauroursodeoxycholic acid

XBP1 X-box-protein 1

UPR Unfolded protein response

UPRE Unfolded protein response element

VCAM1 Vascular adhesion molecule 1

xix

CHAPTER I

Molecular Chaperones and the UPR in Development and Metabolism:

Background, Significance and Hypothesis

1 1. Molecular Chaperones in the Endoplasmic Reticulum

Nearly one third of proteins destined for the cell surface, intracellular organelles, or secretion enter the endoplasmic reticulum (ER) to undergo and post- translational modifications1. Molecular chaperones are crucial components of the ER that direct protein transport across membranes, assist in folding/refolding of nascent proteins, regulate protein-protein interactions, and remove misfolded proteins from the

ER lumen for proteasomal degradation. The first identified ER chaperone, immunoglobulin binding protein (BiP), was discovered nearly three decades ago (refer to section 2a). Since then, BiP has been shown to cooperate with a diverse family of

DnaJ cochaperones, whose importance for ER homeostasis is only beginning to be appreciated.

Our laboratory identified the BiP cochaperone (ERdj4/DNAJB9) as one of the most highly upregulated in response to expression of two disease-associated mutations in surfactant protein C (SP-C) that are prone to misfolding and retention in the

ER lumen. Unlike other DnaJ proteins, ERdj4 preferentially associated with misfolded

SP-C, facilitating its removal from the ER via ER-associated degradation (ERAD)2. The significance of these findings prompted the study of ERdj4 in cellular homeostasis and function in vivo.

1a. BiP: an ER-localized HSP70 chaperone

BiP belongs to the HSP70 family of molecular chaperones, which are highly homologous in structure and function within and across species. HSP70 family members are a ubiquitous class of chaperones present in the cytosol, nucleus,

2 mitochondria and ER. BiP is involved in multiple ER processes, which include regulating the translocon pore3, folding nascent proteins4,5, targeting misfolded proteins for proteasomal degradation6,7, maintaining ER calcium stores8 and activating the unfolded protein response (UPR) under conditions of ER stress9,10. All of these functions, with the exception of maintaining ER calcium stores, require the ATPase activity of BiP, a conserved feature among HSP70 chaperones.

Like other HSP70 proteins, BiP contains an N-terminal nucleotide binding domain

(NBD) and C-terminal substrate binding domain (SBD), which actively communicate via an interdomain linker11. The SBD is in an open configuration when ATP is bound to the nucleotide binding domain (NBD), allowing for transient, low affinity interactions with hydrophobic residues of client proteins. ATP hydrolysis closes the SBD, resulting in tight association with bound substrate. When ADP is exchanged for ATP, the client is released and given the opportunity to fold. Several cycles of client-chaperone binding and release occur until the protein achieves a stable conformation. The ATPase activity of BiP requires cochaperones of the DnaJ family and nucleotide exchange factors

(NEFs) to stimulate client capture and dissociation, respectively (Figure 1). It has been postulated that the multifunctionality of BiP (and other HSP70 family members) is achieved through interaction with various DnaJ proteins12.

1b. DnaJ family of cochaperones

DnaJ proteins were discovered as cofactors for HSP70 in 1978 when the heat shock proteins GrpE and DnaJ were shown to modulate the ATPase activity of DnaK, an

HSP70 family member in Escherichia coli13-16. Since then, DnaJ proteins have emerged

3 Native protein

! DnaJ

Unfolded g SBD ! protein ! !

J domain ! ATP

a !

! BiP ! ADP ATP ATP f SBD ! !

b NBD ATP

ADP

! ! ATP ATP c ! ! Pi e ADP d

! NEF ! ! !

Figure 1. The BiP/HSP70 chaperone cycle. Unfolded protein interacts with the SBD of DnaJ proteins (a) or BiP (b). The J domain of DnaJ proteins associates with the NBD of BiP to stimulate ATPase activity (c), thereby resulting in ATP hydrolysis and closed conformation of the SBD of BiP (d). NEFs associate with BiP (e) to catalyze the exchange of ADP for ATP, which opens the SBD (f) and releases client protein. The client protein either achieves native conformation or reenters the chaperone cycle to continue folding (g). SBD, substrate binding domain; NBD, nucleotide binding domain;

NEF, nucleotide exchange factor.

4 as a structurally and functionally diverse family of cochaperones that play an essential role in many cellular processes.

1bi. Structure and classification

DnaJ proteins have been loosely categorized into three classes based on the presence or absence of structural domains. Class I members contain an N-terminal J domain, a glycine phenylalanine (Gly/Phe)-rich region, two zinc finger motifs and two carboxy- terminal domains (CTDI and CTDII). Class II members have a J domain and Gly/Phe- rich region, but do not possess zinc finger motifs, while class III members contain only a

J domain and do not fit the requirements for class I or class II17.

All DnaJ proteins encode the highly conserved J domain that contains a His, Pro,

Asp (HPD) motif necessary for the stimulation of the ATPase activity of BiP18,19. This interaction is also dependent on a positively charged helix located in the J domain that associates with a negatively charged loop on the NBD of BiP20. The functionality of the

Gly/Phe-rich region remains unclear, while the zinc finger motifs have been shown to be important for client binding and the ATPase activity of BiP21,22. The CTD domains have low sequence identity among DnaJ proteins, but are structurally similar, often by forming a hydrophobic pocket for association with client proteins23.

1bii. Functionality

The structural diversity of DnaJ proteins enables participation in a variety of cellular functions with or without binding to client proteins. Often their subcellular localization and J domain are sufficient; DnaJ proteins are positioned at ribosomes and translocons

5 of mitochondrial and ER membranes to recruit BiP and stimulate ATPase activity, which results in protein folding and/or polypeptide import. In addition to serving as a cochaperone to BiP, DnaJ proteins can interact with the client itself for many cellular processes, including transport across the ER membrane, folding of nascent proteins, remodeling of multimeric protein complexes and degrading unfolded/misfolded proteins12.

1biii. ER-localized DnaJ proteins

Yeast contain three ER-localized DnaJ proteins: Sec63p, Jem1p, and Scj1p. Sec63p is anchored in the ER lumen as a component of the translocon and functions to recruit BiP and assist in the translocation of nascent proteins18. Jem1p and Scj1p are luminal ER proteins upregulated under conditions of ER stress24. Yeast mutants deficient in both

Jem1p and Scj1p exhibited growth arrest, but deletion of either Jem1p or Scj1p individually did not affect cell growth25. Under conditions of ER stress, Scj1p interacts with BiP to facilitate protein folding26. Jem1p and Scj1p also cooperate with BiP to retrotranslocate misfolded proteins from the ER for degradation7,27.

To date, seven mammalian ER-localized DnaJ proteins have been identified and are ubiquitously expressed in vertebrate tissues28 (Figure 2). ERdj1 and ERdj2 are transmembrane proteins positioned at the translocon. ERdj1 recruits BiP to translating ribosomes, but also inhibits protein translation in the absence of BiP29-31. The function of ERdj2 at the translocon remains unclear; ERdj2 does not associate with translocation machinery in the presence of ribosomes32. ERdj3 and ERdj6 are soluble ER luminal proteins that bind unfolded/misfolded substrates and recruit BiP to assist in protein

6 Function UPR induction recruits BiP to ERdj1 No the translocon

ERdj2 unknown No

ERdj3 protein folding Yes

ERdj4 ERAD Yes

ERdj5 ERAD Yes

ERdj6 protein folding Yes

ERdj7 unknown No

J domain Gly/Phe region ER Thioredoxin box Dimerization domain SANT domain Carboxy-terminal domain Sec63 domain Transmembrane domain

Figure 2. The mammalian ER-localized DnaJ proteins. Seven DnaJ proteins have been identified in the ER, playing an important role in transport, protein folding and degradation. The structural diversity among ERdj proteins likely drives diverse BiP- dependent and independent cellular processes. All members contain the highly conserved J domain; ERdj3 and ERdj4 are classified as class I and class II DnaJ proteins, respectively, while the remaining ERdjs are classified as class III members.

7 folding33,34. ERdj4 and ERdj5 are also soluble luminal proteins, but associate with

ERAD machinery to facilitate the removal of unfolded/misfolded proteins from the ER2,35-

37. ERdj7 is a transmembrane protein with unknown function, but reducing expression of ERdj7 did not induce ER stress, suggesting that it plays a minor role in protein folding38,39.

1biv. ERdj4

Hendershot et al. identified ERdj4 as a class II DnaJ family member through bioinformatic methods36. ERdj4 is a soluble ER luminal protein containing the signature

J domain, a Gly/Phe-rich region, and a C-terminal substrate binding domain36 (Figure

2). The HPD motif in the J domain of ERdj4 associates with BiP, thereby assisting BiP in achieving stable conformation with client proteins by stimulating ATP hydrolysis.

ERdj4 also interacts directly with unfolded (insulin2WT) and misfolded substrates

(insulin2C96Y, SP-CΔexon4), and reducing levels of ERdj4 slowed degradation of these substrates in vitro2,36. Further, ERdj4 associates with Derlin-1 and p97, components of

ERAD machinery2,35, to facilitate removal of clients for degradation by the cytosolic proteasome. The mechanism by which ERdj4 discriminates between unfolded and terminally misfolded substrates destined for ERAD remains unclear.

ERdj4 is ubiquitously expressed at low levels, but highly upregulated under conditions of ER stress2,36 by the UPR signal transducer X-box-protein 1 (XBP1)40,41.

The in vivo function of ERdj4 is entirely unknown, but given that ERdj4 mRNA is detected in secretory tissues28,36, it may play an important role in maintaining ER homeostasis in secretory cell types.

8 2. The Unfolded Protein Response

The UPR is an intracellular signaling pathway that is activated when protein load exceeds protein folding capacity in the ER lumen – a condition referred to as ER stress.

ER stress can result from pathological conditions that cause proteins to misfold, including genetic mutations, toxic compounds and oxidative stress. Physiologic stimuli such as nutrients, cytokines and hormones, can also cause ER stress by increasing the demand for protein synthesis. The UPR restores ER homeostasis by increasing ER size, attenuating protein translation and upregulating protein folding and degradation machinery.

2a. Discovery

The discovery of the UPR dates to 1974 when two 78 and 94 kDa proteins were induced in fibroblasts upon transformation with avian sarcoma virus42. These proteins were later designated as the glucose-regulated proteins GRP78 and GRP94, because their upregulation in tumor cells occurred in response to glucose deprivation in the media rather than an effect of cellular transformation43. Several other inducers of

GRP78 and GRP94 were identified including inhibitors of glycosylation, calcium ionophores and amino-acid analogues44. In 1983, Hass et al. independently discovered

GRP78 as an ER-localized protein that interacted with immunoglobulin heavy chain intermediates to facilitate assembly, thereby identifying the first ER chaperone termed

BiP, or immunoglobulin heavy chain binding protein45. The ‘unfolded protein response’ was finally coined in the early 1990’s by Gething and Sambrook46 after demonstrating that accumulation of malfolded influenza hemagglutinin protein in the ER efficiently induced GRP78/BiP and GRP9447. Conserved regulatory domains among GRPs

9 provided further evidence for a signaling pathway induced by ER stress47. However, despite identification of several UPR inducers, the molecular pathway(s) underlying the upregulation of ER chaperones was entirely unknown.

2b. Initial discovery of UPR signaling components in yeast

In Saccharomyces cerevisiae, BiP is encoded by the KAR2 gene, which contains an unfolded protein response element (UPRE) that induces BiP mRNA expression in response to accumulation of unfolded proteins48. Using the UPRE to select yeast mutants with defects in the UPR signaling pathway led to discovery of the first UPR signaling component, Ire1p49,50. Ire1p is a transmembrane receptor kinase, composed of an N-terminal domain located in the ER lumen that senses unfolded protein and a C- terminal protein kinase domain in the cytosol that activates transcription of KAR2 and other UPRE-containing genes. A basic leucine zipper (bZIP) transcription factor Hac1p was then identified as a downstream target of Ire1p by isolating genes that activated the

UPR when overexpressed in Ire1p-deleted yeast mutants51. Upon activation of the

UPR, cytosolic Hac1p mRNA is spliced51 by the endoribonuclease activity of Ire1p and re-ligated by Rlg1p, a tRNA ligase52. Spliced Hac1p mRNA is then translated and subsequently activates transcription of ER-resident genes including KAR2, PDI1a and

SCJ153, via direct binding to the UPRE51. Activation of the Ire1p/Rlg1p/Hac1p signaling pathway induces over 5% of the yeast genome, highlighting its importance in cellular function and homeostasis54.

2c. The mammalian UPR

10 2ci. The ER stress sensors: ATF6, PERK and IRE1

In contrast to yeast, which contains a single UPR signaling pathway

(Ire1p/Rlg1p/Hac1p), mammalian cells express three known UPR signal transducers:

ATF6, PERK and IRE1. These components were identified by Kaufman, Mori and Ron in the late 1990’s, laying the foundation for understanding how the mammalian UPR signaling pathway senses and responds to ER stress in order to maintain cellular homeostasis.

ATF6 is a type II transmembrane protein that contains an ER stress-sensing domain in the ER lumen. Following activation, ATF6 shuttles to the Golgi complex where it is cleaved by S1P and S2P proteases resulting in release of a cytosolic ATF6 fragment that contains a bZIP transcription factor55,56. The cytosolic ATF6 fragment then translocates to the nucleus to activate transcription of ER chaperones and components of ERAD by interacting with ER stress response elements (ERSE)57-59.

Mice ubiquitously express two isoforms of ATF6, ATF6α and ATFβ. Mice deficient in either isoform of ATF6 developed normally, while mice deficient in both isoforms were embryonic lethal, suggesting that ATF6α and ATFβ contain an overlapping function that is crucial for development58,60 (Figure 3).

The second ER stress sensor PERK is localized in the ER as a type I transmembrane protein with a luminal stress-sensing domain and a cytosolic protein kinase domain. In response to ER stress, oligomerization and trans- autophosphorylation of PERK results in phosphorylation of eukaryotic initiation factor 2

(eIF2α). eIF2α phosphorylation reduces protein load in the ER lumen by inhibiting the translation-initiation complex61. Further, phosphorylation of eIF2α upregulates the

11

ER lumen protein BiP

P IRE1 P P ! PERK P ATF6

P eIF2! ATF6 uXBP-1 cleavage ATF4 Golgi

Nutrient uptake, metabolism and apoptosis sXBP-1

Protein folding and degradation

Protein folding and degradation

Nucleus

Figure 3. The mammalian UPR. Unfolded/misfolded protein is detected by BiP in the

ER lumen; disassociation of BiP from the ER stress sensors IRE1α, ATF6 and PERK activates the UPR signaling cascade, ultimately resulting in an increase in ER size, attenuation of protein translation and upregulation of ER chaperones and ERAD machinery.

12 transcription factor ATF4, which induces genes involved in amino acid metabolism, redox status and apoptosis62. Approximately half of PERK-deficient mice died perinatally; surviving mice had postnatal growth restriction with defects in endocrine and exocrine pancreatic function and skeletal development63,64 (Figure 3).

Structurally similar to PERK, the third ER stress sensor IRE1 is also a type I transmembrane protein that contains a stress-sensing domain in the ER lumen and a protein kinase domain in the cytosol. IRE1 functions uniquely in the mammalian UPR by encoding a C-terminal endoribonuclease domain (similar to yeast Ire1p). Upon activation, IRE1 oligomerizes, trans-autophosphorylates and excises a 26 intron in the mRNA of XBP1 (the mammalian homolog of Hac1p). Spliced XBP1 protein contains a C-terminal transactivation domain that interacts with UPREs of ER chaperones, ERAD machinery and genes involved in lipid biosynthesis65,66. The protein kinase activity of activated IRE1 also associates with TNF-receptor-associated factor 2

(TRAF2), which promotes apoptosis through JNK signaling67 (Figure 3).

There are two isoforms of IRE1: IRE1α and IRE1β. Although functionally identical, IRE1α is expressed ubiquitously, while IRE1β is predominantly expressed in the intestinal epithelium. IRE1α deficiency in mice resulted in embryonic lethality at

E10.5 due to defects in placental development68,69. XBP1-deficient mice also died during embryonic development at E12.5 due to liver hypoplasia70. When XBP1 was selectively expressed in the liver of XBP1-deficient mice to rescue embryonic lethality, postnatal lethality occurred within a few days of birth due to impaired production of pancreatic digestive enzymes71.

13 2cii. ER stress-induced apoptosis

During conditions of ER stress, the UPR restores cellular homeostasis by increasing the size of the ER (i.e. increased membrane synthesis), attenuating protein translation and upregulating protein folding/degradation machinery. Failure to resolve ER stress results in UPR-mediated activation of cell death signaling pathways. Phosphorylation of eIF2α by PERK induces transcription of the cell death mediator C/EBP-homologous protein

(CHOP)72, which downregulates the anti-apoptotic molecule BCL-273. IRE1α associates with TRAF2, ultimately resulting in activation of the proapoptotic molecule apoptosis- signal-regulator (ASK1) through c-Jun N-terminal kinase (JNK)67,74. IRE1α signaling through TRAF2 also activates caspase-12, which executes cell death under conditions of ER stress75.

3. β-cell Stress, the UPR and Diabetes

Pancreatic β-cells are highly secretory, insulin-producing cells, which frequently encounter physiological conditions that perturb ER homeostasis. Insulin biosynthesis occurs acutely at the translational level and maturation of proinsulin and insulin processing enzymes is a critical process that occurs within the ER76. Glucose can stimulate a 25-fold increase in insulin biosynthesis77,78, imposing a heavy load of unfolded/misfolded protein on the ER, leading to activation of stress pathways. The

UPR reduces ER burden of proinsulin biosynthesis by attenuating protein translation and increasing the expression of ER chaperones and ERAD machinery to assist in protein folding and degradation. However, excess nutrients (metabolic stress) and/or

14 insulin resistance induces chronic ER stress and hyperactivation of the UPR leading to

β-cell dysfunction and apoptosis, a hallmark of type 2 diabetes.

3a. Inducers of ER stress in β-cells

3ai. Glucose

Insulin resistance in type 2 diabetes is associated with chronic elevation of blood glucose levels, which compromises β-cell function and survival, a phenomenon known as glucotoxicity. Hyperglycemia results in an increase in reactive oxygen species

(ROS), which is associated with β-cell pathology79. High levels of glucose generate

ROS through several mechanisms, including glyceraldehyde autooxidation, glucosamine and hexosamine metabolism, sorbitol formation and oxidative phosphorylation80. Oxidative stress in β-cells is associated with a decrease in the insulin regulatory proteins, PDX-1 and MafA, resulting in reduced insulin gene expression, as well as impaired β-cell proliferation and survival81,82. Antioxidant treatment in rodent models of type 2 diabetes reduced circulating ROS and attenuated hyperglycemia79. Glucotoxicity is also associated with ER stress and UPR activation, which allows for increased proinsulin biosynthesis. However, upregulation of folding enzymes and chaperones by the UPR signaling pathway also contributes to ROS production and β-cell demise by increasing the oxidative protein folding machinery that drives disulfide bond formation83. Further, ROS production can inhibit protein folding by modifying substrates directly or inactivating chaperones. Since β-cells express low levels of antioxidant enzymes, proteins within the ER are particularly vulnerable to oxidation84 (Figure 4).

15 3aii. Free fatty acids

High fat diets and obesity are associated with increased levels of plasma free fatty acids

(FFAs)85. The saturated FFA palmitate causes ER stress and β-cell dysfunction by (1) reducing protein trafficking from the ER to the Golgi; (2) depleting ER calcium stores; (3) lowering fatty acid oxidation and triglyceride synthesis and (4) activating PKC-δ-JNK- mediated apoptosis86,87. Several studies have demonstrated that saturated FFAs activate the UPR, although the underlying mechanisms remain unclear. Palmitate activates the PERK/eIF2α signaling axis, resulting in upregulation of ATF4 and CHOP88, which mediates apoptosis under conditions of ER stress. There are discrepancies in the literature on whether palmitate activates the IRE1α/XBP1 and ATF6α signaling pathways89,90. However, Laybutt et al. reported activation of several XBP1-dependent genes in palmitate-treated insulinoma cells, including Edem1, Herpud1 and ERdj4, which assist in protein folding/degradation to reduce ER stress90-92 (Figure 4).

3aiii. Proinflammatory cytokines

Obesity is associated with chronic, low grade , due to proinflammatory cytokine production by adipocytes and leukocytes in adipose tissue93. Obesity causes an increase in circulating levels of FFAs, which can promote proinflammatory cytokine production by inducing the nuclear factor-κb (NF-κB) signaling pathway94.

Proinflammatory cytokines, including IL-1β, TNFα and IFNγ, stimulate the production of nitric oxide (NO) in pancreatic β-cells, which depletes intracellular calcium stores and causes ER stress95. Cytokine-induced NO production also activates the PERK/eIF2α signaling pathway, thereby inducing CHOP-mediated β-cell death96.

16

Figure 4. Mechanisms of ER stress-induced β-cell dysfunction and apoptosis.

Chronic glucose stimulation induces protein biosynthesis, which increases oxidative protein folding (generating ROS) and accumulation of unfolded/misfolded protein in the

ER. ROS production inhibits mobilization of calcium, ultimately resulting in mitochondrial-induced apoptosis. Hyperactivation of the UPR signaling cascade by either excess lipids or glucose results in degradation of proinsulin mRNA and ER stress- induced apoptosis (Modified from Back and Kaufman, Annual Review of Biochemistry,

2012).

17 3aiv. Misfolded protein

Mutations in the insulin gene that affect protein folding cause ER stress, β-cell failure and diabetes. The Akita and Munich diabetic mouse models have mutations in critical cysteine residues (C96Y and C95S) that are necessary for disulfide bond formation and proper folding of proinsulin97,98. Interestingly, 26 mutations in the human insulin gene are associated with autosomal dominant diabetes and are referred to as mutant INS- gene-induced diabetes of youth (MIDY). Similar to the mutations in mouse insulin,

MIDY mutations (one of which is C96Y) impair proinsulin folding in the ER often by disrupting disulfide bond formation. These insulin mutations in mice and humans resulted in proinsulin accumulation and unresolvable ER stress, leading to β-cell failure and apoptosis99 (Figure 4).

3av. Islet amyloid polypeptide

Deposition of islet amyloid polypeptide (IAPP) in pancreatic islets is strongly associated with the of type 2 diabetes in humans. IAPP is a hormone that is normally co- secreted with insulin to regulate digestion. Human IAPP can form toxic amyloid deposits, which are associated with increased β-cell apoptosis through the activation of

CHOP and caspase-12100. Multiple studies have reported that human IAPP activated the UPR100,101; however, these findings are controversial as one study found that physiological levels of human IAPP did not induce ER stress102 (Figure 4).

3b. The UPR in β-cell dysfunction and diabetes

3bi. PERK/eIF2α

18 Genetic mutations in several components of the UPR are linked to diabetes in humans and mice103 (Table 1), suggesting that the UPR is a necessary component for β-cell development, homeostasis and survival. Humans with Wolcott-Rallison syndrome have an inactivating mutation in PERK that results in neonatal diabetes due to pancreatic hypoplasia and β-cell apoptosis104,105. A similar phenotype was observed in PERK- deficient mice; approximately 50% of PERK-deficient mice died neonatally, but surviving mice developed severe hyperglycemia within four weeks of birth due to impaired β-cell development63,64. PERK/eIF2α signaling was shown to be necessary for β-cell differentiation and proliferation during the fetal period in order to prevent postnatal diabetes. Further, conditional deletion of PERK after the neonatal stage did not affect

β-cell development or glucose homeostasis106. Mice harboring a homozygous mutation in the phosphorylation site of eIF2α (Ser51Ala; A/A mutants), the downstream effector of PERK signaling, die neonatally due to severe hypoglycemia associated with reduced glycogen storage, impaired gluconeogenesis and pancreatic β-cell deficiency107. The phenotype of PERK-deficient mice and eIF2α A/A mutants are different, suggesting that kinases other than PERK are important for eIF2α signal transduction.

3bii. IRE1α/XBP1

Several studies have indicated that the IRE1α/XBP1 signaling pathway is important for proinsulin biosynthesis and β-cell homeostasis. Under high glucose conditions for short periods of time (≤ 1 hour), IRE1α activation stimulated proinsulin biosynthesis. In contrast, prolonged elevation of glucose (≥ 24 hours) resulted in ER stress and IRE1α hyperactivation that suppressed proinsulin biosynthesis and upregulated CHOP, an

19 Table 1. Diabetes phenotypes due to genetic alteration of ER homeostasis Gene Mutation Genotype Species Effect Phenotype Client protein Ins Missense Het Human Misfolded proinsulin Neonatal diabetes Ins2 Akita/Munich Het Mouse Misfolded proinsulin Young adult onset diabetes Iapp hIAPP Transgene Mouse Aggregated hIAPP Young adult onset diabetes UPR signaling Perk Null Hom Mouse Deletion Young adult onset diabetes Perk Missense/deletion Hom Human Loss function Wolcott-Rallison syndrome Perk SNPs Polymorphism Human ? Type 1 diabetes eIF2! Ser51Ala Hom Mouse No phos. !-cell deficiency eIF2! Ser51Ala Het Mouse Reduced phos. High fat diet-induced diabetes Ire1! Null Hom Mouse Deletion Embryonic lethal, E10.5 Xbp1 Null Hom Mouse Deletion Embryonic lethal, E12.5 Xbp1 Null Het Mouse Deletion High fat diet-ins. resistance Xbp1 Null LivXBP1 transgene Mouse Xbp1 in liver only Neo. exocrine pancreas defects Xbp1 Conditional Xbp1 f/f;RIP-cre Mouse Xbp1 -/- !-cells !-cell deficiency and diabetes Atf6! Null Hom Mouse Deletion Normal Atf6" Null Hom Mouse Deletion Normal Atf6!/" Null Dual hom Mouse Deletion Embryonic lethal, E8 Atf6 SNPs Polymorphism Human ? Type 2 diabetes ER resident/chaperone Grp78 Null Het Mouse Deletion Attenuates HFD-induced obesity ERdj6/P58IPK Null Hom Mouse Deletion Adult diabetes Wfs1 Null Hom Mouse Deletion Adult diabetes Wfs1 Missense Hom Human Loss function Wolfram syndrome Het, Heterozygote; Hom, homozygote; Phos, phosphorylation; ?, unknown effect; ins, insulin; neo, neonatal Modifed from Scheuner and Kaufman, Endocrine Reviews, 2008 inducer of apoptosis108,109. Further, the endoribonuclease activity of IRE1α was capable of degrading insulin, insulin processing enzymes and ER chaperone mRNAs108,110.

Given that global deficiency of IRE1α or XBP1 was embryonic lethal in mice69,111, it has been difficult to assess the role of these signal transducers in vivo. Transgenic mice that selectively expressed XBP1 in the liver (XBP1-/-LivXBP1) to rescue embryonic lethality had normal islet structure and morphology at birth, but died shortly thereafter due to impaired production of pancreatic digestive enzymes71. Selective deletion of

XBP1 in β-cells of adult mice resulted in hyperglycemia associated with decreased production of proinsulin and β-cell deficiency. Interestingly, XBP1 deficiency in β-cells also resulted in hyperactivation of IRE1α, which negatively affected insulin production by stimulating degradation of insulin processing enzymes, ER chaperones and insulin itself110.

20 3biii. ATF6α

The role of ATF6α in β-cell homeostasis remains unclear. Two separate cohort studies had shown that single nucleotide pair (SNP) mutations in human ATF6α were linked to glucose intolerance and type 2 diabetes112,113. However, no β-cell dysfunction or diabetic phenotype has been described in ATF6α-deficient mice58,60. Interestingly, glucotoxicity upregulated ATF6α (as well as spliced XBP1, phosphorylated eIF2α and

CHOP), resulting in increased expression of a small heterodimer partner (SHP) that suppressed insulin gene expression114,115. Further, increased levels of activated ATF6α in β-cells caused apoptosis115. These data suggest that, under conditions of ER stress

(e.g. glucotoxicity), upregulation of the ATF6α signaling pathway contributes to β-cell dysfuncton and diabetes.

3biv. ER chaperones

ER chaperones also play an important role in maintaining β-cell homeostasis and function. Humans with a loss of function mutation in Wfs1, an ER-localized chaperone, developed Wolfram syndrome, a disease characterized by juvenile diabetes and neurological disorders. Wfs1 negatively regulates UPR signaling by facilitating the removal of ATF6α from the ER for proteasomal degradation116. ATF6α accumulated in the pancreas of Wfs1-deficient mice, which likely contributed to β-cell pathogenesis.

Wfs1-deficient mice were hyperglycemic, exhibiting increased ER stress and β-cell apoptosis117.

Mice deficient in another ER-resident protein, ERdj6/p58IPK, were also hyperglycemic due to increased β-cell apoptosis118. ERdj6/p58IPK has multiple functions

21 in the ER, including protein folding and negatively regulating eIF2α kinase. Since deficiency of ERdj6/p58IPK worsened the diabetic phenotype in Akita mice, which expressed mutant proinsulin119, it is likely that this chaperone plays an important role in proinsulin folding.

3c. The UPR and insulin resistance

Several UPR signal transducers have been associated with ER stress and insulin resistance in peripheral tissues. PERK, eIF2α and BiP were increased in liver and adipose tissue of either high-fat diet (HFD)-induced or genetic (ob/ob) mouse models of obesity. Increased ER stress was associated with impaired insulin signaling, which was mediated through the IRE1α-JNK signaling cascade. XBP1 heterozygous mice carrying a null mutation in one allele were both hyperglycemic and hyperinsulinemic after exposure to HFD, consistent with impaired insulin signaling in peripheral tissues120.

Interestingly, mice with a heterozygous null mutation in GRP78/BiP were resistant to

HFD-induced obesity due to increased energy expenditure and glucose metabolism121.

In summary, several physiological and pathological stimuli activate the UPR in order to alleviate the ER burden associated with insulin biosynthesis. However, hyperactivation of the UPR is linked to β-cell dysfunction and apoptosis. Disruption of the UPR causes diabetes in mice and humans due to defects in β-cell proliferation or survival, highlighting the importance of the UPR in β-cell homeostasis and function.

4. B cell Subsets and Effector Functions

22 B cells are a crucial component of the immune system, possessing both innate and adaptive properties. B cells present antigens to T cells and NK cells, as well as secrete cytokines and other soluble factors to modulate immune responses. During an adaptive immune response, B cells undergo clonal expansion to generate high affinity, antigen- specific antibodies of unique istoypes. B cells can be classified into B1 and B2 lineages, which are distinguished by anatomical location, cell surface phenotype and effector function.

4a. B1 cells

B1 cells make up approximately 5% of total B cells in mice and reside mainly in the pleural and peritoneal cavities, as well as the intestinal lamina propria. In the pleural and peritoneal cavities, B1 cells are classified as CD11b+IgMhiIgDlow and can be further divided into B1a (CD5+) and B1b (CD5-) subsets122,123. The B cell receptor (BCR) of B1 cells is more restricted due to reduced expression of terminal deoxynucleotidyl transferase (TdT), an enzyme that inserts nucleotides into D-J and V(DJ) regions of immunoglobulin heavy (IgH) and light (IgL) chains124. Upon activation, B1a cells migrate to the spleen and downregulate CD5 before differentiating into plasma cells125.

B1 cells are effectors of innate immunity, responding predominantly to TI-2 antigensa.

Haas et al. eloquently demonstrated the functional differences between B1a and B1b cells; after infection with Streptococcus pneumoniae, B1a cells rapidly secreted natural

IgM antibodies for early protection, while B1b cells generated antigen-specific antibodies necessary for pathogen clearance and long-term immunity126.

a T cell-independent type 2 (TI-2) antigens: polysaccharide antigens that engage the BCR to generate antigen-specific antibody responses

23 4b. B2 cells: Follicular and marginal zone B cells

B2 cells are subdivided into follicular and marginal zone B cells, which develop from transitional B cells and differ significantly in effector function. Both express the B cell lineage markers CD19 and B220, but otherwise are defined as IgMlowIgDhiCD21intCD23hi and IgMhiIgDlowCD21hiCD23low, respectively.

4bi. Follicular B cells

Follicular B cells continuously circulate through the follicles of the spleen and lymph nodes, responding to T cell-dependent and T cell-independent antigens. T cell- dependent antigens induce germinal center reactions in the follicles, where B cells rapidly proliferate while undergoing affinity maturation and class switch recombination of immunoglobulins. As a result, plasma cells and memory B cells exit the germinal center expressing high-affinity BCRs and immunoglobulins of different isotypes127.

4bii. Marginal zone B cells

Marginal zone B cells reside in the white pulp of the spleen adjacent to the marginal sinus, enabling quick responses to blood-borne pathogens in the absence of T cells.

They represent only 5-10% of total B cells in the spleen and differ from follicular B cells in their low threshold for activation, proliferation and differentiation to antigen. Similar to

B1 cells, marginal zone B cells respond to TI-2 antigens to differentiate into short-lived plasma cells that secrete IgM128. Marginal zone B cells can present protein antigens through MHC class II molecules to activate CD4+ T cells129 and lipid antigens through

CD1d (an MHC class I-like molecule) to natural killer T (NKT) cells130. Mice deficient in

24 marginal zone B cells had an increased susceptibility to bacterial infections, implicating their importance in innate and adaptive immunity131.

5. B Cell Development

B1 and B2 cell development occurs in sequential, overlapping waves beginning early in mouse embryogenesis (Figure 5). B1 cell progenitors can be generated from the yolk sac and para-aortic splanchnopleura (PAS) at day 9.5 of gestation before emergence of hematopoietic stem cells (HSCs); thus, the precursors that give rise to B1 cell progenitors in the yolk sac and PAS are unknown. Later in gestation, HSCs give rise to both B1 and B2 cells in the fetal liver132,133, but B1 cell development predominates.

Once HSCs begin to seed the bone marrow by day 15 of gestation, it becomes the primary location for B2 cell development throughout postnatal life122,134.

5a. B1 cell development

B1 cell development occurs predominantly during embryogenesis in the fetal liver. A B1 cell progenitor was recently identified as Lin-AA4.1+CD45-/lowCD19+, which was detected earliest in the yolk sac, peaked in number in the fetal liver and then declined postnatally135,136. B1 cell progenitors proliferated in response to thymic stromal lymphopoietin (TSLP) and differentiated only into B1a and B1b subsets136 (Figure 6). B cell progenitors in adult bone marrow that are CD19+B220- also gave rise to B1b cells, but not B2 cells136.

During the neonatal period, transitional B cells in the spleen primarily generate

B1 cells. Unlike transitional B2 cells (section 5biii), transitional B1 cells do not require B

25

Figure 5. B1 and B2 development occur in distinct, overlapping waves. The first developmental wave (1) initiates in the YS and the PSp before emergence of HSCs. It is unclear (dashed line) whether B1 progenitors in the YS and PSp develop into mature adult B1 cells. The second wave of development (2) begins in the fetal liver and fetal bone marrow where HSCs predominantly generate B1 progenitors and subsequently, mature B1 cells. Whether B1 and B2 cells develop from a single HSC or multiple stem cells remains unknown. Some B-ALLs, which are a B1 cell malignancy, may arise in utero during peak B1 cell development. The third developmental wave (3) occurs in adult bone marrow and results primarily in the production of B2 cells. YS, yolk sac; Psp, para-aortic splanchnopleura; HSCs, hematopoietic stem cells; CLP, common lymphoid progenitor; B-ALL, B cell acute lymphocytic leukemia134.

26 Fetal liver Neonatal Spleen Serous cavities

B1a IgM+ Transitional Natural IgM antibody strong BCR and production in response to T NF-!b signaling cell-independent antigens IgMhigh IgDlow AA4.1+ CD11b+ IgM+ CD5+ TSLP CD21int CD23-/+ B1b

Generate antigen-specific antibodies for long-term B1 immunity

progenitor high - IgM Lin low + IgD AA4.1 + -/low CD11b B220 CD5- CD19+

Figure 6. B1 cell development. B1 progenitors in the fetal liver respond to TSLP to differentiate into IgM+ B1 cells that are later detected in the neonatal spleen as transitional B1 cells. Transitional B1 cells are dependent on strong BCR engagement and classical NF-κb signaling in order to develop into B1a or B1b subsets, which home to the peritoneal and pleural cavities.

27 cell activating factor (BAFF) for survival137, but rather strong BCR engagement and the classical NF-κb signaling pathway136,138 (Figure 6).

5b. B2 cell development

5bi. Early B2 cell differentiation in the bone marrow

A seminal paper by Hardy et al. in the early 1990’s defined the stages of B2 cell development in adult mouse bone marrow based on the following: (1) cell surface phenotype, (2) differences in functional activity and (3) immunoglobulin gene rearrangement status139. Based on these criteria, Hardy described six different stages of B cell development in adult mouse bone marrow, referred to as fraction A through fraction F (Figure 7). Immediately prior to B lineage commitment, common lymphoid progenitors (CLPs) have the ability to differentiate into B cells, T cells and NK cells, but cannot differentiate into myeloid or erythroid lineages140. These cells lack lineage specific surface markers (B220, CD11b, Gr1 and Ter119), but express IL-7Rα and c-kit.

Pre-pro B cells (fraction A) are the earliest defined stage of B cell development based on the expression of the B lineage marker B220 (but not CD19) and the absence of immunoglobulin gene rearrangement. Since NK and T cells also express B220, the pre- pro B cells are distinguished from these cell types by the cell surface marker AA4.1, which is expressed on the earliest B cell progenitors through the immature stage of development. The pre-pro B cells then differentiate into the mitotically active pro-B cells

(fractions B/C), which undergo DJ or (V)DJ IgH rearrangements by upregulating expression of recombination activating genes (RAG). Progression from pro-B cells

(fraction B/C) to pre-B cells (fraction D) is dependent on a fully functional pre-B cell

28 Large Small Pre-Pro B Pro-B Pro-B Pre-B Pre-B Immature Mature

pre-BCR IgM IgM

A B C C’ D E F IgD

Figure 7. B2 cell development in the bone marrow. Fraction A through fraction F are defined based on cell surface phenotype, differences in functional activity and immunoglobulin gene rearrangement status. RAG, recombination activating gene; IgH, immunoglobulin heavy chain; IgL, immunoglobulin light chain.

29 receptor (pre-BCR). The pre-BCR complex consists of the rearranged IgH and a surrogate light chain (SLC), which is comprised of λ5 and V-preB, as well a non- covalent Igα and Igβ heterodimer. Signaling through the pre-BCR is a critical checkpoint during B cell development; pre-B cells expressing rearranged IgH that form poor BCRs are eliminated, while those that signal effectively temporarily downregulate

RAG and undergo rapid proliferation. Mice deficient in genes that are crucial for IgH gene rearrangement (RAG1, RAG2, scid) or components of the pre-BCR signaling complex (λ5, Igβ) have a block in B cell development at the pro-B cell stage (Figure 7).

After proliferation, resting pre-B cells (fraction D) increase RAG expression to undergo

IgL gene rearrangement and differentiate into immature B cells (fraction E), which express IgM on the cell surface. The transition from immature (fraction E) to mature

(fraction F) B cells is another critical checkpoint where the B cell undergoes selection with self-antigen. Crosslinking the BCR with antigen results in elimination, anergy, or receptor editing of autoreactive clones. Receptor editing involves an additional round(s) of IgL gene rearrangement to eliminate self-reactivity. The degree to which the BCR binds antigen ultimately determines B cell fate; high affinity binding to self-antigen results in elimination, whereas low affinity or no interaction with self-antigen typically results in survival, anergy, or receptor editing (Figure 7).

5bii. Transcriptional regulation of early B2 cell development

Several transcription factors are important for B2 cell development by influencing lineage commitment, immunoglobulin gene rearrangement and induction of signaling receptors141. The transcription factors PU.1 and Ikaros are essential for the

30 differentiation of CLPs. Low levels of PU.1 drive B lineage commitment and regulate the expression of fms-related tyrosine kinase 3 (FLT3), IL-7Rα and c-kit, key signaling receptors necessary for B cell development. Ikaros directly regulates TdT and λ5, genes essential for immunoglobulin gene rearrangement and pre-BCR signaling, respectively. The transcription factors E2A, early B cell factor (EBF), interferon regulatory factor 4 (IRF4), IRF8 and paired box protein 5 (PAX5) are necessary for immunoglobulin gene rearrangement and pre-BCR signaling. E2A induces recombination at the IgH locus through chromatin remodeling, while EBF influences the expression of Igα, Igβ and VpreB genes, components of the pre-BCR complex. IRF4 and IRF8 bind directly to the IgL locus to induce DNA gene rearrangement142. PAX5 is crucial for V-DJ IgH gene rearrangement and also regulates B cell-specific genes including CD19 and Igα. Expression of PAX5 is also necessary for B lineage commitment; deletion of PAX5 resulted in the ability of pro-B cells to differentiate into osteoclasts, macrophages, dendritic cells (DCs) and NK cells143.

5biii. Transitional B cells

Immature B cells leave the bone marrow and migrate to the spleen where they go through transitional stages (T1 and T2) before differentiation into mature follicular or marginal zone B cells. T1 and T2 cells are defined by cell surface phenotype, a short half-life and an increased susceptibility to apoptosis. T1 cells (AA4.1+IgM+IgD-

CD21lowCD23-) enter the spleen through the central arterioles and marginal sinus, where they migrate to B cell follicles in response to CXCL13144. In the follicles, T2 cells

31 (AA4.1+IgMhiIgDhiCD21midCD23+) gain the ability to recirculate and upregulate IgD and

CD23, but still express the immature cell surface marker AA4.1145.

T1 cell differentiation into T2 cells is dependent on BCR engagement, BAFF and non-canonical NF-κb signaling (Figure 8). BAFF production by stromal and myeloid cells promotes survival by inducing expression of anti-apoptotic proteins, including BCL-

2, BCL-xL and MCL-1146. BAFF deficiency blocks T1 to T2 differentiation, resulting in dramatic deficiencies in mature B cells147. Signaling through the BAFF receptor

(BAFFR) predominantly activates the noncanonical NF-κb signaling pathway, leading to induction of p52 through NF-κb-inducing kinase (NIK) and IκB kinase α (IKKα). NIK- deficient bone marrow chimeras lacked marginal zone B cells, while IKKα-deficient bone marrow chimeras had reduced follicular B cells148,149, suggesting that the noncanonical

NF-κb signaling pathway plays a role in follicular and marginal B cell development.

5biv. Follicular versus marginal zone B cell fate

Follicular versus marginal zone B cell development from the T2 cell stage is dependent on BCR engagement and signaling through Notch2, NF-κb and BAFFR (Figure 8).

Defects in the BCR signaling components Bruton’s tyrosine kinase (BTK) and phospholipase Cγ2 (PLCγ2) were associated with reduced numbers of follicular B cells, but normal numbers of marginal zone B cells150,151. In contrast, genetic alterations that increased BCR signaling, such as CD22b-deficient mice, drastically reduced marginal zone B cells152. Thus, weak BCR signaling favors marginal zone B cell development, while stronger BCR signaling favors follicular B cell development153.

b CD22 inhibits BCR signaling

32 Adult Spleen

- Antigen presentation MZ - Respond to TI-2 antigens of blood-borne pathogens to Tonic BCR secrete IgM BAFF and NF-!b signaling weak BCR, BAFF IgMhigh !b and Notch signaling low NF- IgD T1 T2 CD21high strong BCR CD23low BAFF and NF- AA4.1+ AA4.1+ !b IgM+ IgMhigh signaling - Germinal center reactions in IgD- IgDhigh FO response to T cell-dependent CD21low CD21int antigens CD23- CD23+ - Respond to T cell- low IgM independent antigens IgDhigh CD21int CD23high

Figure 8. Marginal and follicular B cell development. Immature B cells from the bone marrow migrate to the spleen to undergo further differentiation into mature B cell subsets. Transitional B cell differentiation requires tonic BCR engagement, BAFF survival signals and the alternative NF-κb pathway. Marginal zone or follicular B cell development depends on the strength of BCR engagement and signaling through

BAFF, NF-κb and/or Notch. This cell fate decision results in two B cell subsets that differ in anatomical location, cell marker expression and function. T1 or T2, transitional

B cell; BAFF, B cell activating factor; MZ, marginal zone B cell; FO, follicular B cell.

33 Once BCR signal strength determines B cell fate, NF-κb, Notch and BAFFR signaling drive the development of follicular or marginal zone B cells. In follicular B cells, strong BCR signaling through BTK and phosphoinositide 3 kinase (PI3K) contributes to activation of the canonical NF-κb signaling components p50 and RelA.

Null mutations in the canonical NF-κb signaling pathway, including the NF-κb essential modulator (Nemo) and IKKβ, led to a dramatic loss in all peripheral B cell subsets, implicating the canonical NF-κb signaling pathway in mature B cell development154.

Marginal B cell development also requires BAFFR signaling and the BCL10-MALT1c-

CARMA1 complex to activate the canonical NF-κb signaling pathway. Mice deficient in

BCL10, MALT1, or CARMA1 had impaired development of B1 and marginal zone B cells155-157.

Notch signaling is also an important signal transduction pathway in the development of marginal zone B cells. Mice deficient in components of the Notch signaling pathway, including Notch2, recombining binding protein suppressor of hairless

(RBP-Jκ) and mastermind-like 1 (MAML1), had defects in marginal zone B cell development131,158-160, while inactivation of the Notch suppressor MSX2-interacting protein (MINT) resulted in increased marginal zone B cells161.

5c. The UPR in B cell development

Several components of the UPR influence both early and late stages of B cell development. A seminal paper by Reimold and Glimcher described a physiological role for the IRE1α/XBP1 signaling pathway in plasma cell differentiation and antibody secretion. XBP1-deficient B cells became activated and proliferated normally in c Mucosa-associated lymphoid tissue lymphoma translocation protein 1 (MALT1)

34 response to antigen, but failed to differentiate into plasma cells. Plasma cell deficiency resulted in lower levels of basal immunoglobulins, as well as an inability to generate immunoglobulins against T cell-dependent or T cell-independent antigens70. IL-4, ATF6 and BLIMP1 induced the expression of spliced XBP1 in primary B cells, driving plasma cell differentiation162-164. Activation of IRE1α/XBP1 in primary B cells promoted ER expansion by upregulating chaperones and enzymes involved in transport, folding, glycosylation and trafficking165. Spliced XBP1 also induced expression of IL-6, a plasma cell growth factor162.

Interestingly, IRE1α signaling has also been shown to be important for early B cell development; pre-B and mature B cells were reduced in the bone marrow of IRE1α-

/-RAG2-/- chimeric mice. The developmental block at the pro-B cell stage was associated with defective IgH gene rearrangement due to reduced expression of TdT,

RAG1 and RAG269. Deficiency of TRAF2, a downstream effector molecule of IRE1α, also caused a block at the pro-B cell stage of development, but a causal relationship with impaired immunoglobulin gene rearrangement was not ascertained166. Spliced

XBP1 was also detected at the pro- and pre-B cell stage of development, but the significance of this finding remains unknown167.

6. Growth Factors and Cellular Niches Required for B Cell Development

Microenvironmental niches in the bone marrow support B cell lineage commitment and differentiation from HSCs. This is a highly dynamic process that depends upon stromal cell contact, soluble growth factors and cytokines throughout each developmental stage168 (Table 2 and Figure 9).

35 6a. B cell growth factors

6ai. CXCL12

Eqawa et al. identified CXC-chemokine ligand 12 (CXCL12) as the first soluble factor required after commitment to the B lineage. CXCL12 is highly expressed by stromal cells in the bone marrow. The primary receptor for CXCL12 is CXC-chemokine receptor

4 (CXCR4)169,170, whose expression is Table 2. B cell defects in mice that are deficient in 167 primarily limited to brain and lymphoid microenvironmental factors in the bone marrow . tissues171,172. Both CXCL12 and

CXCR4-deficient mice died perinatally and had identical defects in neural and vascular tissue173,174. Deficiency of either CXCL12 or CXCR4 resulted in severely reduced numbers of pre-pro-

B cells in the bone marrow of fetal and adult mice173-175 (Table 2). Ma et al. further demonstrated that B cell precursors were generated in the bone marrow of CXCR4- deficient mice, but egressed to the periphery before maturation was complete176. Thus,

CXCL12 secretion by stromal cells attracts and retains B cell precursors for development in the bone marrow. Further, plasma cell localization in the bone marrow is also dependent on CXCL12177,178.

6aii. IL-7

IL-7 was identified in 1988 by Namen et al. as the first environmental factor to induce rapid proliferation of B cell precursors179. The IL-7 receptor is comprised of the IL-7Rα

36 Bone marrow *!+%

Dendritic cells Mature Immature B B cells

Blood Megakaryocyte vessel IL-7-secreting cell

Pre-B

Eosinophil !"#&% !"#$% Plasma cell

'()!"%

,-,"./% Pro-B CAR cell

Pre-pro-B

HSC CLP

Osteoblast Bone

Figure 9. Microenvironmental niches for B cell development in the bone marrow.

Stromal and immune cell types support B cell development by expressing cell-surface molecules and secreting growth factors and cytokines that promote retention, survival and differentiation in the bone marrow. CAR, CXCL12-expressing reticular cells; HSCs, hematopoietic stem cells; CLP, common lymphoid progenitor; MIF, migration inhibitory factor; APRIL, a proliferation-inducing ligand.

37 chain and the common γ chain, which also signals for IL-2, IL-4, IL-9 and IL-15. IL-7 signaling in B cell precursors induces IgH gene rearrangement180, the pro-survival factor

MCL-1181 and EBF to maintain B cell differentiation182. Deficiency of either IL-7 or IL-

7Rα resulted in severe defects in both T and B cell development; B cell precursors were drastically reduced starting at the pro-B cell stage (Table 2)183,184. Although the number of CLPs and pre-pro-B cells were normal in IL-7 and IL-7Rα-deficient mice, their ability to differentiate in vitro was severely impaired185, implicating that IL-7 signaling is necessary starting at the CLP stage of development.

6aiii. FLT3L

The fms-like-tyrosine-3 ligand (FLT3L) is a cell surface protein that often undergoes proteolytic cleavage to generate a soluble form, both of which are active in vivo186,187.

FLT3L-deficient mice had drastically reduced CLPs in the bone marrow188, suggesting that FLT3L promotes survival of CLPs (Table 2). The receptor for FLT3L is highly expressed during early lymphoid development through the pre-B cell stage187,189,190.

FLT3 receptor-deficient mice had a milder defect in B lymphopoiesis; pre-pro-B, pro-B and pre-B cells were significantly reduced, while immature and mature B cell populations were normal191. Interestingly, mice double deficient in IL-7Rα and FLT3L had a more severe defect in the early and late stages of B cell development than single deficient mice, suggesting that IL-7 and FLT3L synergistically regulate B lymphopoiesis192.

6aiv. RANKL

38 The receptor activator of nuclear factor-κB ligand (RANKL) is predominantly expressed by osteocytes193 and drives osteoclast differentiation by interacting with the RANKL receptor (RANK) on osteoclast precursors. Mice deficient in either RANK or RANKL had osteopetrosis due to defective osteoclastogenesis, but also lacked all lymph nodes and had severe defects in early T and B cell development194,195 (Table 2). B cell development is dependent on RANKL expression by cells of the hematopoietic lineage195, despite much lower RANKL expression than that of bone cells193.

Reconstitution of RAG1-deficient mice with RANKL-deficient bone marrow resulted in reduced pre-B and immature B cells, while wild-type bone marrow transplanted into

RANKL-deficient recipients rescued B cell development195. The cellular source of

RANKL and downstream effects of RANKL signaling in B cell development remains entirely unclear.

6av. SCF

Stem cell factor (SCF) is a ligand for the tyrosine kinase receptor c-kit, which is highly expressed by hematopoietic progenitor cells and fully differentiated mast cells. The

SCF-c-kit signaling axis is crucial for the development of many hematopoietic cell lineages including multipotential progenitors (MPPs), T cells, mast cells and erythrocytes. Although B cell development in c-kit-deficient mice was normal during the fetal and neonatal period196, several studies indicate a role for SCF-c-kit signaling in adult B lymphopoiesis. Mice with spontaneous null mutations in c-kit and SCF died from anemia within 10 days of birth197, but overexpression of erythropoietin in c-kit- deficient mice (Wepo mice) rescued lethality198. Adult Wepo mice had normal numbers

39 of pre-pro B cells, but reduced CLPs, pro-B and pre-B cells in the bone marrow198

(Table 2). Further, discovery of a viable and healthy c-kit-deficient mouse that arose from heterozygous crossing of c-kit mutants had a similar phenotype as Wepo mice198.

6b. Bone marrow niches that support B cell development

6bi. CAR niche: reticular cells expressing CXCL12

The CXCL12-CXCR4 signaling axis is crucial for both early and late B cell development

(section 6ai). Mice containing a green fluorescent protein (GFP) gene knocked into the

CXCL12 locus (CXCL12-GFP mice) were used to identify the distribution of cells that expressed CXCL12 in the bone marrow during B cell development. CXCL12-GFP expression was detected on vascular adhesion molecule 1 (VCAM-1)-positive reticular cells, termed CAR cells, which make up approximately 0.27% of all nucleated cells in the bone marrow. CAR cells are uniformly distributed throughout the bone marrow and are morphologically similar to dendritic cells, forming a matrix with their wide net of cytoplasmic processes178,199. CXCL12-expressing cells are found in tight association with pre-pro B cells and plasma cells. Conditional depletion of CAR cells impaired

CXCL12 and SCF production, which was associated with lower numbers of HSCs,

CLPs and pro-B cells200. Secretion of CXCL12 by CAR cells recruits eosinophils and megakaryocytes, which also support the plasma cell niche by secreting a proliferation- inducing ligand (APRIL) and/or IL-6201,202 (Figure 9).

6bii. IL-7-expressing cells

40 IL-7 signaling plays an important role in early B cell development by influencing CLP and pro-B cell differentiation (section 6aii). Similar to CAR cells, IL-7-expressing cells are scattered throughout the bone marrow and are VCAM-1-positive, but they have a fibroblast-like morphology, lacking the long dendrites of CAR cells. While CAR cells interact specifically with pre-pro B cells and plasma cells, IL-7-expressing cells associate tightly with pro-B cells in the bone marrow178 (Figure 9).

6biii. Dendritic cells

Resident dendritic cells are clustered around blood vessels in the blood marrow, secreting migration-inhibitory factor 1 (MIF1), which supports the survival of recirculating mature B cells203 (Figure 9). Ablation of bone marrow dendritic cells resulted in reduced levels of mature B cells, which was rescued by expression BCL-2, an anti- apoptotic protein.

6biv. Osteoblasts

Bone-forming cells of the osteoblast lineage express VCAM-1, CXCL12 and IL-7, which are sufficient to support all stages of B cell development from HSCs in vitro204 (Figure

9). Induction of CXCL12 and IL-7 in primary osteoblasts was dependent on parathyroid hormone (PTH)204,205 and ablation of the PTH signaling pathway in osteoprogenitors reduced pro-B and pre-B cells in the bone marrow206. Conditional depletion of osteoblasts decreased bone marrow cellularity, which was associated with reduced numbers of HSCs, B cells and erythrocytes207.

41 Osteoblasts can differentiate into osteocytes, which regulate bone resorption by osteoclasts through RANKL193. Inhibiting bone resorption in mice with zoledronic acid treatment impaired all stages of B cell development in the bone marrow, while HSCs and CLPs were normal208. However, treatment with zoledronic acid was also associated with reduced numbers of osteoblasts, which likely contributed to the B cell phenotype208.

7. The UPR in Osteoblast Differentiation

Osteoblasts develop from mesenchymal stem cells (MSCs) and commitment to the osteoblast lineage requires the transcription factors Runx2 and Osterix and the canonical Wnt signaling pathway. The primary function of osteoblasts is the synthesis, deposition and mineralization of bone, although they also play an important role in hematopoiesis (section 6biv). Mature osteoblasts can become either quiescent bone- lining cells, undergo apoptosis, or differentiate into osteocytes, which are surrounded by extracellular matrix and regulate bone remodeling. Several components of the UPR are essential for osteoblast differentiation and function (Figure 10).

7a. PERK/eIF2α

A role for the UPR (section 2ci) in skeletal development was revealed when a loss of function mutation in human PERK resulted in growth retardation, epiphyseal dysplasia and osteoporosis104,105. A PERK-deficient mouse model duplicated the skeletal defects observed in humans, providing a resource to study the underlying mechanisms of PERK in skeletal development. Neonatal PERK-deficient mice had reduced mineralization in

42 Runx2

MSC Osteoblast Osteocyte ATF6 Bone-lining cell

Osterix

IRE1!/XBP1 ATF4 PERK/eIF2!

Figure 10. The UPR in osteogenesis. The UPR drives osteoblast differentiation by regulating Osterix, a transcription factor necessary for mature osteoblast development.

ATF6 induction by Runx2 influences osteoblast function by controlling the expression of osteocalcin, an extracellular matrix protein. Genetic alterations affecting the UPR cause growth retardation and skeletal dysplasias in mice and humans.

43 flat and long bones, implicating impaired ossification. Osteoblasts were decreased in bone tissue of PERK-deficient mice, which was associated with impaired proliferation and differentiation of primary osteoblasts in vitro209. PERK-deficient osteoblasts also had signs of increased ER stress including ER distention and retention of type I collagen, which surprisingly, did not activate the UPR or increase cell death64,209.

7b. ATF4

ATF4 is a downstream mediator of PERK/eIF2α signaling and a crucial component of osteoblast biology. ATF4 influences bone formation by regulating osteoblast-specific gene expression, amino acid transport and synthesis of type I collagen210. Osteoblast differentiation is also dependent on ATF4, which regulates the expression of Osterix

(Figure 10), an essential transcription factor of mature osteoblast development211.

ATF4-deficient osteoblasts had defects in proliferation and survival, implicating that

ATF4 is necessary for osteoblast homeostasis211,212.

Although PERK/eIF2α can induce ATF4, there is conflicting data on whether

ATF4 functions downstream of PERK in osteoblasts. Downstream target genes of

ATF4 that regulate amino acid metabolism (and ultimately, osteoblast function) were decreased in ATF4-deficient osteoblasts, but were unchanged in PERK-deficient osteoblasts209. Further, PERK deficiency resulted in accumulation of type I collagen209, while ATF4-deficient osteoblasts had impaired production of type I collagen210.

However, other studies reported that ATF4 activation was impaired in PERK-deficient osteoblasts and ATF4 rescued the defects in mineralization and osteoblast differentiation observed in PERK-deficient osteoblasts213. Further studies are needed to

44 identify the underlying mechanisms involving PERK and ATF4 in osteoblast differentiation and function.

7c. ATF6

A recent study identified a role for the UPR signal transducer ATF6 in osteoblast differentiation. The transcription factor Runx2, which induces the differentiation of

MSCs into osteoblasts, increases expression of ATF6 by binding to the ATF6 promoter region. Activation of ATF6 stimulates osteoblast differentiation through upregulation of osteocalcin and increases extracellular matrix production214. However, defects in skeletal development in ATF6-deficient mice have not been described.

The old astrocyte specifically-induced substance (OASIS) is structurally homologous to ATF6 and modulates UPR signaling by activating the transcription of genes containing ERSE and cyclic AMP-responsive elements (CRE)215. OASIS regulates type I collagen production by binding to an ERSE in the type I collagen promoter. OASIS deficiency impaired osteoblast differentiation and function in vitro and

OASIS-deficient mice were osteopenic in association with decreased type I collagen in bone tissue216.

7d. IRE1α/XBP1

The IRE1α/XBP1 signaling pathway is upregulated during bone morphogenetic protein

2 (BMP2)-induced osteoblast differentiationd. XBP1 induces the expression of Osterix, a transcription factor necessary for osteoblast differentiation217. IRE1α or XBP1

d BMP2 induces osteoblast differentiation by upregulating osteoblast-specific genes including Runx2 and Osterix

45 deficiency impaired osteoblast differentiation in vitro. However, given that both IRE1α and XBP1-deficient mice were embryonic lethal69,70, it remains unclear whether this arm of the UPR plays a role in postnatal skeletal development in vivo.

8. Summary and Hypothesis

Highly secretory cells, including pancreatic β-cells, plasma cells and osteoblasts, require the UPR to maintain ER homeostasis in the face of increased protein synthesis.

The UPR signal transducers PERK/eIF2α, ATF6 and IRE1α/XBP1 reduce protein load by attenuating protein translation and upregulating protein folding and degradation machinery. Genetic mutations affecting the UPR cause defects in glucose metabolism, skeletal development and antibody-mediated immune responses in mice and humans, highlighting the importance of this signaling pathway in highly secretory cells.

ERdj4 was identified a decade ago by Shen et al. as a BiP/GRP78 cochaperone expressed most highly in secretory tissues and significantly upregulated in response to

ER stress36. Since then, our laboratory and others have demonstrated that ERdj4 is induced by the UPR to assist in the removal of unfolded and misfolded substrates from the ER lumen. This conclusion is supported by: (1) upregulation of ERdj4 by

IRE1α/XBP1 signaling in response to terminally misfolded protein, (2) association of

ERdj4 with unfolded (insulin2WT) and misfolded (insulin2C96Y, SP-CΔexon4) substrates, (3) interaction of ERdj4 with ERAD machinery and (4) accumulation of unfolded/misfolded protein in the absence of ERdj42,35.

Although previous studies identified the function of ERdj4 in vitro, the biological relevance of ERdj4 was entirely unknown. The essential role of the UPR in highly

46 secretory cells, as well as prominent expression of ERdj4 in secretory tissues, led to the generation of the central hypothesis that underlies the studies in this thesis: the chaperone activity of ERdj4 is required for homeostasis and function of secretory cells in vivo.

47 References

1. Kanapin, A. et al. Mouse proteome analysis. Genome Res 13, 1335-1344 (2003).

2. Dong, M., Bridges, J. P., Apsley, K., Xu, Y. & Weaver, T. E. ERdj4 and ERdj5 are required for endoplasmic reticulum-associated protein degradation of misfolded surfactant protein C. Mol Biol Cell 19, 2620-2630 (2008).

3. Hamman, B. D., Hendershot, L. M. & Johnson, A. E. BiP maintains the permeability barrier of the ER membrane by sealing the lumenal end of the translocon pore before and early in translocation. Cell 92, 747-758 (1998).

4. Hendershot, L. et al. Inhibition of immunoglobulin folding and secretion by dominant negative BiP ATPase mutants. Proc Natl Acad Sci U S A 93, 5269-5274 (1996).

5. Simons, J. F., Ferro-Novick, S., Rose, M. D. & Helenius, A. BiP/Kar2p serves as a molecular chaperone during carboxypeptidase Y folding in yeast. J Cell Biol 130, 41-49 (1995).

6. Brodsky, J. L. et al. The requirement for molecular chaperones during endoplasmic reticulum-associated protein degradation demonstrates that protein export and import are mechanistically distinct. J Biol Chem 274, 3453-3460 (1999).

7. Plemper, R. K., Bohmler, S., Bordallo, J., Sommer, T. & Wolf, D. H. Mutant analysis links the translocon and BiP to retrograde protein transport for ER degradation. Nature 388, 891-895 (1997).

8. Lievremont, J. P., Rizzuto, R., Hendershot, L. & Meldolesi, J. BiP, a major chaperone protein of the endoplasmic reticulum lumen, plays a direct and important role in the storage of the rapidly exchanging pool of Ca2+. J Biol Chem 272, 30873-30879 (1997).

9. Bertolotti, A., Zhang, Y., Hendershot, L. M., Harding, H. P. & Ron, D. Dynamic interaction of BiP and ER stress transducers in the unfolded-protein response. Nat Cell Biol 2, 326-332 (2000).

10. Shen, J., Chen, X., Hendershot, L. & Prywes, R. ER stress regulation of ATF6 localization by dissociation of BiP/GRP78 binding and unmasking of Golgi localization signals. Dev Cell 3, 99-111 (2002).

11. Jiang, J., Prasad, K., Lafer, E. M. & Sousa, R. Structural basis of interdomain communication in the Hsc70 chaperone. Mol Cell 20, 513-524 (2005).

12. Kampinga, H. H. & Craig, E. A. The HSP70 chaperone machinery: J proteins as drivers of functional specificity. Nat Rev Mol Cell Biol 11, 579-592 (2010).

48

13. Georgopoulos, C. The emergence of the chaperone machines. Trends Biochem Sci 17, 295-299 (1992).

14. Liberek, K., Marszalek, J., Ang, D., Georgopoulos, C. & Zylicz, M. Escherichia coli DnaJ and GrpE heat shock proteins jointly stimulate ATPase activity of DnaK. Proc Natl Acad Sci U S A 88, 2874-2878 (1991).

15. Osipiuk, J., Georgopoulos, C. & Zylicz, M. Initiation of lambda DNA replication. The Escherichia coli small heat shock proteins, DnaJ and GrpE, increase DnaK's affinity for the lambda P protein. J Biol Chem 268, 4821-4827 (1993).

16. Yochem, J. et al. Genetic analysis of two genes, dnaJ and dnaK, necessary for Escherichia coli and bacteriophage lambda DNA replication. Mol Gen Genet 164, 9-14 (1978).

17. Cheetham, M. E. & Caplan, A. J. Structure, function and evolution of DnaJ: conservation and adaptation of chaperone function. Cell Stress Chaperones 3, 28- 36 (1998).

18. Feldheim, D., Rothblatt, J. & Schekman, R. Topology and functional domains of Sec63p, an endoplasmic reticulum membrane protein required for secretory protein translocation. Mol Cell Biol 12, 3288-3296 (1992).

19. Wall, D., Zylicz, M. & Georgopoulos, C. The NH2-terminal 108 amino acids of the Escherichia coli DnaJ protein stimulate the ATPase activity of DnaK and are sufficient for lambda replication. J Biol Chem 269, 5446-5451 (1994).

20. Ahmad, A. et al. 70 kDa chaperone/DnaJ cochaperone complex employs an unusual dynamic interface. Proc Natl Acad Sci U S A 108, 18966-18971 (2011).

21. Fan, C. Y., Ren, H. Y., Lee, P., Caplan, A. J. & Cyr, D. M. The type I Hsp40 zinc finger-like region is required for Hsp70 to capture non-native polypeptides from Ydj1. J Biol Chem 280, 695-702 (2005).

22. Szabo, A., Korszun, R., Hartl, F. U. & Flanagan, J. A zinc finger-like domain of the molecular chaperone DnaJ is involved in binding to denatured protein substrates. EMBO J 15, 408-417 (1996).

23. Kota, P., Summers, D. W., Ren, H. Y., Cyr, D. M. & Dokholyan, N. V. Identification of a consensus motif in substrates bound by a Type I Hsp40. Proc Natl Acad Sci U S A 106, 11073-11078 (2009).

24. Schlenstedt, G., Harris, S., Risse, B., Lill, R. & Silver, P. A. A yeast DnaJ homologue, Scj1p, can function in the endoplasmic reticulum with BiP/Kar2p via a

49

conserved domain that specifies interactions with . J Cell Biol 129, 979-988 (1995).

25. Nishikawa, S. & Endo, T. The yeast JEM1p is a DnaJ-like protein of the endoplasmic reticulum membrane required for nuclear fusion. J Biol Chem 272, 12889-12892 (1997).

26. Silberstein, S., Schlenstedt, G., Silver, P. A. & Gilmore, R. A role for the DnaJ homologue Scj1p in protein folding in the yeast endoplasmic reticulum. J Cell Biol 143, 921-933 (1998).

27. Nishikawa, S. I., Fewell, S. W., Kato, Y., Brodsky, J. L. & Endo, T. Molecular chaperones in the yeast endoplasmic reticulum maintain the solubility of proteins for retrotranslocation and degradation. J Cell Biol 153, 1061-1070 (2001).

28. Hageman, J. & Kampinga, H. H. Computational analysis of the human HSPH/HSPA/DNAJ family and cloning of a human HSPH/HSPA/DNAJ expression library. Cell Stress Chaperones 14, 1-21 (2009).

29. Chevalier, M., Rhee, H., Elguindi, E. C. & Blond, S. Y. Interaction of murine BiP/GRP78 with the DnaJ homologue MTJ1. J Biol Chem 275, 19620-19627 (2000).

30. Dudek, J. et al. A novel type of co-chaperone mediates transmembrane recruitment of DnaK-like chaperones to ribosomes. EMBO J 21, 2958-2967 (2002).

31. Dudek, J. et al. ERj1p has a basic role in protein biogenesis at the endoplasmic reticulum. Nat Struct Mol Biol 12, 1008-1014 (2005).

32. Meyer, H. A. et al. Mammalian Sec61 is associated with Sec62 and Sec63. J Biol Chem 275, 14550-14557 (2000).

33. Rutkowski, D. T. et al. The role of p58IPK in protecting the stressed endoplasmic reticulum. Mol Biol Cell 18, 3681-3691 (2007).

34. Shen, Y. & Hendershot, L. M. ERdj3, a stress-inducible endoplasmic reticulum DnaJ homologue, serves as a cofactor for BiP's interactions with unfolded substrates. Mol Biol Cell 16, 40-50 (2005).

35. Lai, C. W., Otero, J. H., Hendershot, L. M. & Snapp, E. ERdj4 protein is a soluble endoplasmic reticulum (ER) DnaJ family protein that interacts with ER-associated degradation machinery. J Biol Chem 287, 7969-7978 (2012).

36. Shen, Y., Meunier, L. & Hendershot, L. M. Identification and characterization of a novel endoplasmic reticulum (ER) DnaJ homologue, which stimulates ATPase

50

activity of BiP in vitro and is induced by ER stress. J Biol Chem 277, 15947-15956 (2002).

37. Ushioda, R. et al. ERdj5 is required as a disulfide reductase for degradation of misfolded proteins in the ER. Science 321, 569-572 (2008).

38. Zahedi, R. P. et al. Analysis of the membrane proteome of canine pancreatic rough microsomes identifies a novel Hsp40, termed ERj7. Proteomics 9, 3463-3473 (2009).

39. Otero, J. H., Lizak, B. & Hendershot, L. M. Life and death of a BiP substrate. Semin Cell Dev Biol 21, 472-478 (2010).

40. Kanemoto, S. et al. XBP1 activates the transcription of its target genes via an ACGT core sequence under ER stress. Biochem Biophys Res Commun 331, 1146-1153 (2005).

41. Lee, A. H., Iwakoshi, N. N. & Glimcher, L. H. XBP-1 regulates a subset of endoplasmic reticulum resident chaperone genes in the unfolded protein response. Mol Cell Biol 23, 7448-7459 (2003).

42. Stone, K. R., Smith, R. E. & Joklik, W. K. Changes in membrane polypeptides that occur when chick embryo fibroblasts and NRK cells are transformed with avian sarcoma viruses. Virology 58, 86-100 (1974).

43. Shiu, R. P., Pouyssegur, J. & Pastan, I. Glucose depletion accounts for the induction of two transformation-sensitive membrane proteinsin Rous sarcoma virus-transformed chick embryo fibroblasts. Proc Natl Acad Sci U S A 74, 3840- 3844 (1977).

44. Lee, A. S. The glucose-regulated proteins: stress induction and clinical applications. Trends Biochem Sci 26, 504-510 (2001).

45. Haas, I. G. & Wabl, M. Immunoglobulin heavy chain binding protein. Nature 306, 387-389 (1983).

46. Gething, M. J. & Sambrook, J. Protein folding in the cell. Nature 355, 33-45 (1992).

47. Resendez, E. J., Wooden, S. K. & Lee, A. S. Identification of highly conserved regulatory domains and protein-binding sites in the promoters of the rat and human genes encoding the stress-inducible 78-kilodalton glucose-regulated protein. Mol Cell Biol 8, 4579-4584 (1988).

48. Mori, K. et al. A 22 bp cis-acting element is necessary and sufficient for the induction of the yeast KAR2 (BiP) gene by unfolded proteins. EMBO J 11, 2583- 2593 (1992).

51

49. Cox, J. S., Shamu, C. E. & Walter, P. Transcriptional induction of genes encoding endoplasmic reticulum resident proteins requires a transmembrane protein kinase. Cell 73, 1197-1206 (1993).

50. Mori, K., Ma, W., Gething, M. J. & Sambrook, J. A transmembrane protein with a cdc2+/CDC28-related kinase activity is required for signaling from the ER to the nucleus. Cell 74, 743-756 (1993).

51. Cox, J. S. & Walter, P. A novel mechanism for regulating activity of a transcription factor that controls the unfolded protein response. Cell 87, 391-404 (1996).

52. Sidrauski, C. & Walter, P. The transmembrane kinase Ire1p is a site-specific endonuclease that initiates mRNA splicing in the unfolded protein response. Cell 90, 1031-1039 (1997).

53. Mori, K., Ogawa, N., Kawahara, T., Yanagi, H. & Yura, T. Palindrome with spacer of one nucleotide is characteristic of the cis-acting unfolded protein response element in Saccharomyces cerevisiae. J Biol Chem 273, 9912-9920 (1998).

54. Travers, K. J. et al. Functional and genomic analyses reveal an essential coordination between the unfolded protein response and ER-associated degradation. Cell 101, 249-258 (2000).

55. Haze, K., Yoshida, H., Yanagi, H., Yura, T. & Mori, K. Mammalian transcription factor ATF6 is synthesized as a transmembrane protein and activated by proteolysis in response to endoplasmic reticulum stress. Mol Biol Cell 10, 3787- 3799 (1999).

56. Ye, J. et al. ER stress induces cleavage of membrane-bound ATF6 by the same proteases that process SREBPs. Mol Cell 6, 1355-1364 (2000).

57. Okada, T., Yoshida, H., Akazawa, R., Negishi, M. & Mori, K. Distinct roles of activating transcription factor 6 (ATF6) and double-stranded RNA-activated protein kinase-like endoplasmic reticulum kinase (PERK) in transcription during the mammalian unfolded protein response. Biochem J 366, 585-594 (2002).

58. Yamamoto, K. et al. Transcriptional induction of mammalian ER quality control proteins is mediated by single or combined action of ATF6alpha and XBP1. Dev Cell 13, 365-376 (2007).

59. Yoshida, H., Haze, K., Yanagi, H., Yura, T. & Mori, K. Identification of the cis-acting endoplasmic reticulum stress response element responsible for transcriptional induction of mammalian glucose-regulated proteins. Involvement of basic leucine zipper transcription factors. J Biol Chem 273, 33741-33749 (1998).

52

60. Wu, J. et al. ATF6alpha optimizes long-term endoplasmic reticulum function to protect cells from chronic stress. Dev Cell 13, 351-364 (2007).

61. Harding, H. P., Zhang, Y. & Ron, D. Protein translation and folding are coupled by an endoplasmic-reticulum-resident kinase. Nature 397, 271-274 (1999).

62. Harding, H. P. et al. An integrated stress response regulates amino acid metabolism and resistance to oxidative stress. Mol Cell 11, 619-633 (2003).

63. Harding, H. P. et al. Diabetes mellitus and exocrine pancreatic dysfunction in perk- /- mice reveals a role for translational control in secretory cell survival. Mol Cell 7, 1153-1163 (2001).

64. Zhang, P. et al. The PERK eukaryotic initiation factor 2 alpha kinase is required for the development of the skeletal system, postnatal growth, and the function and viability of the pancreas. Mol Cell Biol 22, 3864-3874 (2002).

65. Shen, X. et al. Complementary signaling pathways regulate the unfolded protein response and are required for C. elegans development. Cell 107, 893-903 (2001).

66. Yoshida, H., Matsui, T., Yamamoto, A., Okada, T. & Mori, K. XBP1 mRNA is induced by ATF6 and spliced by IRE1 in response to ER stress to produce a highly active transcription factor. Cell 107, 881-891 (2001).

67. Urano, F. et al. Coupling of stress in the ER to activation of JNK protein kinases by transmembrane protein kinase IRE1. Science 287, 664-666 (2000).

68. Iwawaki, T., Akai, R., Yamanaka, S. & Kohno, K. Function of IRE1 alpha in the placenta is essential for placental development and embryonic viability. Proc Natl Acad Sci U S A 106, 16657-16662 (2009).

69. Zhang, K. et al. The unfolded protein response sensor IRE1alpha is required at 2 distinct steps in B cell lymphopoiesis. J Clin Invest 115, 268-281 (2005).

70. Reimold, A. M. et al. Plasma cell differentiation requires the transcription factor XBP-1. Nature 412, 300-307 (2001).

71. Lee, A. H., Chu, G. C., Iwakoshi, N. N. & Glimcher, L. H. XBP-1 is required for biogenesis of cellular secretory machinery of exocrine glands. EMBO J 24, 4368- 4380 (2005).

72. Harding, H. P. et al. Regulated translation initiation controls stress-induced gene expression in mammalian cells. Mol Cell 6, 1099-1108 (2000).

53

73. McCullough, K. D., Martindale, J. L., Klotz, L. O., Aw, T. Y. & Holbrook, N. J. Gadd153 sensitizes cells to endoplasmic reticulum stress by down-regulating Bcl2 and perturbing the cellular redox state. Mol Cell Biol 21, 1249-1259 (2001).

74. Nishitoh, H. et al. ASK1 is essential for endoplasmic reticulum stress-induced neuronal cell death triggered by expanded polyglutamine repeats. Genes Dev 16, 1345-1355 (2002).

75. Yoneda, T. et al. Activation of caspase-12, an endoplastic reticulum (ER) resident caspase, through tumor factor receptor-associated factor 2-dependent mechanism in response to the ER stress. J Biol Chem 276, 13935-13940 (2001).

76. Rhodes, C. J., Shoelson, S. & Halban, P. A. Insulin biosynthesis, processing, and chemistry. Joslin’s diabetes mellitus. 14th ed. Philadelphia: Lippincott Williams & Wilkins 65-82 (2005).

77. Schuit, F. C., In't Veld, P. A. & Pipeleers, D. G. Glucose stimulates proinsulin biosynthesis by a dose-dependent recruitment of pancreatic beta cells. Proc Natl Acad Sci U S A 85, 3865-3869 (1988).

78. Welsh, M., Scherberg, N., Gilmore, R. & Steiner, D. F. Translational control of insulin biosynthesis. Evidence for regulation of elongation, initiation and signal- recognition-particle-mediated translational arrest by glucose. Biochem J 235, 459- 467 (1986).

79. Poitout, V. & Robertson, R. P. Glucolipotoxicity: fuel excess and beta-cell dysfunction. Endocr Rev 29, 351-366 (2008).

80. Robertson, R. P. Chronic oxidative stress as a central mechanism for glucose toxicity in pancreatic islet beta cells in diabetes. J Biol Chem 279, 42351-42354 (2004).

81. Harmon, J. S., Stein, R. & Robertson, R. P. Oxidative stress-mediated, post- translational loss of MafA protein as a contributing mechanism to loss of insulin gene expression in glucotoxic beta cells. J Biol Chem 280, 11107-11113 (2005).

82. Robertson, R. P., Zhang, H. J., Pyzdrowski, K. L. & Walseth, T. F. Preservation of insulin mRNA levels and insulin secretion in HIT cells by avoidance of chronic exposure to high glucose concentrations. J Clin Invest 90, 320-325 (1992).

83. Tu, B. P. & Weissman, J. S. Oxidative protein folding in eukaryotes: mechanisms and consequences. J Cell Biol 164, 341-346 (2004).

84. Lenzen, S., Drinkgern, J. & Tiedge, M. Low antioxidant enzyme gene expression in pancreatic islets compared with various other mouse tissues. Free Radic Biol Med 20, 463-466 (1996).

54

85. Kahn, S. E., Hull, R. L. & Utzschneider, K. M. Mechanisms linking obesity to insulin resistance and type 2 diabetes. Nature 444, 840-846 (2006).

86. Preston, A. M., Gurisik, E., Bartley, C., Laybutt, D. R. & Biden, T. J. Reduced endoplasmic reticulum (ER)-to-Golgi protein trafficking contributes to ER stress in lipotoxic mouse beta cells by promoting protein overload. Diabetologia 52, 2369- 2373 (2009).

87. Back, S. H. & Kaufman, R. J. Endoplasmic reticulum stress and type 2 diabetes. Annu Rev Biochem 81, 767-793 (2012).

88. Cnop, M. et al. Selective inhibition of eukaryotic translation initiation factor 2 alpha dephosphorylation potentiates fatty acid-induced endoplasmic reticulum stress and causes pancreatic beta-cell dysfunction and apoptosis. J Biol Chem 282, 3989- 3997 (2007).

89. Lai, E., Bikopoulos, G., Wheeler, M. B., Rozakis-Adcock, M. & Volchuk, A. Differential activation of ER stress and apoptosis in response to chronically elevated free fatty acids in pancreatic beta-cells. Am J Physiol Endocrinol Metab 294, E540-50 (2008).

90. Laybutt, D. R. et al. Influence of diabetes on the loss of beta cell differentiation after islet transplantation in rats. Diabetologia 50, 2117-2125 (2007).

91. Karaskov, E. et al. Chronic palmitate but not oleate exposure induces endoplasmic reticulum stress, which may contribute to INS-1 pancreatic beta-cell apoptosis. Endocrinology 147, 3398-3407 (2006).

92. Kharroubi, I. et al. Free fatty acids and cytokines induce pancreatic beta-cell apoptosis by different mechanisms: role of nuclear factor-kappaB and endoplasmic reticulum stress. Endocrinology 145, 5087-5096 (2004).

93. Anderson, E. K., Gutierrez, D. A. & Hasty, A. H. Adipose tissue recruitment of leukocytes. Curr Opin Lipidol 21, 172-177 (2010).

94. Boden, G. Obesity and free fatty acids. Endocrinol Metab Clin North Am 37, 635- 46, viii-ix (2008).

95. Oyadomari, S. et al. Nitric oxide-induced apoptosis in pancreatic beta cells is mediated by the endoplasmic reticulum stress pathway. Proc Natl Acad Sci U S A 98, 10845-10850 (2001).

96. Cardozo, A. K. et al. Cytokines downregulate the sarcoendoplasmic reticulum pump Ca2+ ATPase 2b and deplete endoplasmic reticulum Ca2+, leading to induction of endoplasmic reticulum stress in pancreatic beta-cells. Diabetes 54, 452-461 (2005).

55

97. Herbach, N. et al. Dominant-negative effects of a novel mutated Ins2 allele causes early-onset diabetes and severe beta-cell loss in Munich Ins2C95S mutant mice. Diabetes 56, 1268-1276 (2007).

98. Yoshioka, M., Kayo, T., Ikeda, T. & Koizumi, A. A novel locus, Mody4, distal to D7Mit189 on 7 determines early-onset NIDDM in nonobese C57BL/6 (Akita) mutant mice. Diabetes 46, 887-894 (1997).

99. Liu, M. et al. Proinsulin misfolding and diabetes: mutant INS gene-induced diabetes of youth. Trends Endocrinol Metab 21, 652-659 (2010).

100. Huang, C. J. et al. Induction of endoplasmic reticulum stress-induced beta-cell apoptosis and accumulation of polyubiquitinated proteins by human islet amyloid polypeptide. Am J Physiol Endocrinol Metab 293, E1656-62 (2007).

101. Huang, C. J. et al. High expression rates of human islet amyloid polypeptide induce endoplasmic reticulum stress mediated beta-cell apoptosis, a characteristic of humans with type 2 but not type 1 diabetes. Diabetes 56, 2016-2027 (2007).

102. Hull, R. L. et al. Amyloid formation in human IAPP transgenic mouse islets and pancreas, and human pancreas, is not associated with endoplasmic reticulum stress. Diabetologia 52, 1102-1111 (2009).

103. Scheuner, D. & Kaufman, R. J. The unfolded protein response: a pathway that links insulin demand with beta-cell failure and diabetes. Endocr Rev 29, 317-333 (2008).

104. Delepine, M. et al. EIF2AK3, encoding translation initiation factor 2-alpha kinase 3, is mutated in patients with Wolcott-Rallison syndrome. Nat Genet 25, 406-409 (2000).

105. Thornton, C. M., Carson, D. J. & Stewart, F. J. Autopsy findings in the Wolcott- Rallison syndrome. Pediatr Pathol Lab Med 17, 487-496 (1997).

106. Zhang, W. et al. PERK EIF2AK3 control of pancreatic beta cell differentiation and proliferation is required for postnatal glucose homeostasis. Cell Metab 4, 491-497 (2006).

107. Scheuner, D. et al. Translational control is required for the unfolded protein response and in vivo glucose homeostasis. Mol Cell 7, 1165-1176 (2001).

108. Han, D. et al. IRE1alpha kinase activation modes control alternate endoribonuclease outputs to determine divergent cell fates. Cell 138, 562-575 (2009).

56

109. Lipson, K. L., Ghosh, R. & Urano, F. The role of IRE1alpha in the degradation of insulin mRNA in pancreatic beta-cells. PLoS One 3, e1648 (2008).

110. Lee, A. H., Heidtman, K., Hotamisligil, G. S. & Glimcher, L. H. Dual and opposing roles of the unfolded protein response regulated by IRE1alpha and XBP1 in proinsulin processing and insulin secretion. Proc Natl Acad Sci U S A 108, 8885- 8890 (2011).

111. Reimold, A. M. et al. An essential role in liver development for transcription factor XBP-1. Genes Dev 14, 152-157 (2000).

112. Meex, S. J. et al. Activating transcription factor 6 polymorphisms and haplotypes are associated with impaired glucose homeostasis and type 2 diabetes in Dutch Caucasians. J Clin Endocrinol Metab 92, 2720-2725 (2007).

113. Thameem, F., Farook, V. S., Bogardus, C. & Prochazka, M. Association of amino acid variants in the activating transcription factor 6 gene (ATF6) on 1q21-q23 with type 2 diabetes in Pima Indians. Diabetes 55, 839-842 (2006).

114. Park, K. G. et al. Glucotoxicity in the INS-1 rat insulinoma cell line is mediated by the orphan nuclear receptor small heterodimer partner. Diabetes 56, 431-437 (2007).

115. Seo, H. Y. et al. Endoplasmic reticulum stress-induced activation of activating transcription factor 6 decreases insulin gene expression via up-regulation of orphan nuclear receptor small heterodimer partner. Endocrinology 149, 3832-3841 (2008).

116. Fonseca, S. G. et al. Wolfram syndrome 1 gene negatively regulates ER stress signaling in rodent and human cells. J Clin Invest 120, 744-755 (2010).

117. Yamada, T. et al. WFS1-deficiency increases endoplasmic reticulum stress, impairs cell cycle progression and triggers the apoptotic pathway specifically in pancreatic beta-cells. Hum Mol Genet 15, 1600-1609 (2006).

118. Ladiges, W. C. et al. Pancreatic beta-cell failure and diabetes in mice with a deletion mutation of the endoplasmic reticulum molecular chaperone gene P58IPK. Diabetes 54, 1074-1081 (2005).

119. Oyadomari, S. et al. Cotranslocational degradation protects the stressed endoplasmic reticulum from protein overload. Cell 126, 727-739 (2006).

120. Ozcan, U. et al. Endoplasmic reticulum stress links obesity, insulin action, and type 2 diabetes. Science 306, 457-461 (2004).

57

121. Ye, R. et al. Grp78 heterozygosity promotes adaptive unfolded protein response and attenuates diet-induced obesity and insulin resistance. Diabetes 59, 6-16 (2010).

122. Dorshkind, K. & Montecino-Rodriguez, E. Fetal B-cell lymphopoiesis and the emergence of B-1-cell potential. Nat Rev Immunol 7, 213-219 (2007).

123. Hardy, R. R. B-1 B cell development. J Immunol 177, 2749-2754 (2006).

124. Li, Y. S., Hayakawa, K. & Hardy, R. R. The regulated expression of B lineage associated genes during B cell differentiation in bone marrow and fetal liver. J Exp Med 178, 951-960 (1993).

125. Bikah, G., Carey, J., Ciallella, J. R., Tarakhovsky, A. & Bondada, S. CD5-mediated negative regulation of antigen receptor-induced growth signals in B-1 B cells. Science 274, 1906-1909 (1996).

126. Haas, K. M., Poe, J. C., Steeber, D. A. & Tedder, T. F. B-1a and B-1b cells exhibit distinct developmental requirements and have unique functional roles in innate and adaptive immunity to S. pneumoniae. Immunity 23, 7-18 (2005).

127. Shapiro-Shelef, M. & Calame, K. Regulation of plasma-cell development. Nat Rev Immunol 5, 230-242 (2005).

128. Martin, F. & Kearney, J. F. Marginal-zone B cells. Nat Rev Immunol 2, 323-335 (2002).

129. Attanavanich, K. & Kearney, J. F. Marginal zone, but not follicular B cells, are potent activators of naive CD4 T cells. J Immunol 172, 803-811 (2004).

130. Roark, J. H. et al. CD1.1 expression by mouse antigen-presenting cells and marginal zone B cells. J Immunol 160, 3121-3127 (1998).

131. Tanigaki, K. et al. Notch-RBP-J signaling is involved in cell fate determination of marginal zone B cells. Nat Immunol 3, 443-450 (2002).

132. Barber, C. L., Montecino-Rodriguez, E. & Dorshkind, K. Reduced production of B- 1-specified common lymphoid progenitors results in diminished potential of adult marrow to generate B-1 cells. Proc Natl Acad Sci U S A 108, 13700-13704 (2011).

133. Kikuchi, K. & Kondo, M. Developmental switch of mouse hematopoietic stem cells from fetal to adult type occurs in bone marrow after birth. Proc Natl Acad Sci U S A 103, 17852-17857 (2006).

134. Montecino-Rodriguez, E. & Dorshkind, K. B-1 B cell development in the fetus and adult. Immunity 36, 13-21 (2012).

58

135. de Andres, B. et al. The first 3 days of B-cell development in the mouse embryo. Blood 100, 4074-4081 (2002).

136. Montecino-Rodriguez, E., Leathers, H. & Dorshkind, K. Identification of a B-1 B cell-specified progenitor. Nat Immunol 7, 293-301 (2006).

137. Montecino-Rodriguez, E. & Dorshkind, K. Formation of B-1 B cells from neonatal B-1 transitional cells exhibits NF-kappaB redundancy. J Immunol 187, 5712-5719 (2011).

138. Casola, S. et al. B cell receptor signal strength determines B cell fate. Nat Immunol 5, 317-327 (2004).

139. Hardy, R. R., Carmack, C. E., Shinton, S. A., Kemp, J. D. & Hayakawa, K. Resolution and characterization of pro-B and pre-pro-B cell stages in normal mouse bone marrow. J Exp Med 173, 1213-1225 (1991).

140. Kondo, M., Weissman, I. L. & Akashi, K. Identification of clonogenic common lymphoid progenitors in mouse bone marrow. Cell 91, 661-672 (1997).

141. Matthias, P. & Rolink, A. G. Transcriptional networks in developing and mature B cells. Nat Rev Immunol 5, 497-508 (2005).

142. Lu, R., Medina, K. L., Lancki, D. W. & Singh, H. IRF-4,8 orchestrate the pre-B-to-B transition in lymphocyte development. Genes Dev 17, 1703-1708 (2003).

143. Nutt, S. L., Heavey, B., Rolink, A. G. & Busslinger, M. Commitment to the B- lymphoid lineage depends on the transcription factor Pax5. Nature 401, 556-562 (1999).

144. Lo, C. G., Lu, T. T. & Cyster, J. G. Integrin-dependence of lymphocyte entry into the splenic white pulp. J Exp Med 197, 353-361 (2003).

145. Allman, D. et al. Resolution of three nonproliferative immature splenic B cell subsets reveals multiple selection points during peripheral B cell maturation. J Immunol 167, 6834-6840 (2001).

146. Woodland, R. T., Schmidt, M. R. & Thompson, C. B. BLyS and B cell homeostasis. Semin Immunol 18, 318-326 (2006).

147. Schiemann, B. et al. An essential role for BAFF in the normal development of B cells through a BCMA-independent pathway. Science 293, 2111-2114 (2001).

148. Kaisho, T. et al. IkappaB kinase alpha is essential for mature B cell development and function. J Exp Med 193, 417-426 (2001).

59

149. Yamada, T. et al. Abnormal immune function of hemopoietic cells from alymphoplasia (aly) mice, a natural strain with mutant NF-kappa B-inducing kinase. J Immunol 165, 804-812 (2000).

150. Hardy, R. R., Hayakawa, K., Parks, D. R. & Herzenberg, L. A. Demonstration of B- cell maturation in X-linked immunodeficient mice by simultaneous three-colour immunofluorescence. Nature 306, 270-272 (1983).

151. Hikida, M., Johmura, S., Hashimoto, A., Takezaki, M. & Kurosaki, T. Coupling between B cell receptor and phospholipase C-gamma2 is essential for mature B cell development. J Exp Med 198, 581-589 (2003).

152. Samardzic, T. et al. Reduction of marginal zone B cells in CD22-deficient mice. Eur J Immunol 32, 561-567 (2002).

153. Pillai, S. & Cariappa, A. The follicular versus marginal zone B lymphocyte cell fate decision. Nat Rev Immunol 9, 767-777 (2009).

154. Pasparakis, M., Schmidt-Supprian, M. & Rajewsky, K. IkappaB kinase signaling is essential for maintenance of mature B cells. J Exp Med 196, 743-752 (2002).

155. Pappu, B. P. & Lin, X. Potential role of CARMA1 in CD40-induced splenic B cell proliferation and marginal zone B cell maturation. Eur J Immunol 36, 3033-3043 (2006).

156. Ruefli-Brasse, A. A., French, D. M. & Dixit, V. M. Regulation of NF-kappaB- dependent lymphocyte activation and development by paracaspase. Science 302, 1581-1584 (2003).

157. Xue, L. et al. Defective development and function of Bcl10-deficient follicular, marginal zone and B1 B cells. Nat Immunol 4, 857-865 (2003).

158. Oyama, T. et al. Mastermind-1 is required for Notch signal-dependent steps in lymphocyte development in vivo. Proc Natl Acad Sci U S A 104, 9764-9769 (2007).

159. Saito, T. et al. Notch2 is preferentially expressed in mature B cells and indispensable for marginal zone B lineage development. Immunity 18, 675-685 (2003).

160. Wu, L., Maillard, I., Nakamura, M., Pear, W. S. & Griffin, J. D. The transcriptional coactivator Maml1 is required for Notch2-mediated marginal zone B-cell development. Blood 110, 3618-3623 (2007).

161. Kuroda, K. et al. Regulation of marginal zone B cell development by MINT, a suppressor of Notch/RBP-J signaling pathway. Immunity 18, 301-312 (2003).

60

162. Iwakoshi, N. N. et al. Plasma cell differentiation and the unfolded protein response intersect at the transcription factor XBP-1. Nat Immunol 4, 321-329 (2003).

163. Shapiro-Shelef, M. et al. Blimp-1 is required for the formation of immunoglobulin secreting plasma cells and pre-plasma memory B cells. Immunity 19, 607-620 (2003).

164. Yoshida, H. et al. ATF6 activated by proteolysis binds in the presence of NF-Y (CBF) directly to the cis-acting element responsible for the mammalian unfolded protein response. Mol Cell Biol 20, 6755-6767 (2000).

165. Shaffer, A. L. et al. XBP1, downstream of Blimp-1, expands the secretory apparatus and other organelles, and increases protein synthesis in plasma cell differentiation. Immunity 21, 81-93 (2004).

166. Yeh, W. C. et al. Early lethality, functional NF-kappaB activation, and increased sensitivity to TNF-induced cell death in TRAF2-deficient mice. Immunity 7, 715-725 (1997).

167. Brunsing, R. et al. B- and T-cell development both involve activity of the unfolded protein response pathway. J Biol Chem 283, 17954-17961 (2008).

168. Nagasawa, T. Microenvironmental niches in the bone marrow required for B-cell development. Nat Rev Immunol 6, 107-116 (2006).

169. Bleul, C. C. et al. The lymphocyte chemoattractant SDF-1 is a ligand for LESTR/fusin and blocks HIV-1 entry. Nature 382, 829-833 (1996).

170. Oberlin, E. et al. The CXC chemokine SDF-1 is the ligand for LESTR/fusin and prevents infection by T-cell-line-adapted HIV-1. Nature 382, 833-835 (1996).

171. Jazin, E. E., Soderstrom, S., Ebendal, T. & Larhammar, D. Embryonic expression of the mRNA for the rat homologue of the fusin/CXCR-4 HIV-1 co-receptor. J Neuroimmunol 79, 148-154 (1997).

172. Moepps, B., Frodl, R., Rodewald, H. R., Baggiolini, M. & Gierschik, P. Two murine homologues of the human chemokine receptor CXCR4 mediating stromal cell- derived factor 1alpha activation of Gi2 are differentially expressed in vivo. Eur J Immunol 27, 2102-2112 (1997).

173. Nagasawa, T. et al. Defects of B-cell lymphopoiesis and bone-marrow myelopoiesis in mice lacking the CXC chemokine PBSF/SDF-1. Nature 382, 635- 638 (1996).

61

174. Zou, Y. R., Kottmann, A. H., Kuroda, M., Taniuchi, I. & Littman, D. R. Function of the chemokine receptor CXCR4 in haematopoiesis and in cerebellar development. Nature 393, 595-599 (1998).

175. Egawa, T. et al. The earliest stages of B cell development require a chemokine stromal cell-derived factor/pre-B cell growth-stimulating factor. Immunity 15, 323- 334 (2001).

176. Ma, Q., Jones, D. & Springer, T. A. The chemokine receptor CXCR4 is required for the retention of B lineage and granulocytic precursors within the bone marrow microenvironment. Immunity 10, 463-471 (1999).

177. Hargreaves, D. C. et al. A coordinated change in chemokine responsiveness guides plasma cell movements. J Exp Med 194, 45-56 (2001).

178. Tokoyoda, K., Egawa, T., Sugiyama, T., Choi, B. I. & Nagasawa, T. Cellular niches controlling B lymphocyte behavior within bone marrow during development. Immunity 20, 707-718 (2004).

179. Namen, A. E. et al. Stimulation of B-cell progenitors by cloned murine interleukin-7. Nature 333, 571-573 (1988).

180. Corcoran, A. E., Riddell, A., Krooshoop, D. & Venkitaraman, A. R. Impaired immunoglobulin gene rearrangement in mice lacking the IL-7 receptor. Nature 391, 904-907 (1998).

181. Opferman, J. T. et al. Development and maintenance of B and T lymphocytes requires antiapoptotic MCL-1. Nature 426, 671-676 (2003).

182. Kikuchi, K., Lai, A. Y., Hsu, C. L. & Kondo, M. IL-7 receptor signaling is necessary for stage transition in adult B cell development through up-regulation of EBF. J Exp Med 201, 1197-1203 (2005).

183. Peschon, J. J. et al. Early lymphocyte expansion is severely impaired in interleukin 7 receptor-deficient mice. J Exp Med 180, 1955-1960 (1994).

184. von Freeden-Jeffry, U. et al. Lymphopenia in interleukin (IL)-7 gene-deleted mice identifies IL-7 as a nonredundant cytokine. J Exp Med 181, 1519-1526 (1995).

185. Dias, S., Silva, H. J., Cumano, A. & Vieira, P. Interleukin-7 is necessary to maintain the B cell potential in common lymphoid progenitors. J Exp Med 201, 971-979 (2005).

186. Lyman, S. D. et al. Molecular cloning of a ligand for the flt3/flk-2 tyrosine kinase receptor: a proliferative factor for primitive hematopoietic cells. Cell 75, 1157-1167 (1993).

62

187. Lyman, S. D. & Jacobsen, S. E. c-kit ligand and Flt3 ligand: stem/progenitor cell factors with overlapping yet distinct activities. Blood 91, 1101-1134 (1998).

188. Sitnicka, E. et al. Key role of flt3 ligand in regulation of the common lymphoid progenitor but not in maintenance of the hematopoietic stem cell pool. Immunity 17, 463-472 (2002).

189. Matthews, W., Jordan, C. T., Wiegand, G. W., Pardoll, D. & Lemischka, I. R. A receptor tyrosine kinase specific to hematopoietic stem and progenitor cell- enriched populations. Cell 65, 1143-1152 (1991).

190. Wasserman, R., Li, Y. S. & Hardy, R. R. Differential expression of the blk and ret tyrosine kinases during B lineage development is dependent on Ig rearrangement. J Immunol 155, 644-651 (1995).

191. Mackarehtschian, K. et al. Targeted disruption of the flk2/flt3 gene leads to deficiencies in primitive hematopoietic progenitors. Immunity 3, 147-161 (1995).

192. Sitnicka, E. et al. Complementary signaling through flt3 and interleukin-7 receptor alpha is indispensable for fetal and adult B cell genesis. J Exp Med 198, 1495- 1506 (2003).

193. Nakashima, T. et al. Evidence for osteocyte regulation of bone homeostasis through RANKL expression. Nat Med 17, 1231-1234 (2011).

194. Dougall, W. C. et al. RANK is essential for osteoclast and lymph node development. Genes Dev 13, 2412-2424 (1999).

195. Kong, Y. Y. et al. OPGL is a key regulator of osteoclastogenesis, lymphocyte development and lymph-node organogenesis. Nature 397, 315-323 (1999).

196. Takeda, S., Shimizu, T. & Rodewald, H. R. Interactions between c-kit and stem cell factor are not required for B-cell development in vivo. Blood 89, 518-525 (1997).

197. Russell, E. S. Hereditary anemias of the mouse: a review for geneticists. Adv Genet 20, 357-459 (1979).

198. Waskow, C., Paul, S., Haller, C., Gassmann, M. & Rodewald, H. R. Viable c- Kit(W/W) mutants reveal pivotal role for c-kit in the maintenance of lymphopoiesis. Immunity 17, 277-288 (2002).

199. Sugiyama, T., Kohara, H., Noda, M. & Nagasawa, T. Maintenance of the hematopoietic stem cell pool by CXCL12-CXCR4 chemokine signaling in bone marrow stromal cell niches. Immunity 25, 977-988 (2006).

63

200. Omatsu, Y. et al. The essential functions of adipo-osteogenic progenitors as the hematopoietic stem and progenitor cell niche. Immunity 33, 387-399 (2010).

201. Chu, V. T. et al. Eosinophils are required for the maintenance of plasma cells in the bone marrow. Nat Immunol 12, 151-159 (2011).

202. Winter, O. et al. Megakaryocytes constitute a functional component of a plasma cell niche in the bone marrow. Blood 116, 1867-1875 (2010).

203. Sapoznikov, A. et al. Perivascular clusters of dendritic cells provide critical survival signals to B cells in bone marrow niches. Nat Immunol 9, 388-395 (2008).

204. Zhu, J. et al. Osteoblasts support B-lymphocyte commitment and differentiation from hematopoietic stem cells. Blood 109, 3706-3712 (2007).

205. Calvi, L. M. et al. Osteoblastic cells regulate the haematopoietic stem cell niche. Nature 425, 841-846 (2003).

206. Wu, J. Y. et al. Osteoblastic regulation of B lymphopoiesis is mediated by Gs{alpha}-dependent signaling pathways. Proc Natl Acad Sci U S A 105, 16976- 16981 (2008).

207. Visnjic, D. et al. Hematopoiesis is severely altered in mice with an induced osteoblast deficiency. Blood 103, 3258-3264 (2004).

208. Mansour, A. et al. Osteoclast activity modulates B-cell development in the bone marrow. Cell Res 21, 1102-1115 (2011).

209. Wei, J., Sheng, X., Feng, D., McGrath, B. & Cavener, D. R. PERK is essential for neonatal skeletal development to regulate osteoblast proliferation and differentiation. J Cell Physiol 217, 693-707 (2008).

210. Yang, X. et al. ATF4 is a substrate of RSK2 and an essential regulator of osteoblast biology; implication for Coffin-Lowry Syndrome. Cell 117, 387-398 (2004).

211. Yu, S. et al. Critical role of activating transcription factor 4 in the anabolic actions of parathyroid hormone in bone. PLoS One 4, e7583 (2009).

212. Zhang, X. et al. Activating transcription factor 4 is critical for proliferation and survival in primary bone marrow stromal cells and calvarial osteoblasts. J Cell Biochem 105, 885-895 (2008).

213. Saito, A. et al. Endoplasmic reticulum stress response mediated by the PERK- eIF2(alpha)-ATF4 pathway is involved in osteoblast differentiation induced by BMP2. J Biol Chem 286, 4809-4818 (2011).

64

214. Jang, W. G. et al. BMP2 protein regulates osteocalcin expression via Runx2- mediated Atf6 gene transcription. J Biol Chem 287, 905-915 (2012).

215. Kondo, S. et al. OASIS, a CREB/ATF-family member, modulates UPR signalling in astrocytes. Nat Cell Biol 7, 186-194 (2005).

216. Murakami, T. et al. Signalling mediated by the endoplasmic reticulum stress transducer OASIS is involved in bone formation. Nat Cell Biol 11, 1205-1211 (2009).

217. Tohmonda, T. et al. The IRE1alpha-XBP1 pathway is essential for osteoblast differentiation through promoting transcription of Osterix. EMBO Rep 12, 451-457 (2011).

65

CHAPTER II

Deficiency of ERdj4 is associated with perinatal lethality,

constitutive ER stress and glucose intolerance

66 Deficiency of ERdj4 is associated with perinatal lethality, constitutive ER stress and glucose intolerance

Jill M. Fritz*, Mei Dong*, Emily P. Martin, Cheng-Lun Na and Timothy E. Weaver

Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, and the

University of Cincinnati College of Medicine, Cincinnati, OH 45229

Correspondence: Timothy E. Weaver, 3333 Burnet Ave., Cincinnati, OH 45229; Phone:

513-636-7223; Email: [email protected]

Supported by NIH awards HL103923 and HL086492

*These authors contributed equally to this study

Manuscript in preparation

67 Abstract

ERdj4 maintains homeostasis during conditions of ER stress by facilitating

removal of unfolded/misfolded substrates from the ER lumen for degradation by the

proteasome. To elucidate the function of ERdj4 in vivo, the ERdj4 allele was disrupted

by a gene trap (ERdj4+/GT) leading to hypomorphic expression of the encoded protein in

mice homozygous for the trapped allele. ERdj4GT/GT mice exhibited significant perinatal lethality associated with growth retardation and hypoglycemia. Immature islets and reduced glucagon and insulin in pancreas of newborn ERdj4GT/GT mice indicated a delay in endocrine pancreatic development. Surviving adult mice exhibited constitutive ER stress in both non-secretory and secretory cell types and an increased susceptibility to cell death following induction of ER stress. ERdj4 deficiency resulted in altered morphology of pancreatic islets including cytoplasmic vacuolation and α-cell hyperplasia. Pancreatic β-cells from ERdj4GT/GT mice exhibited evidence of ER stress

including ER dilation and increased expression of spliced XBP1. Accumulation of

proinsulin in the ER and an increased number of immature granules in β-cells from

ERdj4GT/GT mice indicated a defect in the insulin secretory pathway. In addition to

insulin, ERdj4 associated with the insulin processing enzymes Pcsk1, Pcsk2 and CPE.

ERdj4GT/GT mice were glucose intolerant, resulting from hypoinsulinemia. Exogenous insulin effectively lowered blood glucose levels consistent with a disruption in the insulin secretory pathway. Together, these results indicate that the chaperone activity of ERdj4 is required for normal growth, development, and metabolism.

68 Introduction

Nascent proteins destined for the cell surface, intracellular organelles or

secretion enter the endoplasmic reticulum (ER) where they undergo chaperone-

mediated protein folding to achieve a stable conformation. Highly secretory cells,

including pancreatic acinar and β-cells, plasma B cells, and serous and mucous cells of

the salivary gland, invoke the unfolded protein response (UPR) to increase ER folding

capacity and maintain homeostasis in the face of increased protein load and

consequent ER stress1-4. The UPR reduces ER burden by attenuating protein translation and upregulating the machinery involved in protein folding and ER-

associated degradation (ERAD). The ER transmembrane sensors, protein kinase RNA

(PKR)-like ER kinase (PERK), activating transcription factor 6 (ATF6) and inositol

requiring kinase 1 (IRE1) detect accumulation of unfolded or misfolded protein in the ER

lumen and transduce signals across the ER membrane to alleviate ER stress. PERK

kinase activation results in phosphorylation of eukaryotic translation initiation factor-2

(eIF2α), which inhibits translation initiation, thereby reducing the load of newly

synthesized proteins within the ER. Activation of ATF6 results in relocation to the Golgi,

proteolytic cleavage and release of a cytosolic ATF6 fragment that translocates to the

nucleus to activate transcription of UPR target genes including ER chaperones. The

activated endoribonuclease domain of IRE1 removes an unspliced intron from X-box

binding protein-1 (XBP1) mRNA leading to translation of a transcription factor that

upregulates chaperones involved in protein folding as well as components of the ERAD

machinery. Failure of the UPR to resolve ER stress can trigger apoptosis5,6.

69 Molecular chaperones are critical components of the ER quality control

machinery that distinguish between unfolded, correctly folded and terminally misfolded proteins within the ER. BiP, an ER-localized member of the Hsp70 family, is a multifunctional chaperone that assists in the folding of nascent proteins7,8, facilitates

targeting of misfolded proteins for proteasomal degradation9,10 and plays a key role in activating the UPR in response to ER stress11,12. BiP contains a C-terminal substrate- binding domain (SBD) that binds unfolded substrates with low affinity via interaction with exposed hydrophobic regions. Hydrolysis of ATP bound to the N-terminal nucleotide- binding domain (NBD) of BiP results in high affinity binding of the SBD with substrate.

Exchange of ADP for ATP releases the unfolded substrate from BiP allowing the substrate to continue folding13. A critical arginine residue located on the surface of the

NBD in the ATP-bound form is necessary for communication with the SBD, as well as

interaction with cochaperones of the DnaJ family14,15.

ER-localized DnaJ (ERdj) homologues induce high affinity substrate binding by

interacting with the NBD of BiP and stimulating ATP hydrolysis. While some ERdjs

interact with BiP independently of substrate, other ERdjs bind substrates directly and

recruit BiP16-19. ERdjs facilitate protein folding and/or degradation of newly translocated

(ERdj1-3), unfolded (ERdj3 and ERdj6) or misfolded (ERdj4 and ERdj5) substrates,

often by interacting with translocation or ERAD machinery20. ERdj4 is a soluble type II

DnaJ protein that binds BiP through a His-Pro-Asp (HPD) motif in the J domain and likely binds directly to misfolded substrates through a C-terminal glycine/phenylalanine- rich region21-23. ERdj4 is ubiquitously expressed at very low levels24,25, but is highly upregulated in response to ER stress21,23 by the UPR signal transducer XBP126,27.

70 ERdj4 binds to unfolded (insulin2WT) or misfolded (insulin2C96Y, SP-CΔexon4 and SP-

CL188Q) substrates to facilitate their removal from the ER for degradation by the proteasome21,22. In the current study, we report that deficiency of ERdj4 is associated with decreased survival, growth retardation and delayed endocrine pancreatic development in newborn mice, and hypoinsulinemia and glucose intolerance in adult

mice. Consistent with these findings, constitutive ER stress was observed in a variety

of tissues and cell types in mutant mice. Taken together, these results reveal an

important and unexpected role for ERdj4 in adaptation to ER stress associated with

growth, development and metabolism.

71 Results

Generation of ERdj4 gene trap mice⎯Since previous studies demonstrated significant expression of ERdj4 in secretory tissues23, we hypothesized that the chaperone activity

of ERdj4 would be important for the homeostasis and function of secretory cells in vivo.

Chimeric mice were generated from embryonic stem (ES) cells harboring a gene trap

(GT) cassette inserted into the ERdj4 locus (Fig. 1A). Offspring carrying the GT allele

were backcrossed eight generations to the 129P2 or C57BL/6 strains. Tail genomic

DNA (gDNA) isolated from the progeny of ERdj4+/GT intercrosses was used to confirm the presence of WT and/or GT alleles (Fig. 1B). Only one or two copies of the GT cassette were present in ERdj4+/GT or ERdj4GT/GT mice, respectively, confirming that

ERdj4 was the only locus that was disrupted (Fig. 1C). Insertion of the GT cassette into intron 1 of the ERdj4 locus (confirmed by sequencing) resulted in decreased expression of ERdj4 mRNA (Fig. 1D) and a corresponding increase in beta-galactosidase mRNA from the GT cassette (Fig. 1E) in MEFs isolated from E13.5 ERdj4GT/GT mice. Variable expression of ERdj4 mRNA in tissues from adult mice indicated that the insertion of the

GT cassette resulted in a hypomorphic allele (Fig. 1F).

Elevated ER stress and increased susceptibility to cell death in ERdj4GT/GT MEFs⎯To

determine if ERdj4 deficiency enhances ER stress, splicing of XBP1 mRNA was

assessed in control and tunicamycin-treated MEFs isolated from ERdj4+/+ and

ERdj4GT/GT mice. The spliced form of XBP1 mRNA was elevated at baseline in

ERdj4GT/GT MEFs compared to ERdj4+/+ controls (Fig. 2A, 0 hrs). Following treatment

with tunicamycin, which induced ER stress by inhibiting N-linked glycosylation, splicing

of XBP1 mRNA was enhanced in both ERdj4GT/GT and ERdj4+/+ MEFs (Fig. 2A).

72 Consistent with constitutive ER stress in ERdj4GT/GT MEFs, BiP and IRE-1α proteins

were elevated at baseline (Fig. 2B, 0 hrs). Treatment with tunicamycin or the proteasome inhibitor MG-132 increased the expression of BiP and IRE-1α in both

ERdj4GT/GT and control MEFs, but the effect was clearly more pronounced in ERdj4GT/GT

MEFs. In contrast, the concentration of another ER chaperone, calnexin, was similarly

regulated in control and ERdj4GT/GT MEFs under both unstressed and stressed

conditions (Fig. 2B). Ultrastructural analyses of ERdj4GT/GT MEFs revealed ER

distention and large Golgi inclusions providing further evidence of constitutive ER stress

(Fig. 2C). In the absence of exogenous stressors, survival of ERdj4GT/GT MEFs and control cells was similar (Fig. 2D). However, when ERdj4GT/GT MEFs were treated with

MG-132, tunicamycin or thapsigargin (an inhibitor of calcium transport) for 48 hours, cell viability was significantly decreased in a dose-dependent manner compared to ERdj4+/+

controls (Fig. 2D). In keeping with this finding, overexpression of ERdj4 was previously

reported to protect against cell death induced by ER stress28.

Hypoglycemia and growth retardation in ERdj4GT/GT mice⎯Perinatal lethality was

observed in ERdj4GT/GT progeny derived from ERdj4+/GT intercrosses of 129P2xC57BL/6,

129P2 and C57BL/6 strains (Table 1). Genotype distribution at E18.5 approximated

Mendelian ratios indicating that lethality resulting from homozygosity of the GT allele

occurred perinatally (Table 2). ERdj4GT/GT embryos were smaller (Fig. 3A) and E18.5 body weights were significantly decreased compared to control animals (Fig. 3B).

Analysis of blood glucose levels in newborn mice indicated that ERdj4GT/GT mice were

severely hypoglycemic (Fig. 3C), something that likely contributed to death. Pancreatic

islets in newborn ERdj4GT/GT mice formed immature cord-like structures in contrast to

73 spherical islets observed in ERdj4+/+ mice (Fig. 3D)29. Glucagon and insulin were also significantly decreased in pancreatic tissues of ERdj4GT/GT neonates (Fig. 3E-F),

providing further evidence of delayed endocrine pancreatic development. Surviving

male and female mice often displayed signs of a vestibular disorder, which included a

head tilt accompanied by abnormal circling behavior and hyperactivity. Surviving

female ERdj4GT/GT mice were infertile. All subsequent experiments were conducted using littermates derived from heterozygous crosses in the mixed 129P2xC57BL/6 genetic background, unless otherwise indicated.

Constitutive ER stress is associated with histological abnormalities in pancreatic islets of ERdj4GT/GT mice⎯In order to determine if ERdj4 deficiency enhanced ER stress in vivo, ERdj4+/GT mice were crossed to ER stress-activated indicator (ERAI) transgenic mice30. GFP protein (resulting from XBP1 splicing and expression of XBP1/GFP fusion

protein) was detected in multiple organs including the lungs and kidneys of

ERdj4GT/GTERAI+ mice (Fig. S1A). Ultrastructural analyses revealed ER distention and

Golgi inclusions in renal tubular epithelial cells (Fig. S1B) and serous cells of the

salivary gland (Fig. S1C). GFP and IRE-1α proteins were also increased in pancreatic

lysates from ERdj4GT/GTERAI+ mice compared to ERdj4+/+ERAI+ controls (Fig. 4A). GFP

immunofluorescence localized to β-cells in the islets of pancreatic sections from both

ERdj4+/+ERAI+ and ERdj4GT/GTERAI+ mice (Fig. 4B), with more GFP-positive cells and

increased fluorescence intensity in mutant mice. GFP fluorescence was not detected in pancreatic α-cells of ERdj4+/+ERAI+ or ERdj4GT/GTERAI+ mice (Fig. S2A) although ultrastructural evidence of ER stress was detected (Fig. S2B). Histological analyses of pancreatic sections from ERdj4GT/GT mice identified vacuolated cells in the islets (Fig.

74 4C). Ultrastructural analyses of islets revealed ER distention, enlarged mitochondria

and Golgi inclusions in both α- and β-cells of ERdj4GT/GT mice (Fig. S2B, S3A). An increased number of glucagon-positive α-cells were detected in the mantle and core of

islets from ERdj4GT/GT mice compared to controls (Fig. 4D and S2A). Glucagon protein was significantly increased in pancreatic extracts of adult ERdj4GT/GT mice (Fig. 4E)

consistent with α-cell hyperplasia and increased glucagon granules (Fig. 4D and S2B).

Further, plasma glucagon was significantly increased in ERdj4GT/GT mice compared to

controls (Fig. 4F). Hypomorphic expression of ERdj4 mRNA was confirmed in islets

isolated from ERdj4GT/GT mice (Fig. 4G), linking the loss of ERdj4 in pancreatic islets to

elevated ER stress.

While histology of ERdj4GT/GT acinar cells appeared normal at 8 weeks of age

(Fig. S4A), cytoplasmic vacuolation was observed in pancreatic acinar cells from 8-

month-old ERdj4GT/GT mice (S4B). Ultrastructural analyses of pancreatic acinar cells revealed dilated ER and multiple immature zymogen granules in ERdj4GT/GT mice (Fig.

S4C), although this did not appear to interfere with acinar cell function, as reflected by similar serum lipase and amylase levels in ERdj4GT/GT and control mice (Fig. S4D).

ERdj4 deficiency impairs insulin biosynthesis⎯Since ERdj4 was previously reported to

associate with insulin21, we assessed whether ERdj4 deficiency caused β-cell stress by

increasing proinsulin retention in the ER. Although total insulin protein in pancreatic

extracts of ERdj4GT/GT mice was normal (Fig. S3B), proinsulin and BiP colocalization was enhanced in pancreatic β-cells, indicating accumulation of proinsulin in the ER (Fig.

5A). Metabolic labeling of islets isolated from ERdj4GT/GT mice showed a modest increase in proinsulin protein compared to controls (Fig. 5B); however, proinsulin was

75 likely underrepresented due to significant α-cell hyperplasia in ERdj4GT/GT islets. To

determine whether ERdj4 associates with components of the insulin/glucagon secretory

pathways, HEK293 cells were cotransfected with ERdj4 and Pcsk1, Pcsk2, glucagon or

CPE. In contrast to insulin, ERdj4 did not associate with glucagon (Fig. 5C), but

consistently formed complexes with the insulin processing enzymes Pcsk1, Pcsk2 and

CPE (Fig. 5C), which generate mature insulin in secretory granules. Ultrastructural analyses of β-cells in ERdj4GT/GT mice revealed an increase in translucent (immature)

insulin granules (Fig. S3A), suggesting a disruption in proinsulin processing by Pcsk1,

Pcsk2 and/or CPE.

Hypoinsulinemia causes glucose intolerance in ERdj4GT/GT mice⎯Since increased ER stress and defective proinsulin processing were observed in β-cells of ERdj4GT/GT mice, endocrine pancreatic function was evaluated by measuring blood glucose and insulin after fasting. Although basal blood glucose levels were normal (Fig. 6A), glucose remained elevated in ERdj4GT/GT mice following glucose administration (Fig. 6B).

Importantly, insulin effectively lowered blood glucose levels in ERdj4GT/GT mice,

suggesting that ERdj4 deficiency leads to a decrease in mature insulin rather than a

defect in the insulin signaling pathway (Fig. 6C). Plasma insulin was significantly

decreased at baseline in ERdj4GT/GT mice (Fig. 6D, 0 min), but did not affect basal blood glucose levels (Fig. 6A). Following glucose administration, plasma insulin increased slightly in ERdj4GT/GT mice, but remained significantly lower than controls (Fig. 6D, 15

and 30 min). Plasma proinsulin:insulin ratios were also significantly increased in

ERdj4GT/GT mice (Fig. 6E), providing further evidence of a disruption in proinsulin processing coincident with ER stress.

76 Discussion

ERdj4 is a soluble ER chaperone protein expressed at low levels in a wide range of cells22-24. In response to ER stress, expression of ERdj4 is rapidly upregulated to assist in the disposal of an as yet poorly defined subset of misfolded proteins via ERAD21,23.

In this study, we showed that hypomorphic expression of ERdj4 in ERdj4GT/GT mice

resulted in perinatal lethality associated with growth retardation and hypoglycemia.

Immature islets and decreased glucagon and insulin in the pancreas of newborn

ERdj4GT/GT mice indicated a delay in endocrine pancreatic development. Constitutive

ER stress was observed in multiple cell types and tissues of surviving ERdj4GT/GT mice

in association with accumulation of immature granules in secretory cells and increased

susceptibility to cell death. These results strongly suggest that ERdj4 plays an

important role in ER homeostasis beyond facilitating disposal of terminally misfolded

proteins. Consistent with this hypothesis, we previously reported that ERdj4 interacts

with wild-type insulin21; in keeping with this observation, insulin accumulated in the ER of β-cells from ERdj4GT/GT mice. A disruption in the insulin secretory pathway elevated

ER stress leading to hypoinsulinemia and glucose intolerance in ERdj4GT/GT mice.

Collectively, these results demonstrate an important and unanticipated role for ERdj4 in

adaptation to ER stress associated with growth, development and metabolism.

Hypomorphic expression of ERdj4 resulted in normal Mendelian distribution of

genotypes at E18.5; in contrast, survival of newborn ERdj4GT/GT pups was significantly

decreased in a strain-dependent manner. Neonatal lethality can arise from defects in

respiration, suckling or metabolism31. ERdj4GT/GT pups did not display overt signs of

respiratory distress and contained milk in their stomachs, consistent with normal

77 respiration and suckling. However, newborn ERdj4GT/GT pups were significantly smaller

and severely hypoglycemic, suggesting that a defect in glucose homeostasis

contributed to lethal nutrient deprivation in the perinatal period. Immature islet

morphology and reduced levels of insulin and glucagon indicated a delay in endocrine

pancreatic development in newborn ERdj4GT/GT mice. Overall, the loss of perinatal

ERdj4 expression was strongly associated with neonatal demise, although the molecular pathway(s) underlying the perinatal survival function of this chaperone remains unclear.

Adult ERdj4GT/GT mice exhibited glucose intolerance resulting from

hypoinsulinemia. Proinsulin accumulated in the ER consistent with ERdj4 associating with insulin and the insulin processing enzymes Pcsk1, Pcsk2 and CPE. Notably, immature insulin granules and plasma proinsulin:insulin ratio were increased suggesting that, in addition to proinsulin, ERdj4 may also facilitate maturation of insulin processing enzymes. A similar phenotype (hypoinsulinemia and increased proinsulin:insulin ratio) was recently described for mice in which XBP1 was selectively deleted in β-cells32.

Dysregulation of proinsulin processing was attributed to degradation of mRNAs

encoding insulin processing enzymes as well as loss of chaperones, including ERdj4.

Thus, retention of proinsulin and possibly its processing enzymes may contribute to β-

cell stress and, ultimately hypoinsulinemia.

In contrast to newborn mice, glucagon was significantly elevated in pancreatic

tissues and plasma of ERdj4GT/GT mice in association with α-cell hyperplasia and ultrastructural evidence of increased glucagon granules. It remains unclear whether

ERdj4 deficiency disrupts proglucagon processing in α-cells. In contrast to insulin,

78 ERdj4 did not associate with glucagon; however, it did interact with Pcsk2 and CPE,

which are responsible for proglucagon processing in α-cells33. To date, we have not

identified any metabolic derangements arising from altered α-cell structure/function in

adult ERdj4GT/GT mice. However, a recent study revealed a possible explanation for α-

cell hyperplasia in ERdj4GT/GT mice by demonstrating that increased metabolic stress

caused β-cells to dedifferentiate into progenitor cells that subsequently adopted the α-

cell fate34.

ERdj4 can directly bind client proteins21, and recruit BiP to promote substrate solubilization and/or folding23. Several lines of evidence previously suggested that

ERdj4 is an ERAD-specific chaperone. First, expression of ERdj4 is normally low and rapidly upregulated in response to ER stress23,28, including stress resulting from

expression of a terminally misfolded protein21. Second, ERdj4 exhibited prolonged association with misfolded SP-C but not with the corresponding wild-type protein21.

Third, misfolded SP-C co-precipitated in a complex containing ERdj4 and the cytosolic

ATPase p97/VCP21. Fourth, ERdj4 was shown to transiently associate with derlin-122.

Taken together, these results suggest that ERdj4 may function in cooperation with the

retrotranslocation machinery late in the ERAD pathway. The results of the current

study, demonstrating widespread ER stress in cells/tissues of ERdj4GT/GT mice, were therefore unexpected. One possible explanation is that ERdj4 associates with proteins that are abnormally prone to misfolding and/or fold more slowly than other substrates, leading to prolonged ERdj4/client interaction and elimination via ERAD under conditions of stress. Alternatively, it is possible that ERdj4 is required for productive folding of

79 some wild-type substrates as well as elimination of unfolded/misfolded substrates from

the ER lumen.

Diabetes pathogenesis has been linked to failure of the UPR to properly manage

ER stress, resulting in β-cell failure or insulin resistance in peripheral tissues. The UPR

is mediated by ER stress sensors that maintain ER homeostasis by attenuating protein

translation (PERK/eIF2α) and upregulating expression of chaperones to assist in protein

folding, maturation and disposal of misfolded substrates via ERAD (IRE-1α/XBP1 and

ATF6)5,6. Mutations in PERK and ATF6α are associated with disrupted glucose homeostasis and diabetes in humans35-39. Furthermore, deficiency of XBP1, PERK or

ERdj6/p58IPK (an XBP1 target gene and ERdj/HSP40 family member) resulted in spontaneous diabetes in mice due to a failure in development and/or function of β- cells3,32,40-42. In this study, we showed that decreased expression of another XBP1

target gene and HSP40 family member, ERdj4, caused glucose intolerance associated

with increased ER stress in pancreatic β-cells. Administration of insulin effectively

reduced blood glucose levels in ERdj4GT/GT mice, indicating that ERdj4 deficiency did

not impair insulin signaling or glucose uptake in peripheral tissues and confirming a

pathologic role for β-cell dysfunction in these mice. Further, increased expression of

sXBP1 was localized to β-cells of ERdj4GT/GT mice, similar to findings in pancreatic islets of type 2 diabetic patients and diabetic mouse models43-45. Overall, ERdj4 plays an important role in β-cell homeostasis by promoting maturation of insulin and possibly insulin processing enzymes. Given the evidence of constitutive ER stress in other cells and organs of ERdj4GT/GT mice, it is likely that the chaperone activity of ERdj4 also

extends to a broad range of unfolded protein substrates.

80 Materials and Methods

Mice. ES cells harboring a gene trap (GT) cassette inserted into the ERdj4 locus (Bay

Genomics, ES cell line KST256) were purchased from the Mutant Mouse Regional

Resource Center. GT ES cells were injected into blastocysts to generate chimeras at

the University of Cincinnati Gene Targeting Facility. Animals were backcrossed eight

generations onto the 129P2 or C57BL/6 genetic backgrounds. With the exception of

perinatal lethality studies, all experiments in this study used 129P2×C57BL/6

littermates. The presence of the GT cassette was confirmed by PCR using primers

designed for WT and GT alleles (WT: 5’-AAG CCC AGT AAT AAC CCA ACT TAT C-3’;

GT: 5’-TGC TGG AGT CTA GCT ACT TAT CCA C-3’; WT/GT: 5’-CTG TCT TTT CTG

TGT TTG GCT AGA A-3’). ERAI mice were provided by RIKEN BRC and the presence

of the ERAI transgene was confirmed by PCR using primers designed by the supplier

(5’-GAA CCA GGA GTT AAG ACA GC-3’; 3’-GAA CAG CTC CTC GCC CTT GC-5’).

All mice were housed in a pathogen-free barrier facility with free access to food and

water. Animal procedures were performed under protocols approved by Cincinnati

Children’s Hospital Medical Center Institutional Animal Care and Use Committee.

RNA Isolation and RT-PCR. Total RNA was isolated from cells and tissues using the

RNeasy Plus Mini Kit (Qiagen) and cDNA was synthesized using iScript cDNA

Synthesis Kit (Bio-Rad). XBP1 was detected in MEFs by standard RT-PCR using

XBP1-specific primers (5’-GTG GTT GAG AAC CAG GAG TTA AGA-3’; 3’-AGA ATC

TGA AGA GGC AAC AGT GTC-3’). PCR products were separated on 4% agarose gels and visualized by ethidium bromide staining. Quantitative RT-PCR was performed with

25 ng of cDNA per reaction on the ABI 7300 system with TaqMan Assays (Applied

81 Biosystems) for mouse ERdj4 (Mm01622956_s1), a custom-designed beta-

galactosidase/neomycin resistance region of the GT cassette and mouse β-actin

(endogenous control, Mm00607939_s1). Relative quantitation was assessed using the

SDS Software (Applied Biosystems).

Gene Copy Number Assay. Tail genomic DNA (gDNA, 20 ng) was prepared in

quadruplicate as recommended by Applied Biosystems. The reaction mix included a

custom-designed TaqMan Copy Number Assay targeted for the beta-galactosidase

region of the GT cassette and a TaqMan Copy Number Reference Assay specific for

the mouse transferrin receptor (Applied Biosystems). Absolute quantitation was performed with the ABI 7300 system followed by gene copy number analysis with SDS and CopyCaller software (Applied Biosystems).

Immunoblot Analysis. Tissue lysate preparation and immunoblot analyses were performed as previously described46. Briefly, tissue was harvested and homogenized in

RIPA buffer (Teknova) containing protease inhibitor cocktail (Sigma). Homogenates were centrifuged at 5,000 g for 10 minutes to clear tissue debris and protein

concentration in the supernatants was assessed using the Micro BCA Kit (Pierce).

Lysates were analyzed by SDS-polyacrylamide gels under reducing conditions,

transferred to nitrocellulose membranes and immunoblotted with antibodies specific for

IRE-1α (Cell Signaling), GFP (Invitrogen), BiP (Cell Signaling), calnexin (Cell Signaling),

and β-actin (Seven Hills Bioreagents).

Cell Culture, Transfection and Coimmunoprecipitation. MEFs were isolated from

ERdj4+/+ and ERdj4GT/GT embryos on day 13.5 as described by Conner47. Cells were

cultured in DMEM with 10% fetal bovine serum, 100 U/ml penicillin, 100 µg/ml

82 streptomycin and 200 mM L-glutamine and passaged at least three times before use in

experiments. Cell viability was assessed (CellTiter 96 AQueous One Solution Cell

Proliferation Assay, Promega) following treatment with tunicamycin (CalBiochem), thapsigargin (CalBiochem) or MG-132 (CalBiochem) for 48 hours.

Human ERdj4-HA was cloned and inserted into a pIRES-EGFP vector

(Clontech), as previously described21. Human Pcsk1 (NM_000439), Pcsk2

(NM_002594), GCG (NM_002054) and CPE (NM_001873) containing C-terminal Myc-

DDK (FLAG) tags in a pCMV6-Entry vector were purchased from OriGene. HEK293 cells per 10cm plate were transfected with 1ug of each construct using Lipofectamine

2000 (Invitrogen) per manufacturer recommendations. Cells were harvested 48 hours after transfection in 50mM Tris, 150 mM NaCl and 1% Triton X-100 with 1X mammalian protease inhibitors (Sigma). Cell lysates were briefly sonicated, centrifuged at 20,000 g for 10 minutes and quantitated using the Direct Detect system (Millipore). Cell lysates were immunoprecipitated with α-FLAG M2 Affinity Gel (Sigma) and eluted proteins were analyzed by SDS-PAGE under reducing electrophoretic conditions followed by Western blotting with antibodies directed against HA (Santa Cruz).

Histology and Electron Microscopy. Tissues were fixed in 4% paraformaldehyde, embedded in paraffin and 5µm sections were cut for H&E staining and immunofluorescence as previously described46. For immunofluorescence, slides underwent antigen retrieval by boiling in 10mM citrate buffer (pH 6.0) for 20 minutes followed by blocking in normal donkey serum (Jackson ImmunoResearch) for two hours. Sections were stained with guinea pig anti-insulin (ab7842, Abcam), rabbit anti- glucagon (ab932, Millapore) and anti-chicken GFP (ab13970, Abcam) followed by

83 application of secondary antibodies conjugated to Alexa Fluor 488 or Alexa Fluor 594

(Invitrogen). Image stacks were acquired by 0.25 µm step distance using a Nikon C1 Si

Confocal Imaging System and processed for deconvolution to remove image blurring

using Autoquant X 2.2 (Media Cybernetics).

For transmission electron microscopy, cells and tissues were processed as previously described48. Briefly, samples were fixed in 2% paraformaldehyde (Electron

Microscopy Sciences ‘EMS’), 2% glutaraldehyde (EMS) and 0.1% calcium chloride in

0.1 M sodium cacodylate buffer (EMS), pH 7.3 for 30 min, followed by transfer to fresh fixative at 4°C overnight. Fixed cells and tissues were cut into 1-2 mm blocks, incubated with 1% aqueous osmium tetroxide (EMS) and 1.5% potassium ferrocyanide

(Sigma) in 0.1M sodium cacodylate buffer, pH 7.3 for 2 hrs, stained en bloc with 4% aqueous ureanyl acetate (EMS) at 4 °C overnight, dehydrated with a graded series of alcohol and embedded in expoxy resin EMbed 812 (EMS) or Quetol 651 (EMS).

Ultrathin sections 90 nm were cut using a Reichert Ultracut E ultramicrotome (Leica).

Electron micrographs were collected using a Hitachi H-7650 TEM (Hitachi High

Technologies America) equipped with a 2,000 by 2,000 pixel AMT TEM CCD camera

(Advanced Microscopy Techniques).

Islet Isolation and Metabolic Labeling. Pancreatic islets were isolated from 6-8-week old mice as previously described49. Briefly, the pancreas was perfused with liberase

(Roche, 0.5 mg/mL) and removed for digestion in a 37°C water-bath for 20 minutes.

After washing and filtering the digest, islets were separated from exocrine cells by

density gradient centrifugation using Histopaque 1077 (Sigma). Islets were collected

from the histopaque/media interface and washed several times before culturing

84 overnight in CRML 1066 (Cellgro) containing 10% FBS, 100 U/ml penicillin and 100

µg/ml streptomycin. For metabolic labeling experiments, 100 islets were hand-picked

from each genotype and repeatedly washed in Kreb’s Ringer-bicarbonate buffer (KRB)

containing 1% BSA. To stimulate insulin biosynthesis, islets were incubated in KRB

containing 2.8 mM glucose for 1 hour at 37°C and then resuspended in KRB containing

16.7 mM glucose and incubated for another hour50,51. During the last 20 minutes of incubation, islets were labeled with 0.5 mCi/ml [35S]methionine/cysteine (MP

Biomedicals) for 60 minutes. Cells were lysed and immunoprecipitated with guinea pig

anti-insulin (abcam, 7842) and protein G plus agarose beads (Santa Cruz) at 4°C

overnight. Immunoprecipitates were analyzed by SDS-PAGE under reducing conditions

and visualized by autoradiography.

Serum Lipase and Amylase Activity. Blood was collected from mice through the

retroorbital sinus into serum separator tubes (BD Biosciences). Serum was submitted

to the Clinical Laboratory at Cincinnati Children’s Hospital for enzyme analyses.

Insulin and Glucagon Assays. Insulin and glucagon were extracted from the

pancreas by repeated homogenization in acid-ethanol (0.18 M HCl in 70% ethanol)52.

Mouse insulin (Crystal Chem) and glucagon (R&D) were assessed by ELISA and

results were normalized to total protein in the pancreatic extracts. Plasma proinsulin

(Alpco), insulin and glucagon were determined by ELISAs in 6 hour-fasted mice at

baseline or after intraperitoneal injection with 10% glucose.

Glucose and Insulin Tolerance Tests. Mice were fasted for 16 hours with free access

to water. Blood glucose levels were determined by collecting blood from the tail vein at

baseline using an ACCU-CHEK Aviva glucose monitor (Roche) before intraperitoneal

85 injection of 10% glucose (1 unit/g of body weight). Blood glucose levels were monitored

overtime following injection. Insulin tolerance tests were performed on 6 hour-fasted

mice by intraperitoneal injection of human insulin (0.75 U/kg, Eli Lilly) followed by blood glucose measurements as described above.

Statistical analyses. All data represent mean ± SEM with a significance difference reported as P ≤ 0.05. Two-way comparisons were analyzed by two-tailed, unpaired

Student’s t test using GraphPad Prism Software.

86 Acknowledgements

We thank Eileen Elfers for assistance with islet isolation, Mary Falconieri for metabolic labeling experiments and Karen Apsley for transfection and coimmunoprecipitation experiments. This study was supported by National Institute of Health Grants

HL103923 (T.E.W.) and HL086492 (T.E.W.).

87 Figure Legends

Figure 1. Generation of ERdj4GT/GT mice. (A) Diagram of the GT allele. The GT

cassette was inserted immediately after adenosine at position 1151 within intron 1 of the

ERdj4 locus (NC_000078.5). SA, splicing acceptor; TM, transmembrane region, β-GEO

(β-galactosidase-neomycin resistance fusion gene); IRES, internal ribosome entry site;

PLAP, placental alkaline phosphatase; pA, polyadenylation signal. (B) PCR genotyping of the WT and GT alleles in tail DNA isolated from the progeny of heterozygous intercrosses. (C) Gene trap copy number was determined in tail gDNA by the TaqMan

Gene Copy Number assay. N = 4 mice/genotype, ***P ≤ 0.001. (D-E) qRT-PCR of

ERdj4 and β-galactosidase mRNAs in ERdj4+/+ and ERdj4GT/GT MEFs. RQ, Relative quantitation. N = 3 samples/group, ***P ≤ 0.001. (F) qRT-PCR of ERdj4 mRNA in tissues isolated from 6-week-old ERdj4+/+ and ERdj4GT/GT littermates; samples were normalized to β-actin. N = 4 mice/genotype, *P ≤ 0.05, **P ≤ 0.01, ***P ≤ 0.001.

Figure 2. Constitutive ER stress and decreased cell viability in ERdj4GT/GT MEFs.

(A) RT-PCR of XBP1 mRNA in ERdj4+/+ and ERdj4GT/GT MEFs treated with tunicamycin

(10 ug/ml) for the indicated periods of time. The PCR products represent hybrid (h),

unspliced (u) and spliced (s) XBP1 species. (B) Western blot analyses of BiP, IRE1α,

calnexin and β-actin (loading control) in ERdj4+/+ and ERdj4GT/GT MEFs treated with indicated doses of tunicamycin or MG-132 (MG, 5 nM, 4 hours). (C) Electron micrographs of ERdj4+/+ and ERdj4GT/GT MEFs. Note dilated ER (arrows) and large inclusions (*) near the Golgi complex in ERdj4GT/GT MEFs. ER: endoplasmic reticulum;

G: Golgi complex; MT: mitochondria; NUC: nucleus. (D) Cell viability of ERdj4+/+ and

ERdj4GT/GT MEFs treated with indicated doses of MG-132, tunicamycin or thapsigargin

88 for 48 hours. Relative values were determined by setting the absorbance of untreated

ERdj4+/+ MEFs to 100%. N = 3 samples/group, *P ≤ 0.05, **P ≤ 0.01, ***P ≤ 0.001.

Figure 3. Hypoglycemia and growth retardation in ERdj4GT/GT mice. Studies assessing perinatal lethality were performed using C57BL/6 littermates. (A)

Comparison of E18.5 embryos from ERdj4+/GT and ERdj4GT/GT mice. (B) Body weights of E18.5 embryos. N = 6-14 mice/genotype, **P ≤ 0.01. (C) Blood glucose levels of

naturally born or caesarean delivered, nonsuckling neonates. N = 10-11

mice/genotype, **P ≤ 0.01, ***P ≤ 0.001. (D) Immunofluorescence of insulin (red) and

glucagon (green) in pancreatic tissue sections from newborn mice. Scale bars are 100

µm. N = 2 mice/genotype. (E-F) Quantitation of insulin and glucagon in pancreatic

extracts from newborn mice. N = 6 mice/genotype, *P ≤ 0.05, **P ≤ 0.01.

Figure 4. Elevated ER stress in pancreatic β-cells of ERdj4GT/GT mice. (A) Western blot analyses of GFP (reporter for XBP1 splicing), IRE-1α and β-actin (loading control) proteins in pancreatic lysates from 6-week-old ERdj4+/+ERAI+ and ERdj4GT/GTERAI+

mice. N = 2-5 mice/genotype. (B) Confocal microscopy of insulin (red) and GFP

(green) protein in pancreatic tissue sections from 8-week-old ERdj4+/+ERAI+ and

ERdj4GT/GTERAI+ mice. Scale bars are 10 µm. N = 4-5 mice/genotype. (C)

Hematoxylin and eosin staining of pancreatic tissue sections from 16-20-week-old

ERdj4+/+ and ERdj4GT/GT mice. Note vacuolated cells in the islets of ERdj4GT/GT mice

(arrows and inset). Scale bars are 10 µm. N = 5 mice/genotype. (D)

Immunofluorescence of insulin (red) and glucagon (green) in pancreatic tissue sections

from 16-20-week-old ERdj4+/+ and ERdj4GT/GT mice. Scale bars are 10 µm. N = 5 mice/genotype. (E) Quantitation of glucagon in pancreatic extracts of 16-20-week-old

89 ERdj4+/+ and ERdj4GT/GT mice. N = 5 mice/genotype, **P ≤ 0.01. (F) Plasma glucagon levels in fasted 8-16-week-old ERdj4+/+ and ERdj4GT/GT mice. N = 11-12 mice/genotype,

*P ≤ 0.05. (G) qRT-PCR of ERdj4 mRNA in islets isolated from 6-week-old, C57BL/6

ERdj4+/+ and ERdj4GT/GT littermates; samples were normalized to β-actin. N = 2-3

samples/genotype.

Figure 5. ERdj4 deficiency impairs insulin biosynthesis. (A) Immunofluorescence of insulin (red) and BiP (green) in pancreatic tissue sections from 16-20-week-old

ERdj4+/+ and ERdj4GT/GT mice. Scale bars are 5 µm. N = 3 mice/genotype. (B)

Metabolic labeling of islets stimulated with 16.7 mM glucose from 6-week-old, C57BL/6 littermates. Equal numbers of trichloroacedic-precipitable cpms were immunoprecipitated with insulin antibody. N = 3 samples/genotype. (C) HEK293 cells were transfected with empty vector or cotransfected with plasmid expressing ERdj4-HA and plasmid encoding Pcsk1, Pcsk2, GCG or CPE with FLAG. Cell lysates were immunoprecipitated with FLAG antibody followed by Western blotting with HA antibody to detect ERdj4.

Figure 6. Hypoinsulinemia causes glucose intolerance in ERdj4GT/GT mice. (A)

Fasting blood glucose levels in 16-20-week-old mice. N = 12-14 mice/genotype. (B)

Blood glucose levels over time after administration of glucose to fasted 12-16-week-old mice. N = 14 mice/genotype, *P ≤ 0.05, **P ≤ 0.01. (C) Blood glucose levels over time after administration of human insulin to fasted 16-20-week-old mice. N = 5 mice/genotype. (D) Plasma insulin levels before and after glucose administration to fasted 12-week-old mice. N = 8 mice/genotype, **P ≤ 0.01, ***P ≤ 0.001. (E) Plasma

proinsulin/insulin ratio in 12-week-old, fasted mice. N = 8 mice/genotype, *P ≤ 0.05.

90 Supplemental Figure 1. Elevated ER stress in tissues of ERdj4GT/GT mice. (A)

Western blot analyses of GFP (reporter for XBP1 splicing), IRE-1α and β-actin (loading control) proteins in kidney and lung lysates from 8-week-old ERdj4+/+ERAI+ and

ERdj4GT/GTERAI+ mice. N = 2-5 mice/genotype. (B) Transmission electron micrographs

of renal tubular epithelial cells in the kidneys of ERdj4+/+ and ERdj4GT/GT mice. Dilated

ER (arrows) was detected in ERdj4GT/GT cells. Scale bars are 1 µm. N = 2 mice/genotype. (C) Transmission electron micrographs of serous cells in the salivary glands of ERdj4+/+ and ERdj4GT/GT mice. Dilated ER (arrows) and Golgi inclusions (*)

were detected in ERdj4GT/GT mice. Scale bars are 1 µm. N = 2 mice/genotype. ER: endoplasmic reticulum; G: Golgi complex; LYS: lysosome; MT: mitochondria; NUC: nucleus.

Supplemental Figure 2. Ultrastructural evidence of ER stress in α-cells of

ERdj4GT/GT mice. (A) Immunofluorescence of glucagon (red) and GFP (green) proteins in pancreatic islets of 16-20-week-old ERdj4+/+ and ERdj4GT/GT mice. Autofluorescence

(yellow in merged panel) was detected in red blood cells. Scale bars are 10 µm. N = 5

mice/genotype. (B) Transmission electron microscopy of α-cells in pancreatic islets of

6-week-old ERdj4+/+ and ERdj4GT/GT mice. Note the pronounced ER dilation and Golgi inclusions in α-cells of ERdj4GT/GT mice. Scale bars are 2 µm. N = 2 mice/genotype.

ER: endoplasmic reticulum; G: Golgi complex; MT: mitochondria; NUC: nucleus; RBC: red blood cell.

Supplemental Figure 3. ER stress in pancreatic β-cells of ERdj4GT/GT mice. (A)

Transmission electron microscopy of β-cells in pancreatic islets of 6-week-old ERdj4+/+

and ERdj4GT/GT mice. Note the pronounced ER dilation, Golgi inclusions and electron

91 translucent secretory granules (*) in β-cells of ERdj4GT/GT mice. Scale bars are 2 µm. N

= 2 mice/genotype. ER: endoplasmic reticulum; G: Golgi complex; MT: mitochondria;

NUC: nucleus; RBC: red blood cells. (B) Quantitation of insulin in pancreatic extracts of

16-20-week-old ERdj4+/+ and ERdj4GT/GT mice. N = 5 mice/genotype.

Supplemental Figure 4. ER stress in pancreatic acinar cells of ERdj4GT/GT mice does not impair function. (A-B) Hematoxylin and eosin staining of pancreatic tissue sections from 8-week-old (A) or 8-month-old (B) ERdj4+/+ and ERdj4GT/GT mice. Note

cytoplasmic vacuolation in the acinar cells of ERdj4GT/GT mice (arrows). Scale bars are

20 µm. N = 3-5 mice/genotype. (C) Transmission electron microscopy of pancreatic acinar cells from 8-week-old ERdj4+/+ and ERdj4GT/GT mice. Note ER dilation (arrows),

Golgi inclusions (*) and immature zymogen granules (IZ). Scale bars are 1 µm. N = 2 mice/genotype. ER: endoplasmic reticulum; Z: zymogen granules; IZ: immature zymogen granules. (D) Amylase and lipase enzyme activity in the serum of 8-week-old

ERdj4+/+ and ERdj4GT/GT mice. N = 4-6 mice/genotype.

92 A A1151 G1152

SA SA TM !-GEO IRES PLAP pA

1 2 3 non-coding coding C

B

+/GT +/GT +/+ +/GT GT/GT

690bp trap 258bp wt

D E

F

Figure 1 93 A XBP-1 mRNA ERdj4+/+ ERdj4GT/GT 0 0.25 0.5 1 2 4 8 0 0.25 0.5 1 2 4 8 hrs h u s

B ERdj4+/+ ERdj4GT/GT TM 0 0.25 0.5 1 4 8 16 MG 0 0.25 0.5 1 4 8 16 MG

BiP

IRE1"

Calnexin

!-actin

C D MEFs ERdj4+/+ ) 125 d *** GT/GT e ERdj4 MT ER t a *** e 100 +/+ r NUC G G t n

u *** 75

% *** ( ** ERdj4

y * t i l

i 50 b a i v

l 25 l e ER C 0 M M M M g/mL g/mL

GT/GT GT/GT TG 1 µ MT Untreated TG 0.5 µ NUC TM 2 µ TM 8 µ * MG-132 1 µ G MG-132 0.5 G µ ERdj4 *

Figure 2 94 A B C ** ERdj4+/GT 1.6 ** ** 100 *** ) g (

t ) 80 l h 1.4 d / g i g e m

w 60 (

y 1.2 e d s o o b

c 40

u l 5 . G 8 1.0

1 20 E 0.8 0

GT/GT +/+ +/+ ERdj4 +/GT +/GT GT/GT GT/GT

ERdj4 ERdj4 ERdj4 ERdj4 ERdj4 ERdj4

D ERdj4+/GT ERdj4GT/GT insulin / glucagon

E F 20 150 ) g

) ** *** µ / g g µ / p g 15 (

t n

( 100

n t e n t e n t o

n 10 c

o c n

o

n 50 i g l a u 5 c s u n l i g 0 0 ERdj4+/GT ERdj4GT/GT ERdj4+/GT ERdj4GT/GT

Figure 3 95 A E 80 Pancreas ) ** g µ /

ERdj4 + + GT GT GT GT GT g n

( 60

t n e GFP t n

o 40 c

n

IRE-1" o g

a 20 c u l

!-actin G 0 ERdj4+/+ ERdj4GT/GT ERdj4+/+ ERdj4GT/GT B F *

) 500 L m /

g 400 p (

n

o 300 g GFP insulin / GFP a c u l 200 g

a

C m s 100 a l p 0 ERdj4+/+ ERdj4GT/GT H&E

G 101

D 0

) 10 Q R (

4 -1 j 10 d R E 10-2 insulin / glucagon 10-3 ERdj4+/+ ERdj4GT/GT

Figure 4 96 A B insulin/BiP ** 1.8

1.5

) +/+ Q

R 1.2 (

n i l ERdj4

u 0.9 s n i o

r 0.6 p 0.3

0.0 ERdj4+/+ ERdj4GT/GT

Islets GT/GT ERdj4

C

FL FL FL FL FL FL FL FL FL + Pcsk1 + GCG + CPE + Pcsk2

+ Pcsk1 + Pcsk2 + GCG + CPE

HA HA HA HA HA HA HA HA HA HA FL FL FL FL ERdj4 Vector Vector ERdj4 ERdj4 ERdj4 ERdj4 ERdj4 ERdj4 Vector Vector GCG ERdj4 Pcsk1 ERdj4 Pcsk2 ERdj4 CPE

!-HA ERdj4 --22 Input IP !-FLAG

Figure 5 97 A B 250 200 ** * ** **

) 200 ** l ) l d / d 150 / g g

m 150 m (

(

e e

100 s s o o

c 100 c +/+ u l u ERdj4 l G G 50 50 ERdj4GT/GT

0 0 ERdj4+/+ ERdj4GT/GT 0 15 30 60 90 120 Time post-glucose injection (min) C D 1.2 175 ERdj4+/+ ) L

) 150 GT/GT l ERdj4 m d

/ 0.8 /

125 g g n ( m

(

100 n

i ** e l s

u ***

o 75 ** s 0.4 c n I u l 50 ERdj4+/+ G 25 ERdj4GT/GT 0 0.0 0 15 30 60 90 120 0 15 30 Time post-insulin injection (min) Time post-glucose injection (min) E 0.8 * n i

l 0.6 u s n i / n i l 0.4 u s n i o r

p 0.2

0.0 ERdj4+/+ ERdj4GT/GT

Figure 6 98 A

Kidney Lung

ERdj4 + + GT GT GT GT GT + + GT GT GT GT GT

GFP

IRE1"

!-actin

B ERdj4+/+ ERdj4GT/GT Kidney

C

G ! ! Salivary Gland

Supplemental Figure S1 99 A Glucagon GFP (sXBP-1) Merge +/+

GT/GT

B ERdj4+/+ ERdj4GT/GT

NUC NUC MT G MT ! -cells ER MT RBC MT

Supplemental Figure S2 100 B Supplemental FigureS3 A

"-cells insulin content (ng/µg) 0.0 0.1 0.2 0.3 0.4 0.5 MT NUC ERdj4 ERdj4 +/+ ERdj4 NUC +/+

GT/GT 101 NUC * * ER G ERdj4 ER

ER MT GT/GT *

* MT A ERdj4+/+ ERdj4GT/GT 8 weeks

B 8 months

C Z Z ER Z ER ER IZ IZ * Z Z

D

Supplemental Figure S4 102 Table 1. Genotype distribution of P21 progeny from heterozygous intercrosses.

103 Table 2. Genotype distribution of day 18.5 embryos from heterozygous intercrosses.

104 References

1. Iwakoshi, N. N., Lee, A. H. & Glimcher, L. H. It's a good year for Blimp-1 (and plasma cells). Immunity 19, 466-468 (2003).

2. Lee, A. H., Chu, G. C., Iwakoshi, N. N. & Glimcher, L. H. XBP-1 is required for biogenesis of cellular secretory machinery of exocrine glands. EMBO J 24, 4368- 4380 (2005).

3. Zhang, P. et al. The PERK eukaryotic initiation factor 2 alpha kinase is required for the development of the skeletal system, postnatal growth, and the function and viability of the pancreas. Mol Cell Biol 22, 3864-3874 (2002).

4. Iwawaki, T., Akai, R. & Kohno, K. IRE1alpha disruption causes histological abnormality of exocrine tissues, increase of blood glucose level, and decrease of serum immunoglobulin level. PLoS One 5, e13052 (2010).

5. Ron, D. & Walter, P. Signal integration in the endoplasmic reticulum unfolded protein response. Nat Rev Mol Cell Biol 8, 519-529 (2007).

6. Schroder, M. & Kaufman, R. J. The mammalian unfolded protein response. Annu Rev Biochem 74, 739-789 (2005).

7. Lee, Y. K., Brewer, J. W., Hellman, R. & Hendershot, L. M. BiP and immunoglobulin light chain cooperate to control the folding of heavy chain and ensure the fidelity of immunoglobulin assembly. Mol Biol Cell 10, 2209-2219 (1999).

8. Wang, N., Daniels, R. & Hebert, D. N. The cotranslational maturation of the type I membrane glycoprotein tyrosinase: the heat shock protein 70 system hands off to the lectin-based chaperone system. Mol Biol Cell 16, 3740-3752 (2005).

9. Molinari, M., Galli, C., Piccaluga, V., Pieren, M. & Paganetti, P. Sequential assistance of molecular chaperones and transient formation of covalent complexes during protein degradation from the ER. J Cell Biol 158, 247-257 (2002).

10. Nishikawa, S. I., Fewell, S. W., Kato, Y., Brodsky, J. L. & Endo, T. Molecular chaperones in the yeast endoplasmic reticulum maintain the solubility of proteins for retrotranslocation and degradation. J Cell Biol 153, 1061-1070 (2001).

11. Bertolotti, A., Zhang, Y., Hendershot, L. M., Harding, H. P. & Ron, D. Dynamic interaction of BiP and ER stress transducers in the unfolded-protein response. Nat Cell Biol 2, 326-332 (2000).

105 12. Shen, J., Chen, X., Hendershot, L. & Prywes, R. ER stress regulation of ATF6 localization by dissociation of BiP/GRP78 binding and unmasking of Golgi localization signals. Dev Cell 3, 99-111 (2002).

13. Mayer, M. P. & Bukau, B. Hsp70 chaperones: cellular functions and molecular mechanism. Cell Mol Life Sci 62, 670-684 (2005).

14. Awad, W., Estrada, I., Shen, Y. & Hendershot, L. M. BiP mutants that are unable to interact with endoplasmic reticulum DnaJ proteins provide insights into interdomain interactions in BiP. Proc Natl Acad Sci U S A 105, 1164-1169 (2008).

15. Jiang, J., Prasad, K., Lafer, E. M. & Sousa, R. Structural basis of interdomain communication in the Hsc70 chaperone. Mol Cell 20, 513-524 (2005).

16. Fewell, S. W., Travers, K. J., Weissman, J. S. & Brodsky, J. L. The action of molecular chaperones in the early secretory pathway. Annu Rev Genet 35, 149- 191 (2001).

17. Jin, Y., Awad, W., Petrova, K. & Hendershot, L. M. Regulated release of ERdj3 from unfolded proteins by BiP. EMBO J 27, 2873-2882 (2008).

18. Petrova, K., Oyadomari, S., Hendershot, L. M. & Ron, D. Regulated association of misfolded endoplasmic reticulum lumenal proteins with P58/DNAJc3. EMBO J 27, 2862-2872 (2008).

19. Shen, Y. & Hendershot, L. M. ERdj3, a stress-inducible endoplasmic reticulum DnaJ homologue, serves as a cofactor for BiP's interactions with unfolded substrates. Mol Biol Cell 16, 40-50 (2005).

20. Otero, J. H., Lizak, B. & Hendershot, L. M. Life and death of a BiP substrate. Semin Cell Dev Biol 21, 472-478 (2010).

21. Dong, M., Bridges, J. P., Apsley, K., Xu, Y. & Weaver, T. E. ERdj4 and ERdj5 are required for endoplasmic reticulum-associated protein degradation of misfolded surfactant protein C. Mol Biol Cell 19, 2620-2630 (2008).

22. Lai, C. W., Otero, J. H., Hendershot, L. M. & Snapp, E. ERdj4 protein is a soluble endoplasmic reticulum (ER) DnaJ family protein that interacts with ER-associated degradation machinery. J Biol Chem 287, 7969-7978 (2012).

23. Shen, Y., Meunier, L. & Hendershot, L. M. Identification and characterization of a novel endoplasmic reticulum (ER) DnaJ homologue, which stimulates ATPase activity of BiP in vitro and is induced by ER stress. J Biol Chem 277, 15947-15956 (2002).

106 24. Hageman, J. & Kampinga, H. H. Computational analysis of the human HSPH/HSPA/DNAJ family and cloning of a human HSPH/HSPA/DNAJ expression library. Cell Stress Chaperones 14, 1-21 (2009).

25. Weitzmann, A., Baldes, C., Dudek, J. & Zimmermann, R. The heat shock protein 70 molecular chaperone network in the pancreatic endoplasmic reticulum - a quantitative approach. FEBS J 274, 5175-5187 (2007).

26. Kanemoto, S. et al. XBP1 activates the transcription of its target genes via an ACGT core sequence under ER stress. Biochem Biophys Res Commun 331, 1146-1153 (2005).

27. Lee, A. H., Iwakoshi, N. N. & Glimcher, L. H. XBP-1 regulates a subset of endoplasmic reticulum resident chaperone genes in the unfolded protein response. Mol Cell Biol 23, 7448-7459 (2003).

28. Kurisu, J. et al. MDG1/ERdj4, an ER-resident DnaJ family member, suppresses cell death induced by ER stress. Genes Cells 8, 189-202 (2003).

29. Miller, K. et al. Islet formation during the neonatal development in mice. PLoS One 4, e7739 (2009).

30. Iwawaki, T., Akai, R., Kohno, K. & Miura, M. A transgenic mouse model for monitoring endoplasmic reticulum stress. Nat Med 10, 98-102 (2004).

31. Turgeon, B. & Meloche, S. Interpreting neonatal lethal phenotypes in mouse mutants: insights into gene function and human diseases. Physiol Rev 89, 1-26 (2009).

32. Lee, A. H., Heidtman, K., Hotamisligil, G. S. & Glimcher, L. H. Dual and opposing roles of the unfolded protein response regulated by IRE1alpha and XBP1 in proinsulin processing and insulin secretion. Proc Natl Acad Sci U S A 108, 8885- 8890 (2011).

33. Bataille, D. Pro-protein convertases in intermediary metabolism: islet hormones, brain/gut hormones and integrated physiology. J Mol Med (Berl) 85, 673-684 (2007).

34. Talchai, C., Xuan, S., Lin, H. V., Sussel, L. & Accili, D. Pancreatic beta Cell Dedifferentiation as a Mechanism of Diabetic beta Cell Failure. Cell 150, 1223- 1234 (2012).

35. Allotey, R. A. et al. The EIF2AK3 gene region and type I diabetes in subjects from South India. Genes Immun 5, 648-652 (2004).

107 36. Delepine, M. et al. EIF2AK3, encoding translation initiation factor 2-alpha kinase 3, is mutated in patients with Wolcott-Rallison syndrome. Nat Genet 25, 406-409 (2000).

37. Meex, S. J. et al. Activating transcription factor 6 polymorphisms and haplotypes are associated with impaired glucose homeostasis and type 2 diabetes in Dutch Caucasians. J Clin Endocrinol Metab 92, 2720-2725 (2007).

38. Senee, V. et al. Wolcott-Rallison Syndrome: clinical, genetic, and functional study of EIF2AK3 mutations and suggestion of genetic heterogeneity. Diabetes 53, 1876- 1883 (2004).

39. Thameem, F., Farook, V. S., Bogardus, C. & Prochazka, M. Association of amino acid variants in the activating transcription factor 6 gene (ATF6) on 1q21-q23 with type 2 diabetes in Pima Indians. Diabetes 55, 839-842 (2006).

40. Harding, H. P. et al. Diabetes mellitus and exocrine pancreatic dysfunction in perk- /- mice reveals a role for translational control in secretory cell survival. Mol Cell 7, 1153-1163 (2001).

41. Ladiges, W. C. et al. Pancreatic beta-cell failure and diabetes in mice with a deletion mutation of the endoplasmic reticulum molecular chaperone gene P58IPK. Diabetes 54, 1074-1081 (2005).

42. Scheuner, D. et al. Translational control is required for the unfolded protein response and in vivo glucose homeostasis. Mol Cell 7, 1165-1176 (2001).

43. Huang, C. J. et al. High expression rates of human islet amyloid polypeptide induce endoplasmic reticulum stress mediated beta-cell apoptosis, a characteristic of humans with type 2 but not type 1 diabetes. Diabetes 56, 2016-2027 (2007).

44. Laybutt, D. R. et al. Endoplasmic reticulum stress contributes to beta cell apoptosis in type 2 diabetes. Diabetologia 50, 752-763 (2007).

45. Marchetti, P. et al. The endoplasmic reticulum in pancreatic beta cells of type 2 diabetes patients. Diabetologia 50, 2486-2494 (2007).

46. Bridges, J. P., Wert, S. E., Nogee, L. M. & Weaver, T. E. Expression of a human surfactant protein C mutation associated with interstitial lung disease disrupts lung development in transgenic mice. J Biol Chem 278, 52739-52746 (2003).

47. Conner, D. A. Mouse embryo fibroblast (MEF) feeder cell preparation. Curr Protoc Mol Biol Chapter 23, Unit 23.2 (2001).

108 48. Ridsdale, R., Na, C. L., Xu, Y., Greis, K. D. & Weaver, T. Comparative proteomic analysis of lung lamellar bodies and lysosome-related organelles. PLoS One 6, e16482 (2011).

49. Zmuda, E. J., Powell, C. A. & Hai, T. A method for murine islet isolation and subcapsular kidney transplantation. J Vis Exp (2011).

50. Skelly, R. H., Bollheimer, L. C., Wicksteed, B. L., Corkey, B. E. & Rhodes, C. J. A distinct difference in the metabolic stimulus-response coupling pathways for regulating proinsulin biosynthesis and insulin secretion that lies at the level of a requirement for fatty acyl moieties. Biochem J 331, 553-561 (1998).

51. Welsh, M., Hammer, R. E., Brinster, R. L. & Steiner, D. F. Stimulation of growth hormone synthesis by glucose in islets of Langerhans isolated from transgenic mice. J Biol Chem 261, 12915-12917 (1986).

52. Sjoholm, A., Arkhammar, P., Berggren, P. O. & Andersson, A. Polyamines in pancreatic islets of obese-hyperglycemic (ob/ob) mice of different ages. Am J Physiol Cell Physiol 280, C317-23 (2001).

109

CHAPTER III

Deficiency of the ER stress chaperone ERdj4 is associated

with defects in osteogenesis and B cell development

110 Deficiency of the ER stress chaperone ERdj4 is associated with defects in

osteogenesis and B cell development

Jill M. Fritz, Mei Dong, Cheng-Lun Na and Timothy E. Weaver

Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center and the

University of Cincinnati College of Medicine, Cincinnati, OH 45229

Correspondence: Timothy E. Weaver, 3333 Burnet Ave., Cincinnati, OH 45229; Phone:

513-636-7223; Email: [email protected]

Supported by NIH awards HL103923 and HL086492

Manuscript in preparation

111 Abstract

Highly secretory cells, including bone-forming osteoblasts and antibody-secreting plasma cells, require the unfolded protein response (UPR) to maintain ER homeostasis during increased protein synthesis. Expression of the UPR chaperone ERdj4 is

upregulated in response to ER stress arising from accumulation of unfolded/misfolded

proteins in the ER lumen. In the current study, we investigated the role of ERdj4 in

osteoblast differentiation and function in hypomorphic ERdj4 gene trap (ERdj4GT/GT) mice. Bone-lining cells, which differentiate from osteoblasts, were reduced in association with increased ER stress in ERdj4GT/GT mice. Primary bone cells isolated from ERdj4GT/GT mice had reduced expression of osteogenic-specific markers. Reduced

osteogenesis, in turn, was associated with decreased numbers of bone marrow B cells

related to increased cell death. Further, numbers of mature B cells in the spleen and

peritoneal cavity were also compromised by ERdj4 deficiency. ERdj4GT/GT bone marrow

transplanted into ERdj4+/+ hosts rescued all B cell compartments, confirming a B cell-

extrinsic defect. Collectively, these data suggest that the loss of ERdj4 impairs the

maturation/survival of osteogenic cells in association with increased ER stress. Further,

deficiency of osteogenic cells in ERdj4GT/GT mice is linked to a defect in B cell development in the bone marrow.

112 Introduction

Highly secretory cells, including bone-forming osteoblasts and antibody-secreting plasma cells, utilize the UPR to alleviate the ER burden imposed by increased protein synthesis. The UPR maintains ER homeostasis by attenuating protein translation, increasing protein folding capacity and removing unfolded/misfolded proteins from the

ER lumen. The UPR signal transducers protein kinase RNA (PKR)-like ER kinase

(PERK), activating transcription factor 6 (ATF6) and inositol-requiring enzyme 1 alpha

(IRE1α) are localized in the ER membrane where they trigger the UPR in response to accumulation of unfolded or misfolded protein in the ER lumen. Activation of UPR sensors occurs when the ER chaperone BiP/GRP78 is recruited away from the luminal domain of PERK, ATF6 and IRE1α to unfolded/misfolded substrates. PERK phosphorylates the alpha subunit of eukaryotic initiation factor 2 (eIF2α) leading to inhibition of protein translation, thereby preventing further protein accumulation in the

ER1. ATF6 results in translocation to the Golgi, where site-specific serine proteases

generate a cytosolic fragment that, in turn, translocates to the nucleus and increases

the expression of ER chaperones2,3. The endocribonuclease activity of IRE1α splices an intron from the mRNA of X-box-protein 1 (XBP1) resulting in translation of a transcription factor that increases the expression of ER chaperones and ER-associated degradation (ERAD) machinery4,5. Failure to restore ER homeostasis results in

apoptosis through PERK/eIF2α-mediated induction of activating transcription factor 4

(ATF4) and C/EBP homologous protein (CHOP)6.

The HSP70 family member BiP/GRP78 is a soluble ER chaperone with multiple functions, including regulation of the translocon pore7, maintenance of ER calcium

113 stores8, protein folding and degradation9-12, and induction of the UPR13,14. The

multifunctionality of BiP is modulated by ER-localized DnaJ (ERdj) proteins, a diverse

class of cochaperones from the HSP40 family. ERdjs stimulate the ATPase activity of

BiP, leading to conformational changes that stabilize client interaction. Although some

ERdjs interact with BiP independently of substrate, others bind substrates directly to

recruit BiP15-18. ERdjs promote protein folding and/or degradation of newly translocated

(ERdj1, ERdj2, ERdj3), unfolded (ERdj3, ERdj4, ERdj6) or misfolded substrates (ERdj4,

ERdj5), often through association with translocation or ERAD machinery19,20.

ERdj4 is a soluble ER luminal protein, containing a J domain that associates with

BiP to stimulate ATP hydrolysis, a glycine-phenylalanine rich region of unknown

function and a substrate binding domain that interacts with misfolded (SP-CΔexon4,

insulin2C96Y) and unfolded (insulin2WT, Pcsk1, Pcsk2, CPE) substrates. ERdj4 is an ER stress-related chaperone21 that facilitates the removal of unfolded/misfolded substrates

from the ER lumen by interacting with ERAD machinery22,23. Although ERdj4

expression is upregulated in response to ER stress arising from expression of terminally

misfolded substrates, recent studies in our laboratory revealed that ERdj4 also plays an

important role in adaptation to ER stress associated with normal growth, development

and metabolism. Hypomorphic expression of ERdj4 in mice resulted in perinatal

lethality associated with growth restriction and hypoglycemia, while surviving adult mice

were glucose intolerant and hypoinsulinemic, with defects in the pancreatic β-cell secretory pathway (Chapter 2).

Osteoblasts are highly secretory cells that are responsible for the synthesis and secretion of extracellular bone matrix. Several components of the UPR regulate the

114 expression of osteoblast-specific genes that influence cell differentiation and function.

ATF4 and XBP1 increase the expression of Osterix, a transcription factor required for

osteoblast maturation. ATF6 and old astrocyte specifically-induced substance (OASIS), an ATF6 structural homologue, regulate expression of osteocalcin and type I collagen, respectively, leading to an increase in extracellular matrix production24,25. Further, the

UPR in osteoblasts is essential for skeletal development in humans and mice. An

inactivating mutation in human PERK caused growth retardation, epiphyseal dysplasia

and osteoporosis26,27. A similar phenotype was observed in PERK-deficient mice and

was associated with decreased numbers of osteoblasts in bone tissue and impaired

osteoblast proliferation and differentiation in vitro28,29. Mice deficient in ATF4, a

transcription factor activated by the PERK signaling cascade, exhibited a delay in

skeletal development during embryogenesis30 and low postnatal bone density due to

impaired osteoblast differentiation and function31.

In addition to bone formation, osteoblasts are an important component of the bone marrow microenvironment, influencing hematopoiesis through cell-surface expression of signaling and adhesion molecules, and secretion of growth factors and cytokines32. Conditional depletion of osteoblasts in vivo reduced hematopoietic stem cells (HSCs), B cells and erythrocytes in the bone marrow, and increased extramedullary hematopoiesis33. Calvarial osteoblasts were shown to support B cell

commitment and maturation from HSCs in vitro through the expression of IL-7, CXC-

chemokine ligand 12 (CXCL12) and vascular adhesion molecule-1 (VCAM-1)34.

Parathyroid hormone (PTH), which regulates osteoblast differentiation, function and

115 survival35, induced IL-7 and CXCL12 production by osteoblasts in vitro34,36,37 and impaired PTH signaling on osteogenic cells reduced B cell development in vivo38.

Since the UPR is required for osteogenesis, we hypothesized that ERdj4

deficiency would impair osteoblast differentiation and/or function. Bone-lining cells of

the osteogenic lineage were reduced in association with increased ER stress in ERdj4

gene trap (ERdj4GT/GT) mice. Primary osteogenic cells isolated from the long bones of

ERdj4GT/GT mice exhibited decreased osteogenic-specific markers consistent with impaired osteogenesis. Deficiency of osteogenic cells in ERdj4GT/GT mice was also associated with reduced B cell development in the bone marrow. Further, ERdj4 deficiency affected mature B cell populations in the spleen and peritoneal cavity.

Transfer of ERdj4GT/GT bone marrow into ERdj4+/+ hosts rescued the defect in all B cell

compartments. Collectively, these results suggest that the loss of ERdj4 in osteogenic

cells impairs maturation and/or survival, which is associated with a defect in B cell

development.

116 Results

Bone-lining cells are reduced in ERdj4GT/GT mice⎯Since several components of the

UPR are essential for skeletal development and osteoblast differentiation28,29,39,40, cells

of the osteogenic lineage were assessed in long bones of adult mice. Bone-lining cells, which are quiescent osteoblasts, were markedly reduced in ERdj4GT/GT mice compared to controls (Fig. 1A). Since ERdj4 deficiency caused constitutive stress in a variety of non-secretory and secretory cell types (Chapter 1), ER stress was assessed in osteogenic cells in vivo by crossing ERdj4+/GT mice to ER stress-activated indicator

(ERAI) transgenic mice41. GFP fluorescence (resulting from XBP1 splicing and

expression of XBP1/GFP fusion protein) was increased in osteocalcin-positive bone-

lining cells of ERdj4GT/GTERAI+ mice (Fig. 1B). These data indicate that bone-lining cells

are reduced in association with increased ER stress in ERdj4GT/GT mice.

Genetic alterations in the UPR caused defects in growth and skeletal development due to impaired osteoblast differentiation and function28,31. Although previous studies in our laboratory demonstrated growth restriction in ERdj4GT/GT

embryos, postnatal compensatory growth resulted in normal weights by 8 weeks of age

(data not shown). Further, bone and cartilage staining of E18.5 embryos revealed no

gross defects in skeletal development in ERdj4GT/GT mice (Supplemental Fig. S1).

Collectively, these data suggest that ERdj4 deficiency does not affect postnatal growth

or osteoblast function.

Primary cells isolated from the long bones of ERdj4+/+ and ERdj4GT/GT mice

varied morphologically in size and staining intensity (Fig. 2A), but were osteocalcin-

positive (Fig. 2B), suggesting that these cells were of the osteogenic lineage.

117 Osteocalcin protein was modestly reduced in osteogenic cells of ERdj4GT/GT mice (Fig.

2B) in association with decreased osteocalcin mRNA (Fig. 2C). Other osteoblast-

specific markers were also significantly reduced in osteogenic cells of ERdj4GT/GT mice, including Osterix and type I collagen (Fig. 2C). RANKL mRNA, an osteocyte marker42,

was also reduced in ERdj4GT/GT mice (Fig. 2C). Treatment with ascorbic acid and beta-

glycerol phosphate, which induces osteoblast differentiation, significantly upregulated

osteocalcin mRNA in ERdj4GT/GT cells (Fig. 2D). Similar to findings in vivo, osteogenic

cells from ERdj4GT/GT mice exhibited increased levels of ER stress (Fig. 2E). Overall, these data suggest that osteogenic cells isolated from the long bones of ERdj4GT/GT

mice were predominantly immature bone cells.

Recent studies demonstrated that UPR deficiency impaired bone morphogenetic protein 2 (BMP2)-induced osteoblast differentiation39,40. BMP2 induces osteogenesis by

activating the Smad-Runx2 signaling pathway that regulates osteoblast-specific genes,

including alkaline phosphatase (ALP), type I collagen and osteocalcin43,44. ERdj4 mRNA was upregulated in a time-dependent manner following BMP2 treatment of

ERdj4+/+ mouse embryonic fibroblasts (MEFs) (Supplemental Fig. S2A). Similarly, BiP

and spliced XBP1 mRNAs were also induced in ERdj4+/+ MEFs after 96 hours of BMP2

treatment (Supplemental Fig. S2B-C), indicating that BMP2 signaling of osteoblast

differentiation upregulates the UPR in ERdj4+/+ MEFs. BMP2 treatment induced expression of BiP and spliced XBP1 mRNAs to a greater extent in ERdj4GT/GT MEFs compared to controls (Supplemental Fig. S2B-C). ALP activity and osteocalcin, Osterix and type I collagen mRNAs, were all increased in BMP2-treated ERdj4GT/GT MEFs

(Supplemental Fig. S2D-E); however, the magnitude of the increase was significantly

118 blunted in ERdj4GT/GT MEFs compared to ERdj4+/+ MEFs. Overall, these results are consistent with a defect in BMP2-induced osteoblast differentiation associated with increased levels of ER stress in ERdj4GT/GT MEFs.

ERdj4 deficiency affects early B lymphopoiesis⎯Several studies have reported that osteoblasts support hematopoiesis33,34,36,38. Since osteogenic cells were reduced in

ERdj4GT/GT mice, hematopoiesis was assessed in the bone marrow of adult mice by flow

cytometry. Bone marrow cellularity was reduced in ERdj4GT/GT mice (Fig. 3A), which correlated with a significant decrease in erythrocytes (Ter119+) and B cells (B220+), but

not macrophages (F4/80+) or granulocytes (Gr-1+) (Supplemental Fig. S3A-C). Based

on the Hardy classification method of B lymphopoiesis45, the frequency and absolute number of pro-B, pre-B, immature and mature B cells was decreased in the bone marrow of ERdj4GT/GT mice compared to controls (Fig. 3B-C). When pro-B cells were further classified into fractions A through C based on HSA and BP-1 expression, there was a significant decrease in the number of cells in fractions B and C, but not fraction A

(Fig. 3D). The frequency of pro-B cells in the blood and proliferation of developing B cells in the bone marrow were normal (Supplemental Fig. S4A-B), but the percentage of

7-AAD+ pro-B cells was significantly increased compared to controls (Fig. 3E).

Collectively, these results indicate a defect in B cell development starting at the pro-B cell stage, which is associated with an increase in pro-B cell death.

ERdj4 deficiency in the bone marrow microenvironment impairs B cell development⎯To

confirm that the defect in B lymphopoiesis was not cell autonomous, ERdj4GT/GT bone

marrow was transplanted into irradiated ERdj4+/+ hosts and B cell development was analyzed 8 weeks post-transplantation. ERdj4GT/GT bone marrow transplanted into

119 ERdj4+/+ hosts rescued the defect in B cell development starting at the pro-B cell stage

when compared to B cell development in ERdj4GT/GT recipients (Fig. 4A-B).

Furthermore, pre-B colony forming units generated from total bone marrow in media containing IL-7 were normal in ERdj4GT/GT mice, suggesting that ERdj4GT/GT B cells

develop normally in the proper microenvironment (Fig. 4C).

Mature B cell populations are reduced in ERdj4GT/GT mice⎯The defect in early B cell

development in the bone marrow prompted analysis of mature B cell subsets in the

periphery. Immature B cells migrate to the spleen where they undergo further

differentiation into follicular and marginal zone B cell subsets46. Total splenocytes were

reduced in ERdj4GT/GT mice (Fig. 5A), which was associated with a decrease in mature

(B220+CD93-) B cells (Fig. 5B-C). Interestingly, immature (B220+CD93+) B cells were

normal in ERdj4GT/GT mice (Fig. 5B-C), despite lower numbers of immature

(B220+IgMlow) B cells in the bone marrow (Fig. 3B-C). The mature B cell subset affected by ERdj4 deficiency was the follicular B cells (B220+CD21lowCD23+), while

marginal zone B cells (B220+CD21+CD23low) were normal (Fig. 5B-C). Transfer of

ERdj4GT/GT bone marrow into irradiated ERdj4+/+ hosts rescued follicular B cell

development, supporting a defect in the microenvironment (Fig. 5D).

B1 cell development occurs predominantly in the fetal liver and neonatal spleen

to generate self-renewing, mature B1 cells that are localized in the pleural and

peritoneal cavities throughout life47. Total B1 cells (CD19+CD11b+) were decreased in the peritoneal cavity of ERdj4GT/GT mice, resulting from lower numbers of B1b (CD5-) cells, while B1a cells were normal (Fig. 5E).

120 The B cell activating factor (BAFF), which influences mature B cell differentiation

and survival in the periphery, was modestly but significantly increased in the serum of

ERdj4GT/GT mice (Fig. 5F). Collectively, these data suggest that mature B cell survival or proliferation is defective in the periphery of ERdj4GT/GT mice even in the face of increased BAFF.

Plasma cell differentiation and antibody secretion are normal in ERdj4GT/GT mice⎯The

UPR signal transducer XBP1 regulates immunoglobulin synthesis and secretion by plasma cells48,49. Given that ERdj4 expression was reduced in LPS-treated XBP1-/- B

cells50, plasma cell differentiation and function was examined in ERdj4GT/GT mice. The

frequency of splenic plasma cells (CD19+CD138+) was normal at baseline in ERdj4GT/GT

mice and treatment with gαmIg-D antibody induced plasma cell differentiation in vivo

(Supplemental Fig. S5A). Basal immunoglobulins, including IgM, IgG, IgA and IgE,

were normal in the serum of ERdj4GT/GT mice (Supplemental Fig. S5B) and ERdj4GT/GT

mice efficiently produced TNP-specific IgM and IgG antibodies to T cell-dependent and

T cell-independent antigens (Supplemental Fig. S5C-D). Therefore, ERdj4 deficiency

does not affect plasma cell differentiation or antibody secretion even though ERdj4 is an

XBP1 target in vivo.

121 Discussion

Osteoblasts are highly secretory cells that require the UPR to induce transcription of genes essential for maturation and function. In the current study, we

demonstrate that ERdj4, a chaperone regulated by the UPR, influences cells of the

osteogenic lineage in vitro and in vivo. ERdj4 expression was upregulated in response

to BMP2, consistent with previous studies demonstrating induction of the UPR during

BMP2-induced osteoblast differentiation39,40. BMP2 supports osteogenesis by activating Runx2 and Osterix, thereby inducing the transcription of several osteoblast- specific genes, including ALP, osteocalcin and type I collagen43,44. The UPR can upregulate transcription of Osterix (XBP1 and ATF4), or function downstream of Runx2

(ATF6) to induce extracellular matrix production24,40,51. BMP2-induced osteoblast

differentiation was impaired in ERdj4GT/GT MEFs in association with increased ER stress.

Previous studies demonstrated that inducers of ER stress decreased cell viability of

MEFs in the absence of ERdj4 (Chapter 1). The loss of ERdj4 chaperone activity likely

resulted in accumulation of unfolded/misfolded protein within the ER lumen, although

these substrates have yet to be identified. Overall, these findings highlight the importance of ERdj4 in maintaining ER homeostasis during BMP2-induced osteoblast differentiation in vitro.

Primary osteogenic cells isolated from the long bones of adult mice were

morphologically heterogeneous, but osteocalcin-positive, suggesting that these cells

were of the osteogenic lineage. Expression of osteoblast-specific markers, including

Osterix, osteocalcin and type I collagen, were decreased in ERdj4GT/GT osteogenic cells,

but upregulated following treatment with differentiation media. These findings suggest

122 that the frequency of immature bone cells was increased in primary osteogenic cells

from ERdj4GT/GT mice. Although these cells can be found within cortical bone52,53,

osteocytes are the predominant postosteoblastic cell type encased within extracellular

bone matrix. Thus, it is possible that osteocytes are decreased in ERdj4GT/GT mice.

Several lines of evidence support this hypothesis, including (1) reduced expression of

RANKL, a marker most highly expressed by osteocytes of the osteogenic lineage42 and

(2) reduced bone-lining cells, another postosteoblast cell type. The majority of osteoblasts undergo apoptosis after bone formation, but some differentiate into osteocytes or bone-lining cells54. Given that ER stress was increased in osteogenic cells of ERdj4GT/GT mice, it is possible that reduced bone-lining cells were the result of

increased apoptosis.

The role of the IRE1α/XBP1 signaling pathway in postnatal growth and skeletal

development has yet to be explored in vivo, impeded by embryonic lethality in both

IRE1α and XBP1-deficient mice55,56. ERdj4, a chaperone induced by the IRE1α/XBP1 signaling pathway, was previously demonstrated to be important for growth during embryogenesis (Chapter 1). Although ERdj4GT/GT embryos were smaller, compensatory

growth during the postnatal period corrected the phenotype. ERdj4GT/GT mice reached normal weight by 8 weeks of age and did not display any gross skeletal defects. In contrast, deficiency of other UPR components, including PERK and ATF4, resulted in multiple skeletal dysplasias due to impaired osteoblast differentiation and function28,31.

Collectively, these data suggest that the loss of ERdj4 does not affect bone formation by

osteoblasts, but instead influences postosteoblasts. Given that postosteoblasts support

123 hematopoiesis52, deficiency of bone-lining cells in ERdj4GT/GT mice may be responsible

for the defect in B cell development.

Osteoblasts are reported to influence B cell development; however, this

conclusion is based on studies that used either heterogeneous osteogenic cultures in

vitro or ablation of multiple cell types within the osteogenic lineage in vivo33,34. In our study, deficiency of postosteoblasts in ERdj4GT/GT mice was associated with reduced B cells and erythrocytes, the same cell lineages affected by conditional depletion of osteoblasts and postosteoblasts in transgenic mice33. As osteoblasts are transient and

short-lived, it is unlikely that they provide sustained support of the hematopoietic

niche52; in contrast, postosteoblasts cover approximately 90% of all bone surfaces and

survive for months to years in humans and mice54. These observations suggest that postosteoblasts influence B cell development, although this hypothesis remains to be directly tested.

Other cellular niches have been described to support B cell development in the bone marrow. Elegant studies by Tokoyoda et al. using CXCL12-GFP reporter mice demonstrated that the earliest and latest stages of B cell development, the pre-pro B cells and plasma cells, respectively, associated with reticular cells that expressed high levels of CXCL12, termed CAR cells. The pro-B cells moved away from CAR cells and interacted with fibroblast-like cells that expressed IL-7, while the pre-B and immature B cells did not associate with either of these stromal cell types57. CAR cells were located

away from the endosteal surface and did not express osteopontin57, indicating that

these were not osteogenic cells. However, the IL-7-expressing cells may be from the

osteogenic lineage38, given that IL-7 expression was induced in osteogenic cells by

124 parathyroid hormone (PTH)34,37 and disruption of the PTH signaling pathway in

osteogenic cells impaired B cell development at the pro-B cell stage38.

Follicular B cells in the spleen were also compromised by ERdj4 deficiency due

to a defect in the microenvironment. Follicular B cells continuously recirculate

throughout the spleen, liver, lymph nodes and bone marrow58,59. In the bone marrow,

mature recirculating B cells occupy the sinusoidal niche and are supported by survival

factors secreted by resident dendritic cells60. Notably, mature recirculating B cells were reduced in the bone marrow of ERdj4GT/GT mice. Sinusoidal formation in the bone marrow is driven by osteocytes through osteoclastogenesis52. Thus, it is possible that

osteocyte deficiency in ERdj4GT/GT mice affected recirculating B cells in the sinusoidal

niche. It remains to be defined whether peritoneal B1b cells were reduced in ERdj4GT/GT

mice due to a defect in the microenvironment. Although B1b cell development occurs

predominantly during embryogenesis47, they can be generated in the adult bone marrow61. Thus, a defect in the bone marrow microenvironment of ERdj4GT/GT mice may contribute to the loss of B1b cells in the peritoneal cavity. Collectively, these data suggest that the loss of ERdj4 affects the maturation/survival of postoosteoblasts in association with increased ER stress. Deficiency of postosteoblasts is linked to a defect in B cell development in the bone marrow.

125 Materials and Methods

Mice. Hypomorphic ERdj4GT/GT mice were generated from an ES cell line harboring a gene trap cassette (GT) in intron 1 (Bay Genomics), as previously described (Chapter

1). Unless otherwise indicated, all mice used in these experiments were 6 to 8-week- old littermates on a mixed 129P2-C57BL/6 genetic background. ERAI mice were obtained from RIKEN BRC. All mice were housed in a pathogen-free barrier facility and studies were conducted with approval from Cincinnati Children’s Hospital Medical

Center Institutional Animal Care and Use Committee.

Histology and Immunohistochemistry. Bone tissue was fixed in 4% paraformaldehyde, decalcified, embedded in paraffin and 5 µm sections were cut for

H&E staining and immunohistochemistry, as previously described62. For immunohistochemistry, endogenous peroxidase activity was quenched in bone tissue/cells by treatment with 1.5% hydrogen peroxide in methanol for 15 minutes.

Slides then underwent antigen retrieval by boiling in 10 mM citrate buffer (pH 6.0) for 20 minutes before blocking in normal goat serum (Jackson ImmunoResearch) for two hours. Bone tissue/cells were incubated with rabbit anti-mouse osteocalcin (1:100,

Enzo Life Sciences), followed by application of biotinylated goat anti-rabbit IgG (1:200,

Vector Laboratories). Bone tissue/cells were visualized with the Vectastain Elite ABC kit (Vector Laboratories) using nickel-diaminobenzidine as a substrate followed by counterstaining with 0.1% nuclear fast red. Images were obtained using a Axio

Imager.A2 microscope (Zeiss) and AxioCam MRc5 digital camera (Zeiss).

For immunofluorescence, slides underwent antigen retrieval followed by blocking in normal donkey serum (Jackson ImmunoResearch) for two hours. Sections were

126 stained with rabbit anti-osteocalcin and anti-mouse GFP (Invitrogen) followed by

application of secondary antibodies conjugated to Alexa Fluor 555 or Alexa Fluor 647

(1:100, Invitrogen). To remove autofluorescence, slides were incubated in 0.1% Sudan

Black in 70% ethanol for 20 minutes. Image stacks were acquired by 0.25 µm step

distance using a Nikon C1 Si Confocal Imaging System and processed for deconvolution to remove image blurring using Autoquant X 2.2 (Media Cybernetics).

Tissue harvest and cell isolation. Mouse femurs were harvested, trimmed and

flushed with media using a 3 cc syringe and a 21 gauge (g) needle to release the bone

marrow. Single cell suspensions were obtained by repeatedly passing the cells through

a syringe. Mouse spleens were harvested and placed on a nylon mesh strainer (100

µm, BD Biosciences) in a 50 mL conical tube. Spleens were gently massaged with the

blunt end of a plunger from a 3 cc syringe and the strainer was then rinsed with media

to release cells. Peritoneal cavity cells were harvested from mice by injecting 3 mL of

cold PBS with a 27 g needle into the peritoneal cavity, gently massaging the peritoneum

and retrieving the fluid with an 18 g needle63.

Flow cytometry. Bone marrow, spleen and peritoneal cavity cells (1x106 cells/100 µl) were incubated in FACS buffer (1X PBS, 1% FBS, 0.05% NaN3) containing CD16/32

Fc-blocking antibody (Biolegend, 93) for 30 minutes at 4°C and then stained with

fluorescently-labeled antibodies for 30 minutes at 4°C. Data was acquired using the

LSR II flow cytometer (BD Biosciences) and analyzed on FlowJo software (TreeStar,

Inc.). The following mouse-specific monoclonal antibodies were used for flow cytometry

with the source and clone indicated in parentheses: B220-Pacific Blue (Biolegend,

RA3-6B2), IgM-PE (Biolegend, RMM-1), CD43-PE/Cy5 (Biolegend, 1B11), CD24-FITC

127 (Biolegend, M1/69), BP-1-Alexa Fluor 647 (Biolegend, 6C3), CD93-APC (Biolegend,

AA4.1), CD21-Pacific Blue (Ebioscience, eBio4E3), CD23-PE (Ebioscience, B3B4),

CD138-APC (BD Pharmingen, 281-2), CD5-PerCP-Cy5.5 (Ebioscience, 53-7.3), CD19-

PE (Ebioscience, eBio1D3), CD11b-eFluor450 (Ebioscience, M1/70), TER119-FITC

(Biolegend, TER119), Gr-1-PE (Biolegend, RB6-8C5) and F4/80-PerCP (Biolegend,

BM8).

Bone marrow transplantation. ERdj4+/+ and ERdj4GT/GT mice were exposed to two

doses of irradiation (7 and 4.75 Gy) and intravenously transplanted with ERdj4GT/GT

bone marrow (5×106 cells/200 µl). B cell populations in the bone marrow and spleen

were analyzed 8 and/or 16 weeks after transplantation by flow cytometry. Bone marrow

aspiration was performed 8 weeks after transplantation by the Cincinnati Children’s

Comprehensive Mouse and Core.

Quantitative Real-Time PCR. Total RNA was extracted from cells using the RNeasy

Plus Mini Kit (Qiagen) and cDNA was synthesized using iScript cDNA Synthesis Kit

(Bio-Rad). Quantitative RT-PCR was performed with 25 ng of cDNA per reaction on the

ABI 7300 system with TaqMan Assays (Applied Biosystems) for mouse ERdj4

(Mm01622956_s1), custom-designed spliced XBP1 (5’-GGC CGG GTC TGC TGA GT-

3’; 3’-CTG AAG AGG CAA CAG TGT CAG AGT; probe, CGC AGC AGG TGC AGG

CCC A), BiP (Hspa5, Mm00517690_g1), osteocalcin (Bglap1, Mm03413826_mH),

Osterix (Sp7, Mm04209856_m1), type 1 collagen (Col1a1, Mm00801666_g1), RANKL

(Tnfsf11, Mm00441906_m1) and mouse β-actin (endogenous control,

Mm00607939_s1). Relative quantitation was determined using the SDS software

(Applied Biosystems).

128 Cell culture. MEFs were isolated from ERdj4+/+ and ERdj4GT/GT embryos and cultured in DMEM, as previously described (Chapter 1). For osteoblast differentiation studies,

MEFs were plated in 24- or 96-well plates and treated with recombinant human BMP2

(Roche, 200 ng/mL) for up to 96 hours. ALP activity was determined by incubating cell lysates with p-nitrophenyl phosphate (Pierce) at room temperature for 30 minutes. The reaction was stopped with addition of 2N NaOH and absorbance was measured at 405 nm on a Synergy 2 microplate reader (BioTek Instraments, Inc.). For pre-B colony forming assays, total bone marrow (1×105 cells) was cultured in MethoCult media (Stem

Cell Technologies, M3630) for 7 days at 37°C before colonies were counted under a light microscope (Zeiss) using low magnification (4X).

Statistical analysis. All experiments were performed at least twice, unless otherwise stated. Data are presented as mean ± SEM and were analyzed by a two-tailed

Student’s t test using GraphPad Prism software, where P ≤ 0.05 was considered statistically significant.

129 Acknowledgements

We thank Jessica Allen for preliminary B cell experiments, Fred Finkelman for providing the IgD antibody and Sheila Bell for skeletal staining. This study was supported by

National Institute of Health Grants HL103923 (T.E.W.) and HL086492 (T.E.W.).

130 Figure Legends

Figure 1. Bone-lining cells are reduced in ERdj4GT/GT mice. (A) H&E staining of

femur tissue from 6-8-week-old mice. Bone-lining cells are present on the cortical bone

surface (arrows). Inset is 100X and scale bars are 20 µm. (B) Confocal microscopy of

osteocalcin (red) and GFP (green) protein in femur tissue sections from 6-8-week-old

ERAI+ mice. Scale bars are 10 µm.

Figure 2. Primary osteogenic cells from ERdj4GT/GT mice have an immature

phenotype. (A) Diff-Quik staining of osteogenic cells cultured in growth media for 16-

18 days. Scale bars are 20 µm. (B) Osteocalcin immunohistochemistry of osteogenic

cells cultured in growth media for 16-18 days. Scale bars are 20 µm. (C) Quantitation of

osteocalcin, Osterix, type 1 collagen (Col1a1) and RANKL mRNAs in osteogenic cells

cultured in growth media for 16-18 days. Mouse β-actin was used as an endogenous

control. N = 3 samples/group, representative of two independent experiments, *P ≤

0.05, **P ≤ 0.01, ***P ≤ 0.001. (D) Quantitation of Osterix and osteocalcin mRNAs in

ERdj4GT/GT osteogenic cells cultured in growth media for 16-18 days (d0) or treated with differentiation media for an additional 21 days (d21). Mouse β-actin was used as an endogenous control. RQ, relative quantitation. N = 3 samples/group, **P ≤ 0.01. (E)

Quantitation of BiP and sXBP1 mRNAs in osteogenic cells cultured in growth media for

16-18 days. Mouse β-actin was used as an endogenous control. N = 8 samples/group, representative of two independent experiments, *P ≤ 0.05, ***P ≤ 0.001.

Figure 3. B cell development is impaired in ERdj4GT/GT mice. (A) Total bone marrow

cells isolated from the femurs of 6-week-old mice. N = 20 mice/genotype, *P ≤ 0.05.

(B) Bone marrow cells isolated from the femurs of 6-week-old mice were stained

131 antibodies against IgM, B220 and CD43. Frequency of bone marrow pro-B (IgM-

B220+CD43+), pre-B (IgM-B220+CD43-), immature (IMB, B220lowIgM+) and mature (MB,

B220highIgM+) B cells were analyzed by flow cytometry. Contour plots are representative of each genotype. (C) Absolute numbers of each bone marrow B cell subset. N = 20 mice/genotype pooled from four independent experiments, *P ≤ 0.05, **P ≤ 0.01, ***P ≤

0.001. (D) Pro-B cells were further differentiated based on HSA and BP-1 expression

(fractions A through C). N = 20 mice/genotype pooled from four independent experiments, *P ≤ 0.05, ***P ≤ 0.001. (E) B cells were isolated from the femurs of 6- week-old mice, stained with 7-AAD and analyzed by flow cytometry. N = 6 mice/genotype, *P ≤ 0.05, **P ≤ 0.01.

Figure 4. ERdj4 deficiency in the bone marrow microenvironment affects B cell development. Recipient mice received two doses of lethal irradiation followed by intravenous injection of ERdj4GT/GT bone marrow cells. (A) Bone marrow pro-B (IgM-

B220+CD43+), pre-B (IgM-B220+CD43-), immature (IMB, B220lowIgM+) and mature (MB,

B220highIgM+) B cells were quantified by flow cytometry 8 weeks after transplantation. N

= 5-6 mice/genotype, *P ≤ 0.05, **P ≤ 0.005. (B) Frequencies of fraction A (BP-1-HSA-),

fraction B (BP-1+HSA-) and fraction C (BP-1+HSA+) were determined by gating on pro-B

cells. N = 5-6 mice/genotype, *P ≤ 0.05. (C) Pre-B colony forming units were assayed

by culturing total bone marrow in MethoCult media containing IL-7. N = 5-7

mice/genotype.

Figure 5. Mature B cells are decreased in the spleen and peritoneal cavity of

ERdj4GT/GT mice. (A) Total splenic leukocytes were isolated from 6-week-old mice. N =

9-12 mice/genotype pooled from two independent experiments, *P ≤ 0.05. (B)

132 Frequency of splenic immature (IMB, B220+CD93+), mature (MB, B220+CD93-),

follicular (FOB, B220+CD21lowCD23+) and marginal zone (MZB, B220+CD21+CD23low) B cells were analyzed by flow cytometry. Contour plots are representative of each genotype. (C) Absolute numbers of splenic B cell subsets. N = 9-12 mice/genotype, **P

≤ 0.01. (D) Recipient mice received two doses of lethal irradiation followed by intravenous injection of ERdj4GT/GT bone marrow cells. Splenic B cell populations were

quantified by flow cytometry 16 weeks after transplantation. N = 5-6 mice/genotype,

***P ≤ 0.001. (E) Peritoneal B1 (CD19+CD11b+) B cells were quantified in 6-week-old mice by flow cytometry. B1a and B1b subsets were differentiated based on CD5 expression. N = 16 mice/genotype pooled from three independent experiments, *P ≤

0.05. (F) Serum BAFF levels were determined in 6-week-old mice by ELISA. N = 6-7

mice/genotype, *P ≤ 0.05.

S1. Skeletal development of ERdj4+/+ and ERdj4GT/GT embryos. Alizarin red (bone)

and Alcian blue (cartilage) staining of mouse skeletons at E18.5. Scale bars are 0.25

cm. N = 4 mice/genotype.

S2. BMP2-treated ERdj4GT/GT MEFs have reduced osteoblast differentiation

potential. (A) Quantitation of ERdj4 mRNA in ERdj4+/+ MEFs treated with rhBMP2.

Mouse β-actin was used as an endogenous control. RQ, relative quantitation. N = 3

samples/group, *P ≤ 0.05, **P ≤ 0.01, ***P ≤ 0.001. (B-C) BiP and sXBP1 mRNA

expression was assessed in ERdj4+/+ and ERdj4GT/GT MEFs treated with rhBMP2 for 96

hours. Mouse β-actin was used as an endogenous control. N = 3 samples/group, *P ≤

0.05, ***P ≤ 0.001. (D) ALP activity was determined in ERdj4+/+ and ERdj4GT/GT MEFs treated with rhBMP2 for 96 hours. N = 3 samples/genotype, **P ≤ 0.01. (E)

133 Quantitation of osteocalcin, Osterix and type 1 collagen (Col1a1) mRNAs by qRT-PCR

in ERdj4+/+ and ERdj4GT/GT MEFs treated with rhBMP2 for 96 hours. Mouse β-actin was used as an endogenous control. ND, not detected. Mean +/- SEM of triplicate cultures are depicted, *P ≤ 0.05, **P ≤ 0.01.

S3. B cells and erythrocytes are decreased in the bone marrow of ERdj4GT/GT

mice. (A-B) Bone marrow cells isolated from the femurs of 6-week-old mice were

stained with antibodies against Gr-1, F4/80, Ter119 and B220 and analyzed by flow

cytometry. N = 6-21 mice/genotype pooled from at least two independent experiments,

*P ≤ 0.05, **P ≤ 0.01, ***P ≤ 0.001. (C) Absolute number of bone marrow B cells from

6-week-old mice. N = 20 mice/genotype pooled from four independent experiments, **P

≤ 0.01, ***P ≤ 0.001.

S4. Bone marrow B cell proliferation and peripheral pro-B cells are normal in

ERdj4GT/GT mice. (A) Mice were intraperitoneally injected with BrdU (1 mg) 24 and 12

hours before sacrifice. Bone marrow B cell incorporation of BrdU was assessed by flow

cytometry. N = 9-10 mice/genotype. (B) Pro-B cells (IgM-B220+CD43+) were assessed

in the peripheral blood of 6-week-old mice by flow cytometry. N = 6 mice/genotype.

S5. Plasma cell differentiation and antibody production are normal in ERdj4GT/GT

mice. (A) Mice were intraperitoneally injected with 200 µl of goat anti-IgD antiserum and

the frequency of splenic plasma cells was determined 8 days post-immunization by flow

cytometry. N = 5-8 mice/genotype, **P ≤ 0.01. (B) Baseline immunoglobulin levels

were determined by ELISA in 4-6-month-old mice. N = 5-9 mice/genotype. (C-D) Mice

were intraperitoneally injected with TNP-CGG (C, 100 µg) or TNP-Ficoll (D, 50 µg) and

134 TNP-specific IgM and IgG antibody production was determined by ELISA. N = 5-7 mice/genotype.

135 A ERdj4+/+ ERdj4GT/GT

B ERAI Osteocalcin Merge +/+ ERdj4 GT/GT GT/GT ERdj4

Figure 1 136 A ERdj4+/+ ERdj4GT/GT

Quik Diff-

B Osteocalcin

C 1.0 ** ***** +/+ ) * * * ERdj4 Q * GT/GT

R 0.8 ERdj4 (

) 0.8 n Q o i R

( 0.6 s

s A 0.6 e N r p R 0.4 x m e

0.4

e 0.2 v i t a

l 0.2

e 0.0

R Osteocalcin Osterix Col1a1 RANKL 0.0 Osteocalcin Osterix Col1a

D 8 E *** d0 ** 2.5 ERdj4+/+ GT/GT

) d21 6 ERdj4 Q 2.0 ) R ( Q

R A *

4 ( 1.5

N A R N

m 1.0 2 ns R m 0 0.5 Osterix Osteocalcin 0.0 ERdj4GT/GT BiP sXBP-1

Figure 2 137 A B Gated on: IgM- Live cells Pre-B Pro-B 40 * 2.6% 21.6% 4.0% MB ) 6 0 1

30 +/+

x ERdj4 (

s

l 8.0% IMB l e

c 20

M B

l a B220 t 10 Pre-B Pro-B o MB T 2.0% 11.4% 1.6%

0 +/+ GT/GT ERdj4 ERdj4 ERdj4GT/GT 5.8% IMB

CD43 IgM C *** ** 30 64 8 * 12 ** 56

) 48

5 6

0 20 8

1 40

x ( 32 4 s l l e 24 10 4 C 16 2 1.0 ** +/+ 8 ) * ERdj4 Q * GT/GT

0 0 R 0 ERdj4

( 0 0.8

Pro-B Pren -B IMB MB o i s

s 0.6

D e r

3.0 p 15 x * 50 *** e

0.4 ) ) ) 4 5 5 2.5 e 0 0 0 v 40 i 1 1 1 t

x x x a ( ( l

( 2.0

10 0.2

- + e + 30 A A A 1.5 R S S S H H H 0.0 - - 20 + 1 1.0 1 5 1 -

- Osteocalcin Osterix Col1a - P P P +/+ B 1.0B 10 ** ) 0.5 B * ERdj4 Q * GT/GT

0.0 R ERdj4 ( 0 0 0.8

Fraction A n Fraction B Fraction C o i s s 0.6 ns E e r

20 ** p * 16 * x e

0.4 e ) v ) 15 i 12 t % % ( a

( l 0.2 + + e D D

10 R 8 A A A - A

0.0 - 7 5 Osteocalcin Osterix7 4 Col1a 1.0 ** +/+ ) * ERdj4 Q * GT/GT

0 R 0 ERdj4 ( Total B220B220++ B220B2200.8lolow B220B220highhi Pro-B n o i s

s 0.6 e r

Figure 3 p x e

0.4

e 138 v i t a

l 0.2 e R 0.0 Osteocalcin Osterix Col1a A B )

GT GT - 15 ! 10 GT ! GT * M *

GT ! WT g I 8 GT ! WT % t (

n 10 ** + e

r 6 3 a 0.07 4 P

D 4 * % 5 C +

0 2 * 2 2

0 B 0 Pro-B Pre-B IMB MB A B C

C

150 s l l e c

5 0

1 100 ! 1 / s U F

C 50

B - e r P 0 ERdj4+/+ ERdj4GT/GT

Figure 4 139 A B ERdj4+/+ ERdj4GT/GT MB MB

) 32.7% 26.8%

7 15 * 0 1

x ( B220

s IMB IMB

e 11.7% 11.5% t 10 y c o k CD93 u

e MZB MZB l 5 4.8% 6.3% c i n e l p CD21 S 0 FOB FOB +/+ GT/GT ERdj4 ERdj4 59.0% 43.5%

CD23 C D *** 50 +/+ GT GT ) ERdj4 60 ! 6 ** ** 0 GT/GT GT ! WT

1 ERdj4

40 x

( 45

s l

l *** 30 e v e i c

L

30 B

20 % c i n e l 10 15 p S 0 0 IMB MB FOB MZB IMB MB FOB MZB

1.0 E ** +/+ F ) * ERdj4 Q * GT/GT

R ERdj4 * ( 0.8 8 n o i * s 35 s 0.6 e ) r * 4 p 30 ) 6 0 x l 1 e

0.4 m x / e

( 25

g v i t s n l ( a l l 0.2 20 4 e e F c

R F 1 0.0 15 A B B Osteocalcin Osterix Col1a

C 10 2 r e

P 5 0 0 B1 B1-a B1-b ERdj4+/+ ERdj4GT/GT

Figure 5 140 A ERdj4+/+ ERdj4GT/GT

Supplemental Figure S1 141 +/+ A MEFs B 2.5 *** ERd+j/4+ 2.5 *** * ERdj4 GT/GT 2.8 * * ERdGjT4/GT ) ERdj4

) 2.0 ) 2.4 Q Q ** 2.0 Q R ( R

( R ***

2.0 ( A *** 1.5 *** A A N 1.5 N 1.6 N R R R

m 1.0 m

1.2

m 1.0

P 4 i j P d i 0.8 B

R 0.5 B

E 0.4 0.5 0.0 0.0 0.0 Untreated BMP2 0 24 48 72 96 Untreated BMP2 Time after BMP2 treatment (hours) MEFs MEFs

C D +/+ 20 4 *** ERdj4 ERdj4+/+ **

GT/GT ) ) *** ERdj4 GT/GT g

Q ERdj4 m R / ( 3 15

U A

*** (

N y

*** t R i

2 v

m 10 i

t 1 c - a P

B 1 P 5 X L s A 0 ** Untreated BMP2 0 Untreated BMP2 MEFs MEFs

E

** * ) 250 +/+ ** 1000 ERdj4+/+ ERdj4 4 +/+ Q

) ERdj4 GT/GT GT/GT ) R

Q ERdj4 GT/GT ( ERdj4 Q 200 ERdj4 800 R R ( A (

3 N A A R N 600 150 N m R R 2 n m m i

c x l 100 400 1 i r a a c e 1 t l

o 1 s o

e 200 50 t O C s *

O ND 0 0 0 Untreated BMP2 Untreated BMP2 Untreated BMP2 MEFs MEFs MEFs

Supplemental Figure S2 142 A 1.0B ** +/+ ) * ERdj4

Q *** 1.0 ** +/+ * GT/GT ) 80 ERdj4 R ERdj4

* (

Q * GT/GT 0.8 30 n

R ERdj4 o ( i

) ** 0.8 s n ) M s 0.6 o 25 e i 60 B r

% s p (

s * e

0.6 x e v s e i r 20

0.4 l l p L e x v e i t

e 40 c %

0.4

a

( 15

l 0.2 e e e v + i v t R 0 i a l 0.2 2 10 L 0.0 e 20 2 Osteocalcin Osterix Col1a ** R B 0.0 5 Osteocalcin Osterix Col1a 0 0 Gr-1+ F4/80+ Ter119+ Total B220+ B220low B220high

1.0 C ** +/+ ) * ERdj4 Q * *** GT/GT

R 100 ERdj4 ( 0.8 n o ** i s

) 80 s

0.6 5 e r 0 p 1 x x e

( 60

0.4 e s l v l i t e a l 0.2 c 40 e M R 0.0 B ** Osteocalcin20 Osterix Col1a 0 Total B220+ B220low B220high

Supplemental Figure S3 143 1.0 ** +/+ A ) * ERdj4 Q * GT/GT

R ERdj4 ( 0.8 72 60 n o i

s 60 s 0.6 e r ) 45 p x

) 48 % e (

0.4

% ( e

+ v +

i 36 U 30 t U d a l r

d 0.2 r e B

B 24 R 15 0.0 Osteocalcin12 Osterix Col1a 0 0 Total B220+ B220low B220high Pro-B Pre-B

B 0.5 ) e v i L 0.4 % (

+

3 0.3 4 D C

+ 0.2 0 2 2 B - 0.1 M g I 0.0 ERdj4+/+ ERdj4GT/GT

Supplemental Figure S4 144 1.0 ** +/+ +/+ ) A * ERdj4 B 2.5 ERdj4

Q * GT/GT GT/GT

R ERdj4

( ERdj4 0.8

n 2.0 o )

i ** l s m s

0.6 ** 4 / 2.5 e 1.5 r

2.5 g ) p n e ) ( x v l

i e

0.4 2.0 E L

m 1.0

e 2.0 / 3 g v I g i % t ( a L m l 0.2 + ( 0.5 1.5 e 1.5 m 8 / g R 3 I 2 g

1

0.0 n D 1.0 m 0.0 1.0

Osteocalcin Osterix Col1a u C r IgE + e

9 1 S 1 0.5 0.5 D C 0.0 0 0.0 Untreated g!mIg-D IgM IgG IgA IgE

ERdj4+/+ GT/GT C ERdj4 2.5 ERdj4+/+ ERdj4GT/GT 2.0 ) l 2.5 ) ) 2.0 m m /

m 1.5 g n n

n

( 0

0 2.0 5 5 1.5 E 1.0 4 4 g I ( (

1.5 M G 0.5 g g I I

1.0 G 1.0 G 0.0 G G

C IgE - C

- 0.5 P 0.5 P N N T 0.0 T 0.0 -3 7 14 21 -3 7 14 21 Time (days) Time (days) D 2.5 ERdj4+/+

) 3.0 ERdj4GT/GT ) 2.5 m 2.0 n ) m l

n 0

m / 5 0 1.5 2.0 g 4 5 n ( (

4

2.0 (

E G 1.0 g g M 1.5 I I

g l I l

l 0.5 o l c

o 1.0 i 1.0 c i F

- 0.0 F - 0.5 P IgE P N N T

T 0.0 0.0 -3 7 14 21 -3 7 14 21 Time (days) Time (days)

Supplemental Figure S5 145 References

1. Harding, H. P., Zhang, Y. & Ron, D. Protein translation and folding are coupled by an endoplasmic-reticulum-resident kinase. Nature 397, 271-274 (1999).

2. Haze, K., Yoshida, H., Yanagi, H., Yura, T. & Mori, K. Mammalian transcription factor ATF6 is synthesized as a transmembrane protein and activated by proteolysis in response to endoplasmic reticulum stress. Mol Biol Cell 10, 3787- 3799 (1999).

3. Yoshida, H., Haze, K., Yanagi, H., Yura, T. & Mori, K. Identification of the cis-acting endoplasmic reticulum stress response element responsible for transcriptional induction of mammalian glucose-regulated proteins. Involvement of basic leucine zipper transcription factors. J Biol Chem 273, 33741-33749 (1998).

4. Shen, X. et al. Complementary signaling pathways regulate the unfolded protein response and are required for C. elegans development. Cell 107, 893-903 (2001).

5. Yoshida, H., Matsui, T., Yamamoto, A., Okada, T. & Mori, K. XBP1 mRNA is induced by ATF6 and spliced by IRE1 in response to ER stress to produce a highly active transcription factor. Cell 107, 881-891 (2001).

6. Back, S. H. & Kaufman, R. J. Endoplasmic reticulum stress and type 2 diabetes. Annu Rev Biochem 81, 767-793 (2012).

7. Hamman, B. D., Hendershot, L. M. & Johnson, A. E. BiP maintains the permeability barrier of the ER membrane by sealing the lumenal end of the translocon pore before and early in translocation. Cell 92, 747-758 (1998).

8. Lievremont, J. P., Rizzuto, R., Hendershot, L. & Meldolesi, J. BiP, a major chaperone protein of the endoplasmic reticulum lumen, plays a direct and important role in the storage of the rapidly exchanging pool of Ca2+. J Biol Chem 272, 30873-30879 (1997).

9. Brodsky, J. L. et al. The requirement for molecular chaperones during endoplasmic reticulum-associated protein degradation demonstrates that protein export and import are mechanistically distinct. J Biol Chem 274, 3453-3460 (1999).

10. Hendershot, L. et al. Inhibition of immunoglobulin folding and secretion by dominant negative BiP ATPase mutants. Proc Natl Acad Sci U S A 93, 5269-5274 (1996).

11. Plemper, R. K., Bohmler, S., Bordallo, J., Sommer, T. & Wolf, D. H. Mutant analysis links the translocon and BiP to retrograde protein transport for ER degradation. Nature 388, 891-895 (1997).

146 12. Simons, J. F., Ferro-Novick, S., Rose, M. D. & Helenius, A. BiP/Kar2p serves as a molecular chaperone during carboxypeptidase Y folding in yeast. J Cell Biol 130, 41-49 (1995).

13. Bertolotti, A., Zhang, Y., Hendershot, L. M., Harding, H. P. & Ron, D. Dynamic interaction of BiP and ER stress transducers in the unfolded-protein response. Nat Cell Biol 2, 326-332 (2000).

14. Shen, J., Chen, X., Hendershot, L. & Prywes, R. ER stress regulation of ATF6 localization by dissociation of BiP/GRP78 binding and unmasking of Golgi localization signals. Dev Cell 3, 99-111 (2002).

15. Fewell, S. W., Travers, K. J., Weissman, J. S. & Brodsky, J. L. The action of molecular chaperones in the early secretory pathway. Annu Rev Genet 35, 149- 191 (2001).

16. Jin, Y., Awad, W., Petrova, K. & Hendershot, L. M. Regulated release of ERdj3 from unfolded proteins by BiP. EMBO J 27, 2873-2882 (2008).

17. Petrova, K., Oyadomari, S., Hendershot, L. M. & Ron, D. Regulated association of misfolded endoplasmic reticulum lumenal proteins with P58/DNAJc3. EMBO J 27, 2862-2872 (2008).

18. Shen, Y. & Hendershot, L. M. ERdj3, a stress-inducible endoplasmic reticulum DnaJ homologue, serves as a cofactor for BiP's interactions with unfolded substrates. Mol Biol Cell 16, 40-50 (2005).

19. Kampinga, H. H. & Craig, E. A. The HSP70 chaperone machinery: J proteins as drivers of functional specificity. Nat Rev Mol Cell Biol 11, 579-592 (2010).

20. Otero, J. H., Lizak, B. & Hendershot, L. M. Life and death of a BiP substrate. Semin Cell Dev Biol 21, 472-478 (2010).

21. Shen, Y., Meunier, L. & Hendershot, L. M. Identification and characterization of a novel endoplasmic reticulum (ER) DnaJ homologue, which stimulates ATPase activity of BiP in vitro and is induced by ER stress. J Biol Chem 277, 15947-15956 (2002).

22. Dong, M., Bridges, J. P., Apsley, K., Xu, Y. & Weaver, T. E. ERdj4 and ERdj5 are required for endoplasmic reticulum-associated protein degradation of misfolded surfactant protein C. Mol Biol Cell 19, 2620-2630 (2008).

23. Lai, C. W., Otero, J. H., Hendershot, L. M. & Snapp, E. ERdj4 protein is a soluble endoplasmic reticulum (ER) DnaJ family protein that interacts with ER-associated degradation machinery. J Biol Chem 287, 7969-7978 (2012).

147 24. Jang, W. G. et al. BMP2 protein regulates osteocalcin expression via Runx2- mediated Atf6 gene transcription. J Biol Chem 287, 905-915 (2012).

25. Murakami, T. et al. Signalling mediated by the endoplasmic reticulum stress transducer OASIS is involved in bone formation. Nat Cell Biol 11, 1205-1211 (2009).

26. Delepine, M. et al. EIF2AK3, encoding translation initiation factor 2-alpha kinase 3, is mutated in patients with Wolcott-Rallison syndrome. Nat Genet 25, 406-409 (2000).

27. Thornton, C. M., Carson, D. J. & Stewart, F. J. Autopsy findings in the Wolcott- Rallison syndrome. Pediatr Pathol Lab Med 17, 487-496 (1997).

28. Wei, J., Sheng, X., Feng, D., McGrath, B. & Cavener, D. R. PERK is essential for neonatal skeletal development to regulate osteoblast proliferation and differentiation. J Cell Physiol 217, 693-707 (2008).

29. Zhang, P. et al. The PERK eukaryotic initiation factor 2 alpha kinase is required for the development of the skeletal system, postnatal growth, and the function and viability of the pancreas. Mol Cell Biol 22, 3864-3874 (2002).

30. Tanaka, T. et al. Targeted disruption of ATF4 discloses its essential role in the formation of eye lens fibres. Genes Cells 3, 801-810 (1998).

31. Yang, X. et al. ATF4 is a substrate of RSK2 and an essential regulator of osteoblast biology; implication for Coffin-Lowry Syndrome. Cell 117, 387-398 (2004).

32. Mercier, F. E., Ragu, C. & Scadden, D. T. The bone marrow at the crossroads of blood and immunity. Nat Rev Immunol 12, 49-60 (2012).

33. Visnjic, D. et al. Hematopoiesis is severely altered in mice with an induced osteoblast deficiency. Blood 103, 3258-3264 (2004).

34. Zhu, J. et al. Osteoblasts support B-lymphocyte commitment and differentiation from hematopoietic stem cells. Blood 109, 3706-3712 (2007).

35. Schmidt, I. U., Dobnig, H. & Turner, R. T. Intermittent parathyroid hormone treatment increases osteoblast number, steady state messenger ribonucleic acid levels for osteocalcin, and bone formation in tibial metaphysis of hypophysectomized female rats. Endocrinology 136, 5127-5134 (1995).

36. Calvi, L. M. et al. Osteoblastic cells regulate the haematopoietic stem cell niche. Nature 425, 841-846 (2003).

148 37. Jung, Y. et al. Regulation of SDF-1 (CXCL12) production by osteoblasts; a possible mechanism for stem cell homing. Bone 38, 497-508 (2006).

38. Wu, J. Y. et al. Osteoblastic regulation of B lymphopoiesis is mediated by Gs{alpha}-dependent signaling pathways. Proc Natl Acad Sci U S A 105, 16976- 16981 (2008).

39. Saito, A. et al. Endoplasmic reticulum stress response mediated by the PERK- eIF2(alpha)-ATF4 pathway is involved in osteoblast differentiation induced by BMP2. J Biol Chem 286, 4809-4818 (2011).

40. Tohmonda, T. et al. The IRE1alpha-XBP1 pathway is essential for osteoblast differentiation through promoting transcription of Osterix. EMBO Rep 12, 451-457 (2011).

41. Iwawaki, T., Akai, R., Kohno, K. & Miura, M. A transgenic mouse model for monitoring endoplasmic reticulum stress. Nat Med 10, 98-102 (2004).

42. Nakashima, T. et al. Evidence for osteocyte regulation of bone homeostasis through RANKL expression. Nat Med 17, 1231-1234 (2011).

43. Chen, D. et al. Bone morphogenetic protein 2 (BMP-2) enhances BMP-3, BMP-4, and bone cell differentiation marker gene expression during the induction of mineralized bone matrix formation in cultures of fetal rat calvarial osteoblasts. Calcif Tissue Int 60, 283-290 (1997).

44. Matsubara, T. et al. BMP2 regulates Osterix through Msx2 and Runx2 during osteoblast differentiation. J Biol Chem 283, 29119-29125 (2008).

45. Hardy, R. R., Carmack, C. E., Shinton, S. A., Kemp, J. D. & Hayakawa, K. Resolution and characterization of pro-B and pre-pro-B cell stages in normal mouse bone marrow. J Exp Med 173, 1213-1225 (1991).

46. Pillai, S. & Cariappa, A. The follicular versus marginal zone B lymphocyte cell fate decision. Nat Rev Immunol 9, 767-777 (2009).

47. Montecino-Rodriguez, E. & Dorshkind, K. B-1 B cell development in the fetus and adult. Immunity 36, 13-21 (2012).

48. Reimold, A. M. et al. Plasma cell differentiation requires the transcription factor XBP-1. Nature 412, 300-307 (2001).

49. Taubenheim, N. et al. High Rate of Antibody Secretion Is not Integral to Plasma Cell Differentiation as Revealed by XBP-1 Deficiency. J Immunol 189, 3328-3338 (2012).

149 50. Lee, A. H., Iwakoshi, N. N. & Glimcher, L. H. XBP-1 regulates a subset of endoplasmic reticulum resident chaperone genes in the unfolded protein response. Mol Cell Biol 23, 7448-7459 (2003).

51. Yu, S. et al. Critical role of activating transcription factor 4 in the anabolic actions of parathyroid hormone in bone. PLoS One 4, e7583 (2009).

52. Bianco, P. Bone and the hematopoietic niche: a tale of two stem cells. Blood 117, 5281-5288 (2011).

53. Short, B. J., Brouard, N. & Simmons, P. J. Prospective isolation of mesenchymal stem cells from mouse compact bone. Methods Mol Biol 482, 259-268 (2009).

54. Franz-Odendaal, T. A., Hall, B. K. & Witten, P. E. Buried alive: how osteoblasts become osteocytes. Dev Dyn 235, 176-190 (2006).

55. Reimold, A. M. et al. An essential role in liver development for transcription factor XBP-1. Genes Dev 14, 152-157 (2000).

56. Zhang, K. et al. The unfolded protein response sensor IRE1alpha is required at 2 distinct steps in B cell lymphopoiesis. J Clin Invest 115, 268-281 (2005).

57. Tokoyoda, K., Egawa, T., Sugiyama, T., Choi, B. I. & Nagasawa, T. Cellular niches controlling B lymphocyte behavior within bone marrow during development. Immunity 20, 707-718 (2004).

58. Cariappa, A. et al. Perisinusoidal B cells in the bone marrow participate in T- independent responses to blood-borne microbes. Immunity 23, 397-407 (2005).

59. Cariappa, A., Chase, C., Liu, H., Russell, P. & Pillai, S. Naive recirculating B cells mature simultaneously in the spleen and bone marrow. Blood 109, 2339-2345 (2007).

60. Sapoznikov, A. et al. Perivascular clusters of dendritic cells provide critical survival signals to B cells in bone marrow niches. Nat Immunol 9, 388-395 (2008).

61. Esplin, B. L., Welner, R. S., Zhang, Q., Borghesi, L. A. & Kincade, P. W. A differentiation pathway for B1 cells in adult bone marrow. Proc Natl Acad Sci U S A 106, 5773-5778 (2009).

62. Tompkins, D. H. et al. Sox2 is required for maintenance and differentiation of bronchiolar Clara, ciliated, and goblet cells. PLoS One 4, e8248 (2009).

63. Ray, A. & Dittel, B. N. Isolation of mouse peritoneal cavity cells. J Vis Exp (2010).

150

CHAPTER IV

Discussion and Future Directions

151 Summary

ERdj4 facilitates the removal of unfolded/misfolded substrates from the ER lumen

for proteasomal degradation. Previous studies identified ERdj4 as a chaperone that

was ubiquitously expressed at low levels under normal conditions, but rapidly

upregulated in response to stress. However, herein we identify an important role for

ERdj4 in postnatal survival, glucose metabolism and B cell development in vivo.

Approximately half of ERdj4 gene trap (ERdj4GT/GT) mice died perinatally in association with growth retardation and hypoglycemia. Altered distribution of pancreatic

α- and β-cells, as well as decreased insulin and glucagon in the pancreas, indicated a

delay in pancreatic development in ERdj4GT/GT neonates. Other common causes of

perinatal lethality, including defects in respiration and suckling, were excluded in

ERdj4GT/GT neonates. Collectively, these results suggest that ERdj4 is required for adaptation to metabolic stress in the neonatal period.

Mice that survived into adulthood were glucose intolerant due to hypoinsulinemia, rather than insulin resistance. Pancreatic β-cells from ERdj4GT/GT mice exhibited

increased levels of ER stress, as indicated by cytoplasmic vacuolation, ER dilation and upregulation of the UPR. A previous study in our laboratory identified insulin as a

substrate for ERdj41; the current work identified other ERdj4 substrates including the

insulin-processing enzymes, Pcsk1, Pcsk2 and carboxypeptidase E (CPE).

Furthermore, the loss of ERdj4 resulted in accumulation of proinsulin in the ER, which

likely contributed to β-cell stress and dysfunction. Overall, these results indicate that

the chaperone activity of ERdj4 is essential for β-cell homeostasis, insulin biogenesis

and glucose metabolism.

152 Since the UPR influences several stages of B cell development2,3, we hypothesized that B cell maturation would also be impaired in ERdj4GT/GT mice. The

loss of ERdj4 resulted in fewer developing B cells in the bone marrow in association

with increased cell death. Further, several mature B cell populations, including follicular

and B1b B cells, were reduced in the spleen and peritoneal cavity, respectively.

Surprisingly, ERdj4 deficiency did not affect antibody secretion by plasma cells, even

though expression of ERdj4 was increased during plasma cell differentiation4.

Bone marrow chimeras revealed that a defect in the microenvironment of

ERdj4GT/GT mice affected B cell development in the bone marrow and periphery. Given

the role of the UPR in osteoblast differentiation and function, we hypothesized that

osteogenic cells were the defective stromal cell type in ERdj4GT/GT mice. Osteogenic

cells support B lymphocyte commitment and development through the expression of

adhesion molecules and cytokines5,6. Osteogenic cells isolated from ERdj4GT/GT mice

exhibited reduced expression of osteoblast-specific markers and increased levels of ER

stress. Importantly, bone-lining cells, which differentiate from osteoblasts, were

reduced in femur tissue sections of ERdj4GT/GT mice. These data support the hypothesis that reduced osteogenic cells in ERdj4GT/GT mice are responsible for the defect in B cell development.

Although previous studies demonstrated that ERdj4 facilitates the removal of unfolded/misfolded proteins from the ER lumen in vitro1,7,8, the biological relevance of

ERdj4 was unknown. This study highlights the significance of ERdj4 in vivo, revealing

an important role in survival, glucose metabolism and B cell development. The

questions raised by these findings will be addressed in the remainder of this discussion.

153 1. Gene trapping ERdj4: a blessing in disguise?

To determine the role of ERdj4 in vivo, ERdj4GT/GT mice were generated from embryonic stem cells harboring a gene trap vector randomly inserted into the ERdj4 locus. Gene trapping is a cost-effective and efficient experimental strategy for gene ablation. The International Gene Trap Consortium (IGTC) provides embryonic stem cells with identified trapped genes, eliminating the need to generate a targeting construct for homologous recombination in embryonic stem cells9,10. When the gene trap is inserted into an intron, the promoter of the “trapped” gene drives transcription of a fusion transcript, consisting of the upstream exon(s) spliced with the gene trap.

Sequencing analyses determined that the gene trap cassette inserted into intron 1 of the ERdj4 locus, which resulted in a hypomorphic allele. We hypothesized that splicing around the gene trap cassette led to transcription of some full-length, wildtype ERdj4; however, the presence of multiple transcripts in ERdj4GT/GT mice remains to be confirmed. A gene copy number assay determined that only one copy of the gene trap was present per haploid genome (Chapter 2, Figure 1C). This data was critical for the credibility of the ERdj4GT/GT phenotype, confirming that the gene trap disrupted only the

ERdj4 locus and not any other unknown loci.

In mice homozygous for the gene trap allele, expression of ERdj4 mRNA differed

among tissues and cell types examined by quantitative RT-PCR, but was 10-100 fold

lower compared to ERdj4+/+ controls (Chapter 2, Figure 1F). It is unclear why the

expression of ERdj4 varied in ERdj4GT/GT mice; however, it is possible that elevated ER

stress induced by ERdj4 deficiency increased ERdj4 promoter activity leading to

transcription of wildtype ERdj4 (related to splicing around the GT cassette).

154 Unfortunately, the level of ERdj4 protein in ERdj4GT/GT mice remains unknown because

of the lack of useful antibodies, including an antibody generated in our laboratory

against recombinant ERdj4. Erik Snapp et al. have alluded to the generation of a monoclonal antibody against ERdj4, which will be described in an upcoming publication and may help to resolve these unanswered questions7.

Approximately half of the ERdj4GT/GT mice died perinatally (Chapter 2, Tables 1-2)

and we hypothesize that hypomorphic expression of ERdj4 likely prevented fully

penetrant lethality. The surviving ERdj4 hypomorphic mice reached adulthood, but had

defects in glucose metabolism (Chapter 2) and B cell development (Chapter 3). Adult

mice also displayed signs of a vestibular disorder and female mice were infertile,

although these defects were not pursued in the current study. Overall, gene ablation of

ERdj4 using gene trapping instead of homologous recombination likely enabled the

study of ERdj4 in adult mice, revealing several interesting phenotypes that have yet to

be fully explored.

2. What is the function of ERdj4 in perinatal survival?

Perinatal lethality of ERdj4GT/GT mice was associated with growth retardation and hypoglycemia (Chapter 2, Figure 3A-C). After birth, pups endure a starvation period before the mother can provide nutrients from suckling. Survival during this time period depends on self-nourishment, which is mediated through glycogenolysis and the balance of insulin, glucagon and glucocorticoids to regulate metabolism11,12.

Interestingly, neonatal lethality and hypoglycemia were also observed in mice

expressing a nonphosphorylatable mutant of eIF2α, a component of the UPR signaling

155 pathway. These neonatal mice had reduced hepatic glycogen, although whether this

was the result of a defect in glycogen synthesis/storage or increased utilization of

glycogen remains unclear13. Glycogen storage was assessed in E18.5 livers of

ERdj4+/+ and ERdj4GT/GT mice by PAS staining (data not shown), but the result was

inconsistent and revealed no discernable difference between genotypes. A more

sensitive method to evaluate glycogen content will be pursued by using an assay that

quantitates glycogen in biological samples based on the production of glucose by

glucoamylase.

The underlying cause(s) of perinatal growth restriction in ERdj4GT/GT mice

(Chapter 2, Figure 3A-B) remains entirely unknown. However, since ERdj4 is highly

expressed in the placenta8, it is possible that defects in placental development affect nutrient and gas exchange leading to fetal growth restriction14. Further, the IRE1α

signaling pathway is essential for development of the labyrinth layer of the placenta,

which is the site of nutrient and gas exchange between the mother and fetus15. Since the IRE1α signaling pathway regulates expression of ERdj4, future studies will investigate whether ERdj4 deficiency affects placental development. Placental morphology will be analyzed by H&E staining in E18.5 ERdj4+/+ and ERdj4GT/GT mice.

Given that fetal essential fatty acids (EFAs) are obtained only from the mother, nutrient transport will be assessed in fetal ERdj4+/+ and ERdj4GT/GT mice by quantitating EFAs

through gas chromatography15. Interestingly, low birth weight is associated with type 2 diabetes in adulthood due to accelerated growth that alters food intake and glucose metabolism16,17. ERdj4GT/GT mice reached normal weight by 8 weeks of age (data not

156 shown), but were glucose intolerant, suggesting that growth retardation during

embryogenesis affects glucose homeostasis in adult ERdj4GT/GT mice.

Several lines of evidence indicated that ERdj4GT/GT neonates have a delay in

pancreatic development, including altered distribution of α- and β-cells and decreased

pancreatic insulin and glucagon (Chapter 2, Figure 3D-F). Further studies are needed

to confirm the defect in pancreatic development, including enumeration of α- and β- cells, as well as assessment of maturation markers (e.g. MafA and MafB). Plasma glucagon and insulin also need to be quantitated in newborn ERdj4GT/GT mice. Given

that ERdj4GT/GT neonates are hypoglycemic, we expect to observe lower levels of circulating glucagon, thereby reducing glycogenolysis in the liver.

Although other possible causes of neonatal lethality, including abnormal feeding and respiration, appeared normal in ERdj4GT/GT neonates (data not shown), it remains

unclear whether hypoglycemia directly caused their demise. To address this question,

glucose will be administered to neonatal mice during the starvation period, a procedure

that has previously been shown to rescue lethal hypoglycemia18. We hypothesize that

ERdj4GT/GT neonates that receive glucose will survive, thus confirming the link between

hypoglycemia and neonatal lethality.

3. ER stress and Type 2 Diabetes

Type 2 diabetes is a heterogeneous group of metabolic illnesses characterized

by high blood glucose levels due to insulin resistance in the periphery and/or impaired

insulin secretion by pancreatic β-cells. Although obesity and insulin resistance are

linked to type 2 diabetes, insulin-resistant individuals only develop the disease with the

157 onset of β-cell failure19. Current lifestyles, with increased consumption of energy-rich foods and reduced physical activity, have contributed to the prevalence of diabetes, affecting approximately 285 million adults worldwide. Further, global estimates predict a

54% increase in diabetes by 2030 due to population growth, ageing and urbanization associated with a sedentary lifestyle20. Although improved survival has been reported for other chronic, noninfectious diseases21, mortality attributed to diabetes increased

29% from 2007 to 2010, a frequency expected to rise as a result of increased

prevalence22. Unfortunately, the pathogenesis of diabetes is not well understood and

further insight is necessary for the prevention and treatment of disease.

Several studies have linked ER stress to β-cell dysfunction and type 2 diabetes23-

25. Chapter 2 highlights the importance of ERdj4 in glucose metabolism in vivo.

However, several questions remain including, whether ERdj4GT/GT mice develop diabetes with metabolic stress, how the loss of ERdj4 causes β-cell failure and whether

rescue of ERdj4 expression in pancreatic β-cells will correct the defects in glucose

metabolism. Further, the role of ERdj4 in pancreatic α-cells remains entirely unclear.

These questions and issues will each be addressed in turn.

3a. Do physiological stressors cause diabetes in ERdj4GT/GT mice?

Although fasting blood glucose levels were normal in young adult ERdj4GT/GT mice, they

were considered pre-diabetic based on glucose intolerance, hypoinsulinemia and an

increased proinsulin to insulin ratio (Chapter 2, Figure 6). It remains unknown whether

increased physiological β-cell stress, including pregnancy26, age27 or high-fat diet

(HFD)24,28, would promote diabetes in ERdj4GT/GT mice. Pregnancy cannot be used as

158 model of metabolic stress because female ERdj4GT/GT mice are infertile; however, aging

and HFD studies are concurrently underway. We hypothesize that age and/or HFD will

result in hyperglycemia in ERdj4GT/GT mice, given the β-cell dysfunction and prediabetic

phenotype already observed in these animals. Since insulin stimulates lipogenesis in

peripheral tissues and ERdj4GT/GT mice are hypoinsulinemic, it is possible that a HFD

will result in increased levels of circulating FFAs, which could promote cardiovascular

disease in ERdj4GT/GT mice.

If ERdj4GT/GT mice do not develop diabetes with physiological stressors, an

alterative approach would be to cross ERdj4GT/GT mice to the Akita mouse model of type

2 diabetes. These mice carry a C96Y mutation in the insulin 2 (insulin2C96Y) gene, resulting in disruption of disulfide bond formation and accumulation of misfolded protein.

Akita mice develop hyperglycemia and hypoinsulinemia at approximately 4 weeks of

age29. Our laboratory previously showed that ERdj4 facilitates the removal of insulin2C96Y from the ER1. Thus, we hypothesize that the loss of ERdj4 in Akita mice

would further increase accumulation of insulin2C96Y in the ER, resulting in an

exacerbated diabetic phenotype.

3b. What causes β-cell failure in ERdj4GT/GT mice?

Cytoplasmic vacuolation, ER dilation and upregulation of the UPR (Chapter 2, Figure 4),

indicated that ERdj4 deficiency caused ER stress in pancreatic β-cells. What remains

unclear, however, is whether increased ER stress results in apoptosis or

dedifferentiation of β-cells in ERdj4GT/GT mice. The UPR activates cell death pathways in response to protein accumulation in the ER that cannot be resolved through

159 attenuation of protein translation or upregulation of protein folding/degradation

machinery (Chapter 1, Section 2cii)24. β-cell death, apparent in several mouse models with genetic alterations in the UPR30,31, will be assessed in pancreatic tissue sections from ERdj4GT/GT mice by using the TUNEL assaya. However, a recent study by Talchai

et al. indicated that enhanced ER stress did not result in apoptosis, but instead caused

β-cell dedifferentiation to a progenitor phenotype, expressing Neurogenin3, Oct4,

Nanog and L-Myc32, that no longer produced insulin. Importantly, β-cell dedifferentiation also led to an increased number of α-cells, resulting in hyperglucagonemia. Based on the α-cell hyperplasia and hyperglucagonemia observed in ERdj4GT/GT mice, we hypothesize that increased ER stress causes β-cell dedifferentiation and not apoptosis in ERdj4GT/GT mice. Thus, we expect to observe an increase in the number of progenitor cells in islets of ERdj4GT/GT mice compared to

controls. These findings will provide a mechanistic understanding of how increased ER

stress causes β-cell failure in ERdj4GT/GT mice.

3c. Does rescue of ERdj4 expression in pancreatic β-cells result in normal insulin

secretion and glucose metabolism in ERdj4GT/GT mice?

Pancreatic β-cells activate the UPR in order to maintain ER homeostasis during

increased insulin biosynthesis. The UPR increases protein folding in the ER through

upregulation of foldases and chaperones24. Our data suggest that the chaperone activity of ERdj4 plays an important role in insulin biosynthesis. ERdj4 associated with insulin, as well as several insulin processing enzymes, including Pcsk1, Pcsk2 and CPE

a The terminal deoxynucleotidyltransferase-mediated dUTP-biotin nick-end labeling (TUNEL) assay detects DNA fragmentation associated with apoptosis.

160 (Chapter 2, Figure 5C). Loss of ERdj4 led to accumulation of proinsulin in the ER,

contributing to increased β-cell stress (Chapter 2, Figures 4 and 5). Reduced insulin

biogenesis resulted in hypoinsulinemia and glucose intolerance in ERdj4GT/GT mice

(Chapter 2, Figure 6). To confirm that ERdj4 deficiency impairs insulin biosynthesis,

proinsulin processing will be assessed by pulse chase analyses using a pancreatic β- cell line transfected with small interfering RNA (siRNA) against ERdj4. We hypothesize that ERdj4 deficiency in β-cells will disrupt proinsulin processing leading to reduced production and secretion of mature insulin.

Since ERdj4 is important for β-cell homeostasis and function, we hypothesize

that rescuing ERdj4 expression in β-cells will enhance insulin secretion and ameliorate

glucose intolerance in ERdj4GT/GT mice. These studies will be performed in vitro by

transfecting ERdj4 into pancreatic islets isolated from ERdj4GT/GT mice33 and analyzing

insulin biosynthesis by pulse chase analyses. We expect that rescue of ERdj4

expression will enhance proinsulin processing and, as a result, increase secretion of

mature insulin. This hypothesis could be further tested in vivo by rescuing ERdj4

expression in pancreatic β-cells of ERdj4GT/GT mice. ERdj4 cDNA would be subcloned

into a plasmid containing the porcine insulin promoter, which directs expression in

mouse pancreatic β-cells34. Expression of the ERdj4 transgene would be confirmed in

vitro by transfecting a pancreatic β-cell line with the recombinant plasmid and assessing

ERdj4 expression by qRT-PCR. The DNA fragment would then be microinjected into

ERdj4+/GT oocytes and fertilized embryos would be transferred into pseudopregnant mice. Founder mice would be assessed for the presence of the ERdj4 transgene by

Southern blot analysis and multiple transgenic lines would be generated to exclude

161 artifacts related to random transgenic integration. ERdj4 mRNA expression would be

analyzed in pancreatic β-cells isolated from transgenic mice by qRT-PCR. We hypothesize that rescuing ERdj4 expression in pancreatic β-cells would reduce ER stress and increase insulin biosynthesis leading to normal glucose tolerance. If

ERdj4GT/GT mice develop diabetes with age and/or HFD (as discussed in section 3a), we

predict that rescuing ERdj4 in pancreatic β-cells would prevent the onset of diabetes.

Alternatively, if rescue of ERdj4 in pancreatic β-cells fails to correct the metabolic

defects in ERdj4GT/GT mice, it is possible that aberrant expression/activity of Pcsk1 indirectly affects insulin secretion. Pcsk1 also processes proglucagon in intestinal L cells to generate glucagon-like peptide (GLP)-135,36, which stimulates glucose- dependent insulin secretion37. Thus, lower levels of GLP-1 in ERdj4GT/GT mice may

cause hypoinsulinemia and glucose intolerance. Overall, these studies would provide

insight into how the absence of ERdj4 causes abnormalities in glucose metabolism.

Since ER stress is associated with obesity, insulin resistance and type 2 diabetes, alleviating ER stress by increasing protein folding capacity may be a novel therapeutic strategy in the treatment of disease. Ozcan et al. tested this hypothesis

using the pharmaceutical chaperones 4-phenyl butyric acid (PBA) and

tauroursodeoxycholic acid (TUDCA) in leptin-deficient mice (ob/ob), a model of obesity

and insulin resistance. PBA and TUDCA are chemical chaperones, approved by the

Federal Drug Administration for use in humans, that function to increase protein folding

38 and degradation in ER . Treatment with either PBA or TUDCA reduced ER stress in peripheral tissues, resulting in normoglycemia with increased insulin sensitivity and resolution of fatty liver disease28. It remains unknown whether PBA or TUDCA are

162 capable of alleviating β-cell stress in other mouse models of type 2 diabetes (e.g. Akita mice). However, since ER stress is tightly associated with disease pathogenesis, restoring ER function by chemical chaperones is a promising therapeutic approach for type 2 diabetes38.

3d. Is the chaperone activity of ERdj4 important in α-cells?

3di. Does loss of ERdj4 affect glucagon biosynthesis?

Glucagon is released from pancreatic α-cells in response to low levels of blood glucose in order to stimulate gluconeogenesis and glycogenolysis. Accumulation of glucagon was associated with increased ER stress in pancreatic α-cells of ERdj4GT/GT mice

(Chapter 2, Figure S2B), suggesting that ERdj4 may be required for glucagon

maturation in the ER. However, co-immunoprecipitation experiments revealed that,

unlike insulin, ERdj4 did not directly associate with glucagon but rather, interacted with

Pcsk2 and CPE, glucagon-processing enzymes (Chapter 2, Figure 5C). Mice deficient

in either Pcsk2 or CPE had defects in proglucagon processing and glucose

metabolism39,40.

Since ERdj4 associated with Pcsk2 and CPE, but not glucagon, we hypothesize

that ERdj4 indirectly affects proglucagon processing in α-cells by influencing Pcsk2 and

CPE maturation. Several experiments are needed to test this hypothesis. First,

glucagon processing and secretion will be assessed by pulse chase analyses using a

pancreatic α-cell lineb transfected with ERdj4 siRNA. We expect that proglucagon will accumulate in α-cells deficient in ERdj4, thus explaining the increase in glucagon

b Islets are not used to assess glucagon processing by pulse chase analysis due to α-cell hyperplasia in ERdj4GT/GT mice.

163 granules observed in pancreatic α-cells of ERdj4GT/GT mice (Chapter 2, Figure S2B).

Second, proglucagon levels will be assessed in the pancreas and serum of ERdj4GT/GT

mice by immunoassay. We hypothesize that proglucagon will be increased in the

pancreas and serum, if expression and/or enzymatic activity of Pcsk2/CPE is altered in

the absence of ERdj4. Third, the maturation of Pcsk2/CPE will be analyzed by pulse

chase analyses in an α-cell line transfected with ERdj4 siRNA. Since ERdj4 interacts

with Pcsk2/CPE, we hypothesize Pcsk2/CPE processing will be reduced in the absence

of ERdj4. These studies will determine if ERdj4 indirectly disrupts glucagon

biosynthesis by affecting the expression and/or maturation of glucagon-processing

enzymes.

ERdj4 also associated with Pcsk1, which processes proglucagon in intestinal L

cells to generate GLP-1 and GLP-235,36 (Chapter 2, Figure 5C). GLP-1 and GLP-2 regulate energy homeostasis by influencing insulin/glucagon secretion and intestinal nutrient absorption, respectively37. The levels of GLP-1 and GLP-2 have not been

assessed in ERdj4GT/GT mice, but it is possible that they are affected by ERdj4 deficiency, thereby contributing to the defects in glucose metabolism (as discussed in section 3c).

3dii. What causes α-cell hyperplasia in ERdj4GT/GT mice?

Defective proglucagon processing is associated with α-cell hyperplasia40, a phenotype

observed in pancreatic islets of ERdj4GT/GT mice (Chapter 2, Figure 4D). Since the

mature form of glucagon negatively inhibits α-cell proliferation, decreased mature

glucagon in ERdj4GT/GT mice may promote an increase in α-cell mass. Thus, assessing

164 proglucagon processing (as described above) will elucidate the mechanisms underlying

α-cell hyperplasia. Insulin also regulates glucagon41, another factor likely contributing to the increased number of α-cells and hyperglucagonemia in hypoinsulinemic ERdj4GT/GT

mice (Chapter 2, Figure 6). Several diabetic mouse models30,31,42, as well as humans

with type 2 diabetes43 report α-cell hyperplasia and hyperglucagonemia as a result of insulin deficiency. Thus, it is possible that dysregulation of glucagon and/or insulin in

ERdj4GT/GT mice leads to α-cell hyperplasia.

4. The role of ERdj4 in the bone marrow microenvironment

4a. Does the loss of ERdj4 in osteogenic cells impair B cell development in ERdj4GT/GT

mice?

4ai. Do ERdj4GT/GT osteogenic cells support B cell maturation in vitro?

Although osteogenic cells are reduced in ERdj4GT/GT mice in association with increased

ER stress (Chapter 3, Figure 1), direct evidence linking ERdj4 deficiency in osteogenic cells to the defect in B cell development is lacking. To address this issue, HSCs will be co-cultured with osteogenic cells isolated from either ERdj4+/+ or ERdj4GT/GT mice. Zhu

et al. demonstrated that osteogenic cells isolated from neonatal calvariae supported B

cell development from HSCs through expression of VCAM-1, CXCL12 and IL-76.

However, their isolation technique resulted in a heterogeneous cell population with the

possibility of fibroblast contamination. Thus, a different approach will be used to obtain

primary osteogenic cells; mesenchymal stem cells (MSCs, Lin-CD45-CD31-Sca1+),

isolated from mouse compact bone by collagenase digestion and fluorescence activated

cell sorting (FACS), will be differentiated into osteogenic cells by treatment with

165 BMP244,45. The ability of ERdj4+/+ and ERdj4GT/GT MSCs to undergo osteogenesis will

be assessed by ALP activity and qRT-PCR of Osterix, osteocalcin and type 1 collagen

mRNAs. Further, B cell adhesion molecules and cytokines, including VCAM-1, CXCL12

and IL-7, will be analyzed by flow cytometry and/or immunoassay. Wild-type HSCs will

then be cultured on osteogenic cells for 7 days before assessing B cell development by

flow cytometry6. We expect that ERdj4 deficiency will impair osteogenesis by affecting

maturation and/or survival of osteogenic cells, thereby affecting B cell development. It

is possible that MSCs from ERdj4GT/GT mice could differentiate into osteogenic cells in vitro, resulting in normal B cell development. These findings would suggest that osteogenic cells from ERdj4GT/GT mice are capable of supporting B cell development, but

ERdj4 deficiency may affect growth factors necessary for osteogenesis in vivo.

4aii. Does administration of PTH rescue the defect in B cell development in ERdj4GT/GT

mice?

PTH promotes osteoblast differentiation, function and survival6,46,47. Importantly, PTH

induces the expression of several B cell growth factors, including CXCL12 and IL-7,

thereby enhancing B cell development in vitro6. Defects in the PTH signaling pathway in osteogenic cells reduced B cell development in the bone marrow beginning at the pro-B cell stage, similar to ERdj4GT/GT mice. We expect that exogenous PTH will

stimulate osteogenesis in ERdj4GT/GT mice, thereby rescuing the defect in B cell

development. A potential pitfall to this experiment is the expression of PTH receptor on

B cells; PTH receptor signaling inhibited B cell proliferation by altering intracellular

calcium metabolism48-50. Thus, it is possible that inhibition of B cell proliferation by PTH

166 will prevent rescue of B cell development via the osteogenic pathway. To address this issue, B cell development will be assessed in the bone marrow of wildtype mice upon treatment with PTH. If PTH disrupts B cell development under normal conditions, alternative approaches would be pursued to identify whether impaired osteogenesis reduces B cell development in ERdj4GT/GT mice (as discussed below).

4aiii. Would rescue of ERdj4 expression in osteogenic cells result in normal B cell development in ERdj4GT/GT mice?

An alternative approach to PTH administration would be to rescue ERdj4 expression in

osteogenic cells of ERdj4GT/GT mice. Mouse ERdj4 cDNA would be subcloned into a

vector containing the rat collagen α1 type I promoter, which directs expression in

osteoblasts, as well as bone-lining cells and osteocytes51,52. The plasmid would be

transfected into an osteoblast cell line to verify expression of the ERdj4 transgene by

qRT-PCR. The DNA fragment would be microinjected into oocytes from ERdj4+/GT mice

and fertilized embryos would be transferred into pseudopregnant mice. Founder mice

would be tested for the presence of the transgene by Southern blot analyses and

multiple transgenic lines would be generated to exclude artifacts related to random

transgenic integration. ERdj4 mRNA expression would be analyzed in osteogenic cells

isolated from transgenic mice by qRT-PCR. Osteogenic cells and B cell development

would then be assessed in transgenic mice by immunohistochemistry and flow

cytometry, respectively. We expect that rescuing ERdj4 expression in osteogenic cells

would increase the number of postosteoblasts resulting in normal B cell development in

167 ERdj4GT/GT mice. These studies would confirm that the loss of ERdj4 in osteogenic cells

causes the defect in B cell development in ERdj4GT/GT mice.

4b. Which cell(s) of the osteogenic lineage affects B cell development?

Osteoblasts are reported to influence hematopoiesis5,53,54. However, this conclusion is based on approaches that affect cells of the osteogenic lineage, not osteoblasts per se.

Conditional depletion of osteoblasts in vivo by expressing herpesvirus thymidine kinase

under control of the rat collagen α1 type I promoter also depleted postosteoblastic cells,

including bone-lining cells and osteocytes55. Ablation of these osteogenic cell types

resulted in a reduction of HSCs, B cells and erythrocytes in the bone marrow5. Further,

Zhu et al. demonstrated that osteoblasts isolated from neonatal calvariae supported B

cell development through VCAM-1, CXCL12 and IL-76. Although these calvarial cells expressed osteogenic-specific markers, the cultures contained multiple cell types within the osteogenic lineage, including MSCs, osteoblasts and osteocytes.

Osteoblasts are active at low frequencies during adulthood; in humans, only 0.1-

7.3% of endosteal surfaces are forming new bone and after bone formation, osteoblasts either undergo cell death or become quiescent bone-lining cells or osteocytes. Thus, if osteoblasts are the only cell in the osteogenic lineage to support hematopoiesis, the hematopoietic niche would be transient, relocating to new areas of bone formation, an unlikely possibility. It is more reasonable to postulate that other cells of the osteogenic lineage, including MSCs, bone-lining cells and osteocytes, influence hematopoiesis56.

MSCs are scattered throughout the bone marrow and a play a pivotal role in

hematopoiesis by stimulating adipogenesis and osteogenesis, as well as the

168 development of bone marrow sinusoids56. Bone-lining cells and osteocytes are the most abundant osteogenic cell types, covering approximately 90% of all bone surfaces57. Osteocytes also express high levels of RANKL, which influences B cell

development58,59. Bone-lining cells were reduced in association with impaired B

lymphopoiesis in ERdj4GT/GT mice (Chapter 3, Figures 1 and 3), although it is possible that these phenotypes are unrelated (addressed experimentally in section 4a).

Unfortunately, the role of MSCs and postosteoblasts in B cell development remains unknown, challenged by the lack of markers defining specific stages of osteogenesis and the plasticity of phenotypes within the osteogenic lineage56.

4c. How does ERdj4 deficiency affect peripheral B cells?

Numbers of follicular and B1b cells were reduced in the spleen and peritoneal cavity of

ERdj4GT/GT mice, respectively. However, not all mature B cell compartments were

compromised; marginal zone and B1a B cells, which function similarly but differ in

anatomical location, were preserved. Similarly, transitional B cells, representing the

most recent bone marrow emigrants, were also unaffected. Bone marrow chimeras

rescued the defect in mature B cell development, supporting a microenvironmental

defect in ERdj4GT/GT mice (Chapter 3, Figure 5).

Follicular B cells continuously circulate throughout the body, visiting the spleen,

lymph nodes, Peyer’s Patches and bone marrow60,61. In the bone marrow, mature B cells reside in the sinusoidal niche, receiving survival signals from dendritic cells through MIF62. Interestingly, the bone marrow sinusoidal niche is formed by osteoclasts, which are regulated by osteocytes56. Thus, it is possible that osteocyte

169 deficiency in ERdj4GT/GT mice affects the sinusoidal niche, thereby impairing the survival of mature recirculating B cells. Experiments addressing whether the loss of ERdj4 in osteogenic cells caused the defect in B cell development are addressed above (section

4a).

Mechanisms for the identified defects in mature peripheral B cells include decreased production, survival and/or proliferation. These possibilities will be explored by using BrdU incorporation to assess cell turnover, as well as cell fate63. BrdU will be

administered to mice through their drinking water for 14 days. Mice will be sacrificed on

days 7 and 14 of the pulse or days 42 or 84 of the chase and mature B cell populations

will be analyzed in the spleen and peritoneal cavity by flow cytometry64. Given that a microenvironmental defect in the bone marrow increased pro-B cell death in ERdj4GT/GT

mice, we expect that BrdU+ mature B cells will decrease more quickly overtime,

indicating a shorter lifespan.

4d. How does the loss of ERdj4 affect postosteoblasts and skeletal development?

4di. Quantitation of osteogenic cells from ERdj4GT/GT mice

Although histological analyses of femur tissue sections revealed a loss of bone-lining

cells in ERdj4GT/GT mice, these cells have yet to be quantitated. To address this issue,

osteoblasts, bone-lining cells and osteocytes will be quantitated using

histomorphometric analyses. Bone sections will be stained with toluidine blue and the

OsteoMeasure system (Osteometrics, Inc.) will be used to count the number of

osteogenic cells per tissue volume65. Anatomical location will distinguish between

osteoblasts, bone-lining cells and osteocytes.

170 4dii. Why are postosteoblasts reduced in ERdj4GT/GT mice?

After bone formation, a majority of osteoblasts undergo apoptosis, while some

differentiate into bone-lining cells and osteocytes57. Bone-lining cells and osteocytes

were reduced in femur tissue sections and primary osteogenic cultures from ERdj4GT/GT

mice, respectively (Chapter 3, Figures 1 and 2). The increased ER stress observed in osteogenic cells in vivo may enhance osteoblast apoptosis, thereby reducing the pool of

cells that become bone-lining cells or osteocytes. Alternatively, postosteoblasts may

have an increased susceptibility to apoptosis. This question will be addressed by using

the TUNEL assay to stain for DNA fragmentation, a marker of apoptosis, on femur

tissue sections from ERdj4GT/GT mice. Osteoblasts, bone-lining cells and osteocytes will be identified by anatomical location. If ERdj4 deficiency does not increase osteoblast

and/or postosteoblast apoptosis, it is possible that osteoblasts fail to differentiate into

bone-lining cells and osteocytes. A block in development would likely result in

accumulation of MSCs and osteoblasts in ERdj4GT/GT mice, which could be assessed by

colony forming assays in vitro.

4diii. Does the loss of postosteoblasts affect skeletal development in ERdj4GT/GT mice?

Postosteoblasts regulate bone remodeling by stimulating osteoclastogenesis through

expression of RANKL. Deficiency of RANKL on osteocytes did not cause any gross

skeletal defects, but resulted in an osteopetrotic phenotype due to reduced

osteoclastogenesis59. Osteogenic cells isolated from ERdj4GT/GT mice expressed significantly lower levels of RANKL (Chapter 3, Figure 2C), suggesting that either osteocytes were reduced within compact bone or expressed lower levels of RANKL.

171 Thus, we hypothesize that reduced osteocytes and/or RANKL expression in ERdj4GT/GT

mice influences osteoclast differentiation and function. To test this possibility,

osteoclasts will be evaluated in femur tissue sections from ERdj4+/+ and ERdj4GT/GT mice

by staining for tartrate-resistant acid phosphatase (TRAP), an osteoclast marker.

Further, microcomputed tomography (µCT) scanning will be performed to assess bone

volume, trabecular thickness/separation and bone mineral content. We expect that a

decrease in osteocytes and/or RANKL in ERdj4GT/GT mice will reduce osteoclastogenesis, leading to a slower rate of bone resorption.

5. Molecular chaperones in the treatment of ER stress-related disease

Several diseases are associated with ER stress, including neurodegenerative diseases, atherosclerosis, interstitial lung disease and diabetes. ER stress results from perturbations that alter redox balance, calcium levels or posttranslational modifications, leading to accumulation of unfolded/misfolded proteins within the ER lumen. The UPR attempts to restore ER homeostasis by attenuating protein translation and upregulating protein folding/degradation machinery. However, a failure to alleviate ER stress results in activation of cell death pathways, thereby contributing to disease pathology.

Molecular chaperones perform a wide variety of functions in the ER including

maintenance of ER calcium stores66, regulation of the translocon pore67, protein folding and degradation68-71, and activation of the UPR72,73. The importance of molecular chaperone activity in cellular homeostasis and function was demonstrated in the current study; ERdj4 deficiency impaired survival, glucose metabolism, osteogenesis and B cell development in vivo.

172 The ability of molecular chaperones to maintain/restore ER homeostasis has

promising therapeutic potential in the treatment of diseases associated with ER stress.

Upregulation of molecular chaperones reduced neurotoxicity in protein folding diseases

such as Alzheimer’s, Huntington’s and Parkinson disease74,75 and therapeutics that induce expression of molecular chaperones are currently being evaluated in clinical trials75-77. This study, clarifying the role of ERdj4 in cellular homeostasis and function,

provides rationale for therapeutic targeting of molecular chaperones in ER stress-

related disease.

6. ERdj4 in cellular function and homeostasis

ERdj4 maintains ER homeostasis by assisting in the removal of unfolded/misfolded

substrates from the ER lumen for proteasomal degradation. ERdj4 deficiency increased

ER stress in a variety of non-secretory and secretory cell types (Chapter 2, Figure S1).

Collectively, these data indicate that the chaperone activity of ERdj4 is important for

cellular homeostasis, but it also suggests that ERdj4 associates with a broad range of

substrates. Our laboratory has identified the only known substrates for ERdj4, which

include unfolded (insulin2WT, Pcsk1, Pcsk2 and CPE) and misfolded (insulin2C96Y and

SP-CΔexon4) proteins that are either non-glycosylated (insulin2 and SP-C) or glycosylated

(Pcsk1, Pcsk2 and CPE) (Chapter 1, Figure 5C)1. Pcsk1, Pcsk2 and CPE play an

important role in glucose metabolism by influencing insulin and glucagon biosynthesis,

but also process many other hormones and neuropeptides in the neuroendocrine

system. Deficiency of Pcsk1, Pcsk2 or CPE in mice resulted in a wide range of neural

and endocrine abnormalities35,36,78. It is possible that ERdj4 deficiency affects the

173 maturation of Pcsk1, Pcsk2 and/or CPE, causing multiple systemic defects in

ERdj4GT/GT mice. Further studies are needed to examine the maturation of these enzymes in the absence of ERdj4 using pulse chase analyses. If Pcsk1, Pcsk2 and/or

CPE fail to achieve native conformation due to the loss of ERdj4, the hormones and neuropeptides processed by these enzymes will be assessed in ERdj4GT/GT mice.

Although several lines of evidence indicate that ERdj4 is a component of ERAD,

it is possible that ERdj4 also promotes the folding and maturation of nascent proteins.

Other ERdjs are known to facilitate protein folding by directly associating with unfolded

substrates and/or the translocon pore, where nascent proteins enter the ER lumen79.

Examining the maturation of Pcsk1, Pcsk2 and CPE will provide evidence as to whether

ERdj4 assists in protein folding, as well as degradation. This work highlights the

importance of ERdj4 chaperone activity in maintaining ER homeostasis under normal

conditions.

Collectively, a great deal has been learned about ERdj4 biology throughout the

course of these studies, revealing novel insights into the role of ERdj4 in survival,

glucose metabolism and B cell development. Future studies hope to address the questions raised in this discussion, as well as explore the other interesting phenotypes resulting from ERdj4 deficiency.

174 References

1. Dong, M., Bridges, J. P., Apsley, K., Xu, Y. & Weaver, T. E. ERdj4 and ERdj5 are required for endoplasmic reticulum-associated protein degradation of misfolded surfactant protein C. Mol Biol Cell 19, 2620-2630 (2008).

2. Reimold, A. M. et al. Plasma cell differentiation requires the transcription factor XBP-1. Nature 412, 300-307 (2001).

3. Zhang, K. et al. The unfolded protein response sensor IRE1alpha is required at 2 distinct steps in B cell lymphopoiesis. J Clin Invest 115, 268-281 (2005).

4. Lee, A. H., Iwakoshi, N. N. & Glimcher, L. H. XBP-1 regulates a subset of endoplasmic reticulum resident chaperone genes in the unfolded protein response. Mol Cell Biol 23, 7448-7459 (2003).

5. Visnjic, D. et al. Hematopoiesis is severely altered in mice with an induced osteoblast deficiency. Blood 103, 3258-3264 (2004).

6. Zhu, J. et al. Osteoblasts support B-lymphocyte commitment and differentiation from hematopoietic stem cells. Blood 109, 3706-3712 (2007).

7. Lai, C. W., Otero, J. H., Hendershot, L. M. & Snapp, E. ERdj4 protein is a soluble endoplasmic reticulum (ER) DnaJ family protein that interacts with ER-associated degradation machinery. J Biol Chem 287, 7969-7978 (2012).

8. Shen, Y., Meunier, L. & Hendershot, L. M. Identification and characterization of a novel endoplasmic reticulum (ER) DnaJ homologue, which stimulates ATPase activity of BiP in vitro and is induced by ER stress. J Biol Chem 277, 15947-15956 (2002).

9. Friedrich, G. & Soriano, P. Promoter traps in embryonic stem cells: a genetic screen to identify and mutate developmental genes in mice. Genes Dev 5, 1513- 1523 (1991).

10. Skarnes, W. C., Auerbach, B. A. & Joyner, A. L. A gene trap approach in mouse embryonic stem cells: the lacZ reported is activated by splicing, reflects endogenous gene expression, and is mutagenic in mice. Genes Dev 6, 903-918 (1992).

11. Desvergne, B., Michalik, L. & Wahli, W. Transcriptional regulation of metabolism. Physiol Rev 86, 465-514 (2006).

12. Turgeon, B. & Meloche, S. Interpreting neonatal lethal phenotypes in mouse mutants: insights into gene function and human diseases. Physiol Rev 89, 1-26 (2009).

175

13. Scheuner, D. et al. Translational control is required for the unfolded protein response and in vivo glucose homeostasis. Mol Cell 7, 1165-1176 (2001). 14. Watson, E. D. & Cross, J. C. Development of structures and transport functions in the mouse placenta. Physiology (Bethesda) 20, 180-193 (2005).

15. Iwawaki, T., Akai, R., Yamanaka, S. & Kohno, K. Function of IRE1 alpha in the placenta is essential for placental development and embryonic viability. Proc Natl Acad Sci U S A 106, 16657-16662 (2009).

16. Forsen, T. et al. The fetal and childhood growth of persons who develop type 2 diabetes. Ann Intern Med 133, 176-182 (2000).

17. Gluckman, P. D. & Hanson, M. A. Living with the past: evolution, development, and patterns of disease. Science 305, 1733-1736 (2004).

18. Scheuner, D. et al. Translational control is required for the unfolded protein response and in vivo glucose homeostasis. Mol Cell 7, 1165-1176 (2001).

19. Kahn, S. E., Hull, R. L. & Utzschneider, K. M. Mechanisms linking obesity to insulin resistance and type 2 diabetes. Nature 444, 840-846 (2006).

20. Shaw, J. E., Sicree, R. A. & Zimmet, P. Z. Global estimates of the prevalence of diabetes for 2010 and 2030. Diabetes Res Clin Pract 87, 4-14 (2010).

21. Tunstall-Pedoe, H. et al. Contribution of trends in survival and coronary-event rates to changes in coronary heart disease mortality: 10-year results from 37 WHO MONICA project populations. Monitoring trends and determinants in cardiovascular disease. Lancet 353, 1547-1557 (1999).

22. Roglic, G. & Unwin, N. Mortality attributable to diabetes: estimates for the year 2010. Diabetes Res Clin Pract 87, 15-19 (2010).

23. Back, S. H., Kang, S. W., Han, J. & Chung, H. T. Endoplasmic reticulum stress in the beta-cell pathogenesis of type 2 diabetes. Exp Diabetes Res 2012, 618396 (2012).

24. Back, S. H. & Kaufman, R. J. Endoplasmic reticulum stress and type 2 diabetes. Annu Rev Biochem 81, 767-793 (2012).

25. Scheuner, D. & Kaufman, R. J. The unfolded protein response: a pathway that links insulin demand with beta-cell failure and diabetes. Endocr Rev 29, 317-333 (2008).

26. Rieck, S. et al. The transcriptional response of the islet to pregnancy in mice. Mol Endocrinol 23, 1702-1712 (2009).

176

27. Rankin, M. M. & Kushner, J. A. Aging induces a distinct gene expression program in mouse islets. Islets 2, 345-352 (2010).

28. Ozcan, U. et al. Endoplasmic reticulum stress links obesity, insulin action, and type 2 diabetes. Science 306, 457-461 (2004).

29. Yoshioka, M., Kayo, T., Ikeda, T. & Koizumi, A. A novel locus, Mody4, distal to D7Mit189 on chromosome 7 determines early-onset NIDDM in nonobese C57BL/6 (Akita) mutant mice. Diabetes 46, 887-894 (1997).

30. Ladiges, W. C. et al. Pancreatic beta-cell failure and diabetes in mice with a deletion mutation of the endoplasmic reticulum molecular chaperone gene P58IPK. Diabetes 54, 1074-1081 (2005).

31. Zhang, P. et al. The PERK eukaryotic initiation factor 2 alpha kinase is required for the development of the skeletal system, postnatal growth, and the function and viability of the pancreas. Mol Cell Biol 22, 3864-3874 (2002).

32. Talchai, C., Xuan, S., Lin, H. V., Sussel, L. & Accili, D. Pancreatic beta Cell Dedifferentiation as a Mechanism of Diabetic beta Cell Failure. Cell 150, 1223- 1234 (2012).

33. Lakey, J. R. et al. Nonviral transfection of intact pancreatic islets. Cell Transplant 10, 697-708 (2001).

34. Grzech, M. et al. Specific transgene expression in mouse pancreatic beta-cells under the control of the porcine insulin promoter. Mol Cell Endocrinol 315, 219-224 (2010).

35. Zhu, X. et al. Severe block in processing of proinsulin to insulin accompanied by elevation of des-64,65 proinsulin intermediates in islets of mice lacking prohormone convertase 1/3. Proc Natl Acad Sci U S A 99, 10299-10304 (2002).

36. Zhu, X. et al. Disruption of PC1/3 expression in mice causes dwarfism and multiple neuroendocrine peptide processing defects. Proc Natl Acad Sci U S A 99, 10293- 10298 (2002).

37. Drucker, D. J. Biologic actions and therapeutic potential of the proglucagon-derived peptides. Nat Clin Pract Endocrinol Metab 1, 22-31 (2005).

38. Engin, F. & Hotamisligil, G. S. Restoring endoplasmic reticulum function by chemical chaperones: an emerging therapeutic approach for metabolic diseases. Diabetes Obes Metab 12 Suppl 2, 108-115 (2010).

177 39. Cawley, N. X. et al. The carboxypeptidase E knockout mouse exhibits endocrinological and behavioral deficits. Endocrinology 145, 5807-5819 (2004).

40. Furuta, M. et al. Defective prohormone processing and altered pancreatic islet morphology in mice lacking active SPC2. Proc Natl Acad Sci U S A 94, 6646-6651 (1997). 41. Bansal, P. & Wang, Q. Insulin as a physiological modulator of glucagon secretion. Am J Physiol Endocrinol Metab 295, E751-61 (2008).

42. Kharouta, M. et al. No mantle formation in rodent islets -- the prototype of islet revisited. Diabetes Res Clin Pract 85, 252-257 (2009).

43. Clark, A. et al. Islet amyloid, increased A-cells, reduced B-cells and exocrine fibrosis: quantitative changes in the pancreas in type 2 diabetes. Diabetes Res 9, 151-159 (1988).

44. Short, B. J., Brouard, N. & Simmons, P. J. Prospective isolation of mesenchymal stem cells from mouse compact bone. Methods Mol Biol 482, 259-268 (2009).

45. Zhao, L. et al. Tumor necrosis factor inhibits mesenchymal stem cell differentiation into osteoblasts via the E3 ligase Wwp1. Stem Cells 29, 1601-1610 (2011).

46. Jilka, R. L. et al. Increased bone formation by prevention of osteoblast apoptosis with parathyroid hormone. J Clin Invest 104, 439-446 (1999).

47. O'Brien, C. A. et al. Control of bone mass and remodeling by PTH receptor signaling in osteocytes. PLoS One 3, e2942 (2008).

48. Alexiewicz, J. M. et al. Parathyroid hormone inhibits B cell proliferation: implications in chronic renal failure. J Am Soc Nephrol 1, 236-244 (1990).

49. Alexiewicz, J. M., Smogorzewski, M., Klin, M., Akmal, M. & Massry, S. G. Effect of treatment of hemodialysis patients with nifedipine on metabolism and function of polymorphonuclear leukocytes. Am J Kidney Dis 25, 440-444 (1995).

50. Alexiewicz, J. M., Smogorzewski, M., Gill, S. K., Akmal, M. & Massry, S. G. Time course of the effect of nifedipine therapy and its discontinuation on [Ca2+]i and phagocytosis of polymorphonuclear leukocytes from hemodialysis patients. Am J Nephrol 17, 12-16 (1997).

51. Kalajzic, I. et al. Use of type I collagen green fluorescent protein transgenes to identify subpopulations of cells at different stages of the osteoblast lineage. J Bone Miner Res 17, 15-25 (2002).

178 52. Zha, L. et al. Collagen1alpha1 promoter drives the expression of Cre recombinase in osteoblasts of transgenic mice. J Genet Genomics 35, 525-530 (2008).

53. Calvi, L. M. et al. Osteoblastic cells regulate the haematopoietic stem cell niche. Nature 425, 841-846 (2003).

54. Zhang, J. et al. Identification of the haematopoietic stem cell niche and control of the niche size. Nature 425, 836-841 (2003).

55. Visnjic, D. et al. Conditional ablation of the osteoblast lineage in Col2.3deltatk transgenic mice. J Bone Miner Res 16, 2222-2231 (2001).

56. Bianco, P. Bone and the hematopoietic niche: a tale of two stem cells. Blood 117, 5281-5288 (2011).

57. Franz-Odendaal, T. A., Hall, B. K. & Witten, P. E. Buried alive: how osteoblasts become osteocytes. Dev Dyn 235, 176-190 (2006).

58. Kong, Y. Y. et al. OPGL is a key regulator of osteoclastogenesis, lymphocyte development and lymph-node organogenesis. Nature 397, 315-323 (1999).

59. Nakashima, T. et al. Evidence for osteocyte regulation of bone homeostasis through RANKL expression. Nat Med 17, 1231-1234 (2011).

60. Cariappa, A., Chase, C., Liu, H., Russell, P. & Pillai, S. Naive recirculating B cells mature simultaneously in the spleen and bone marrow. Blood 109, 2339-2345 (2007).

61. Cariappa, A. et al. Perisinusoidal B cells in the bone marrow participate in T- independent responses to blood-borne microbes. Immunity 23, 397-407 (2005).

62. Sapoznikov, A. et al. Perivascular clusters of dendritic cells provide critical survival signals to B cells in bone marrow niches. Nat Immunol 9, 388-395 (2008).

63. Tough, D. F., Sprent, J. & Stephens, G. L. Measurement of T and B cell turnover with bromodeoxyuridine. Curr Protoc Immunol Chapter 4, Unit 4.7 (2007).

64. Sasaki, Y. et al. Canonical NF-kappaB activity, dispensable for B cell development, replaces BAFF-receptor signals and promotes B cell proliferation upon activation. Immunity 24, 729-739 (2006).

65. Liu, Y. et al. Intracellular VEGF regulates the balance between osteoblast and adipocyte differentiation. J Clin Invest 122, 3101-3113 (2012).

66. Lievremont, J. P., Rizzuto, R., Hendershot, L. & Meldolesi, J. BiP, a major chaperone protein of the endoplasmic reticulum lumen, plays a direct and

179 important role in the storage of the rapidly exchanging pool of Ca2+. J Biol Chem 272, 30873-30879 (1997).

67. Hamman, B. D., Hendershot, L. M. & Johnson, A. E. BiP maintains the permeability barrier of the ER membrane by sealing the lumenal end of the translocon pore before and early in translocation. Cell 92, 747-758 (1998).

68. Brodsky, J. L. et al. The requirement for molecular chaperones during endoplasmic reticulum-associated protein degradation demonstrates that protein export and import are mechanistically distinct. J Biol Chem 274, 3453-3460 (1999). 69. Hendershot, L. et al. Inhibition of immunoglobulin folding and secretion by dominant negative BiP ATPase mutants. Proc Natl Acad Sci U S A 93, 5269-5274 (1996).

70. Plemper, R. K., Bohmler, S., Bordallo, J., Sommer, T. & Wolf, D. H. Mutant analysis links the translocon and BiP to retrograde protein transport for ER degradation. Nature 388, 891-895 (1997).

71. Simons, J. F., Ferro-Novick, S., Rose, M. D. & Helenius, A. BiP/Kar2p serves as a molecular chaperone during carboxypeptidase Y folding in yeast. J Cell Biol 130, 41-49 (1995).

72. Bertolotti, A., Zhang, Y., Hendershot, L. M., Harding, H. P. & Ron, D. Dynamic interaction of BiP and ER stress transducers in the unfolded-protein response. Nat Cell Biol 2, 326-332 (2000).

73. Shen, J., Chen, X., Hendershot, L. & Prywes, R. ER stress regulation of ATF6 localization by dissociation of BiP/GRP78 binding and unmasking of Golgi localization signals. Dev Cell 3, 99-111 (2002).

74. Barral, J. M., Broadley, S. A., Schaffar, G. & Hartl, F. U. Roles of molecular chaperones in protein misfolding diseases. Semin Cell Dev Biol 15, 17-29 (2004).

75. Broadley, S. A. & Hartl, F. U. The role of molecular chaperones in human misfolding diseases. FEBS Lett 583, 2647-2653 (2009).

76. Lavu, S., Boss, O., Elliott, P. J. & Lambert, P. D. Sirtuins--novel therapeutic targets to treat age-associated diseases. Nat Rev Drug Discov 7, 841-853 (2008).

77. Westerheide, S. D., Anckar, J., Stevens, S. M. J., Sistonen, L. & Morimoto, R. I. Stress-inducible regulation of heat shock factor 1 by the deacetylase SIRT1. Science 323, 1063-1066 (2009).

78. Wang, J. et al. The prohormone convertase enzyme 2 (PC2) is essential for processing pro-islet amyloid polypeptide at the NH2-terminal cleavage site. Diabetes 50, 534-539 (2001).

180

79. Otero, J. H., Lizak, B. & Hendershot, L. M. Life and death of a BiP substrate. Semin Cell Dev Biol 21, 472-478 (2010).

181