Skip to main content
Physiological Reviews logoLink to Physiological Reviews
. 2018 May 2;98(3):1143–1167. doi: 10.1152/physrev.00034.2016

(Re)generating Human Beta Cells: Status, Pitfalls, and Perspectives

Luc Baeyens 1, Marie Lemper 1, Willem Staels 1, Sofie De Groef 1, Nico De Leu 1, Yves Heremans 1, Michael S German 1, Harry Heimberg 1
PMCID: PMC6088144  PMID: 29717931

Abstract

Diabetes mellitus results from disturbed glucose homeostasis due to an absolute (type 1) or relative (type 2) deficiency of insulin, a peptide hormone almost exclusively produced by the beta cells of the endocrine pancreas in a tightly regulated manner. Current therapy only delays disease progression through insulin injection and/or oral medications that increase insulin secretion or sensitivity, decrease hepatic glucose production, or promote glucosuria. These drugs have turned diabetes into a chronic disease as they do not solve the underlying beta cell defects or entirely prevent the long-term complications of hyperglycemia. Beta cell replacement through islet transplantation is a more physiological therapeutic alternative but is severely hampered by donor shortage and immune rejection. A curative strategy should combine newer approaches to immunomodulation with beta cell replacement. Success of this approach depends on the development of practical methods for generating beta cells, either in vitro or in situ through beta cell replication or beta cell differentiation. This review provides an overview of human beta cell generation.

I. KEY POINTS

  • • 

    Current knowledge on human endocrine pancreas development, gained from scarce histopathology samples, indicates some analogy with rodents.

  • • 

    Human beta cells are long-lived with limited expansion potential. Nevertheless, the adult human beta cell mass retains plasticity to adapt to changing metabolic needs.

  • • 

    High-content screening has identified several novel adult beta cell mitogens. However, a lack of cell specificity warrants caution for clinical translation.

  • • 

    Breakthroughs in directed differentiation of human embryonic stem/induced pluripotent stem cells create hope for cell therapy in diabetes. Overcoming the risk for teratoma formation and immune rejection will determine its true clinical potential.

  • • 

    Multiple approaches, including transdifferentiation of adult human non-beta cells, harbor potential for clinical translation as an alternative to stem cell-based therapy. A particularly attractive strategy would be in vivo reprogramming of human acinar cells.

II. INTRODUCTION

Diabetes mellitus is a chronic metabolic disorder characterized by elevated blood glucose levels and increased risk of micro- and macrovascular complications. Currently 415 million people have diabetes globally, and that number is expected to rise to 642 million by 2040 (116a). Several subtypes of diabetes can be clinically distinguished, including polygenic/multifactorial, monogenic, and secondary forms (165). The two most common forms of diabetes, type 1 (T1DM) and type 2 (T2DM) diabetes, are polygenic and multifactorial. A large number of rarer monogenic and secondary varieties together explain roughly 10% of diabetes (79, 246). Gestational diabetes [GDM, prevalence of around 10% of pregnancies (19, 138)], defined as maternal diabetes commencing during pregnancy, represents a transient form of polygenic and multifactorial diabetes caused by situational, relative insulin deficiency.

T1DM is characterized by an absolute shortage of circulating insulin due to progressive autoimmune destruction of the insulin-producing pancreatic beta cells (3). In contrast, T2DM results from a relative shortage of circulating insulin due to 1) failure of the beta cell mass to adapt to rising metabolic demands, most often caused by obesity-related peripheral insulin resistance; and 2) increased beta cell death and dysfunction (41, 109, 183, 278). Reduced insulin signaling (whether from insulin resistance or beta cell failure), results in decreased tissue uptake of glucose and increased glucose release by the liver and kidney. The resulting chronic hyperglycemia in turn damages blood vessels and nerves, culminating in multiorgan damage (83, 108).

Current pharmacological therapy for diabetes consists of exogenous insulin injections and/or oral glucose-lowering medications. None of these therapies directly addresses the underlying beta cell deficit in most patients with diabetes. Beta cell replacement therapies could effectively restore and protect the functional beta cell mass. Transplantation of cadaveric human donor islets represents one approach to beta cell replacement.

The landmark Edmonton protocol overcame prior poor success rates by using improved islet isolation techniques, fresh islet preparations, and a steroid-free immunosuppressive regimen to significantly improve transplantation outcomes (220, 233, 234). This study pioneered a series of reproducibility trials and improved protocols worldwide, leading to insulin-independence durations of at least 5 yr in 50% of engrafted patients (20). Nevertheless, shortage of donor islets severely limits more widespread clinical use and has lowered the goal from achieving insulin independence to reducing glycemic variability and elimination of hypoglycemia unawareness (107).

Therefore, the need for alternative supply of human islet cells is obvious. Insulin-producing cells can be generated by replication of preexisting beta cells and/or by (trans)differentiation of non-beta cells (38, 125). Since rodent models remain the most prevalent and accessible tool for preclinical research, insights obtained from rodent pancreas development have guided the design of human beta cell generation protocols. Although some analogy exists between human and rodent pancreas development and function, notable interspecies differences remain. Understanding the pathways that control human beta cell generation will be critical to devising a successful beta cell therapy for diabetes.

III. HUMAN PANCREAS DEVELOPMENT AND POSTNATAL HOMEOSTASIS

A. Human Pancreas Development

1. Specification and early differentiation of multipotent pancreatic progenitor cells

Similar to rodent, the human embryonic pancreas develops from the primitive gut tube early in development. The timeline of human development is defined according to the Carnegie system (Carnegie Stage, CS), based on age estimates until 60 days post conception (dpc) when recognizable human features become apparent and nomenclature is switched from embryo to fetus (185). The human embryonic pancreas evolves as one dorsal and two ventral buds from the primitive foregut at 30–33 dpc (CS12–13) (118, 119, 198). After regression of the left ventral bud, the right ventral bud fuses with the dorsal bud around 58 dpc (CS22–23) (169, 198, 200) (FIGURE 1).

FIGURE 1.

FIGURE 1.

Schematic representation of human pancreas development. Top panel: human pancreas development is shown in relation to the Carnegie classification system (CS; numbers 9 to 23). Embryos are drawn proportional to the UNSW Human Embryo Resource. Middle panel: pancreas morphogenesis is not depicted proportionally but is intended to frame the key developmental steps (bottom panel) in the development of the organ. The human embryonic pancreas evolves as one dorsal and two ventral buds from the primitive foregut (yellow tube) at CS12–13. Prior to budding, the dorsal prepancreatic endoderm is in contact with the notochord (red line). After regression of the left ventral bud, the right ventral bud fuses with the dorsal bud around CS22–23. Bottom panel: key developmental steps include branching morphogenesis (green, tip cells; orange, trunk cells) (A), the first appearance of NEUROG3+ cells (pink) (B), formation of the initial hormone+ cells (green) (C), and establishment of lineage-committed monohormonal endocrine cells (green, red, brown, purple) and formation of the islet cytoarchitecture (D).

Due to limited access to relevant early developmental human pancreatic tissue, insights into the earliest developmental stages of gut tube regionalization and pancreas specification rely almost solely on model organisms. Prior to budding (25–27 dpc, CS10) and similar to pancreas development in chick (104, 136) and mouse (135), the notochord directly contacts the dorsal prepancreatic endoderm, leading to regional repression of sonic hedgehog (SHH) (118). This notochord-endoderm interaction is interrupted upon fusion of the paired dorsal aortas around 29–31 dpc (CS11) (FIGURE 1). In contrast to mouse where the developmental transcription factor pancreatic and duodenal homeobox 1 (PDX1) can be detected at the earliest stage of pancreas development (E8.0) when endoderm is still in contact with the notochord (85, 124, 186, 251), PDX1 does not appear in human pancreas until 29–31 dpc (CS12) when the notochord-endoderm interaction is disrupted (118, 240).

At 30–33 dpc (CS13), the stratified epithelium of the developing pancreatic expresses the same early pancreatic transcription factors–PDX1, pancreas specific transcription factor 1A (PTF1A), forkhead box A2 (FOXA2), GATA binding protein 4 (GATA4), Nirenberg and Kim homeobox factor 6.1 (NKX6.1), and sex determining region Y-box 9 (SOX9)–that define the multipotent pancreatic progenitor cells (MPPCs) that give rise to all pancreatic cell lineages in the developing murine pancreas (FIGURE 2) (57, 118, 160, 198). In analogy to murine development, mesenchyme-derived fibroblast growth factor (FGF)7 and FGF10 amplify the MPPC pool by stimulating proliferation of PDX1+ cytokeratin+ MPPCs, resulting in substantial organ growth between 33 and 45 dpc (CS14–18) (FIGURE 1) (25, 27, 282). Expression of genes involved in WNT signaling including numerous noncanonical WNT regulators (45) increases in MPPCs during 37–45 dpc (CS16–18), before significant acinar and endocrine cell differentiation. From 37 to 40 dpc (CS16) onwards, active growth and branching results in a lobular epithelium (118, 200). Starting around 45–47 dpc (CS19), the growing epithelial branches begin to organize into distinct trunk and tip domains (FIGURE 1) (118). The progenitor cells in the tip domain give rise to acinar cells while the trunk progenitors yield ductal and endocrine cells. At 14 wk post conception (wpc), tip-trunk segregation is complete and trunk cells lose GATA4 expression while tip cells cease SOX9 expression (118).

FIGURE 2.

FIGURE 2.

Overview of transcription factor expression during human beta cell development. Schematic overview of the signaling cascade governing human pancreas development including currently known transcription factors responsible for cell type fate determination. Transcription factors depicted in green are associated with the development of monogenic diabetes; in red are the transcription factors known to cause MODY.

Notably, the succession of appearance of endocrine cells in the early developing human pancreas differs from mouse. Rather than the glucagon+ that appear first in the mouse, insulin+ cells appear first in the developing human pancreas, around 49–52 dpc (CS21) (FIGURE 1), 3 wk after budding, followed by glucagon+ and somatostatin+ cells at 8.5 wpc and by pancreatic polypeptide+ and ghrelin+ cells at 9 wpc (118, 160, 198, 214). At mid-gestation (14–16 wpc), a near 1:1 ratio between insulin+ and glucagon+ cells is observed that increases with time to approximate 1.5:1 postnatally (120). While 20–40% of the early endocrine cells coexpressed insulin and glucagon between 9 and 16 wpc, this fraction declines by 21 wpc such that the vast majority of fetal endocrine cells is monohormonal (120, 214). Notably, ghrelin does not colocalize with insulin or glucagon, suggesting a separate ontology of epsilon cells (214). While clusters of fetal beta cells are present by 10 wpc (118), true islet-like structures only appear from 12 wpc onward (198), reaching a maximum average size at 14 wpc (169).

Rodent studies have demonstrated the importance of the vasculature in pancreatic endocrine development (70, 146, 287; reviewed in Ref. 272). In human pancreas, endothelial cells localize near small beta cell clusters at 10 wpc (198) and, by 14 wpc, vascular structures invade the fetal islets (169, 198). Beyond this observation, more work is needed to fully understand the role of endothelial cell differentiation and blood vessel formation in the developing human pancreas.

2. Proliferation of MPPCs and early endocrine cells

Reported cell proliferation rates in pancreas range widely, at least partially due to the use of different proliferation markers (191). As the human fetal pancreas grows, proliferation of the epithelial cells drops significantly, with highest rates seen around the expanding periphery of the organ while endocrine differentiation from progenitor cells occurs more centrally (119, 198). Both exocrine and endocrine cell replication rates decrease between 8 and 34 wpc (169). However, a transient burst of proliferation occurs neonatally in differentiated duct and beta cells (95).

As noted above, mesenchymal signals, especially Fgf7 (74) and Fgf10 (25), drive the expansion of the MPPCs in rodents. Human pancreatic mesenchyme also expresses both growth factors, their levels parallel MPPC expansion (22), and they activate the proliferation of human pancreatic epithelial cells in vitro (282). Together these data suggest a similar role for FGF7 and FGF10 in both human and rodent MMPC growth.

A careful analysis of the transcriptional regulatory program in human MPPCs revealed a role for noncanonical WNT regulatory genes, similar to developing mouse pancreas, and the Hippo signaling effector TEAD1 and its coactivator YAP in human MPPC expansion, mediated in part through the transcription factor SOX9 (45), which regulates MPPC growth in mouse and human (119, 197, 232). In addition, the WNT agonist R-spondin1 (RSPO1), FGF10 and epidermal growth factor (EGF) enhance human MPPC expansion in long-term cultures (27). Although Notch and retinoic acid signaling also increase mouse MPPC proliferation (reviewed in Refs. 126, 192), their role in human pancreas development is unknown. Taken together, despite evidence for the involvement of specific extracellular signals in human MMPC expansion, a comprehensive picture is still lacking.

3. Monogenic diabetes and transcription factor hierarchy in human pancreas development

The spatiotemporal expression pattern of transcription factors in the developing human pancreas taken together with the discovery of monogenic forms of diabetes caused by mutations in many of the same transcription factors confirm their importance in human pancreatic development (reviewed in Ref. 57). The hierarchy of transcription factors guiding human pancreas development has been partially disclosed by genetic studies of permanent neonatal diabetes (PNDM). Genes that cause pancreas agenesis and therefore likely contribute to early pancreas specification in human include PDX1 (251), PTF1A (230, 276), and GATA6 (49, 63, 147). Defects in endocrine cell development and manifestation of PNDM or MODY are linked to mutations in GATA4 (236), SOX9 (197), RFX6 (242), GLIS3 (66, 231), MNX1 (28, 81), NEUROD1 (216), and NKX2.2 (81). In addition, heterozygous loss-of-function mutations of PDX1 and HNF1b are associated with MODY4 (250) and MODY5 (69, 286), respectively.

NEUROG3, a basic helix-loop-helix transcription factor that acts as the initiating pro-endocrine transcription factor in rodents (7, 94, 229), is transiently expressed in scattered epithelial cells during human pancreas development around 8 wpc, concomitant with the appearance of the first hormone-expressing cells (FIGURE 2) (118). NEUROG3 expression in the developing pancreas peaks between 10 and 14 wpc, declines after 18 wpc on (57, 118, 222), and disappears around 35–41 wpc (222). This wave of NEUROG3 expression overlaps with the appearance of differentiated endocrine cells in the pancreas (120, 222, 225). In addition, NEUROG3+/PDX1+, NEUROG3+/INS+, and NEUROG3+/GLUC+ cells can be detected briefly during these early stages of development (44, 160). However, in contrast to the biphasic Neurog3 expression pattern that parallels the first and second transition of endocrine differentiation in murine pancreatic development (271), only a single phase of NEUROG3 expression is observed in the developing human pancreas (271). Interestingly, the lack of an early phase of NEUROG3 expression and of a separate primary transition of endocrine differentiation may result from differences in the vascular signals: in human development, the paired dorsal aortas do not have early contact with the dorsal endoderm as they do in mouse embryos, underscoring the potential importance of vascular-derived signals in development of the human endocrine pancreas (118).

Based on the hierarchy of transcription factors during human pancreas development, the success of endocrine (trans)differentiation protocols can be gauged as follows (118): 1) foregut endoderm should express FOXA2 and SOX17 (dorsal endoderm) or SHH (ventral endoderm); 2) endoderm fated to become pancreas should express PDX1, FOXA2, GATA4, NKX6.1, and SOX9; 3) trunk progenitors that give rise to ductal and endocrine cells should express PDX1, FOXA2, SOX9, and NKX6.1; and 4) transient expression of NEUROG3 to initiate endocrine differentiation should correlate with loss of SOX9 and with gain of NKX2.2 and ISL1 to define fetal beta cells (118). Additional transcription factors such as PAX6, RFX6, PTF1A, HNF1B, GATA6, ONECUT1, and MNX1 should be added to this checklist since clinical case reports have demonstrated their necessity for human islet development (81).

4. A ductal niche for MPPCs

Because the MPPCs form the branching, ductlike epithelium where NEUROG3+ cells and differentiated endocrine cells first appear in the developing human pancreas, duct cells have been proposed as endocrine progenitors (32). In addition, during early fetal human development, all pancreatic epithelial cells express the ductal marker keratin 19 (KRT19) while some single endocrine cells and small islet cell clusters express both duct and endocrine markers (32, 33, 169). Many transcription factors that are confined to the postnatal ductal compartment are also expressed in MPPCs during development. Furthermore, lineage tracings using ‟duct-specificˮ promoters during early mouse pancreas development suggest a ductal origin of endocrine cells (86, 140, 141, 244). However, lineage tracing past the MPPC stage, when terminal differentiation is established, argues against a duct cell origin of new endocrine cells after that time (86, 140, 141, 244). Moreover, the proximity of two cells types cannot distinguish an important anatomical niche from an actual lineage relationship in which one cell type differentiates into another. Location alone is never sufficient; formal proof should be obtained from carefully designed lineage tracing experiments.

B. Postnatal Human Beta Cell Homeostasis

In humans, the adult beta cell mass roughly constitutes 2% of the total pancreas mass, corresponding to 1–2 g of tissue (278). Under normal conditions, beta cell turnover is tightly regulated. Beta cell expansion in human mainly occurs during the perinatal period, i.e., 2 mo before and after birth, when ~3% of the insulin+ cells express Ki67, a marker of active cell cycling (131, 168, 169). This expansion decreases rapidly with age such that, by adulthood and under normal physiological conditions, human beta cells are mostly quiescent with replication rates below 0.1% (194). Consistent with a very low rate of replacement, human beta cells have a long half-life (54) and rarely undergo cell death (41, 168).

Notably, rates of adult beta cell proliferation are substantially lower in humans than in rodents. This difference in replicative potential between human and rodent beta cells might be explained by species-specific replicative aging: in humans, telomere shortening limits replication and leads to senescence (279). This mechanism is considered a critical anti-tumor defense, selected under evolutionary pressure to compensate for the much larger human body and its longer lifespan (237).

However, as in rodents, human beta cells can respond to increased metabolic demands, such as obesity (101) or pregnancy (40, 212), by numeric or functional compensation. In humans, the source of new beta cells in settings of increased metabolic demand or pancreatic damage remains controversial (91), but in mice, after birth, lineage tracing indicates that in most circumstances new beta cells come from the proliferation of preexisting beta cells (68, 264, 282).

In general, nutrients, especially glucose, activate signaling pathways that can mediate beta cell growth (288). Hypoglycemia leads to beta cell atrophy, whereas persistent hyperglycemia provokes hypertrophy and hyperplasia (129, 162). Beta cells are uniquely designed to detect even small changes in extracellular glucose concentrations. For instance, in mice, a 4-day-long infusion of 50% glucose results in mild hyperglycemia, hyperinsulinemia, and increased beta cell replication (3), the latter correlating with increased levels and nuclear localization of cyclin D2 in beta cells (3). Glucose metabolism also shortens the refractory period of beta cells, primarily by increasing the abundance of cyclin D2 postmitosis (223). Notably, aging negatively correlates with the mitogenic effects of glucose on the beta cell in vitro, possibly due to decreased PDX1 expression (161). A mitogenic effect of glucose on human beta cells has been observed both in vitro and after transplantation under the kidney capsule of immune-deficient mice (154, 174, 248).

Glucose phosphorylation by glucokinase and subsequent ATP-dependent closure of potassium (KATP) channels and membrane depolarization are crucial for glucose-mediated beta cell replication (201). Haploinsufficiency for glucokinase in mouse beta cells blunts beta cell hyperplasia following high-fat diet (261). In humans, a glucokinase-activating mutation was associated with abnormally large islets and increased beta cell proliferation in pancreatic tissue resected from a 3 yr old (130), and treatment with a glucokinase-activating drug induced the proliferation of human beta cells via protein kinase C (PKC)-ζ (145). The glucose-sensing transcription factor carbohydrate response element-binding protein (ChREBP) also plays a role in glucose-stimulated expression of cell cycle regulatory genes and beta cell proliferation (174, 293).

The increase in insulin production and secretion induced by glucose catabolism also contributes to beta cell expansion through the induction of compensatory endoplasmic reticulum signals (235), or possibly through autocrine stimulation of the insulin receptor itself (11). Glucose catabolism also activates insulin receptor substrate 2 (IRS-2) via the Ca2+/calcineurin/NFAT pathway (64) and regulates mTOR and cyclin D2 via activation of PKC-ζ to promote beta cell replication independent of the insulin receptor (145, 248).

These homeostatic mechanisms for balancing beta cell mass with metabolic demands suggest that human beta cell replication can be triggered even when aging and maturation have forced them into a dormant quiescent state. If we can better understand and control these pathways, we may develop clinically useful tools for controlled beta cell expansion.

1. Postnatal human beta cell neogenesis

Despite indirect evidence, neogenesis of beta cells in postnatal humans has never been directly demonstrated. The indirect evidence consists of histological observations of single insulin+ cells dispersed over the pancreatic parenchyma (31), of cells coexpressing insulin and the duct marker, and of insulin+ cells within ducts (29). However, as noted before, conclusions based on histology alone can be misleading and do not provide definitive proof.

Beta cell neogenesis from a non-beta cell also requires further steps of maturation to yield a normally functioning adult beta cell that can correctly regulate insulin secretion in response to glucose and other physiological signals. This maturation process is still poorly understood. Relative to mature beta cells, FACS-purified immature human beta cells (PDX1+/INSlow) are enriched for transcripts encoding proteins characteristic of beta cell development, cell cycling, apoptosis, and other islet cell types. Thus the heterogeneous expression pattern of INS and PDX1, as described previously in vitro (255), also occurs in vivo (256). This subset of PDX1+/INSlow beta cells also contain C-peptide and GLUT2 (42). More extensive knowledge of human beta cell maturation will further refine strategies for in vitro generation of fully functional beta cells for the treatment of diabetes mellitus.

2. Postnatal human beta cell replication

Given the technical hurdles to studying the human pancreas in vivo, available data on human beta cell mass and turnover are very scarce. Most studies are limited to analysis of post mortem tissue samples or to in vitro study of cells isolated from cadaveric pancreases. Human pancreas samples are inherently variable, mandating the analysis of a sufficient number of, preferably healthy, donors to draw valid conclusions (115, 131, 168, 215). Furthermore, ante mortem events such as prolonged life support and post mortem handling of the tissues can alter assessment of beta cell proliferation as assessed by Ki67 staining (115, 254).

Notwithstanding these obstacles, we know that the highest rates of human beta cell replication occur during infancy (0–3 yr), thereafter declining from youth to adulthood (131, 168, 275), with the peak in beta cell mass per kilogram body weight preceding the increase in insulin demand that occurs during puberty (177). The increase in beta cell mass during infancy mainly results from an increase in the number of beta cells per islet rather than the number of islets (168). A study using thymidine analogs administered 8 days to 4 yr before death found that beta cell labeling could be detected only in the youngest patients (18–20 yr old) and was absent from subjects over 30 yr of age (194), confirming that the human beta cell mass is largely established by the age of 20 (55).

Similar to other cell populations, the beta cell mass is not only determined by hyperplasia but also balanced by cell death through apoptosis, a process collectively referred to as “remodeling” (90). The frequency of beta cell apoptosis in pancreases obtained from normal human fetuses and infants is low during weeks 17–32 of gestation, increases perinatally, and declines again after 6 mo of age, adding to the relative increase in beta cell abundance at early childhood.

The limited proliferative capacity of adult human beta cells implies they have an extended lifespan. Cellular longevity can be deduced from intracellular lipofuscin body content. Lipofuscin bodies are multivesicular storage organelles from the lysosomal system, and their accumulation in a cell is an indication of cell aging. An age-related increase in the fraction of lipofuscin body-positive beta cells has been observed in human (89% in 1 yr olds, 95% in 5 yr olds, 97% in 20 yr olds, and 98% after age 50), again supporting the conclusion that the human beta cell mass is largely established by the age of 20 (54, 55).

Despite the low rates of beta cell replication in adult humans, some research efforts have focused on the stimulation of beta cell replication as a means of beta cell replacement. Although a number of growth factors have been proposed to drive beta cell replication in vivo, few, if any, have been unequivocally validated as effective human beta cell mitogens. In mice, beta cell proliferation and mass expansion can be stimulated by infusion with the insulin receptor antagonist S961. Initially, these effects were attributed to a liver-derived mRNA encoding angiopoietin-like 8 (ANGPTL8, renamed ‟betatrophinˮ) (122, 283). However, S961 failed to induce a similar proliferative response in human beta cells engrafted into mice (77), and elevated plasma concentrations of betatrophin were found in patients with longstanding T1DM, suggesting that betatrophin alone may not protect or expand human beta cell (76). Subsequently, repeated studies could not confirm the betatrophin effects in rodents that could not attribute the effects of the insulin receptor antagonist to betatrophin (60, 98, 284, 285). The effects of S961, however, were reproducible in rodents, and other studies suggest that the liver secretes the protease inhibitor SerpinB1 in response to insulin resistance and obesity in mouse, zebrafish, and human (73). SerpinB1 acts as an inhibitor of elastase and modulates phosphorylation of several factors related to the beta cell cycle and survival, including p-GSK3, p-MAPK3, and p-PRKAR2B (73).

As it stands, the list of mouse beta cell mitogens that fail to induce a comparable human beta cell proliferative response is growing and, among others, includes growth hormone, prolactin, insulin-like growth factor I (IGF-I), placental lactogen, betacellulin, hepatocyte growth factor, serotonin, gastrin, gamma-aminobutyric acid, osteocalcin, and adenosine kinase inhibitor (24, 33, 249). Nevertheless, the search for effective human beta cell mitogens to restore an adequate beta cell mass remains important, given the presence of residual insulin+ cells in most diabetic patients, even in those with longstanding T1DM (167). Importantly, studies that evaluate cell proliferation should examine the presence of mitotic figures and positivity for the G2 to M transition marker phosphohistone H3 to prove that thymidine analog or Ki67-positive cells indeed are driven towards a proliferative pathway, rather than toward a polyploid state that is relatively frequent in normal human pancreas (72). Obviously, any clinical application of human beta cell mitogens will require concurrent inhibition of the original cause of beta cell loss, and must be monitored for aberrant cell growth and tumorigenesis.

C. Pregnancy-Associated Changes in Human Beta Cell Mass

Proper fetal growth relies on a constant and adequate flow of nutrients from the mother to the fetus. The largest portion of the energy required by the growing fetus comes from glucose diffusion across the placenta (17, 18, 103). During early pregnancy, a passive glucose gradient is maintained by the fetal beta cells through high basal insulin secretion (10, 78). However, as the fetus grows, the fraction of maternal glucose diverted to the fetus grows. To guarantee a sufficiently high maternal glucose concentration to maintain this glucose gradient in the later part of pregnancy, hormones secreted by the placenta induce maternal insulin resistance (144, 184). To prevent excessive nutrient flows to the fetus after meals, the maternal peripheral insulin resistance is balanced by an increase in maternal beta cell mass in both rodents and humans (1, 40, 262, 265, 268, 280). In pregnant rodents, the maternal beta cell mass expansion occurs mainly, if not exclusively, by beta cell replication (1, 262, 265, 280). The dominant mechanism in human, be it replication, neogenesis, or a combination of both, remains unclear.

In rodents, lactogenic hormones from the pituitary and placenta trigger beta cell replication as demonstrated by the necessity of an intact prolactin receptor (PRLR) (5, 84, 93, 113, 206) and by the effects of prolactin and placental lactogen in vitro (3537, 245). PRLR signaling induces key genes that control serotonin production in the maternal beta cells (133, 148, 213, 228). In addition, a beta cell-specific switch in serotonin receptor expression occurs during rodent pregnancy: expression of the Gαq-linked HTR2B receptor increases while expression of the Gαi-linked HTR1D receptor (133) falls. These changes result in increased beta cell replication driven by the HTR2B receptor. At the end of pregnancy and post partum, HTR2B expression is downregulated and HTR1D expression reinstated, causing a drop in beta cell replication and increased beta cell apoptosis. In addition, serotonin signaling causes enhanced insulin secretion by mildly depolarizing the cells through upregulation of the ionotropic HTR3A receptor (187). During rodent pregnancy, PRLR-induced JAK2/STAT5 signaling impacts many other signaling pathway components that may contribute to beta cell adaptation, including insulin receptor substrate 1 (IRS1), insulin receptor substrate 2 (IRS2), phosphatidylinositol 3-kinase (PI3K), AKT, p70S6K, mTOR FOXM1, HNF4a, FOXD3, and menin (5, 6, 97, 114, 128, 199, 289, 291, 292).

The recent, rapid, and independent evolution of the placenta and the lactogenic hormones and their receptors makes direct comparisons among the placental mammals difficult (53, 99). In addition, compared with rodent, human beta cells are much more resistant to mitogenic stimuli and rarely divide (275). However, we do know that, as in rodents, beta cell mass increases in pregnant women, based on two autopsy studies. The first reported a 1.4-fold increase in human beta cell mass during pregnancy, did not detect an increase in beta cell proliferation, and concluded that new beta cells were generated by neogenesis (40). However, as noted above, heterogeneity among pathological samples may complicate correct interpretation of histological observations. Also, a temporal peak in human beta cell replication during pregnancy is easily missed if sampling does not span the entire period of pregnancy. The second study only assessed maternal pancreases from the latter half of and detected a 2.4-fold increase in human beta cell mass (268).

It remains to be determined whether the increase in beta cells during human pregnancy results from increases in proliferation or in neogenesis, or whether any of the pathways that contribute to murine maternal beta cell mass adaptation are active in human pregnancy. Treatment of human islet cells in vitro with prolactin, placental lactogen, or growth hormone increases glucose-stimulated insulin secretion (36), but beta cell proliferation is unaffected (50, 193). Overexpression of murine instead of human STAT5 in human beta cells bypasses this restraint on human beta cell proliferation, leading to the hypothesis that human beta cells have inherent defects in prolactin signaling (50).

The design of experimental models, in vivo or in vitro, that mimic pregnancy-mediated human beta cell adaptation will help to unravel the molecular mechanisms underlying human beta cell expansion and function. The use of serum from pregnant women at various time points throughout pregnancy or the use of placental cell types as signaling source may be a first step in better understanding the effects of pregnancy on human beta cells.

D. Obesity-Associated Changes in Human Beta Cell Mass

Ample studies in rodent models have demonstrated beta cell adaptation and increases in beta cell mass in response to increased adiposity and insulin resistance (8, 61, 294). Lineage tracing has demonstrated that the new beta cells generated in these settings come from the proliferation of preexisting beta cells (264, 282).

Human beta cell mass also correlates with adiposity: nondiabetic, obese humans have ~50% more beta cell mass relative to lean nondiabetic individuals based on autopsy studies (204, 221). This increase apparently depends on genetic ethnicity, since surgically resected pancreases from obese Japanese patients have almost no increase in beta cell mass compared with lean Japanese patients (116). Adiposity-driven beta cell mass expansion is not due to cellular hypertrophy (221), but just as with gestational increases in beta cell mass, the source of the increased numbers of beta cells in obese humans remains uncertain in the absence of lineage tracing. Some evidence suggests that the additional beta cells come from neogenesis (41, 101), while other evidence supports a role for proliferation in the generation of these new beta cells (61).

Several mechanisms have been implicated in obesity-induced beta cell adaptation and replication. As discussed earlier, glucose can promote beta cell proliferation through its catabolism in beta cells (145, 248). However, in nondiabetic obese individuals with normal glucose levels, increases in beta cell proliferation cannot be attributed to hyperglycemia.

To prevent hyperglycemia, however, beta cells must produce and secrete more insulin to compensate for the increasing insulin resistance that occurs as central adiposity increases. In mice, beta cell-specific knockout of the insulin receptor disrupted beta cell replication in response to a high-fat diet, supporting the conclusion that insulin signaling plays a role in compensatory beta cell replication during obesity and insulin resistance (186). Insulin and insulin-like growth factors (IGFs) mediate their proliferative actions via IRS2-PI3K/AKT axis. Downstream of AKT [also known as protein kinase B (PKB)], phosphorylation of p27Kip1 and of the antiproliferative transcription factor FOXO1 promotes nuclear exclusion of the latter, thereby allowing beta cell replication (137).

In addition, the obesity-induced increase in insulin synthesis and processing in high-fat diet or db/db mice induces endoplasmic reticulum (ER) stress and the adaptive unfolded protein response (UPR), which promotes beta cell proliferation via activating transcription factor 6 (ATF6). The adaptive UPR also elevates thymidine-analog labeling in human beta cells upon exposure to high glucose in vitro (235).

In addition to glucose, SerpinB1, and insulin, other obesity-induced, extrapancreatic beta cell mitogens have been proposed, including growth hormone (GH) (111), incretins (GLP-1 and GIP) (270), WNTs (96, 142, 164, 207, 227), and hepatocyte growth factor (HGF) (4, 8, 173).

Finally, microRNAs (miRNAs) also regulate the compensatory beta cell expansion in response to obesity in mice (182). Beta cell-specific disruption of Dicer-1, a key processing enzyme required for the synthesis of all miRNA, caused glucose intolerance due to a significant decrease in pancreas insulin content. A more specific knockdown of miR‐24, miR‐26, miR‐182, and miR‐148 in isolated murine islets enhanced insulin expression, at least in part, by repressing insulin gene transcriptional repressors (172). Other miRNAs, including miR-375 (202) and miR338–3p (117), play a direct role in compensatory beta cell proliferation by regulating a cluster of genes controlling cell cycle and growth. Additional evidence for the role of miRNAs in adaptive beta cell proliferation comes from the reduced compensatory beta cell expansion in the absence of Argonaut 2 (Ago2), which facilitates interactions of miRNAs with their target mRNAs (260).

Taken together, it is becoming increasingly clear that beta cells integrate various extra- and intracellular signals to adjust their mass in response to the increased insulin demand of obesity.

IV. STRATEGIES FOR GENERATING AN ABUNDANT SUPPLY OF TRANSPLANTABLE HUMAN BETA CELLS

A. In Vitro Beta Cell Replication

Transplantation of in vitro expanded human beta cells could offer a logical approach to beta cell replacement in people with diabetes. However, as discussed above, human beta cells are terminally differentiated and long-lived cells that become established in the first few years of life followed by very low proliferation rates in adult humans (54, 194). The age-dependent decline in replicative potential represents a major obstacle to therapeutic beta cell expansion since currently the principal source of human beta cells is adult cadaveric donors.

In contrast to rodent beta cells, human beta cells have been proven difficult to maintain, let alone expand in vitro (71, 193). In addition, often when replication of human beta cells has been achieved, glucose-stimulated secretion from the proliferated cells has declined (217, 226). For example, while glucose is clearly mitogenic for rodent beta cells, it exerts very limited effects on human beta cell cycling, depending on donor age, duration of glucose exposure, and glucose concentration (161).

The low rates of basal and glucose-stimulated proliferation in human beta cells have led to efforts to identify other and better human beta cell mitogens (267). Among candidate human beta cell growth factors, those signaling through IRS2 have attracted particular interest. Even though the efforts to understand the involvement of IRS2 signaling by IGF-I mainly focused on rodent beta cells (158, 203, 211, 257), in vitro propagation of human beta cells will most likely depend on the identification of growth factors that maintain activation of such a key signal transducer. However, mitogens in this pathway may not display much beta cell specificity.

Recent advances in high-throughput screening with large libraries of compounds have identified a number of factors capable of stimulating significant human beta cell proliferation (239, 274) (TABLE 1). One such molecule, WS6, can stimulate proliferation of human beta cells in a dose-responsive manner (239). Although its mechanism of action is largely unknown, involvement of Erb3 binding protein (EBP1) and of the IκB kinase IKKε has been suggested (239). In additional screens, small molecule inhibitors of the DYRK1A kinase were identified as potent inductors of human beta cell proliferation (67, 238, 274). Two of these, harmine and INDY, inhibit DYRK1A by inserting into its ATP-binding pocket. The pro-proliferative effects of the DYRK1A inhibitors have been linked to the calmodulin-calcineurin-cMyc pathway.

Table 1.

Postnatal sources of new human beta(-like) cells

Method of Beta Cell Generation Cell of Origin Trigger Mechanism Reference Nos.
Replication Beta cell Glucokinase Signals via protein kinase C-ζ 130, 145
Harmine Calmodulin-calcineurin-cMyc pathway 274
INDY (harmine analog) Calmodulin-calcineurin-cMyc pathway 274
WS6 Involvement of Erb3 binding protein (EBP1) and the IkB kinase IKKe has been suggested 239
Osteoprotegerin (and denosumab) Inhibits glycogen synthase kinase-3 (GSK3), stimulates cAMP response element-binding protein (CREB), and inhibits RANKL/RANK pathway 139
SerpinB1 Activation of proteins in insulin/IGF-I signaling pathway, altered phosphorylation of MAPK, PRKAR2B, and GSK3 subunits 73
Differentiation of ES/iPS cells ES/iPS 7-stage protocol Neurog3 induction, thyroid hormone, and a gamma secretase inhibitor addition to induce Nkx6, 1/ins+ cells, and MAFA induction for maturation 208
Sequential treatment with multiple growth and differentiation factors over a period of 5 wk Progenitor differentiation by modulating signaling by Wnt, activin, hedgehog, EGF, TGF-β, thyroid hormone, and retinoic acid, as well as γ-secretase inhibition 190
15- to 21-day protocol to generate monohormonal endocrine cells Omission of BMP inhibitors at the pancreas progenitor stage, addition of EGF+KGF for rapid Nkx6.1 induction, and BMP+ALK inhibition to synchronize Neurog3 expression 218
Transdifferentiation of adult stem cells Pancreas-derived multipotent precursors from adult human pancreas Facultative stem cells within the beta cell population Spontaneous expansion and differentiation upon isolation and cell culture 243
Islet-derived mesenchymal stem cells Mix of appropriate factors under serum-free conditions Directed differentiation 87
Human bone marrow-derived MSCs Exposure to specific growth factors, mimicking diabetic hyperglycemic niche, supplementation of fetal pancreatic extract Directed differentiation 195
Human bone marrow-derived MSCs Pdx1 overexpression, transplantation into hyperglycemic niche Forced differentiation by transcription factor overexpression 127
Human multipotent adipose tissue-derived stem cells Multi-step protocol Directed differentiation 48, 134, 176, 188
Human multipotent adipose tissue-derived stem cells PDX1 overexpression Forced differentiation by transcription factor overexpression 151
Reprogramming of nonpancreatic cell types Hepatocytes Ectopic expression of PDX1 Inhibition of liver-gene expression by inhibition of C/EBPβ 23, 170, 171, 181, 224, 290
Hepatocytes Overexpression of PDX1 + exendin-4 Exendin-4 induces liver cell proliferation and maturation of transdifferentiated cells 13
Keratinocytes Overexpression of PDX1 Forced differentiation by transcription factor overexpression 166
Fibroblasts Combination of growth factors, chemical compounds, and nonintegrative episomal reprogramming factors Directed differentiation 296
Reprogramming of pancreatic cell types Duct cells PDX1/NEUROG3/MAFA/PAX4 + epigenetic signaling events Forced differentiation by transcription factor overexpression 157
DPP-4i, PPI Directed differentiation 252, 253
Acinar cells MAPK/STAT3 overexpression in an in vitro setting using sequential free-floating and 3D matrix culture Constitutively active MAPK+STAT3 overexpression 152
Alpha cells Nonspecific methyltransferase inhibitor Potential role for p53 34, 82

Overview of the currently known sources of new human beta(-like) cells in vitro and the mechanisms used to stimulate beta cell generation. See text for definitions.

It is clear, however, that these molecules are not beta cell specific but rather trigger cell cycle activation in most cell types, warranting extreme caution and emphasizing the need of beta cell specific delivery of these mitogens. Cell specificity can be enhanced by using combinations of drugs. For example, combining a selective inhibitor of the endocrine tumor suppressor menin combined with a GLP1 receptor agonist (exendin-4) yields potent beta cell-specific mitogenic activity (47).

These reports are encouraging and may lead to strategies to expand human beta cells in vitro or even in vivo. Small molecule screening also identified compounds with activity similar to SerpinB1, the protease inhibitor with beta cell mitogenic activity induced in the liver in response to insulin resistance and obesity (73). Osteoprotegerin (OPG) is yet another factor whose concentration increases in models of beta cell expansion, including obesity and pregnancy. In response to lactogens, OPG induces beta cell proliferation without dedifferentiation, by modulating proliferative pathways in rodent and human islets (139). OPG inhibits glycogen synthase kinase-3 (GSK3), stimulates cAMP response element-binding protein (CREB), and inhibits the RANKL/RANK pathway that acts as a brake on beta cell proliferation (139). Other recently identified human beta cell mitogens were recently reviewed by Shirakawa et al. (241).

Despite the availability of numerous rodent beta cell lines, the first functional human beta cell line (EndoC-βH1) was generated only recently by introducing immortalizing transgenes in human fetal pancreatic tissue (205). This cell line has been further optimized through Cre-mediated excision of the immortalizing transgenes, resulting in a phenotype more similar to genuine human beta cells (22, 226). Despite the potential to generate an unlimited supply of transplantable human beta cells with this or similar lines, the use of lentiviral vectors for immortalization, combined with the potential for incomplete excision of the immortalizing oncogenes in every cell, prevents the direct application of EndoC cells to human therapy, although they remain a uniquely useful tool for drug development and preclinical studies of cell replacement therapy in diabetes.

In contrast to murine beta cells (12, 178, 277), epithelial-to-mesenchymal transition (EMT) of adult human beta cells generates proliferative fibroblast-like cells that, upon serum removal and addition of growth factors, appear to redifferentiate into insulin-producing isletlike structures that can replace beta cell function in hyperglycemic mice (15, 92, 149, 189). The origin, identity, and ultimate fate of these cells have provoked vigorous debate. Eventually the beta cell origin of the proliferating cells was confirmed by genetic lineage tracing (217, 219). Reexpression of insulin and other beta cell-specific genes occurs only in the beta cell-derived mesenchymal cells, demonstrating that these cells arise by redifferentation rather than by de novo differentiation from non-beta cells. Inhibition of NOTCH (15) and Wnt signaling (153) synergistically promote endocrine redifferentiation. Although these cells can further mature in vivo into functional beta-like cells (15), the reproducibility of this approach has been questioned (132). In addition, detailed characterization of the expansion capacity of this approach and the fidelity of the beta cell characteristics of the resulting cells has not been fully established. Also, it remains unknown whether EMT of human beta cells occurs in vivo and, if so, whether it could be therapeutically exploited.

Unrestrained metabolic stress alters the expression of many functionally important and cell type-specific genes in mature beta cells, in a process sometimes broadly labeled “dedifferentiation” or “loss of cell type identity.” This process can lead to cells that express typically progenitor- or alpha cell genes and lose glucose-stimulated insulin secretion. These pathological changes may contribute to the beta cell failure in T2DM (123, 258). In this regard, treatment of beta cell dysfunction should aim for reduction of metabolic stress, thereby potentially correcting these pathological beta cell changes and reestablishing metabolic control.

B. In Vitro (Trans)differentiation to Beta Cells

1. Differentiation of embryonic stem and induced pluripotent stem cells to beta cells

Directed differentiation of human embryonic stem (hES) cells and induced pluripotent stem (iPS) cells into glucose-responsive, insulin-producing beta cells represents a promising route for generating an unlimited source of human beta cells for replacement therapy in people with diabetes (FIGURE 3) (TABLE 1). The most successful protocols use a stepwise process that manipulates the cellular micro- and macroenvironment in stages, based on our understanding of pancreatic development gained from developmental studies. Nevertheless, this approach remains empirical. The first major breakthrough in the field of directed human stem cell differentiation mimicked the normal developmental program, guiding hES cells through formation of definitive endoderm to pancreatic endoderm and subsequently to endocrine progenitor and beta cell-like stages (62). Early protocols generated cells that often expressed multi-hormones and had poor (62, 196) to moderate (121) glucose-stimulated human insulin C-peptide secretion. Careful analysis of the insulin-expressing cells after in vitro differentiation identified many differences from normal adult beta cells on the levels of function, transcriptome proteome, and epigenome (16). Further maturation to glucose responsive, beta-like cells capable of correcting experimental diabetes was achieved by long-term engraftment in immune-deficient mice (39, 143). In vivo, the polyhormonal cells mainly differentiated to a glucagon-positive phenotype while the progenitors were not immediately able to reverse hyperglycemia and engrafted mice required exogenous insulin until the transplanted cells functionally matured and produced sufficient amounts of insulin (209). Notably, besides characterization of the resulting cells at the transcript, protein, and functional levels, epigenetic profiling may reveal further abnormalities caused by a failure to eliminate polycomb group-mediated repression of endocrine-specific genes (281).

FIGURE 3.

FIGURE 3.

Nonpancreatic cell sources for beta cell generation. Illustration of the potential use of nonpancreatic cell types to generate new beta(-like) cells in vitro based on hES/hIPS directed differentiation or postnatal donor cell reprogramming. The cells of origin depicted represent healthy donor cells. Functional beta(-like) cells could be reintroduced to replenish the beta cell pool lost in diabetes patients.

The development of patient-derived iPS cells now allows the generation of autologous cells for transplantation (163), although this approach does not solve the problem of autoimmunity in patients with type 1 diabetes. The availability of genome-wide reference maps of DNA methylation and gene expression of hES and iPS cells facilitates in-depth characterization of different cell lines (26). Genome-wide transcriptional analysis of insulin+ cells derived from three independent human iPS lines resulted in cells resembling human fetal beta cells rather than adult beta cells. Similar to hES, in vivo maturation was necessary for iPS-derived cells to advance to a more adult beta cell phenotype (112).

Several groups have reported the generation entirely in vitro of hES-derived cells with more similarities to mature, functional cells (190, 208) reported optimized protocols for the in vitro generation of mature, functional beta-like cells from hES cells. The first described a seven-stage protocol resulting in the efficient conversion (>50%) of hES cells to monohormonal, phenotypically mature, and glucose responsive beta-like cells, stably expressing NKX6.1, insulin, and MAFA. Although these cells closely resemble adult beta cells, important differences remained. Most notably, the hES-derived beta-like cells showed a blunted and delayed response during dynamic glucose stimulation with normal Ca2+ oscillations observed in only a fraction of the insulin+ cells. Nonetheless, when transplanted into mice, these cells reversed diabetes within 40 days (208).

The second study reported a scalable three-dimensional culture system using sequential treatment with multiple growth and differentiation factors over a period of 5 wk to transform hES cells into monohormonal, functional beta-like cells. Insulin+ cells (33% of final cells) displayed key features of mature human beta cells including normal Ca2+ oscillations and glucose responsiveness, expression of mature beta cell markers (PDX1, NKX6.1, insulin, C-peptide, ZnT8), and ultrastructural features reminiscent of adult beta cells. When transplanted into mice, these cells produced detectable circulating human insulin within 2 wk (190).

Recent reports demonstrated that synchronization of NEUROG3 expression following initial induction of PDX1+NKX6.1+ MPPCs prevents preemptive endocrine differentiation and generation of dysfunctional polyhormonal cells, allowing greater yields of functional beta-like cells (210, 218). A followup report described the generation of functional beta-like cells by a similar protocol from iPS cells derived from a patient with T1DM (175).

The generation of functional, mature beta-like cells from hES/human iPS cells has moved stem cell-derived beta cells closer to clinical application. The United States Food and Drug Administration approved an Investigational New Drug (IND) application for a phase1/2 safety, tolerability, and efficacy trial of stem cell-derived encapsulated cell replacement therapy in adult patients with T1DM in 2014.

Despite advancements, several concerns regarding the use of pluripotent-stem cell-derived beta cells in patients remain. These concerns include the risk of tumor formation, either from undifferentiated pluripotent cells that might remain among the differentiated beta-like cells or from oncogenic mutations that accumulate during the differentiation process. Also, cells with unregulated insulin secretion could cause hypoglycemia in the recipient.

In addition, transplanted beta-like cells must be protected from allograft rejection (for beta-like cells not derived from the recipient) and autoimmune destruction (in recipients with T1DM) by blocking or modifying the immune response. Clinical application will also require cell culture on an industrial scale, and potentially the generation of an hESC bank based on growth, differentiation, and HLA-type matching (43, 179, 266). Whether patient-derived iPS cells can replace hES cells for cell therapy remains to be determined, but already patient-derived iPS cells are contributing to disease modeling.

2. Differentiation of adult stem cells to beta cells

The end of the last century witnessed much excitement over the possibility that adult stem cells and hematopoietic stem cells (HSCs) might have the capacity to differentiate across tissue lineages and even across germ layer boundaries (FIGURE 3). For example, HSCs were thought to transdifferentiate into lung, gut, skin, muscle, liver, and neurons. In 2003, bone marrow cells transplanted into lethally irradiated mice reportedly generated “glucose-competent” beta cells, but subsequent studies could not replicate these unexpected findings (52, 150, 259). In general, these observations of adult stem cell plasticity were often over-interpreted or could be explained by experimental artifacts (273).

Interestingly, a subsequent study showed that bone marrow-derived stem cells could induce pancreatic regeneration after injection in diabetic mice in a non-cell autonomous manner (110). Rather than transdifferentiating into beta cells, the injected cells acquired an endothelial cell phenotype and stimulated beta cell proliferation (110). Although the exact mechanism remains unclear, this observation fits well with the role of signals from mesoderm-derived tissues in pancreas development and adult beta cell proliferation and function.

Autologous non-myeloablative HSC transplantation led to insulin independence with good glycemic control in a series of patients with newly diagnosed T1DM (58). However, this exciting finding could not be corroborated in patients with T1DM of more than 5 yr duration (75) or when umbilical cord blood was infused intravenously in young children with T1DM (100). This difference in outcomes suggests that the preparatory treatment with cyclophosphamide and rabbit anti-thymocyte globulin altered the disease course in new-onset patients, while the subsequent stem cell infusion had no measurable impact on the disease.

Recent research focus has shifted to the isolation and study of other populations of adult human stem cells besides the well-characterized HSCs. For example, so-called “pancreas-derived multipotent precursors” (PMPs) were isolated from islets of adult human and mouse pancreas, independent of age (243). These PMP cells proliferate and differentiate into multiple differentiated neuronal and pancreatic cell types. The generated beta cells contain about one-third of the amount of insulin of a primary beta cell. After transplantation into mice, these cells produced circulating human C-peptide and ameliorated experimental diabetes. Immunohistochemistry of the graft detected human insulin+ cells (243). Independent confirmation and further characterization of the PMPs and PMP-derived beta cells and their molecular mechanism of differentiation will help determine the utility of this approach to beta cell replacement therapy.

Mesenchymal stem cells (MSCs) isolated from various tissues have also been suggested as a potential source of endocrine progenitors. For example, human islet-derived MSCs develop into isletlike clusters when exposed to a cocktail of differentiation factors under serum-free conditions. After differentiation, expression of glucagon, somatostatin, and transcription factor Islet 1 (ISL1) was noted. Although the resulting cells stained for C-peptide, they did not secrete insulin in a glucose-regulated manner (87).

Human bone marrow-derived MSCs (hBM-MSC) form isletlike aggregates after exposure to a differentiation cocktail. However, significant maturation, secretion of human C-peptide, and normalization of glycemia were only observed after transplantation in mice with lowered beta cell mass after pancreatectomy or following streptozotocin-induced beta cell ablation (195). Similar findings were previously reported using hBM-MSCs ectopically expressing PDX1 (127).

Finally, human multipotent adipose tissue-derived stem cells (hASCs) have been proposed as a readily accessible source of stem cells for beta cell generation (134). Following a multistep differentiation protocol, abdominal fat tissue obtained by liposuction generated isletlike aggregates containing insulin+ cells that secreted insulin in vitro (48, 176, 188). Two to three weeks after transplantation in diabetic mice, these cells could restore near-normoglycemia for up to 8 wk (48). As with hBM-MSCs, overexpression of PDX1 in hASCs enhanced pancreatic differentiation. The resulting cells secreted insulin in response to glucose stimulation in vitro and reduced blood glucose levels in diabetic mice without achieving normoglycemia (151) (TABLE 1).

Despite continued interest in generating beta cells from adult stem cells due to their ready availability and practicality, it remains unclear how similar the derived cells are to normal beta cells at the level of gene expression, stability, or function or how well they compare with beta-like cells derived from hES or human IPS cells.

3. Transdifferentiation of nonpancreatic cells to beta cells

Transdifferentiation of differentiated adult human nonpancreatic cells represents another potential route to new beta cells (TABLE 1). Because pancreas and liver cells originally derive from bipotent progenitor cells in the gut endoderm during embryonic development (65), transdifferentiation of liver cells became a major focus in the quest for beta cell replacement (FIGURE 3). In the first experiment of its kind, ectopic expression of PDX1 in adult human hepatocytes induced insulin synthesis, processing, storage, and glucose-regulated release. In diabetic mice, these cells reduce blood glucose levels (13, 23, 127, 170, 171, 180, 181, 224, 290). Treatment with GLP1 agonist exendin-4 further enhanced this transdifferentiation to insulin-producing cells (13). The reprogrammed human liver cells expressed pancreas-specific genes, and cell lineage tracing showed that the insulin+ cells originated from genuine hepatocytes. Further study, however, suggested that these insulin+ cells appear only in culture after undergoing EMT and that a similar phenomenon may not occur in vivo (170). More recently, the TALE homeodomain transcription factor Tgif2 was used to reprogram mouse liver cells to pancreas progenitor-like cells that express Pdx1, Ptf1a, Neurod1, Pax6, MafA, Isl1, and Insm1, a potential starting point for generation of beta(-like) cells (46).

Similar to hepatocytes, human keratinocytes ectopically expressing PDX1 also transformed into glucose-responsive beta-like cells (166). The pool of potential nonpancreatic cell sources for beta cell generation was even further enlarged by the derivation, from human fibroblasts, of proliferative endodermal progenitors and, subsequently, pancreatic beta-like cells that exhibit glucose-stimulated insulin secretion in vivo (296). Despite the range of nonpancreatic cell sources (many of which fall beyond the scope of this review), efforts to transdifferentiate nonpancreatic cells towards beta cells have recently dwindled, possibly because these efforts have failed to generate functionally equivalent beta-like cells. Increasing evidence suggests that tissue-specific resident or facultative stem cells may provide a more successful starting cell, and newer protocols using hES/iPS cells are generating cells increasingly similar to normal adult human beta cells.

4. Transdifferentiation of pancreatic cell types to beta cells

Because pancreatic endocrine cells arise during human pancreas development from branching pancreatic endoderm comprised of epidermal MPPCs that share many similarities with adult human pancreatic duct cells, the duct cells are often considered candidates for transdifferentiation towards endocrine cells. Adult human pancreatic duct cells express a network of transcription factors that also play a role in the maintenance of progenitor cell characteristics in embryonic MPPCs (105, 159).

Several studies have shown that adult human duct cells can propagate in vitro as well as generate new isletlike cells (FIGURE 4) (30, 89). These conclusions were based on the observation that isletlike structures could bud from three-dimensional ductal cysts (30, 89). Notably, when contaminating endocrine cells were depleted from these cultures, the original findings were no longer reproducible, suggesting that beta cell dedifferentiation rather than neogenesis may explain the former conclusions (88). In a proof-of-concept study, ectopic expression of Neurog3 in adult human duct cells has shown a capacity for adult duct cells to differentiate into endocrine-like cells (106). Other reports show how cotransplantation with fetal cells (102) or engraftment followed by introduction of gastrin/GLP1 appears to induce beta cell generation from human ducts (252) (253). Importantly, none of these studies included lineage tracing, and in adult mice, lineage tracing with different duct-specific promoters has not supported a duct-related origin of regenerating endocrine cells (86, 140, 141, 244). Working with human cells, lineage tracing is imperative to rule out redifferentiation of residual degranulated or fully dedifferentiated beta cells rather than actual transdifferentiation of exocrine cells.

FIGURE 4.

FIGURE 4.

Strategies to increase human beta cell numbers in vitro from pancreatic cell types. Schematic overview of the strategies used to generate new beta(-like) cells in vitro using cell types of pancreatic origin. Black arrows represent strategies that have been documented; red arrow depicts a potential strategy that has yet to be shown using human cells.

Human acinar cells were genetically traced and shown to undergo EMT in vitro (156). Upon ectopic expression of NEUROG3, PDX1, MAFA, and PAX4 and a series of (epigenetic) signaling events, the human exocrine cells could give rise to insulin+ cells capable of normalizing blood glucose levels (156), thereby extrapolating the initial findings documented in mice in vivo (295). Of note, in vitro transdifferentiation towards beta-like cells seemed most efficient using freshly isolated tissue under conditions whereby EMT was inhibited. Supplementing this approach with ARX inhibition yielded a monohormonal cell population containing 40% C-peptide+, 4% glucagon+, and <2% somatostatin+ cells. The resulting beta-like cells were glucose responsive and expressed insulin protein levels at 15% of adult human islets, similar to hIPS cell-derived beta-like cells. When grafted in hyperglycemic mice, an immediate and prolonged effect on normalization of blood sugar levels was noted (157).

Another approach was based on ectopic expression of activated mitogen-activated protein kinase and STAT3 to induce human acinar-to-beta cell conversion through a NEUROG3+ intermediate (152). The expression of the pro-endocrine factor NEUROG3 was activated in more than half of transduced exocrine cells. Genetic lineage tracing identified human acinar cells as a source of NEUROG3+ and insulin+ cells. However, the number of insulin+ cells in vitro only increased following sequential suspension and three-dimensional Matrigel culture. Long-term engraftment into immune-compromised mice amplified the efficiency of reprogramming to insulin+ cells. Under normoglycemic conditions, the engrafted mice displayed an increase in circulating human C-peptide, starting at around day 90 posttransplantation. Upon chemical destruction of the endogenous rodent beta cells, human C-peptide levels further spiked, and the sharp increase in blood glucose observed in controls was attenuated in engrafted mice. Removal of the graft unequivocally identified the human cells as the source of C-peptide and blood glucose control (152).

These data demonstrate that exocrine cells from human pancreas can be transdifferentiated to transplantable insulin-producing cells that acquire functionality (FIGURE 4). Given the large number of exocrine cells in a donor pancreas, this approach presents a novel strategy to expand beta cell therapy in diabetes. Efficient transdifferentiation of human acinar cells would be able to augment the beta cell number by 10-fold or more. Can exocrine cells, shown to be surprisingly plastic in the rodent pancreas, be the key to resolving the shortage in human donor beta cells? Future experiments will need to find the definitive answers. However, acinar cells also force us to be cautious as emerging evidence identifies these cells as potential origin of pancreatic ductal adenocarcinoma (PDAC), one of the most aggressive pancreas malignancies in humans.

Finally, since the endocrine lineages are closely related, it seems likely that, analogous to rodents, non-beta endocrine cells represent another ideal source for generating new beta cells (FIGURE 4). Indeed, similar to rodents, human alpha and beta cells share a close epigenetic relationship making human alpha cells perhaps even more amenable to convert into beta cells compared with other pancreatic cell types (34, 82). Treatment of adult human islets with a nonspecific histone methyltransferase inhibitor generates GCG+/INS+ and GCG+/PDX1+ cells (34). A potential role for p53 in insulin expression by adult human alpha cells was suggested (82). Vice versa, adult human beta cells were recently converted to alpha-like cells in a model of islet cell cluster formation (247). These observations lend evidence to the notion that chromatin-modifying compounds affect plasticity of endocrine cells.

Regarding the potential of pancreatic cell types to serve as a beta cell replacement (TABLE 1), we can conclude that, historically, human duct cells were considered the cell type with the highest probability for harboring transdifferentiation potential. This notion is mainly due to histological studies observing single insulin+ cells as well as whole islets near ducts in the human pancreas and the detection of endocrine progenitors in the lining of embryonic ductules. However, current evidence is still limited to suggest that duct cells have an increased capacity over other cell types to respond to cell transdifferentiation signals. Recently, alpha, delta, or acinar cells (14, 51, 263) have received increasing attention following their successful transdifferentiation in rodent studies. Human alpha cells bear bivalent epigenetic marks, allowing easier transdifferentiation into beta cells. However, given the relatively small fraction of alpha cells in human islets (9), this approach would generate only a limited amount of transplantable beta cells unless the alpha cell pool would be replenished. This is the case by continuous mobilization of duct-lining progenitor-like cells in mice with ectopic expression of Pax4 (2, 56) or inactivation of Arx (59) selectively in alpha cells. Interestingly, GABA signaling was identified as an inducer of alpha- to beta-like cell conversion (21), and artemisinins were reported to functionally repress Arx via enhancement of GABAA receptor signaling, also in human cells (155). However, conversion of alpha to mature beta cells through artemisinins has just recently been disproven (269).

V. CONCLUSIONS AND PERSPECTIVES

Given the critical importance of the beta cells in metabolic homeostasis, it is not surprising that the complex forces control the size and function of the beta cell population in the pancreas. Recent evidence suggests that multiple signals orchestrated by the interplay among various organs as well as contributions from autocrine and paracrine signals within the pancreas regulate beta cell mass. Thus it seems possible that new beta cells can be generated via multiple routes, a conclusion that provides optimism that we can develop successful therapies for beta cell regeneration. However, despite important conceptual progress, major practical challenges remain before we can meet the ambitious goal of beta cell (re)generation for people with diabetes.

GRANTS

L. Baeyens is supported by European Research Council Grant ERC DLV-677555, the Research Foundation–Flanders (FWO-Odysseus), and JDRF Grants 3-APF-2014–183-A-N and 1-FAC-2018–538-A-N. M. S. German is supported by National Institute of Diabetes and Digestive and Kidney Diseases Grant P30 DK063720, Larry L. Hillblom Grant 2014-D-004-NET, and Nora Eccles Treadwell Grant 10568. H. Heimberg is supported by the VUB-Research Council, the Research Foundation-Flanders (FWO), the DON Foundation, the Interuniversity Attraction Pole Network VII-P07 DevRepair, and the European Union Sixth and Seventh Framework Program.

DISCLOSURES

M. S. German owns stock in Viacyte, Inc. The authors declare no other conflicts of interest, financial or otherwise.

ACKNOWLEDGMENTS

L. Baeyens and M. Lemper contributed equally to this work.

Address for reprint requests and other correspondence: H. Heimberg, Beta Cell Neogenesis (BENE), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium (e-mail: harry.heimberg@vub.ac.be).

REFERENCES

  • 1.Abouna S, Old RW, Pelengaris S, Epstein D, Ifandi V, Sweeney I, Khan M. Non-β-cell progenitors of β-cells in pregnant mice. Organogenesis 6: 125–133, 2010. doi: 10.4161/org.6.2.10374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Al-Hasani K, Pfeifer A, Courtney M, Ben-Othman N, Gjernes E, Vieira A, Druelle N, Avolio F, Ravassard P, Leuckx G, Lacas-Gervais S, Ambrosetti D, Benizri E, Hecksher-Sorensen J, Gounon P, Ferrer J, Gradwohl G, Heimberg H, Mansouri A, Collombat P. Adult duct-lining cells can reprogram into β-like cells able to counter repeated cycles of toxin-induced diabetes. Dev Cell 26: 86–100, 2013. doi: 10.1016/j.devcel.2013.05.018. [DOI] [PubMed] [Google Scholar]
  • 3.Alonso LC, Yokoe T, Zhang P, Scott DK, Kim SK, O’Donnell CP, Garcia-Ocaña A. Glucose infusion in mice: a new model to induce beta-cell replication. Diabetes 56: 1792–1801, 2007. doi: 10.2337/db06-1513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Alvarez-Perez JC, Ernst S, Demirci C, Casinelli GP, Mellado-Gil JM, Rausell-Palamos F, Vasavada RC, Garcia-Ocaña A. Hepatocyte growth factor/c-Met signaling is required for β-cell regeneration. Diabetes 63: 216–223, 2014. doi: 10.2337/db13-0333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Amaral ME, Cunha DA, Anhê GF, Ueno M, Carneiro EM, Velloso LA, Bordin S, Boschero AC. Participation of prolactin receptors and phosphatidylinositol 3-kinase and MAP kinase pathways in the increase in pancreatic islet mass and sensitivity to glucose during pregnancy. J Endocrinol 183: 469–476, 2004. doi: 10.1677/joe.1.05547. [DOI] [PubMed] [Google Scholar]
  • 6.Amaral MEC, Ueno M, Carvalheira JB, Carneiro EM, Velloso LA, Saad MJA, Boschero AC. Prolactin-signal transduction in neonatal rat pancreatic islets and interaction with the insulin-signaling pathway. Horm Metab Res 35: 282–289, 2003. doi: 10.1055/s-2003-41303. [DOI] [PubMed] [Google Scholar]
  • 7.Apelqvist A, Li H, Sommer L, Beatus P, Anderson DJ, Honjo T, Hrabe de Angelis M, Lendahl U, Edlund H. Notch signalling controls pancreatic cell differentiation. Nature 400: 877–881, 1999. doi: 10.1038/23716. [DOI] [PubMed] [Google Scholar]
  • 8.Araújo TG, Oliveira AG, Carvalho BM, Guadagnini D, Protzek AO, Carvalheira JB, Boschero AC, Saad MJ. Hepatocyte growth factor plays a key role in insulin resistance-associated compensatory mechanisms. Endocrinology 153: 5760–5769, 2012. doi: 10.1210/en.2012-1496. [DOI] [PubMed] [Google Scholar]
  • 9.Arrojo e Drigo R, Ali Y, Diez J, Srinivasan DK, Berggren PO, Boehm BO. New insights into the architecture of the islet of Langerhans: a focused cross-species assessment. Diabetologia 58: 2218–2228, 2015. doi: 10.1007/s00125-015-3699-0. [DOI] [PubMed] [Google Scholar]
  • 10.Asplund K, Westman S, Hellerström C. Glucose stimulation of insulin secretion from the isolated pancreas of foetal and newborn rats. Diabetologia 5: 260–262, 1969. doi: 10.1007/BF01212095. [DOI] [PubMed] [Google Scholar]
  • 11.Assmann A, Ueki K, Winnay JN, Kadowaki T, Kulkarni RN. Glucose effects on beta-cell growth and survival require activation of insulin receptors and insulin receptor substrate 2. Mol Cell Biol 29: 3219–3228, 2009. doi: 10.1128/MCB.01489-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Atouf F, Park CH, Pechhold K, Ta M, Choi Y, Lumelsky NL. No evidence for mouse pancreatic beta-cell epithelial-mesenchymal transition in vitro. Diabetes 56: 699–702, 2007. doi: 10.2337/db06-1446. [DOI] [PubMed] [Google Scholar]
  • 13.Aviv V, Meivar-Levy I, Rachmut IH, Rubinek T, Mor E, Ferber S. Exendin-4 promotes liver cell proliferation and enhances the PDX-1-induced liver to pancreas transdifferentiation process. J Biol Chem 284: 33509–33520, 2009. doi: 10.1074/jbc.M109.017608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Baeyens L, Lemper M, Leuckx G, De Groef S, Bonfanti P, Stangé G, Shemer R, Nord C, Scheel DW, Pan FC, Ahlgren U, Gu G, Stoffers DA, Dor Y, Ferrer J, Gradwohl G, Wright CV, Van de Casteele M, German MS, Bouwens L, Heimberg H. Transient cytokine treatment induces acinar cell reprogramming and regenerates functional beta cell mass in diabetic mice. Nat Biotechnol 32: 76–83, 2014. doi: 10.1038/nbt.2747. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 15.Bar Y, Russ HA, Sintov E, Anker-Kitai L, Knoller S, Efrat S. Redifferentiation of expanded human pancreatic β-cell-derived cells by inhibition of the NOTCH pathway. J Biol Chem 287: 17269–17280, 2012. doi: 10.1074/jbc.M111.319152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Basford CL, Prentice KJ, Hardy AB, Sarangi F, Micallef SJ, Li X, Guo Q, Elefanty AG, Stanley EG, Keller G, Allister EM, Nostro MC, Wheeler MB. The functional and molecular characterisation of human embryonic stem cell-derived insulin-positive cells compared with adult pancreatic beta cells. Diabetologia 55: 358–371, 2012. doi: 10.1007/s00125-011-2335-x. [DOI] [PubMed] [Google Scholar]
  • 17.Battaglia FC. Principal substrates of fetal metabolism: fuel and growth requirements of the ovine fetus. Ciba Found Symp 63: 57–74, 1978. [DOI] [PubMed] [Google Scholar]
  • 18.Baumann MU, Deborde S, Illsley NP. Placental glucose transfer and fetal growth. Endocrine 19: 13–22, 2002. doi: 10.1385/ENDO:19:1:13. [DOI] [PubMed] [Google Scholar]
  • 19.Bellamy L, Casas JP, Hingorani AD, Williams D. Type 2 diabetes mellitus after gestational diabetes: a systematic review and meta-analysis. Lancet 373: 1773–1779, 2009. doi: 10.1016/S0140-6736(09)60731-5. [DOI] [PubMed] [Google Scholar]
  • 20.Bellin MD, Barton FB, Heitman A, Harmon JV, Kandaswamy R, Balamurugan AN, Sutherland DE, Alejandro R, Hering BJ. Potent induction immunotherapy promotes long-term insulin independence after islet transplantation in type 1 diabetes. Am J Transplant 12: 1576–1583, 2012. doi: 10.1111/j.1600-6143.2011.03977.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Ben-Othman N, Vieira A, Courtney M, Record F, Gjernes E, Avolio F, Hadzic B, Druelle N, Napolitano T, Navarro-Sanz S, Silvano S, Al-Hasani K, Pfeifer A, Lacas-Gervais S, Leuckx G, Marroquí L, Thévenet J, Madsen OD, Eizirik DL, Heimberg H, Kerr-Conte J, Pattou F, Mansouri A, Collombat P. Long-Term GABA Administration Induces Alpha Cell-Mediated Beta-like Cell Neogenesis. Cell 168: 73–85.e11, 2017. doi: 10.1016/j.cell.2016.11.002. [DOI] [PubMed] [Google Scholar]
  • 22.Benazra M, Lecomte MJ, Colace C, Müller A, Machado C, Pechberty S, Bricout-Neveu E, Grenier-Godard M, Solimena M, Scharfmann R, Czernichow P, Ravassard P. A human beta cell line with drug inducible excision of immortalizing transgenes. Mol Metab 4: 916–925, 2015. doi: 10.1016/j.molmet.2015.09.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Ber I, Shternhall K, Perl S, Ohanuna Z, Goldberg I, Barshack I, Benvenisti-Zarum L, Meivar-Levy I, Ferber S. Functional, persistent, and extended liver to pancreas transdifferentiation. J Biol Chem 278: 31950–31957, 2003. doi: 10.1074/jbc.M303127200. [DOI] [PubMed] [Google Scholar]
  • 24.Bernal-Mizrachi E, Kulkarni RN, Scott DK, Mauvais-Jarvis F, Stewart AF, Garcia-Ocaña A. Human β-cell proliferation and intracellular signaling part 2: still driving in the dark without a road map. Diabetes 63: 819–831, 2014. doi: 10.2337/db13-1146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Bhushan A, Itoh N, Kato S, Thiery JP, Czernichow P, Bellusci S, Scharfmann R. Fgf10 is essential for maintaining the proliferative capacity of epithelial progenitor cells during early pancreatic organogenesis. Development 128: 5109–5117, 2001. [DOI] [PubMed] [Google Scholar]
  • 26.Bock C, Kiskinis E, Verstappen G, Gu H, Boulting G, Smith ZD, Ziller M, Croft GF, Amoroso MW, Oakley DH, Gnirke A, Eggan K, Meissner A. Reference Maps of human ES and iPS cell variation enable high-throughput characterization of pluripotent cell lines. Cell 144: 439–452, 2011. doi: 10.1016/j.cell.2010.12.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Bonfanti P, Nobecourt E, Oshima M, Albagli-Curiel O, Laurysens V, Stangé G, Sojoodi M, Heremans Y, Heimberg H, Scharfmann R. Ex Vivo Expansion and Differentiation of Human and Mouse Fetal Pancreatic Progenitors Are Modulated by Epidermal Growth Factor. Stem Cells Dev 24: 1766–1778, 2015. doi: 10.1089/scd.2014.0550. [DOI] [PubMed] [Google Scholar]
  • 28.Bonnefond A, Vaillant E, Philippe J, Skrobek B, Lobbens S, Yengo L, Huyvaert M, Cavé H, Busiah K, Scharfmann R, Polak M, Abdul-Rasoul M, Froguel P, Vaxillaire M. Transcription factor gene MNX1 is a novel cause of permanent neonatal diabetes in a consanguineous family. Diabetes Metab 39: 276–280, 2013. doi: 10.1016/j.diabet.2013.02.007. [DOI] [PubMed] [Google Scholar]
  • 29.Bonner-Weir S, Li WC, Ouziel-Yahalom L, Guo L, Weir GC, Sharma A. Beta-cell growth and regeneration: replication is only part of the story. Diabetes 59: 2340–2348, 2010. doi: 10.2337/db10-0084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Bonner-Weir S, Taneja M, Weir GC, Tatarkiewicz K, Song KH, Sharma A, O’Neil JJ. In vitro cultivation of human islets from expanded ductal tissue. Proc Natl Acad Sci USA 97: 7999–8004, 2000. doi: 10.1073/pnas.97.14.7999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Bouwens L, Klöppel G. Islet cell neogenesis in the pancreas. Virchows Arch 427: 553–560, 1996. doi: 10.1007/BF00202885. [DOI] [PubMed] [Google Scholar]
  • 32.Bouwens L, Lu WG, De Krijger R. Proliferation and differentiation in the human fetal endocrine pancreas. Diabetologia 40: 398–404, 1997. doi: 10.1007/s001250050693. [DOI] [PubMed] [Google Scholar]
  • 33.Bouwens L, Rooman I. Regulation of pancreatic beta-cell mass. Physiol Rev 85: 1255–1270, 2005. doi: 10.1152/physrev.00025.2004. [DOI] [PubMed] [Google Scholar]
  • 34.Bramswig NC, Everett LJ, Schug J, Dorrell C, Liu C, Luo Y, Streeter PR, Naji A, Grompe M, Kaestner KH. Epigenomic plasticity enables human pancreatic α to β cell reprogramming. J Clin Invest 123: 1275–1284, 2013. doi: 10.1172/JCI66514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Brelje TC, Allaire P, Hegre O, Sorenson RL. Effect of prolactin versus growth hormone on islet function and the importance of using homologous mammosomatotropic hormones. Endocrinology 125: 2392–2398, 1989. doi: 10.1210/endo-125-5-2392. [DOI] [PubMed] [Google Scholar]
  • 36.Brelje TC, Scharp DW, Lacy PE, Ogren L, Talamantes F, Robertson M, Friesen HG, Sorenson RL. Effect of homologous placental lactogens, prolactins, and growth hormones on islet B-cell division and insulin secretion in rat, mouse, and human islets: implication for placental lactogen regulation of islet function during pregnancy. Endocrinology 132: 879–887, 1993. doi: 10.1210/endo.132.2.8425500. [DOI] [PubMed] [Google Scholar]
  • 37.Brelje TC, Sorenson RL. Role of prolactin versus growth hormone on islet B-cell proliferation in vitro: implications for pregnancy. Endocrinology 128: 45–57, 1991. doi: 10.1210/endo-128-1-45. [DOI] [PubMed] [Google Scholar]
  • 38.Brennand K, Melton D. Slow and steady is the key to beta-cell replication. J Cell Mol Med 13: 472–487, 2009. doi: 10.1111/j.1582-4934.2008.00635.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Bruin JE, Erener S, Vela J, Hu X, Johnson JD, Kurata HT, Lynn FC, Piret JM, Asadi A, Rezania A, Kieffer TJ. Characterization of polyhormonal insulin-producing cells derived in vitro from human embryonic stem cells. Stem Cell Res (Amst) 12: 194–208, 2014. doi: 10.1016/j.scr.2013.10.003. [DOI] [PubMed] [Google Scholar]
  • 40.Butler AE, Cao-Minh L, Galasso R, Rizza RA, Corradin A, Cobelli C, Butler PC. Adaptive changes in pancreatic beta cell fractional area and beta cell turnover in human pregnancy. Diabetologia 53: 2167–2176, 2010. doi: 10.1007/s00125-010-1809-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Butler AE, Janson J, Bonner-Weir S, Ritzel R, Rizza RA, Butler PC. Beta-cell deficit and increased beta-cell apoptosis in humans with type 2 diabetes. Diabetes 52: 102–110, 2003. doi: 10.2337/diabetes.52.1.102. [DOI] [PubMed] [Google Scholar]
  • 42.Cabrera O, Berman DM, Kenyon NS, Ricordi C, Berggren PO, Caicedo A. The unique cytoarchitecture of human pancreatic islets has implications for islet cell function. Proc Natl Acad Sci USA 103: 2334–2339, 2006. doi: 10.1073/pnas.0510790103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Campbell PM, Salam A, Ryan EA, Senior P, Paty BW, Bigam D, McCready T, Halpin A, Imes S, Al Saif F, Lakey JR, Shapiro AM. Pretransplant HLA antibodies are associated with reduced graft survival after clinical islet transplantation. Am J Transplant 7: 1242–1248, 2007. doi: 10.1111/j.1600-6143.2007.01777.x. [DOI] [PubMed] [Google Scholar]
  • 44.Castaing M, Duvillié B, Quemeneur E, Basmaciogullari A, Scharfmann R. Ex vivo analysis of acinar and endocrine cell development in the human embryonic pancreas. Dev Dyn 234: 339–345, 2005. doi: 10.1002/dvdy.20547. [DOI] [PubMed] [Google Scholar]
  • 45.Cebola I, Rodríguez-Seguí SA, Cho CHH, Bessa J, Rovira M, Luengo M, Chhatriwala M, Berry A, Ponsa-Cobas J, Maestro MA, Jennings RE, Pasquali L, Morán I, Castro N, Hanley NA, Gomez-Skarmeta JL, Vallier L, Ferrer J. TEAD and YAP regulate the enhancer network of human embryonic pancreatic progenitors. Nat Cell Biol 17: 615–626, 2015. doi: 10.1038/ncb3160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Cerdá-Esteban N, Naumann H, Ruzittu S, Mah N, Pongrac IM, Cozzitorto C, Hommel A, Andrade-Navarro MA, Bonifacio E, Spagnoli FM. Stepwise reprogramming of liver cells to a pancreas progenitor state by the transcriptional regulator Tgif2. Nat Commun 8: 14127, 2017. doi: 10.1038/ncomms14127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Chamberlain CE, Scheel DW, McGlynn K, Kim H, Miyatsuka T, Wang J, Nguyen V, Zhao S, Mavropoulos A, Abraham AG, O’Neill E, Ku GM, Cobb MH, Martin GR, German MS. Menin determines K-RAS proliferative outputs in endocrine cells. J Clin Invest 124: 4093–4101, 2014. doi: 10.1172/JCI69004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Chandra V, Swetha G, Muthyala S, Jaiswal AK, Bellare JR, Nair PD, Bhonde RR. Islet-like cell aggregates generated from human adipose tissue derived stem cells ameliorate experimental diabetes in mice. PLoS One 6: e20615, 2011. doi: 10.1371/journal.pone.0020615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Chao CS, McKnight KD, Cox KL, Chang AL, Kim SK, Feldman BJ. Novel GATA6 mutations in patients with pancreatic agenesis and congenital heart malformations. PLoS One 10: e0118449, 2015. doi: 10.1371/journal.pone.0118449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Chen H, Kleinberger JW, Takane KK, Salim F, Fiaschi-Taesch N, Pappas K, Parsons R, Jiang J, Zhang Y, Liu H, Wang P, Bender AS, Frank SJ, Stewart AF. Augmented Stat5 Signaling Bypasses Multiple Impediments to Lactogen-Mediated Proliferation in Human β-Cells. Diabetes 64: 3784–3797, 2015. doi: 10.2337/db15-0083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Chera S, Baronnier D, Ghila L, Cigliola V, Jensen JN, Gu G, Furuyama K, Thorel F, Gribble FM, Reimann F, Herrera PL. Diabetes recovery by age-dependent conversion of pancreatic δ-cells into insulin producers. Nature 514: 503–507, 2014. doi: 10.1038/nature13633. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Choi JB, Uchino H, Azuma K, Iwashita N, Tanaka Y, Mochizuki H, Migita M, Shimada T, Kawamori R, Watada H. Little evidence of transdifferentiation of bone marrow-derived cells into pancreatic beta cells. Diabetologia 46: 1366–1374, 2003. doi: 10.1007/s00125-003-1182-9. [DOI] [PubMed] [Google Scholar]
  • 53.Chuong EB, Hannibal RL, Green SL, Baker JC. Evolutionary perspectives into placental biology and disease. Appl Transl Genomics 2: 64–69, 2013. doi: 10.1016/j.atg.2013.07.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Cnop M, Hughes SJ, Igoillo-Esteve M, Hoppa MB, Sayyed F, van de Laar L, Gunter JH, de Koning EJ, Walls GV, Gray DW, Johnson PR, Hansen BC, Morris JF, Pipeleers-Marichal M, Cnop I, Clark A. The long lifespan and low turnover of human islet beta cells estimated by mathematical modelling of lipofuscin accumulation. Diabetologia 53: 321–330, 2010. doi: 10.1007/s00125-009-1562-x. [DOI] [PubMed] [Google Scholar]
  • 55.Cnop M, Igoillo-Esteve M, Hughes SJ, Walker JN, Cnop I, Clark A. Longevity of human islet α- and β-cells. Diabetes Obes Metab 13, Suppl 1: 39–46, 2011. doi: 10.1111/j.1463-1326.2011.01443.x. [DOI] [PubMed] [Google Scholar]
  • 56.Collombat P, Xu X, Ravassard P, Sosa-Pineda B, Dussaud S, Billestrup N, Madsen OD, Serup P, Heimberg H, Mansouri A. The ectopic expression of Pax4 in the mouse pancreas converts progenitor cells into alpha and subsequently beta cells. Cell 138: 449–462, 2009. doi: 10.1016/j.cell.2009.05.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Conrad E, Stein R, Hunter CS. Revealing transcription factors during human pancreatic β cell development. Trends Endocrinol Metab 25: 407–414, 2014. doi: 10.1016/j.tem.2014.03.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Couri CE, Oliveira MC, Stracieri AB, Moraes DA, Pieroni F, Barros GM, Madeira MI, Malmegrim KC, Foss-Freitas MC, Simões BP, Martinez EZ, Foss MC, Burt RK, Voltarelli JC. C-peptide levels and insulin independence following autologous nonmyeloablative hematopoietic stem cell transplantation in newly diagnosed type 1 diabetes mellitus. JAMA 301: 1573–1579, 2009. doi: 10.1001/jama.2009.470. [DOI] [PubMed] [Google Scholar]
  • 59.Courtney M, Gjernes E, Druelle N, Ravaud C, Vieira A, Ben-Othman N, Pfeifer A, Avolio F, Leuckx G, Lacas-Gervais S, Burel-Vandenbos F, Ambrosetti D, Hecksher-Sorensen J, Ravassard P, Heimberg H, Mansouri A, Collombat P. The inactivation of Arx in pancreatic α-cells triggers their neogenesis and conversion into functional β-like cells. PLoS Genet 9: e1003934, 2013. doi: 10.1371/journal.pgen.1003934. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Cox AR, Barrandon O, Cai EP, Rios JS, Chavez J, Bonnyman CW, Lam CJ, Yi P, Melton DA, Kushner JA. Resolving Discrepant Findings on ANGPTL8 in β-Cell Proliferation: A Collaborative Approach to Resolving the Betatrophin Controversy. PLoS One 11: e0159276, 2016. doi: 10.1371/journal.pone.0159276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Cox AR, Lam CJ, Rankin MM, King KA, Chen P, Martinez R, Li C, Kushner JA. Extreme obesity induces massive beta cell expansion in mice through self-renewal and does not alter the beta cell lineage. Diabetologia 59: 1231–1241, 2016. doi: 10.1007/s00125-016-3922-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.D’Amour KA, Bang AG, Eliazer S, Kelly OG, Agulnick AD, Smart NG, Moorman MA, Kroon E, Carpenter MK, Baetge EE. Production of pancreatic hormone-expressing endocrine cells from human embryonic stem cells. Nat Biotechnol 24: 1392–1401, 2006. doi: 10.1038/nbt1259. [DOI] [PubMed] [Google Scholar]
  • 63.De Franco E, Shaw-Smith C, Flanagan SE, Edghill EL, Wolf J, Otte V, Ebinger F, Varthakavi P, Vasanthi T, Edvardsson S, Hattersley AT, Ellard S. Biallelic PDX1 (insulin promoter factor 1) mutations causing neonatal diabetes without exocrine pancreatic insufficiency. Diabet Med 30: e197–e200, 2013. doi: 10.1111/dme.12122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Demozay D, Tsunekawa S, Briaud I, Shah R, Rhodes CJ. Specific glucose-induced control of insulin receptor substrate-2 expression is mediated via Ca2+-dependent calcineurin/NFAT signaling in primary pancreatic islet β-cells. Diabetes 60: 2892–2902, 2011. doi: 10.2337/db11-0341. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Deutsch G, Jung J, Zheng M, Lóra J, Zaret KS. A bipotential precursor population for pancreas and liver within the embryonic endoderm. Development 128: 871–881, 2001. [DOI] [PubMed] [Google Scholar]
  • 66.Dimitri P, Warner JT, Minton JA, Patch AM, Ellard S, Hattersley AT, Barr S, Hawkes D, Wales JK, Gregory JW. Novel GLIS3 mutations demonstrate an extended multisystem phenotype. Eur J Endocrinol 164: 437–443, 2011. doi: 10.1530/EJE-10-0893. [DOI] [PubMed] [Google Scholar]
  • 67.Dirice E, Walpita D, Vetere A, Meier BC, Kahraman S, Hu J, Dančík V, Burns SM, Gilbert TJ, Olson DE, Clemons PA, Kulkarni RN, Wagner BK. Inhibition of DYRK1A Stimulates Human β-Cell Proliferation. Diabetes 65: 1660–1671, 2016. doi: 10.2337/db15-1127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Dor Y, Brown J, Martinez OI, Melton DA. Adult pancreatic beta-cells are formed by self-duplication rather than stem-cell differentiation. Nature 429: 41–46, 2004. doi: 10.1038/nature02520. [DOI] [PubMed] [Google Scholar]
  • 69.Edghill EL, Bingham C, Slingerland AS, Minton JA, Noordam C, Ellard S, Hattersley AT. Hepatocyte nuclear factor-1 beta mutations cause neonatal diabetes and intrauterine growth retardation: support for a critical role of HNF-1beta in human pancreatic development. Diabet Med 23: 1301–1306, 2006. doi: 10.1111/j.1464-5491.2006.01999.x. [DOI] [PubMed] [Google Scholar]
  • 70.Edsbagge J, Johansson JK, Esni F, Luo Y, Radice GL, Semb H. Vascular function and sphingosine-1-phosphate regulate development of the dorsal pancreatic mesenchyme. Development 132: 1085–1092, 2005. doi: 10.1242/dev.01643. [DOI] [PubMed] [Google Scholar]
  • 71.Efrat S. Ex-vivo Expansion of Adult Human Pancreatic Beta-Cells. Rev Diabet Stud 5: 116–122, 2008. doi: 10.1900/RDS.2008.5.116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Ehrie MG, Swartz FJ. Diploid, tetraploid and octaploid beta cells in the islets of Langerhans of the normal human pancreas. Diabetes 23: 583–588, 1974. doi: 10.2337/diab.23.7.583. [DOI] [PubMed] [Google Scholar]
  • 73.El Ouaamari A, Dirice E, Gedeon N, Hu J, Zhou JY, Shirakawa J, Hou L, Goodman J, Karampelias C, Qiang G, Boucher J, Martinez R, Gritsenko MA, De Jesus DF, Kahraman S, Bhatt S, Smith RD, Beer HD, Jungtrakoon P, Gong Y, Goldfine AB, Liew CW, Doria A, Andersson O, Qian WJ, Remold-O’Donnell E, Kulkarni RN. SerpinB1 Promotes Pancreatic β Cell Proliferation. Cell Metab 23: 194–205, 2016. doi: 10.1016/j.cmet.2015.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Elghazi L, Cras-Méneur C, Czernichow P, Scharfmann R. Role for FGFR2IIIb-mediated signals in controlling pancreatic endocrine progenitor cell proliferation. Proc Natl Acad Sci USA 99: 3884–3889, 2002. doi: 10.1073/pnas.062321799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Esmatjes E, Montaña X, Real MI, Blanco J, Conget I, Casamitjana R, Rovira M, Gomis R, Marin P. Regeneration of insulin production by autologous bone marrow blood autotransplantation in patients with type 1 diabetes. Diabetologia 53: 786–789, 2010. doi: 10.1007/s00125-010-1660-9. [DOI] [PubMed] [Google Scholar]
  • 76.Espes D, Lau J, Carlsson PO. Increased circulating levels of betatrophin in individuals with long-standing type 1 diabetes. Diabetologia 57: 50–53, 2014. doi: 10.1007/s00125-013-3071-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Espes D, Martinell M, Carlsson PO. Increased circulating betatrophin concentrations in patients with type 2 diabetes. Int J Endocrinol 2014: 323407, 2014. doi: 10.1155/2014/323407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Espinosa de los Monteros A, Driscoll SG, Steinke J. Insulin release from isolated human fetal pancreatic islets. Science 168: 1111–1112, 1970. doi: 10.1126/science.168.3935.1111. [DOI] [PubMed] [Google Scholar]
  • 79.Fajans SS, Bell GI, Polonsky KS. Molecular mechanisms and clinical pathophysiology of maturity-onset diabetes of the young. N Engl J Med 345: 971–980, 2001. doi: 10.1056/NEJMra002168. [DOI] [PubMed] [Google Scholar]
  • 81.Flanagan SE, De Franco E, Lango Allen H, Zerah M, Abdul-Rasoul MM, Edge JA, Stewart H, Alamiri E, Hussain K, Wallis S, de Vries L, Rubio-Cabezas O, Houghton JA, Edghill EL, Patch AM, Ellard S, Hattersley AT. Analysis of transcription factors key for mouse pancreatic development establishes NKX2-2 and MNX1 mutations as causes of neonatal diabetes in man. Cell Metab 19: 146–154, 2014. doi: 10.1016/j.cmet.2013.11.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Fomina-Yadlin D, Kubicek S, Vetere A, He KH, Schreiber SL, Wagner BK. GW8510 increases insulin expression in pancreatic alpha cells through activation of p53 transcriptional activity. PLoS One 7: e28808, 2012. doi: 10.1371/journal.pone.0028808. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Forbes JM, Cooper ME. Mechanisms of diabetic complications. Physiol Rev 93: 137–188, 2013. doi: 10.1152/physrev.00045.2011. [DOI] [PubMed] [Google Scholar]
  • 84.Freemark M, Avril I, Fleenor D, Driscoll P, Petro A, Opara E, Kendall W, Oden J, Bridges S, Binart N, Breant B, Kelly PA. Targeted deletion of the PRL receptor: effects on islet development, insulin production, and glucose tolerance. Endocrinology 143: 1378–1385, 2002. doi: 10.1210/endo.143.4.8722. [DOI] [PubMed] [Google Scholar]
  • 85.Fujitani Y, Fujitani S, Boyer DF, Gannon M, Kawaguchi Y, Ray M, Shiota M, Stein RW, Magnuson MA, Wright CVE. Targeted deletion of a cis-regulatory region reveals differential gene dosage requirements for Pdx1 in foregut organ differentiation and pancreas formation. Genes Dev 20: 253–266, 2006. doi: 10.1101/gad.1360106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Furuyama K, Kawaguchi Y, Akiyama H, Horiguchi M, Kodama S, Kuhara T, Hosokawa S, Elbahrawy A, Soeda T, Koizumi M, Masui T, Kawaguchi M, Takaori K, Doi R, Nishi E, Kakinoki R, Deng JM, Behringer RR, Nakamura T, Uemoto S. Continuous cell supply from a Sox9-expressing progenitor zone in adult liver, exocrine pancreas and intestine. Nat Genet 43: 34–41, 2011. doi: 10.1038/ng.722. [DOI] [PubMed] [Google Scholar]
  • 87.Gallo R, Gambelli F, Gava B, Sasdelli F, Tellone V, Masini M, Marchetti P, Dotta F, Sorrentino V. Generation and expansion of multipotent mesenchymal progenitor cells from cultured human pancreatic islets. Cell Death Differ 14: 1860–1871, 2007. doi: 10.1038/sj.cdd.4402199. [DOI] [PubMed] [Google Scholar]
  • 88.Gao R, Ustinov J, Korsgren O, Otonkoski T. In vitro neogenesis of human islets reflects the plasticity of differentiated human pancreatic cells. Diabetologia 48: 2296–2304, 2005. doi: 10.1007/s00125-005-1935-8. [DOI] [PubMed] [Google Scholar]
  • 89.Gao R, Ustinov J, Pulkkinen MA, Lundin K, Korsgren O, Otonkoski T. Characterization of endocrine progenitor cells and critical factors for their differentiation in human adult pancreatic cell culture. Diabetes 52: 2007–2015, 2003. doi: 10.2337/diabetes.52.8.2007. [DOI] [PubMed] [Google Scholar]
  • 90.Georgia S, Bhushan A. Beta cell replication is the primary mechanism for maintaining postnatal beta cell mass. J Clin Invest 114: 963–968, 2004. doi: 10.1172/JCI22098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.German MS. Anonymous sources: where do adult β cells come from? J Clin Invest 123: 1936–1938, 2013. doi: 10.1172/JCI69297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Gershengorn MC, Hardikar AA, Wei C, Geras-Raaka E, Marcus-Samuels B, Raaka BM. Epithelial-to-mesenchymal transition generates proliferative human islet precursor cells. Science 306: 2261–2264, 2004. doi: 10.1126/science.1101968. [DOI] [PubMed] [Google Scholar]
  • 93.Goyvaerts L, Lemaire K, Arijs I, Auffret J, Granvik M, Van Lommel L, Binart N, in’t Veld P, Schuit F, Schraenen A. Prolactin receptors and placental lactogen drive male mouse pancreatic islets to pregnancy-related mRNA changes. PLoS One 10: e0121868, 2015. doi: 10.1371/journal.pone.0121868. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Gradwohl G, Dierich A, LeMeur M, Guillemot F. Neurogenin3 is required for the development of the four endocrine cell lineages of the pancreas. Proc Natl Acad Sci USA 97: 1607–1611, 2000. doi: 10.1073/pnas.97.4.1607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Gregg BE, Moore PC, Demozay D, Hall BA, Li M, Husain A, Wright AJ, Atkinson MA, Rhodes CJ. Formation of a human β-cell population within pancreatic islets is set early in life. J Clin Endocrinol Metab 97: 3197–3206, 2012. doi: 10.1210/jc.2012-1206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Guan B, Li W, Li F, Xie Y, Ni Q, Gu Y, Li X, Wang Q, Zhang H, Ning G. Sfrp5 mediates glucose-induced proliferation in rat pancreatic β-cells. J Endocrinol 229: 73–83, 2016. doi: 10.1530/JOE-15-0535. [DOI] [PubMed] [Google Scholar]
  • 97.Gupta RK, Gao N, Gorski RK, White P, Hardy OT, Rafiq K, Brestelli JE, Chen G, Stoeckert CJ Jr, Kaestner KH. Expansion of adult beta-cell mass in response to increased metabolic demand is dependent on HNF-4alpha. Genes Dev 21: 756–769, 2007. doi: 10.1101/gad.1535507. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Gusarova V, Alexa CA, Na E, Stevis PE, Xin Y, Bonner-Weir S, Cohen JC, Hobbs HH, Murphy AJ, Yancopoulos GD, Gromada J. ANGPTL8/betatrophin does not control pancreatic beta cell expansion. Cell 159: 691–696, 2014. doi: 10.1016/j.cell.2014.09.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Haig D. Placental growth hormone-related proteins and prolactin-related proteins. Placenta 29, Suppl A: S36–S41, 2008. doi: 10.1016/j.placenta.2007.09.010. [DOI] [PubMed] [Google Scholar]
  • 100.Haller MJ, Wasserfall CH, Hulme MA, Cintron M, Brusko TM, McGrail KM, Sumrall TM, Wingard JR, Theriaque DW, Shuster JJ, Atkinson MA, Schatz DA. Autologous umbilical cord blood transfusion in young children with type 1 diabetes fails to preserve C-peptide. Diabetes Care 34: 2567–2569, 2011. doi: 10.2337/dc11-1406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Hanley SC, Austin E, Assouline-Thomas B, Kapeluto J, Blaichman J, Moosavi M, Petropavlovskaia M, Rosenberg L. beta-Cell mass dynamics and islet cell plasticity in human type 2 diabetes. Endocrinology 151: 1462–1472, 2010. doi: 10.1210/en.2009-1277. [DOI] [PubMed] [Google Scholar]
  • 102.Hao E, Tyrberg B, Itkin-Ansari P, Lakey JR, Geron I, Monosov EZ, Barcova M, Mercola M, Levine F. Beta-cell differentiation from nonendocrine epithelial cells of the adult human pancreas. Nat Med 12: 310–316, 2006. doi: 10.1038/nm1367. [DOI] [PubMed] [Google Scholar]
  • 103.Hay WW., Jr Energy and substrate requirements of the placenta and fetus. Proc Nutr Soc 50: 321–336, 1991. doi: 10.1079/PNS19910042. [DOI] [PubMed] [Google Scholar]
  • 104.Hebrok M, Kim SK, Melton DA. Notochord repression of endodermal Sonic hedgehog permits pancreas development. Genes Dev 12: 1705–1713, 1998. doi: 10.1101/gad.12.11.1705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Heimberg H, Bouwens L, Heremans Y, Van De Casteele M, Lefebvre V, Pipeleers D. Adult human pancreatic duct and islet cells exhibit similarities in expression and differences in phosphorylation and complex formation of the homeodomain protein Ipf-1. Diabetes 49: 571–579, 2000. doi: 10.2337/diabetes.49.4.571. [DOI] [PubMed] [Google Scholar]
  • 106.Heremans Y, Van De Casteele M, in’t Veld P, Gradwohl G, Serup P, Madsen O, Pipeleers D, Heimberg H. Recapitulation of embryonic neuroendocrine differentiation in adult human pancreatic duct cells expressing neurogenin 3. J Cell Biol 159: 303–312, 2002. doi: 10.1083/jcb.200203074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Hering BJ, Clarke WR, Bridges ND, Eggerman TL, Alejandro R, Bellin MD, Chaloner K, Czarniecki CW, Goldstein JS, Hunsicker LG, Kaufman DB, Korsgren O, Larsen CP, Luo X, Markmann JF, Naji A, Oberholzer J, Posselt AM, Rickels MR, Ricordi C, Robien MA, Senior PA, Shapiro AMJ, Stock PG, Turgeon NA; Clinical Islet Transplantation Consortium . Phase 3 Trial of Transplantation of Human Islets in Type 1 Diabetes Complicated by Severe Hypoglycemia. Diabetes Care 39: 1230–1240, 2016. doi: 10.2337/dc15-1988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Herman MA, Kahn BB. Glucose transport and sensing in the maintenance of glucose homeostasis and metabolic harmony. J Clin Invest 116: 1767–1775, 2006. doi: 10.1172/JCI29027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Herold KC, Vignali DA, Cooke A, Bluestone JA. Type 1 diabetes: translating mechanistic observations into effective clinical outcomes. Nat Rev Immunol 13: 243–256, 2013. doi: 10.1038/nri3422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Hess D, Li L, Martin M, Sakano S, Hill D, Strutt B, Thyssen S, Gray DA, Bhatia M. Bone marrow-derived stem cells initiate pancreatic regeneration. Nat Biotechnol 21: 763–770, 2003. doi: 10.1038/nbt841. [DOI] [PubMed] [Google Scholar]
  • 111.Höglund E, Mattsson G, Tyrberg B, Andersson A, Carlsson C. Growth hormone increases beta-cell proliferation in transplanted human and fetal rat islets. JOP 10: 242–248, 2009. [PubMed] [Google Scholar]
  • 112.Hrvatin S, O’Donnell CW, Deng F, Millman JR, Pagliuca FW, DiIorio P, Rezania A, Gifford DK, Melton DA. Differentiated human stem cells resemble fetal, not adult, β cells. Proc Natl Acad Sci USA 111: 3038–3043, 2014. doi: 10.1073/pnas.1400709111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Huang C, Snider F, Cross JC. Prolactin receptor is required for normal glucose homeostasis and modulation of beta-cell mass during pregnancy. Endocrinology 150: 1618–1626, 2009. doi: 10.1210/en.2008-1003. [DOI] [PubMed] [Google Scholar]
  • 114.Hughes E, Huang C. Participation of Akt, menin, and p21 in pregnancy-induced beta-cell proliferation. Endocrinology 152: 847–855, 2011. doi: 10.1210/en.2010-1250. [DOI] [PubMed] [Google Scholar]
  • 115.In’t Veld P, De Munck N, Van Belle K, Buelens N, Ling Z, Weets I, Haentjens P, Pipeleers-Marichal M, Gorus F, Pipeleers D. Beta-cell replication is increased in donor organs from young patients after prolonged life support. Diabetes 59: 1702–1708, 2010. doi: 10.2337/db09-1698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Inaishi J, Saisho Y, Sato S, Kou K, Murakami R, Watanabe Y, Kitago M, Kitagawa Y, Yamada T, Itoh H. Effects of Obesity and Diabetes on α- and β-Cell Mass in Surgically Resected Human Pancreas. J Clin Endocrinol Metab 101: 2874–2882, 2016. doi: 10.1210/jc.2016-1374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116a.International Diabetes Federation. Diabetes Atlas. https://www.idf.org/diabetesatlas. [October, 10, 2016].
  • 117.Jacovetti C, Abderrahmani A, Parnaud G, Jonas JC, Peyot ML, Cornu M, Laybutt R, Meugnier E, Rome S, Thorens B, Prentki M, Bosco D, Regazzi R. MicroRNAs contribute to compensatory β cell expansion during pregnancy and obesity. J Clin Invest 122: 3541–3551, 2012. doi: 10.1172/JCI64151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Jennings RE, Berry AA, Kirkwood-Wilson R, Roberts NA, Hearn T, Salisbury RJ, Blaylock J, Piper Hanley K, Hanley NA. Development of the human pancreas from foregut to endocrine commitment. Diabetes 62: 3514–3522, 2013. doi: 10.2337/db12-1479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Jennings RE, Berry AA, Strutt JP, Gerrard DT, Hanley NA. Human pancreas development. Development 142: 3126–3137, 2015. doi: 10.1242/dev.120063. [DOI] [PubMed] [Google Scholar]
  • 120.Jeon J, Correa-Medina M, Ricordi C, Edlund H, Diez JA. Endocrine cell clustering during human pancreas development. J Histochem Cytochem 57: 811–824, 2009. doi: 10.1369/jhc.2009.953307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Jiang J, Au M, Lu K, Eshpeter A, Korbutt G, Fisk G, Majumdar AS. Generation of insulin-producing islet-like clusters from human embryonic stem cells. Stem Cells 25: 1940–1953, 2007. doi: 10.1634/stemcells.2006-0761. [DOI] [PubMed] [Google Scholar]
  • 122.Jiao Y, Le Lay J, Yu M, Naji A, Kaestner KH. Elevated mouse hepatic betatrophin expression does not increase human β-cell replication in the transplant setting. Diabetes 63: 1283–1288, 2014. doi: 10.2337/db13-1435. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Jonas JC, Sharma A, Hasenkamp W, Ilkova H, Patanè G, Laybutt R, Bonner-Weir S, Weir GC. Chronic hyperglycemia triggers loss of pancreatic beta cell differentiation in an animal model of diabetes. J Biol Chem 274: 14112–14121, 1999. doi: 10.1074/jbc.274.20.14112. [DOI] [PubMed] [Google Scholar]
  • 124.Jonsson J, Carlsson L, Edlund T, Edlund H. Insulin-promoter-factor 1 is required for pancreas development in mice. Nature 371: 606–609, 1994. doi: 10.1038/371606a0. [DOI] [PubMed] [Google Scholar]
  • 125.Jopling C, Boue S, Izpisua Belmonte JC. Dedifferentiation, transdifferentiation and reprogramming: three routes to regeneration. Nat Rev Mol Cell Biol 12: 79–89, 2011. doi: 10.1038/nrm3043. [DOI] [PubMed] [Google Scholar]
  • 126.Jørgensen MC, Ahnfelt-Rønne J, Hald J, Madsen OD, Serup P, Hecksher-Sørensen J. An illustrated review of early pancreas development in the mouse. Endocr Rev 28: 685–705, 2007. doi: 10.1210/er.2007-0016. [DOI] [PubMed] [Google Scholar]
  • 127.Karnieli O, Izhar-Prato Y, Bulvik S, Efrat S. Generation of insulin-producing cells from human bone marrow mesenchymal stem cells by genetic manipulation. Stem Cells 25: 2837–2844, 2007. doi: 10.1634/stemcells.2007-0164. [DOI] [PubMed] [Google Scholar]
  • 128.Karnik SK, Chen H, McLean GW, Heit JJ, Gu X, Zhang AY, Fontaine M, Yen MH, Kim SK. Menin controls growth of pancreatic beta-cells in pregnant mice and promotes gestational diabetes mellitus. Science 318: 806–809, 2007. doi: 10.1126/science.1146812. [DOI] [PubMed] [Google Scholar]
  • 129.Karunakaran U, Kim HJ, Kim JY, Lee IK. Guards and culprits in the endoplasmic reticulum: glucolipotoxicity and β-cell failure in type II diabetes. Exp Diabetes Res 2012: 639762, 2012. doi: 10.1155/2012/639762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Kassem S, Bhandari S, Rodríguez-Bada P, Motaghedi R, Heyman M, García-Gimeno MA, Cobo-Vuilleumier N, Sanz P, Maclaren NK, Rahier J, Glaser B, Cuesta-Muñoz AL. Large islets, beta-cell proliferation, and a glucokinase mutation. N Engl J Med 362: 1348–1350, 2010. doi: 10.1056/NEJMc0909845. [DOI] [PubMed] [Google Scholar]
  • 131.Kassem SA, Ariel I, Thornton PS, Scheimberg I, Glaser B. Beta-cell proliferation and apoptosis in the developing normal human pancreas and in hyperinsulinism of infancy. Diabetes 49: 1325–1333, 2000. doi: 10.2337/diabetes.49.8.1325. [DOI] [PubMed] [Google Scholar]
  • 132.Kayali AG, Flores LE, Lopez AD, Kutlu B, Baetge E, Kitamura R, Hao E, Beattie GM, Hayek A. Limited capacity of human adult islets expanded in vitro to redifferentiate into insulin-producing beta-cells. Diabetes 56: 703–708, 2007. doi: 10.2337/db06-1545. [DOI] [PubMed] [Google Scholar]
  • 133.Kim H, Toyofuku Y, Lynn FC, Chak E, Uchida T, Mizukami H, Fujitani Y, Kawamori R, Miyatsuka T, Kosaka Y, Yang K, Honig G, van der Hart M, Kishimoto N, Wang J, Yagihashi S, Tecott LH, Watada H, German MS. Serotonin regulates pancreatic beta cell mass during pregnancy. Nat Med 16: 804–808, 2010. doi: 10.1038/nm.2173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Kim SC, Han DJ, Lee JY. Adipose tissue derived stem cells for regeneration and differentiation into insulin-producing cells. Curr Stem Cell Res Ther 5: 190–194, 2010. doi: 10.2174/157488810791268717. [DOI] [PubMed] [Google Scholar]
  • 135.Kim SK, Hebrok M, Li E, Oh SP, Schrewe H, Harmon EB, Lee JS, Melton DA. Activin receptor patterning of foregut organogenesis. Genes Dev 14: 1866–1871, 2000. [PMC free article] [PubMed] [Google Scholar]
  • 136.Kim SK, Hebrok M, Melton DA. Notochord to endoderm signaling is required for pancreas development. Development 124: 4243–4252, 1997. [DOI] [PubMed] [Google Scholar]
  • 137.Kitamura T, Nakae J, Kitamura Y, Kido Y, Biggs WH III, Wright CV, White MF, Arden KC, Accili D. The forkhead transcription factor Foxo1 links insulin signaling to Pdx1 regulation of pancreatic beta cell growth. J Clin Invest 110: 1839–1847, 2002. doi: 10.1172/JCI200216857. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Kjos SL, Buchanan TA. Gestational diabetes mellitus. N Engl J Med 341: 1749–1756, 1999. doi: 10.1056/NEJM199912023412307. [DOI] [PubMed] [Google Scholar]
  • 139.Kondegowda NG, Fenutria R, Pollack IR, Orthofer M, Garcia-Ocaña A, Penninger JM, Vasavada RC. Osteoprotegerin and Denosumab Stimulate Human Beta Cell Proliferation through Inhibition of the Receptor Activator of NF-κB Ligand Pathway. Cell Metab 22: 77–85, 2015. doi: 10.1016/j.cmet.2015.05.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Kopinke D, Brailsford M, Shea JE, Leavitt R, Scaife CL, Murtaugh LC. Lineage tracing reveals the dynamic contribution of Hes1+ cells to the developing and adult pancreas. Development 138: 431–441, 2011. doi: 10.1242/dev.053843. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Kopp JL, Dubois CL, Schaffer AE, Hao E, Shih HP, Seymour PA, Ma J, Sander M. Sox9+ ductal cells are multipotent progenitors throughout development but do not produce new endocrine cells in the normal or injured adult pancreas. Development 138: 653–665, 2011. doi: 10.1242/dev.056499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Kozinski K, Jazurek M, Dobrzyn P, Janikiewicz J, Kolczynska K, Gajda A, Dobrzyn A. Adipose- and muscle-derived Wnts trigger pancreatic β-cell adaptation to systemic insulin resistance. Sci Rep 6: 31553, 2016. doi: 10.1038/srep31553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Kroon E, Martinson LA, Kadoya K, Bang AG, Kelly OG, Eliazer S, Young H, Richardson M, Smart NG, Cunningham J, Agulnick AD, D’Amour KA, Carpenter MK, Baetge EE. Pancreatic endoderm derived from human embryonic stem cells generates glucose-responsive insulin-secreting cells in vivo. Nat Biotechnol 26: 443–452, 2008. doi: 10.1038/nbt1393. [DOI] [PubMed] [Google Scholar]
  • 144.Lain KY, Catalano PM. Metabolic changes in pregnancy. Clin Obstet Gynecol 50: 938–948, 2007. doi: 10.1097/GRF.0b013e31815a5494. [DOI] [PubMed] [Google Scholar]
  • 145.Lakshmipathi J, Alvarez-Perez JC, Rosselot C, Casinelli GP, Stamateris RE, Rausell-Palamos F, O’Donnell CP, Vasavada RC, Scott DK, Alonso LC, Garcia-Ocaña A. PKCζ Is Essential for Pancreatic β-Cell Replication During Insulin Resistance by Regulating mTOR and Cyclin-D2. Diabetes 65: 1283–1296, 2016. doi: 10.2337/db15-1398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Lammert E, Cleaver O, Melton D. Induction of pancreatic differentiation by signals from blood vessels. Science 294: 564–567, 2001. doi: 10.1126/science.1064344. [DOI] [PubMed] [Google Scholar]
  • 147.Lango Allen H, Flanagan SE, Shaw-Smith C, De Franco E, Akerman I, Caswell R, Ferrer J, Hattersley AT, Ellard S; International Pancreatic Agenesis Consortium . GATA6 haploinsufficiency causes pancreatic agenesis in humans. Nat Genet 44: 20–22, 2012. doi: 10.1038/ng.1035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Layden BT, Durai V, Newman MV, Marinelarena AM, Ahn CW, Feng G, Lin S, Zhang X, Kaufman DB, Jafari N, Sørensen GL, Lowe WL Jr. Regulation of pancreatic islet gene expression in mouse islets by pregnancy. J Endocrinol 207: 265–279, 2010. doi: 10.1677/JOE-10-0298. [DOI] [PubMed] [Google Scholar]
  • 149.Lechner A, Nolan AL, Blacken RA, Habener JF. Redifferentiation of insulin-secreting cells after in vitro expansion of adult human pancreatic islet tissue. Biochem Biophys Res Commun 327: 581–588, 2005. doi: 10.1016/j.bbrc.2004.12.043. [DOI] [PubMed] [Google Scholar]
  • 150.Lechner A, Yang YG, Blacken RA, Wang L, Nolan AL, Habener JF. No evidence for significant transdifferentiation of bone marrow into pancreatic beta-cells in vivo. Diabetes 53: 616–623, 2004. doi: 10.2337/diabetes.53.3.616. [DOI] [PubMed] [Google Scholar]
  • 151.Lee J, Kim SC, Kim SJ, Lee H, Jung EJ, Jung SH, Han DJ. Differentiation of human adipose tissue-derived stem cells into aggregates of insulin-producing cells through the overexpression of pancreatic and duodenal homeobox gene-1. Cell Transplant 22: 1053–1060, 2013. doi: 10.3727/096368912X657215. [DOI] [PubMed] [Google Scholar]
  • 152.Lemper M, Leuckx G, Heremans Y, German MS, Heimberg H, Bouwens L, Baeyens L. Reprogramming of human pancreatic exocrine cells to β-like cells. Cell Death Differ 22: 1117–1130, 2015. doi: 10.1038/cdd.2014.193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Lenz A, Toren-Haritan G, Efrat S. Redifferentiation of adult human β cells expanded in vitro by inhibition of the WNT pathway. PLoS One 9: e112914, 2014. doi: 10.1371/journal.pone.0112914. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Levitt HE, Cyphert TJ, Pascoe JL, Hollern DA, Abraham N, Lundell RJ, Rosa T, Romano LC, Zou B, O’Donnell CP, Stewart AF, Garcia-Ocaña A, Alonso LC. Glucose stimulates human beta cell replication in vivo in islets transplanted into NOD-severe combined immunodeficiency (SCID) mice. Diabetologia 54: 572–582, 2011. doi: 10.1007/s00125-010-1919-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Li J, Casteels T, Frogne T, Ingvorsen C, Honoré C, Courtney M, Huber KVM, Schmitner N, Kimmel RA, Romanov RA, Sturtzel C, Lardeau CH, Klughammer J, Farlik M, Sdelci S, Vieira A, Avolio F, Briand F, Baburin I, Májek P, Pauler FM, Penz T, Stukalov A, Gridling M, Parapatics K, Barbieux C, Berishvili E, Spittler A, Colinge J, Bennett KL, Hering S, Sulpice T, Bock C, Distel M, Harkany T, Meyer D, Superti-Furga G, Collombat P, Hecksher-Sørensen J, Kubicek S. Artemisinins Target GABAA Receptor Signaling and Impair α Cell Identity. Cell 168: 86–100.e15, 2017. doi: 10.1016/j.cell.2016.11.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Lima MJ, Muir KR, Docherty HM, Drummond R, McGowan NW, Forbes S, Heremans Y, Houbracken I, Ross JA, Forbes SJ, Ravassard P, Heimberg H, Casey J, Docherty K. Suppression of epithelial-to-mesenchymal transitioning enhances ex vivo reprogramming of human exocrine pancreatic tissue toward functional insulin-producing β-like cells. Diabetes 62: 2821–2833, 2013. doi: 10.2337/db12-1256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Lima MJ, Muir KR, Docherty HM, McGowan NWA, Forbes S, Heremans Y, Heimberg H, Casey J, Docherty K. Generation of Functional Beta-Like Cells from Human Exocrine Pancreas. PLoS One 11: e0156204, 2016. doi: 10.1371/journal.pone.0156204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Lingohr MK, Dickson LM, McCuaig JF, Hugl SR, Twardzik DR, Rhodes CJ. Activation of IRS-2-mediated signal transduction by IGF-1, but not TGF-alpha or EGF, augments pancreatic beta-cell proliferation. Diabetes 51: 966–976, 2002. doi: 10.2337/diabetes.51.4.966. [DOI] [PubMed] [Google Scholar]
  • 159.Lynn FC, Smith SB, Wilson ME, Yang KY, Nekrep N, German MS. Sox9 coordinates a transcriptional network in pancreatic progenitor cells. Proc Natl Acad Sci USA 104: 10500–10505, 2007. doi: 10.1073/pnas.0704054104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Lyttle BM, Li J, Krishnamurthy M, Fellows F, Wheeler MB, Goodyer CG, Wang R. Transcription factor expression in the developing human fetal endocrine pancreas. Diabetologia 51: 1169–1180, 2008. doi: 10.1007/s00125-008-1006-z. [DOI] [PubMed] [Google Scholar]
  • 161.Maedler K, Schumann DM, Schulthess F, Oberholzer J, Bosco D, Berney T, Donath MY. Aging correlates with decreased beta-cell proliferative capacity and enhanced sensitivity to apoptosis: a potential role for Fas and pancreatic duodenal homeobox-1. Diabetes 55: 2455–2462, 2006. doi: 10.2337/db05-1586. [DOI] [PubMed] [Google Scholar]
  • 162.Maedler K, Sergeev P, Ris F, Oberholzer J, Joller-Jemelka HI, Spinas GA, Kaiser N, Halban PA, Donath MY. Glucose-induced beta cell production of IL-1beta contributes to glucotoxicity in human pancreatic islets. J Clin Invest 110: 851–860, 2002. doi: 10.1172/JCI200215318. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Maehr R, Chen S, Snitow M, Ludwig T, Yagasaki L, Goland R, Leibel RL, Melton DA. Generation of pluripotent stem cells from patients with type 1 diabetes. Proc Natl Acad Sci USA 106: 15768–15773, 2009. doi: 10.1073/pnas.0906894106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Maschio DA, Oliveira RB, Santos MR, Carvalho CP, Barbosa-Sampaio HC, Collares-Buzato CB. Activation of the Wnt/β-catenin pathway in pancreatic beta cells during the compensatory islet hyperplasia in prediabetic mice. Biochem Biophys Res Commun 478: 1534–1540, 2016. doi: 10.1016/j.bbrc.2016.08.146. [DOI] [PubMed] [Google Scholar]
  • 165.Masharani U, German MS. Pancreatic hormones and Diabetes Mellitus. In: Greenspan’s Basic and Clinical Endocrinology (10th ed.), edited by Gardner DG, Shoback D. New York: McGraw-Hill Education, 2017, p. 595–682. [Google Scholar]
  • 166.Mauda-Havakuk M, Litichever N, Chernichovski E, Nakar O, Winkler E, Mazkereth R, Orenstein A, Bar-Meir E, Ravassard P, Meivar-Levy I, Ferber S. Ectopic PDX-1 expression directly reprograms human keratinocytes along pancreatic insulin-producing cells fate. PLoS One 6: e26298, 2011. doi: 10.1371/journal.pone.0026298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Meier JJ, Bhushan A, Butler AE, Rizza RA, Butler PC. Sustained beta cell apoptosis in patients with long-standing type 1 diabetes: indirect evidence for islet regeneration? Diabetologia 48: 2221–2228, 2005. doi: 10.1007/s00125-005-1949-2. [DOI] [PubMed] [Google Scholar]
  • 168.Meier JJ, Butler AE, Saisho Y, Monchamp T, Galasso R, Bhushan A, Rizza RA, Butler PC. Beta-cell replication is the primary mechanism subserving the postnatal expansion of beta-cell mass in humans. Diabetes 57: 1584–1594, 2008. doi: 10.2337/db07-1369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Meier JJ, Köhler CU, Alkhatib B, Sergi C, Junker T, Klein HH, Schmidt WE, Fritsch H. Beta-cell development and turnover during prenatal life in humans. Eur J Endocrinol 162: 559–568, 2010. doi: 10.1530/EJE-09-1053. [DOI] [PubMed] [Google Scholar]
  • 170.Meivar-Levy I, Sapir T, Berneman D, Weissbach T, Polak-Charcon S, Ravassard P, Tzakis AG, Mor E, Ricordi C, Ferber S. Human liver cells expressing albumin and mesenchymal characteristics give rise to insulin-producing cells. J Transplant 2011: 252387, 2011. doi: 10.1155/2011/252387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Meivar-Levy I, Sapir T, Gefen-Halevi S, Aviv V, Barshack I, Onaca N, Mor E, Ferber S. Pancreatic and duodenal homeobox gene 1 induces hepatic dedifferentiation by suppressing the expression of CCAAT/enhancer-binding protein beta. Hepatology 46: 898–905, 2007. doi: 10.1002/hep.21766. [DOI] [PubMed] [Google Scholar]
  • 172.Melkman-Zehavi T, Oren R, Kredo-Russo S, Shapira T, Mandelbaum AD, Rivkin N, Nir T, Lennox KA, Behlke MA, Dor Y, Hornstein E. miRNAs control insulin content in pancreatic β-cells via downregulation of transcriptional repressors. EMBO J 30: 835–845, 2011. doi: 10.1038/emboj.2010.361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Mellado-Gil J, Rosa TC, Demirci C, Gonzalez-Pertusa JA, Velazquez-Garcia S, Ernst S, Valle S, Vasavada RC, Stewart AF, Alonso LC, Garcia-Ocaña A. Disruption of hepatocyte growth factor/c-Met signaling enhances pancreatic beta-cell death and accelerates the onset of diabetes. Diabetes 60: 525–536, 2011. doi: 10.2337/db09-1305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Metukuri MR, Zhang P, Basantani MK, Chin C, Stamateris RE, Alonso LC, Takane KK, Gramignoli R, Strom SC, O’Doherty RM, Stewart AF, Vasavada RC, Garcia-Ocaña A, Scott DK. ChREBP mediates glucose-stimulated pancreatic β-cell proliferation. Diabetes 61: 2004–2015, 2012. doi: 10.2337/db11-0802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Millman JR, Xie C, Van Dervort A, Gürtler M, Pagliuca FW, Melton DA. Generation of stem cell-derived β-cells from patients with type 1 diabetes. Nat Commun 7: 11463, 2016. doi: 10.1038/ncomms11463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Mohamad Buang ML, Seng HK, Chung LH, Saim AB, Idrus RB. In vitro generation of functional insulin-producing cells from lipoaspirated human adipose tissue-derived stem cells. Arch Med Res 43: 83–88, 2012. doi: 10.1016/j.arcmed.2012.01.012. [DOI] [PubMed] [Google Scholar]
  • 177.Moran A, Jacobs DR Jr, Steinberger J, Hong CP, Prineas R, Luepker R, Sinaiko AR. Insulin resistance during puberty: results from clamp studies in 357 children. Diabetes 48: 2039–2044, 1999. doi: 10.2337/diabetes.48.10.2039. [DOI] [PubMed] [Google Scholar]
  • 178.Morton RA, Geras-Raaka E, Wilson LM, Raaka BM, Gershengorn MC. Endocrine precursor cells from mouse islets are not generated by epithelial-to-mesenchymal transition of mature beta cells. Mol Cell Endocrinol 270: 87–93, 2007. doi: 10.1016/j.mce.2007.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Muir KR, Lima MJ, Docherty HM, Docherty K. Cell therapy for type 1 diabetes. QJM 107: 253–259, 2014. doi: 10.1093/qjmed/hcu025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Muniappan L, Ozcan S. Induction of insulin secretion in engineered liver cells by nitric oxide. BMC Physiol 7: 11, 2007. doi: 10.1186/1472-6793-7-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Nakajima-Nagata N, Sakurai T, Mitaka T, Katakai T, Yamato E, Miyazaki J, Tabata Y, Sugai M, Shimizu A. In vitro induction of adult hepatic progenitor cells into insulin-producing cells. Biochem Biophys Res Commun 318: 625–630, 2004. doi: 10.1016/j.bbrc.2004.04.059. [DOI] [PubMed] [Google Scholar]
  • 182.Nesca V, Guay C, Jacovetti C, Menoud V, Peyot M-L, Laybutt DR, Prentki M, Regazzi R. Identification of particular groups of microRNAs that positively or negatively impact on beta cell function in obese models of type 2 diabetes. Diabetologia 56: 2203–2212, 2013. doi: 10.1007/s00125-013-2993-y. [DOI] [PubMed] [Google Scholar]
  • 183.Nolan CJ, Damm P, Prentki M. Type 2 diabetes across generations: from pathophysiology to prevention and management. Lancet 378: 169–181, 2011. doi: 10.1016/S0140-6736(11)60614-4. [DOI] [PubMed] [Google Scholar]
  • 184.Nolan CJ, Proietto J. The feto-placental glucose steal phenomenon is a major cause of maternal metabolic adaptation during late pregnancy in the rat. Diabetologia 37: 976–984, 1994. doi: 10.1007/BF00400460. [DOI] [PubMed] [Google Scholar]
  • 185.O’Rahilly R, Müller F. Developmental stages in human embryos: revised and new measurements. Cells Tissues Organs 192: 73–84, 2010. doi: 10.1159/000289817. [DOI] [PubMed] [Google Scholar]
  • 186.Offield MF, Jetton TL, Labosky PA, Ray M, Stein RW, Magnuson MA, Hogan BL, Wright CV. PDX-1 is required for pancreatic outgrowth and differentiation of the rostral duodenum. Development 122: 983–995, 1996. [DOI] [PubMed] [Google Scholar]
  • 187.Ohara-Imaizumi M, Kim H, Yoshida M, Fujiwara T, Aoyagi K, Toyofuku Y, Nakamichi Y, Nishiwaki C, Okamura T, Uchida T, Fujitani Y, Akagawa K, Kakei M, Watada H, German MS, Nagamatsu S. Serotonin regulates glucose-stimulated insulin secretion from pancreatic β cells during pregnancy. Proc Natl Acad Sci USA 110: 19420–19425, 2013. doi: 10.1073/pnas.1310953110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Okura H, Komoda H, Fumimoto Y, Lee CM, Nishida T, Sawa Y, Matsuyama A. Transdifferentiation of human adipose tissue-derived stromal cells into insulin-producing clusters. J Artif Organs 12: 123–130, 2009. doi: 10.1007/s10047-009-0455-6. [DOI] [PubMed] [Google Scholar]
  • 189.Ouziel-Yahalom L, Zalzman M, Anker-Kitai L, Knoller S, Bar Y, Glandt M, Herold K, Efrat S. Expansion and redifferentiation of adult human pancreatic islet cells. Biochem Biophys Res Commun 341: 291–298, 2006. doi: 10.1016/j.bbrc.2005.12.187. [DOI] [PubMed] [Google Scholar]
  • 190.Pagliuca FW, Millman JR, Gürtler M, Segel M, Van Dervort A, Ryu JH, Peterson QP, Greiner D, Melton DA. Generation of functional human pancreatic β cells in vitro. Cell 159: 428–439, 2014. doi: 10.1016/j.cell.2014.09.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Pan FC, Brissova M. Pancreas development in humans. Curr Opin Endocrinol Diabetes Obes 21: 77–82, 2014. doi: 10.1097/MED.0000000000000047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Pan FC, Wright C. Pancreas organogenesis: from bud to plexus to gland. Dev Dyn 240: 530–565, 2011. doi: 10.1002/dvdy.22584. [DOI] [PubMed] [Google Scholar]
  • 193.Parnaud G, Bosco D, Berney T, Pattou F, Kerr-Conte J, Donath MY, Bruun C, Mandrup-Poulsen T, Billestrup N, Halban PA. Proliferation of sorted human and rat beta cells. Diabetologia 51: 91–100, 2008. doi: 10.1007/s00125-007-0855-1. [DOI] [PubMed] [Google Scholar]
  • 194.Perl S, Kushner JA, Buchholz BA, Meeker AK, Stein GM, Hsieh M, Kirby M, Pechhold S, Liu EH, Harlan DM, Tisdale JF. Significant human beta-cell turnover is limited to the first three decades of life as determined by in vivo thymidine analog incorporation and radiocarbon dating. J Clin Endocrinol Metab 95: E234–E239, 2010. doi: 10.1210/jc.2010-0932. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Phadnis SM, Joglekar MV, Dalvi MP, Muthyala S, Nair PD, Ghaskadbi SM, Bhonde RR, Hardikar AA. Human bone marrow-derived mesenchymal cells differentiate and mature into endocrine pancreatic lineage in vivo. Cytotherapy 13: 279–293, 2011. doi: 10.3109/14653249.2010.523108. [DOI] [PubMed] [Google Scholar]
  • 196.Phillips BW, Hentze H, Rust WL, Chen QP, Chipperfield H, Tan EK, Abraham S, Sadasivam A, Soong PL, Wang ST, Lim R, Sun W, Colman A, Dunn NR. Directed differentiation of human embryonic stem cells into the pancreatic endocrine lineage. Stem Cells Dev 16: 561–578, 2007. doi: 10.1089/scd.2007.0029. [DOI] [PubMed] [Google Scholar]
  • 197.Piper K, Ball SG, Keeling JW, Mansoor S, Wilson DI, Hanley NA. Novel SOX9 expression during human pancreas development correlates to abnormalities in Campomelic dysplasia. Mech Dev 116: 223–226, 2002. doi: 10.1016/S0925-4773(02)00145-4. [DOI] [PubMed] [Google Scholar]
  • 198.Piper K, Brickwood S, Turnpenny LW, Cameron IT, Ball SG, Wilson DI, Hanley NA. Beta cell differentiation during early human pancreas development. J Endocrinol 181: 11–23, 2004. doi: 10.1677/joe.0.1810011. [DOI] [PubMed] [Google Scholar]
  • 199.Plank JL, Frist AY, LeGrone AW, Magnuson MA, Labosky PA. Loss of Foxd3 results in decreased β-cell proliferation and glucose intolerance during pregnancy. Endocrinology 152: 4589–4600, 2011. doi: 10.1210/en.2010-1462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Polak M, Bouchareb-Banaei L, Scharfmann R, Czernichow P. Early pattern of differentiation in the human pancreas. Diabetes 49: 225–232, 2000. doi: 10.2337/diabetes.49.2.225. [DOI] [PubMed] [Google Scholar]
  • 201.Porat S, Weinberg-Corem N, Tornovsky-Babaey S, Schyr-Ben-Haroush R, Hija A, Stolovich-Rain M, Dadon D, Granot Z, Ben-Hur V, White P, Girard CA, Karni R, Kaestner KH, Ashcroft FM, Magnuson MA, Saada A, Grimsby J, Glaser B, Dor Y. Control of pancreatic β cell regeneration by glucose metabolism. Cell Metab 13: 440–449, 2011. doi: 10.1016/j.cmet.2011.02.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Poy MN, Hausser J, Trajkovski M, Braun M, Collins S, Rorsman P, Zavolan M, Stoffel M. miR-375 maintains normal pancreatic alpha- and beta-cell mass. Proc Natl Acad Sci USA 106: 5813–5818, 2009. doi: 10.1073/pnas.0810550106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Rafacho A, Ribeiro DL, Boschero AC, Taboga SR, Bosqueiro JR. Increased pancreatic islet mass is accompanied by activation of the insulin receptor substrate-2/serine-threonine kinase pathway and augmented cyclin D2 protein levels in insulin-resistant rats. Int J Exp Pathol 89: 264–275, 2008. doi: 10.1111/j.1365-2613.2008.00588.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Rahier J, Guiot Y, Goebbels RM, Sempoux C, Henquin JC. Pancreatic beta-cell mass in European subjects with type 2 diabetes. Diabetes Obes Metab 10, Suppl 4: 32–42, 2008. doi: 10.1111/j.1463-1326.2008.00969.x. [DOI] [PubMed] [Google Scholar]
  • 205.Ravassard P, Hazhouz Y, Pechberty S, Bricout-Neveu E, Armanet M, Czernichow P, Scharfmann R. A genetically engineered human pancreatic β cell line exhibiting glucose-inducible insulin secretion. J Clin Invest 121: 3589–3597, 2011. doi: 10.1172/JCI58447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Rawn SM, Huang C, Hughes M, Shaykhutdinov R, Vogel HJ, Cross JC. Pregnancy Hyperglycemia in Prolactin Receptor Mutant, but Not Prolactin Mutant, Mice and Feeding-Responsive Regulation of Placental Lactogen Genes Implies Placental Control of Maternal Glucose Homeostasis. Biol Reprod 93: 75, 2015. doi: 10.1095/biolreprod.115.132431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Rebuffat SA, Oliveira JM, Altirriba J, Palau N, Garcia A, Esteban Y, Nadal B, Gomis R. Downregulation of Sfrp5 promotes beta cell proliferation during obesity in the rat. Diabetologia 56: 2446–2455, 2013. doi: 10.1007/s00125-013-3030-x. [DOI] [PubMed] [Google Scholar]
  • 208.Rezania A, Bruin JE, Arora P, Rubin A, Batushansky I, Asadi A, O’Dwyer S, Quiskamp N, Mojibian M, Albrecht T, Yang YH, Johnson JD, Kieffer TJ. Reversal of diabetes with insulin-producing cells derived in vitro from human pluripotent stem cells. Nat Biotechnol 32: 1121–1133, 2014. doi: 10.1038/nbt.3033. [DOI] [PubMed] [Google Scholar]
  • 209.Rezania A, Bruin JE, Riedel MJ, Mojibian M, Asadi A, Xu J, Gauvin R, Narayan K, Karanu F, O’Neil JJ, Ao Z, Warnock GL, Kieffer TJ. Maturation of human embryonic stem cell-derived pancreatic progenitors into functional islets capable of treating pre-existing diabetes in mice. Diabetes 61: 2016–2029, 2012. doi: 10.2337/db11-1711. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Rezania A, Bruin JE, Xu J, Narayan K, Fox JK, O’Neil JJ, Kieffer TJ. Enrichment of human embryonic stem cell-derived NKX6.1-expressing pancreatic progenitor cells accelerates the maturation of insulin-secreting cells in vivo. Stem Cells 31: 2432–2442, 2013. doi: 10.1002/stem.1489. [DOI] [PubMed] [Google Scholar]
  • 211.Rhodes CJ. IGF-I and GH post-receptor signaling mechanisms for pancreatic beta-cell replication. J Mol Endocrinol 24: 303–311, 2000. doi: 10.1677/jme.0.0240303. [DOI] [PubMed] [Google Scholar]
  • 212.Rieck S, Kaestner KH. Expansion of beta-cell mass in response to pregnancy. Trends Endocrinol Metab 21: 151–158, 2010. doi: 10.1016/j.tem.2009.11.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Rieck S, White P, Schug J, Fox AJ, Smirnova O, Gao N, Gupta RK, Wang ZV, Scherer PE, Keller MP, Attie AD, Kaestner KH. The transcriptional response of the islet to pregnancy in mice. Mol Endocrinol 23: 1702–1712, 2009. doi: 10.1210/me.2009-0144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Riedel MJ, Asadi A, Wang R, Ao Z, Warnock GL, Kieffer TJ. Immunohistochemical characterisation of cells co-producing insulin and glucagon in the developing human pancreas. Diabetologia 55: 372–381, 2012. doi: 10.1007/s00125-011-2344-9. [DOI] [PubMed] [Google Scholar]
  • 215.Ritzel RA, Butler AE, Rizza RA, Veldhuis JD, Butler PC. Relationship between beta-cell mass and fasting blood glucose concentration in humans. Diabetes Care 29: 717–718, 2006. doi: 10.2337/diacare.29.03.06.dc05-1538. [DOI] [PubMed] [Google Scholar]
  • 216.Rubio-Cabezas O, Minton JA, Kantor I, Williams D, Ellard S, Hattersley AT. Homozygous mutations in NEUROD1 are responsible for a novel syndrome of permanent neonatal diabetes and neurological abnormalities. Diabetes 59: 2326–2331, 2010. doi: 10.2337/db10-0011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Russ HA, Bar Y, Ravassard P, Efrat S. In vitro proliferation of cells derived from adult human beta-cells revealed by cell-lineage tracing. Diabetes 57: 1575–1583, 2008. doi: 10.2337/db07-1283. [DOI] [PubMed] [Google Scholar]
  • 218.Russ HA, Parent AV, Ringler JJ, Hennings TG, Nair GG, Shveygert M, Guo T, Puri S, Haataja L, Cirulli V, Blelloch R, Szot GL, Arvan P, Hebrok M. Controlled induction of human pancreatic progenitors produces functional beta-like cells in vitro. EMBO J 34: 1759–1772, 2015. doi: 10.15252/embj.201591058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Russ HA, Sintov E, Anker-Kitai L, Friedman O, Lenz A, Toren G, Farhy C, Pasmanik-Chor M, Oron-Karni V, Ravassard P, Efrat S. Insulin-producing cells generated from dedifferentiated human pancreatic beta cells expanded in vitro. PLoS One 6: e25566, 2011. doi: 10.1371/journal.pone.0025566. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Ryan EA, Paty BW, Senior PA, Bigam D, Alfadhli E, Kneteman NM, Lakey JR, Shapiro AM. Five-year follow-up after clinical islet transplantation. Diabetes 54: 2060–2069, 2005. doi: 10.2337/diabetes.54.7.2060. [DOI] [PubMed] [Google Scholar]
  • 221.Saisho Y, Butler AE, Manesso E, Elashoff D, Rizza RA, Butler PC. β-Cell mass and turnover in humans: effects of obesity and aging. Diabetes Care 36: 111–117, 2013. doi: 10.2337/dc12-0421. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Salisbury RJ, Blaylock J, Berry AA, Jennings RE, De Krijger R, Piper Hanley K, Hanley NA. The window period of NEUROGENIN3 during human gestation. Islets 6: e954436, 2014. doi: 10.4161/19382014.2014.954436. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223.Salpeter SJ, Klein AM, Huangfu D, Grimsby J, Dor Y. Glucose and aging control the quiescence period that follows pancreatic beta cell replication. Development 137: 3205–3213, 2010. doi: 10.1242/dev.054304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Sapir T, Shternhall K, Meivar-Levy I, Blumenfeld T, Cohen H, Skutelsky E, Eventov-Friedman S, Barshack I, Goldberg I, Pri-Chen S, Ben-Dor L, Polak-Charcon S, Karasik A, Shimon I, Mor E, Ferber S. Cell-replacement therapy for diabetes: generating functional insulin-producing tissue from adult human liver cells. Proc Natl Acad Sci USA 102: 7964–7969, 2005. doi: 10.1073/pnas.0405277102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.Sarkar SA, Kobberup S, Wong R, Lopez AD, Quayum N, Still T, Kutchma A, Jensen JN, Gianani R, Beattie GM, Jensen J, Hayek A, Hutton JC. Global gene expression profiling and histochemical analysis of the developing human fetal pancreas. Diabetologia 51: 285–297, 2008. doi: 10.1007/s00125-007-0880-0. [DOI] [PubMed] [Google Scholar]
  • 226.Scharfmann R, Pechberty S, Hazhouz Y, von Bülow M, Bricout-Neveu E, Grenier-Godard M, Guez F, Rachdi L, Lohmann M, Czernichow P, Ravassard P. Development of a conditionally immortalized human pancreatic β cell line. J Clin Invest 124: 2087–2098, 2014. doi: 10.1172/JCI72674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Schinner S, Ulgen F, Papewalis C, Schott M, Woelk A, Vidal-Puig A, Scherbaum WA. Regulation of insulin secretion, glucokinase gene transcription and beta cell proliferation by adipocyte-derived Wnt signalling molecules. Diabetologia 51: 147–154, 2008. doi: 10.1007/s00125-007-0848-0. [DOI] [PubMed] [Google Scholar]
  • 228.Schraenen A, Lemaire K, de Faudeur G, Hendrickx N, Granvik M, Van Lommel L, Mallet J, Vodjdani G, Gilon P, Binart N, in’t Veld P, Schuit F. Placental lactogens induce serotonin biosynthesis in a subset of mouse beta cells during pregnancy. Diabetologia 53: 2589–2599, 2010. doi: 10.1007/s00125-010-1913-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Schwitzgebel VM, Scheel DW, Conners JR, Kalamaras J, Lee JE, Anderson DJ, Sussel L, Johnson JD, German MS. Expression of neurogenin3 reveals an islet cell precursor population in the pancreas. Development 127: 3533–3542, 2000. [DOI] [PubMed] [Google Scholar]
  • 230.Sellick GS, Barker KT, Stolte-Dijkstra I, Fleischmann C, Coleman RJ, Garrett C, Gloyn AL, Edghill EL, Hattersley AT, Wellauer PK, Goodwin G, Houlston RS. Mutations in PTF1A cause pancreatic and cerebellar agenesis. Nat Genet 36: 1301–1305, 2004. doi: 10.1038/ng1475. [DOI] [PubMed] [Google Scholar]
  • 231.Senée V, Chelala C, Duchatelet S, Feng D, Blanc H, Cossec JC, Charon C, Nicolino M, Boileau P, Cavener DR, Bougnères P, Taha D, Julier C. Mutations in GLIS3 are responsible for a rare syndrome with neonatal diabetes mellitus and congenital hypothyroidism. Nat Genet 38: 682–687, 2006. doi: 10.1038/ng1802. [DOI] [PubMed] [Google Scholar]
  • 232.Seymour PA, Freude KK, Tran MN, Mayes EE, Jensen J, Kist R, Scherer G, Sander M. SOX9 is required for maintenance of the pancreatic progenitor cell pool. Proc Natl Acad Sci USA 104: 1865–1870, 2007. doi: 10.1073/pnas.0609217104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233.Shapiro AM, Lakey JR, Ryan EA, Korbutt GS, Toth E, Warnock GL, Kneteman NM, Rajotte RV. Islet transplantation in seven patients with type 1 diabetes mellitus using a glucocorticoid-free immunosuppressive regimen. N Engl J Med 343: 230–238, 2000. doi: 10.1056/NEJM200007273430401. [DOI] [PubMed] [Google Scholar]
  • 234.Shapiro AM, Ricordi C, Hering BJ, Auchincloss H, Lindblad R, Robertson RP, Secchi A, Brendel MD, Berney T, Brennan DC, Cagliero E, Alejandro R, Ryan EA, DiMercurio B, Morel P, Polonsky KS, Reems JA, Bretzel RG, Bertuzzi F, Froud T, Kandaswamy R, Sutherland DE, Eisenbarth G, Segal M, Preiksaitis J, Korbutt GS, Barton FB, Viviano L, Seyfert-Margolis V, Bluestone J, Lakey JR. International trial of the Edmonton protocol for islet transplantation. N Engl J Med 355: 1318–1330, 2006. doi: 10.1056/NEJMoa061267. [DOI] [PubMed] [Google Scholar]
  • 235.Sharma RB, O’Donnell AC, Stamateris RE, Ha B, McCloskey KM, Reynolds PR, Arvan P, Alonso LC. Insulin demand regulates β cell number via the unfolded protein response. J Clin Invest 125: 3831–3846, 2015. doi: 10.1172/JCI79264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236.Shaw-Smith C, De Franco E, Lango Allen H, Batlle M, Flanagan SE, Borowiec M, Taplin CE, van Alfen-van der Velden J, Cruz-Rojo J, Perez de Nanclares G, Miedzybrodzka Z, Deja G, Wlodarska I, Mlynarski W, Ferrer J, Hattersley AT, Ellard S. GATA4 mutations are a cause of neonatal and childhood-onset diabetes. Diabetes 63: 2888–2894, 2014. doi: 10.2337/db14-0061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.Shay JW, Wright WE. Aging. When do telomeres matter? Science 291: 839–840, 2001. doi: 10.1126/science.1058546. [DOI] [PubMed] [Google Scholar]
  • 238.Shen W, Taylor B, Jin Q, Nguyen-Tran V, Meeusen S, Zhang YQ, Kamireddy A, Swafford A, Powers AF, Walker J, Lamb J, Bursalaya B, DiDonato M, Harb G, Qiu M, Filippi CM, Deaton L, Turk CN, Suarez-Pinzon WL, Liu Y, Hao X, Mo T, Yan S, Li J, Herman AE, Hering BJ, Wu T, Martin Seidel H, McNamara P, Glynne R, Laffitte B. Inhibition of DYRK1A and GSK3B induces human β-cell proliferation. Nat Commun 6: 8372, 2015. doi: 10.1038/ncomms9372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239.Shen W, Tremblay MS, Deshmukh VA, Wang W, Filippi CM, Harb G, Zhang YQ, Kamireddy A, Baaten JE, Jin Q, Wu T, Swoboda JG, Cho CY, Li J, Laffitte BA, McNamara P, Glynne R, Wu X, Herman AE, Schultz PG. Small-molecule inducer of β cell proliferation identified by high-throughput screening. J Am Chem Soc 135: 1669–1672, 2013. doi: 10.1021/ja309304m. [DOI] [PubMed] [Google Scholar]
  • 240.Sherwood RI, Chen TY, Melton DA. Transcriptional dynamics of endodermal organ formation. Dev Dyn 238: 29–42, 2009. doi: 10.1002/dvdy.21810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241.Shirakawa J, Kulkarni RN. Novel factors modulating human β-cell proliferation. Diabetes Obes Metab 18, Suppl 1: 71–77, 2016. doi: 10.1111/dom.12731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242.Smith SB, Qu HQ, Taleb N, Kishimoto NY, Scheel DW, Lu Y, Patch AM, Grabs R, Wang J, Lynn FC, Miyatsuka T, Mitchell J, Seerke R, Désir J, Vanden Eijnden S, Abramowicz M, Kacet N, Weill J, Renard ME, Gentile M, Hansen I, Dewar K, Hattersley AT, Wang R, Wilson ME, Johnson JD, Polychronakos C, German MS. Rfx6 directs islet formation and insulin production in mice and humans. Nature 463: 775–780, 2010. doi: 10.1038/nature08748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243.Smukler SR, Arntfield ME, Razavi R, Bikopoulos G, Karpowicz P, Seaberg R, Dai F, Lee S, Ahrens R, Fraser PE, Wheeler MB, van der Kooy D. The adult mouse and human pancreas contain rare multipotent stem cells that express insulin. Cell Stem Cell 8: 281–293, 2011. doi: 10.1016/j.stem.2011.01.015. [DOI] [PubMed] [Google Scholar]
  • 244.Solar M, Cardalda C, Houbracken I, Martín M, Maestro MA, De Medts N, Xu X, Grau V, Heimberg H, Bouwens L, Ferrer J. Pancreatic exocrine duct cells give rise to insulin-producing beta cells during embryogenesis but not after birth. Dev Cell 17: 849–860, 2009. doi: 10.1016/j.devcel.2009.11.003. [DOI] [PubMed] [Google Scholar]
  • 245.Sorenson RL, Brelje TC. Adaptation of islets of Langerhans to pregnancy: beta-cell growth, enhanced insulin secretion and the role of lactogenic hormones. Horm Metab Res 29: 301–307, 1997. doi: 10.1055/s-2007-979040. [DOI] [PubMed] [Google Scholar]
  • 246.Sperling MA. ATP-sensitive potassium channels–neonatal diabetes mellitus and beyond. N Engl J Med 355: 507–510, 2006. doi: 10.1056/NEJMe068142. [DOI] [PubMed] [Google Scholar]
  • 247.Spijker HS, Ravelli RB, Mommaas-Kienhuis AM, van Apeldoorn AA, Engelse MA, Zaldumbide A, Bonner-Weir S, Rabelink TJ, Hoeben RC, Clevers H, Mummery CL, Carlotti F, de Koning EJ. Conversion of mature human β-cells into glucagon-producing α-cells. Diabetes 62: 2471–2480, 2013. doi: 10.2337/db12-1001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248.Stamateris RE, Sharma RB, Kong Y, Ebrahimpour P, Panday D, Ranganath P, Zou B, Levitt H, Parambil NA, O’Donnell CP, García-Ocaña A, Alonso LC. Glucose Induces Mouse β-Cell Proliferation via IRS2, MTOR, and Cyclin D2 but Not the Insulin Receptor. Diabetes 65: 981–995, 2016. doi: 10.2337/db15-0529. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249.Stewart AF. Betatrophin versus bitter-trophin and the elephant in the room: time for a new normal in β-cell regeneration research. Diabetes 63: 1198–1199, 2014. doi: 10.2337/db14-0009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 250.Staffers DA, Ferrer J, Clarke WL, Habener JF. Early-onset type-II diabetes mellitus (MODY4) linked to IPF1. Nat Genet 17: 138–139, 1997. doi: 10.1038/ng1097-138. [DOI] [PubMed] [Google Scholar]
  • 251.Stoffers DA, Zinkin NT, Stanojevic V, Clarke WL, Habener JF. Pancreatic agenesis attributable to a single nucleotide deletion in the human IPF1 gene coding sequence. Nat Genet 15: 106–110, 1997. doi: 10.1038/ng0197-106. [DOI] [PubMed] [Google Scholar]
  • 252.Suarez-Pinzon WL, Lakey JR, Rabinovitch A. Combination therapy with glucagon-like peptide-1 and gastrin induces beta-cell neogenesis from pancreatic duct cells in human islets transplanted in immunodeficient diabetic mice. Cell Transplant 17: 631–640, 2008. doi: 10.3727/096368908786092775. [DOI] [PubMed] [Google Scholar]
  • 253.Suarez-Pinzon WL, Rabinovitch A. Combination therapy with a dipeptidyl peptidase-4 inhibitor and a proton pump inhibitor induces β-cell neogenesis from adult human pancreatic duct cells implanted in immunodeficient mice. Cell Transplant 20: 1343–1349, 2011. doi: 10.3727/096368910X557263. [DOI] [PubMed] [Google Scholar]
  • 254.Sullivan BA, Hollister-Lock J, Bonner-Weir S, Weir GC. Reduced Ki67 staining in the postmortem state calls into question past conclusions about the lack of turnover of adult human beta cells. Diabetes 64: 1698–1702, 2015. doi: 10.2337/db14-1675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255.Szabat M, Luciani DS, Piret JM, Johnson JD. Maturation of adult beta-cells revealed using a Pdx1/insulin dual-reporter lentivirus. Endocrinology 150: 1627–1635, 2009. doi: 10.1210/en.2008-1224. [DOI] [PubMed] [Google Scholar]
  • 256.Szabat M, Pourghaderi P, Soukhatcheva G, Verchere CB, Warnock GL, Piret JM, Johnson JD. Kinetics and genomic profiling of adult human and mouse β-cell maturation. Islets 3: 175–187, 2011. doi: 10.4161/isl.3.4.15881. [DOI] [PubMed] [Google Scholar]
  • 257.Takamoto I, Terauchi Y, Kubota N, Ohsugi M, Ueki K, Kadowaki T. Crucial role of insulin receptor substrate-2 in compensatory beta-cell hyperplasia in response to high fat diet-induced insulin resistance. Diabetes Obes Metab 10, Suppl 4: 147–156, 2008. doi: 10.1111/j.1463-1326.2008.00951.x. [DOI] [PubMed] [Google Scholar]
  • 258.Talchai C, Xuan S, Lin HV, Sussel L, Accili D. Pancreatic β cell dedifferentiation as a mechanism of diabetic β cell failure. Cell 150: 1223–1234, 2012. doi: 10.1016/j.cell.2012.07.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 259.Taneera J, Rosengren A, Renstrom E, Nygren JM, Serup P, Rorsman P, Jacobsen SE. Failure of transplanted bone marrow cells to adopt a pancreatic beta-cell fate. Diabetes 55: 290–296, 2006. doi: 10.2337/diabetes.55.02.06.db05-1212. [DOI] [PubMed] [Google Scholar]
  • 260.Tattikota SG, Rathjen T, McAnulty SJ, Wessels HH, Akerman I, van de Bunt M, Hausser J, Esguerra JL, Musahl A, Pandey AK, You X, Chen W, Herrera PL, Johnson PR, O’Carroll D, Eliasson L, Zavolan M, Gloyn AL, Ferrer J, Shalom-Feuerstein R, Aberdam D, Poy MN. Argonaute2 mediates compensatory expansion of the pancreatic β cell. Cell Metab 19: 122–134, 2014. doi: 10.1016/j.cmet.2013.11.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 261.Terauchi Y, Takamoto I, Kubota N, Matsui J, Suzuki R, Komeda K, Hara A, Toyoda Y, Miwa I, Aizawa S, Tsutsumi S, Tsubamoto Y, Hashimoto S, Eto K, Nakamura A, Noda M, Tobe K, Aburatani H, Nagai R, Kadowaki T. Glucokinase and IRS-2 are required for compensatory beta cell hyperplasia in response to high-fat diet-induced insulin resistance. J Clin Invest 117: 246–257, 2007. doi: 10.1172/JCI17645. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 262.Teta M, Rankin MM, Long SY, Stein GM, Kushner JA. Growth and regeneration of adult beta cells does not involve specialized progenitors. Dev Cell 12: 817–826, 2007. doi: 10.1016/j.devcel.2007.04.011. [DOI] [PubMed] [Google Scholar]
  • 263.Thorel F, Népote V, Avril I, Kohno K, Desgraz R, Chera S, Herrera PL. Conversion of adult pancreatic alpha-cells to beta-cells after extreme beta-cell loss. Nature 464: 1149–1154, 2010. doi: 10.1038/nature08894. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 264.Todorov I, Omori K, Pascual M, Rawson J, Nair I, Valiente L, Vuong T, Matsuda T, Orr C, Ferreri K, Smith CV, Kandeel F, Mullen Y. Generation of human islets through expansion and differentiation of non-islet pancreatic cells discarded (pancreatic discard) after islet isolation. Pancreas 32: 130–138, 2006. doi: 10.1097/01.mpa.0000202945.78331.93. [DOI] [PubMed] [Google Scholar]
  • 265.Toselli C, Hyslop CM, Hughes M, Natale DR, Santamaria P, Huang CT. Contribution of a non-β-cell source to β-cell mass during pregnancy. PLoS One 9: e100398, 2014. doi: 10.1371/journal.pone.0100398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 266.Turner M, Leslie S, Martin NG, Peschanski M, Rao M, Taylor CJ, Trounson A, Turner D, Yamanaka S, Wilmut I. Toward the development of a global induced pluripotent stem cell library. Cell Stem Cell 13: 382–384, 2013. doi: 10.1016/j.stem.2013.08.003. [DOI] [PubMed] [Google Scholar]
  • 267.Tyrberg B, Ustinov J, Otonkoski T, Andersson A. Stimulated endocrine cell proliferation and differentiation in transplanted human pancreatic islets: effects of the ob gene and compensatory growth of the implantation organ. Diabetes 50: 301–307, 2001. doi: 10.2337/diabetes.50.2.301. [DOI] [PubMed] [Google Scholar]
  • 268.Van Assche FA, Aerts L, De Prins F. A morphological study of the endocrine pancreas in human pregnancy. Br J Obstet Gynaecol 85: 818–820, 1978. doi: 10.1111/j.1471-0528.1978.tb15835.x. [DOI] [PubMed] [Google Scholar]
  • 269.Van der Meulen T, Lee S, Noordeloos E, Donaldson CJ, Adams MW, Noguchi GM, Mawla AM, Huising MO. Artemether Does Not Turn α Cells into β Cells. Cell Metab 27: 218–225.e4, 2018. doi: 10.1016/j.cmet.2017.10.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 270.Vasu S, Moffett RC, Thorens B, Flatt PR. Role of endogenous GLP-1 and GIP in beta cell compensatory responses to insulin resistance and cellular stress. PLoS One 9: e101005, 2014. doi: 10.1371/journal.pone.0101005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 271.Villasenor A, Chong DC, Cleaver O. Biphasic Ngn3 expression in the developing pancreas. Dev Dyn 237: 3270–3279, 2008. doi: 10.1002/dvdy.21740. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 272.Villasenor A, Cleaver O. Crosstalk between the developing pancreas and its blood vessels: an evolving dialog. Semin Cell Dev Biol 23: 685–692, 2012. doi: 10.1016/j.semcdb.2012.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 273.Wagers AJ, Weissman IL. Plasticity of adult stem cells. Cell 116: 639–648, 2004. doi: 10.1016/S0092-8674(04)00208-9. [DOI] [PubMed] [Google Scholar]
  • 274.Wang P, Alvarez-Perez JC, Felsenfeld DP, Liu H, Sivendran S, Bender A, Kumar A, Sanchez R, Scott DK, Garcia-Ocaña A, Stewart AF. A high-throughput chemical screen reveals that harmine-mediated inhibition of DYRK1A increases human pancreatic beta cell replication. Nat Med 21: 383–388, 2015. doi: 10.1038/nm.3820. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 275.Wang P, Fiaschi-Taesch NM, Vasavada RC, Scott DK, García-Ocaña A, Stewart AF. Diabetes mellitus–advances and challenges in human β-cell proliferation. Nat Rev Endocrinol 11: 201–212, 2015. doi: 10.1038/nrendo.2015.9. [DOI] [PubMed] [Google Scholar]
  • 276.Weedon MN, Cebola I, Patch AM, Flanagan SE, De Franco E, Caswell R, Rodríguez-Seguí SA, Shaw-Smith C, Cho CH, Allen HL, Houghton JA, Roth CL, Chen R, Hussain K, Marsh P, Vallier L, Murray A, Ellard S, Ferrer J, Hattersley AT; International Pancreatic Agenesis Consortium . Recessive mutations in a distal PTF1A enhancer cause isolated pancreatic agenesis. Nat Genet 46: 61–64, 2014. doi: 10.1038/ng.2826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 277.Weinberg N, Ouziel-Yahalom L, Knoller S, Efrat S, Dor Y. Lineage tracing evidence for in vitro dedifferentiation but rare proliferation of mouse pancreatic beta-cells. Diabetes 56: 1299–1304, 2007. doi: 10.2337/db06-1654. [DOI] [PubMed] [Google Scholar]
  • 278.Weir GC, Bonner-Weir S. Islet β cell mass in diabetes and how it relates to function, birth, and death. Ann N Y Acad Sci 1281: 92–105, 2013. doi: 10.1111/nyas.12031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 279.Wright WE, Shay JW. Historical claims and current interpretations of replicative aging. Nat Biotechnol 20: 682–688, 2002. doi: 10.1038/nbt0702-682. [DOI] [PubMed] [Google Scholar]
  • 280.Xiao X, Chen Z, Shiota C, Prasadan K, Guo P, El-Gohary Y, Paredes J, Welsh C, Wiersch J, Gittes GK. No evidence for β cell neogenesis in murine adult pancreas. J Clin Invest 123: 2207–2217, 2013. doi: 10.1172/JCI66323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 281.Xie W, Schultz MD, Lister R, Hou Z, Rajagopal N, Ray P, Whitaker JW, Tian S, Hawkins RD, Leung D, Yang H, Wang T, Lee AY, Swanson SA, Zhang J, Zhu Y, Kim A, Nery JR, Urich MA, Kuan S, Yen CA, Klugman S, Yu P, Suknuntha K, Propson NE, Chen H, Edsall LE, Wagner U, Li Y, Ye Z, Kulkarni A, Xuan Z, Chung WY, Chi NC, Antosiewicz-Bourget JE, Slukvin I, Stewart R, Zhang MQ, Wang W, Thomson JA, Ecker JR, Ren B. Epigenomic analysis of multilineage differentiation of human embryonic stem cells. Cell 153: 1134–1148, 2013. doi: 10.1016/j.cell.2013.04.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 282.Ye F, Duvillié B, Scharfmann R. Fibroblast growth factors 7 and 10 are expressed in the human embryonic pancreatic mesenchyme and promote the proliferation of embryonic pancreatic epithelial cells. Diabetologia 48: 277–281, 2005. doi: 10.1007/s00125-004-1638-6. [DOI] [PubMed] [Google Scholar]
  • 283.Yi P, Park JS, Melton DA. Betatrophin: a hormone that controls pancreatic β cell proliferation. Cell 153: 747–758, 2013. doi: 10.1016/j.cell.2013.04.008. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 284.Yi P, Park JS, Melton DA. Perspectives on the activities of ANGPTL8/betatrophin. Cell 159: 467–468, 2014. doi: 10.1016/j.cell.2014.09.028. [DOI] [PubMed] [Google Scholar]
  • 285.Yi P, Park JS, Melton DA. Retraction Notice to: Betatrophin: A Hormone that Controls Pancreatic β Cell Proliferation. Cell 168: 326, 2017. doi: 10.1016/j.cell.2016.12.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 286.Yorifuji T, Kurokawa K, Mamada M, Imai T, Kawai M, Nishi Y, Shishido S, Hasegawa Y, Nakahata T. Neonatal diabetes mellitus and neonatal polycystic, dysplastic kidneys: phenotypically discordant recurrence of a mutation in the hepatocyte nuclear factor-1beta gene due to germline mosaicism. J Clin Endocrinol Metab 89: 2905–2908, 2004. doi: 10.1210/jc.2003-031828. [DOI] [PubMed] [Google Scholar]
  • 287.Yoshitomi H, Zaret KS. Endothelial cell interactions initiate dorsal pancreas development by selectively inducing the transcription factor Ptf1a. Development 131: 807–817, 2004. doi: 10.1242/dev.00960. [DOI] [PubMed] [Google Scholar]
  • 288.Yuan HX, Xiong Y, Guan KL. Nutrient sensing, metabolism, and cell growth control. Mol Cell 49: 379–387, 2013. doi: 10.1016/j.molcel.2013.01.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 289.Zahr E, Molano RD, Pileggi A, Ichii H, Jose SS, Bocca N, An W, Gonzalez-Quintana J, Fraker C, Ricordi C, Inverardi L. Rapamycin impairs in vivo proliferation of islet beta-cells. Transplantation 84: 1576–1583, 2007. doi: 10.1097/01.tp.0000296035.48728.28. [DOI] [PubMed] [Google Scholar]
  • 290.Zalzman M, Anker-Kitai L, Efrat S. Differentiation of human liver-derived, insulin-producing cells toward the beta-cell phenotype. Diabetes 54: 2568–2575, 2005. doi: 10.2337/diabetes.54.9.2568. [DOI] [PubMed] [Google Scholar]
  • 291.Zhang H, Ackermann AM, Gusarova GA, Lowe D, Feng X, Kopsombut UG, Costa RH, Gannon M. The FoxM1 transcription factor is required to maintain pancreatic beta-cell mass. Mol Endocrinol 20: 1853–1866, 2006. doi: 10.1210/me.2006-0056. [DOI] [PubMed] [Google Scholar]
  • 292.Zhang H, Zhang J, Pope CF, Crawford LA, Vasavada RC, Jagasia SM, Gannon M. Gestational diabetes mellitus resulting from impaired beta-cell compensation in the absence of FoxM1, a novel downstream effector of placental lactogen. Diabetes 59: 143–152, 2010. doi: 10.2337/db09-0050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 293.Zhang P, Kumar A, Katz LS, Li L, Paulynice M, Herman MA, Scott DK. Induction of the ChREBPβ Isoform Is Essential for Glucose-Stimulated β-Cell Proliferation. Diabetes 64: 4158–4170, 2015. doi: 10.2337/db15-0239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 294.Zhang T, Kim DH, Xiao X, Lee S, Gong Z, Muzumdar R, Calabuig-Navarro V, Yamauchi J, Harashima H, Wang R, Bottino R, Alvarez-Perez JC, Garcia-Ocaña A, Gittes G, Dong HH. FoxO1 Plays an Important Role in Regulating β-Cell Compensation for Insulin Resistance in Male Mice. Endocrinology 157: 1055–1070, 2016. doi: 10.1210/en.2015-1852. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 295.Zhou Q, Brown J, Kanarek A, Rajagopal J, Melton DA. In vivo reprogramming of adult pancreatic exocrine cells to beta-cells. Nature 455: 627–632, 2008. doi: 10.1038/nature07314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 296.Zhu S, Russ HA, Wang X, Zhang M, Ma T, Xu T, Tang S, Hebrok M, Ding S. Human pancreatic beta-like cells converted from fibroblasts. Nat Commun 7: 10080, 2016. doi: 10.1038/ncomms10080. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Physiological Reviews are provided here courtesy of American Physiological Society

RESOURCES