Abstract
The pancreatic β-cells are essential for regulating glucose homeostasis through the coordinated release of the insulin hormone. Dysfunction of the highly specialized β-cells results in diabetes mellitus, a growing global health epidemic. In this review, we describe the development and function of β-cells the emerging concept of heterogeneity within insulin-producing cells, and the potential of other cell types to assume β-cell functionality via transdifferentiation. We also discuss emerging routes to design cells with minimal β-cell properties and human stem cell differentiation efforts that carry the promise to restore normoglycemia in patients suffering from diabetes.
THE β-CELL
Pancreatic β-cells are vital to the maintenance of systemic glucose homeostasis. In response to elevated blood glucose levels, β-cells secrete the peptide hormone insulin, which stimulates glucose uptake by peripheral tissues such as skeletal muscle, liver, and adipose tissue. Glucose-stimulated insulin secretion (GSIS) occurs when β-cells sense and absorb glucose from the bloodstream through specific glucose transporters. Internalized glucose is metabolized, largely within the mitochondria, to generate ATP. This increase in the intracellular ATP/ADP ratio induces closure of KATP-sensitive channels and plasma membrane depolarization, causing Ca2+ channels along the cell surface to open. The resulting influx of Ca2+ ions induces exocytosis of insulin granules from the β-cell interior (Fig. 1). Tight regulation of insulin secretion is critical; constitutive insulin secretion results in chronic low blood glucose, or hypoglycemia, which poses a significant immediate health risk. In contrast, perturbation of β-cell function and diminished insulin secretion results in diabetes and its associated long-term complications.
Figure 1.
Schematic of key steps of insulin secretion mechanism in mature β-cells with emphasis on key steps. (1) Rise in extracellular glucose concentration leads to its uptake via the glucose transporter GLUT2 (GLUT1 in human β-cells), which is phosphorylated by glucokinase, to (2) glucose-6-phosphate (G-6-P) entering the glycolysis pathway to generate pyruvate, ATP, and NADH. Pyruvate is channeled to mitochondria to undergo metabolism by the Krebs cycle and subsequently generating more ATP (3), which together with ATP generated from glycolysis, increases the ATP/ADP ratio. This causes closure of ATP-dependent K+ (KATP) channels (4), leading to reduced K+ efflux, plasma membrane depolarization (5), and opening of voltage-dependent Ca2+ channels (6). This results in Ca2+ influx and rise in cytosolic free Ca2+ concentration [Ca2+], which acts as a triggering signal for insulin secretion (7). High levels of ATP further facilitate insulin vesicle transport and docking on the plasma membrane and subsequent release into the cytosol (8). (ER) Endoplasmic reticulum.
According to the WHO report (2021), diabetes is an evolving disease affecting 422 million people worldwide and causing 1.6 million deaths annually, with the prevalence rate increasing steadily over the past few decades majorly in middle- and low-income countries. The most prevalent and well-characterized forms of diabetes are type 1 diabetes and type 2 diabetes, which account for nearly all diagnosed cases; however, there exist multiple additional subtypes, including the monogenetic mature onset diabetes of the young (MODY) and transient gestational diabetes (Ahlqvist et al. 2020). Type 1 diabetes is an autoimmune disease caused by the destruction of β-cells by activated immune cells. This wholesale elimination of β-cells results in a near-complete lack of circulating insulin, necessitating treatment with exogenous insulin or a whole-pancreas or islet transplant. Type 2 diabetes, however, is driven by two primary factors: an acquired resistance to insulin by the peripheral tissues responsible for glucose uptake and β-cell dysfunction leading to an inability to produce and secrete sufficient insulin. This review will explore various aspects of β-cell biology: physiology, development, and intra- and intercellular interactions. We will also delve into the concepts of β-cell heterogeneity and plasticity. Finally, we will touch upon modern strategies to generate β-cells in vitro and the novel, emergent technologies designed to improve and protect β-cells.
HISTOLOGY AND ARCHITECTURE OF THE PANCREATIC ISLET
The pancreas includes two distinct functional compartments. The exocrine pancreas, which accounts for the vast majority of the pancreas, includes the acinar and ductal cells responsible for producing, secreting, and transporting digestive enzymes (Gardner and Jackson 1977; Kern 1993; Williams 2010; Reichert and Rustgi 2011). The endocrine pancreas refers to the scattered clusters of endocrine cells, vasculature, and neurons, otherwise known as islets of Langerhans, which produce and secrete various peptide hormones (Logothetopoulos et al. 1965; Orci and Unger 1975). The cell-type and functional diversity present within the islet has resulted in their conceptualization as “micro-organs.” Insulin-secreting β-cells are the predominant endocrine cell type present in the islet; others include α- and δ-cells, which produce and secrete glucagon and somatostatin, respectively (Lazarow 1957; Da Silva Xavier 2018). Islets also possess a small subpopulation of rare endocrine cells known as pancreatic polypeptide (PP) cells and epsilon cells, which secrete ghrelin (Prado et al. 2004). The cell types present within the islet are similar across mammalian species. It is, however, important to note the existence of species-specific differences in islet architecture, particularly relating to cellular proportions and spatial distribution. Given the prevalence of rodent models of pancreatic biology, comparison studies have predominantly focused on differences and commonalities between human and rodent islets (Cabrera et al. 2006; Kim et al. 2009; Steiner et al. 2010; Bonner-Weir et al. 2015; Dolenšek et al. 2015; Tsuchitani et al. 2016). Immunohistochemical analysis of human and rodent islets has revealed that rodent islets are composed of a clustered “core” of β-cells (∼80%–90%) surrounded by a “mantle” of α-, δ-, and PP cells at the islet periphery. Human islets, however, possess approximately equivalent numbers of β- and non-β-endocrine cells intermingled within the islet proper, although they also demonstrate a notable degree of islet heterogeneity (Dybala and Hara 2019). These discrepancies must be considered when attempting to extrapolate human β-cell biology from rodent studies.
INTRA-ISLET COMMUNICATION
Disruption of islet architecture in vitro significantly diminishes β-cell function (Dunbar and Walsh 1982; Pipeleers et al. 1982; Weir et al. 1984; Hopcroft et al. 1985). Islet structure lends itself to extensive intercellular communication: signaling between β-cells and neighboring cells may occur through hormones from neighboring α- and δ-cells, angiogenic factors from endothelial cells, neurotransmitters from resident autonomic nerves, and physical cell–cell contact.
Islet Hormones
The hormones produced by α-, β-, and δ-cells are primarily designated for endocrine signaling via secretion into the circulatory system and transport to distant organ systems. However, these hormones also serve as paracrine signaling molecules, facilitating critical intercellular cross talk and intra-islet regulation. This is apparent in the intricate regulatory patterns of glucagon, insulin, and somatostatin. Glucagon regulates glucose homeostasis both globally and within the islet (Tellez et al. 2020). During periods of hypoglycemia, α-cells release glucagon to stimulate hepatic glycogenolysis and gluconeogenesis, thereby raising blood glucose levels (Gromada et al. 2007). However, glucagon has also been shown to promote insulin secretion through interaction with the glucagon receptor (GcgR) or the GLP-1 receptor (GLP-1R) at the β-cell surface (Kieffer et al. 1996; Moens et al. 1998; Huypens et al. 2000; Svendsen et al. 2018). Given the function of insulin as a negative regulator of glucagon, this suggests that glucagon secretion promotes its own negative feedback loop to prevent glucagon-induced hyperglycemia. Conversely, somatostatin is both stimulated by and inhibitory of glucagon and insulin secretion (Koerker et al. 1974; Taborsky et al. 1978; Hauge-Evans et al. 2009), thus providing an additional layer of control to ensure tight regulation of physiological glucose levels.
Vasculature and Nerves
The islet is heavily vascularized, reflecting its endocrine function, nutrient requirements, and robust oxygen demand (Bonner-Weir and Orci 1982; Ballian and Brunicardi 2007; Nyman et al. 2008). Limited vascularization greatly hinders human islet transplants through significant β-cell death and graft failure (Brissova et al. 2004, 2005; Zhang et al. 2004). Type 2 diabetic islets further display perturbed capillary morphology and density (Brissova et al. 2014). Overexpression of VEGF-A in rodent β-cells increases vascularization, disrupts islet morphogenesis, reshapes endothelial ultrastructure, and drastically alters β-cell proliferation (Cai et al. 2012; Brissova et al. 2014). Islet vascularization is also inextricably linked to innervation: islet-associated endothelial cells produce proneuronal migration molecules, with intra-islet capillaries acting as physical scaffolding for the ingrowth of autonomic nerves (Reinert et al. 2014). Importantly, establishment of islet architecture depends upon interaction between β-cells and local sympathetic nerves; depletion of sympathetic innervation disrupts rodent islet cytoarchitecture and diminishes GSIS (Borden et al. 2013).
Physical Cell–Cell Contact
Physical endocrine cell–cell contact is critical to intra-islet cross talk. These interactions are mediated by a wide array of proteins at the cell surface, including gap junction proteins, cell-adhesion molecules (CAMs), and receptor tyrosine kinases (RTKs).
Gap junctions are formed via the tubular amalgamation of connexin proteins and act as intercellular channels. Particular attention has been devoted to connexin 36 (Cx36), which is preferentially expressed in both rodent and human β-cells (Serre-Beinier et al. 2000, 2009). An increase in gap junctions between neighboring β-cells has been shown to occur during both functional maturation and insulin secretion (Carvalho et al. 2010). Subsequent studies have demonstrated that Cx36 gap junctions, specifically, regulate the synchronization of Ca2+ oscillations that occur during GSIS (Calabrese et al. 2003; Ravier et al. 2005; Benninger et al. 2011).
CAMs refer to four distinct protein families—integrins, selectins, immunoglobulins, and cadherins—expressed on the cell surface for the purpose of facilitating cell–cell adhesion, signal transduction from the extracellular environment, and downstream intracellular events. Particularly promising have been cadherins, transmembrane glycoproteins that mediate homophilic Ca2+-dependent cell–cell interactions. Loss of E-cadherin has been shown to disrupt gap junction formation, and therefore Ca2+ signaling and insulin secretion (Yamagata et al. 2002; Rogers et al. 2007).
Ephrin receptors constitute the largest family of RTKs, unique for their ability to engage in bidirectional signaling. EphA5 receptors and their cognate ephrin-A5 ligands are both expressed in human islets (Konstantinova et al. 2007). Strikingly, loss of forward and reverse signaling between EphA5 and ephrin-A5 resulted in opposite effects: reverse signaling through ephrin-A5 increased insulin secretion during both phases of the glucose response, whereas forward signaling through EphA5 reduced insulin secretion. These effects were attributed to reduced granule fusion events on the β-cell surface, rather than alterations in upstream components of the insulin secretion pathway. Recent studies have found that ablation of the primary cilia located at the β-cell plasma membrane resulted in hyperphosphorylation of the EphA2/3 receptors and a subsequent decrease in glucose tolerance and insulin secretion, similar to that observed upon activation of EphA5 reverse signaling (Volta et al. 2019). The importance of primary cilia to the β-cells has been further bolstered by studies indicating that dysregulation of Hedgehog signaling resulting in part from primary cilia ablation induces diminished glucose tolerance and degradation of β-cell identity (Cervantes et al. 2010; Landsman et al. 2011). Taken together, these findings point to the importance of a wide array of cell-surface proteins and physical cell–cell communication in regulating β-cell function.
ISLET AND β-CELL DEVELOPMENT
β-cell development is regulated in utero by complex interplay between transcription factors and dynamic changes in gene regulatory networks. Postnatal β-cell maturation continues with molecular and metabolic events that enable the β-cell to achieve GSIS. Illuminating the process of β-cell development and maturation is critical to our understanding of β-cell biology and efforts to counteract the degradation of β-cell identity in addition to mechanisms that occurs during physiological stress and diabetes (Weir and Bonner-Weir 2004; Chakravarthy et al. 2017). Understanding β-cell development and maturation is also vital to the advancement of directed β-cell differentiation protocols, which aim to artificially recapitulate such processes in vitro.
Most of our understanding of β-cell differentiation comes from rodent studies, although recent work has shown that the rodent and human endocrine compartments diverge at certain stages of development (Larsen and Grapin-Botton 2017). In rodents, endocrine differentiation occurs at two stages of embryonic development: the primary transition (E9.0–E12.5), where the main differentiated endocrine cells are α-cells, and secondary transition (E12.5–E15.5), at which point other endocrine cell types, including β-cells, are formed (Pan and Wright 2011; Shih et al. 2013; Nair and Hebrok 2015; Bastidas-Ponce et al. 2017). Human endocrine development is thought to exclusively entail the secondary transition, which gives rise to all hormone-secreting endocrine cells (Jennings et al. 2013); however, it is known that all endocrine cell types arise from a common progenitor pool in both rodent and human. Endocrine development begins with the emergence of endocrine progenitors lowly expressing the transcription factor Neurogenin-3 (NGN3low). NGN3 is indispensable for endocrine cell formation; an increase in NGN3 expression generates NGN3high endocrine precursors. From these precursors arises cells positive for the transcription factor Fev, which differentiate to form endocrine cells that express hormones and possess the requisite secretory machinery (Wang et al. 2010; Bechard et al. 2016; Byrnes et al. 2018). At this stage, multiple transcription factors begin to drive the establishment of terminal endocrine cell identities. Expression of ARX favors α-cells; PAX4 favors the formation of β- and δ-cells. Adoption of the β-cell fate additionally depends on the expression of several other transcription factors, such as FOXA2, PDX1, NKX6.1, NEUROD1, NKX2.2, and MNX1 (Arda et al. 2013).
All β-cells express certain genes that are required for the establishment and maintenance of β-cell identity at birth, including NKX6.1, PDX1, and NEUROD1. The initial postnatal maturation phase is largely characterized by the induction of additional β-cell markers and an increase in endocrine cell mass due to the highly, although progressively declining, proliferative nature of immature β-cells (Blum et al. 2012). The Maf family, in particular, is important for the establishment of β-cell identity and functionality (Hang and Stein 2011); in rodents, the switch from MafB to MafA expression heralds β-cell maturation, a clear distinction to the human situation in which MAFB expression is retained in mature β-cells (Arda et al. 2016; Cyphert et al. 2019; Russell et al. 2020). During this stage, β-cells further demonstrate an increase in Urocortin3 (UCN3) and SYT4 of the Synaptotagmin (Syt) family, with a concurrent reduction in Neuropeptide Y (NPY) (Blum et al. 2012; van der Meulen et al. 2012; Rodnoi et al. 2017). During the second phase of maturation, β-cells shift from relying predominantly on glycolytic metabolism to favoring aerobic respiration and mitochondrial metabolism, a change that underlies functional maturation (Bonner-Weir et al. 2016). Insulin processing, glucose sensing, and granule exocytosis genes such as GLUT2, GLP-1 receptor, prohormone convertase-1/3, and pyruvate carboxylase are up-regulated (Artner et al. 2007; Nishimura et al. 2015). Intriguingly, the surface markers ST8SIA1 and CD9 additionally delineate four antigenic subgroups of β-cells in human islets upon maturation. These subgroups demonstrate differential expression of genes involved in insulin secretion, such as GLUT2, PPP1R1A, SUR2, and G6PC2, but similarly express primary β-cell markers (Dorrell et al. 2016). This finding reflects our growing understanding of transcriptional and functional heterogeneity within the β-cell population.
FUNCTIONAL HETEROGENEITY OF β-CELLS
β-cells exhibit heterogeneous properties both within the individual islets and between islets localized to different areas of the pancreas (Dorrell et al. 2016). Single-cell transcriptomic and proteomic data, as well as experimental studies, suggest that this heterogeneity encompasses both gene expression and β-cell function, including processes such as glucose sensing, intracellular calcium signaling, metabolism, insulin biosynthesis, and insulin secretion (Pipeleers et al. 1994; van der Meulen et al. 2017; Mawla and Huising 2019). From these differences arise a diverse, hierarchical network of β-cells, all contributing to the maintenance of glucose homeostasis (Johnston et al. 2016; Salem et al. 2019).
Recent evidence from rodents indicates the presence of a subpopulation of β-cells known as “hub cells,” which demonstrate increased expression of glucokinase and glucose oxidation genes, hyperpolarized mitochondria, reduced PDX1 and NKX6.1, and higher metabolic activity despite fewer insulin granules. These cells have been proposed to make connections with, and control the activity of, other “follower” β-cells (Johnston et al. 2016; Xin et al. 2016; Salem et al. 2019). Although these cells have not yet been identified in human islets, the identification and characterization of hub cells reveals a novel mechanism for concerted insulin secretion across β-cells within rodent islets. These hub cells constitute 1%–10% of islet surface, display many electrically coupled cellular connections, and exhibit accelerated and sustained Ca2+ signaling upon glucose stimulation, which precedes that of the “follower” cells. Hub-cell inactivation by photoablation performed in zebra fish islets disrupted the synchronization of Ca2+ signaling across the islet, whereas similar perturbation had no effect in follower cells. Hub cells may therefore serve as “pacemakers” to synchronize glucose-stimulated Ca2+ signaling and insulin granule exocytosis across networks of follower cells throughout the islet. The difference in the glucose responsiveness between the hub and follower cells may be due to varying levels of glucose uptake, unknown mechanisms of Ca2+ signal synchronization, or changes in islet architecture upon isolation. Interestingly, hub cells seem to largely escape immune destruction in type 1 diabetes (Liu and Hebrok 2017; Oram et al. 2019).
Emerging from these studies is the concept that the function of individual β-cells is determined by intercellular and intracellular signaling events, with a subset of β-cells more extensively involved in the coordination of insulin secretion necessary for ensuring that the islet functions as an integrated unit, despite the diversity of cell types that comprise it. Loss of these interactions has been shown to perturb glucose stimulation threshold and the kinetics of insulin secretion; furthermore, altered distribution of islet cell populations has been implicated as a hallmark of obesity, aging, and type 2 diabetes (Dorrell et al. 2016). However, several questions regarding β-cell diversity remain unanswered, including whether they arise from subtle differences in developmental programs or settle into subpopulations at later stages of maturation, where they are localized in the 3D islet architecture, how that localization relates to their function, and, finally, whether subpopulations that are fluent with cells within one population can assume properties of the others (Liu and Hebrok 2017).
PANCREAS REGENERATION
Defining and replicating the regenerative properties of the pancreas has been a tantalizing goal. The different cellular components of the pancreas, exocrine and endocrine, display different regenerative potentials (Zhou and Melton 2018). Furthermore, regenerative capacity is linked to the particular damage inflicted upon the organ.
Regeneration of Exocrine Pancreas
Acinar cell regeneration has been studied in detail upon treatment with caerulein, a human cholecystokinin analog, that results in pancreatitis-like phenotypes and a transient dedifferentiation of acinar cells toward a duct-like identity. Short-term caerulein treatment appears to be completely reversible within several days (Jensen et al. 2005; Morris et al. 2010). The regenerative process involves transient phases of inflammation, metaplasia, and redifferentiation. Two distinct modes of regeneration have been proposed to occur in models of pancreatitis (Murtaugh and Keefe 2015): the generation of new acinar cells from preexisting acinar cells (Desai et al. 2007; Strobel et al. 2007; Blaine et al. 2010) or the redifferentiation of degranulated and duct-like acinar cells toward their normal functional state. The study of acinar redifferentiation has led to the identification of proinflammatory and developmentally regulated genes that guide this process. Deletion of key components of the Hedgehog, Notch, and Wnt pathways as well as NR5A2 and PTF1A transcription factors from acinar cells severely disrupts exocrine regeneration (David et al. 2005; Fendrich et al. 2008; Siveke et al. 2008; Morris et al. 2010; Stanger and Hebrok 2013; Von Figura et al. 2014; Hoang et al. 2016). Of note, the regenerative process is highjacked in the presence of oncogenic Kras, the predominant oncogene in pancreatic ductal adenocarcinoma (Morris et al. 2010).
Regeneration of the Endocrine Pancreas
Extensive research has been conducted to determine the extent to which endocrine cells within the pancreas can regenerate upon injury. Early islet regeneration studies relied on rodent injury models, including pancreatectomy, pancreatic duct ligation, and chemical ablation of β-cells with toxins streptozotocin (STZ) or alloxan. In particular, pancreatic duct ligation has been used extensively to demonstrate regenerative potential of acinar and insulin-producing β-cells in rodents (Lehv and Fitzgerald 1968; Bonner Weir et al. 1983). With regard to β-cell regeneration, early studies implicated pancreatic duct cells as progenitors for endocrine cells (Bonner-Weir et al. 1993). Follow-up studies, however, supported the notion that existing β-cells, and not pancreatic duct cells, serve as progenitors for regenerating β-cells after pancreatic duct ligation (Solar et al. 2009).
Pancreatic β-cell ablation using STZ, a chemical toxin that structurally mimics glucose and is imported into the β-cells through GLUT2 receptors (Szkudelski 2001; Graham et al. 2011), or alloxan, a selective inhibitor of glucokinase, have been extensively used to ablate rodent β-cells (Lenzen 2008). Depending on drug dosage, the entire β-cell mass can be partially or nearly completely ablated in a few days. Extensive studies have found no convincing evidence for extensive β-cell regeneration in adult animals following chemical ablation (Rankin and Kushner 2009; Tschen et al. 2009). However, β-cells in young rodents have high proliferative capacity that declines rapidly with age (Byrne et al. 1995; Montanya et al. 2000; Teta et al. 2005). β-cell replacement has been observed in young animals, but the source of these new β-cells after injury was identified as replication from preexisting β-cells (Dor et al. 2004). In humans, removal of 50% of the pancreas leads to insulin-dependent diabetes (Kumar et al. 2008; Menge et al. 2008). Although juvenile human pancreas offers some regeneration after pancreatectomy, this capacity rapidly declines with age and appears completely absent in adult humans (Berrocal et al. 2005; Menge et al. 2008; Rankin and Kushner 2009).
More recently, transdifferentiation of α- or δ-cells into β-cells has been reported upon diphtheria toxin-based β-cell ablation in mice (Thorel et al. 2010; Chera et al. 2014). The molecular mechanism of this conversion between islet cell types has yet to be uncovered, and the extent to which new β-cells form from their endocrine relatives appears to be rather small. Furthermore, replicative capacity of adult human β-cells is still a matter of debate; however, islet hyperplasia has been reported during pregnancy, stimulated by placental lactogen and prolactin hormones (Kalousova et al. 2010; Kim et al. 2010; Ohara-Imaizumi et al. 2013). High-fat diet-induced obesity and insulin resistance condition also leads to increase in β-cell mass (Rieck and Kaestner 2010; Mezza and Kulkarni 2014; Wang et al. 2015a,b; Saunders and Powers 2016). The molecular pathways that drive these increases in β-cell mass in obesity and insulin resistance have yet to be fully elucidated. Studies using insulinoma samples have implicated the dual-specificity tyrosine-regulated kinase-1a (DYRK1A) and its inhibitor Harmine (Wang et al. 2014) as novel regulators of human β-cell proliferation. Considering that Harmine elicits proliferative responses in other cells, combining it with glucagon-like peptide receptor agonists allows for more specifically targeting β-cells to induce proliferation (Ackeifi et al. 2020). Additional studies will be necessary to further refine safe stimulation of endogenous human β-cell expansion.
β-CELL DEDIFFERENTIATION, REDIFFERENTIATION, AND TRANS-DIFFERENTIATION
In addition to the presence of defined β-cell subpopulations, emerging evidence indicates the propensity for insulin-producing cells to lose or regain their differentiation state.
Dedifferentiation
Dedifferentiation may be defined as the loss of the mature β-cell transcriptional profile (Puri et al. 2015) and resulting functional decline. Unlike many other terminally differentiated cell types, β-cells are plastic and may undergo de-differentiation under certain pathophysiological conditions, such as chronic hyperglycemia, obesity, and aging (Kayali et al. 2007; Dhawan et al. 2011; Puri et al. 2013; John et al. 2018). Maintaining the β-cell identity requires the induction and repression of genes characteristic of mature and immature β-cells, respectively. Dedifferentiation entails the down-regulation of canonical β-cell maturity genes, such as PDX1, NKX6.1, MAFA, MAFB, and GLUT2, as well as genes involved in mitochondrial metabolism and insulin biosynthesis and secretion. Conversely, dedifferentiated β-cells exhibit increased levels of “disallowed” genes expressed in immature β- and endocrine progenitor cells, including NGN3, LDHA, MCT4, and SOX9. These transcriptional changes are accompanied by a loss in β-cell mass, which is attributable specifically to β-cell dedifferentiation, rather than increased apoptosis (Talchai et al. 2012).
Single-cell RNA sequencing of human islets has revealed that islet cells can dedifferentiate ex vivo; furthermore, β-cells from type 2 diabetic patients have been found to adopt the gene expression profile of endocrine progenitors, which is suggestive of dedifferentiation. (Talchai et al. 2012; Guo et al. 2013a; Brereton et al. 2014; Wang et al. 2014; Cinti et al. 2016; Jacobson and Liu 2016; Dirice et al. 2019). Although the precise mechanism of β-cells dedifferentiation remains unknown, potential players include hypoxia, oxidative stress and reactive oxygen species (ROS) accumulation, and inflammation-induced endoplasmic reticulum (ER) stress (Sherry et al. 2006; Puri et al. 2009, 2013; Hasnain et al. 2014; Nordmann et al. 2017).
Redifferentiation
Prior to the advent of stem cell–derived β-cells, one potential strategy for overcoming the shortage of cadaveric donor islets was in vitro expansion of human β-cells (Bar et al. 2012). However, this process resulted in β-cell dedifferentiation and epithelial-to-mesenchymal transition, likely due to the loss of islet architecture and the induction of nonphysiological proliferation (Puri et al. 2018). Subsequent research demonstrated that human β-cells could be partially redifferentiated in vitro postexpansion through a variety of methods, including inhibition of the Notch signaling pathway, overexpression of miR-375, or culture within specialized 3D hydrogel systems (Bar et al. 2012; Nathan et al. 2015; Aloy-Reverté et al. 2018). Despite this, the use of human β-cells expanded in vitro soon lost appeal as a therapeutic option due to issues with efficiency, propensity for repeated dedifferentiation, and lack of evidence of functionality (Baeyens et al. 2018; Brovkina and Dashinimaev 2020).
Trans-Differentiation of β-Cells from α- and Exocrine Cells
β-cell trans-differentiation, or the direct interconversion between β-cells and other cell types, is thought to represent a potential therapeutic strategy for diabetes (Bouwens et al. 2013; Aguayo-Mazzucato and Bonner-Weir 2018). However, efforts to take advantage of this phenomenon have been hampered by a number of scientific and logistical concerns, including the lack of a clear understanding of pancreatic plasticity, insufficient characterization of trans-differentiated cells, and difficulty of inducing cellular trans-differentiation with spatial and temporal precision.
α- to β-Cell
Interconversion between α- and β-cells has generated particular excitement due to their derivation from the same lineage, localization within the islet, overlap in a number of endocrine and secretory genes, and relative abundance in type 1 diabetic pancreata. Given that these cell types originate from a common endocrine progenitor cell, two theories regarding the broad mechanism of trans-differentiation have evolved: first, that glucagon-positive α-cells, at least in humans, become insulin-positive β-cells through an intermediate, double hormone-positive stage; and second, that α-cells revert back into endocrine progenitors, which then proceed to adopt the β-cell phenotype (van der Meulen and Huising 2015). Trans-differentiation has been shown to occur between pancreatic α- and β-cells upon damage to the pancreas and ablation of endogenous β-cells, as well as through various genetic manipulations. Depletion of rodent β-cells through acute treatment with diphtheria toxin, as well as pancreatic ductal ligation combined with alloxan-induced β-cell ablation, both result in β-cell neogenesis from α-cells, as confirmed by lineage tracing (Chung et al. 2010; Thorel et al. 2010). Genetically engineered mouse models have revealed that ectopic overexpression of the early β-cell transcription factor PAX4, or knockout of the α-cell transcription factor ARX, have each been shown to promote α- to β-cell trans-differentiation (Collombat et al. 2010; Courtney et al. 2013). Promising research has shown that endogenous rodent α-cells can be reprogrammed into β-cells and used to cure autoimmune diabetes through viral gene therapy and overexpression of PDX1 and MAFA (Xiao et al. 2018). Furthermore, Tang and colleagues recently demonstrated that reversal of diabetic autoimmunity through induction of haploidentical mixed chimerism, followed by administration of gastrin and epidermal growth factor (EGF), can result in rodent β-cell neogenesis via α- to β-cell trans-differentiation (Tang et al. 2020). This trans-differentiation, combined with reactivation of insulin-low dysfunctional β-cells, was able to achieve normoglycemia in mice models of type 1 diabetes.
Exocrine to β-Cell
Pancreatic exocrine cells have demonstrated a degree of plasticity and the ability to adopt β-cell characteristics under certain conditions. Early experiments demonstrated that rodent pancreatic exocrine cells could be isolated and cultured in vitro with leukemia inhibitory factor (LIF) and EGF to generate insulin-producing cells (Baeyens et al. 2005); reprogramming of endogenous rodent exocrine cells in vivo was later achieved through the adenoviral delivery and induction of one or more key β-cell transcription factors NGN3, PDX1, and MAFA (Zhou et al. 2008; Miyazaki et al. 2021). These and subsequent studies have found that pancreatic acinar, not ductal cells, were the predominant targets for exocrine- to β-cell trans-differentiation in both experimental models and type 1 diabetic patients (Masini et al. 2017). Recent research demonstrating that culturing human acinar and ductal cells together in 3D cultures known as “pancreatospheres” results in acinar, but not ductal cells; adopting a progenitor-like transcriptional profile may offer a potential explanation for the discrepancy in trans-differentiation capacity between these two cell types (Baldan et al. 2019). Whereas ductal cells have been shown to potentially possess some measure of trans-differentiation potential (Yatoh et al. 2007), the increased efficacy of acinar- to β-cell trans-differentiation and surplus of acinar cells within the human pancreas have resulted in ductal cells largely being sidelined as potential trans-differentiation targets.
Hepatocyte to β-Cell
Research efforts have additionally focused on hepatocytes as a potential source for β-cell trans-differentiation. Hepatocytes can be harvested directly from type 1 diabetic patients, thereby bypassing the issue of immune rejection; accordingly, these cells represent particularly appealing prospects for expansion and trans-differentiation. Ectopic expression of PDX1 in both rodent liver in vivo and human hepatocytes in vitro results in up-regulation of critical β-cell markers, such as the transcription factors NKX6.1 and NKX2.2, as well as genes critical for insulin biosynthesis and processing, such as INS1, INS2, and PC1/3 (Ferber et al. 2000; Sapir et al. 2005). The efficiency of this process was found to be improved treatment with the GLP-1R agonist extendin 4 or the growth factor PGDF (Aviv et al. 2009; Chang et al. 2016). However, there remain significant hurdles to the production of mature β-cells from hepatocytes, including the use of viral vectors for delivery and induction of ectopic PDX1, the propensity for transdifferentiated cells to express pancreatic progenitor or exocrine markers, and relatively low GSIS. Furthermore, the capacity for trans-differentiation has been found to be restricted to a small population of trans-differentiation-competent hepatocytes enriched for Wnt signaling (Cohen et al. 2018).
GENERATION OF HUMAN STEM CELL–DERIVED β-CELLS IN VITRO
Transplantation of cadaveric human islets has long been used as a treatment strategy for type 1 diabetes. Despite its success, this procedure comes with numerous challenges, including a limited supply of donor islets, engraftment failure, and immune rejection. The last several decades have therefore seen significant efforts to establish protocols for generation of functional β-cells from human pluripotent stem cells (hPSCs) for the dual purposes of replacing cadaveric islets and serving as platforms to interrogate β-cell biology in human cells. The majority of this work was performed using human embryonic stem cells (hESCs); however, the use of induced pluripotent stem cells (iPSCs), derived from adult somatic cells, has become increasingly common.
Initial attempts to generate insulin-producing cells relied on the spontaneous differentiation of hESCs enriched for Nestin, an intermediate filament protein expressed in neural stem cells and pancreatic progenitors (Hunziker and Stein 2000; Assady et al. 2001). However, the cells resulting from these protocols proved incapable of insulin biosynthesis (Rajagopal et al. 2003) Attention therefore turned toward the establishment of directed differentiation protocols based on known principles of rodent pancreatic development. Selective introduction of growth factors, agonists, and small-molecule inhibitors facilitated the stepwise progression of hESCs through definitive endoderm, to pancreatic and endocrine progenitors, ultimately resulting in the generation of insulin-producing β-cells (D'Amour et al. 2005; Guo et al. 2013b; Rezania et al. 2013, 2014; Pagliuca et al. 2014; Russ et al. 2015). The cells yielded by these protocols largely resembled immature β-cells, as characterized by their inability to engage in GSIS.
Recent work has led to the development of novel protocols for the purpose of driving further maturation of these immature β-cells, thereby allowing them to better recapitulate those found in human islets. Isolation, purification, and reaggregation of immature β-cells has been found to produce enriched clusters of insulin-positive β-cells that strongly resemble human islets in terms of transcriptional profile, dynamic GSIS, Ca2+ signaling, mitochondrial metabolism, and glucose regulation upon transplantation into diabetic mice (Nair et al. 2019). Further strategies for improving hPSC-derived β-cells have focused on modulation of signaling pathways such as transforming growth factor β (TGF-β), metabolic augmentation, and purification of distinct endodermal progenitor populations prior to differentiation (Velazco-Cruz et al. 2019; Davis et al. 2020; Mahaddalkar et al. 2020).
Despite significant improvements in the production and transplantation of hPSC-derived β-cells, autoimmune rejection continues to pose a threat to their engraftment and survival. Immune rejection may be avoided by generating β-cells from patient-specific iPSCs, but such an approach is prohibitively difficult to scale and economically unviable. Under consideration are biobanks containing a limited number of iPSC lines with human leukocyte antigen (HLA) types that match the majority of potential recipients within specific ethnic populations (Nakatsuji et al. 2008; Gourraud et al. 2012; Taylor et al. 2012). Unfortunately, this strategy fails to ameliorate concerns regarding autoimmune rejection in patients with type 1 diabetes. For that reason, gene-editing strategies for immunomodulation of hPSC-derived β-cells are currently under active investigation. One such method, referred to as “immune cloaking,” entails removing the MHC class I genes (HLA) that control presentation of auto- and allo-antigens to provoke an immune response (Fig. 2; van der Torren et al. 2017; Deuse et al. 2019; Han et al. 2019). Deletion of all HLA proteins could further confer total immune protection; it was recently shown that abolishment of HLA-A, HLA-B, and partial HLA-C expression through deletion of β-2-microglobulin, paired with continued expression of HLA E/G, conferred hypo-immunogenicity and allowed hPSC-derived β-cells to evade T-cell attack (Xu et al. 2019). Similar immune evasion strategies include T-cell inactivation and subsequent prevention of β-cell loss via β-cell-specific overexpression of PDL1-CTLA4Ig molecules (El Khatib et al. 2015). Importantly, reducing immunogenicity of transplanted hPSC-derived β-cells also increases the probability of neoplastic growths and pathogenic infections. This issue may be tackled through the use of genetic “kill switches,” as discussed below.
Figure 2.
Advances in β-cell therapy. (1) Designer β-cells with synthetic gene circuits for precise control of developmental, metabolic, and/or signaling pathways. (2) Immunomodulation approaches to generate β-cells invisible to the host immune system. (3) Bioprocessing technologies for scalable manufacture of β-cells. (4) Use of patient-specific iPS cells for generation of β-cells for allogenic transplantation. (5) Encapsulation and scaffold-based β-cell devices for delivery, immune suppression, and drug delivery. (6) Recapitulating the minimal β-cell machinery for insulin release with artificial β-cells. (HLA) Human leukocyte antigen.
ADVANCES IN β-CELL TECHNOLOGIES
Research is progressively focusing on the generation of stem cell–derived β-cells as a viable large-scale treatment option for type 1 diabetes. Despite significant advancements in β-cell differentiation protocols, the requirement for multiple complex media formulations and the proven establishment of heterogeneous end-stage cell populations continues to render the process both challenging and inconsistent. Furthermore, the propensity for variability across differentiations, threat of autoimmune graft rejection, and safety risks inherent to stem cell transplantation pose significant hurdles to their use as a therapeutic strategy. To address these concerns, multiple synthetic biology and technological approaches, including the creation of synthetic gene networks for lineage control, genetic “kill switches” to bolster transplant safety, completely artificial β-cells, and β-cell encapsulation devices are currently under development.
Programmable, Synthetic Gene Circuits, and Lineage Control Networks
Synthetic gene circuits are designed to facilitate dynamic gene expression through the coupling of signaling cascades and transcription factor–based gene switches with reversible and differential sensitivity to chemical compounds (Fig. 2). Creating synthetic gene circuits involves assembling multicomponent genetic constructs specifically designed to reprogram cells for the purpose of enacting a lineage specific developmental program and, potentially, enable therapeutic function. These gene circuits create “smarter” cells and allow for the development of gene therapies with increased efficacy, precision, and control. Significant efforts are currently underway to use synthetic gene circuits to create functional, pre-programmed β-cell differentiation protocols that coordinate the timely induction and repression of multiple cell fate regulators at various stages of pancreatic development. Zhou et al. (2008) and Ariyachet et al. (2016) have generated a synthetic lineage-control network with mutually exclusive expression switches for the transcription factors NGN3, PDX1, and MAFA to drive differentiation of pancreatic progenitor cells into β-like cells. Saxena et al. (2016) designed a genetic circuit triggered by the common food additive vanillic acid and characterized by time delay and feedforward amplifier properties. In their system, this allowed for timely and controlled expression and repression of NGN3, PDX1, and MAFA. Such synthetic lineage control networks have an advantage of using the endogenous pathways rather than external signaling inputs as well as more precise feedback.
Genetic Kill Switches
Whereas hPSC-derived β-cells hold great promise for treatment of type 1 diabetes, safety concerns surrounding their introduction to an uncontrolled physiological environment remain. One such risk is the indiscriminate growth of uncommitted progenitors, leading to the formation of teratomas. These concerns necessitate the incorporation of a genetic safety mechanism deemed a “kill” or “suicide” switch that can be activated to selectively eliminate aberrant or malfunctioning transplanted cells (Bonini et al. 2007). Current mechanisms designed to achieve this targeted cellular ablation include the CD20-based complementation system, the ganciclovir-dependent herpes simplex virus thymidine kinase (HSV-TK) system (Jones et al. 2014), and drug-inducible Cas9 suicide gene cassettes (Yagyu et al. 2015). It is important to note that these approaches entail off-target effects and may fall prey to the development of drug resistance. The latter concern may potentially be mitigated by the development of a dual kill switch based on the HSV-TK model and other genes such as the cell-cycle regulator CDK1 (Kotini et al. 2016; Liang et al. 2018). A microRNA-based system reliant upon microRNAs expressed in undifferentiated cells, such as Let7, is also under further clinical investigation (Qadir et al. 2019).
β-Cell Devices
The use of β-cell encapsulation devices has emerged as a promising delivery mechanism and a potential strategy for overcoming the requirement for immunosuppressants upon β-cell transplantation (Sneddon et al. 2018). Extensive efforts have focused on the generation of ideal approaches to cell encapsulation, including optimization of encapsulation material and topography, transplantation site, configuration to improve vascularization, and immune modulation (Fig. 2). Current strategies include cellular macro-encapsulation in devices made of polymers such as polytetrafluoroethylene or polycaprolactone, or microencapsulation in materials including alginate, polyacrylate, or collagen. These biocompatible encapsulation devices are designed to provide an ideal environment for optimal β-cell function, sequestering the immune response and providing vascularization and effective nutrient and waste exchange (Scharp and Marchetti 2014). Also under investigation are drug- and cytokine-releasing scaffolds, as well as procedures involving cotransplantation with mesenchymal stem cells (Desai and Shea 2017).
Artificial β-Cells
Artificial β-cells are biomimetic assemblies designed to recapitulate key β-cell functions (Fig. 2). They possess a vesicle superstructure—notably, an outer large vesicle mimicking the plasma membrane paired with small, inner liposomal vesicles loaded with insulin (Chen et al. 2018). These synthetic bodies are equipped with glucose-responsive metabolic systems and pH-sensitive membrane-fusion machinery to facilitate glucose sensing, signal transduction, and insulin release through vesicle fusion. The reversible nature of pH-sensitive polyethylene glycol (PEG) attachment and detachment causes random collision of the small, liposomal insulin vesicles with the outer vesicle plasma membrane, thereby leading to sustained, dynamic insulin release in response to glucose stimulation. The primary advantage of artificial β-cells is that they can either be encapsulated within an injectable gel and transplanted directly or delivered through transcutaneous micro-needle patches, thereby avoiding the requirement for immunosuppressive drugs. Further advancements will likely include improvements in the sensor structure to increase longevity and decrease sensor delay.
SUMMARY
Loss of functional β-cells is the central event leading to type 1 and type 2 diabetes mellitus. According to the World Health Organization (WHO) report, the number of people with diabetes rose from 108 million in 1980 to 422 million in 2014 while its prevalence has been rising more rapidly in low- and middle-income countries leading to premature deaths as well as several other comorbid conditions. Understanding β-cell development and function is key to our efforts in devising strategies in combating diabetes. Over the past decade, research focus on the pathogenesis of diabetes mellitus has shifted from animal models to morphological and functional studies on human islet cells. The tremendous strides made in the fields of pancreatic and β-cell biology discussed in this review—islet architecture and communication, β-cell development, and heterogeneity, and various forms of cellular differentiation—have positioned us to both understand this disease to a greater degree than ever before and develop innovative new strategies, such as in vitro differentiation of hPSCs into β-cells and increasingly advanced β-cell technologies for tackling and circumventing it. These studies have helped us in developing novel therapies to prevent β-cell death or to restore lost β-cell mass in advanced disease. While much remains to be done, through these research efforts we may begin to imagine a society in which diabetes is treated with far more efficacy than previously achieved—and, quite possibly, cured entirely.
COMPETING INTEREST STATEMENT
M.H. is on the SAB of Encellin Inc. and Thymmune Therapeutics Inc., holds stocks in Encellin Inc., Thymmune Therapeutics Inc., and Viacyte Inc., and has received research support from Eli Lilly. He is the co-founder, SAB member of Minutia Inc., and EndoCrine, and holds stocks and options in both companies.
ACKNOWLEDGMENTS
We thank the members of the Hebrok laboratory for helpful discussions.
Footnotes
Editors: Kenneth D. Poss and Donald T. Fox
Additional Perspectives on Regeneration available at www.cshperspectives.org
REFERENCES
- Ackeifi C, Wang P, Karakose E, Manning Fox JE, González BJ, Liu H, Wilson J, Swartz E, Berrouet C, Li Y, et al. 2020. GLP-1 receptor agonists synergize with DYRK1A inhibitors to potentiate functional human β cell regeneration. Sci Transl Med 12: aeew996. 10.1126/scitranslmed.aaw9996 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Aguayo-Mazzucato C, Bonner-Weir S. 2018. Pancreatic β cell regeneration as a possible therapy for diabetes. Cell Metab 27: 57–67. 10.1016/j.cmet.2017.08.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ahlqvist E, Prasad RB, Groop L. 2020. Subtypes of type 2 diabetes determined from clinical parameters. Diabetes 69: 2086–2093. 10.2337/dbi20-0001 [DOI] [PubMed] [Google Scholar]
- Aloy-Reverté C, Moreno-Amador JL, Nacher M, Montanya E, Semino CE. 2018. Use of RGD-functionalized sandwich cultures to promote redifferentiation of human pancreatic β cells after in vitro expansion. Tissue Eng Part A 24: 394–406. 10.1089/ten.tea.2016.0493 [DOI] [PubMed] [Google Scholar]
- Arda HE, Benitez CM, Kim SK. 2013. Gene regulatory networks governing pancreas development. Dev Cell 25: 5–13. 10.1016/j.devcel.2013.03.016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Arda HE, Li L, Tsai J, Torre EA, Rosli Y, Peiris H, Spitale RC, Dai C, Gu X, Qu K, et al. 2016. Age-dependent pancreatic gene regulation reveals mechanisms governing human β cell function. Cell Metab 23: 909–920. 10.1016/j.cmet.2016.04.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ariyachet C, Tovaglieri A, Xiang G, Lu J, Shah MS, Richmond CA, Verbeke C, Melton DA, Stanger BZ, Mooney D, et al. 2016. Reprogrammed stomach tissue as a renewable source of functional β cells for blood glucose regulation. Cell Stem Cell 18: 410–421. 10.1016/j.stem.2016.01.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Artner I, Blanchi B, Raum JC, Guo M, Kaneko T, Cordes S, Sieweke M, Stein R. 2007. MafB is required for islet β cell maturation. Proc Natl Acad Sci 104: 3853–3858. 10.1073/pnas.0700013104 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Assady S, Maor G, Amit M, Itskovitz-Eldor J, Skorecki K, Tzukerman M. 2000. Insulin production by human embryonic stem cells. Diabetes 50: 1691–1697. 10.2337/diabetes.50.8.1691 [DOI] [PubMed] [Google Scholar]
- Aviv V, Meivar-Levy I, Rachmut IH, Rubinek T, Mor E, Ferber S. 2009. Exendin-4 promotes liver cell proliferation and enhances the PDX-1-induced liver to pancreas transdifferentiation process. J Biol Chem 284: 33509–33520. 10.1074/jbc.M109.017608 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Baeyens L, De Breuck S, Lardon J, Mfopou JK, Rooman I, Bouwens L. 2005. In vitro generation of insulin-producing β cells from adult exocrine pancreatic cells. Diabetologia 48: 49–57. 10.1007/s00125-004-1606-1 [DOI] [PubMed] [Google Scholar]
- Baeyens L, Lemper M, Staels W, De Groef S, De Leu N, Heremans Y, German MS, Heimberg H. 2018. (Re)generating human β cells: status, pitfalls, and perspectives. Physiol Rev 98: 1143–1167. 10.1152/physrev.00034.2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Baldan J, Houbracken I, Rooman I, Bouwens L. 2019. Adult human pancreatic acinar cells dedifferentiate into an embryonic progenitor-like state in 3D suspension culture. Sci Rep 9: 1–12. 10.1038/s41598-019-40481-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ballian N, Brunicardi FC. 2007. Islet vasculature as a regulator of endocrine pancreas function. World J Surg 31: 705–714. 10.1007/s00268-006-0719-8 [DOI] [PubMed] [Google Scholar]
- Bar Y, Russ HA, Sintov E, Anker-Kitai L, Knoller S, Efrat S. 2012. Redifferentiation of expanded human pancreatic β-cell-derived cells by inhibition of the NOTCH pathway. J Biol Chem 287: 17269–17280. 10.1074/jbc.M111.319152 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bastidas-Ponce A, Scheibner K, Lickert H, Bakhti M. 2017. Cellular and molecular mechanisms coordinating pancreas development. Development 144: 2873–2888. 10.1242/dev.140756 [DOI] [PubMed] [Google Scholar]
- Bechard ME, Bankaitis ED, Hipkens SB, Ustione A, Piston DW, Yang YP, Magnuson MA, Wright CVE. 2016. Precommitment low-level neurog3 expression defines a long-lived mitotic endocrine-biased progenitor pool that drives production of endocrine-committed cells. Genes Dev 30: 1852–1865. 10.1101/gad.284729.116 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Benninger RKP, Head WS, Zhang M, Satin LS, Piston DW. 2011. Gap junctions and other mechanisms of cell–cell communication regulate basal insulin secretion in the pancreatic islet. J Physiol 589: 5453–5466. 10.1113/jphysiol.2011.218909 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Berrocal T, Pajares MP, Zubillaga AF. 2005. Pancreatic cystosis in children and young adults with cystic fibrosis: sonographic, CT, and MRI findings. Am J Roentgenol 184: 1305–1309. 10.2214/ajr.184.4.01841305 [DOI] [PubMed] [Google Scholar]
- Blaine SA, Ray KC, Anunobi R, Gannon MA, Washington MK, Means AL. 2010. Adult pancreatic acinar cells give rise to ducts but not endocrine cells in response to growth factor signaling. Development 137: 2289–2296. 10.1242/dev.048421 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Blum B, Hrvatin S, Schuetz C, Bonal C, Rezania A, Melton DA. 2012. Functional β-cell maturation is marked by an increased glucose threshold and by expression of urocortin 3. Nat Biotechnol 30: 261–264. 10.1038/nbt.2141 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bonini C, Bondanza A, Perna SK, Kaneko S, Traversari C, Ciceri F, Bordignon C. 2007. The suicide gene therapy challenge: how to improve a successful gene therapy approach. Mol Ther 15: 1248–1252. 10.1038/sj.mt.6300190 [DOI] [PubMed] [Google Scholar]
- Bonner-Weir S, Orci L. 1982. New perspectives on the microvasculature of the islets of Langerhans in the rat. Diabetes 31: 883–889. 10.2337/diab.31.10.883 [DOI] [PubMed] [Google Scholar]
- Bonner Weir S, Trent DF, Weir GC. 1983. Partial pancreatectomy in the rat and subsequent defect in glucose-induced insulin release. J Clin Invest 71: 1544–1553. 10.1172/JCI110910 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bonner-Weir S, Baxter L, Schuppin G, Smith F. 1993. A second pathway for regeneration of adult exocrine and endocrine pancreas. A possible recapitulation of embryonic development. Diabetes 42: 1715–1720. 10.2337/diab.42.12.1715 [DOI] [PubMed] [Google Scholar]
- Bonner-Weir S, Sullivan BA, Weir GC. 2015. Human islet morphology revisited: human and rodent islets are not so different after all. J Histochem Cytochem 63: 604–612. 10.1369/0022155415570969 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bonner-Weir S, Aguayo-Mazzucato C, Weir GC. 2016. Dynamic development of the pancreas from birth to adulthood. Ups J Med Sci 121: 155–158. 10.3109/03009734.2016.1154906 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Borden P, Houtz J, Leach SD, Kuruvilla R. 2013. Sympathetic innervation during development is necessary for pancreatic islet architecture and functional maturation. Cell Rep 4: 287–301. 10.1016/j.celrep.2013.06.019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bouwens L, Houbracken I, Mfopou JK. 2013. The use of stem cells for pancreatic regeneration in diabetes mellitus. Nat Rev Endocrinol 9: 598–606. 10.1038/nrendo.2013.145 [DOI] [PubMed] [Google Scholar]
- Brereton MF, Iberl M, Shimomura K, Zhang Q, Adriaenssens AE, Proks P, Spiliotis II, Dace W, Mattis KK, Ramracheya R, et al. 2014. Reversible changes in pancreatic islet structure and function produced by elevated blood glucose. Nat Commun 5: 4639. 10.1038/ncomms5639 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brissova M, Fowler M, Wiebe P, Shostak A, Shiota M, Radhika A, Lin PC, Gannon M, Powers AC. 2004. Intraislet endothelial cells contribute to revascularization of transplanted pancreatic islets. Diabetes 53: 1318–1325. 10.2337/diabetes.53.5.1318 [DOI] [PubMed] [Google Scholar]
- Brissova M, Fowler MJ, Nicholson WE, Chu A, Hirshberg B, Harlan DM, Powers AC. 2005. Assessment of human pancreatic islet architecture and composition by laser scanning confocal microscopy. J Histochem Cytochem 53: 1087–1097. 10.1369/jhc.5C6684.2005 [DOI] [PubMed] [Google Scholar]
- Brissova M, Aamodt K, Brahmachary P, Prasad N, Hong JY, Dai C, Mellati M, Shostak A, Poffenberger G, Aramandla R, et al. 2014. Islet microenvironment, modulated by vascular endothelial growth factor-A signaling, promotes β cell regeneration. Cell Metab 19: 498–511. 10.1016/j.cmet.2014.02.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brovkina O, Dashinimaev E. 2020. Advances and complications of regenerative medicine in diabetes therapy. Peer J 8: e9746. 10.7717/peerj.9746 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Byrne MM, O'Meara JSNM, Polonsky KS. 1995. Insulin secretion in insulin-resistant women with a history of gestational diabetes. Metab Clin Exp 44: 1067–1073. 10.1016/0026-0495(95)90106-X [DOI] [PubMed] [Google Scholar]
- Byrnes LE, Wong DM, Subramaniam M, Meyer NP, Gilchrist CL, Knox SM, Tward AD, Ye CJ, Sneddon JB. 2018. Lineage dynamics of murine pancreatic development at single-cell resolution. Nat Commun 9: 1–17. 10.1038/s41467-018-06176-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cabrera O, Berman DM, Kenyon NS, Ricordi C, Berggren PO, Caicedo A. 2006. The unique cytoarchitecture of human pancreatic islets has implications for islet cell function. Proc Natl Acad Sci 103: 2334–2339. 10.1073/pnas.0510790103 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cai Q, Brissova M, Reinert RB, Cheng Pan F, Brahmachary P, Jeansson M, Shostak A, Radhika A, Poffenberger G, et al. 2012. Enhanced expression of VEGF-A in β cells increases endothelial cell number but impairs islet morphogenesis and β cell proliferation. Dev Biol 367: 40–54. 10.1016/j.ydbio.2012.04.022 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Calabrese A, Zhang M, Serre-Beinier V, Caton D, Mas C, Satin LS, Meda P. 2003. Connexin 36 controls synchronization of Ca2+ oscillations and insulin secretion in MIN6 cells. Diabetes 52: 417–424. 10.2337/diabetes.52.2.417 [DOI] [PubMed] [Google Scholar]
- Carvalho CPF, Barbosa HCL, Britan A, Santos-Silva JCR, Boschero AC, Meda P, Collares-Buzato CB. 2010. β Cell coupling and connexin expression change during the functional maturation of rat pancreatic islets. Diabetologia 53: 1428–1437. 10.1007/s00125-010-1726-8 [DOI] [PubMed] [Google Scholar]
- Cervantes S, Lau J, Cano DA, Borromeo-Austin C, Hebrok M. 2010. Primary cilia regulate Gli/hedgehog activation in pancreas. Proc Natl Acad Sci 107: 10109–10114. 10.1073/pnas.0909900107 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chakravarthy H, Gu X, Enge M, Dai X, Wang Y, Damond N, Downie C, Liu K, Wang J, Xing Y, et al. 2017. Converting adult pancreatic islet α cells into β cells by targeting both Dnmt1 and Arx. Cell Metab 25: 622–634. 10.1016/j.cmet.2017.01.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chang FP, Cho CHH, Shen CR, Chien CY, Ting LW, Lee HS, Shen CN. 2016. PDGF facilitates direct lineage reprogramming of hepatocytes to functional β-like cells induced by Pdx1 and Ngn3. Cell Transplant 25: 1893–1909. 10.3727/096368916X691439 [DOI] [PubMed] [Google Scholar]
- Chen Z, Wang J, Sun W, Archibong E, Kahkoska AR, Zhang X, Lu Y, Ligler FS, Buse JB, Gu Z. 2018. Synthetic β cells for fusion-mediated dynamic insulin secretion. Nat Chem Biol 14: 86–93. 10.1038/nchembio.2511 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chera S, Baronnier D, Ghila L, Cigliola V, Jensen JN, Gu G, Furuyama K, Thorel F, Gribble FM, Reimann F, et al. 2014. Diabetes recovery by age-dependent conversion of pancreatic δ-cells into insulin producers. Nature 514: 503–507. 10.1038/nature13633 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chung CH, Hao E, Piran R, Keinan E, Levine F. 2010. Pancreatic β-cell neogenesis by direct conversion from mature α-cells. Stem Cells 28: 1630–1638. 10.1002/stem.482 [DOI] [PubMed] [Google Scholar]
- Cinti F, Bouchi R, Kim-Muller JY, Ohmura Y, Sandoval PR, Masini M, Marselli L, Suleiman M, Ratner LE, Marchetti P, et al. 2016. Evidence of β-cell dedifferentiation in human type 2 diabetes. J Clin Endocrinol Metab 101: 1044–1054. 10.1210/jc.2015-2860 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cohen H, Barash H, Meivar-Levy I, Molakandov K, Ben-Shimon M, Gurevich M, Zoabi F, Har-Zahav A, Gebhardt R, Gaunitz F, et al. 2018. The Wnt/β-catenin pathway determines the predisposition and efficiency of liver-to-pancreas reprogramming. Hepatology 68: 1589–1603. 10.1002/hep.29827 [DOI] [PubMed] [Google Scholar]
- Collombat P, Xu X, Ravassard P, Sosa-pineda B, Dussaud S, Billestrup N, Madsen OD, Serup P. 2010. NIH Public Access 138: 449–462. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Courtney M, Gjernes E, Druelle N, Ravaud C, Vieira A, Ben-Othman N, Pfeifer A, Avolio F, Leuckx G, Lacas-Gervais S, et al. 2013. The inactivation of Arx in pancreatic α-cells triggers their neogenesis and conversion into functional β-like cells. PLoS Genet 9: e1003934. 10.1371/journal.pgen.1003934 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cyphert HA, Walker EM, Hang Y, Dhawan S, Haliyur R, Bonatakis L, Avrahami D, Brissova M, Kaestner KH, Bhushan A, et al. 2019. Examining how the MAFB transcription factor affects islet β-cell function postnatally. Diabetes 68: 337–348. 10.2337/db18-0903 [DOI] [PMC free article] [PubMed] [Google Scholar]
- D'Amour KA, Agulnick AD, Eliazer S, Kelly OG, Kroon E, Baetge EE. 2005. Efficient differentiation of human embryonic stem cells to definitive endoderm. Nat Biotechnol 23: 1534–1541. 10.1038/nbt1163 [DOI] [PubMed] [Google Scholar]
- Da Silva Xavier G. 2018. The cells of the islets of Langerhans. J Clin Med 7: 54. 10.3390/jcm7030054 [DOI] [PMC free article] [PubMed] [Google Scholar]
- David SP, Murthy NV, Rabiner EA, Munafó MR, Johnstone EC, Jacob R, Walton RT, Grasby PM. 2005. A functional genetic variation of the serotonin (5-HT) transporter affects 5-HT1A receptor binding in humans. J Neurosci 25: 2586–2590. 10.1523/JNEUROSCI.3769-04.2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Davis JC, Alves TC, Helman A, Chen JC, Kenty JH, Cardone RL, Liu DR, Kibbey RG, Melton DA. 2020. Glucose response by stem cell-derived β cells in vitro is inhibited by a bottleneck in glycolysis. Cell Rep 31: 107623. 10.1016/j.celrep.2020.107623 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Desai T, Shea LD. 2017. Advances in islet encapsulation technologies. Nat Rev Drug Discov 16: 338–350. 10.1038/nrd.2016.232 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Desai BM, Oliver-Krasinski J, De Leon DD, Farzad C, Hong N, Leach SD, Stoffers DA. 2007. Preexisting pancreatic acinar cells contribute to acinar cell, but not islet β cell, regeneration. J Clin Invest 117: 971–977. 10.1172/JCI29988 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Deuse T, Hu X, Gravina A, Wang D, Tediashvili G, De C, Thayer WO, Wahl A, Garcia JV, Reichenspurner H, et al. 2019. Hypoimmunogenic derivatives of induced pluripotent stem cells evade immune rejection in fully immunocompetent allogeneic recipients. Nat Biotechnol 37: 252–258. 10.1038/s41587-019-0016-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dhawan S, Georgia S, Tschen S, Fan G, Bhushan A. 2011. Pancreatic β cell identity is maintained by DNA methylation-mediated repression of Arx. Dev Cell 20: 419–429. 10.1016/j.devcel.2011.03.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dirice E, Kahraman S, De Jesus DF, El Ouaamari A, Basile G, Baker RL, Yigit B, Piehowski PD, Kim MJ, Dwyer AJ, et al. 2019. Increased β-cell proliferation before immune cell invasion prevents progression of type 1 diabetes. Nat Metab 1: 509–518. 10.1038/s42255-019-0061-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dolenšek J, Rupnik MS, Stožer A. 2015. Structural similarities and differences between the human and the mouse pancreas. Islets 7: e1024405. 10.1080/19382014.2015.1024405 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dor Y, Brown J, Martinez OI, Melton DA. 2004. Adult pancreatic β-cells are formed by self-duplication rather than stem-cell differentiation. Nature 429: 41–46. 10.1038/nature02520 [DOI] [PubMed] [Google Scholar]
- Dorrell C, Schug J, Canaday PS, Russ HA, Tarlow BD, Grompe MT, Horton T, Hebrok M, Streeter PR, Kaestner KH, et al. 2016. Human islets contain four distinct subtypes of β cells. Nat Commun 7: 1–9. 10.1038/ncomms11756 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dunbar JC, Walsh MF. 1982. Glucagon and insulin secretion by dispersed islet cells: possible paracrine relationships. Horm Res 16: 257–267. 10.1159/000179510 [DOI] [PubMed] [Google Scholar]
- Dybala MP, Hara M. 2019. Heterogeneity of the human pancreatic islet. Diabetes 68: 1230–1239. 10.2337/db19-0072 [DOI] [PMC free article] [PubMed] [Google Scholar]
- El Khatib MM, Sakuma T, Tonne JM, Mohamed MS, Holditch SJ, Lu B, Kudva YC, Ikeda Y. 2015. β-Cell-targeted blockage of PD1 and CTLA4 pathways prevents development of autoimmune diabetes and acute allogeneic islets rejection. Gene Ther 22: 430–438. 10.1038/gt.2015.18 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fendrich V, Esni F, Garay MVR, Feldmann G, Habbe N, Jensen JN, Dor Y, Stoffers D, Jensen J, Leach SD, et al. 2008. Hedgehog signaling is required for effective regeneration of exocrine pancreas. Gastroenterology 135: 621–631.e8. 10.1053/j.gastro.2008.04.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ferber S, Halkin A, Cohen H, Ber I, Einav Y, Goldberg I, Barshack I, Seijffers R, Kopolovic J, Kaiser N, et al. 2000. Pancreatic and duodenal homeobox gene 1 induces expression of insulin genes in liver and ameliorates streptozotocin-induced hyperglycemia. Nat Med 6: 568–572. 10.1038/75050 [DOI] [PubMed] [Google Scholar]
- Gardner JD, Jackson MJ. 1977. Regulation of amylase release from dispersed pancreatic acinar cells. J Physiol 270: 439–454. 10.1113/jphysiol.1977.sp011961 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gourraud PA, Gilson L, Girard M, Peschanski M. 2012. The role of human leukocyte antigen matching in the development of multiethnic “haplobank” of induced pluripotent stem cell lines. Stem Cells 30: 180–186. 10.1002/stem.772 [DOI] [PubMed] [Google Scholar]
- Graham ML, Janecek JL, Kittredge JA, Hering BJ, Schuurman HJ. 2011. The streptozotocin-induced diabetic nude mouse model: differences between animals from different sources. Comp Med 61: 356–360. [PMC free article] [PubMed] [Google Scholar]
- Gromada J, Franklin I, Wollheim CB. 2007. Α-cells of the endocrine pancreas: 35 years of research but the enigma remains. Endocr Rev 28: 84–116. 10.1210/er.2006-0007 [DOI] [PubMed] [Google Scholar]
- Guo S, Dai C, Guo M, Taylor B, Harmon JS, Sander M, Robertson RP, Powers AC, Stein R. 2013a. Inactivation of specific β cell transcription factors in type 2 diabetes. J Clin Invest 123: 3305–3316. 10.1172/JCI65390 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guo T, Landsman L, Li N, Hebrok M. 2013b. Factors expressed by murine embryonic pancreatic mesenchyme enhance generation of insulin-producing cells from hESCs. Diabetes 62: 1581–1592. 10.2337/db12-0167 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Han X, Wang M, Duan S, Franco PJ, Kenty JHR, Hedrick P, Xia Y, Allen A, Ferreira LMR, Strominger JL, et al. 2019. Generation of hypoimmunogenic human pluripotent stem cells. Proc Natl Acad Sci 116: 10441–10446. 10.1073/pnas.1902566116 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hang Y, Stein R. 2011. Mafa and MafB activity in pancreatic β cells. Trends Endocrinol Metab 22: 364–373. 10.1016/j.tem.2011.05.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hasnain SZ, Borg DJ, Harcourt BE, Tong H, Sheng YH, Ng CP, Das I, Wang R, Chen ACH, Loudovaris T, et al. 2014. Glycemic control in diabetes is restored by therapeutic manipulation of cytokines that regulate β cell stress. Nat Med 20: 1417–1426. 10.1038/nm.3705 [DOI] [PubMed] [Google Scholar]
- Hauge-Evans AC, King AJ, Carmignac D, Richardson CC, Robinson ICAF, Low MJ, Christie MR, Persaud SJ, Jones PM. 2009. Somatostatin secreted by islet δ-cells fulfills multiple roles as a paracrine regulator of islet function. Diabetes 58: 403–411. 10.2337/db08-0792 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hoang CQ, Hale MA, Azevedo-Pouly AC, Elsässer HP, Deering TG, Willet SG, Pan FC, Magnuson MA, Wright CVE, Swift GH, et al. 2016. Transcriptional maintenance of pancreatic acinar identity, differentiation, and homeostasis by PTF1A. Mol Cell Biol 36: 3033–3047. 10.1128/MCB.00358-16 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hopcroft DW, Mason DR, Scott RS. 1985. Structure-function relationships in pancreatic islets: support for intraislet modulation of insulin secretion. Endocrinology 117: 2073–2080. 10.1210/endo-117-5-2073 [DOI] [PubMed] [Google Scholar]
- Huypens P, Ling Z, Pipeleers D, Schuit F. 2000. Glucagon receptors on human islet cells contribute to glucose competence of insulin release. Diabetologia 43: 1012–1019. 10.1007/s001250051484 [DOI] [PubMed] [Google Scholar]
- Hunziker E, Stein M. 2000. Nestin-expressing cells in the pancreatic islets of Langerhans. Biochem Biophys Res Commun 271: 116–119. [DOI] [PubMed] [Google Scholar]
- Jacobson K, Liu P. 2016. Complexity revealed: a hierarchy of clustered membrane proteins. Biophys J 111: 1–2. 10.1016/j.bpj.2016.05.045 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jennings RE, Berry AA, Kirkwood-Wilson R, Roberts NA, Hearn T, Salisbury RJ, Blaylock J, Hanley KP, Hanley NA. 2013. Development of the human pancreas from foregut to endocrine commitment. Diabetes 62: 3514–3522. 10.2337/db12-1479 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jensen JN, Cameron E, Garay MVR, Starkey TW, Gianani R, Jensen J. 2005. Recapitulation of elements of embryonic development in adult mouse pancreatic regeneration. Gastroenterology 128: 728–741. 10.1053/j.gastro.2004.12.008 [DOI] [PubMed] [Google Scholar]
- John AN, Ram R, Jiang FX. 2018. RNA-seq analysis of islets to characterise the dedifferentiation in type 2 diabetes model mice db/db. Endocr Pathol 29: 207–221. 10.1007/s12022-018-9523-x [DOI] [PubMed] [Google Scholar]
- Johnston NR, Mitchell RK, Haythorne E, Pessoa MP, Semplici F, Ferrer J, Piemonti L, Marchetti P, Bugliani M, Bosco D, et al. 2016. β Cell hubs dictate pancreatic islet responses to glucose. Cell Metab 24: 389–401. 10.1016/j.cmet.2016.06.020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jones BS, Lamb LS, Goldman F, Di Stasi A. 2014. Improving the safety of cell therapy products by suicide gene transfer. Front Pharmacol 5: 1–8. 10.3389/fphar.2014.00254 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kalousova A, Mavropoulos A, Adams BA, Nekrep N, Li Z, Krauss S, Stainier DY, German MS. 2010. Dachshund homologues play a conserved role in islet cell development. Dev Biol 348: 143–152. 10.1016/j.ydbio.2010.09.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kayali AG, Flores LE, Lopez AD, Kutlu B, Baetge E, Kitamura R, Hao E, Beattie GM, Hayek A. 2007. Limited capacity of human adult islets expanded in vitro to redifferentiate into insulin-producing β-cells. Diabetes 56: 703–708. 10.2337/db06-1545 [DOI] [PubMed] [Google Scholar]
- Kern HF. 1993. Fine structure of the human exocrine pancreas. Pancreas Biol Pathobiol Dis 2: 9–19. [Google Scholar]
- Kieffer J, Heller RS, Unson CG, Weir C, Habener JF. 1996. Distribution of glucagon receptors on hormone-specific endocrine cells of rat pancreatic islets. Endocrinology 137: 5119–5125. 10.1210/endo.137.11.8895386 [DOI] [PubMed] [Google Scholar]
- Kim A, Miller K, Jo J, Kilimnik G, Wojcik P, Hara M. 2009. Islet architecture: a comparative study. Islets 1: 129–136. 10.4161/isl.1.2.9480 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim H, Toyofuku Y, Lynn FC, Chak E, Uchida T, Mizukami H, Fujitani Y, Kawamori R, Miyatsuka T, Kosaka Y, et al. 2010. Serotonin regulates pancreatic β cell mass during pregnancy. Nat Med 16: 804–808. 10.1038/nm.2173 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Koerker DJ, Ruch W, Chideckel E, Palmer J, Charles J. 1974. Somatostatin: hypothalamic inhibitor of the endocrine pancreas. Science 184: 482–484. 10.1126/science.184.4135.482 [DOI] [PubMed] [Google Scholar]
- Konstantinova I, Nikolova G, Ohara-Imaizumi M, Meda P, Kučera T, Zarbalis K, Wurst W, Nagamatsu S, Lammert E. 2007. EphA-Ephrin-A-mediated β cell communication regulates insulin secretion from pancreatic islets. Cell 129: 359–370. 10.1016/j.cell.2007.02.044 [DOI] [PubMed] [Google Scholar]
- Kotini AG, de Stanchina E, Themeli M, Sadelain M, Papapetrou EP. 2016. Escape mutations, ganciclovir resistance, and teratoma formation in human iPSCs expressing an HSVtk suicide gene. Mol Ther Nucleic Acids 5: e284. 10.1038/mtna.2015.57 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kumar AF, Gruessner RWG, Seaquist ER. 2008. Risk of glucose intolerance and diabetes in hemipancreatectomized donors selected for normal preoperative glucose metabolism. Diabetes Care 31: 1639–1643. 10.2337/dc07-2453 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Landsman L, Parent A, Hebrok M. 2011. Elevated Hedgehog/Gli signaling causes β-cell dedifferentiation in mice. Proc Natl Acad Sci 108: 17010–17015. 10.1073/pnas.1105404108 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Larsen HL, Grapin-Botton A. 2017. The molecular and morphogenetic basis of pancreas organogenesis. Semin Cell Dev Biol 66: 51–68. 10.1016/j.semcdb.2017.01.005 [DOI] [PubMed] [Google Scholar]
- Lazarow A. 1957. Cell types of the islets of Langerhans and the hormones they produce. Diabetes 6: 222–232. 10.2337/diab.6.3.222 [DOI] [PubMed] [Google Scholar]
- Lehv M, Fitzgerald PJ. 1968. Pancreatic acinar cell regeneration. IV: Regeneration after resection. Am J Pathol 53: 513–535. [PMC free article] [PubMed] [Google Scholar]
- Lenzen S. 2008. The mechanisms of alloxan- and streptozotocin-induced diabetes. Diabetologia 51: 216–226. 10.1007/s00125-007-0886-7 [DOI] [PubMed] [Google Scholar]
- Liang Q, Monetti C, Shutova MV, Neely EJ, Hacibekiroglu S, Yang H, Kim C, Zhang P, Li C, Nagy K, et al. 2018. Linking a cell-division gene and a suicide gene to define and improve cell therapy safety. Nature 563: 701–704. 10.1038/s41586-018-0733-7 [DOI] [PubMed] [Google Scholar]
- Liu JSE, Hebrok M. 2017. All mixed up: defining roles for β-cell subtypes in mature islets. Genes Dev 31: 228–240. 10.1101/gad.294389.116 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Logothetopoulos J, Davidson JK, Haist RE, Best CH. 1965. Degranulation of β cells and loss of pancreatic insulin after infusions of insulin antibody or glucose. Diabetes 14: 493–500. 10.2337/diab.14.8.493 [DOI] [PubMed] [Google Scholar]
- Mahaddalkar PU, Scheibner K, Pfluger S, Ansarullah, Sterr M, Beckenbauer J, Irmler M, Beckers J, Knöbel S, Lickert H. 2020. Generation of pancreatic β cells from CD177+ anterior definitive endoderm. Nat Biotechnol 38: 1061–1072. 10.1038/s41587-020-0492-5 [DOI] [PubMed] [Google Scholar]
- Masini M, Marselli L, Himpe E, Martino L, Bugliani M, Suleiman M, Boggi U, Filipponi F, Occhipinti M, Bouwens L, et al. 2017. Co-localization of acinar markers and insulin in pancreatic cells of subjects with type 2 diabetes. PLoS ONE 12: e0179398. 10.1371/journal.pone.0179398 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mawla AM, Huising MO. 2019. Navigating the depths and avoiding the shallows of pancreatic islet cell transcriptomes. Diabetes 68: 1380–1393. 10.2337/dbi18-0019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Menge BA, Tannapfel A, Belyaev O, Drescher R, Müller C, Uhl W, Schmidt WE, Meier JJ. 2008. Partial pancreatectomy in adult humans does not provoke β-cell regeneration. Diabetes 57: 142–149. 10.2337/db07-1294 [DOI] [PubMed] [Google Scholar]
- Mezza T, Kulkarni RN. 2014. The regulation of pre- and post-maturational plasticity of mammalian islet cell mass. Diabetologia 57: 1291–1303. 10.1007/s00125-014-3251-7 [DOI] [PubMed] [Google Scholar]
- Miyazaki S, Tashiro F, Tsuchiya T, Sasaki K, Miyazaki J. 2021. Establishment of a long-term stable β-cell line and its application to analyze the effect of Gcg expression on insulin secretion. Sci Rep 11: 1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moens K, Flamez D, Van Schravendijk C, Ling Z, Pipeleers D, Schuit F. 1998. Dual glucagon recognition by pancreatic β-cells via glucagon and glucagon-like peptide 1 receptors. Diabetes 47: 66–72. 10.2337/diab.47.1.66 [DOI] [PubMed] [Google Scholar]
- Montanya E, Nacher V, Biarnes M, Soler J. 2000. Linear correlation between β-cell mass and body weight throughout the lifespan in Lewis rats. Role of β-cell hyperplasia and hypertrophy. Diabetes 49: 1341–1346. 10.2337/diabetes.49.8.1341 [DOI] [PubMed] [Google Scholar]
- Morris JP IV, Cano DA, Sekine S, Wang SC, Hebrok M. 2010. β-Catenin blocks Kras-dependent reprogramming of acini into pancreatic cancer precursor lesions in mice. J Clin Invest 120: 508–520. 10.1172/JCI40045 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Murtaugh LC, Keefe MD. 2015. Regeneration and repair of the exocrine pancreas. Annu Rev Physiol 77: 229–249. 10.1146/annurev-physiol-021014-071727 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nair G, Hebrok M. 2015. Islet formation in mice and men: lessons for the generation of functional insulin-producing β-cells from human pluripotent stem cells. Curr Opin Genet Dev 32: 171–180. 10.1016/j.gde.2015.03.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nair GG, Liu JS, Russ HA, Tran S, Saxton MS, Chen R, Juang C, Li M, Nguyen VQ, Giacometti S, et al. 2019. Recapitulating endocrine cell clustering in culture promotes maturation of human stem-cell-derived β cells. Nat Cell Biol 21: 263–274. 10.1038/s41556-018-0271-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nakatsuji N, Nakajima F, Tokunaga K. 2008. HLA-haplotype banking and iPS cells. Nat Biotechnol 26: 739–740. 10.1038/nbt0708-739 [DOI] [PubMed] [Google Scholar]
- Nathan G, Kredo-Russo S, Geiger T, Lenz A, Kaspi H, Hornstein E, Efrat S. 2015. MiR-375 promotes redifferentiation of adult human β cells expanded in vitro. PLoS ONE 10: 1–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nishimura W, Takahashi S, Yasuda K. 2015. Mafa is critical for maintenance of the mature β cell phenotype in mice. Diabetologia 58: 566–574. 10.1007/s00125-014-3464-9 [DOI] [PubMed] [Google Scholar]
- Nordmann TM, Dror E, Schulze F, Traub S, Berishvili E, Barbieux C, Böni-Schnetzler M, Donath MY. 2017. The role of inflammation in β-cell dedifferentiation. Sci Rep 7: 1–10. 10.1038/s41598-017-06731-w [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nyman LR, Wells KS, Head WS, McCaughey M, Ford E, Brissova M, Piston DW, Powers AC. 2008. Real-time, multidimensional in vivo imaging used to investigate blood flow in mouse pancreatic islets. J Clin Invest 118: 3790–3797. 10.1172/JCI36209 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ohara-Imaizumi M, Kim H, Yoshida M, Fujiwara T, Aoyagi K, Toyofuku Y, Nakamichi Y, Nishiwaki C, Okamura T, Uchida T, et al. 2013. Serotonin regulates glucose-stimulated insulin secretion from pancreatic β cells during pregnancy. Proc Natl Acad Sci 110: 19420–19425. 10.1073/pnas.1310953110 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Oram RA, Sims EK, Evans-Molina C. 2019. β Cells in type 1 diabetes: mass and function; sleeping or dead? Diabetologia 62: 567–577. 10.1007/s00125-019-4822-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Orci L, Unger RH. 1975. Functional subdivision of islets of Langerhans and possible role of D cells. Lancet 306: 1243–1244. 10.1016/S0140-6736(75)92078-4 [DOI] [PubMed] [Google Scholar]
- Pagliuca FW, Millman JR, Gürtler M, Segel M, Van Dervort A, Ryu JH, Peterson QP, Greiner D, Melton DA. 2014. Generation of functional human pancreatic β cells in vitro. Cell 159: 428–439. 10.1016/j.cell.2014.09.040 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pan FC, Wright C. 2011. Pancreas organogenesis: from bud to plexus to gland. Dev Dyn 240: 530–565. 10.1002/dvdy.22584 [DOI] [PubMed] [Google Scholar]
- Pipeleers D, In't Veld P, Maes E, Van De Winkel M. 1982. Glucose-induced insulin release depends on functional cooperation between islet cells. Proc Natl Acad Sci 79: 7322–7325. 10.1073/pnas.79.23.7322 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pipeleers D, Kiekens R, Ling Z, Wilikens A, Schuit F. 1994. Physiologic relevance of heterogeneity in the pancreatic β-cell population. Diabetologia 37: S57–S64. 10.1007/BF00400827 [DOI] [PubMed] [Google Scholar]
- Prado CL, Pugh-Bernard AE, Elghazi L, Sosa-Pineda B, Sussel L. 2004. Ghrelin cells replace insulin-producing β cells in two mouse models of pancreas development. Proc Natl Acad Sci 101: 2924–2929. 10.1073/pnas.0308604100 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Puri S, Cano DA, Hebrok M. 2009. A role for von Hippel-Lindau protein in pancreatic β-cell function. Diabetes 58: 433–441. 10.2337/db08-0749 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Puri S, Akiyama H, Hebrok M. 2013. VHL-mediated disruption of Sox9 activity compromises β-cell identity and results in diabetes mellitus. Genes Dev 27: 2563–2575. 10.1101/gad.227785.113 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Puri S, Folias AE, Hebrok M. 2015. Plasticity and dedifferentiation within the pancreas: development, homeostasis, and disease. Cell Stem Cell 16: 18–31. 10.1016/j.stem.2014.11.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Puri S, Roy N, Russ HA, Leonhardt L, French EK, Roy R, Bengtsson H, Scott DK, Stewart AF, Hebrok M. 2018. Replication confers β cell immaturity. Nat Commun 9: 485. 10.1038/s41467-018-02939-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Qadir MMF, Álvarez-Cubela S, Belle K, Sapir T, Messaggio F, Johnson KB, Umland O, Hardin D, Klein D, Pérez-Álvarez I, et al. 2019. A double fail-safe approach to prevent tumorigenesis and select pancreatic β cells from human embryonic stem cells. Stem Cell Reports 12: 611–623. 10.1016/j.stemcr.2019.01.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rajagopal J, Anderson WJ, Kume S, Martinez OI, Melton DA. 2003. Insulin staining of ES cell progeny from insulin uptake. Science 299: 363. 10.1126/science.1077838 [DOI] [PubMed] [Google Scholar]
- Rankin MM, Kushner JA. 2009. Adaptive β-cell proliferation is severely restricted with advanced age. Diabetes 58: 1365–1372. 10.2337/db08-1198 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ravier M, Gu M, Charollais A, Gjinovci A, Wollheim CB, Willecke K, Henquin J, Meda P. 2005. Loss of Connexin36 channels alters β-cell coupling, islet synchronization of glucose-induced Ca2+ and insulin oscillations, and basal insulin release. Diabetes 54: 1798–1807. 10.2337/diabetes.54.6.1798 [DOI] [PubMed] [Google Scholar]
- Reichert M, Rustgi AK. 2011. Pancreatic ductal cells in development, regeneration, and neoplasia. J Clin Invest 121: 4572–4578. 10.1172/JCI57131 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reinert RB, Cai Q, Hong JY, Plank JL, Aamodt K, Prasad N, Aramandla R, Dai C, Levy SE, Pozzi A, et al. 2014. Vascular endothelial growth factor coordinates islet innervation via vascular scaffolding. Development 141: 1480–1491. 10.1242/dev.098657 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rezania A, Bruin JE, Xu J, Narayan K, Fox JK, O'Neil JJ, Kieffer TJ. 2013. 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. 10.1002/stem.1489 [DOI] [PubMed] [Google Scholar]
- Rezania A, Bruin JE, Arora P, Rubin A, Batushansky I, Asadi A, O'Dwyer S, Quiskamp N, Mojibian M, Albrecht T, et al. 2014. Reversal of diabetes with insulin-producing cells derived in vitro from human pluripotent stem cells. Nat Biotechnol 32: 1121–1133. 10.1038/nbt.3033 [DOI] [PubMed] [Google Scholar]
- Rieck S, Kaestner KH. 2010. Expansion of β-cell mass in response to pregnancy. Trends Endocrinol Metab 21: 151–158. 10.1016/j.tem.2009.11.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rodnoi P, Rajkumar M, Moin ASM, Georgia SK, Butler AE, Dhawan S. 2017. Neuropeptide Y expression marks partially differentiated β cells in mice and humans. JCI Insight 2: e94005. 10.1172/jci.insight.94005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rogers GJ, Hodgkin MN, Squires PE. 2007. E-cadherin and cell adhesion: a role in architecture and function in the pancreatic islet. Cell Physiol Biochem 20: 987–994. 10.1159/000110459 [DOI] [PubMed] [Google Scholar]
- Russ HA, Parent AV, Ringler JJ, Hennings TG, Nair GG, Shveygert M, Guo T, Puri S, Haataja L, Cirulli V, et al. 2015. Controlled induction of human pancreatic progenitors produces functional β-like cells in vitro. EMBO J 34: 1759–1772. 10.15252/embj.201591058 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Russell R, Carnese PP, Hennings TG, Walker EM, Russ HA, Liu JS, Giacometti S, Stein R, Hebrok M. 2020. Loss of the transcription factor MAFB limits β-cell derivation from human PSCs. Nat Commun 11: 1–15. 10.1038/s41467-020-16550-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Salem V, Silva LD, Suba K, Georgiadou E, Gharavy SNM, Akhtar N, Martin-Alonso A, Gaboriau DCA, Rothery SM, Stylianides T, et al. 2019. Leader β-cells coordinate Ca2+ dynamics across pancreatic islets in vivo. Nat Metab 1: 615–629. 10.1038/s42255-019-0075-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sapir T, Shternhall K, Meivar-Levy I, Blumenfeld T, Cohen H, Skutelsky E, Eventov-Friedman S, Barshack I, Goldberg I, Pri-Chen S, et al. 2005. Cell-replacement therapy for diabetes: generating functional insulin-producing tissue from adult human liver cells. Proc Natl Acad Sci 102: 7964–7969. 10.1073/pnas.0405277102 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Saunders D, Powers AC. 2016. Replicative capacity of β-cells and type 1 diabetes. J Autoimmun 71: 59–68. 10.1016/j.jaut.2016.03.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Saxena P, Heng BC, Bai P, Folcher M, Zulewski H, Fussenegger M. 2016. A programmable synthetic lineage-control network that differentiates human IPSCs into glucose-sensitive insulin-secreting β-like cells. Nat Commun 7: 1–14. 10.1038/ncomms11247 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Scharp DW, Marchetti P. 2014. Encapsulated islets for diabetes therapy: history, current progress, and critical issues requiring solution. Adv Drug Deliv Rev 67-68: 35–73. 10.1016/j.addr.2013.07.018 [DOI] [PubMed] [Google Scholar]
- Serre-Beinier V, Le Gurun S, Belluardo N, Trovato-Salinaro A, Charollais A, Haefliger JA, Condorelli DF, Meda P. 2000. Cx36 preferentially connects β-cells within pancreatic islets. Diabetes 49: 727–734. 10.2337/diabetes.49.5.727 [DOI] [PubMed] [Google Scholar]
- Serre-Beinier V, Bosco D, Zulianello L, Charollais A, Caille D, Charpantier E, Gauthier BR, Diaferia GR, Giepmans BN, Lupi R, et al. 2009. Cx36 makes channels coupling human pancreatic β-cells, and correlates with insulin expression. Hum Mol Genet 18: 428–439. 10.1093/hmg/ddn370 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sherry NA, Kushner JA, Glandt M, Kitamura T, Brillantes AMB, Herold KC. 2006. Effects of autoimmunity and immune therapy on β-cell turnover in type 1 diabetes. Diabetes 55: 3238–3245. 10.2337/db05-1034 [DOI] [PubMed] [Google Scholar]
- Shih HP, Wang A, Sander M. 2013. Pancreas organogenesis: from lineage determination to morphogenesis. Annu Rev Cell Dev Biol 29: 81–105. 10.1146/annurev-cellbio-101512-122405 [DOI] [PubMed] [Google Scholar]
- Siveke JT, Lubeseder-Martellato C, Lee M, Mazur PK, Nakhai H, Radtke F, Schmid RM. 2008. Notch signaling is required for exocrine regeneration after acute pancreatitis. Gastroenterology 134: 544–555.e3. 10.1053/j.gastro.2007.11.003 [DOI] [PubMed] [Google Scholar]
- Sneddon JB, Tang Q, Stock P, Bluestone JA, Roy S, Desai T, Hebrok M. 2018. Stem cell therapies for treating diabetes: progress and remaining challenges. Cell Stem Cell 22: 810–823. 10.1016/j.stem.2018.05.016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Solar M, Cardalda C, Houbracken I, Martin M, Maestro MA, de Medts N, Xu X, Grau V, Heimberg H, Bouwens L, et al. 2009. Pancreatic exocrine duct cells give rise to insulin-producing β cells during embryogenesis but not after birth. Dev Cell 17: 849–860. 10.1016/j.devcel.2009.11.003 [DOI] [PubMed] [Google Scholar]
- Stanger BZ, Hebrok M. 2013. Control of cell identity in pancreas development and regeneration. Gastroenterology 144: 1170–1179. 10.1053/j.gastro.2013.01.074 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Steiner DJ, Kim A, Miller K, Hara M. 2010. Pancreatic islet plasticity: interspecies comparison of islet architecture and composition. Islets 2: 135–145. 10.4161/isl.2.3.11815 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Strobel O, Dor Y, Alsina J, Stirman A, Lauwers G, Trainor A, Del Castillo CF, Warshaw AL, Thayer SP. 2007. In vivo lineage tracing defines the role of acinar-to-ductal transdifferentiation in inflammatory ductal metaplasia. Gastroenterology 133: 1999–2009. 10.1053/j.gastro.2007.09.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Svendsen B, Larsen O, Gabe MBN, Christiansen CB, Rosenkilde MM, Drucker DJ, Holst JJ. 2018. Insulin secretion depends on intra-islet glucagon signaling. Cell Rep 25: 1127–1134.e2. 10.1016/j.celrep.2018.10.018 [DOI] [PubMed] [Google Scholar]
- Szkudelski T. 2001. The mechanism of alloxan and streptozotocin action in B cells of the rat pancreas. Physiol Res 50: 537–546. [PubMed] [Google Scholar]
- Taborsky GJ, Smith PH, Porte D. 1978. Interaction of somatostatin with the A and B cells of the endocrine pancreas. Metab Clin Exp 27: 1299–1302. 10.1016/0026-0495(78)90062-8 [DOI] [PubMed] [Google Scholar]
- Talchai C, Xuan S, Lin HV, Sussel L, Accili D. 2012. Pancreatic β cell dedifferentiation as a mechanism of diabetic β cell failure. Cell 150: 1223–1234. 10.1016/j.cell.2012.07.029 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tang S, Zhang M, Zeng S, Huang Y, Qin M, Nasri U, Santamaria P, Riggs AD, Jin L, Zeng D. 2020. Reversal of autoimmunity by mixed chimerism enables reactivation of β cells and transdifferentiation of α cells in diabetic NOD mice. Proc Natl Acad Sci 117: 31219–31230. 10.1073/pnas.2012389117 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Taylor CJ, Peacock S, Chaudhry AN, Bradley JA, Bolton EM. 2012. Generating an iPSC bank for HLA-matched tissue transplantation based on known donor and recipient HLA types. Cell Stem Cell 11: 147–152. 10.1016/j.stem.2012.07.014 [DOI] [PubMed] [Google Scholar]
- Tellez K, Hang Y, Gu X, Chang CA, Stein RW, Kim SK. 2020. In vivo studies of glucagon secretion by human islets transplanted in mice. Nat Metab 2: 547–557. 10.1038/s42255-020-0213-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- Teta M, Long SY, Wartschow LM, Rankin MM, Kushner JA. 2005. Very slow turnover of β-cells in aged adult mice. Diabetes 54: 2557–2567. 10.2337/diabetes.54.9.2557 [DOI] [PubMed] [Google Scholar]
- Thorel F, Népote V, Avril I, Kohno K, Desgraz R, Chera S, Herrera PL. 2010. Conversion of adult pancreatic α-cells to Β-cells after extreme Β-cell loss. Nature 464: 1149–1154. 10.1038/nature08894 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tschen SI, Dhawan S, Gurlo T, Bhushan A. 2009. Age-dependent decline in β-cell proliferation restricts the capacity of β-cell regeneration in mice. Diabetes 58: 1312–1320. 10.2337/db08-1651 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tsuchitani M, Sato J, Kokoshima H. 2016. A comparison of the anatomical structure of the pancreas in experimental animals. J Toxicol Pathol 29: 147–154. 10.1293/tox.2016-0016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- van der Meulen T, Huising MO. 2015. Role of transcription factors in the transdifferentiation of pancreatic islet cells. J Mol Endocrinol 54: R103–R117. 10.1530/JME-14-0290 [DOI] [PMC free article] [PubMed] [Google Scholar]
- van der Meulen T, Xie R, Kelly OG, Vale WW, Sander M, Huising MO. 2012. Urocortin 3 marks mature human primary and embryonic stem cell-derived pancreatic α and β cells. PLoS ONE 7: e52181. 10.1371/journal.pone.0052181 [DOI] [PMC free article] [PubMed] [Google Scholar]
- van der Meulen T, Mawla AM, DiGruccio MR, Adams MW, Nies V, Dólleman S, Liu S, Ackermann AM, Cáceres E, Hunter AE, et al. 2017. Virgin β cells persist throughout life at a neogenic niche within pancreatic islets. Cell Metab 25: 911–926.e6. 10.1016/j.cmet.2017.03.017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- van der Torren CR, Zaldumbide A, Duinkerken G, Brand-Schaaf SH, Peakman M, Stangé G, Martinson L, Kroon E, Brandon EP, Pipeleers D, et al. 2017. Immunogenicity of human embryonic stem cell-derived β cells. Diabetologia 60: 126–133. 10.1007/s00125-016-4125-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- Velazco-Cruz L, Song J, Maxwell KG, Goedegebuure MM, Augsornworawat P, Hogrebe NJ, Millman JR. 2019. Acquisition of dynamic function in human stem cell-derived β cells. Stem Cell Reports 12: 351–365. 10.1016/j.stemcr.2018.12.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Volta F, Scerbo MJ, Seelig A, Wagner R, O'Brien N, Gerst F, Fritsche A, Häring HU, Zeigerer A, Ullrich S, et al. 2019. Glucose homeostasis is regulated by pancreatic β-cell cilia via endosomal EphA-processing. Nat Commun 10: 1–17. 10.1038/s41467-019-12953-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Von Figura G, Morris JP IV, Wright CVE, Hebrok M. 2014. Nr5a2 maintains acinar cell differentiation and constrains oncogenic Kras-mediated pancreatic neoplastic initiation. Gut 63: 656–664. 10.1136/gutjnl-2012-304287 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang S, Yan J, Anderson DA, Xu Y, Kanal MC, Cao Z, Wright CVE, Gu G. 2010. Neurog3 gene dosage regulates allocation of endocrine and exocrine cell fates in the developing mouse pancreas. Dev Biol 339: 26–37. 10.1016/j.ydbio.2009.12.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang Z, York NW, Nichols CG, Remedi MS. 2014. Pancreatic β cell dedifferentiation in diabetes and redifferentiation following insulin therapy. Cell Metab 19: 872–882. 10.1016/j.cmet.2014.03.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang P, Alvarez-Perez JC, Felsenfeld DP, Liu H, Sivendran S, Bender A, Kumar A, Sanchez R, Scott DK, Garcia-Ocaña A, et al. 2015a. A high-throughput chemical screen reveals that harmine-mediated inhibition of DYRK1A increases human pancreatic β cell replication. Nat Med 21: 383–388. 10.1038/nm.3820 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang P, Fiaschi-Taesch NM, Vasavada RC, Scott DK, García-Ocaña A, Stewart AF. 2015b. Diabetes mellitus—advances and challenges in human β-cell proliferation. Nat Rev Endocrinol 11: 201–212. 10.1038/nrendo.2015.9 [DOI] [PubMed] [Google Scholar]
- Weir GC, Bonner-Weir S. 2004. Five of stages of evolving β-cell dysfunction during progression to diabetes. Diabetes 53 (Suppl 3): S16–S21. 10.2337/diabetes.53.suppl_3.s16 [DOI] [PubMed] [Google Scholar]
- Weir GC, Halban PA, Meda P, Wollheim CB, Orci L, Renold AE. 1984. Dispersed adult rat pancreatic islet cells in culture: A, B, and D cell function. Metab Clin Exp 33: 447–453. 10.1016/0026-0495(84)90146-X [DOI] [PubMed] [Google Scholar]
- Williams JA. 2010. Regulation of acinar cell function in the pancreas. Curr Opin Gastroenterol 26: 478–483. 10.1097/MOG.0b013e32833d11c6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xiao X, Guo P, Shiota C, Zhang T, Coudriet GM, Fischbach S, Prasadan K, Fusco J, Ramachandran S, Witkowski P, et al. 2018. Endogenous reprogramming of α cells into β cells, induced by viral gene therapy, reverses autoimmune diabetes. Cell Stem Cell 22: 78–90.e4. 10.1016/j.stem.2017.11.020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xin Y, Kim J, Okamoto H, Ni M, Wei Y, Adler C, Murphy AJ, Yancopoulos GD, Lin C, Gromada J. 2016. RNA sequencing of single human islet cells reveals type 2 diabetes genes. Cell Metab 24: 608–615. 10.1016/j.cmet.2016.08.018 [DOI] [PubMed] [Google Scholar]
- Xu H, Wang B, Ono M, Kagita A, Fujii K, Sasakawa N, Ueda T, Gee P, Nishikawa M, Nomura M, et al. 2019. Targeted disruption of HLA genes via CRISPR-Cas9 generates iPSCs with enhanced immune compatibility. Cell Stem Cell 24: 566–578.e7. 10.1016/j.stem.2019.02.005 [DOI] [PubMed] [Google Scholar]
- Yagyu S, Hoyos V, Del Bufalo F, Brenner MK. 2015. An inducible caspase-9 suicide gene to improve the safety of therapy using human induced pluripotent stem cells. Mol Ther 23: 1475–1485. 10.1038/mt.2015.100 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yamagata K, Nammo T, Moriwaki M, Ihara A, Iizuka K, Yang Q, Satoh T, Li M, Uenaka R, Okita K, et al. 2002. Overexpression of dominant-negative mutant hepatocyte nuclear factor-1α in pancreatic β-cells causes abnormal islet architecture with decreased expression of E-cadherin, reduced β-cell proliferation, and diabetes. Diabetes 51: 114–123. 10.2337/diabetes.51.1.114 [DOI] [PubMed] [Google Scholar]
- Yatoh S, Dodge R, Akashi T, Omer A, Sharma A, Weir GC, Bonner-weir S. 2007. Differentiation of affinity-purified human pancreatic duct cells to β-cells. Diabetes 56: 1802–1809. 10.2337/db06-1670 [DOI] [PubMed] [Google Scholar]
- Zhang N, Richter A, Suriawinata J, Harbaran S, Altomonte J, Cong L, Zhang H, Song K, Meseck M, Bromberg J, et al. 2004. Elevated vascular endothelial growth factor production in islets improves islet graft vascularization. Diabetes 53: 963–970. 10.2337/diabetes.53.4.963 [DOI] [PubMed] [Google Scholar]
- Zhou Q, Melton DA. 2018. Pancreas regeneration. Nature 557: 351–358. 10.1038/s41586-018-0088-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhou Q, Brown J, Kanarek A, Rajagopal J, Melton DA. 2008. In vivo reprogramming of adult pancreatic exocrine cells to β-cells. Nature 455: 627–632. 10.1038/nature07314 [DOI] [PMC free article] [PubMed] [Google Scholar]


