Skip to main content
Endocrinology logoLink to Endocrinology
. 2022 Nov 22;164(1):bqac193. doi: 10.1210/endocr/bqac193

Therapeutic Strategies Targeting Pancreatic Islet β-Cell Proliferation, Regeneration, and Replacement

Roy A Goode 1, Julia M Hum 2, Michael A Kalwat 3,4,
PMCID: PMC9923807  PMID: 36412119

Abstract

Diabetes results from insufficient insulin production by pancreatic islet β-cells or a loss of β-cells themselves. Restoration of regulated insulin production is a predominant goal of translational diabetes research. Here, we provide a brief overview of recent advances in the fields of β-cell proliferation, regeneration, and replacement. The discovery of therapeutic targets and associated small molecules has been enabled by improved understanding of β-cell development and cell cycle regulation, as well as advanced high-throughput screening methodologies. Important findings in β-cell transdifferentiation, neogenesis, and stem cell differentiation have nucleated multiple promising therapeutic strategies. In particular, clinical trials are underway using in vitro–generated β-like cells from human pluripotent stem cells. Significant challenges remain for each of these strategies, but continued support for efforts in these research areas will be critical for the generation of distinct diabetes therapies.

Keywords: pancreatic islet beta cells, induced pluripotent stem cells, proliferation, diabetes

Introduction

Pancreatic Islets of Langerhans and the Burden of Diabetes Mellitus

Pancreatic islets of Langerhans are key regulators of glucose homeostasis containing insulin-producing β-cells, glucagon-producing α-cells, somatostatin-producing δ-cells, and to a lesser extent pancreatic polypeptide (PP)–producing cells and ghrelin-producing ε-cells (1). Islets release insulin and glucagon, among other hormones, in response to a variety of metabolic, endocrine, and neuronal inputs to maintain glucose homeostasis (2). Diabetes mellitus is characterized by an impairment in the production or response to insulin, leading to chronic hyperglycemia and subsequent comorbidities, including cardiovascular disease, retinopathy, nephropathy, and neuropathy (3). In type 1 diabetes mellitus (T1D), the accepted view is that β-cells are destroyed via autoimmunity. This immune attack and loss of β-cell mass can occur in juveniles, adolescents, or adults (4, 5). While the exact causes of T1D are not understood, certain genetic variations can predispose individuals to T1D and environmental factors can trigger autoimmune responses (6, 7). For example, infections with certain viruses have been associated with increased T1D risk (8); however, this idea is still under investigation (9). It is also possible that the β-cell itself instigates its own autoimmune attack (10, 11). In type 2 diabetes mellitus (T2D), genetic and environmental circumstances contribute to β-cell dysfunction and peripheral insulin resistance (12-14). During this process, β-cells are thought to compensate for insulin resistance by increasing their mass and insulin output. Indeed, genome-wide association studies indicate that many of the genes associated with T2D risk are involved in β-cell function (15-18). As the disease develops, β-cell compensation eventually fails and there is loss of functional β-cell mass leading to T2D (19, 20). The resulting hyperglycemia has long-term deleterious effects in T1D and T2D patients, including microvascular and macrovascular complications (21).

In 2021, it was estimated that 537 million adults (≥18 years old) worldwide had diabetes, and projections indicate that by 2045 this number is expected to be near 783 million adults worldwide (22). Furthermore, medical care for those with diabetes presents a significant economic burden. Recent reports indicate that the cost of medical care to those with diabetes is rising in the United States. In 2017, the total cost dedicated to diabetes in the United States was estimated to be $327 billion (23). The rising cost of diabetes is increasing at a greater rate than other diseases, suggesting its treatment is becoming more expensive. From 1987 to 2011, the excess cost of care for a patient with diabetes essentially doubled when compared with a patient without diabetes (24).

Current Diabetes Therapies

While currently there is no cure for diabetes, patients with T1D and late stage T2D are treated using an exogenous subcutaneous insulin regimen to maintain glycemic control (25). Although life-saving and effective, subcutaneous insulin treatments have limitations. When compared with endogenous insulin secreted by pancreatic islet β-cells, recombinant exogenous insulin treatments are inferior in terms of the temporality and fine-tuning involved in the precise regulation of glycaemia (26). For instance, delivery of too much insulin may result in hypoglycemic episodes, which can be life-threatening (27). Prior to development of insulin dependence in T2D, a variety of pharmaceutical treatments exist to help lower insulin resistance and mitigate or delay endogenous β-cell failure. Such therapies include intensive insulin therapy (28), metformin (29), sulfonylureas (30), thiazolidinediones (31), glucagon-like peptide 1 receptor (GLP1R) agonists (32), dipeptidyl peptidase 4 inhibitors (33), and sodium/glucose cotransporter 2 inhibitors (30); many of these therapeutics have been reviewed in detail (30, 34). Artificial pancreas technologies such as closed-loop systems using glucose sensors and insulin pumps are also among strategies currently pursued (35). There are also multiple other pharmaceutical targets and approaches under investigation for T2D including glucokinase activators, GPR40 agonists, and alternative incretin axis stimulators (36). In T1D, major areas of focus for therapeutics include prevention of β-cell loss or replacement of β-cell mass, and, to this end, advances in immunotherapies are promising. Teplizumab is an anti-CD3 monoclonal antibody designed to bind CD3 at the cell surface and consequently activate the T cell receptor/CD3 signaling (37). Activation of the T cell receptor in this way can improve self-tolerance, preservation of regulatory T cells (38). Teplizumab slowed the reduction of β-cell function in new-onset T1D patients for 2 years after clinical diagnosis (39). Additionally, teplizumab delayed time to diagnosis by 24 months in a phase II trial conducted in individuals at high-risk for developing T1D (40). A decision from the FDA is anticipated in late 2022 on the approval of teplizumab for use in T1D patients (41). A second immunotherapy, golimumab, is human antitumor necrosis factor (TNF)α monoclonal antibody designed to abrogate interaction of TNFα with its receptor, thereby preventing inflammatory signaling downstream of the TNFα receptor (42). Golimumab is an FDA-approved for use in multiple autoimmune diseases including rheumatoid arthritis and ulcerative colitis (43). Although golimumab was recently shown to improve endogenous β-cell function in recently diagnosed T1D patients (44), it has not yet been approved by the FDA for use in T1D.

Another approach is the use of anti-interleukin (IL)-21 antibody in combination with the GLP1R agonist, liraglutide. This combination strategy was recently used in a clinical trial in individuals recently diagnosed with T1D (45). GLP1R agonists are known to mitigate β-cell apoptosis under stress (46) and IL-21 promotes recruitment of cytotoxic CD8+ T cells to pancreatic islets (47). The anti-IL-21 and liraglutide combination significantly blunted the T1D-related decrease in β-cell function after 1 year of treatment. There is also recent evidence that oral administration of the voltage-dependent Ca2+ channel blocker verapamil can delay T1D progression by up to 2 years and may also improve outcomes in combination therapy in T2D (48, 49). The relative contribution of cell types and pathways involved in the effects of verapamil are not yet clear, but may be due to a combination of effects on β-cells and immune cells.

In addition to methods which protect endogenous β-cells, the Edmonton protocol is an islet transplantation method that has been used to successfully treat T1D for extended durations (Fig. 1) (50). This method is useful to a subgroup of T1D patients with refractory hypoglycemia. However, the number of islet transplants performed annually is small due to limited donors, specific selection criteria, and damage during processing (51). Methods to enhance viability and function of islet tissues in vitro prior to transplantation or to promote engraftment and protect against immune destruction in vivo have also been investigated (52, 53). Examples of these approaches include antibodies against the proinflammatory cytokine interferon-γ-induced protein 10 (54), inhibitors of proapoptotic pathways (eg, caspases, JNK), catalytic antioxidants (55, 56), and anti-inflammatory treatments (α-1 antitrypsin (57, 58)). A variety of G protein–coupled receptor (GPCR) pathways also represent potential targets given their protective properties in β-cells exposed to inflammatory cytokines, including agonism of GLP1R (32, 59-61) and cholecystokinin receptors (62), modulation of neuropeptide Y receptors (63, 64), or antagonism of proinflammatory GPCRs like GPR31 (65). In a complementary approach to these methods, new approaches focused on replacing or regenerating endogenous β-cells have been under active development with potential applications in both T1D and T2D patients.

Figure 1.

Figure 1.

Current and future strategies for β-cell replacement therapies. (A) Remaining endogenous β-cells may be coaxed into self-renewal via replication-inducing drugs targeted in a cell type–specific manner. Pharmacological stimulation of β-cell neogenesis or transdifferentiation from progenitor or islet endocrine cells, respectively, is also under investigation. (B) Currently, donor human islet tissue can be transplanted into the liver through the Edmonton protocol. There is potential for expansion of islet endocrine or progenitor cells in vitro using newly discovered small molecules or hormones. Differentiation or transdifferentiation of these expanded cell populations into β-like cells could provide a supplemental source of transplantable tissues. New trials are underway utilizing stem cell–derived pancreatic endoderm or β-like cell clusters either in a ‘naked’ format transplanted via the liver portal vein, or contained within macroencapsulation devices.

Regenerative medicine is aimed at replacing or regenerating human cells with the goal of restoring or establishing normal function (66). A major goal in diabetes research is aimed at applying these methods to replace or replenish lost β-cells by promoting their proliferation or regeneration either in vivo or ex vivo, or by generating de novo β-cells from induced pluripotent stem cells. In the following sections we will begin with the process of β-cell maturation to highlight the functional characteristics and identifying features of an adult β-cell, as well as the heterogeneity within mature β-cell populations. We will also provide an in-depth discussion of β-cell proliferation and regeneration, including recent advancements in this area and how these findings may lead to a successful treatment or cure for diabetes.

β-Cell Development and Maturation

Juvenile, Immature, and Mature β-Cells

Juvenile β-cells (in humans from 1-9 years of age) respond similarly to adult β-cells in glucose-stimulated insulin secretion assays, except that juvenile human islets release less insulin (67). Additionally, juvenile human β-cells respond normally to canonical insulin secretagogues in vitro including glucose and amino acids (68). Juvenile β-cells are therefore considered functionally mature after approximately the first year of development. Mature human β-cells have an increased threshold for glucose-stimulated insulin secretion and express maturation markers, including the gene urocortin 3 (69, 70). Immature β-cells lacking urocortin 3 persist in adult mice and humans within a neogenic niche in the islet (71). These virgin cells could potentially be harnessed for β-cell neogenesis. Transcription factors have been identified that are critical during β-cell maturation, such as NKX6-1 (72), as well as those unique to mature adult β-cells, including MAFA (67) and SIX2/SIX3 (73). β-Cell differentiation is dependent upon lineage-determining transcription factors, but the development of β-cell identity is not sufficient for glucose-stimulated insulin secretion (74). Glucose-stimulated insulin secretion is dependent upon signal-dependent transcription factors, which are regulated via environmental signals (74). Guided by lineage-determining transcription factors, signal-dependent transcription factors mediate the required fine-tuning to confer functional and metabolic properties of β-cells.

β-Cell heterogeneity

Adult β-cells are heterogeneous in their functions and abilities. Some adult β-cells may have greater capacity to secrete insulin than others, and this may change over time (75). Such functional heterogeneity has been described in the case of leader/first-responder β-cells (76, 77) and hub β-cells (78) in both mouse and human islets. In addition, there is heterogeneity in the proliferative capacity of adult β-cells. Based upon this notion, the defined “maturity” of a β-cell cannot be determined only by the amount of insulin expression or the presence of specific transcription factors. For instance, 1 subpopulation of β-cells is known as “hub”’ or “leader” cells. These cells contain a lesser amount of insulin, PDX1, and NKX6.1 when compared with other β-cells (78). Hub β-cells are under active investigation, and transcriptomic distinctions between hub/leader and follower cells implicated differences in primary cilia number and length may be involved. Furthermore, 2 other subpopulations of β-cells differ in functionality and were identified in mice marked by the Wnt/planar cell polarity effector Flattop. Flattop-negative β-cells have a greater proliferative ability, while Flattop-positive cells have better functional responses (79). During times of need, such as pregnancy, Flattop-negative β-cells can proliferate and change to Flattop-positive cells to handle the altered metabolic state (79). In times of physiological stress or disease, this variability within β-cell populations is imperative in order to maintain normoglycemia (75). However, the exact mechanisms underlying the origin and durability of different β-cell subpopulations is not fully understood (75, 78, 80-85), and further investigation is warranted.

Avenues to Formation of New β-Cells

The major paths to β-cell replacement include proliferation, transdifferentiation, neogenesis, replication, and stem cell–based approaches (86-88) (Fig. 1). In replication (also referred to as proliferation), a β-cell divides into 2 new β-cells (88). During transdifferentiation, other pancreatic cells, such as ductal cells (89) or islet endocrine cells (90), can convert into β-cells. In the process of neogenesis, new β-cells are created from undifferentiated progenitors (69, 71, 85). Although transdifferentiation and neogenesis describe different mechanisms, they encompass the same outcome, which is generation of new insulin-expressing β-cells from noninsulin-expressing cells (91). In taking advantage of β-cell neogenesis and replication, the therapeutic goal is to essentially re-enact the signaling mechanisms that occur during the development of the pancreas. β-Like cells can also be generated in vitro from either human embryonic stem cells (hESCs) or human induced pluripotent stem cells (hiPSCs). hESCs are uniquely able to indefinitely self-renew and to differentiate into tissues from all 3 germ layers (mesoderm, endoderm, ectoderm) (92). hiPSCs are patient-derived somatic cells reprogrammed into pluripotent cells. Stem cell–based strategies are performed in vitro with the goal of transplanting functional β-cells into humans (93). The following 3 sections will discuss how recent findings in these areas are being developed toward diabetes therapies.

β-Cell Proliferation/Replication

Overview of Cell Cycle Regulation and β-Cell Replication

The cell cycle is a tightly controlled process. By analogy, the cyclin-dependent kinases (CDKs) can be thought of as engines of the cell cycle and the cyclins as the gas pedal, which is either engaged, to power the engine through the cell cycle, or disengaged, allowing the engine to remain in an idle state. First discovered in yeast as Cdc2 (94), CDK1 was later identified in humans in 1987 (95). CDKs drive the cell cycle through their kinase activity and are regulated by the binding of distinct cyclin proteins (96). In β-cells, the cell cycle and proliferation are regulated by many factors including, but not exclusively, nutrients (eg, glucose, amino acids), growth factors (eg, IGF-I, lactogens), and GPCRs; subsequent signaling including Akt, mTOR, MAP kinases, cAMP-PKA; and downstream effects on transcriptional regulators such as FoxO1, STAT proteins, CREB, and β-catenin (for comprehensive review see (97-99)). In most cases of diabetes, including T1D, a small population of β-cells can persist (100). Considering this, it may be possible to stimulate the replication of these cells to treat diabetes. A major roadblock to this approach is that β-cells are postmitotic and have a low replication rate throughout adult life, estimated to be ∼0.1% in mice (101). Recent work using multi-isotope imaging mass spectrometry–electron microscopy indicated that many islet β-cells are as old as the animal itself (102). These findings are in agreement with earlier estimates that adult human β-cells are long-lived and the cell population is largely stable after 20 years of age in healthy individuals (103). In the fetal and neonatal stages of development, β-cells replicate rapidly, and as adulthood is reached β-cells become quiescent (104). However, adult β-cells do replicate under certain conditions, such as pregnancy and the initial compensatory response to obesity (105).

β-Cell Replication and Quiescence

Challenges in β-cell replication

There are multiple barriers to exploiting endogenous β-cell proliferation for diabetes therapy. One challenge is that in order for this process to be possible there must be pre-existing β-cells to proliferate or β-cell precursors to differentiate. The smaller the population of remaining β-cells in a patient with diabetes, the more difficult this treatment process will be. Another challenge is that there have been no clinical trials on humans to determine an optimal therapeutic rate of β-cell replication. For example, a patient with T1D with a small population of β-cells would most likely need to take a putative β-cell proliferative drug for much longer than a T2D patient who may still have half of their β-cells remaining (20). These therapeutic rates will need to be determined via clinical trials.

As mentioned previously, there is also the issue of β-cell heterogeneity, the existence of β-cell subpopulations, and their relevance to diabetes disease states (83, 106, 107). For instance, molecular markers like Flattop mark nonproliferative islet cells (79), urocortin 3 marks mature β-cells (70), and RBP4+ β-cells correlate with reduced function (108). Determining the specific subtypes of β-cells that have the ability to proliferate may be critical when populations of these cells are small, as in diabetes. Such knowledge would inform studies with proliferative drugs which could be most effective if they are designed to target those specific subtypes. Further, inflammation in T1D is not uniform throughout the islet and some regions may be free of inflammation (109). These findings support the idea that there may be a subset of cells which are resistant to immune killing. A 2017 study identified a subpopulation of β-cells during the progression of T1D in NOD (nonobese diabetic) mice that survived immune attack (110). These novel β-cells had stem-like features, were less differentiated, had lower expression of mature β-cell markers, and contained lower granularity (110). In line with this idea, the autoimmune destruction of human β-cells in T1D is not always complete, and some individuals with longstanding disease retain detectable insulin concentrations (111, 112). As researchers continue to identify and characterize β-cells in these systems, proliferative treatments may be more likely to succeed. A final issue which requires consideration is that any drug which induces proliferation also has the potential to cause cancer. Hence, it is imperative that these drugs be extremely specific and well tested.

Mechanisms underlying β-cell replication and quiescence

After maturation, β-cells eventually become quiescent (104). Knowledge of the mechanistic underpinnings of β-cell quiescence is important in the quest to determine methods to stimulate β-cell proliferation. Over the course of evolution, adaptations arose to prevent the proliferation of certain cell types (including β-cells) in adults. In the absence of these mechanisms, unregulated β-cell mass expansion can cause hyperinsulinemia and result in dangerous hypoglycemia. Examples of this can be found in endocrine cancers, such as insulinomas (113, 114), or in nesidioblastosis (115). Different pairs of CDKs and cyclins have been shown to induce β-cell proliferation (Cdk4-cyclin D1) (116), or to traffic to the β-cell cytoplasm (Cdk6-cyclin D3) (117, 118). When cyclin D3 (Ccnd3) and Cdk6 are overexpressed, they can traffic to the nucleus to cause phosphorylation of cell cycle inhibitors (eg, retinoblastoma protein Rb) and drive entry of β-cells into the cell cycle (118). Furthermore, mitogenic stimuli have been shown to “awaken” quiescent β-cells by means of controlling the expression and activation of specific cyclins and CDKs, allowing β-cells to re-enter the cell cycle. For example, GLP1R ligands can act as mitogens which promote β-cell proliferation and this has been shown to require epidermal growth factor (EGF) receptors (119-121). The effects of GLP1R agonism on proliferation were primarily observed in juvenile β-cells and required calcineurin/NFAT (nuclear factor of activated T cells) signaling to induce cyclin A (CCNA1) and CDK1 expression (122).

Development of Pharmacological β-Cell Protectants and Proliferation Inducers

Early efforts to stimulate β-cell proliferation via pharmacological methods were quite challenging, but the advent of high throughput chemical screening identified agents that stimulate human β-cell proliferation in vitro (123, 124). Small molecules modulating a variety of targets have been discovered to alter β-cell proliferation including the kinases DYRK1A, adenosine kinase, SIK, and glucokinase; receptors for transforming growth factor (TGF)β, EGF, insulin, glucagon, GLP1, and prolactin; and oligonucleotide-mediated depletion of cell cycle regulators (Table 1).

Table 1.

β-Cell protection, proliferation and regeneration modulating agents

Agent/chemical/factor Agent family Target(s) or Pathway(s) Target class Effect of agent at target Target tissue(s) Effects on tissue References
Aminopyrazine (GNF7156, GNF4877) Small molecule DYRK1A, GSK3β Kinase Inhibition β-Cell ↑ β-cell proliferation (130)
6-Azaindole derivative (GNF2133) Small molecule DYRK1A, GSK3β Kinase Inhibition β-Cell ↑ β-cell proliferation (130)
CC-401 Small molecule DYRK1A/B Kinase Inhibition β-Cell ↑ β-cell proliferation (130)
1,5-Naphthridine Small molecule DYRK1A Kinase Inhibition β-Cell ↑ β-cell proliferation (130)
Denosumab Monoclonal antibody RANKL Cytokine Inhibition circulation ↑ β-cell proliferation (190)
Osteoprotegerin Endogenous humoral RANKL, GSK3 Kinase Inhibition β-Cell ↑ β-cell proliferation (190)
Harmine Small molecule DYRK1A Kinase Inhibition β-Cell ↑ β-cell proliferation (130)
5-Iodotubercidin (5-IT) Small molecule DYRK1A/adenosine kinase Kinase Inhibition β-Cell ↑ β-cell proliferation (130, 146)
Serpin B1 Endogenous humoral proteases Protease Inhibition β-Cell ↑ β-cell proliferation (85, 167)
GLP1R agonist/DYRK1A inhibitor combination Pharmacological co-treatment GLP1R/DYRK1A GPCR/kinase Inhibition β-Cell ↑ β-cell proliferation (137)
HB-EGF Endogenous humoral EGFR Kinase Inhibition β-Cell ↑ β-cell proliferation (134)
WS6 Small molecule EGFR/NFkB pathways Kinase Activation β-Cell ↑ β-cell proliferation (288, 289)
Glucose Endogenous metabolite glycolysis, IRS2, mTOR, cyclin D2 Metabolism Activation β-Cell ↑ β-cell proliferation (290-293)
Glucokinase activators (eg, TTP399; Ro28) Small molecule glucokinase Glycolysis Activation β-Cell, liver ↑ β-cell proliferation (149-151, 294)
Placental lactogen; prolactin Endogenous humoral Prolactin receptor and growth hormone receptor (JAK-STAT signaling) Kinase Inhibition β-Cell ↑ β-cell proliferation (175, 295)
S961; OSI-906 Synthetic peptide Insulin/IGF1 receptors Kinase Inhibition Liver, adipose ↑ β-cell proliferation (162-164)
ABT-702 Small molecule Adenosine kinase Kinase Activation ↑ β-cell proliferation (146)
A-134974 Small molecule Adenosine kinase Kinase Inhibition β-Cell ↑ β-cell proliferation
5′-N-ethylcarboxamide adenosine (NECA) Small molecule ADORA2A Gpcr Inhibition β-Cell ↑ β-cell proliferation (147)
HG-9-91-01 Small molecule SIK1/2; RIPK3 Kinase Inhibition β-Cell ↑ β-cell proliferation (concentration-dependent) (140-142)
GLP1R agonist (eg, exendin-4; liraglutide) Synthetic peptide GLP1R Gpcr Synergistic activation β-Cell ↑ β-cell proliferation; β-cell protection (46, 119, 121, 122, 296)
AS1842856 Small molecule FoxO1 Transcription factor Inhibition Multiple ↑ β-cell transdifferentiation (225, 226)
IGFBP1 Endogenous humoral IGFR signaling Kinase α-Cell, β-cell ↑ β-cell transdifferentiation (206)
Glucagon receptor antibodies Antibody GCGR signaling Gpcr Activation Liver, α-cell (indirect) ↑ α-cell proliferation, β-cell transdifferentiation (211, 215, 216)
GLP1-estrogen Small molecule-linked peptide GLP1R/ER GPCR/nuclear receptor Inhibition β-Cell, brain β-Cell protection (195-197)
GLP1-ASOs Oligonucleotide-linked peptide CHOP Transcription factor Activation β-Cell β-Cell protection (188)
Anti-IL-21/liraglutide Peptide/antibody co-treatment IL-21 Cytokine; GPCR Inhibition; activation Circulation; β-cell β-Cell protection (45)
Golimumab Antibody TNFα Cytokine Inhibition Circulation β-Cell protection (42-44)
Verapamil Small molecule L-type Ca2+ channel Ca2+ channels Inhibition Multiple β-Cell protection (48, 49)
sCCK-8 Synthetic peptide CCKRB Gpcr Activation β-Cell β-Cell protection (62)
Neuropeptide Y Synthetic peptide NPYRs Gpcr Activation/inhibition β-Cell β-Cell protection (63, 64)
Anti-IP-10 Antibody IP-10/CXCL10 Cytokine Inhibition Circulation β-Cell protection (54)
α-1 Antitrypsin Purified protein Proteases Protease Inhibition Circulation β-Cell protection (57, 58)
BMS-986165 Small molecule TYK2 Kinase Inhibition Multiple β-Cell protection (281)
Teplizumab Antibody CD3 T cell receptor Activation Regulatory T cells ↑ immune tolerance; β-cell protection (37, 39-41)
Fabkin Endogenous humoral P2Y1 Gpcr β-Cell ↓ β-cell function ↑ β-cell death (148)
Insulin Endogenous humoral Insulin receptor signaling Kinase Inhibition Liver, β-cell Regulate β-cell mass in some models (160, 291, 295, 297)
CID661578 Small molecule MNK2 Kinase Block binding to eif4g β-Cell ↑ β-cell neogenesis from duct (251)
(R)-DRF053; roscovitine Small molecule Cdk5 Kinase Multiple ↑ β-cell neogenesis from duct (232)
GABA Endogenous small molecule GABAAR; GABABR; metabolism Ligand-gated Cl channel, GPCR Synergistic activation/inhibition β-Cell Variable observations in β-cell neogenesis (178, 219-222, 224)

Different small molecules, peptides, proteins, antibodies, oligonucleotides, or combination therapeutics have been demonstrated to affect β-cell proliferation, regeneration/neogenesis, or to protect β-cell function or viability from stressors. The targets of these agents fall into a variety of classes including GPCRs, kinases, proteases, or other circulating factors. While some combinations of agents have been tested, most have been tested alone, leaving the possibility of synergy between multiple different agents targeted against independent pathways for selective β-cell effects.

Kinase targets in β-cell proliferation

DYRK1A

Inhibitors of DYRK1A (dual-specificity tyrosine phosphorylation regulated kinase 1A) have been widely studied (80, 125-128) and have been shown to stimulate the highest β-cell proliferation rates through inhibition of calcineurin/NFAT/DYRK1A signaling (125-127, 129). DYRK1A inhibitors include aminopyrazines, harmine, 5-iodotubercidin, GNF2133, 1,5-naphridine, and CC-401 (85, 130). In 2015, Wang et al used a high-throughput chemical screen to identify analogs of harmine that could be useful therapeutic drugs in the treatment of diabetes (125). This study identified the likely target of harmine to be DYRK1A and downstream signaling to NFAT as the mechanism by which it induced proliferation and differentiation of mouse and human β-cells. Harmine treatment caused an expansion of islet mass and improved glucose control in mouse models of diabetes (125). Harmine was further confirmed to induce human β-cell proliferation (127, 131). Harmine is also known to target multiple proteins, warranting the development of analogs that may improve specificity and potency (132, 133). A recent study further investigated the effects of harmine, glucose, and heparin-binding epidermal growth factor-like growth factor (HB-EGF) (134). β-Cells were stimulated by harmine and HB-EGF, but these molecules also both increased the proliferation of insulin-negative and glucagon-negative cells. Harmine also greatly stimulated the proliferation of α-cells. Proliferation due to the presence of glucose was dependent upon both the donor and the performed assay. This study also highlights the need for in-depth identification of cell types using multiple markers, as non-β-cells may be misidentified as β-cells (134). DYRK1A is not β-cell specific, but rather expressed widely and is known to be associated with other diseases (135). This is a safety issue that must be overcome with future research. One strategy is to exploit synergistic effects of multiple β-cell proliferation-inducing pathways. For example, GLP1R agonists have been attributed to the promotion of cell survival as well as reducing β-cell ER stress (46, 136). There is a potentiating effect when using DYRK1A inhibitors in combination with GLP1R agonists (137), as well as when combined with TGFβ pathway inhibitors (130, 138). This combination method also allowed for lower dosage of each agent, limiting unintended effects because these are not β-cell–specific pathways. This is a significant finding, considering that a deficiency, excess, or dysregulation of DYRK1A has been associated with central nervous system effects (139). Although effective at stimulating β-cell proliferation, it is not known whether DYRK1A inhibitors will be safe in humans. The utility of DYRK1A inhibition will be unclear until there are data on human safety and preclinical studies on animals (130).

Salt-inducible kinases

Among the 3 salt-inducible kinases (SIKs) (SIK1, SIK2, SIK3), SIK2 has been shown to be important for β-cell function in mice, as β-cell–specific Sik2 knockout animals become glucose intolerant due to impaired insulin secretion (140). Nevertheless, recent work from Charbord et al found pharmacological inhibition of SIKs to induce human β-cell proliferation (141). In a separate study by Iorio et al, human β-cell proliferation was shown to be induced via targeting the GPCR GPR3-SIK2 pathway (142). In their study, GPR3 silencing induced human β-cell proliferation by increasing SIK2 activity. Silencing of SIK2 only, but not SIK1 or SIK3, prevented increased proliferation rates in GPR3-silenced β-cells (142). Overexpression of SIK2 had the opposite effect. Interestingly, in the study by Iorio et al, the pan-SIK inhibitor HG-9-91-01 at 2 µM reduced proliferation caused by GPR3 silencing, while in the study by Charbord et al, the same inhibitor (but at 0.1-1 µM) was found to induce proliferation in zebrafish, mouse, and human β-cells; at 3 µM HG-9-91-01 had little impact on proliferation. It is possible these discrepancies are due to differential effects of SIK family members SIK1 (143) and SIK2 (140), acute sensitivity of β-cells to the concentration of HG-9-91-01, the potential polypharmacology of the drug, or different proliferation assay protocols (SV40 T-antigen expression and EdU+/Ins+ incorporation in Iorio vs Ki67+/Ins+ staining in Charbord). Indeed, HG-9-91-01 was also recently shown to inhibit the kinase RIPK3 at similar dose ranges (0.5-5 µM) (144), and RIPK3 is a critical regulator of TNFα-induced β-cell death in T1D mouse models (145). Future studies of the SIK family kinases and their related pathways will be important for progressing potential human β-cell therapeutics.

Adenosine kinase

Inhibitors of adenosine kinase were discovered as β-cell regeneration/proliferation inducers using a primary rat islet screening platform (146) and through a high-throughput in vivo zebrafish screen (147). In the rat islet screen, 2 adenosine kinase inhibitors were identified, 5-iodotubercidin and ABT-702, and were shown to act on β-cell proliferation via a nuclear isoform of ADK and in an mTOR-dependent manner (146). In the zebrafish screen, among the top hits were adenosine kinase inhibitor A-134974 and adenosine receptor agonist 5′-N-ethylcarboxamide adenosine (NECA). The authors focused on NECA which they determined to promote mouse β-cell proliferation/regeneration in an ADORA2A-dependent mechanism (147). Related to findings with adenosine kinase, a recent investigation discovered a circulating complex, named Fabkin, comprised of fatty acid–binding protein 4, adenosine kinase, and nucleoside diphosphate kinase (148). This complex was suggested to inhibit β-cell function and promote cell death, in part through alteration of ADP/ATP ratio near β-cells and modulation of signaling through purinergic receptors. Overall, targeting adenosine signaling pathway components appears to be a promising area of research which contributes to our understanding of β-cell proliferation and regeneration.

Glucokinase

Glucokinase has also been shown to be critical in β-cell proliferation using rodent models, where deletion prevented β-cell replication and provision of a small molecule glucokinase activator (Ro28-1675) enhanced replication (149). While most glucokinase activator trials have been aimed at human T2D (36), there have been clinical trials in T1D using a hepatoselective activator, TTP399, which resulted in improved glycemic control (150, 151) (ClinicalTrials.gov identifier: NCT03335371). Other glucokinase activators (Table 1) have been shown to enhance human β-cell proliferation in a PKCζ-dependent manner (152), but have yet to be tested in human trials.

Humoral factors in β-cell proliferation

Insulin

While insulin is an endocrine growth factor released from β-cells, it can also signal in an autocrine/paracrine manner. It follows that components of the insulin receptor signaling pathway have been found to be involved in regulation of β-cell mass and expansion. As examples, β-cell mass has been shown to be decreased in a variety of models including β-cell–specific insulin receptor knockout (βIRKO) mice (153, 154), IRS-2 whole-body knockout mice (155), β-cell–specific PDK1 knockout mice (156), and S6K1 knockout mice (157). Alternatively, heterozygous deletion of insulin receptors, IRS-1 and IRS-2, cause increases in β-cell mass (158), while dominant-negative Akt transgenic animals had no β-cell mass changes (159). In the case of insulin receptor, careful investigations in newer β-cell–specific genetic mouse models have found a more nuanced role for β-cell insulin receptor. Skovso et al deleted insulin receptor from β-cells using Ins1-Cre and observed no alterations in β-cell mass, but instead observed increased glucose-stimulated insulin secretion, in support of β-cell insulin resistance causing hyperinsulinemia (160).

Insulin resistance also plays a role in the proliferation of β-cells. This is shown, for example, by studies in mice with liver-specific insulin receptor knockout (161), or antagonist peptides S961 against the insulin receptor (162) and OSI-906 against both insulin receptor and insulin-like growth factor 1 receptor (163, 164). Under these types of conditions, there is evidence that the liver may generate a signal that leads to β-cell expansion (165, 166), but the identities of putative secreted factors are still under investigation. One candidate, Serpin B1, a hepatocyte-secreted protease inhibitor, is a molecule which has been linked to β-cell proliferation in humans, mice, pigs, and zebrafish (167, 168) (Table 1). Additionally, serum Serpin B1 was also found to be decreased in T2D subjects (169), and, under conditions of insulin resistance, a deficiency of Serpin B1 has been shown to be related to reduced β-cell proliferation (167). Because Serpin B1 is a protease inhibitor, the implication is there is a specific protease involved in the inhibition of β-cell proliferation. Indeed, Serpin B1 was found to inhibit the protease elastase to promote human β-cell proliferation (167). Recent studies using of S961 and OSI-906 in β-cell–specific insulin receptor knockout mice have identified the existence of an adipocyte-derived factor which acts through β-cell E2F transcription factor 1 (E2F1) to induce proliferation (170). While the identity of the putative circulating factor is not known, the β-cell proliferative effects required CENP-A and E2F1, potentially upstream of the master regulator transcription factor FoxM1 (171, 172).

Other humoral contributors

Early studies of nerve growth factor suggested that it enhanced β-cell proliferation via promoting production of activin A and betacellulin, an EGF receptor ligand (173). Additionally, EGF-mediated expansion of other cell types, such as pancreatic duct cells (174), has been explored for its transdifferentiation potential. More recently, heparin-binding EGF was shown to induce both EdU incorporation and Ki67+ staining of human β-cells, but also caused proliferation of other non-β-cell types (134). Indeed, as noted earlier, EGF receptor can also be transactivated by GLP1R agonists to induce β-cell proliferation (119, 120).

Multiple other circulating factors have been explored for their potential to induce β-cell proliferation/regeneration, including lactogens (175, 176) as well as neurotransmitters like serotonin (177) and γ-amino butyric acid (GABA) (178). However, later testing of many of these factors did not induce proliferation in human β-cells using Ki67+ as a readout, while the DYRK1A inhibitor harmine did (128). A recent study using single-cell RNA sequencing of human stem cell–derived β-cells found a subpopulation of proliferative cells which responded to leukemia inhibitor factor (179). This work revealed additional targetable pathways including STAT3 and CEBPD downstream of leukemia inhibitor factor which could be exploited for future β-cell proliferative strategies.

UPRER-related factors in β-cell proliferation

Insulin hypersecretion is known to elicit the unfolded protein response of the endoplasmic reticulum (UPRER) in β-cells (180), and the UPRER is involved in disease progression in T1D β-cells (181). MANF, previously known as ARMET, is a secreted protein and a prosurvival UPRER factor in β-cells (182-184). Whole-body deletion or β-cell–specific deletion of MANF in mice leads to reduced β-cell mass and diabetes (184, 185). Stimulation with MANF in vitro stimulated mouse β-cell proliferation (185), and also human β-cell proliferation under conditions of TGFβ inhibition (183). Unmitigated β-cell ER stress will lead to a terminal UPRER, involving the transcription factor C/EBP homologous protein (CHOP), encoded by Ddit3/DDIT3 (186). Prolonged CHOP expression will eventually induce apoptosis in β-cells. In mice with β-cell–specific depletion of CHOP, or knockdown of CHOP by GLP1-linked antisense oligonucleotides (GLP1-ASOs), expansion of β-cell mass and improved function is observed, respectively (187, 188). Alternatively, suppressing β-cell activity using inhibitory Gi/o–GPCR signaling, for instance through the α2-adrenergic receptor, restricts β-cell expansion perinatally and inhibition of this pathway specifically in β-cells can promote their expansion (189).

Other β-cell protectants and proliferation inducers

Inhibitors of the NFκB pathway, specifically osteoprotegerin and denosumab (Table 1), have also effectively induced proliferation of β-cells (190). Similarly, suppression of the cell cycle regulators CDKN2C/p18 and CDKN1A/p21 successfully induced replication of human β-cells (191). Under diabetic conditions, the high demand for secretion of insulin itself may also cause ER stress and this is relieved by the genetic deletion of both isoforms of insulin in mice, thereby promoting the ability of β-cells to proliferate via the Akt-Cyclin D1 axis (192).

Given the separately known β-cell protective effects of both GLP1 (46) and estrogen (193, 194), these 2 molecules have been covalently linked and tested by multiple groups in diabetes models (195-197). The combined results have shown significant body weight reduction in rodents (197) combined with protection of β-cells in streptozotocin-treated mouse models (195) and cytokine-treated human islet β-cells (196). Recent findings also show that suppressing expression of DDIT3, which encodes the terminal UPRER transcription factor CHOP, in vivo in β-cells using GLP1-ASOs prevented loss of β-cell function in mouse models of diabetes (188). GLP1-ASOs activate GLP1R and become internalized to deliver the conjugated oligonucleotide (198). In addition, GLP1 has been covalently linked to PPARα/γ dual agonists as a diabetes therapy (199). Each of these efforts support the overall concept that linking GLP1 to protective or proliferation-inducing therapies can be used to more specifically target the β-cell in vivo.

Advances in β-Cell Transdifferentiation and Neogenesis

Stimulating proliferation of endogenous insulin-producing β-cells may be a promising approach to treat diabetes. However, in millions of diabetes cases, most notably T1D, β-cells are nearly completely absent. Without sufficient existing β-cells to proliferate, this approach becomes hampered. Therefore, generation of new β-cells via transdifferentiation or neogenesis may be a viable approach. Sources of cells that have been demonstrated to have β-cell transdifferentiation potential include α-cells, δ-cells, PP cells, and acinar cells; while cells with neogenic potential include pancreatic ductal progenitors and niches of immature β-cell populations (Fig. 1).

Transdifferentiation of β-Cells

The posterior foregut region of the developing embryo is the region which gives rise to the pancreas. The posterior foregut is also the origin of the liver, the posterior part of the stomach, and the proximal region of the gut (200). Considering that these regions are similarly related in their cellular makeup, focusing on cell types from these regions is a rational approach to identifying potential candidates to transdifferentiate into functional β-cells.

α-Cells

A major cell type in β-cell transdifferentiation research is the pancreatic islet α-cell, which can convert to insulin-producing β-cells under certain conditions. Additionally, cells in an intermediate phase of α to β transdifferentiation have been shown to comprise a neogenic niche of immature β-cells at the islet periphery, representing a potentially targetable pool of β-cell progenitors in humans and mice (69-71). To understand mechanisms of α- to β-cell transdifferentiation, studies of transcription factors involved in islet endocrine cell differentiation found opposing roles for Aristaless-related homeobox (Arx) and Pax4 (201). Arx expression positively correlated with α-cell number, while Pax4 repressed α-cell number and increased the numbers of β- and δ-cells. These findings indicate that the relative amount of α- and β-cells is related to the expression of Arx or Pax4, respectively. Indeed, a gain-of-function mutation in mouse Arx resulted in the loss of β- and δ-cells and a gain in the number of α-cells and PP cells (202). Alternatively, in a mouse model of β-cell ablation, lineage-tracing showed many β-cells regenerated from α-cells (90). Deleting Arx specifically in α-cells early in development promoted their transdifferentiation into β-cells (203). In conditional Arx α-cell knockout mice, α-cells readily converted to β-cells even upon Arx deletion in adult animals. Consequently, inhibition of Arx was suggested to be a promising area of future research and in the treatment of diabetes. Further work in adult mice indicated that α-cell–specific deletion of both Arx and DNA methyltransferase 1 (Dnmt1) promoted α- to β-cell conversion (204). α-Cell–specific deletion of Arx alone caused a loss of α-cell identity, while loss of Dnmt1 alone did not impact α-cell identity. Glucose-stimulated insulin release was also observed after this conversion, suggesting the generation of functional β-cells (204). In T1D, some glucagon+ cells show a loss of ARX and DNMT1, and produce β-cell products such as insulin. These results suggest that, in humans, ARX and DNMT1 are associated with the maintenance of α-cell identity (204). Notably, another DNA methyltransferase, Dnmt3a, is important for methylating the Arx promoter in rodent β-cells to maintain β-cell identity (205). α-Cells may also be transdifferentiated to β-cells by endogenous factors. The secreted protein IGFBP1 was found in zebrafish to promote α- to β-cell transdifferentiation, and circulating IGFBP1 levels in humans were inversely correlated with T2D risk (206). IGFBP1 was presumed to act through suppressing IGF receptor signaling, as β-cell proliferation effects were also seen with 2 other IGF receptor inhibitors (206).

An in vivo mouse study in 2018 showed adenoviral expression of Pdx1 and MafA delivered to the pancreas via the bile duct caused α to β conversion and reversed diabetes (207). In vitro, overexpression of Pdx1 and MafA in human islets induced insulin expression in α-cells (207). In a separate study, Pdx1 and MafA overexpression in human α-cells resulted in the production and glucose-sensitive secretion of insulin, but the cells maintained α-cell characteristics including expression of Arx (208). Transplantation of these reprogrammed α-cells into streptozotocin-treated mice mitigated diabetes and the grafts continued to express insulin for at least 6 months (208).

Another approach under investigation to regenerate β-cells is via modulating α-cell function. Beneficial effects have been found for injected antiglucagon receptor antibodies in humans with T1D (209) as well as nonhuman primates (210, 211) and rodents (212). Disruption of glucagon receptor activation either genetically or through use of injected antibodies results in α-cell hyperplasia (213, 214), offering a potential source of new β-cells if they can be transdifferentiated. Multiple recent studies in mouse models of T1D and T2D have suggested that a combination of α- to β-cell transdifferentiation and β-cell replication can occur in response to glucagon receptor antagonist antibodies (211, 215-217). These promising findings await further confirmation in humans (218).

Lastly, GABA has been studied for its potential impacts on α to β transdifferentiation. GABA induced β-cell proliferation of human islets transplanted into streptozotocin-treated NOD-SCID mice and in vitro (219). Ben-Othman et al found that long-term GABA administration was also suggested to induce β-cell neogenesis from α-cells both in mice and in transplanted human islets (178). A different study published in the same issue suggested the artemisinin class of antimalaria drugs could promote α to β transdifferentiation through activation of GABA receptor signaling (220). However, follow-up studies indicated that artemisinins promote islet endocrine cell dedifferentiation and not β-cell transdifferentiation (221) and that long-term treatment with GABA or artemisinins had no impact on α to β transdifferentiation in mice (222). Nevertheless, recent clinical trials (ClinicalTrials.gov identifier: NCT03635437 and ClinicalTrialsregister.eu identifier: EudraCT2018-001115-73) found GABA administration to be effective for improving the counterregulatory response to hypoglycemia in humans with T1D (223), but direct effects on α- or β-cells were not studied. Despite disparate findings with regard to transdifferentiation, GABA clearly has a role in islet function (see detailed review (224)).

δ-Cells and PP cells

Besides α-cells, islets contain other endocrine cell types that can transdifferentiate into β-cells, including δ-cells and γ-cells (also known as PP cells).

While α to β transdifferentiation occurs in adult animals after β-cell ablation (90), in juvenile mice β-cell transdifferentiation was found to occur through δ-cells and not α-cells (225). This process involved suppression of amitogenic FoxO1 signaling; pharmacological FoxO1 inhibition (AS1842856 (226)) potentiated the transdifferentiation process. A similar process has been described in zebrafish, where β-cell ablation led to the appearance of somatostatin+/insulin+ cells which derived from a distinct δ-cell subtype with pro-β identity (227, 228). As these animals recovered normoglycemia, it was suggested that bihormonal cells are functional and important. Single-cell transcriptomic comparison analysis of zebrafish islets to human islet cells suggested this subtype of δ-cell has similarity to human γ-cells (227). Interestingly, lineage-tracing studies of mouse γ-cells showed these cells can also transdifferentiate to different bihormonal cell types including PPY+/insulin+ cells, and these bihormonal γ-cells were also found in human islets (229).

Acinar and antral stomach cells

Viral gene therapy is an approach that has been successful in mouse models of T1D. In vivo transduction of pancreatic acinar cells with the combination of Ngn3, Pdx1, and MafA led to cellular reprogramming and expression of a β-cell–like phenotype (230). Another source of tissue for β-cell production is the antral stomach. Transgenic overexpression of this same trio of factors, Pdx1, MafA, and Ngn3, in antral stomach tissue resulted in the induction of insulin-positive cells with the ability to rescue mice from streptozotocin-induced diabetes (231). The therapeutic value of this approach was demonstrated by isolation of the transgenic stomach tissue, growth as organoids, and transplantation into diabetic mice prior to induction of Pdx1, MafA, and Ngn3. Mice were rescued from diabetes in those experiments (231).

Differentiation of β-Cells From Progenitors

Cdk5 inhibitors have recently been shown to induce β-cell differentiation from ductal progenitor cells in zebrafish, mice and hiPSCs (232). Additionally, when an adult mouse pancreas is injured, Ngn3+ endocrine progenitors have the ability to differentiate into all of the islet cell types through the Notch signaling pathway (233-235). Although these findings arose from studies on rodents, they provide valuable insight that may be translated to human diabetes research. There is an evident role of multipotent pancreatic progenitors during embryonic development. While the existence of a pancreatic islet stem cell that could be targeted in adult humans remains controversial (91), some studies suggest this possibility. A 2020 study used single-cell RNA sequencing to identify a population of mouse islet cells termed protein C receptor-positive (Procr+) cells (236). These Procr+ cells were suggested to act as progenitors with the ability to be passaged as organoids in vitro and differentiate into each of the islet endocrine cell types. However, other analyses of mouse islet single-cell RNA-sequencing did not detect this Procr+ population (237).

The potential of pancreatic ductal cells as a source of β-cell progenitors has been investigated and debated (238-242). Overall, observing substantial β-cell neogenesis from ductal cells seems to require special conditions, such as surgical damage to the pancreas or genetic modifications. Pancreatic ductal cells have the ability to regenerate both endocrine and exocrine tissue in rodents after duct ligation or pancreatectomy (234, 243), or by diphtheria toxin–based ablation of exocrine and ductal tissue (244). Sancho et al found that adult ductal cells can be reprogrammed to multiple islet endocrine cell types (α, β, δ) by deleting Fbw7 in the pancreas (245). They also found that as a ubiquitin ligase component, Fbw7 regulates the stability of Ngn3. Ngn3 is a critical transcription factor required for the development of the pancreatic endocrine compartment (246). Additionally, lineage tracing showed some ductal cells express Ngn3, and over time these Ngn3+ ductal cells expressed somatostatin and a subset eventually expressed insulin, suggesting a role in β-cell neogenesis (247). Most of this understanding comes from work in rodents; however, human pancreatic ductal cells can gain β-cell identity when transplanted into mouse models of insulin resistance (248), and ductal cells can express immature β-cell markers in pregnant humans (249). There is now mounting evidence in the field for these ductal progenitors in human pancreas (250), and it may be possible to target such progenitors in diabetic patients to generate new β-cells (Fig. 1). Indeed, pharmacological modulation of MAPK interacting Ser/Thr kinases 2 (MNK2) was discovered to be responsible for β-cell neogenesis from pancreatic ductal cells in vivo (zebrafish) and in vitro (pigs and humans) (251). A role for MNK2 was discovered in this study by deconvoluting the target of the small molecule CID661578. CID661578 turned out to be a MNK2-interacting compound which increased protein synthesis by preventing MNK2 from binding eIF4G of the translation initiation complex. This alteration of MNK2 signaling and enhanced cap-dependent mRNA translation was hypothesized to contribute to the β-cell neogenic effect.

Pluripotent Stem Cell–Derived β-Cells to Replace Endogenous β-Cell Mass

hESCs and hiPSCs are similar in their abilities to differentiate into many different cell types (93). Therefore, these stem cells are excellent candidates for generating insulin producing β-cells. In 1998, Thomson and colleagues derived hESCs, a pluripotent stem cell derived from the inner cell mass of the blastocyst with the ability to give rise to any cell type of the 3 human germ layers (252). Thomson used donated human embryos, which became available to him from unused fertility treatments (252). Despite ethical implications, multiple developments have been made using hESCs as precursors in the formation of insulin producing β-cells (253). Alternatively, hiPSCs are reprogrammed from somatic dermal fibroblasts or peripheral blood mononuclear cells (254). In 2006-2007, the Yamanaka laboratory pioneered a somatic cell reprogramming process by expressing 4 specific genes in mouse and human fibroblasts (255, 256). The overexpression of these specific genes (OCT4, SOX2, KLF4, and CMYC) was necessary and sufficient to induce pluripotency (256). These studies were a milestone in stem cell research, as it enabled the creation of pluripotent stem cells without the use of human embryos. Like hESCs, hiPSCs have the ability to differentiate into multiple cell types. hESCs and hiPSCs can be coaxed through a complex multistage process to result in insulin-producing β-like cells. Typically stem cells must be subjected to a weeks-long multi-step process first requiring generation of definitive endoderm (257), followed by primitive gut tube cells and pancreatic progenitors (258), and finally immature pancreatic endocrine cells before differentiation of β-like cells (259-261). There have been limitations with stem cell–derived β-like cells, notably reduced insulin production and secretion as compared to primary β-cells. However, the process has been further refined to permit more steps to occur in monolayer culture (262, 263), and recent work has shown stem cell–derived islet function quite close to that of primary human islets (264). Additional work has demonstrated generation of enhanced β-cell clusters (eBCs) by reaggregating FACS-enriched hESC-derived β-like cells (265). These eBCs had improved mitochondrial respiration and increased similarity to endogenous β-cells in terms of function, such as dynamic insulin secretion. Further work with eBCs determined that self-aggregation yielded development of more mature self-enriched β-cell clusters (seBCs) (266). It was discovered that the most mature of these β-like cells are marked by a cell surface protein, ENTPD3. This study, and others like it (267), which provide large transcriptomic datasets will aid in discovering relevant pathways and markers to improve mature β-cell differentiation in vitro.

The multistep differentiation protocols which coax hESCs and hiPSCs into β-like cells require the modulation of many signaling pathways at each step. Furthering understanding of the signaling pathways governing β-cell differentiation and maturation provides the necessary data to improve stem cell–derived β-cell generation. Recent investigations of TGFβ, Wnt, and the F-actin cytoskeleton are prime examples. TGFβ signaling has been shown to have an effect on developing and adult β-cells, as removing the actions of TGFβ inhibitors can promote the β-cell maturation process (268). Indeed, the TGFB2 gene was found to be upregulated in ENTPD3+ eBCs (266). Wnt pathway signaling is also involved in pancreatic differentiation and β-cell maturation (79, 269, 270). The Wnt family member Wnt4 is highly expressed in human islets and inclusion of Wnt4 in the terminal stages of hiPSC-derived β-cell maturation improved production of insulin and urocortin-3 (269). Wnt4 was also recently shown to be involved in β-cell heterogeneity, with Wnt4+ cells being more mature and Wnt4 cells being more proliferative (271). Finally, a very useful discovery was made by the Millman laboratory, which found that adding the F-actin depolymerizing agent latrunculin A at a precise stage of the differentiation protocol improved expression of NEUROG3 and subsequent efficiency of stem cell–derived β-cells from both hESCs and hiPSCs (262, 263). Identifying new markers of β-cell maturation and innovative technical advancements like above will advance the creation of replacement cell therapies that are increasingly similar to endogenous human islets.

Completed and ongoing clinical trials are testing the potential to use stem cell–derived pancreatic endoderm and β-like cells by transplanting either encapsulated or naked tissues. For example, ViaCyte has developed pancreatic endoderm cells (PEC-01) from an hESC line CyT49 (272); the PEC-01 cells mature to β-cells after implantation. Initial tested devices for implanting PEC-01 cells (VC-01) failed due to insufficient function after implantation due to hypoxia and necrosis (ClinicalTrials.gov identifier: NCT02239354). Therefore, a new PEC-Encap (VC01-103) semipermeable macroencapsulation device was designed to prevent access to the immune system, but retain improved permeability to prevent hypoxia within the implant. The improved PEC-Encap device is in phase 2 clinical trials with results anticipated in 2023 (ClinicalTrials.gov identifier: NCT04678557). In addition, ViaCyte is testing PEC-01 cells contained an alternate device with pores for capillary access and improved β-cell maturation (VC-02) (273, 274), which is also in phase 2 trials (ClinicalTrials.gov identifier: NCT03163511). VC-02 requires immunosuppression because of the circulatory access of the device. Finally, a new experimental treatment, VCTX210, developed in a collaboration between ViaCyte and CRISPR Therapeutics, uses the CyT49 hESCs (272) engineered with CRISPR gene editing to be immune evasive and differentiated to pancreatic endoderm (PEC-QT), which are contained within a macroencapsulation device for implantation. VCTX201 is currently being tested in an ongoing clinical trial (ClinicalTrials.gov identifier: NCT05210530).

An alternative strategy from Vertex Pharmaceuticals, VX-880, uses naked stem cell–derived β-like cells transplanted into the liver via the portal vein, similar to how primary human islet transplants are performed (ClinicalTrials.gov identifier: NCT04786262). Therefore, this approach still requires immunosuppressive drugs to prevent tissue destruction. Current results are limited to only a few patients, and while the study has not been peer-reviewed, the grafts are reported to be tolerated and yield reduced reliance on exogenous insulin. Interestingly, Vertex recently acquired ViaCyte (275), forecasting multiple potential combination approaches between macroencapsulation technology and CRISPR-edited stem cell–derived functional β-like cells.

Ultimately, the final products of any cell replacement approach must be adapted to mass production for clinical usage, protected from the immune system either with physical barriers or immunosuppression and also ensured not to spread to the recipient any potential teratogenic or tumorigenic cells (93, 276). In addition to the kinases and signaling pathways discussed targeting β-cell proliferation or transdifferentiation, there have been advancements in targeting immune response pathways in the β-cell to protect them from immune destruction. For example, the Janus kinase family member tyrosine kinase 2 (TYK2), which mediates signaling downstream of interferon receptors, has been associated with multiple autoimmune diseases, including T1D (277-279). Knockout or pharmacological inhibition of TYK2 protects β-cells from interferon-α–mediated effects (280, 281). Other strategies to protect β-cells include genetic modification of stem cells prior to differentiation to β-like cells. Deletion of multiple human leukocyte antigen (HLA) genes, importantly while retaining one (HLA-A2), reduced immune rejection of transplanted hiPSC-derived islet cells (282).

Additional consideration should be given to islet innovation and vascularization. Endogenous islets pancreatic islets are highly innervated, influencing islet development and function (283). Islets are also vascularized continuously with the pancreatic exocrine tissue where islet-resident pericytes help regulate capillary constriction (284, 285). Recapitulating this endogenous vascularization and innervation represents a potential hurdle for transplanted tissue aimed at replacing endogenous islet function. These are challenges that must be overcome in order for such methods to be effective clinically. Advancements in producing large quantities of functional β-cells that can safely avoid destruction after engraftment are imperative for future successful treatments.

Conclusion

The potential for regenerative medicine to treat diabetes has progressed significantly in the last 40 years. Endogenous β-cell regeneration can occur via numerous methods; however, a majority of these methods have succeeded in animals but have not yet led to accessible human therapies (85). A major issue in humans is the immune attack of transplanted β-cells or the autoimmune destruction of β-cells derived from neogenesis in T1D. Strategies are under continued development to defend β-cells against these insults, including encapsulation technologies (286) and immunotherapies replenishing anti-inflammatory T regulatory cells to confer tolerance to new or transplanted β-cells (287). Despite the challenges, the future is bright in the field of regenerative medicine to treat diabetes.

Acknowledgments

We thank Dr. Barak Blum at University of Wisconsin, and Dr. Andrew Templin, Dr. Erica Cai, Dr. Li Zhang at the Indiana Bioscinces Research Institute (IBRI) for helpful discussions. We also appreciate the reviewers’ critiques, each of which improved this manuscript. During the course of this work M.A.K. had support from a Diabetes Research Connection (Project #33), Juvenile Diabetes Research Foundation Innovative Award (1-INO-2022-1113-A-N), internal support from the IBRI, and by a Pilot and Feasibility Award within the CDMD NIH/NIDDK Grant Number P30 DK097512.

Abbreviations

CDK

cyclin-dependent kinase

CHOP

C/EBP homologous protein

eBC

enhanced β-cell cluster

EGF

epidermal growth factor

GABA

γ-amino butyric acid

GLP1R

glucagon-like peptide 1 receptor

GPCR

G protein–coupled receptor

HB-EGF

heparin-binding epidermal growth factor-like growth factor

hESC

human embryonic stem cells

hiPSC

human induced pluripotent stem cell

IL

interleukin

NECA

5′-N-ethylcarboxamide adenosine

NFAT

nuclear factor of activated T cells

PP

pancreatic polypeptide

SIK

salt-inducible kinase

T1D

type 1 diabetes mellitus

T2D

type 2 diabetes mellitus

TGF

transforming growth factor

TNF

tumor necrosis factor

UPRER

unfolded protein response of the endoplasmic reticulum

Contributor Information

Roy A Goode, Division of Biomedical Sciences, College of Osteopathic Medicine, Marian University, Indianapolis, IN, USA.

Julia M Hum, Division of Biomedical Sciences, College of Osteopathic Medicine, Marian University, Indianapolis, IN, USA.

Michael A Kalwat, Lilly Diabetes Center of Excellence, Indiana Biosciences Research Institute, Indianapolis, IN, USA; Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA.

Disclosures

The authors have nothing to disclose.

Data Availability

Data sharing is not applicable to this article as no datasets were generated or analyzed during the current study.

References

  • 1. Da Silva Xavier G. The cells of the islets of Langerhans. J Clin Med. 2018;7(3):54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Kalwat MA, Cobb MH. Mechanisms of the amplifying pathway of insulin secretion in the beta cell. Pharmacol Ther. 2017;179(November):17–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. LeRoith D, Taylor SI, Olefsky JM. Diabetes Mellitus: A Fundamental and Clinical Text. 3rd ed. Lippincott Williams & Wilkins, 2004. [Google Scholar]
  • 4. Rogers MAM, Kim C, Banerjee T, Lee JM. Fluctuations in the incidence of type 1 diabetes in the United States from 2001 to 2015: a longitudinal study. BMC Med. 2017;15(1):199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Centers for Disease Control and Prevention . National Diabetes Statistics Report. Centers for Disease Control and Prevention, U.S. Dept of Health and Human Services; 2020.
  • 6. Ramos-Rodriguez M, Pérez-González B, Pasquali L. The beta-cell genomic landscape in T1D: Implications for disease pathogenesis. Curr Diab Rep. 2021;21(1):1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Nyaga DM, Vickers MH, Jefferies C, Perry JK, O'Sullivan JM. Type 1 diabetes mellitus-associated genetic variants contribute to overlapping immune regulatory networks. Front Genet. 2018;9:535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Dotta F, Censini S, van Halteren AG, Marselli L, Masini M, Dionisi S, et al. Coxsackie B4 virus infection of beta cells and natural killer cell insulitis in recent-onset type 1 diabetic patients. Proc Natl Acad Sci U S A. 2007;104(12):5115–5120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Roep BO. A viral link for type 1 diabetes. Nat Med. 2019;25(12):1816–1818. [DOI] [PubMed] [Google Scholar]
  • 10. Roep BO, Thomaidou S, van Tienhoven R, Zaldumbide A. Type 1 diabetes mellitus as a disease of the beta-cell (do not blame the immune system)? Nat Rev Endocrinol. 2020;17(3):150–161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Mallone R, Halliez C, Rui J, Herold KC. The beta-cell in type 1 diabetes pathogenesis: a victim of circumstances or an instigator of tragic events? Diabetes. 2022;71(8):1603–1610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Kahn SE, Chen YC, Esser N, Taylor AJ, van Raalte DH, Zraika S, et al. The beta cell in diabetes: integrating biomarkers with functional measures. Endocr Rev. 2021;42(5):528–583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Esser N, Utzschneider KM, Kahn SE. On the causal relationships between hyperinsulinaemia, insulin resistance, obesity and dysglycaemia in type 2 diabetes: reply to Johnson JD. Diabetologia. 2021;64(10):2345–2347. [DOI] [PubMed] [Google Scholar]
  • 14. Johnson JD. On the causal relationships between hyperinsulinaemia, insulin resistance, obesity and dysglycaemia in type 2 diabetes. Diabetologia. 2021;64(10):2138–2146. [DOI] [PubMed] [Google Scholar]
  • 15. Voight BF, Scott LJ, Steinthorsdottir V, Morris AP, Dina C, Welch RP, et al. Twelve type 2 diabetes susceptibility loci identified through large-scale association analysis. Nat Genet. 2010;42(7):579–589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Dupuis J, Langenberg C, Prokopenko I, Saxena R, Soranzo N, Jackson AU, et al. New genetic loci implicated in fasting glucose homeostasis and their impact on type 2 diabetes risk. Nat Genet. 2010;42(2):105–116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Deshmukh HA, Madsen AL, Vinuela A, Have CT, Grarup N, Tura A, et al. Genome-wide association analysis of pancreatic beta cell glucose sensitivity. J Clin Endocrinol Metab. 2020;106(1):80–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Locke JM, Hysenaj G, Wood AR, Weedon MN, Harries LW. Targeted allelic expression profiling in human islets identifies cis-regulatory effects for multiple variants identified by type 2 diabetes genome-wide association studies. Diabetes. 2015;64(4):1484–1491. [DOI] [PubMed] [Google Scholar]
  • 19. Prentki M, Nolan CJ. Islet beta cell failure in type 2 diabetes. J Clin Invest. 2006;116(7):1802–1812. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Butler AE, Janson J, Bonner-Weir S, Ritzel R, Rizza RA, Butler PC. Beta-cell deficit and increased beta-cell apoptosis in humans with type 2 diabetes. Diabetes. 2003;52(1):102–110. [DOI] [PubMed] [Google Scholar]
  • 21. Kreider KE, Gabrielski AA, Hammonds FB. Hyperglycemia syndromes. Nurs Clin North Am. 2018;53(3):303–317. [DOI] [PubMed] [Google Scholar]
  • 22. IDF . Diabetes Atlas. 10th edn. International Diabetes Federation, 2021. [Google Scholar]
  • 23. American Diabetes Association . Economic costs of diabetes in the U.S. in 2017. Diabetes Care. 2018;41(5):917–928. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Peter P, Lipska K. The rising cost of diabetes care in the USA. Lancet Diabetes Endocrinol. 2016;4(6):479–480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Silver B, Ramaiya K, Andrew SB, Fredrick O, Bajaj S, Kalra S, et al. EADSG guidelines: insulin therapy in diabetes. Diabetes Ther. 2018;9(2):449–492. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Unger RH, Cherrington AD. Glucagonocentric restructuring of diabetes: a pathophysiologic and therapeutic makeover. J Clin Invest. 2012;122(1):4–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Cryer PE. Mechanisms of hypoglycemia-associated autonomic failure in diabetes. N Engl J Med. 2013;369(4):362–372. [DOI] [PubMed] [Google Scholar]
  • 28. Brown RJ, Rother KI. Effects of beta-cell rest on beta-cell function: a review of clinical and preclinical data. Pediatr Diabetes. 2008;9(3 Pt 2):14–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Pernicova I, Korbonits M. Metformin–mode of action and clinical implications for diabetes and cancer. Nat Rev Endocrinol. 2014;10(3):143–156. [DOI] [PubMed] [Google Scholar]
  • 30. Clapham JC. Sixty years of drug discovery for type 2 diabetes: where are we now? Methods Mol Biol. 2020;2076:1–30. [DOI] [PubMed] [Google Scholar]
  • 31. Davidson MA, Mattison DR, Azoulay L, Krewski D. Thiazolidinedione drugs in the treatment of type 2 diabetes mellitus: past, present and future. Crit Rev Toxicol. 2018;48(1):52–108. [DOI] [PubMed] [Google Scholar]
  • 32. Drucker DJ. Mechanisms of action and therapeutic application of glucagon-like peptide-1. Cell Metab. 2018;27(4):740–756. [DOI] [PubMed] [Google Scholar]
  • 33. Meier JJ, Nauck MA. Incretin-based therapies: where will we be 50 years from now? Diabetologia. 2015;58(8):1745–1750. [DOI] [PubMed] [Google Scholar]
  • 34. Satin LS, Soleimanpour SA, Walker EM. New aspects of diabetes research and therapeutic development. Pharmacol Rev. 2021;73(3):1001–1015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Reiss AL, Jo B, Arbelaez AM, Tsalikian E, Buckingham B, Weinzimer SA, et al. A pilot randomized trial to examine effects of a hybrid closed-loop insulin delivery system on neurodevelopmental and cognitive outcomes in adolescents with type 1 diabetes. Nat Commun. 2022;13(1):4940. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Perreault L, Skyler JS, Rosenstock J. Novel therapies with precision mechanisms for type 2 diabetes mellitus. Nat Rev Endocrinol. 2021;17(6):364–377. [DOI] [PubMed] [Google Scholar]
  • 37. Herold KC, Hagopian W, Auger JA, Poumian-Ruiz E, Taylor L, Donaldson D, et al. Anti-CD3 monoclonal antibody in new-onset type 1 diabetes mellitus. N Engl J Med. 2002;346(22):1692–1698. [DOI] [PubMed] [Google Scholar]
  • 38. Bluestone JA, Herold K, Eisenbarth G. Genetics, pathogenesis and clinical interventions in type 1 diabetes. Nature. 2010;464(7293):1293–1300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Herold KC, Gitelman SE, Ehlers MR, Gottlieb PA, Greenbaum CJ, Hagopian W, et al. Teplizumab (anti-CD3 mAb) treatment preserves C-peptide responses in patients with new-onset type 1 diabetes in a randomized controlled trial: metabolic and immunologic features at baseline identify a subgroup of responders. Diabetes. 2013;62(11):3766–3774. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Sims EK, Bundy BN, Stier K, Serti E, Lim N, Long SA, et al. Teplizumab improves and stabilizes beta cell function in antibody-positive high-risk individuals. Sci Transl Med. 2021;13(583):eabc8980. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Provention Bio, Inc. 2022. Provention Bio Announces Extension of FDA User Fee Goal Date for Teplizumab to November 17, 2022. Accessed August 26, 2022. Available from: https://investors.proventionbio.com/2022-06-30-Provention-Bio-Announces-Extension-of-FDA-User-Fee-Goal-Date-for-Teplizumab-to-November-17,-2022
  • 42. Tracey D, Klareskog L, Sasso EH, Salfeld JG, Tak PP. Tumor necrosis factor antagonist mechanisms of action: a comprehensive review. Pharmacol Ther. 2008;117(2):244–279. [DOI] [PubMed] [Google Scholar]
  • 43. Mazumdar S, Greenwald D. Golimumab. MAbs. 2009;1(5):422–431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Quattrin T, Haller MJ, Steck AK, Felner EI, Li Y, Xia Y, et al. Golimumab and beta-cell function in youth with new-onset type 1 diabetes. N Engl J Med. 2020;383(21):2007–2017. [DOI] [PubMed] [Google Scholar]
  • 45. von Herrath M, Bain SC, Bode B, Clausen JO, Coppieters K, Gaysina L, et al. Anti-interleukin-21 antibody and liraglutide for the preservation of beta-cell function in adults with recent-onset type 1 diabetes: a randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Diabetes Endocrinol. 2021;9(4):212–224. [DOI] [PubMed] [Google Scholar]
  • 46. Yusta B, Baggio LL, Estall JL, Koehler JA, Holland DP, Li H, et al. GLP-1 receptor activation improves beta cell function and survival following induction of endoplasmic reticulum stress. Cell Metab. 2006;4(5):391–406. [DOI] [PubMed] [Google Scholar]
  • 47. Van Belle TL, Nierkens S, Arens R, von Herrath MG. Interleukin-21 receptor-mediated signals control autoreactive T cell infiltration in pancreatic islets. Immunity. 2012;36(6):1060–1072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Xu G, Grimes TD, Grayson TB, Chen J, Thielen LA, Tse HM, et al. Exploratory study reveals far reaching systemic and cellular effects of verapamil treatment in subjects with type 1 diabetes. Nat Commun. 2022;13(1):1159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Wang CY, Huang KC, Lu CW, Chu CH, Huang CN, Chen HS, et al. A randomized controlled trial of R-form verapamil added to ongoing metformin therapy in patients with type 2 diabetes. J Clin Endocrinol Metab. 2022;107(10):e4063–e4071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Shapiro AM, Lakey JR, Ryan EA, Korbutt GS, Toth E, Warnock GL, et al. Islet transplantation in seven patients with type 1 diabetes mellitus using a glucocorticoid-free immunosuppressive regimen. N Engl J Med. 2000;343(4):230–238. [DOI] [PubMed] [Google Scholar]
  • 51. Shapiro AM, Ricordi C, Hering BJ, Auchincloss H, Lindblad R, Robertson RP, et al. International trial of the Edmonton protocol for islet transplantation. N Engl J Med. 2006;355(13):1318–1330. [DOI] [PubMed] [Google Scholar]
  • 52. Shindo Y, Kalivarathan J, Saravanan PB, Levy MF, Kanak MA. Assessment of culture/preservation conditions of human islets for transplantation. Cell Transplant. 2022;31:9636897221086966. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Carvalho AM, Paulino da Silva Filho O, van Apeldoorn A. Protection factors used to improve in vivo islet function. J Immunol Regen Med. 2021;14:100051. [Google Scholar]
  • 54. Yoshimatsu G, Kunnathodi F, Saravanan PB, Shahbazov R, Chang C, Darden CM, et al. Pancreatic beta-cell-derived IP-10/CXCL10 isletokine mediates early loss of graft function in islet cell transplantation. Diabetes. 2017;66(11):2857–2867. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Bottino R, Balamurugan AN, Bertera S, Pietropaolo M, Trucco M, Piganelli JD. Preservation of human islet cell functional mass by anti-oxidative action of a novel SOD mimic compound. Diabetes. 2002;51(8):2561–2567. [DOI] [PubMed] [Google Scholar]
  • 56. Sklavos MM, Bertera S, Tse HM, Bottino R, He J, Beilke JN, et al. Redox modulation protects islets from transplant-related injury. Diabetes. 2010;59(7):1731–1738. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Wang J, Sun Z, Gou W, Adams DB, Cui W, Morgan KA, et al. Alpha-1 antitrypsin enhances islet engraftment by suppression of instant blood-mediated inflammatory reaction. Diabetes. 2017;66(4):970–980. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Song L, Gou W, Wang J, Wei H, Lee J, Strange C, et al. Overexpression of alpha-1 antitrypsin in mesenchymal stromal cells improves their intrinsic biological properties and therapeutic effects in nonobese diabetic mice. Stem Cells Transl Med. 2021;10(2):320–331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Cornu M, Modi H, Kawamori D, Kulkarni RN, Joffraud M, Thorens B. Glucagon-like peptide-1 increases beta-cell glucose competence and proliferation by translational induction of insulin-like growth factor-1 receptor expression. J Biol Chem. 2010;285(14):10538–10545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Cornu M, Yang JY, Jaccard E, Poussin C, Widmann C, Thorens B. Glucagon-like peptide-1 protects beta-cells against apoptosis by increasing the activity of an IGF-2/IGF-1 receptor autocrine loop. Diabetes. 2009;58(8):1816–1825. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Ding M, Fang QH, Cui YT, Shen QL, Liu Q, Wang PH, et al. Liraglutide prevents beta-cell apoptosis via inactivation of NOX2 and its related signaling pathway. J Diabetes Complications. 2019;33(4):267–277. [DOI] [PubMed] [Google Scholar]
  • 62. Kim HT, Desouza AH, Umhoefer H, Han J, Anzia L, Sacotte SJ, et al. Cholecystokinin attenuates beta-cell apoptosis in both mouse and human islets. Transl Res. 2022;243:1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Franklin ZJ, Tsakmaki A, Fonseca Pedro P, King AJ, Huang GC, Amjad S, et al. Islet neuropeptide Y receptors are functionally conserved and novel targets for the preservation of beta-cell mass. Diabetes Obes Metab. 2018;20(3):599–609. [DOI] [PubMed] [Google Scholar]
  • 64. Yang CH, Ann-Onda D, Lin X, Fynch S, Nadarajah S, Pappas EG, et al. Neuropeptide Y1 receptor antagonism protects beta-cells and improves glycemic control in type 2 diabetes. Mol Metab. 2022;55:101413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Hernandez-Perez M, Kulkarni A, Samala N, Sorrell C, El K, Haider I, et al. A 12-lipoxygenase-Gpr31 signaling axis is required for pancreatic organogenesis in the zebrafish. FASEB J. 2020;34(11):14850–14862. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Mason C, Dunnill P. A brief definition of regenerative medicine. Regen Med. 2008;3(1):1–5. [DOI] [PubMed] [Google Scholar]
  • 67. Arda HE, Li L, Tsai J, Torre EA, Rosli Y, Peiris H, et al. Age-dependent pancreatic gene regulation reveals mechanisms governing human beta cell function. Cell Metab. 2016;23(5):909–920. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Henquin JC, Nenquin M. Dynamics and regulation of insulin secretion in pancreatic islets from normal young children. PLoS One. 2016;11(11):e0165961. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Lee S, Zhang J, Saravanakumar S, Flisher MF, Grimm DR, van der Meulen T, Huising MO. Virgin beta-cells at the neogenic niche proliferate normally and mature slowly. Diabetes. 2021;70(5):1070–1083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. van der Meulen T, Xie R, Kelly OG, Vale WW, Sander M, Huising MO. Urocortin 3 marks mature human primary and embryonic stem cell-derived pancreatic alpha and beta cells. PLoS One. 2012;7(12):e52181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. van der Meulen T, Mawla AM, DiGruccio MR, Adams MW, Nies V, Dolleman S, et al. Virgin beta cells persist throughout life at a neogenic niche within pancreatic islets. Cell Metab. 2017;25(4):911–926.e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Nelson SB, Schaffer AE, Sander M. The transcription factors Nkx6.1 and Nkx6.2 possess equivalent activities in promoting beta-cell fate specification in Pdx1+ pancreatic progenitor cells. Development. 2007;134(13):2491–2500. [DOI] [PubMed] [Google Scholar]
  • 73. Bevacqua RJ, Lam JY, Peiris H, Whitener RL, Kim S, Gu X, et al. SIX2 and SIX3 coordinately regulate functional maturity and fate of human pancreatic beta cells. Genes Dev. 2021;35(3-4):234–249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Wortham M, Sander M. Transcriptional mechanisms of pancreatic β-cell maturation and functional adaptation. Trends Endocrinol Metab. 2021;32(7):474–487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Nasteska D, Hodson DJ. The role of beta cell heterogeneity in islet function and insulin release. J Mol Endocrinol. 2018;61(1):R43–R60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Salem V, Silva LD, Suba K, Georgiadou E, Neda Mousavy Gharavy S, Akhtar N, et al. Leader β-cells coordinate Ca2+ dynamics across pancreatic islets in vivo. Nat Metab. 2019;1(6):615–629. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Kravets V, Dwulet JM, Schleicher WE, Hodson DJ, Davis AM, Pyle L, et al. Functional architecture of pancreatic islets identifies a population of first responder cells that drive the first-phase calcium response. PLoS Biol. 2022;20(9):e3001761. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Johnston NR, Mitchell RK, Haythorne E, Pessoa MP, Semplici F, Ferrer J, et al. Beta cell hubs dictate pancreatic islet responses to glucose. Cell Metab. 2016;24(3):389–401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Bader E, Migliorini A, Gegg M, Moruzzi N, Gerdes J, Roscioni SS, et al. Identification of proliferative and mature beta-cells in the islets of Langerhans. Nature. 2016;535(7612):430–434. [DOI] [PubMed] [Google Scholar]
  • 80. Wang YJ, Golson ML, Schug J, Traum D, Liu C, Vivek K, et al. Single-cell mass cytometry analysis of the human endocrine pancreas. Cell Metab. 2016;24(4):616–626. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Salomon D, Meda P. Heterogeneity and contact-dependent regulation of hormone secretion by individual B cells. Exp Cell Res. 1986;162(2):507–520. [DOI] [PubMed] [Google Scholar]
  • 82. Avrahami D, Klochendler A, Dor Y, Glaser B. Beta cell heterogeneity: an evolving concept. Diabetologia. 2017;60(8):1363–1369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Dorrell C, Schug J, Canaday PS, Russ HA, Tarlow BD, Grompe MT, et al. Human islets contain four distinct subtypes of beta cells. Nat Commun. 2016;7:11756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Enge M, Arda HE, Mignardi M, Beausang J, Bottino R, Kim SK, et al. Single-cell analysis of human pancreas reveals transcriptional signatures of aging and somatic mutation patterns. Cell. 2017;171(2):321–330.e14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Zhong F, Jiang Y. Endogenous pancreatic beta cell regeneration: a potential strategy for the recovery of beta cell deficiency in diabetes. Front Endocrinol (Lausanne). 2019;10:101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Bonner-Weir S, Guo L, Li WC, Ouziel-Yahalom L, Lysy PA, Weir GC, et al. Islet neogenesis: a possible pathway for beta-cell replenishment. Rev Diabet Stud. 2012;9(4):407–416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Docherty FM, Sussel L. Islet regeneration: endogenous and exogenous approaches. Int J Mol Sci. 2021;22(7):3306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Levine F, Itkin-Ansari P. Beta-cell regeneration: neogenesis, replication or both? J Mol Med (Berl). 2008;86(3):247–258. [DOI] [PubMed] [Google Scholar]
  • 89. Téllez N, Montanya E. Gastrin induces ductal cell dedifferentiation and beta-cell neogenesis after 90% pancreatectomy. J Endocrinol. 2014;223(1):67–78. [DOI] [PubMed] [Google Scholar]
  • 90. Thorel F, Nepote V, Avril I, Kohno K, Desgraz R, Chera S, et al. Conversion of adult pancreatic alpha-cells to beta-cells after extreme beta-cell loss. Nature. 2010;464(7292):1149–1154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Aguayo-Mazzucato C, Bonner-Weir S. Pancreatic beta cell regeneration as a possible therapy for diabetes. Cell Metab. 2018;27(1):57–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Pera MF. Defining pluripotency. Nat Methods. 2010;7(11):885–887. [DOI] [PubMed] [Google Scholar]
  • 93. Nair GG, Tzanakakis ES, Hebrok M. Emerging routes to the generation of functional beta-cells for diabetes mellitus cell therapy. Nat Rev Endocrinol. 2020;16(9):506–518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Nurse P. Genetic control of cell size at cell division in yeast. Nature. 1975;256(5518):547–551. [DOI] [PubMed] [Google Scholar]
  • 95. Lee MG, Nurse P. Complementation used to clone a human homologue of the fission yeast cell cycle control gene cdc2. Nature. 1987;327(6117):31–35. [DOI] [PubMed] [Google Scholar]
  • 96. Evans T, Rosenthal ET, Youngblom J, Distel D, Hunt T. Cyclin: a protein specified by maternal mRNA in sea urchin eggs that is destroyed at each cleavage division. Cell. 1983;33(2):389–396. [DOI] [PubMed] [Google Scholar]
  • 97. Kulkarni RN, Mizrachi EB, Ocana AG, Stewart AF. Human beta-cell proliferation and intracellular signaling: driving in the dark without a road map. Diabetes. 2012;61(9):2205–2213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Bernal-Mizrachi E, Kulkarni RN, Scott DK, Mauvais-Jarvis F, Stewart AF, Garcia-Ocaña A. Human beta-cell proliferation and intracellular signaling part 2: still driving in the dark without a road map. Diabetes. 2014;63(3):819–831. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Stewart AF, Hussain MA, Garcia-Ocana A, Vasavada RC, Bhushan A, Bernal-Mizrachi E, et al. Human beta-cell proliferation and intracellular signaling: part 3. Diabetes. 2015;64(6):1872–1885. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Campbell-Thompson M, Fu A, Kaddis JS, Wasserfall C, Schatz DA, Pugliese A, et al. Insulitis and beta-cell mass in the natural history of type 1 diabetes. Diabetes. 2016;65(3):719–731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Teta M, Long SY, Wartschow LM, Rankin MM, Kushner JA. Very slow turnover of beta-cells in aged adult mice. Diabetes. 2005;54(9):2557–2567. [DOI] [PubMed] [Google Scholar]
  • 102. Arrojo e Drigo R, Lev-Ram V, Tyagi S, Ramachandra R, Deerinck T, Bushong E, et al. Age mosaicism across multiple scales in adult tissues. Cell Metabolism. 2019;30(2):343–351.e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Cnop M, Hughes SJ, Igoillo-Esteve M, Hoppa MB, Sayyed F, van de Laar L, et al. The long lifespan and low turnover of human islet beta cells estimated by mathematical modelling of lipofuscin accumulation. Diabetologia. 2010;53(2):321–330. [DOI] [PubMed] [Google Scholar]
  • 104. Gregg BE, Moore PC, Demozay D, Hall BA, Li M, Husain A, et al. Formation of a human beta-cell population within pancreatic islets is set early in life. J Clin Endocrinol Metab. 2012;97(9):3197–3206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Weir GC, Bonner-Weir S. Five stages of evolving beta-cell dysfunction during progression to diabetes. Diabetes. 2004;53(Suppl 3):S16–S21. [DOI] [PubMed] [Google Scholar]
  • 106. Da Silva Xavier G, Rutter GA. Metabolic and functional heterogeneity in pancreatic beta cells. J Mol Biol. 2020;432(5):1395–1406. [DOI] [PubMed] [Google Scholar]
  • 107. Dominguez-Gutierrez G, Xin Y, Gromada J. Heterogeneity of human pancreatic beta-cells. Mol Metab. 2019;27S:S7–S14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Camunas-Soler J, Dai XQ, Hang Y, Bautista A, Lyon J, Suzuki K, et al. Patch-Seq links single-cell transcriptomes to human islet dysfunction in diabetes. Cell Metab. 2020;31(5):1017–1031.e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Atkinson MA, Bluestone JA, Eisenbarth GS, Hebrok M, Herold KC, Accili D, et al. How does type 1 diabetes develop? : the notion of homicide or beta-cell suicide revisited. Diabetes. 2011;60(5):1370–1379. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Rui J, Deng S, Arazi A, Perdigoto AL, Liu Z, Herold KC. beta Cells that resist immunological attack develop during progression of autoimmune diabetes in NOD mice. Cell Metab. 2017;25(3):727–738. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Keenan HA, Sun JK, Levine J, Doria A, Aiello LP, Eisenbarth G, et al. Residual insulin production and pancreatic ss-cell turnover after 50 years of diabetes: Joslin Medalist Study. Diabetes. 2010;59(11):2846–2853. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Liu EH, Digon BJ III, Hirshberg B, Chang R, Wood BJ, Neeman Z, et al. Pancreatic beta cell function persists in many patients with chronic type 1 diabetes, but is not dramatically improved by prolonged immunosuppression and euglycaemia from a beta cell allograft. Diabetologia. 2009;52(7):1369–1380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Karakose E, Ackeifi C, Wang P, Stewart AF. Advances in drug discovery for human beta cell regeneration. Diabetologia. 2018;61(8):1693–1699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Okabayashi T, Shima Y, Sumiyoshi T, Kozuki A, Ito S, Ogawa Y, et al. Diagnosis and management of insulinoma. World J Gastroenterol. 2013;19(6):829–837. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Service GJ, Thompson GB, Service FJ, Andrews JC, Collazo-Clavell ML, Lloyd RV. Hyperinsulinemic hypoglycemia with nesidioblastosis after gastric-bypass surgery. N Engl J Med. 2005;353(3):249–254. [DOI] [PubMed] [Google Scholar]
  • 116. Cozar-Castellano I, Takane KK, Bottino R, Balamurugan AN, Stewart AF. Induction of beta-cell proliferation and retinoblastoma protein phosphorylation in rat and human islets using adenovirus-mediated transfer of cyclin-dependent kinase-4 and cyclin D1. Diabetes. 2004;53(1):149–159. [DOI] [PubMed] [Google Scholar]
  • 117. Fiaschi-Taesch NM, Kleinberger JW, Salim FG, Troxell R, Wills R, Tanwir M, et al. Human pancreatic beta-cell G1/S molecule cell cycle atlas. Diabetes. 2013;62(7):2450–2459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Fiaschi-Taesch NM, Kleinberger JW, Salim FG, Troxell R, Wills R, Tanwir M, et al. Cytoplasmic-nuclear trafficking of G1/S cell cycle molecules and adult human beta-cell replication: a revised model of human beta-cell G1/S control. Diabetes. 2013;62(7):2460–2470. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Fusco J, Xiao X, Prasadan K, Sheng Q, Chen C, Ming YC, et al. GLP-1/Exendin-4 induces beta-cell proliferation via the epidermal growth factor receptor. Sci Rep. 2017;7(1):9100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. Buteau J, Foisy S, Joly E, Prentki M. Glucagon-like peptide 1 induces pancreatic beta-cell proliferation via transactivation of the epidermal growth factor receptor. Diabetes. 2003;52(1):124–132. [DOI] [PubMed] [Google Scholar]
  • 121. Chen S, Bastarrachea RA, Roberts BJ, Voruganti VS, Frost PA, Nava-Gonzalez EJ, et al. Successful beta cells islet regeneration in streptozotocin-induced diabetic baboons using ultrasound-targeted microbubble gene therapy with cyclinD2/CDK4/GLP1. Cell Cycle. 2014;13(7):1145–1151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Dai C, Hang Y, Shostak A, Poffenberger G, Hart N, Prasad N, et al. Age-dependent human β cell proliferation induced by glucagon-like peptide 1 and calcineurin signaling. J Clin Invest. 2017;127(10):3835–3844. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Walpita D, Hasaka T, Spoonamore J, Vetere A, Takane KK, Fomina-Yadlin D, et al. A human islet cell culture system for high-throughput screening. J Biomol Screen. 2012;17(4):509–518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. Wagner BK. Small-molecule discovery in the pancreatic beta cell. Curr Opin Chem Biol. 2022;68:102150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125. Wang P, Alvarez-Perez JC, Felsenfeld DP, Liu H, Sivendran S, Bender A, et al. A high-throughput chemical screen reveals that harmine-mediated inhibition of DYRK1A increases human pancreatic beta cell replication. Nat Med. 2015;21(4):383–388. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Shen W, Taylor B, Jin Q, Nguyen-Tran V, Meeusen S, Zhang YQ, et al. Inhibition of DYRK1A and GSK3B induces human beta-cell proliferation. Nat Commun. 2015;6:8372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Dirice E, Walpita D, Vetere A, Meier BC, Kahraman S, Hu J, et al. Inhibition of DYRK1A stimulates human beta-cell proliferation. Diabetes. 2016;65(6):1660–1671. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. Aamodt KI, Aramandla R, Brown JJ, Fiaschi-Taesch N, Wang P, Stewart AF, et al. Development of a reliable automated screening system to identify small molecules and biologics that promote human beta-cell regeneration. Am J Physiol Endocrinol Metab. 2016;311(5):E859–E868. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Shirakawa J, Kulkarni RN. Novel factors modulating human beta-cell proliferation. Diabetes Obes Metab. 2016;18(Suppl 1):71–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. Kumar K, Suebsuwong C, Wang P, Garcia-Ocana A, Stewart AF, DeVita RJ. DYRK1A inhibitors as potential therapeutics for beta-cell regeneration for diabetes. J Med Chem. 2021;64(6):2901–2922. [DOI] [PubMed] [Google Scholar]
  • 131. Title AC, Karsai M, Mir-Coll J, Grining OY, Rufer C, Sonntag S, et al. Evaluation of the effects of harmine on beta-cell function and proliferation in standardized human islets using 3D high-content confocal imaging and automated analysis. Front Endocrinol (Lausanne). 2022;13:854094. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. Ball KA, Webb KJ, Coleman SJ, Cozzolino KA, Jacobsen J, Jones KR, et al. An isothermal shift assay for proteome scale drug-target identification. Commun Biol. 2020;3(1):75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133. Kumar K, Wang P, Swartz EA, Khamrui S, Secor C, Lazarus B, et al. Structure-activity relationships and biological evaluation of 7-substituted harmine analogs for human beta-cell proliferation. Molecules. 2020;25(8):1983. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. Maachi H, Ghislain J, Tremblay C, Poitout V. Pronounced proliferation of non-beta cells in response to beta-cell mitogens in isolated human islets of Langerhans. Sci Rep. 2021;11(1):11283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Jarhad DB, Mashelkar KK, Kim HR, Noh M, Jeong LS. Dual-specificity tyrosine phosphorylation-regulated kinase 1A (DYRK1A) inhibitors as potential therapeutics. J Med Chem. 2018;61(22):9791–9810. [DOI] [PubMed] [Google Scholar]
  • 136. Farilla L, Bulotta A, Hirshberg B, Li Calzi S, Khoury N, Noushmehr H, et al. Glucagon-like peptide 1 inhibits cell apoptosis and improves glucose responsiveness of freshly isolated human islets. Endocrinology. 2003;144(12):5149–5158. [DOI] [PubMed] [Google Scholar]
  • 137. Ackeifi C, Wang P, Karakose E, Manning Fox JE, Gonzalez BJ, Liu H, et al. GLP-1 receptor agonists synergize with DYRK1A inhibitors to potentiate functional human beta cell regeneration. Sci Transl Med. 2020;12(530):eaaw9996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138. Xiao X, Wiersch J, El-Gohary Y, Guo P, Prasadan K, Paredes J, et al. TGF β receptor signaling is essential for inflammation-induced but not β-cell workload-induced β-cell proliferation. Diabetes. 2013;62(4):1217–1226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139. Arbones ML, Thomazeau A, Nakano-Kobayashi A, Hagiwara M, Delabar JM. DYRK1A and cognition: a lifelong relationship. Pharmacol Ther. 2019;194:199–221. [DOI] [PubMed] [Google Scholar]
  • 140. Sakamaki J, Fu A, Reeks C, Baird S, Depatie C, Al Azzabi M, et al. Role of the SIK2-p35-PJA2 complex in pancreatic beta-cell functional compensation. Nat Cell Biol. 2014;16(3):234–244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. Charbord J, Ren L, Sharma RB, Johansson A, Ågren R, Chu L, et al. In vivo screen identifies a SIK inhibitor that induces β cell proliferation through a transient UPR. Nature Metabolism. 2021;3(5):682–700. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142. Iorio C, Rourke JL, Wells L, Sakamaki JI, Moon E, Hu Q, et al. Silencing the G-protein coupled receptor 3-salt inducible kinase 2 pathway promotes human beta cell proliferation. Commun Biol. 2021;4(1):907. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143. Kim MJ, Park SK, Lee JH, Jung CY, Sung DJ, Park JH, et al. Salt-inducible kinase 1 terminates cAMP signaling by an evolutionarily conserved negative-feedback loop in beta-cells. Diabetes. 2015;64(9):3189–3202. [DOI] [PubMed] [Google Scholar]
  • 144. Huang D, Chen P, Huang G, Sun H, Luo X, He C, et al. Salt-inducible kinases inhibitor HG-9-91-01 targets RIPK3 kinase activity to alleviate necroptosis-mediated inflammatory injury. Cell Death Dis. 2022;13(2):188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145. Contreras CJ, Mukherjee N, Branco RCS, Lin L, Hogan MF, Cai EP, et al. RIPK1 and RIPK3 regulate TNFα-induced β-cell death in concert with caspase activity. Mol Metab. 2022;65:101582. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146. Annes JP, Ryu JH, Lam K, Carolan PJ, Utz K, Hollister-Lock J, et al. Adenosine kinase inhibition selectively promotes rodent and porcine islet beta-cell replication. Proc Natl Acad Sci U S A. 2012;109(10):3915–3920. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147. Andersson O, Adams BA, Yoo D, Ellis GC, Gut P, Anderson RM, et al. Adenosine signaling promotes regeneration of pancreatic beta cells in vivo. Cell Metab. 2012;15(6):885–894. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148. Prentice KJ, Saksi J, Robertson LT, Lee GY, Inouye KE, Eguchi K, et al. A hormone complex of FABP4 and nucleoside kinases regulates islet function. Nature. 2021;600(7890):720–726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149. Porat S, Weinberg-Corem N, Tornovsky-Babaey S, Schyr-Ben-Haroush R, Hija A, Stolovich-Rain M, et al. Control of pancreatic beta cell regeneration by glucose metabolism. Cell Metab. 2011;13(4):440–449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150. Klein KR, Boeder SC, Freeman JLR, Dunn I, Dvergsten C, Madduri S, et al. Impact of the hepatoselective glucokinase activator TTP399 on ketoacidosis during insulin withdrawal in people with type 1 diabetes. Diabetes Obes Metab. 2022;24(8):1439–1447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151. Klein KR, Freeman JLR, Dunn I, Dvergsten C, Kirkman MS, Buse JB, et al. The SimpliciT1 Study: a randomized, double-blind, placebo-controlled phase 1b/2 adaptive Study of TTP399, a hepatoselective glucokinase activator, for adjunctive treatment of type 1 diabetes. Diabetes Care. 2021;44(4):960–968. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152. Lakshmipathi J, Alvarez-Perez JC, Rosselot C, Casinelli GP, Stamateris RE, Rausell-Palamos F, et al. PKCζ is essential for pancreatic β-cell replication during insulin resistance by regulating mTOR and cyclin-D2. Diabetes. 2016;65(5):1283–1296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153. Kulkarni RN, Brüning JC, Winnay JN, Postic C, Magnuson MA, Kahn CR. Tissue-specific knockout of the insulin receptor in pancreatic β cells creates an insulin secretory defect similar to that in type 2 diabetes. Cell. 1999;96(3):329–339. [DOI] [PubMed] [Google Scholar]
  • 154. Otani K, Kulkarni RN, Baldwin AC, Krutzfeldt J, Ueki K, Stoffel M, et al. Reduced beta-cell mass and altered glucose sensing impair insulin-secretory function in betaIRKO mice. Am J Physiol Endocrinol Metab. 2004;286(1):E41–49. [DOI] [PubMed] [Google Scholar]
  • 155. Withers DJ, Gutierrez JS, Towery H, Burks DJ, Ren JM, Previs S, et al. Disruption of IRS-2 causes type 2 diabetes in mice. Nature. 1998;391(6670):900–904. [DOI] [PubMed] [Google Scholar]
  • 156. Hashimoto N, Kido Y, Uchida T, Asahara S, Shigeyama Y, Matsuda T, et al. Ablation of PDK1 in pancreatic beta cells induces diabetes as a result of loss of beta cell mass. Nat Genet. 2006;38(5):589–593. [DOI] [PubMed] [Google Scholar]
  • 157. Pende M, Kozma SC, Jaquet M, Oorschot V, Burcelin R, Le Marchand-Brustel Y, et al. Hypoinsulinaemia, glucose intolerance and diminished beta-cell size in S6K1-deficient mice. Nature. 2000;408(6815):994–997. [DOI] [PubMed] [Google Scholar]
  • 158. Kido Y, Burks DJ, Withers D, Bruning JC, Kahn CR, White MF, et al. Tissue-specific insulin resistance in mice with mutations in the insulin receptor, IRS-1, and IRS-2. J Clin Invest. 2000;105(2):199–205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159. Bernal-Mizrachi E, Fatrai S, Johnson JD, Ohsugi M, Otani K, Han Z, et al. Defective insulin secretion and increased susceptibility to experimental diabetes are induced by reduced Akt activity in pancreatic islet beta cells. J Clin Invest. 2004;114(7):928–936. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160. Skovso S, Panzhinskiy E, Kolic J, Cen HH, Dionne DA, Dai XQ, et al. Beta-cell specific Insr deletion promotes insulin hypersecretion and improves glucose tolerance prior to global insulin resistance. Nat Commun. 2022;13(1):735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161. Michael MD, Kulkarni RN, Postic C, Previs SF, Shulman GI, Magnuson MA, et al. Loss of insulin signaling in hepatocytes leads to severe insulin resistance and progressive hepatic dysfunction. Molecular Cell. 2000;6(1):87–97. [PubMed] [Google Scholar]
  • 162. Gusarova V, Alexa CA, Na E, Stevis PE, Xin Y, Bonner-Weir S, et al. ANGPTL8/betatrophin does not control pancreatic beta cell expansion. Cell. 2014;159(3):691–696. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163. Shirakawa J, Okuyama T, Yoshida E, Shimizu M, Horigome Y, Tuno T, et al. Effects of the antitumor drug OSI-906, a dual inhibitor of IGF-1 receptor and insulin receptor, on the glycemic control, beta-cell functions, and beta-cell proliferation in male mice. Endocrinology. 2014;155(6):2102–2111. [DOI] [PubMed] [Google Scholar]
  • 164. Tajima K, Shirakawa J, Togashi Y, Yamazaki S, Okuyama T, Kyohara M, et al. Metabolic recovery of lipodystrophy, liver steatosis, and pancreatic beta cell proliferation after the withdrawal of OSI-906. Sci Rep. 2017;7(1):4119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165. El Ouaamari A, Kawamori D, Dirice E, Liew CW, Shadrach JL, Hu J, et al. Liver-derived systemic factors drive beta cell hyperplasia in insulin-resistant states. Cell Rep. 2013;3(2):401–410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166. Flier SN, Kulkarni RN, Kahn CR. Evidence for a circulating islet cell growth factor in insulin-resistant states. Proc Natl Acad Sci U S A. 2001;98(13):7475–7480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167. El Ouaamari A, Dirice E, Gedeon N, Hu J, Zhou JY, Shirakawa J, et al. SerpinB1 Promotes Pancreatic beta Cell Proliferation. Cell Metab. 2016;23(1):194–205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168. Xu S, Qin D, Yang H, He C, Liu W, Tian N, et al. SerpinB1 promotes the proliferation of porcine pancreatic stem cells through the STAT3 signaling pathway. J Steroid Biochem Mol Biol. 2020;198:105537. [DOI] [PubMed] [Google Scholar]
  • 169. Kamal MM, Adel A, Sayed GH, Ragab S, Kassem DH. New emerging roles of the novel hepatokine SERPINB1 in type 2 diabetes mellitus: crosstalk with beta-cell dysfunction and dyslipidemia. Transl Res. 2020;231:1–12. [DOI] [PubMed] [Google Scholar]
  • 170. Shirakawa J, Togashi Y, Basile G, Okuyama T, Inoue R, Fernandez M, et al. E2F1 transcription factor mediates a link between fat and islets to promote beta cell proliferation in response to acute insulin resistance. Cell Rep. 2022;41(1):111436. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171. Ke XY, Chen Y, Tham VY, Lin RY, Dakle P, Nacro K, et al. MNK1 and MNK2 enforce expression of E2F1, FOXM1, and WEE1 to drive soft tissue sarcoma. Oncogene. 2021;40(10):1851–1867. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172. Liao GB, Li XZ, Zeng S, Liu C, Yang SM, Yang L, et al. Regulation of the master regulator FOXM1 in cancer. Cell Commun Signal. 2018;16(1):57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173. Gezginci-Oktayoglu S, Karatug A, Bolkent S. Nerve growth factor neutralization suppresses β-cell proliferation through activin A and Betacellulin. Pancreas. 2015;44(2):243–249. [DOI] [PubMed] [Google Scholar]
  • 174. Rescan C, Le Bras S, Lefebvre VH, Frandsen U, Klein T, Foschi M, et al. EGF-induced proliferation of adult human pancreatic duct cells is mediated by the MEK/ERK cascade. Lab Invest. 2005;85(1):65–74. [DOI] [PubMed] [Google Scholar]
  • 175. Parsons JA, Brelje TC, Sorenson RL. Adaptation of islets of Langerhans to pregnancy: increased islet cell proliferation and insulin secretion correlates with the onset of placental lactogen secretion. Endocrinology. 1992;130(3):1459–1466. [DOI] [PubMed] [Google Scholar]
  • 176. Li R, Kondegowda NG, Filipowska J, Hampton RF, Leblanc S, Garcia-Ocana A, et al. Lactogens reduce endoplasmic reticulum stress-induced rodent and human beta-cell death and diabetes incidence in Akita mice. Diabetes. 2020;69(7):1463–1475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177. Kim H, Toyofuku Y, Lynn FC, Chak E, Uchida T, Mizukami H, et al. Serotonin regulates pancreatic beta cell mass during pregnancy. Nat Med. 2010;16(7):804–808. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178. Ben-Othman N, Vieira A, Courtney M, Record F, Gjernes E, Avolio F, et al. Long-term GABA administration induces alpha cell-mediated beta-like cell neogenesis. Cell. 2017;168(1-2):73–85.e11. [DOI] [PubMed] [Google Scholar]
  • 179. Rosado-Olivieri EA, Aigha II, Kenty JH, Melton DA. Identification of a LIF-responsive, replication-competent subpopulation of human β cells. Cell Metabolism. 2020;31(2):327–338.e6. [DOI] [PubMed] [Google Scholar]
  • 180. Kalwat MA, Scheuner D, Rodrigues-Dos-Santos K, Eizirik DL, Cobb MH. The pancreatic ss-cell response to secretory demands and adaption to stress. Endocrinology. 2021;162(11):bqab173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181. Engin F, Yermalovich A, Nguyen T, Hummasti S, Fu W, Eizirik DL, et al. Restoration of the unfolded protein response in pancreatic beta cells protects mice against type 1 diabetes. Sci Transl Med. 2013;5(211):211ra156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182. Tang Q, Li Y, He J. MANF: an emerging therapeutic target for metabolic diseases. Trends Endocrinol Metab. 2022;33(4):236–246. [DOI] [PubMed] [Google Scholar]
  • 183. Hakonen E, Chandra V, Fogarty CL, Yu NY, Ustinov J, Katayama S, et al. MANF protects human pancreatic beta cells against stress-induced cell death. Diabetologia. 2018;61(10):2202–2214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184. Lindahl M, Danilova T, Palm E, Lindholm P, Voikar V, Hakonen E, et al. MANF is indispensable for the proliferation and survival of pancreatic beta cells. Cell Rep. 2014;7(2):366–375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185. Danilova T, Belevich I, Li H, Palm E, Jokitalo E, Otonkoski T, et al. MANF is required for the postnatal expansion and maintenance of pancreatic beta-cell mass in mice. Diabetes. 2019;68(1):66–80. [DOI] [PubMed] [Google Scholar]
  • 186. Marciniak SJ, Yun CY, Oyadomari S, Novoa I, Zhang Y, Jungreis R, et al. CHOP induces death by promoting protein synthesis and oxidation in the stressed endoplasmic reticulum. Genes Dev. 2004;18(24):3066–3077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187. Song B, Scheuner D, Ron D, Pennathur S, Kaufman RJ. Chop deletion reduces oxidative stress, improves beta cell function, and promotes cell survival in multiple mouse models of diabetes. J Clin Invest. 2008;118(10):3378–3389. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188. Yong J, Parekh VS, Reilly SM, Nayak J, Chen Z, Lebeaupin C, et al. Chop/Ddit3 depletion in beta cells alleviates ER stress and corrects hepatic steatosis in mice. Sci Transl Med. 2021;13(604):eaba9796. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189. Berger M, Scheel DW, Macias H, Miyatsuka T, Kim H, Hoang P, et al. Gα i/o-coupled receptor signaling restricts pancreatic β-cell expansion. Proc Natl Acad Sci U S A. 2015;112(9):2888–2893. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190. Kondegowda NG, Fenutria R, Pollack IR, Orthofer M, Garcia-Ocana A, Penninger JM, et al. Osteoprotegerin and denosumab stimulate human beta cell proliferation through inhibition of the receptor activator of NF-κB ligand pathway. Cell Metab. 2015;22(1):77–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191. Robitaille K, Rourke JL, McBane JE, Fu A, Baird S, Du Q, et al. High-throughput functional genomics identifies regulators of primary human beta cell proliferation. J Biol Chem. 2016;291(9):4614–4625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192. Szabat M, Page MM, Panzhinskiy E, Skovso S, Mojibian M, Fernandez-Tajes J, et al. Reduced insulin production relieves endoplasmic reticulum stress and induces beta cell proliferation. Cell Metab. 2016;23(1):179–193. [DOI] [PubMed] [Google Scholar]
  • 193. Le May C, Chu K, Hu M, Ortega CS, Simpson ER, Korach KS, et al. Estrogens protect pancreatic beta-cells from apoptosis and prevent insulin-deficient diabetes mellitus in mice. Proc Natl Acad Sci U S A. 2006;103(24):9232–9237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194. Liu S, Mauvais-Jarvis F. Minireview: Estrogenic protection of beta-cell failure in metabolic diseases. Endocrinology. 2010;151(3):859–864. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195. Fuselier T, Mota de Sa P, Qadir MMF, Xu B, Allard C, Meyers MM, et al. Efficacy of glucagon-like peptide-1 and estrogen dual agonist in pancreatic islets protection and pre-clinical models of insulin-deficient diabetes. Cell Rep Med. 2022;3(4):100598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196. Sachs S, Bastidas-Ponce A, Tritschler S, Bakhti M, Bottcher A, Sanchez-Garrido MA, et al. Targeted pharmacological therapy restores beta-cell function for diabetes remission. Nat Metab. 2020;2(2):192–209. [DOI] [PubMed] [Google Scholar]
  • 197. Finan B, Yang B, Ottaway N, Stemmer K, Muller TD, Yi CX, et al. Targeted estrogen delivery reverses the metabolic syndrome. Nat Med. 2012;18(12):1847–1856. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198. Ammala C, Drury WJ III, Knerr L, Ahlstedt I, Stillemark-Billton P, Wennberg-Huldt C, et al. Targeted delivery of antisense oligonucleotides to pancreatic beta-cells. Sci Adv. 2018;4(10):eaat3386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199. Quarta C, Stemmer K, Novikoff A, Yang B, Klingelhuber F, Harger A, et al. GLP-1-mediated delivery of tesaglitazar improves obesity and glucose metabolism in male mice. Nat Metab. 2022;4(8):1071–1083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200. Nair G, Hebrok M. Islet formation in mice and men: lessons for the generation of functional insulin-producing beta-cells from human pluripotent stem cells. Curr Opin Genet Dev. 2015;32:171–180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201. Collombat P, Mansouri A, Hecksher-Sorensen J, Serup P, Krull J, Gradwohl G, et al. Opposing actions of Arx and Pax4 in endocrine pancreas development. Genes Dev. 2003;17(20):2591–2603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202. Collombat P, Hecksher-Sorensen J, Krull J, Berger J, Riedel D, Herrera PL, et al. Embryonic endocrine pancreas and mature beta cells acquire alpha and PP cell phenotypes upon Arx misexpression. J Clin Invest. 2007;117(4):961–970. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203. Courtney M, Gjernes E, Druelle N, Ravaud C, Vieira A, Ben-Othman N, et al. The inactivation of Arx in pancreatic alpha-cells triggers their neogenesis and conversion into functional beta-like cells. PLoS Genet. 2013;9(10):e1003934. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204. Chakravarthy H, Gu X, Enge M, Dai X, Wang Y, Damond N, et al. Converting adult pancreatic islet alpha cells into beta cells by targeting both Dnmt1 and Arx. Cell Metab. 2017;25(3):622–634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205. Papizan JB, Singer RA, Tschen SI, Dhawan S, Friel JM, Hipkens SB, et al. Nkx2.2 repressor complex regulates islet beta-cell specification and prevents beta-to-alpha-cell reprogramming. Genes Dev. 2011;25(21):2291–2305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206. Lu J, Liu KC, Schulz N, Karampelias C, Charbord J, Hilding A, et al. IGFBP1 increases beta-cell regeneration by promoting alpha- to beta-cell transdifferentiation. EMBO J. 2016;35(18):2026–2044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207. Xiao X, Guo P, Shiota C, Zhang T, Coudriet GM, Fischbach S, et al. Endogenous reprogramming of alpha cells into beta cells, induced by viral gene therapy, reverses autoimmune diabetes. Cell Stem Cell. 2018;22(1):78–90.e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208. Furuyama K, Chera S, van Gurp L, Oropeza D, Ghila L, Damond N, et al. Diabetes relief in mice by glucose-sensing insulin-secreting human alpha-cells. Nature. 2019;567(7746):43–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209. Pettus J, Reeds D, Cavaiola TS, Boeder S, Levin M, Cava E, et al. REMD-477, a human glucagon receptor (GCGR) antibody, reduces daily insulin requirements and improves glycemic control in people with type 1 diabetes (T1D). Diabetes. 2017;66(Suppl 1):A100. [Google Scholar]
  • 210. Okamoto H, Kim J, Aglione J, Lee J, Cavino K, Na E, et al. Glucagon receptor blockade with a human antibody normalizes blood glucose in diabetic mice and monkeys. Endocrinology. 2015;156(8):2781–2794. [DOI] [PubMed] [Google Scholar]
  • 211. Xi Y, Song B, Ngan I, Solloway MJ, Humphrey M, Wang Y, et al. Glucagon-receptor-antagonism-mediated beta-cell regeneration as an effective anti-diabetic therapy. Cell Rep. 2022;39(9):110872. [DOI] [PubMed] [Google Scholar]
  • 212. Sharma AX, Quittner-Strom EB, Lee Y, Johnson JA, Martin SA, Yu X, et al. Glucagon receptor antagonism improves glucose metabolism and cardiac function by promoting AMP-mediated protein kinase in diabetic mice. Cell Reports. 2018;22(7):1760–1773. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213. Dean ED, Li M, Prasad N, Wisniewski SN, Von Deylen A, Spaeth J, et al. Interrupted glucagon signaling reveals hepatic alpha cell axis and role for L-glutamine in alpha cell proliferation. Cell Metab. 2017;25(6):1362–1373.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214. Kim J, Okamoto H, Huang Z, Anguiano G, Chen S, Liu Q, et al. Amino acid transporter Slc38a5 controls glucagon receptor inhibition-induced pancreatic alpha cell hyperplasia in mice. Cell Metab. 2017;25(6):1348–1361.e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215. Wang MY, Dean ED, Quittner-Strom E, Zhu Y, Chowdhury KH, Zhang Z, et al. Glucagon blockade restores functional beta-cell mass in type 1 diabetic mice and enhances function of human islets. Proc Natl Acad Sci U S A. 2021;118(9):e2022142118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216. Cui X, Feng J, Wei T, Gu L, Wang D, Lang S, et al. Pro-alpha-cell-derived beta-cells contribute to beta-cell neogenesis induced by antagonistic glucagon receptor antibody in type 2 diabetic mice. iScience. 2022;25(7):104567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217. Wei R, Gu L, Yang J, Yang K, Liu J, Le Y, et al. Antagonistic glucagon receptor antibody promotes alpha-cell proliferation and increases beta-cell mass in diabetic mice. iScience. 2019;16:326–339. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218. Rubin de Celis MF, Bonner-Weir S. Glucagon receptor antagonists might stimulate beta-cell expansion. Nat Rev Endocrinol. 2022;18(11):659–660. [DOI] [PubMed] [Google Scholar]
  • 219. Purwana I, Zheng J, Li X, Deurloo M, Son DO, Zhang Z, et al. GABA promotes human beta-cell proliferation and modulates glucose homeostasis. Diabetes. 2014;63(12):4197–4205. [DOI] [PubMed] [Google Scholar]
  • 220. Li J, Casteels T, Frogne T, Ingvorsen C, Honore C, Courtney M, et al. Artemisinins target GABAA receptor signaling and impair alpha cell identity. Cell. 2017;168(1-2):86–100.e15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221. van der Meulen T, Lee S, Noordeloos E, Donaldson CJ, Adams MW, Noguchi GM, et al. Artemether Does Not Turn alpha Cells into beta Cells. Cell Metab. 2017;27(1):218–225.e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222. Ackermann AM, Moss NG, Kaestner KH. GABA and artesunate do not induce pancreatic alpha-to-beta cell transdifferentiation in vivo. Cell Metab. 2018;28(5):787–792.e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223. Espes D, Liljeback H, Hill H, Elksnis A, Caballero-Corbalan J, Carlsson PO. GABA induces a hormonal counter-regulatory response in subjects with long-standing type 1 diabetes. BMJ Open Diabetes Res Care. 2021;9(1):e002442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224. Hagan DW, Ferreira SM, Santos GJ, Phelps EA. The role of GABA in islet function. Front Endocrinol. 2022;13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225. Chera S, Baronnier D, Ghila L, Cigliola V, Jensen JN, Gu G, et al. Diabetes recovery by age-dependent conversion of pancreatic delta-cells into insulin producers. Nature. 2014;514(7523):503–507. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226. Nagashima T, Shigematsu N, Maruki R, Urano Y, Tanaka H, Shimaya A, et al. Discovery of novel forkhead box O1 inhibitors for treating type 2 diabetes: improvement of fasting glycemia in diabetic db/db mice. Mol Pharmacol. 2010;78(5):961–970. [DOI] [PubMed] [Google Scholar]
  • 227. Singh SP, Chawla P, Hnatiuk A, Kamel M, Silva LD, Spanjaard B, et al. A single-cell atlas of de novo beta-cell regeneration reveals the contribution of hybrid beta/delta-cells to diabetes recovery in zebrafish. Development. 2022;149(2):dev199853. [DOI] [PubMed] [Google Scholar]
  • 228. Carril Pardo CA, Massoz L, Dupont MA, Bergemann D, Bourdouxhe J, Lavergne A, et al. A delta-cell subpopulation with a pro-beta-cell identity contributes to efficient age-independent recovery in a zebrafish model of diabetes. Elife. 2022;11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229. Perez-Frances M, van Gurp L, Abate MV, Cigliola V, Furuyama K, Bru-Tari E, et al. Pancreatic Ppy-expressing gamma-cells display mixed phenotypic traits and the adaptive plasticity to engage insulin production. Nat Commun. 2021;12(1):4458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230. Zhou Q, Brown J, Kanarek A, Rajagopal J, Melton DA. In vivo reprogramming of adult pancreatic exocrine cells to beta-cells. Nature. 2008;455(7213):627–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231. Ariyachet C, Tovaglieri A, Xiang G, Lu J, Shah MS, Richmond CA, et al. Reprogrammed stomach tissue as a renewable source of functional beta cells for blood glucose regulation. Cell Stem Cell. 2016;18(3):410–421. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232. Liu KC, Leuckx G, Sakano D, Seymour PA, Mattsson CL, Rautio L, et al. Inhibition of Cdk5 promotes beta-cell differentiation from ductal progenitors. Diabetes. 2018;67(1):58–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233. Gu G, Dubauskaite J, Melton DA. Direct evidence for the pancreatic lineage: NGN3+ cells are islet progenitors and are distinct from duct progenitors. Development. 2002;129(10):2447–2457. [DOI] [PubMed] [Google Scholar]
  • 234. Xu X, D'Hoker J, Stange G, Bonne S, De Leu N, Xiao X, et al. Beta cells can be generated from endogenous progenitors in injured adult mouse pancreas. Cell. 2008;132(2):197–207. [DOI] [PubMed] [Google Scholar]
  • 235. Apelqvist A, Li H, Sommer L, Beatus P, Anderson DJ, Honjo T, et al. Notch signalling controls pancreatic cell differentiation. Nature. 1999;400(6747):877–881. [DOI] [PubMed] [Google Scholar]
  • 236. Wang D, Wang J, Bai L, Pan H, Feng H, Clevers H, et al. Long-term expansion of pancreatic islet organoids from resident Procr+ progenitors. Cell. 2020;180(6):1198–1211.e19. [DOI] [PubMed] [Google Scholar]
  • 237. Yu XX, Qiu WL, Yang L, Wang YC, He MY, Wang D, et al. Sequential progenitor states mark the generation of pancreatic endocrine lineages in mice and humans. Cell Res. 2021;31(8):886–903. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238. Seaberg RM, Smukler SR, Kieffer TJ, Enikolopov G, Asghar Z, Wheeler MB, et al. Clonal identification of multipotent precursors from adult mouse pancreas that generate neural and pancreatic lineages. Nat Biotechnol. 2004;22(9):1115–1124. [DOI] [PubMed] [Google Scholar]
  • 239. Li WC, Rukstalis JM, Nishimura W, Tchipashvili V, Habener JF, Sharma A, et al. Activation of pancreatic-duct-derived progenitor cells during pancreas regeneration in adult rats. J Cell Sci. 2010;123(Pt 16):2792–2802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240. Bonner-Weir S, Inada A, Yatoh S, Li WC, Aye T, Toschi E, et al. Transdifferentiation of pancreatic ductal cells to endocrine beta-cells. Biochem Soc Trans. 2008;36(Pt 3):353–356. [DOI] [PubMed] [Google Scholar]
  • 241. El-Gohary Y, Wiersch J, Tulachan S, Xiao X, Guo P, Rymer C, et al. Intra-islet pancreatic ducts can give rise to insulin-positive cells. Endocrinology. 2015;157(1):166–175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242. Xiao X, Chen Z, Shiota C, Prasadan K, Guo P, El-Gohary Y, et al. No evidence for beta cell neogenesis in murine adult pancreas. J Clin Invest. 2013;123(5):2207–2217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243. Bonner-Weir S, Toschi E, Inada A, Reitz P, Fonseca SY, Aye T, et al. The pancreatic ductal epithelium serves as a potential pool of progenitor cells. Pediatr Diabetes. 2004;5(Suppl 2):16–22. [DOI] [PubMed] [Google Scholar]
  • 244. Criscimanna A, Speicher JA, Houshmand G, Shiota C, Prasadan K, Ji B, et al. Duct cells contribute to regeneration of endocrine and acinar cells following pancreatic damage in adult mice. Gastroenterology. 2011;141(4):1451–1462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 245. Sancho R, Gruber R, Gu G, Behrens A. Loss of Fbw7 reprograms adult pancreatic ductal cells into alpha, delta, and beta cells. Cell Stem Cell. 2014;15(2):139–153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 246. Gradwohl G, Dierich A, LeMeur M, Guillemot F. neurogenin3 is required for the development of the four endocrine cell lineages of the pancreas. Proc Natl Acad Sci U S A. 2000;97(4):1607–1611. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247. Gribben C, Lambert C, Messal HA, Hubber EL, Rackham C, Evans I, et al. Ductal Ngn3-expressing progenitors contribute to adult beta cell neogenesis in the pancreas. Cell Stem Cell. 2021;28(11):2000–2008.e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248. Dirice E, De Jesus DF, Kahraman S, Basile G, Ng RW, El Ouaamari A, et al. Human duct cells contribute to beta cell compensation in insulin resistance. JCI Insight. 2019;4(8):e99576. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249. Dirice E, Basile G, Kahraman S, Diegisser D, Hu J, Kulkarni RN. Single-nucleus RNA-sequencing reveals singular gene signatures of human ductal cells during adaptation to insulin resistance. JCI Insight. 2022;7(16):e153877. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 250. Qadir MMF, Alvarez-Cubela S, Klein D, van Dijk J, Muniz-Anquela R, Moreno-Hernandez YB, et al. Single-cell resolution analysis of the human pancreatic ductal progenitor cell niche. Proc Natl Acad Sci U S A. 2020;117(20):10876–10887. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 251. Karampelias C, Watt K, Mattsson CL, Ruiz AF, Rezanejad H, Mi J, et al. MNK2 deficiency potentiates beta-cell regeneration via translational regulation. Nat Chem Biol. 2022;18(9):942–953. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 252. Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA, Swiergiel JJ, Marshall VS, et al. Embryonic stem cell lines derived from human blastocysts. Science. 1998;282(5391):1145–1147. [DOI] [PubMed] [Google Scholar]
  • 253. Basile G, Qadir MMF, Mauvais-Jarvis F, Vetere A, Shoba V, Modell AE, et al. Emerging diabetes therapies: Bringing back the beta-cells. Mol Metab. 2022;60:101477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 254. Lewandowski J, Kurpisz M. Techniques of human embryonic stem cell and induced pluripotent stem cell derivation. Arch Immunol Ther Exp (Warsz). 2016;64(5):349–370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255. Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007;131(5):861–872. [DOI] [PubMed] [Google Scholar]
  • 256. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126(4):663–676. [DOI] [PubMed] [Google Scholar]
  • 257. D'Amour KA, Agulnick AD, Eliazer S, Kelly OG, Kroon E, Baetge EE. Efficient differentiation of human embryonic stem cells to definitive endoderm. Nat Biotechnol. 2005;23(12):1534–1541. [DOI] [PubMed] [Google Scholar]
  • 258. D'Amour KA, Bang AG, Eliazer S, Kelly OG, Agulnick AD, Smart NG, et al. Production of pancreatic hormone-expressing endocrine cells from human embryonic stem cells. Nat Biotechnol. 2006;24(11):1392–1401. [DOI] [PubMed] [Google Scholar]
  • 259. Rezania A, Bruin JE, Arora P, Rubin A, Batushansky I, Asadi A, et al. Reversal of diabetes with insulin-producing cells derived in vitro from human pluripotent stem cells. Nat Biotechnol. 2014;32(11):1121–1133. [DOI] [PubMed] [Google Scholar]
  • 260. Russ HA, Parent AV, Ringler JJ, Hennings TG, Nair GG, Shveygert M, et al. Controlled induction of human pancreatic progenitors produces functional beta-like cells in vitro. EMBO J. 2015;34(13):1759–1772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 261. Pagliuca FW, Millman JR, Gurtler M, Segel M, Van Dervort A, Ryu JH, et al. Generation of functional human pancreatic beta cells in vitro. Cell. 2014;159(2):428–439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 262. Hogrebe NJ, Maxwell KG, Augsornworawat P, Millman JR. Generation of insulin-producing pancreatic beta cells from multiple human stem cell lines. Nat Protoc. 2021;16(9):4109–4143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 263. Hogrebe NJ, Augsornworawat P, Maxwell KG, Velazco-Cruz L, Millman JR. Targeting the cytoskeleton to direct pancreatic differentiation of human pluripotent stem cells. Nat Biotechnol. 2020;38(4):460–470. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 264. Balboa D, Barsby T, Lithovius V, Saarimaki-Vire J, Omar-Hmeadi M, Dyachok O, et al. Functional, metabolic and transcriptional maturation of human pancreatic islets derived from stem cells. Nat Biotechnol. 2022;40(7):1042–1055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 265. Nair GG, Liu JS, Russ HA, Tran S, Saxton MS, Chen R, et al. Recapitulating endocrine cell clustering in culture promotes maturation of human stem-cell-derived beta cells. Nat Cell Biol. 2019;21(2):263–274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 266. Docherty FM, Riemondy KA, Castro-Gutierrez R, Dwulet JM, Shilleh AH, Hansen MS, et al. ENTPD3 marks mature stem cell-derived beta-cells formed by self-aggregation in vitro. Diabetes. 2021;70(11):2554–2567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 267. Weng C, Xi J, Li H, Cui J, Gu A, Lai S, et al. Single-cell lineage analysis reveals extensive multimodal transcriptional control during directed beta-cell differentiation. Nat Metab. 2020;2(12):1443–1458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 268. Velazco-Cruz L, Song J, Maxwell KG, Goedegebuure MM, Augsornworawat P, Hogrebe NJ, et al. Acquisition of dynamic function in human stem cell-derived β cells. Stem Cell Reports. 2019;12(2):351–365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 269. Yoshihara E, O'Connor C, Gasser E, Wei Z, Oh TG, Tseng TW, et al. Immune-evasive human islet-like organoids ameliorate diabetes. Nature. 2020;586(7830):606–611. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 270. Mahaddalkar PU, Scheibner K, Pfluger S, Sterr M, Beckenbauer J, et al. Generation of pancreatic beta cells from CD177+ anterior definitive endoderm. Nat Biotechnol. 2020;38(9):1061–1072. [DOI] [PubMed] [Google Scholar]
  • 271. Katsumoto K, Yennek S, Chen C, Silva LFD, Traikov S, Sever D, et al. Wnt4 is heterogeneously activated in maturing beta-cells to control calcium signaling, metabolism and function. Nat Commun. 2022;13(1):6255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 272. Schulz TC, Young HY, Agulnick AD, Babin MJ, Baetge EE, Bang AG, et al. A scalable system for production of functional pancreatic progenitors from human embryonic stem cells. PLoS One. 2012;7(5):e37004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 273. Ramzy A, Thompson DM, Ward-Hartstonge KA, Ivison S, Cook L, Garcia RV, et al. Implanted pluripotent stem-cell-derived pancreatic endoderm cells secrete glucose-responsive C-peptide in patients with type 1 diabetes. Cell Stem Cell. 2021;28(12):2047–2061.e5. [DOI] [PubMed] [Google Scholar]
  • 274. Shapiro AMJ, Thompson D, Donner TW, Bellin MD, Hsueh W, Pettus J, et al. Insulin expression and C-peptide in type 1 diabetes subjects implanted with stem cell-derived pancreatic endoderm cells in an encapsulation device. Cell Rep Med. 2021;2(12):100466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 275. Vertex Pharmaceuticals Inc . 2022. Vertex to Acquire ViaCyte, With the Goal of Accelerating its Potentially Curative VX-880 Programs in Type 1 Diabetes. Accessed August 29, 2022. Available from: https://investors.vrtx.com/news-releases/news-release-details/vertex-acquire-viacyte-goal-accelerating-its-potentially
  • 276. Desai T, Shea LD. Advances in islet encapsulation technologies. Nat Rev Drug Discov. 2017;16(5):338–350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 277. Wallace C, Smyth DJ, Maisuria-Armer M, Walker NM, Todd JA, Clayton DG. The imprinted DLK1-MEG3 gene region on chromosome 14q32.2 alters susceptibility to type 1 diabetes. Nat Genet. 2010;42(1):68–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 278. Westra HJ, Martinez-Bonet M, Onengut-Gumuscu S, Lee A, Luo Y, Teslovich N, et al. Fine-mapping and functional studies highlight potential causal variants for rheumatoid arthritis and type 1 diabetes. Nat Genet. 2018;50(10):1366–1374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 279. Marroqui L, Dos Santos RS, Op de Beeck A, Coomans de Brachene A, Marselli L, Marchetti P, et al. Interferon-alpha mediates human beta cell HLA class I overexpression, endoplasmic reticulum stress and apoptosis, three hallmarks of early human type 1 diabetes. Diabetologia. 2017;60(4):656–667. [DOI] [PubMed] [Google Scholar]
  • 280. Coomans de Brachene A, Castela A, Op de Beeck A, Mirmira RG, Marselli L, Marchetti P, et al. Preclinical evaluation of tyrosine kinase 2 inhibitors for human beta-cell protection in type 1 diabetes. Diabetes Obes Metab. 2020;22(10):1827–1836. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 281. Chandra V, Ibrahim H, Halliez C, Prasad RB, Vecchio F, Dwivedi OP, et al. The type 1 diabetes gene TYK2 regulates beta-cell development and its responses to interferon-alpha. Nat Commun. 2022;13(1):6363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 282. Parent AV, Faleo G, Chavez J, Saxton M, Berrios DI, Kerper NR, et al. Selective deletion of human leukocyte antigens protects stem cell-derived islets from immune rejection. Cell Rep. 2021;36(7):109538. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 283. Hampton RF, Jimenez-Gonzalez M, Stanley SA. Unravelling innervation of pancreatic islets. Diabetologia. 2022;65(7):1069–1084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 284. Dybala MP, Kuznetsov A, Motobu M, Hendren-Santiago BK, Philipson LH, Chervonsky AV, et al. Integrated pancreatic blood flow: bidirectional microcirculation between endocrine and exocrine pancreas. Diabetes. 2020;69(7):1439–1450. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 285. Almaça J, Weitz J, Rodriguez-Diaz R, Pereira E, Caicedo A. The pericyte of the pancreatic islet regulates capillary diameter and local blood flow. Cell Metab. 2018;27(3):630–644.e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 286. Vegas AJ, Veiseh O, Gurtler M, Millman JR, Pagliuca FW, Bader AR, et al. Long-term glycemic control using polymer-encapsulated human stem cell-derived beta cells in immune-competent mice. Nat Med. 2016;22(3):306–311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 287. Bluestone JA, Tang Q. Treg cells-the next frontier of cell therapy. Science. 2018;362(6411):154–155. [DOI] [PubMed] [Google Scholar]
  • 288. Boerner BP, George NM, Mir SUR, Sarvetnick NE. WS6 induces both alpha and beta cell proliferation without affecting differentiation or viability. Endocr J. 2015;62(4):379–386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 289. Shen W, Tremblay MS, Deshmukh VA, Wang W, Filippi CM, Harb G, et al. Small-molecule inducer of beta cell proliferation identified by high-throughput screening. J Am Chem Soc. 2013;135(5):1669–1672. [DOI] [PubMed] [Google Scholar]
  • 290. Alonso LC, Yokoe T, Zhang P, Scott DK, Kim SK, O'Donnell CP, et al. Glucose infusion in mice: a new model to induce beta-cell replication. Diabetes. 2007;56(7):1792–1801. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 291. Bernard C, Thibault C, Berthault MF, Magnan C, Saulnier C, Portha B, et al. Pancreatic beta-cell regeneration after 48-h glucose infusion in mildly diabetic rats is not correlated with functional improvement. Diabetes. 1998;47(7):1058–1065. [DOI] [PubMed] [Google Scholar]
  • 292. Bonner-Weir S, Deery D, Leahy JL, Weir GC. Compensatory growth of pancreatic beta-cells in adult rats after short-term glucose infusion. Diabetes. 1989;38(1):49–53. [DOI] [PubMed] [Google Scholar]
  • 293. Stamateris RE, Sharma RB, Kong Y, Ebrahimpour P, Panday D, Ranganath P, et al. Glucose induces mouse beta-cell proliferation via IRS2, MTOR, and Cyclin D2 but not the insulin receptor. Diabetes. 2016;65(4):981–995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 294. Terauchi Y, Takamoto I, Kubota N, Matsui J, Suzuki R, Komeda K, et al. Glucokinase and IRS-2 are required for compensatory beta cell hyperplasia in response to high-fat diet-induced insulin resistance. J Clin Invest. 2007;117(1):246–257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 295. Sachdeva MM, Stoffers DA. Minireview: meeting the demand for insulin: molecular mechanisms of adaptive postnatal beta-cell mass expansion. Mol Endocrinol. 2009;23(6):747–758. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 296. Yesildag B, Mir-Coll J, Neelakandhan A, Gibson CB, Perdue NR, Rufer C, et al. Liraglutide protects beta-cells in novel human islet spheroid models of type 1 diabetes. Clin Immunol. 2022;244:109118. [DOI] [PubMed] [Google Scholar]
  • 297. Paris M, Bernard-Kargar C, Berthault MF, Bouwens L, Ktorza A. Specific and combined effects of insulin and glucose on functional pancreatic beta-cell mass in vivo in adult rats. Endocrinology. 2003;144(6):2717–2727. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Data sharing is not applicable to this article as no datasets were generated or analyzed during the current study.


Articles from Endocrinology are provided here courtesy of The Endocrine Society

RESOURCES