Next Article in Journal
Ashwagandha-Induced Programmed Cell Death in the Treatment of Breast Cancer
Next Article in Special Issue
Capsaicin: Emerging Pharmacological and Therapeutic Insights
Previous Article in Journal
The Promising Potential of Caulerpa microphysa in Dermatology
Previous Article in Special Issue
The Latest Look at PDT and Immune Checkpoints
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Targeting β-Cell Plasticity: A Promising Approach for Diabetes Treatment

Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
*
Authors to whom correspondence should be addressed.
Curr. Issues Mol. Biol. 2024, 46(7), 7621-7667; https://doi.org/10.3390/cimb46070453
Submission received: 24 June 2024 / Revised: 11 July 2024 / Accepted: 15 July 2024 / Published: 18 July 2024
(This article belongs to the Special Issue Latest Review Papers in Molecular Biology 2024)

Abstract

:
The β-cells within the pancreas play a pivotal role in insulin production and secretion, responding to fluctuations in blood glucose levels. However, factors like obesity, dietary habits, and prolonged insulin resistance can compromise β-cell function, contributing to the development of Type 2 Diabetes (T2D). A critical aspect of this dysfunction involves β-cell dedifferentiation and transdifferentiation, wherein these cells lose their specialized characteristics and adopt different identities, notably transitioning towards progenitor or other pancreatic cell types like α-cells. This process significantly contributes to β-cell malfunction and the progression of T2D, often surpassing the impact of outright β-cell loss. Alterations in the expressions of specific genes and transcription factors unique to β-cells, along with epigenetic modifications and environmental factors such as inflammation, oxidative stress, and mitochondrial dysfunction, underpin the occurrence of β-cell dedifferentiation and the onset of T2D. Recent research underscores the potential therapeutic value for targeting β-cell dedifferentiation to manage T2D effectively. In this review, we aim to dissect the intricate mechanisms governing β-cell dedifferentiation and explore the therapeutic avenues stemming from these insights.

Graphical Abstract

1. Introduction

Globally, around 422 million people have Diabetes Mellitus (DM or diabetes), with most living in low- and middle-income countries. Diabetes causes 1.5 million deaths annually worldwide. In the United States, approximately 37.3 million people, or about 11% of the population, have diabetes, with Type 2 Diabetes (T2D) accounting for 90–95% of cases. Projections indicate that the global number of adults with diabetes will rise to 643 million by 2030 and 783 million by 2045.
Findings indicate that changing β-cell plasticity plays an important role in the incidence of all three main forms of diabetes (see Box 1). In T2D, for example, β-cell dedifferentiation and transdifferentiation are more critical than β-cell loss [1,2]. Various factors, including metabolic stress, inflammation [3], and viral infection [4], contribute to these processes. They exert their effects through pathways such as the mTOR pathway, ER stress, and by altering the transcriptional activity of β-cells, targeting key transcription and epigenetic factors (Figure 1).
The concept of β-cell dedifferentiation was introduced by Gershengorn et al. in 2004 after they observed that β-cells in human pancreatic islets vanished and were replaced by fibroblast-like precursor cells. They also noted that these cells could redifferentiate into α- and β-cells under certain growth conditions. This concept was further confirmed by the findings of Dor et al. (2007), Accili et al. (2012), Wang et al. (2014), and Cinti et al. (2016) [5]. β-cell dedifferentiation is characterized by the appearance of progenitor-like biomarkers, such as Ngn3, Oct4, Aldh1a3, Nanog, L-Myc, and Sox9, and the disappearance or downregulation of β-cell-enriched biomarkers, including MafA, Ins, Nkx6.1, Glut2, and Pdx-1. This shift results in β-cells losing their functionality, which can ultimately lead to the incidence of diabetes [5,6]. Similar to rodents, β-cell dedifferentiation occurs in human pancreatic islets. However, unlike in rodents, Ngn3 is not upregulated in human pancreatic islets during dedifferentiation. Aldh1a3, on the other hand, is upregulated in both rodents and humans [7]. The transdifferentiation of β-cells into other pancreatic cell types, such as α- and δ-cells, is another form of β-cell plasticity that occurs in both rodents and humans, much like dedifferentiation [6]. The transdifferentiation of β-cells into α-cells is typically marked by the loss of β-cell differentiation markers and the acquisition of α-cell biomarkers, such as glucagon and the Arx transcription factor [8]. The transdifferentiation of pancreatic endocrine cells, including α- and δ-cells, into β-cells is observed alongside reports of pancreatic exocrine cells, like acinar cells, also transdifferentiating into β-cells. This suggests a novel approach for treating diabetes [9].
The dedifferentiation and transdifferentiation of β-cells are observed across all diabetes types. In Type I Diabetes (T1D), inflammatory reactions are pivotal in triggering these processes, inducing ER stress and modifying gene expression and protein synthesis in β-cells [10]. In T2D, metabolic stresses linked to obesity, insulin resistance, and high levels of glucose and lipids similarly contribute to β-cell dedifferentiation and transdifferentiation [11].
Mitigating and reversing β-cell dedifferentiation and transdifferentiation offer promising treatments for all three main types of diabetes. Various approaches, including natural products, dietary interventions, and common antidiabetic therapies, can help mitigate β-cell dedifferentiation and transdifferentiation.
In addition, in vitro β-cell neogenesis followed by the transplantation of newly differentiated β-cells is a promising method for treating all types of diabetes, particularly T1D. We have highlighted new findings in this area.
β-cell regeneration and proliferation represent another strategy for treating all types of diabetes. Suppressing Dual-Specificity Tyrosine-Regulated Kinase 1A (DYRK1A) [12], antagonizing glucagon receptors [13,14,15,16], and inhibiting polyamine biosynthesis [17] are potential treatments to promote β-cell regeneration and proliferation.
Box 1. Definitions.
 β-cell plasticity refers to the ability of β-cells to alter their identity and function in response to specific conditions. This trait is not exclusive to β-cells; other pancreatic cells, such as α-cells, also exhibit this adaptability. The primary aspects of β-cell plasticity are β-cell dedifferentiation and transdifferentiation [18].
 β-cell dedifferentiation is triggered by chronic metabolic stresses like hyperglycemia, endoplasmic reticulum (ER) stress, and inflammatory cytokines. This process involves mature β-cells losing their differentiated characteristics and reverting to a progenitor-like or undifferentiated state [2].
 β-cell transdifferentiation involves the conversion of mature β-cells into other islet cell types, such as α-cells, under certain conditions.
 β-cell regeneration is the process of restoring or increasing the number of functional pancreatic β-cells, which produce insulin. This process can occur through the body’s natural mechanisms, involving two main pathways:
 1—β-cell proliferation: Existing β-cells divide to increase their numbers.
 2—Neogenesis from progenitor cells: Pancreatic stem or progenitor cells differentiate into new β-cells [19].
 T1D—Type 1 Diabetes: an autoimmune condition where the immune system attacks insulin-producing β-cells in the pancreas, leading to an inability to produce insulin. It accounts for about 10% of diabetes cases and is usually diagnosed in childhood or adolescence, though it can occur at any age [20].
 T2D—Type 2 Diabetes: the most common form caused by insulin resistance and relative insulin deficiency. It is often associated with obesity, a sedentary lifestyle, and genetic factors. While it is typically diagnosed in adulthood, it is increasingly seen in children and teens due to rising obesity rates. Management can involve lifestyle changes, medications, or insulin [21,22].
 GDM—Gestational Diabetes Mellitus arises when the body struggles to produce sufficient insulin to counteract the insulin resistance induced by placental hormones such as estrogen, cortisol, and human placental lactogen [23].
In this review, we explore the mechanisms behind β-cell dedifferentiation and transdifferentiation and provide insights into the possibility of mitigating these processes by identifying new therapeutic targets, medications, and treatments. Additionally, we highlight recent discoveries related to β-cell neogenesis and regeneration as potential treatments for all three types of diabetes.

2. The Main Mechanisms Involved in β-Cell Function and Identity

2.1. Inflammation

Inflammation, whether originating from the β-cells themselves or from resident macrophages in the pancreas, plays a critical role in the function and maintenance of β-cells, thereby contributing to the development of diabetes [3]. Pro-inflammatory cytokines like IL-1β, IL-6, and TNFα induce β-cell dedifferentiation in cultured human cells, human islets, and mouse islets, contributing to β-cell dysfunction in T2D. IL-1β is the most potent among them, downregulating Forkhead Box O1 (FoxO1), a key transcription factor for maintaining β-cell identity. Additionally, treatment with proinflammatory cytokines was associated with a translocation of FoxO1 to the nucleus, the loss of nuclear Nkx6.1 expression, and the emergence of Aldh1a3, indicating β-cell dedifferentiation [24]. Accordingly, in vivo, anti-inflammatory treatments with anti-IL-1β, anti-TNFα, or NF-kB inhibiting sodium salicylate improved insulin secretion from isolated islets. While anti-TNFα therapy partially prevented the loss of β-cell identity gene expression, the combined antagonism of IL-1β and TNFα produced a synergistic effect in restoring insulin secretion. Although inflammation plays a significant role in β-cell dedifferentiation in vitro, its impact on in vivo insulin secretion improvement appears to be more complex, with additional factors potentially involved [25].
The pro-inflammatory factor BMP-2, upregulated in the islets of diabetic individuals, contributes to the inhibition of β-cell function and proliferation. BMP-2 disrupts β-cell maturity through epigenetic changes and decreased NeuroD1 activity, contributing to β-cell dysfunction in diabetes. Exposure to BMP-2 for 10 days impaired glucose-stimulated insulin secretion (GSIS) and β-cell proliferation, correlating with decreased expressions of key β-cell genes like Ins1, Ucn3, and Ki67. BMP-2-induced β-cell dysfunction was also linked to an increased expression of dedifferentiation regulators (Id1-4, Hes-1, and Hey-1), a reduction in H3K27 acetylation, and decreased NeuroD1 DNA binding activity [26].
TGF-β, a versatile cytokine, coordinates various cellular processes such as growth, proliferation, differentiation, apoptosis, and immune responses. It plays a crucial role in regulating β-cell dedifferentiation, preserving β-cell function, and managing β-cell mass [27]. Inhibitors targeting TGF-β receptor I (TGFβ1) have shown potential in restoring β-cell identity by influencing the expression of β-cell-specific transcription factors under severe diabetic conditions [28].
Patients with chronic pancreatitis (CP) are at risk of developing diabetes due to the gradual loss of functional β-cells. In a mouse model for CP-related diabetes (CPRD), researchers found that β-cell loss is driven by TGFβ1-induced epithelial–mesenchymal transition (EMT), not apoptosis. They also observed increased angiogenesis in CP patients’ inflamed pancreata and in mice subjected to pancreatic duct ligation, a surgical procedure used in research to induce pancreatitis. However, when angiogenesis was blocked with an anti-vascular endothelial growth factor receptor 2 antibody (DC101), the EMT process reverted to β-cell apoptosis. This indicates that angiogenesis helps β-cells survive in the inflamed pancreas, suggesting a potential therapeutic target for CPRD [29].
Activin is a protein that belongs to the TGF-β superfamily. It plays crucial roles in various biological processes, including embryonic development, cell proliferation, differentiation, and tissue homeostasis. Its signaling influences the balance between α- and β-cell populations, impacting glucose homeostasis. Accordingly, it has been found that Activin can suppress genes in α-cells such as Arx4 and upregulate β-cell genes such as Pax4, which accelerates the process of transdifferentiation from α- to β-cells [30].
In diabetes, stromal cell-derived factor-1 (SDF-1), a chemokine, is implicated in the dedifferentiation of islet β-cells. Elevated SDF-1 levels correlate with the increased dedifferentiation of β-cells in pancreatic tissues. Experimental inhibition of SDF-1 led to a higher proportion of dedifferentiated cells, while overexpression reduced their numbers. Despite the diabetes-induced upregulation of SDF-1, its activity may decline, thereby impacting dedifferentiation. SDF-1 exerts its effects through CXCR4, activating AKT and FoxO1 [31].
T2D exhibits hyperglycemia and inflammation, paralleled by the upregulation of prostaglandin E2 (PGE2). A systemic blockade of the PGE2 receptor EP3 promotes β-cell proliferation and mass without affecting α-cells. EP3 blockade reverses islet gene expression changes in diabetic mice, restoring normal islet architecture. Additionally, it increases Glp1r expression and activates Nrf2, potentially protecting β-cells from oxidative stress. EP3 blockade shows promise for enhancing β-cell mass in T2D [32].
Insulitis, or inflammation of β-cells, is most commonly associated with T1D. Under this condition, β-cells are usually exposed to a variety of cytokines, including IFNγ, IL-1β, TNFα, and IL-17, all of which are able to trigger ER stress and β-cell dedifferentiation [10]. There is some evidence indicating that the direct interaction of autoimmune T cells with β-cells may be responsible, but not sufficient, for changes in the β-cell identity in T1D patients [33]. However, the islet microenvironment in T1D has been reported to generate CD4 T cells that target proinsulin, suggesting that proinflammatory mediators from islets may cause β-cell dedifferentiation in individuals with T1D [34]. Moreover, it has been found that exposing β-cells to double-stranded RNA, mimicking a viral infection that could trigger T1D, reduces the expressions of β-cell-enriched genes like MafA and Ins while increasing progenitor markers. This effect is driven by NF-kB within the β-cells and the interferon alpha released by neighboring cells [35,36].
The ablation of β-cells, which is associated with T1D, induces the transdifferentiation of α-cells into β-cells [37]. This indicates that cell plasticity could be used as a potential treatment for T1D. It is worth mentioning that in the context of T1D, the immune system does not attack α-cells [38].

2.2. Metabolic Stress

Metabolic stressors, including glucotoxicity, lipotoxicity, and glucolipotoxicity, have been demonstrated to impair β-cell function and identity by disrupting key metabolic pathways.
Glucotoxicity has been shown to inhibit β-cell regeneration. In a mouse model, where β-cells are ablated, the usual regenerative response includes β-cell proliferation and the normalization of blood glucose levels. However, if the β-cell loss leads to severe hyperglycemia (above 28 mmol/L), regeneration fails, causing permanent diabetes, despite some surviving β-cells entering the cell cycle. Reducing hyperglycemia with insulin, SGLT2 inhibitors, or a ketogenic diet for about three weeks can partially restore β-cell mass and aid in diabetes recovery, indicating that extreme glucotoxicity hinders regeneration. Gene expression analysis suggests that high glucose levels cause metabolic stress and suppress key cell-cycle genes, potentially blocking regeneration [39].
Persistent hyperglycemia maintains a cycle of β-cell dysfunction. Using advanced optical imaging, researchers found that streptozotocin (STZ) predominantly damages larger islets in the pancreatic core, leading to a significant reduction in β-cell mass. However, even in hyperglycemic STZ-treated mice, a considerable number of β-cells remain but with a decreased expression of Glut2 (Slc2a2), indicating impaired β-cell maturity. Islet gene expression studies confirm this downregulation and suggest that hyperglycemia causes β-cell dysfunction more than ablation. Islet transplantation partially restored some markers of islet function but not the overall β-cell mass [40].
The hyperactive isoform of carbohydrate response-element-binding protein (ChREBPβ) plays a crucial role in the response of β-cells to hyperglycemic stress. It is involved in adaptive β-cell expansion when faced with metabolic challenges like prolonged glucose exposure and HFD. However, chronic overexpression of ChREBPβ in β-cells can lead to a loss of β-cell identity, increased apoptosis, reduced β-cell mass, and diabetes. Deleting ChREBPβ can prevent β-cell “glucolipotoxicity”, while the overexpression of ChREBPα or activation of the antioxidant Nrf2 pathway can mitigate ChREBPβ-induced β-cell death [41].
Stearoyl CoA desaturase (SCD), an enzyme pivotal in lipid metabolism, is highly expressed in β-cells and human islets. The silencing of SCD prompts inflammation, ER stress, and elevated IAPP mRNA levels. Conversely, treatment with SCD products counteracts these effects. Moreover, when SCD is knocked down and exposed to palmitate, dedifferentiation markers are induced. Notably, SCD knockdown alone disrupts β-cell identity, diminishing mature β-cell markers and compromising insulin secretion [42].
Accordingly, it has been found that the inhibition of stearoyl-CoA desaturase 1 (SCD1) is associated with increased methylation in Pdx1 and MafA promoter regions. This inhibition also skews pancreas development towards α-cell formation, disrupting the α-cell-to-β-cell ratio and islet structure. Consequently, islet dysfunction occurs, characterized by impaired insulin secretion and elevated glucagon levels [43].
Oxidative stress originating from hyperglycemia and hyperlipidemia can cause a significant decline in human pancreatic β-cell function. This decline is characterized by the activation of ER stress and the nuclear translocation of FoxO1. This stress response results in the downregulation of the maturity genes MafA and Pdx1 along with the upregulation of the progenitor markers Sox9 and Hes1, suggesting a potential process of partial dedifferentiation. These findings highlight the critical importance of targeting antioxidant mechanisms to preserve functional β-cell mass, particularly in the early stages of T1D [44].
During the insulin resistance phase of T2D, β-cells exhibit persistently high calcium levels. Initially, this adaptation may assist β-cells in managing the metabolic stress induced by hyperglycemia. However, prolonged exposure to elevated calcium levels may lead to alterations in β-cell function and identity, as well as diabetic complications. Single-cell RNA sequencing of pancreatic islets from prediabetic and diabetic mice unveiled a diabetes-specific transcriptomic profile. It highlighted Anxa10, a gene linked to prediabetes, as being upregulated by persistently high calcium levels in β-cells. Pseudotime analysis (computational ordering of cells in the progression of differentiation) suggested that disturbances in calcium influx during the insulin resistance phase and heightened Anxa10 expression could induce mitochondrial dysfunction, ER stress, and β-cell transdifferentiation into acinar cells, offering new insights into diabetes progression [45].
PARP1 is related to metabolic stress through its involvement in DNA repair mechanisms and cellular responses to stressors such as oxidative stress and inflammation. During metabolic stress, such as high glucose levels or lipid overload, PARP1 can be activated excessively, leading to the increased production of poly(ADP-ribose) polymers (PARylation). Research shows that PARP1 knockout (KO) mice exhibit resistance to diabetes, and inhibiting PARP1, particularly with PJ34, alleviates complications in type 2 diabetes patients. Complete PARP1 depletion halted β-cell formation, whereas its inhibition enhanced β-cell differentiation and maturation via p38 MAPK phosphorylation and Ngn3 reactivation, implying PARP1’s necessity for proper β-cell development [46].
Maintaining lysosomal activity and mitophagy is crucial for the differentiation and maturation of β-cells. The Niemann-Pick C1 protein (NPC1) on the lysosome membrane is essential for intracellular cholesterol transport and lysosome function. Research links NPC1 mutations to pancreatic islet dysfunction in T2D. NPC1 depletion in mice impairs β-cell viability, expansion, and GSIS, disrupting mitochondrial function through compromised lysosomal activity and mitophagy, indicating metabolic stress [47].
Insulin granule maturation in pancreatic β-cells relies heavily on phosphatidylinositol transfer protein α (PITPNA). Reduced PITPNA levels in β-cells from individuals with T2D result in impaired GSIS, diminished β-cell mass, and increased ER stress, reflecting metabolic stress. The conditional deletion of PITPNA in mice causes hyperglycemia and compromised insulin processing. Silencing PITPNA in human islets yields similar defects that can be reversed by restoring PITPNA levels [48].

2.3. Age-Related Dynamics of β-Cell Dedifferentiation and Transdifferentiation

The impact of age on β-cell dedifferentiation and transdifferentiation is increasingly recognized, with studies revealing a significant rise in dedifferentiated cells among older age groups. This phenomenon is accompanied by diminished FoxO1 activity and reduced UCN3 expression in aging β-cells along with a decline in adaptive unfolded protein response (UPR) components. Glucose-regulated protein 94 (GRP94), also known as heat shock protein 90B1 (HSP90B1), is an ER chaperone that plays a crucial role in protein folding, quality control, and the maintenance of ER homeostasis. Lowering GRP94 levels has been shown to induce dedifferentiation, while restoring GRP94 levels preserves β-cell identity in aging cells [49].
β-cell regeneration becomes challenging with advancing age, particularly in the context of adult-onset diabetes. In middle-aged rats, β-cell aging is notably influenced by compromised duct cell plasticity and β-cell dedifferentiation. Despite observed increases in hypertrophy and hyperplasia, the regenerative potential of functional β-cell mass is limited. This implies the critical role of β-cell dedifferentiation and potential transdifferentiation in the aging process [50].
Excessive glycolysis may contribute to age-related β-cell dysfunction and the loss of cellular identity. Both aged and diabetic mouse β-cells exhibit common dysfunctional traits, including heightened glucose sensitivity and compromised cellular integrity. They also share an increase in glycolysis driven by elevated Nmnat2 expression, which is crucial for NAD synthesis. Knocking out Got1, a gene associated with glycolysis, replicated these traits, which are observed in aging and diabetes. Restoring the β-cell function and integrity was achieved by modulating glycolysis or the Nmnat2 activity in Got1 KO β-cells [51].

2.4. Sex-Dependent Variations in Pancreatic Islet Plasticity

Sex influences β-cell plasticity, as evidenced by the differences observed in pancreatic islet structures between male and female mice. Male control mice exhibit larger pancreatic islets and experience a greater reduction in islet and β-cell areas after STZ exposure compared to females. Conversely, female mice treated with STZ show lower rates of β-cell apoptosis. Hydrocortisone (HC) administration results in a higher proportion of α-cells in female islets, which is accompanied by a slight increase in β-cell apoptosis. Healthy female mice demonstrate a higher rate of α-to-β-cell transdifferentiation, which decreases with HC treatment, while STZ treatment increases α-cell transdifferentiation to β-cells in males but not females. These findings underscore the impact of sex hormones on islet morphology and function [52].

2.5. The Impacts of Chemicals on β-Cell Plasticity

Compounds like ethanol, STZ, glucocorticoids, and environmental pollutants impact different aspects of diabetes, including the dedifferentiation and transdifferentiation of β-cells.
Alcohol addiction is a serious global issue, responsible for 5.9% of all deaths worldwide. Regular alcohol consumption heightens the risk of metabolic diseases, including T2DM, compared to nonconsumption. Chronic alcohol consumption results in reduced insulin levels, β-cell apoptosis, and decreased β-cell mass along with upregulated Fibroblast Growth Factor 21 (FGF21) and its receptor mRNA levels, indicating FGF21 resistance. Short-term ethanol exposure does not immediately impair β-cell function, but prolonged exposure progresses to significant dysfunction. Chronic binge drinking is linked to reductions in insulin, the insulin-to-glucagon ratio, the GSIS response, and NKX2.2 and PDX1 levels, highlighting alcohol’s impact on β-cell dedifferentiation [53]. Additionally, there is a negative correlation between alcohol consumption and blood adiponectin levels [54], which diminish insulin expression and secretion [55] as well as β-cell survival [56].
STZ is a naturally occurring compound used to induce diabetes in experimental models by selectively destroying insulin-producing β-cells in the pancreas through DNA alkylation and oxidative stress. It targets β-cells specifically due to their high expression of the Glut2 glucose transporter, which facilitates STZ uptake and leads to cell death. However, some β-cells with lower Glut2 expression survive the initial damage but experience long-term functional and phenotypic changes, remaining in a low-Glut2 state. It has been found that some interventions, such as islet transplantation and glucose normalization, can partially restore β-cell function and Glut2 expression [57,58]. STZ treatment potentially promotes β-cell regeneration and transdifferentiation in pancreatic islets. This includes the emergence of vimentin+/MafB+ cells, which can differentiate into insulin-producing β-like cells, and the possibility of α-cells transdifferentiating into β-cells under specific conditions [1,59].
Glucocorticoids such as dexamethasone and hydrocortisone can induce β-cell dedifferentiation through various mechanisms, including triggering ER stress, changes in gene expression [60], impaired β-cell glucose metabolism, and reduced sensitivity to glucose stimulation, leading to altered insulin secretion patterns and decreased GSIS [61,62]. Furthermore, excess glucocorticoid exposure during fetal development can permanently alter β-cell mass and function, resulting in long-term consequences for glucose homeostasis [63].
Bisphenols (BPs) and phthalates are classes of industrial chemicals widely used in the production of plastics and other consumer products. These chemicals have been linked to higher diabetes rates, particularly affecting β-cells. Exposure to these chemicals disrupts electrical activity, mitochondrial function, gene expression, and β-cell function, leading to altered GSIS and impaired β-cell proliferation [64]. Studies show mixed results, with some indicating heightened insulin secretion and content, while others suggest diminished GSIS or insulin levels. These substances can also impact β-cell survival, with higher doses or prolonged exposure often leading to increased cell death [65]. BPs induce ER stress, mitochondrial dysfunction, and oxidative stress in β-cells, contributing to cellular damage, apoptosis, and plasticity [66,67]. The effects on β-cell proliferation vary, with some studies showing increased proliferation at lower doses [68] and others observing reduced growth at higher concentrations [69]. Bisphenol A (BPA) acts as an endocrine disruptor, altering gene expression in β-cells during development and producing long-term metabolic disturbances. Early-life exposure to BPA increases β-cell mass and proliferation through estrogen receptor β (ERβ), resulting in hyperinsulinemia that precedes insulin resistance [70,71].
Phthalates affect insulin secretion in β-cells variably depending on the duration and concentration of exposure. Acute high-concentration exposure increases basal insulin secretion (BIS) without affecting GSIS. In contrast, prolonged exposure typically inhibits both BIS and GSIS, indicating impaired insulin synthesis and β-cell function, often coupled with reduced β-cell survival and increased cellular stress [64,72].

3. Molecular Pathways Involved in β-Cell Plasticity

3.1. ER Stress

ER stress results from the buildup of misfolded proteins, activating the UPR to restore normalcy. Environmental factors like inflammation and oxidative stress can trigger ER stress, which is implicated in T1D and T2D onset [73,74]. ER stress activates the UPR, which is mediated by sensors like IRE1α, PERK, and ATF6, which aim to restore ER homeostasis [75].
Prolonged ER stress can lead to cellular dysfunction and apoptosis, particularly affecting pancreatic β-cells. When β-cells experience ER stress, they need to activate mechanisms to adapt and manage this stress effectively. Chronic stress induces transcriptional and translational changes, impairing β-cell function and identity regulators. Despite having some capacity to adapt to stress, β-cells under prolonged stress can eventually lose their functional identity. This means they no longer produce insulin efficiently, which is crucial for regulating blood sugar levels.
Single-cell RNA sequencing from T1D patients reveals severe disruptions in stress adaptation, indicating a role for β-cell adaptive exhaustion in diabetes progression [76].
A study by Hugo Lee et al. (2020) discovered that the β-cell deletion of IRE1α in Non-Obese Diabetic (NOD) mice, in the T1D model, led to transient β-cell dedifferentiation, temporarily protecting against immune-mediated β-cell destruction. Additionally, IRE1α influences the stability of TXNIP mRNA, impacting NLRP3 inflammasome activation and cell death pathways [77].
The X-box binding protein 1 (XBP1) serves as a pivotal regulator of the UPR within the ER. Extensive evidence implies the crucial role of XBP1 in preserving β-cell identity and function, especially during periods of metabolic stress. The depletion of XBP1 results in β-cell dysfunction, dedifferentiation, and heightened vulnerability to diabetes, underscoring its significance in maintaining β-cell homeostasis and regulating insulin secretion [78].
Like T1D, T2D is also characterized by increased ER stress and altered insulin processing, leading to a loss of β-cell identity and function. The transition from impaired glucose tolerance to diabetes, as a hallmark of T2D, is marked by a significant increase in ER stress markers and a decrease in β-cell phenotype markers. β-cell dedifferentiation appears as the disease progresses, with islets showing reduced expressions of key genes such as Nkx6.1 and MafA. Additionally, these changes were accompanied by an increase in altered proinsulin processing and intracellular localization, indicating a deterioration of the β-cell secretory machinery [73].
ATF4, a key regulator of ER stress responses, contributes to pancreatic β-cell dysfunction in T2D. It induces phosphodiesterase 4D (PDE4D) expression, disrupting cAMP signaling and accelerating T2D progression. Pharmacological inhibition of the ATF4 pathway reduces PDE4D expression, improving glucose tolerance and insulin secretion. The administration of Sephin1, a selective inhibitor of a holophosphatase with a modulatory impact on ER stress, enhances glucose metabolism in Akita mice by increasing insulin-positive areas and ATF4 expression. Conversely, β-cell-specific ATF4 knockout (βAtf4-KO) mice exhibit exacerbated diabetes, with increased dedifferentiation markers and a reduced expression of the Atonal homolog 8 transcription factor (Atoh8) [79,80].
ATF4 is also a key player in integrated stress response (ISR). ISR is a complex cellular signaling pathway in eukaryotic cells that manages and mitigates various stress stimuli. Essential for maintaining cellular homeostasis, the ISR can be triggered by intrinsic and extrinsic factors such as hypoxia, amino acid deprivation, glucose deprivation, viral infections, and oxidative stress [81]. The ISR is driven by the phosphorylation of eukaryotic translation initiation factor 2 alpha (eIF2α) by one of four kinases: PERK, GCN2, PKR, or HRI. This phosphorylation reduces global protein synthesis, conserving resources and alleviating the load on the protein-folding machinery. At the same time, it promotes the translation of specific genes, such as the transcription factor ATF4, which, in turn, aids in cellular adaptation to stress by upregulating the genes involved in both protein folding and amino acid metabolism [82]. The activation of the ISR in β-cells is found to regulate oxidative stress, a key player in β-cell dedifferentiation. This supports the observations indicating that the phosphorylation of eIF2α has a protective impact on β-cells against oxidative stress caused by insulin resistance through an ATF4-dependent mechanism [83]. Amino acid deficiency, another inducer of the ISR, stimulates the binding of uncharged tRNA to GCN2, leading to the autophosphorylation of GCN2 and subsequent phosphorylation of eIF2α [84]. This phosphorylation of eIF2α is associated with an increase in ATF4 levels, which upregulates the genes involved in the synthesis and metabolism of amino acids [85]. The upregulation of ATF4 also increases the expression of sesterin2, thereby inhibiting mTORC1. It is noteworthy that the chronic activation of mTORC1 during the insulin resistance phase negatively impacts β-cell mass and proliferation [86].
In summary, ATF4 upregulation, whether through ER stress or through the ISR, has a great impact on the β-cell fate.
Both high-glucose- and cytokine-induced ER stress activate intracellular calcium and reactive oxygen species (ROS) signals in pancreatic β-cells. These signals trigger calcineurin (CN)/NFATc2 and PI3K/AKT pathways, regulating gene expression that is critical for β-cell function. While inducing the endocrine differentiation gene RFX6, they suppress “disallowed” genes like MCT1, which are detrimental to β-cell identity. CN/NFATc2, along with co-activator p300 and co-repressor HDAC1, orchestrates these gene expression changes. Prolonged stress or disrupted CN/NFATc2 signaling leads to β-cell dedifferentiation, marked by reduced RFX6 and elevated MCT1 levels [87].

3.2. Angiotensin Receptors

Angiotensin receptors are cell surface receptors that bind and respond to angiotensin peptides produced by the renin–angiotensin system (RAS). There is an interaction between the renin–angiotensin aldosterone system (RAAS) and ER stress in pancreatic β-cells, particularly in the context of T2D [88]. The activation of the Angiotensin II (Ang II) receptor is linked to the initiation of β-cell dedifferentiation and the development of T2D. This occurs through the activation of the RAS and subsequent NFκB activation [89].
The activation of Angiotensin II exerts its negative impact on β-cell plasticity by increasing ER stress markers and reducing insulin secretion in clonal INS-1E β-cells and human islets. Accordingly, it has been found that the inhibition of Ang II production or the blockade of its receptors alleviates ER stress and restores insulin secretion. These findings elucidate a potential mechanism linking pancreatic RAAS activation to β-cell dysfunction in T2D, highlighting the therapeutic potential of RAAS inhibitors in T2D management [88].

3.3. mTOR Pathway

The protein kinase mTOR (the mechanistic target of rapamycin) regulates cell growth, proliferation, and survival, impacting cellular metabolism and protein synthesis. mTOR activity decreases in mice fed a high-fat diet (HFD), leading to the initial enlargement but subsequent diminishment of pancreatic islets. This decrease in mTOR activity correlates with increased β-cell death, reduced levels of myeloid cell leukemia 1 (Mcl-1), a Bcl-2 family member crucial for pancreatic β-cell survival, and impaired insulin secretion in response to glucose. Additionally, the activation of AMPK by palmitic acid (PA) degrades Mcl-1 via pathways such as ERK and GSK3β, further contributing to β-cell dysfunction and loss of identity [90].
The growth factor receptor-bound protein 10 (GRB10), a negative regulator of insulin and mTORC1 signaling, is significantly elevated in the islets of diabetic mice and insulinoma cells treated with high glucose. β-cell-specific knockout of GRB10 in mice increased β-cell mass and function along with enhanced mTORC1 signaling and reduced β-cell dedifferentiation, reversible by rapamycin. Conversely, the overexpression of GRB10 in MIN6 cells induced β-cell dedifferentiation, indicating potential therapeutic targeting of GRB10 to address β-cell dysfunction in diabetes [91].
The deletion of the β-cell-specific gene Raptor, a component of mTORC1, resulted in decreased β-cell mass, the adoption of α-cell characteristics, and loss of β-cell identity, even after glucose normalization. A Raptor deficiency directly impaired β-cell identity, mitochondrial function, and protein synthesis, leading to β-cell dysfunction. Additionally, Raptor loss activated MafB, a transcription factor associated with α-cells, and genes specific to α-cells, promoting a transition from β- to α-cell identity. These findings highlight mTORC1’s role in maintaining β-cell identity and suppressing α-cell characteristics under normal glucose conditions [92].
Additionally, the mTORC1 pathway regulates β-cell functional maturation through epigenetic mechanisms. Utilizing microarray, MeDIP-seq, and ATAC-seq analyses, it has been uncovered that DNMT3A-dependent DNA methylation and PRC2-dependent H3K27me3 modification are key silencing mechanisms in immature β-cells lacking Raptor. Overexpressing DNMT3A partially reverses the immature transcriptome pattern and restores impaired GSIS in Raptor-deficient β-cells. Furthermore, Raptor directly regulates PRC2/EED and H3K27me3 expressions, suggesting a complex interplay between nutrient sensing, epigenetic modifications, and β-cell function, with implications for T2D [93].
Rictor, a key component of mTORC2, regulates β-cell proliferation and function, with long chain acyl-CoA synthetase 4 (Acsl4) potentially downstream. Acsl4 is positively regulated by Rictor at both transcriptional and post-translational levels in mouse β-cells. Adenovirus-mediated Acsl4 expression rescued β-cell dysplasia in βRicKO islets but not dysfunction. However, Acsl4 could not fully restore glucose oxidation in Rictor-deficient β-cells, instead promoting lipid oxidation. Epigenetic analyses revealed signatures of β-cell dedifferentiation and an altered oxidative response in Rictor-lacking cells expressing Acsl4, suggesting a context-dependent role for Acsl4 in β-cell dedifferentiation under reduced Rictor/mTORC2 activity [94].
mTORC1 activation under chronic metabolic stress leads to increased levels of the phosphatases PHLPP1 and PHLPP2 in diabetic β-cells. These elevated phosphatases contribute to β-cell failure in diabetes by regulating survival kinases such as AKT and MST1. Inhibiting PHLPPs genetically enhances β-cell survival and function in diabetic models and human T2D islets, suggesting a potential therapeutic target for diabetes [95].

3.4. Role of Ubiquitin–Proteasome System in the Pancreatic β-Cell Plasticity

The ubiquitin–proteasome system (UPS) regulates cellular protein degradation, impacting various biological processes like inflammation and apoptosis. Emerging evidence suggests its crucial involvement in pancreatic β-cell transdifferentiation in diabetes, presenting a potential treatment avenue [96,97].
The UPS, which includes E3 ligases and deubiquitinases (DUBs), regulates several aspects of β-cell function. The inhibition of deubiquitinase USP1, a key player in β-cell dedifferentiation, either through genetic intervention or using the small-molecule inhibitor ML323, restored the epithelial phenotype of β-cells. Conversely, the overexpression of USP1 induced dedifferentiation, which was mediated by modulating the expression of the inhibitor of differentiation 2 (ID2). In an STZ-induced mouse model of β-cell dedifferentiation, treatment with ML323 improved hyperglycemia [98].
Ubiquitin-Specific Peptidase 7 (USP7) stabilizes Ngn3 by deubiquitination. The conditional knockout of Usp7 in mice reduces islet formation and causes hyperglycemia due to impaired Ngn3-mediated endocrine specification. Pharmacological inhibition of USP7 in human iPSC models also decreases Ngn3 progenitors and impairs β-cell differentiation, highlighting the importance of the USP7-Ngn3 axis in preventing β-cell dedifferentiation [99].
Ubiquitin-mediated degradation of Ngn3, regulated by PRMT1-mediated arginine 65 methylation, is essential for β-cell differentiation. Protein arginine methyltransferase-1 (PRMT1) knockout in human embryonic stem cells (hESCs) leads to Ngn3 accumulation in the cytoplasm, reducing its transcriptional activity and preventing endocrine cell differentiation. This methylation acts as a molecular switch, ensuring Ngn3’s proper localization and function, thus preventing β-cell dedifferentiation [100].

4. Transcriptional Regulation in β-Cell Plasticity

Transcription factors are crucial for establishing and maintaining the mature phenotype of β-cells as well as regulating their plasticity through processes such as dedifferentiation, transdifferentiation, and regeneration. Key transcription factors, including Pdx1, Ngn3, Pax4, MafA, FoxO1, and Nkx6.1, are essential for β-cell identity, function, and survival. The dysregulation of these factors can result in dedifferentiation, where β-cells lose their mature state and revert to a progenitor-like condition.
Furthermore, alterations in the transcriptional network can drive the transdifferentiation of β-cells into other islet cell types, such as α- or δ-cells. Modulating transcription factors like Pdx1, Ngn3, and MafA shows potential for reprogramming other cell types into insulin-producing β-like cells, thereby facilitating β-cell regeneration [101]. The information on key transcription factors involved in dedifferentiation (De), transdifferentiation (Tra), neogenesis (Neo), and proliferation (Pro) is summarized in Table 1.
The table is divided into seven columns: “Transcription Factors” lists the names of the transcription factors involved in β-cell dedifferentiation, transdifferentiation, neogenesis, or proliferation (De/Tra/Neo/Pro). “Full Name” provides the full names of these transcription factors. “Target Genes” describes the genes regulated by the transcription factors. “Impact on Genes” explains how each transcription factor affects the target genes (induction (I) or repression (R)). “De/Tra/Neo/Pro” indicates how the transcription factors impact β-cell plasticity and regeneration. “Impact on β-Cell Plasticity/Regeneration” describes whether the transcription factors have a positive (P) or negative (N) effect on β-cell plasticity/regeneration. “References” lists the sources used for each transcription factor’s explanation. Abbreviations: P: Positive, N: Negative, NM: Not Mentioned, I: induction, R: Repression.

5. Translational Control

The eukaryotic initiation factor 5A (eIF5A), crucial for β-cell identity and function, undergoes a post-translational modification by deoxyhypusine synthase (DHPS) to activate its translational activity. In mice lacking DHPS, there is a significant reduction in the proteins essential for β-cell identity, leading to rapid-onset diabetes during β-cell maturation. Further investigation revealed that eIF5A regulates the translation of key genes like Ins1, Glut2, Ucn3, and Chga during β-cell maturation, underscoring the pathway’s significance in β-cell function maintenance. Targeting mRNA translation in β-cells, particularly by modulating the eIF5A pathway, could present a therapeutic approach to correcting diabetes-related defects [189].

6. Epigenetic-Related Targets

6.1. Histon Modifications

Histone modifications act as epigenetic regulators that control the expression of key β-cell genes. The disruption of these histone marks contributes to β-cell dedifferentiation, while resetting them is crucial for transdifferentiation approaches aimed at generating functional β-like cells [190].

6.2. Histone Acetylation

Sirtuin 3 (Sirt3) is a major mitochondrial deacetylase that is synthesized in the cytoplasm and imported into the mitochondrial matrix. It plays a key role in mitochondrial homeostasis by regulating mitochondrial protein acetylation levels and responding to cellular energy status [191]. It inhibits the ATX-LPA pathway to preserve β-cell identity and function. The depletion of Sirt3 enhances autotaxin (ATX) levels, leading to increased lysophosphatidic acid (LPA) production, which induces β-cell dedifferentiation and impairs insulin secretion. Inhibiting ATX in vivo mitigates β-cell dysfunction in HFD-fed mice lacking Sirt3, suggesting a potential therapeutic strategy for managing diabetes-associated β-cell dysfunction [192].
Sirt3 depletion is also attributed to the reduced deacetylation of the FoxO1 gene and the subsequent decrease in its transcriptional activity. This suggests that Sirt3 downregulation contributes to β-cell dedifferentiation via FoxO1 [120].
CBP/p300, acting as transcriptional co-activators and histone acetyltransferases, are crucial for β-cell function. Treatment with A-485, a selective inhibitor of CBP/p300 HAT activity, impairs insulin secretion and downregulates β- and α-cell identity genes without inducing dedifferentiation. A-485 also affects genes related to glucose sensing but not glycolysis. In prediabetic db/db mice, decreased CBP/p300 levels impact β-cell function genes, possibly involving histone H3K27 deacetylation and transcription factors like Hnf1α and FoxO1. This highlights the critical role of CBP/p300 HAT in maintaining β-cell identity [193].

6.3. Histone Methylation

Histone H3 lysine 4 (H3K4) methylation is crucial for sustaining gene expression related to insulin production and glucose responsiveness, with deficient methylation leading to an altered epigenome profile. Moreover, in diabetic models, there is a shift favoring inactive genes, potentially contributing to β-cell dysfunction [194].
Utilizing GSK126 and Tazemetostat (Taz) inhibitors of histone methyltransferase EZH2, researchers induced a phenotypic shift towards a β-like cell identity in exocrine cells from T1D donors. This transition was facilitated by chromatin modifications, particularly H3K27me3 and H3K4me3, suggesting EZH2 to be pivotal regulator of β-cells’ regenerative potential. Reprogrammed pancreatic ductal cells demonstrated insulin production and secretion in response to physiological glucose levels ex vivo [195].

6.4. DNA Methylation

Proper DNA methylation patterns are required to maintain the mature β-cell identity. Disruption of these methylation dynamics contributes to β-cell dedifferentiation in diabetes [196].
DNA methylation plays a significant role in regulating tyrosine hydroxylase (TH), an enzyme known to suppress insulin secretion, during β-cell differentiation. The ablation of the de novo DNA methyltransferase Dnmt3a in embryonic progenitors increases TH+ β-cells, reducing insulin secretion. However, postnatal β-cell-specific Dnmt3a ablation does not have the same effect. This suggests that maintaining a lower proportion of TH+ β-cells is vital for proper insulin secretion and that embryonic DNA methylation patterns regulate β-cell heterogeneity. Chronic overnutrition can lead to a loss of TH promoter methylation, increasing TH+ β-cells and compromising β-cell function, linking embryonic epigenetic regulation to postnatal β-cell identity and diabetes risk [197].

6.5. Chromatin Remodeling

Chromatin remodeling events play a crucial role in facilitating β-cell plasticity by regulating the expression of lineage-specific transcription factors and epigenetic modifiers.
The ARID1A subunit of the SWI/SNF chromatin remodeling complex regulates β-cell regeneration, with its expression naturally decreasing during β-cell proliferation. ARID1A knockout mice show increased protection against diabetes, indicating its role in regeneration. The specific deletion of Arid1a in β-cells enhances regeneration via neuregulin–ERBB–NR4A signaling. Blocking ERBB or NR4A1 disrupts this regeneration, suggesting that mSWI/SNF complex activity limits β-cell regeneration in health and disease [198].
It has also been found that Pdx1 recruits Swi/Snf to regulate pancreas development and β-cell function by influencing gene expression and chromatin structure. The loss of Swi/Snf in β-cells leads to reduced insulin production and impaired β-cell maturation, highlighting its crucial role. The interaction between Pdx1 and Swi/Snf is vital for β-cell proliferation and the expression of insulin-related genes [199].
Chromodomain Helicase DNA-Binding Protein 4 (Chd4), a subunit of the nucleosome remodeling and deacetylase complex, is found to be crucial for maintaining β-cell identity and function. An inducible β-cell-specific Chd4 knockout mouse model showed that the loss of Chd4 led to glucose intolerance and impaired insulin secretion. Chd4-deficient β-cells exhibited an increased ratio of immature-to-mature insulin granules and elevated proinsulin levels. RNA sequencing revealed that Chd4 deficiency altered chromatin accessibility and the expressions of key β-cell genes like MafA, Glut2, Chga, and Chgb. Similar defects were observed in human β-cells with Chd44 knockdown, underscoring its importance in β-cell function. Additionally, it has been found that Chd4 is involved in the transcriptional activity of Pdx1 [200,201].

6.6. MicroRNAs in Pancreatic β-Cell Function and Identity

MicroRNAs (miRNAs) are critical for regulating many cellular functions, such as insulin signaling and glucose metabolism. Pancreatic β-cells depend on a network of miRNAs and their target mRNAs, which influence insulin production, secretion, cell growth, differentiation, survival, and apoptosis. Important miRNAs, including miR-7, miR-9, miR-375, miR-130, and miR-124, are abundant in β-cells and significantly impact their function. In diabetes, shifts in miRNA expression can lead to various complications such as β-cell dedifferentiation and transdifferentiation [202,203]. Table 2 provides detailed information on the impacts of various key miRNAs on β-cell dedifferentiation, transdifferentiation, neogenesis, and proliferation.
During the birth-to-weaning period, pancreatic β-cell growth and maturation are influenced by epigenetic programs activated by nutrients in human milk, including milk-derived exosomes (MEXs). These MEXs, containing miRNAs, drive β-cell proliferation, while reducing differentiation and mitochondrial function. Changes in MEX miRNAs during weaning may trigger β-cell differentiation, yet continuous exposure to bovine MEX miRNAs via cow’s milk could lead to β-cell dedifferentiation in adults. This suggests a potential link between prolonged bovine MEX exposure and an increased risk of diabetes [204].
Pancreatic β-cells undergo distinct metabolic changes during breastfeeding. Initially, they exhibit an immature state with high proliferation driven by mTORC1 activation and AMPK suppression. Upon weaning, β-cells transition to a mature state with reduced proliferation and altered mTORC1/AMPK activity, facilitating glucose-stimulated insulin secretion.
Table 2. miRNAs involved in β-cell dedifferentiation, transdifferentiation, neogenesis, or proliferation (De/Tra/Pro/Neo).
Table 2. miRNAs involved in β-cell dedifferentiation, transdifferentiation, neogenesis, or proliferation (De/Tra/Pro/Neo).
miRNAAffected GenesDe/Tra/Pro/NeoType of ImpactReferences
miRNA-148aPten, AmpkDeP[205]
microRNA-483Aldh1a3, Socs3DeN[206,207,208]
miR-29-aCdc42, Irs1, AKTDeP[209,210]
ProN
miR-23aSDF-1αTraN[211]
miR-24Ire1α, MafADeP[212]
miR-7Pdx1, Isl1NeoP[213,214]
miR-25/miR-92bNeurod1, Mcl1DeP[215]
miR-9Stxbp1De/TraP[216]
miR-96PAK1β-cell dysfunctionN[217]
miR-33a-5pAbca1DePositive and negative[218]
miR-302sNeuroD1 and Kat2bDeP[203]
miR-203Zbtb20DeP[219]
miR-802NeuroD1, Fzd5DeP[220]
miR-30dSocs3, Glp1r, Igf1r, MafA, Pdx1, Nkx6.1TraP[221]
miR-212/132Fbw7NeoP[222]
miR-690Sox9, Ngn3, Pdx1NeoN[223]
miR-223FoxO1, Sox6ProP[121]
miR-155Pdx1DeP[224]
miR-153SNAREs, MafA, NeuroD1, Bcl2, Ero1lb, Igf1r, Irs2DeP[225]
miR-320aMafFDeP[226]
miR-17-92 clusterPten, Cdkn1a, p57, Bcl2L11ProP[227]
miR-21Tgfb2, Fgfr3, Smad2DeP[228,229]
miR-184-3pCrtc1, Slc25A2De and lossP[230]
miR-199b-5pMlk3ProP[231]
miR-212-5pSirt2Insulin secretion and functionN[232]
miR-375Motn, Mapkap 1, Pdx1, Pdpk1, Notch2, Hnf1β, Cadm1, Pax6, CremDeN[233,234,235,236,237,238,239]
ProP
NeoP
miR-200Zeb1DeP[240]
miR-146aNumbDeN[241]
miR-195Mfn2DeP[242]
The table is divided into five columns: “miRNA” lists the names of the miRNAs involved in β-cell De/Tra/Neo/Pro. “Affected Genes” describes the genes regulated by these miRNAs. “De/Tra/Neo/Pro” indicates the specific processes of β-cell plasticity and regeneration influenced by the miRNAs. “Type of Impact” describes whether the miRNAs have a positive or negative effect on β-cell plasticity and regeneration. “References” lists the sources used for each miRNA’s explanation. Abbreviations: P: Positive, N: Negative.

6.7. Long Non-Coding RNAs (LncRNAs)

Long non-coding RNAs (LncRNAs) have become crucial regulators of gene expression and cellular activity in pancreatic β-cells, profoundly influencing their function, adaptability, and growth. Recent studies have revealed several LncRNAs that are vital for preserving β-cell identity, controlling β-cell mass, and facilitating β-cell proliferation and regeneration. These discoveries highlight the significance of LncRNAs in β-cell biology and their potential as targets for diabetes treatment. Table 3 provides a summary of these key LncRNAs and their roles in β-cell regulation.
The table is organized into five columns: “LncRNAs” lists the names of the lncRNAs involved in β-cell De/Tra/Neo/Pro. “Direct and Indirect Targets” describes the miRNAs and genes regulated by the lncRNAs. “De/Tra/Neo/Pro” specifies the particular processes of β-cell plasticity and regeneration affected by the LncRNAs. “Type of effect” indicates whether the LncRNAs have a positive or negative effect on β-cell plasticity and regeneration. “References” provides the sources used for the information. Abbreviations: P: Positive, N: Negative.

7. Reprogramming and Neogenesis of β-Cells

Evidence suggests that newly emerged cells following β-cell destruction differ from original β-cells, demonstrating bihormonal activity by simultaneously producing both somatostatin and insulin. This dual hormone production may contribute to the management of hyperglycemia. Their rapid appearance following β-cell destruction, regardless of age, indicates a promising avenue for further investigation. Moreover, research indicates that these cells likely originate from Somatostatin 1.1 (sst1.1) δ-cells with a β-cell pre-identity, offering a potential source for β-cell regeneration after β-cell destruction [262]. Using single-cell transcriptomics in zebrafish during diabetes induction and recovery, researchers identified two distinct somatostatin-producing δ-cells, denoted as δ1 and δ2. Additionally, a small subset of glucose-responsive islet cells displayed characteristics of both β- and δ1-cells. These hybrid cells emerged spontaneously and gained glucose responsiveness, crucial for insulin expression during recovery. Furthermore, the overexpression of Dickkopf-related protein 3 (DKK3), a member of the Dickkopf (DKK) family involved in Wnt signaling pathway regulation, enhanced the formation of these hybrid cells without harming β-cells [263].
The abundance of these δ-cells, as observed in T1D, could potentially result from α- to β- to δ-cell conversion [264,265]. Supporting this notion, it has been discovered that α-cells could also contribute to β-cell regeneration [264,266,267]. α-cells, especially those secreting glucagon, have the capacity to spontaneously transform into insulin-producing cells, thereby aiding in diabetes recovery, particularly post-puberty. A novel mechanism, partially governed by FoxO1, facilitates this transformation of somatostatin-producing δ-cells, particularly in juvenile individuals [266]. Additionally, a novel β-cell progenitor type displaying mesenchymal characteristics and expressing α-cell markers has been identified, potentially influencing β-cell regeneration in STZ-treated mice. These new β-like cells emerged within 48 h and, initially, were unresponsive to glucose. Insulin treatment normalized the glucose levels, preserving the neogenic β-like cells and gradually maturing the islet function. Intermediate cells expressing vimentin and MafB appeared within 16 h, serving as a major source of insulin-producing cells at 24 h [268].
In mice, α-cells and δ-cells can transform into insulin-producing β-cells after β-cell ablation, aiding in diabetes recovery, but whether this occurs in humans is unclear. This study demonstrates that non-β-cells from deceased human donors, including α-cells and γ-cells, can be reprogrammed to produce insulin using the transcription factors Pdx1 and MafA. Transplanted human α-cells reverse diabetes in mice and sustain insulin production for at least six months, retaining their original cell identity markers [269].
In addition to δ- and α-cells, scientists also discovered that ghrelin-producing ε-cells contribute to β-cell regeneration following significant β-cell loss. Augmenting ghrelin levels or expanding ε-cell populations supports this regeneration [270].
Beyond the conventional pancreatic cell types like α-, δ-, and ε-cells, ductal cells also play a role in contributing to the β-cell population, especially under diabetic conditions.
Investigations revealed that there are two distinct niches for β-cell neogenesis: “β duct cells” near ducts and “β vessel cells” near blood vessels. Single-cell transcriptomic analysis revealed five unique populations among newborn β-cells, shedding light on potential regulatory mechanisms, including the role of MafB-expressing cells in β-cell specification. Additionally, comparative analysis with human embryonic stem cell-derived β-like cells highlighted similarities in the transcriptional profiles, suggesting shared regulatory pathways in β-cell maturation between mice and humans [271].
It is important to note that the evidence suggests that β-cell transdifferentiation plays a larger role in regenerating β-cells following extreme conditions like partial pancreatectomy, pancreatic duct ligation, and β-cell ablation with STZ, mimicking T1D, compared to neogenesis [264]. Conversely, β-cell neogenesis is more prominent in regenerating β-cells during insulin resistance in T2D, where an increase in β-cell mass is needed to counter hyperglycemia [272].
The process of neogenesis from β duct cells involves the expression of the endocrine master transcription factor Ngn3. Lineage tracing and single-cell sequencing (scRNA-seq) have unveiled that duct-derived somatostatin-expressing cells undergo a transition into β-cells, signifying a physiological mechanism for β-cell maintenance beyond mere proliferation. Moreover, the study has noted the presence of bihormonal cells within ducts, suggesting a ductal origin for certain islet cells and the potential for differentiation into various endocrine cell types. This underscores the significance of ductal cells in pancreatic regeneration and β-cell neogenesis [273].
The inhibition of MAP kinase-interacting serine/threonine kinase 2 (MNK2) with the small molecule CID661578 has been identified as a potent stimulator of β-cell regeneration. This small molecule facilitated β-cell neogenesis from ductal cells across various models, including zebrafish and human pancreatic organoids, by enhancing protein synthesis via modulation of the eIF4E-eIF4G interaction. MNK2 inhibition by CID661578 augmented eIF4E phosphorylation, promoting hypertranslation and β-cell regeneration, highlighting a potential therapeutic avenue for diabetes management [274].

8. Mitigation of β-Cell Dedifferentiation and Transdifferentiation

β-cell dedifferentiation and transdifferentiation can be reversed through various treatments, including dietary supplements, natural products, dietary interventions, hormones, and antidiabetic medications. These approaches offer promising strategies to restore β-cell function and mitigate diabetes.

8.1. Dietary Supplements

Glutathione is a crucial antioxidant synthesized within cells, playing a vital role in maintaining cellular health and defending against oxidative stress. Prolonged exposure of rats to long-term oscillating glucose (LOsG) triggers β-cell dysfunction through ROS stress. This stress disrupts FoxO1 signaling, leading to an elevation in thioredoxin interacting protein (TXNIP) levels and a reduction in insulin and SOD-2 expression. Consequently, β-cell dedifferentiation and dysfunction occur. However, the timely administration of glutathione effectively prevents LOsG-induced β-cell failure [275].
Vitamin D3 therapy may offer a promising avenue for attenuating β-cell dedifferentiation. Research indicates that the administration of vitamin D3 and its analogues to the high-glucose-induced mouse insulinoma cell line (MIN6) results in the upregulation of the vitamin D3 receptor. This upregulation enhances β-cell function and maintenance by increasing insulin release and upregulating key transcription factors such as Pdx1 and MafA. Consequently, there is an observed augmentation in the expressions of Ins-1 and Ins-2 genes [276].
Omega-3 polyunsaturated fatty acids (PUFAs) are known to have anti-hyperglycemic effects. Transgenic mice (mfat-1) that overexpress an enzyme converting omega-6 to omega-3 polyunsaturated fatty acids (PUFAs) were found to have significantly higher levels of eicosapentaenoic acid (EPA) in pancreatic tissues compared to wild-type mice. After STZ treatment, these transgenic mice did not develop diabetes and maintained normal insulin and blood glucose levels. The EPA metabolites from the CYP 450 pathway promoted insulin expression and increased β-cell markers in α-cells. This suggests that omega-3 PUFAs and their metabolites can promote α-to-β-cell transdifferentiation [277].
Folinic acid treatment, or the overexpression of folate receptor 1 (folr1), has been shown to promote β-cell differentiation in a zebrafish model of β-cell loss. This effect extends to cultures of neonatal pig islets, suggesting its potential applicability in mammals. Notably, the source of increased β-cell differentiation in both zebrafish and pig islets was traced back to ductal cells. Additionally, comparative metabolomic analysis suggested that the regenerative effect of folinic acid might be linked to alterations in the pyrimidine, carnitine, and serine pathways [278].
Treatment with nicotinamide (NA), also known as niacinamide, a form of vitamin B3, significantly increased the number of insulin/PCNA, insulin/glucagon, and insulin/somatostatin double-positive cells compared to untreated groups. Notably, NA-treated rats showed enhanced insulin and Pdx1-positive cells, located within small clusters or scattered throughout the exocrine tissue and around ducts. Additionally, NA treatment increased Notch1 expression while decreasing caspase-3 and TUNEL-positive β-cell numbers, suggesting that NA induces β-cell regeneration through ductal progenitor cell differentiation, acinar cell transdifferentiation, and the activation of Notch signaling [279].
Cathelicidin-based peptides like LLKKK18, a class of peptides derived from the cathelicidin family of antimicrobial peptides, have shown promise for T1D therapy. When delivered via poly (lactic-co-glycolic acid) (PLGA) nanoparticles (NPs), these peptides enhance β-cell function and neogenesis. In vitro studies have demonstrated glucose-stimulated insulin release and strong NP association with β-cells. In diabetic rats, these NPs effectively reduced hyperglycemia, improved pancreatic insulin content and glucose tolerance, and notably increased β-cell mass [280].

8.2. Dietary Interventions and β-Cell Function, Dedifferentiation, and Transdifferentiation

Intermittent fasting and calorie restriction have been found to regulate not only blood glucose and insulin levels, triglyceride and cholesterol levels, and insulin resistance but also insulin secretion, GSIS, and the modulation of β-cell transcription factors such as Pdx1, Nkx6.1, and MafA in obesity-induced diabetes [281].
Intermittent protein restriction (IPR) has also been found to significantly reduce hyperglycemia, protect pancreatic islets, and increase β-cell proliferation in diabetic Zucker fatty rats. IPR has been shown to reverse β-cell dedifferentiation, as evidenced by decreased expressions of dedifferentiation markers like Cd81 and stress-related genes, while increasing markers of β-cell function such as islet amyloid polypeptide and chromogranin. Furthermore, IPR altered the expression of transcription factors, mitigating diabetes-associated changes in D-box binding PAR bZIP transcription factor (Dbp) and immediate-early response genes [282].
The ketogenic diet has also been shown to improve fasting blood glucose and insulin levels, β-cell mass, and identity by restoring the β/α-cell ratio, rearranging β-cells in the pancreas, and modulating β-cell transcription factors in diabetic mice [283].
A Chinese medical nutrition therapy incorporates medicinal herbs, whole grains, and intermittent energy restriction. It has demonstrated effectiveness in alleviating T2D. This therapy reduces fasting blood glucose levels, improves insulin sensitivity, enhances insulin production and secretion, modulates the gut microbiota, reduces pancreatic macrophage infiltration, and restores β-cell identity and morphology [284].
Increasing dietary fiber intake has been demonstrated to alleviate the progression of T2D and β-cell pathophysiological alterations, underscoring the therapeutic potential of dietary interventions. This is supported by research indicating that dietary fiber intake can safeguard and enhance insulin secretion through its influence on the intestinal microbiome. Specifically, it encourages the proliferation of short-chain fatty acid (SCFA)-producing strains and enhances the production of GLP-1, a hormone that stimulates insulin secretion [285].

8.3. Physical Activity and β-Cell Plasticity

Physical activity has been demonstrated to enhance pancreatic β-cell function and may play a crucial role in preventing or reversing β-cell dedifferentiation in the context of T2D. Exercise promotes β-cell function in T2D patients by increasing β-cell mass, improving glucose sensing, enhancing insulin secretion, and augmenting insulin content [285,286]. The protective effects of exercise on β-cells are mediated through multiple mechanisms: 1. Reducing metabolic stress: Physical activity facilitates increased glucose uptake in peripheral tissues, thereby lowering blood glucose levels and reducing the metabolic load on β-cells [287]; 2. Alleviating ER and oxidative stress: Exercise may attenuate ER stress and oxidative stress in β-cells, which are critical contributors to β-cell dysfunction; and 3. Modulating inflammation: Physical activity has the potential to modulate the immune environment of the pancreas, thereby reducing inflammatory processes that contribute to β-cell damage [286]. IL-6, which is produced by muscle cells during contraction, is associated with elevated levels of IL-1RA, IL-10, and GLP-1, which regulate abdominal fat lipolysis and promote β-cell protection, growth, and mass [288,289]. Additionally, certain myokines like angiopoietin and osteoprotegerin, specifically produced by triceps, exhibit anti-inflammatory effects [290]. Exercise can also increase levels of adiponectin, an adipokine synthesized by adipose tissue with anti-inflammatory properties [291]. Adiponectin has been shown to induce a shift of M1 (proinflammatory) macrophages to the M2 (anti-inflammatory) phenotype by reducing proinflammatory cytokines, including TNFα [292]. Notably, inflammatory responses by macrophages resident in the pancreas play a critical role in the fate of β-cells [3].
Besides IL-6, exercise also positively influences brain-derived neurotrophic factor (BDNF) [293] and C-X3-C motif chemokine ligand 1 (CX3CL1) [294]. Similar to GLP-1, these factors have beneficial effects on insulin secretion, glucose-stimulated insulin secretion (GSIS), and β-cell function [293,294].
Four exerkines elevated during exercise known to enhance β-cell proliferation, function, and maintenance include the following: irisin, an exerkine released by muscle tissue; follistatin, which is induced by exercise and secreted by the liver and muscles; osteocalcin, which is produced by bone marrow; and FGF21, which is produced snd decreted by hepatic tissues [295,296,297,298,299,300].
β-cell dedifferentiation and exercise: Although direct evidence linking exercise to the prevention or reversal of β-cell dedifferentiation is limited, various studies suggest that exercise may support the maintenance of β-cell identity and function. In animal models, voluntary running has been shown to prevent β-cell failure in susceptible islets [301]. Apelin, an exerkine that is involved in various physiological processes and is produced and secreted by various tissues, including adipose tissue, the heart, and skeletal muscle, has been shown to reverse β-cell dedifferentiation [302]. Exercise is also linked to the activation of the GABAergic system [303]. There is evidence suggesting that GABA and the activation of the GABAergic system might trigger the conversion of α-cells into β-cells, presenting a potential treatment avenue for Type 1 Diabetes (T1D) [304,305].
Combined effects of diet and exercise: The synergistic effects of dietary intervention and physical exercise on β-cell health are notable. Both strategies can alleviate metabolic stress on β-cells, and their combination, particularly through diet-induced weight loss, may enhance the benefits of each intervention. This dual approach has the potential to mitigate the adverse effects of T2D and β-cell identity [286].
Muscle mass reduction can significantly impact glucose metabolism and increase the risk of diabetes. Since approximately 80% of glucose clearance occurs in muscle tissue under normal glycemic conditions, any decrease in muscle mass or quality can adversely affect blood glucose regulation, leading to hyperglycemia, insulin resistance, metabolic stress, and impaired β-cell function and maintenance. Consequently, a correlation has been observed between sarcopenia (age-related loss of muscle mass and function) and the incidence of T2D. To address this issue, dietary supplements containing specific amino acids such as HMB (β-hydroxy β-methylbutyrate), leucine, glutamine, and arginine have been proposed as potential interventions. These supplements may help prevent sarcopenia and, consequently, reduce the risk of developing insulin resistance and T2D. By maintaining muscle mass and function, these amino acids could support better glucose regulation and metabolic health, particularly in aging populations or those at risk for muscle loss [306].

8.4. Antidiabetic Medications

8.4.1. Glucagon-like Peptide-1 Receptor Agonist (GLP-1 RA)

Liraglutide, classified as a GLP-1 RA, stimulates GSIS. Sitagliptin, categorized as a dipeptidyl peptidase-4 inhibitor (DPP-4 inhibitor), elevates endogenous GLP-1 and glucose-dependent insulinotropic peptide (GIP) levels. These actions enhance insulin secretion and inhibit glucagon release. Both liraglutide and sitagliptin treatments have demonstrated the ability to reverse β-cell dedifferentiation in STZ-, high-fat-feeding- or HC-induced mice. Moreover, liraglutide administration has been observed to attenuate β-cell transdifferentiation into α-cells [307].

8.4.2. Sodium–Glucose Co-Transporter 2 (SGLT2) Inhibitors

Dapagliflozin, a medication employed in the treatment of T2D, falls within the category of sodium–glucose co-transporter 2 (SGLT2) inhibitors. Emerging evidence suggests that dapagliflozin administration might mitigate transdifferentiation in diabetic mouse models. Interestingly, this effect appears to occur through a mechanism independent of reducing ambient blood glucose levels [308].
Luseogliflozin, another SGLT2 inhibitor, was tested on the pancreatic β-cell mass in db/db mice of various ages. The 6-week-old mice treated with luseogliflozin showed a significant increase in β-cell mass and upregulation of key genes compared to controls. While β-cell mass declined with age in the control group, luseogliflozin preserved the β-cell mass, especially in the early stages of diabetes. Immunohistochemistry and gene expression analyses supported these findings [309].

8.4.3. Dipeptidyl Peptidase-4 Inhibitor (DPP-4 Inhibitor)

DPP-4 inhibitor Sitagliptin, known for its β-cell regenerative and immunoregulatory properties, and melatonin, a hormone primarily produced by the pineal gland that regulates sleep–wake cycles and possesses antioxidant, anti-inflammatory, and potential immunomodulatory properties, were studied for their combined effect on β-cell regeneration under glucotoxic stress in diabetic mouse models and humanized islet transplant models. The combination therapy demonstrated additive effects in promoting β-cell regeneration, reducing apoptosis, and improving glucose tolerance. In young diabetic mice, monotherapies induced β-cell transdifferentiation, while in older mice, they promoted β-cell proliferation and reduced apoptosis. Overall, the combination therapy proved superior, suggesting significant potential for future diabetes treatment [310].

8.5. Hormones

Hormones such as insulin, glucagon, and somatostatin play a crucial role in regulating β-cell dedifferentiation and transdifferentiation processes [190,311].
Glucagon has been identified as a key factor triggering the dedifferentiation of β-cells, with FoxO1 playing a partial role in this process. This interaction sheds light on the intriguing interplay between α- and β-cells. Blocking the glucagon receptor with a monoclonal antibody (mAb) has been found to reverse the effects induced by glucagon. Consequently, this intervention leads to improvements in β-cell size and function, while also preventing dedifferentiation in mice with T2D. Conversely, the inhibition of FoxO1 mimics the effects of glucagon, whereas its overexpression counteracts dedifferentiation [123]
Gastrin, a hormone primarily secreted by G cells in the stomach and duodenum, plays a key role in stimulating gastric acid secretion to aid in digestion. Recent studies have revealed that gastrin administration can induce the transformation and reprogramming of regenerative ductal cells in rats with 95% pancreatectomy, leading to the enhanced formation and expansion of β-cells. This process appears to involve the downregulation of keratin 20 and β-catenin, representative of ductal dedifferentiation, and the upregulation of Ngn 3 and Nkx6-1, indicating an endocrine progenitor phenotype [312].
The long-acting xenin analogue xenin-25[Lys13 PAL], a modified form of xenin-25 and peptide hormone that influences gastrointestinal function and glucose metabolism, has antidiabetic effects, promoting insulin secretion and improving pancreatic islet architecture. In high-fat-fed and STZ-induced insulin-deficient mice, xenin-25[Lys13 PAL] treatment reversed STZ-induced body weight loss, increased insulin levels, and reduced blood glucose. It also decreased pancreatic α-cell areas, reduced β-cell apoptosis, and promoted β-cell growth. Additionally, xenin-25[Lys13 PAL] decreased the transition of insulin-positive to glucagon-positive cells, suggesting that it helps maintain β-cell identity and function [313].
Administering T3, a thyroid hormone variant, efficiently counteracts the harmful effects of β-cell dedifferentiation and transdifferentiation. This corrective action not only boosts insulin secretion and restores vital markers but also safeguards pancreatic islet structure by halting β-cell polyploidization. T3 treatment has been shown to diminish the transformation of β-cells into α-cells, thus upholding insulin levels. Conversely, blocking T3 exacerbates transdifferentiation [314].
Angiotensin (1–7) (Ang (1–7)), a peptide hormone derived from the cleavage of Angiotensin II by enzymes such as ACE2, has demonstrated potential in mitigating high-glucose-induced apoptosis and dedifferentiation of pancreatic β-cells through its interaction with MAS receptors. In the presence of high glucose levels, the β-cell morphology deteriorates, insulin secretion diminishes, and markers associated with mature β-cells (Pdx1 and MafA) decrease, while those linked to endocrine progenitor cells (Oct4 and Nanog) increase. However, treatment with Ang (1–7) significantly reverses these detrimental changes. This reversal is attributed to the specific interaction between Ang (1–7) and its MAS receptors, which subsequently impacts the Wnt/β-catenin/FoxO1 pathway, ultimately safeguarding the β-cell function and identity against the adverse effects of high glucose [315].
Adipsin, also referred to as complement factor D, is a serine protease enzyme encoded by the ADIPOQ gene. Its pivotal role lies within the alternative complement pathway, an integral part of the innate immune system. Predominantly synthesized by adipocytes, adipsin actively participates in adipose tissue metabolism, exerting influence over energy regulation and adipocyte differentiation. Additionally, adipsin contributes to the generation of complement component C3a, which exhibits insulin secretagogue activity. Emerging evidence suggests that elevated levels of blood adipsin/C3a may uphold β-cell identity amidst diabetic conditions [316].

8.6. Natural Products

Sangzhi alkaloids (SZ-A), extracted from Morus alba L. (mulberry twig), have shown promise in mitigating hyperglycemia and glucose intolerance in diabetic mice. Moreover, they have been found to restore β-cell identity and function by modulating the levels of β-cell-specific transcription factors under diabetic conditions [317].
Eugenol (4-allyl-2-methoxyphenol) is a naturally derived compound present in clove, basil, cinnamon, and nutmeg. When administered to Ins-1 β-cells exposed to high glucose and lipid levels, eugenol demonstrated the ability to partially restore the expressions of certain β-cell-specific genes, including Glut2, NeuroD1, and FoxO1 [318].
Piperine, a compound in black pepper, offers numerous health benefits, including anti-inflammatory, antioxidant, anti-obesity, and antidiabetic effects. In a mouse model of obesity, a 10-week piperine treatment improved glycolipid metabolism, insulin resistance, and metabolic inflammation. Piperine enhanced glucose metabolism, reduced inflammation, and prevented β-cell dedifferentiation while also decreasing M1-like macrophage polarization in islets and visceral adipose tissue [319].
Psilocybin, a natural compound from Psilocybe mushrooms, acts as an agonist for serotonin receptors. It has been shown to reduce β-cell loss induced by high-glucose–high-lipid (HG-HL) conditions by mitigating apoptosis, likely through modulating TXNIP and STAT pathways. Additionally, psilocybin affects key genes related to β-cell dedifferentiation, such as Pou5f1 and Nanog, indicating its potential therapeutic role in protecting against β-cell dysfunction and dedifferentiation in T2D [320].
Phytocannabinoids are a group of secondary metabolites produced by Cannabis sativa. We recently studied the impacts of various phytocannabinoids on β-cell survival, function, and dedifferentiation in high-glucose–high-lipid-induced INS-1 β-cells. Our results indicate that certain phytocannabinoids can restore an impaired β-cell identity by reactivating β-cell-enriched transcription factors [321].
Gymnemic acid (GA) is a compound found in the leaves of Gymnema sylvestre, a plant native to India and Africa. It is known for its unique property of temporarily suppressing the taste of sweetness, making sweet foods taste less appealing. GA treatment demonstrated promising outcomes by maintaining normoglycemia in diabetic rats through enhanced insulin secretion via the upregulation of β-cell regeneration markers such as Pdx1, Neurognin 3, MafA, and NeuroD1. Additionally, GA facilitated proliferation by activating the PI3K/AKT pathway and promoting cell adhesion through the increased expression of ECAD and β-catenin, thereby preserving islet architecture [322].
Treatment with magainin-AM2, an analog of magainins, which are antimicrobial peptides originally discovered in the skin of the African clawed frog, Xenopus laevis, and growth hormone improved glucose tolerance and fasting blood glucose levels post-STZ treatment and increased the total cell counts and insulin+ and glucagon+ cells per islet. The expressions of key factors for α-to-β-like cell conversion and cell proliferation/differentiation were significantly upregulated, suggesting a potential for the pharmaceutical induction of α-cell production and their transdifferentiation to functional β-like cells, with magainin-AM2 showing the greatest efficacy [323].
The administration of a Fufang-Zhenzhu-Tiaozhi capsule (FTZ), a traditional Chinese medicine prescription commonly used in clinical practice, increased insulin levels, reduced glucose levels, and promoted β-cell regeneration in T1DM mice. Additionally, FTZ inhibited inflammatory cell invasion and islet cell apoptosis, preserving the quantity and quality of β-cells. The promotion of β-cell regeneration by FTZ was associated with the upregulation of key transcription factors (Pdx1, MafA, and Ngn3), suggesting FTZ as a potential therapeutic drug for T1DM [324].

8.7. The Role of the Gut Microbiome in β-Cell Plasticity

The gut microbiome appears to play an important role in shaping β-cell biology through bacterial metabolites, proteins, extracellular vesicles, and immunomodulatory effects [325].
Short-chain fatty acids (SCFAs) produced by gut bacteria help regulate metabolism. SCFAs such as sodium acetate, sodium propionate, and sodium butyrate (SB) did not immediately affect insulin release from pancreatic islets. However, prolonged exposure to SCFAs improved the function of β-cells. An analysis of RNA levels showed that SB-treated islets had lower expression levels of genes related to insulin secretion and β-cell identity, such as Pdx1 and MafA. SB increased oxygen consumption but reduced glucose-stimulated oxygen consumption in islets, with no changes in genes related to glycolysis or the tricarboxylic acid cycle. Additionally, SB lowered the expression of Kcnj11, which encodes a channel involved in regulating insulin release, increased calcium levels, and promoted insulin gene expression by affecting the histone modification H3K18bu on its promoter. This suggests that while SB improves islet function, it also alters the expressions of genes related to β-cell identity [326].
Additionally, butyrate demonstrates anti-inflammatory properties, preserving the β-cell function against IL-1β-induced dysfunction. Butyrate exerts these effects through the suppression of IL-1β-induced inflammatory genes and nitric oxide production. Notably, butyrate inhibits NF-κB activity via histone deacetylase inhibition without affecting NF-κB nuclear translocation. These findings highlight butyrate’s potential in mitigating β-cell dysfunction through NF-κB pathway modulation, offering insights into T2D prevention and treatment strategies [327].
An altered gut microbiota is linked to T1D, influencing SCFAs and glucose homeostasis. A dietary supplement called HAMSAB, designed to increase serum acetate and butyrate, improved glycemic control in NOD mice and patients with established T1D. Single-cell RNA sequencing on islet cells from NOD mice fed a HAMSAB or control diet revealed that HAMSAB reduced immune-cell infiltration in the pancreas, preserved β-cell function and identity, and decreased cellular stress. This suggests that SCFAs have potential anti-inflammatory effects and that HAMSAB may be a promising approach to managing T1D [328].

8.8. Other Interventions

Following bariatric surgery, rats undergoing Roux-en-Y gastric bypass (RYGB) and sleeve gastrectomy exhibited significant improvements in metabolic parameters and an increase in pancreatic β-cell proportion compared to Sham-operated rats. Both the RYGB and Sleeve groups demonstrated a decrease in the α-to-β-cell ratio. The study identified Gcg+/Ppy+ and Ins+/Gcg+/Ppy+ multiple hormone-expressing cells post-surgery, suggesting potential transdifferentiation events. Notably, the discovery of Ins+/Gcg+/Ppy+ cells, displaying features of both α- and β-cells, implies a role for transdifferentiation in enhancing β-cell function following bariatric surgery [329].

9. Advancements in Stem Cell-Based Therapies for Diabetes Mellitus

Recent studies have detailed the generation of functional β-cells from both embryonic and induced pluripotent stem cells. Adipose-derived mesenchymal stem cells (Ad-MSCs) are potential candidates for generating insulin-producing cells (IPCs), irrespective of donor age, gender, or health status. These cells demonstrated insulin secretion in response to glucose and dysfunction with palmitate exposure. Gene expression analysis showed over 5000 differentially regulated genes, aligning closely with native β-cells. Pathway analysis confirmed relevance to stem cell differentiation and pancreatic development, indicating that adipose-derived β-cells may be a promising clinical therapy for diabetes [330].
Research indicates that manipulating the FoxO1-1 gene during the final stages of differentiation may enhance IPC generation from Ad-MSCs. While FoxO1 is essential for early β-cell development, its overexpression in mature β-cells can cause glucose intolerance and hyperglycemia. Similarly, in Ad-MSCs, FoxO1 supports early differentiation, but reducing its expression at later stages results in more mature and functional IPCs [331].
In addition to Ad-MSCs, bone marrow-derived mesenchymal stem cells (bm-MSCs) can potentially be transdifferentiated into islet-like cells for treating T1DM. In a research study, this process was induced using high glucose, nicotinamide, β-mercaptoethanol, betacellulin, and IGF-1, and transdifferentiation was confirmed through glucose challenge assays and gene expression analysis. The transdifferentiated cells were then microencapsulated with 1% alginate and cultured in a fluidized-bed bioreactor to ensure stability and proliferation. After transplantation into the omentum of STZ-induced diabetic Wistar rats, blood glucose levels significantly decreased within a week, and the treated rats’ weight, insulin, and C-peptide levels returned to normal [332]. Moreover, there is evidence suggesting that exosomes derived from bm-MSCs, known as bm-MSC-derived exosomes (bmMDEs), could alleviate β-cell dedifferentiation and enhance their function in type II diabetic rats as well as primary islets exposed to high glucose levels. This therapeutic effect is facilitated by miR-146a, which acts on the NUMB/β-catenin signaling pathway [241].
A protocol for the transformation of human fibroblasts into endodermal cells using non-integrative episomal reprogramming factors, growth factors, and chemical compounds has been demonstrated. The differentiated cells undergo a progression from definitive endodermal progenitors to posterior foregut-like progenitors, then to pancreatic endodermal progenitors, with significant expansion potential. Through screening, specific compounds were identified that enhance the differentiation of pancreatic endodermal progenitors into functional β-like cells in laboratory settings. When transplanted, these β-like cells exhibit GSIS in animal models, offering protection against chemically induced diabetes [333].
Human umbilical cord mesenchymal stem cells (hUC-MSCs) have shown promising potential in reverting β-cell dedifferentiation when cocultured with purified human T2D mellitus (hT2DM) islets. This reversal is orchestrated through a targeted interaction facilitated by the secretion of interleukin-1 receptor antagonist (IL-1Ra) from hUC-MSCs. This mechanism plays a pivotal role in modulating the overall inflammatory responses within diabetic islets, highlighting the therapeutic significance of hUC-MSC-based interventions in diabetes management [334].
MSC secretomes, which are collections of molecules including proteins, cytokines, and other factors secreted by MSCs, show higher levels of total protein and IL-4 when derived from 3D cultures compared to 2D cultures. In vivo, these secretomes induce regulatory T cells, modulate cytokine release, and increase β-cell regeneration, particularly those from 3D cultures. This demonstrates the dual therapeutic potential of MSC secretomes in T1D, promoting both immunomodulation and β-cell regeneration. Culturing MSCs in 3D scaffolds enhances these effects, suggesting a promising approach for T1D therapy [335].
hTERT-MSC (human telomerase reverse transcriptase MSC) transplantation, both intrapancreatically (IPR) and intravenously (IVR), in pancreatectomized mice, leads to the increased proliferation of pancreatic β-cells. IPR demonstrates moderate advantages over IVR. These cells activate pancreatic progenitor cell characteristics through the AKT/Pdx1/FoxO1 signaling pathway and modulate growth factors and proinflammatory cytokines [119].

10. β-Cell Proliferation and Regeneration

β-cell regeneration and proliferation are essential for diabetes treatment, restoring insulin production, and managing blood glucose levels. Enhancing these processes can potentially reverse diabetes, reducing the need for external insulin therapy. Strategies include stimulating endogenous β-cell proliferation, neogenesis, and transdifferentiation using small molecules and transcription factors, as well as deriving β-cells from pluripotent stem cells [336,337].

10.1. The Suppression of Dual-Specificity Tyrosine-Regulated Kinase 1A (DYRK1A)

The DREAM complex plays a pivotal role in maintaining quiescence in human β-cells. Comprising transcriptionally repressive proteins, this complex forms in response to DYRK1A kinase activity, enforcing cellular quiescence. When DYRK1A activity is inhibited, DREAM subunits transition into the pro-proliferative MMB complex. Small molecule DYRK1A inhibitors stimulate replication in human β-cells by shifting the DREAM complex from its repressive state to the proliferative MMB configuration [338].
Small-scale suppression of DYRK1A holds promise for pharmaceutical intervention in β-cell regeneration for diabetes, addressing a significant therapeutic gap. However, due to DYRK1A’s involvement in critical signaling pathways, it is essential to maintain its levels similar to those found in healthy individuals. In silico tests have identified several plant-based compounds as potential ligands for β-cell regeneration. These compounds include 3-[6-(3-methyl-but-2-enyl)-1H-indolyl]-6-(3-methyl-but-2-enyl)-1H-indole, Lanceolatin B, Lysicamine, Pratorinine, Pratorimine, Lanceolatin A, Lanuginosine, Hippacine, (-)-Semiglabrin, Aegyptinone B, 3′-Prenylnaringenin, and 8-C-p-hydroxybenzylluteolin. Computational analyses suggest these compounds have good intestinal absorption and favorable drug-like properties, making them promising candidates for new medications, pending further in vivo validation [12].
Various synthetic small molecules have shown potential for promoting β-cell regeneration by inhibiting DYRK1A activity. AC27, a compound with superior selectivity compared to another DYRK1A inhibitor, harmine, not only inhibits DYRK1A but also reduces its cellular levels, resulting in increased β-cell proliferation and enhanced insulin and C-peptide secretion in response to glucose. These effects were stable over a 12-day period and persisted after the inhibitor was withdrawn. AC27’s benefits were validated in insulinoma cell lines, iPSC-derived β-cell organoids, and isolated mouse pancreatic islets, with its effects further enhanced by TGF-β inhibition [339].
Gua et al. conducted a chemical screen in zebrafish to identify small molecules that increase the number of incretin-expressing cells. One of the identified compounds, AZ Dyrk1B 33, was found to be a potent inhibitor of DYRKs and effectively boosted incretin+ cell populations in zebrafish. Moreover, it demonstrated improved glucose regulation in mouse models. The research suggests that AZ Dyrk1B 33 likely influences enteroendocrine cell differentiation through NFAT signaling pathways. Unlike the DYRK1A inhibitors known to enhance β-cell proliferation, AZ Dyrk1B 33 did not affect β-cell mass in this study, highlighting its specific role in enteroendocrine cells regulated by DYRK1B [340].
While DYRK1A inhibitors such as AC27 hold potential for enhancing β-cell proliferation and function by targeting β-cell cycle pathways, significant obstacles persist. These include achieving β-cell specificity, refining evaluation techniques, and overcoming the low proliferation rates of human β-cells. To address these issues, structural modifications are needed to improve targeting and minimize off-target effects. Furthermore, integrating DYRK1A inhibitors with other antidiabetic medications could enhance their therapeutic efficacy [340,341].
Common DYRK1A inhibitors can induce β-cell proliferation, but with low rates and limited specificity. It has been shown that combining any GLP1R agonist with any DYRK1A inhibitor results in a significant synergistic increase in human β-cell replication (5 to 6%), leading to an actual increase in human β-cell count. This combination did not cause β-cell dedifferentiation and was effective for both normal human β-cells and those derived from individuals with T2D [342].
Molecular docking analysis revealed diosmin, a flavonoid compound found in various plants such as Zanthoxylum chalybeum Engl., as a potential inhibitor of DYRK1A. In vitro experiments showed that the extract promoted cell viability and proliferation, particularly under conditions of palmitate exposure. However, in vivo results indicated only mild β-cell regenerative potential, suggesting further investigation is needed to understand the molecular interactions underlying these effects, particularly regarding diosmin’s role as a DYRK1A inhibitor [343].

10.2. Glucagon Receptor Antagonism in β-Cell Regeneration

Research indicates that the glucagon receptor antibody (anti-GCGR) has significant potential for promoting β-cell regeneration in diabetes models. Anti-GCGR treatment effectively reverses hyperglycemia, elevates plasma insulin levels, and restores β-cell mass through both β-cell proliferation and α-to-β-cell transdifferentiation [13,14,15,16]. Using lineage-tracing techniques, researchers found that GCGR mAb treatment causes α-cells to regress to progenitor states and reactivates Ngn3 progenitors, promoting their transdifferentiation into β-cells. This treatment also upregulates the genes associated with β-cell regeneration and improves insulin secretion in primary mouse islets [14].
Treatment using REMD 2.59, a competitive human GCGR antagonist, lowered blood glucose levels while elevating the plasma levels of glucagon and somatostatin. Notably, the administration of a GCGR monoclonal antibody substantially increased α-cell and δ-cell populations in both normal and T1D mice. This increase in δ-cell mass was linked to enhanced proliferation and neogenesis originating from pancreatic ducts. Remarkably, the observed δ-to-β-cell transdifferentiation in T1D mice post-treatment suggests a promising avenue for restoring β-cell numbers [344].
Additionally, it has been found that REMD 2.59 induces α-cell regression to progenitors, facilitating β-cell neogenesis and upregulating regeneration-related genes in mouse islets. This highlights the GCGR mAb’s potential for β-cell regeneration in T2D, supporting α-cell to β-cell conversion and progenitor-driven β-cell formation [345].
GCGR mAb treatment in PANIC-ATTAC mice, which triggers caspase-8–mediated apoptosis specifically within the pancreatic β-cell, stimulates α-cell to β-cell conversion, leading to sustained improvements in glycemic control and effectively curing diabetes in this model. While α-cell hyperplasia may pose challenges in T2D, in T1D, it offers a potential source for generating new insulin-producing cells [346].
FGF21 has been shown to play a crucial role in the regenerative effects of GCGR antagonism on β-cells. The levels of FGF21 increase in both the plasma and liver in response to GCGR antagonism. The inhibition of FGF21 activity diminishes the upregulation of β-cell identity genes induced by the GCGR mAb. Furthermore, the increases in the β-cell number and proliferation driven by the GCGR mAb are less pronounced in mice lacking FGF21 or hepatic FGF21 [16].
Interestingly, anti-GCGR-induced α-cell hyperplasia can occur independently of β-cell regeneration. Furthermore, this therapy has shown efficacy in non-human primates, and when combined with anti-CD3, it significantly enhances β-cell mass and reverses diabetes in T1D models [13].
GCGR antagonism also stimulates the secretion of glucagon and glucagon-like peptide 1 (GLP-1) [347,348], which promotes β-cell regeneration. Blocking GLP-1 receptor (GLP-1R) signaling reduces these regenerative effects, suggesting that GLP-1R activation by glucagon is crucial for the β-cell regenerative effects induced by the GCGR mAb [15].
GLP1 induces insulin–glucagon-positive cells in rat pancreatic islets, potentially indicating α- to β-cell transdifferentiation. Molecular analyses suggest that GLP1 promotes β-cell function through the PI3K/AKT/FOXO1 pathway, enhancing Pdx1 and MafA expressions while reducing MafB, suggesting a mechanism for α-cell inhibition and β-cell promotion by GLP1 [349].
The effect of Liraglutide and the GCGR mAb in promoting pancreatic β-cell neogenesis in T1D mice has been examined. Liraglutide treatment expanded β-cell mass through self-replication, differentiation, and transdifferentiation from α-cells, while GCGR mAb treatment significantly increased β-cell mass. However, the combination of both drugs did not further enhance the β-cell area but reduced plasma glucagon levels and increased the proportion of β-cells to α-cells [350].
Dapagliflozin has been found to enhance the expression of pancreatic endocrine progenitor and β-cell markers, including Pdx1, in cultured primary rodent islets and αTC1.9 cells under hyperglycemic conditions. This regenerative effect is partially mediated by GLP-1 secreted from α-cells [351].
The stimulatory impact of dapagliflozin on GLP-1 secretion is attributed to the reshaping of the gut microbiota and the modulation of metabolites involved in tryptophan metabolism, particularly l-tryptophan, which augments GLP-1 secretion and insulin levels. These beneficial effects are mediated by gut microbiota and tryptophan metabolism and are attenuated by GLP-1 receptor antagonism or pancreatic Glp1r knockout [352].
The GLP-1–oestrogen conjugate yielded significant benefits in managing diabetes. The GLP-1–oestrogen conjugate notably reduced daily insulin needs by 60% and triggered the endoplasmic-reticulum-associated protein-degradation system. This intervention improved β-cell survival and regeneration. Moreover, the GLP-1–oestrogen conjugate shielded human β-cells from dysfunction induced by cytokines [353].
The impacts of γ-aminobutyric acid (GABA) alone or in combination with the GCGR mAb on β-cell and α-cell mass in T1D mice have also been studied. Treatment with GABA or the GCGR mAb individually improved hyperglycemia and increased β-cell mass, with similar effects observed in the combination treatment. Interestingly, the combined treatment attenuated GCGR mAb-induced α-cell hyperplasia [354].
In summary, anti-GCGR treatment and GCGR antagonism promote β-cell regeneration through multiple mechanisms, including α-to-β-cell transdifferentiation, the upregulation of β-cell genes, and involvement of GLP-1 and FGF21 pathways. These findings highlight the potential of GCGR antagonists in enhancing β-cell mass and function in diabetes therapy.

10.3. The Polyamine Biosynthesis Pathway

The polyamine biosynthesis pathway is a conserved metabolic process in all organisms, involving key enzymes like aminopropyltransferases (spermidine and spermine synthases). This pathway is essential for cellular functions, including growth, proliferation, and stress responses. Research has shown that inhibitors of polyamine biosynthesis, such as difluoromethylornithine (DFMO), enhance β-cell regeneration without affecting overall growth or pancreas length, while Imatinib mesylate, a tyrosine kinase inhibitor that inhibits polyamine biosynthesis pathway, has no impact. These findings suggest that targeting polyamine biosynthesis, especially with DFMO, holds promise for stimulating β-cell regeneration in diabetes [17].
In the context of the polyamine pathway, administering spermidine to NOD mice heightened the diabetes occurrence, despite unchanging insulitis levels. This elevation was correlated with increased pancreatic tissue polyamines and markers of autophagy. Remarkably, spermidine induced shifts in the T-cell composition within pancreatic lymph nodes, amplifying pro-inflammatory T-cell populations in diabetic mice and demonstrating differing impacts on regulatory T cells depending on disease onset, ultimately exacerbating T1D incidence [355].
In preclinical models, DFMO has also been shown to delay T1D onset by reducing β-cell stress. A clinical trial with recent-onset T1D patients showed that DFMO is safe and tolerable, reducing urinary putrescine levels and preserving C-peptide at higher doses. Transcriptomic and proteomic analyses suggest DFMO preserves β-cell function by altering mRNA translation, protein transport, and secretion in stressed islets [356].

10.4. Other Pathways Involved in β-Cell Proliferation

In addition to the aforementioned methods to induce β-cell proliferation and regeneration, there are other approaches, such as inhibiting TBK1/IKKε and salt-inducible kinases (SIKs).
Xu et al. found TBK1/IKKε inhibitors (TBK1/IKKε-Is) to be potent enhancers of β-cell regeneration in a zebrafish model of T1D, validated across mammalian systems including human and rat β-cells, and STZ-induced diabetic mice. The mechanism involves TBK1/IKKε inhibition, which promotes β-cell proliferation via the cAMP-PKA-mTOR signaling axis through PDE3, highlighting a novel role for TBK1/IKKε in β-cell mass modulation. Notably, TBK1/IKKε-Is like amlexanox and PIAA selectively enhance β-cell proliferation without inducing proliferation in other cell types, showing promise for targeted therapy [357].
An in vivo study by Charbord et al. identified HG-9-91-01(HG) as a β-cell mitogen that enhances β-cell proliferation in zebrafish, mouse, and human β-cells by inhibiting salt-inducible kinases (SIKs), specifically SIK1–SIK3, members of the AMPK-related protein kinase family. HG stimulates β-cell proliferation through UPR activation via ATF6–IRE1 pathways. Despite showing specificity for SIK1, HG’s pharmacokinetic limitations currently preclude its consideration as a drug candidate [358].

11. Conclusions

β-cell dedifferentiation and transdifferentiation are critical factors in the progression of both Type 1 and T2D (T1D and T2D). Metabolic stress and inflammation significantly contribute to β-cell dedifferentiation and transdifferentiation by dysregulating various pathways, including ER stress, the mTOR pathway, the endocannabinoid system, viral infections, and aging. Targeting these processes is considered a potential therapeutic strategy for diabetes. Various natural products, hormones, and dietary treatments have shown promise in mitigating β-cell dedifferentiation and transdifferentiation.
β-cell regeneration and neogenesis present potential treatments for diabetes, particularly T1D. Research has demonstrated that various pancreatic cells, including δ-, ε-, and α-cells, can participate in β-cell regeneration under diabetic conditions. Treatments such as glucagon receptor antagonism, both alone and in combination with GLP-1 agonism and DYRK1A inhibition, have been effective in regenerating ablated β-cells in diabetic models. Additionally, differentiating MSCs into β-cells, followed by the transplantation of these newly differentiated cells, offers a promising approach for diabetes treatment, especially for T1D. It has been shown that specific inducers containing certain components can effectively promote the neogenesis of MSCs into β-cells.

12. Limitations

  • Lack of review on experimental models: In this study, we did not review the experimental models and the methods used to model β-cell dedifferentiation and transdifferentiation. A comprehensive examination of the various approaches to model β-cell dedifferentiation and transdifferentiation in both in vitro and in vivo settings would be highly beneficial.
  • Limited scope on cell plasticity: This review primarily focused on β-cell plasticity and did not cover the plasticities of other cell types. It would be valuable to briefly explore the plasticities of other cells such as hepatocytes, hepatic stellate cells, epidermal stem cells, melanocytes, astrocytes, intestinal stem cells, cardiac fibroblasts, club cells, podocytes, and others. Investigating the plasticities of these diverse cell types could offer insights into the broader applications and implications of cellular plasticity.
  • Context of cell plasticity in other diseases: While this review centered on β-cell plasticity, it would have been beneficial to study cell plasticity in the context of other diseases such as liver diseases, neurodegenerative diseases, and cancer. Understanding how cellular plasticity manifests and contributes to the pathology of these diseases could inform the development of new therapeutic strategies and enhance our knowledge of disease progression and treatment.

13. Unanswered Questions

Even though the progress in diabetes research and the role of β-cell plasticity in it have been substantial, many questions have remained unanswered:
Combinatorial effects of treatments: What are the combined impacts of various effective natural products and common antidiabetic medications on β-cell dedifferentiation, transdifferentiation, regeneration, and neogenesis?
Endocannabinoid system and β-cell plasticity: How does the endocannabinoid system influence β-cell plasticity? Can the modulation of this system induce β-cell regeneration and neogenesis?
Serotonin signaling pathway and β-cell plasticity: How does the serotonin signaling pathway affect β-cell plasticity? Can targeting this pathway enhance β-cell regeneration and neogenesis?
Long-term effects of β-cell plasticity interventions: What are the long-term effects of interventions aimed at promoting β-cell plasticity on the overall pancreatic function and glucose homeostasis?

Author Contributions

Conceptualization, E.G.G. and I.K.; literature review and curation, S.R., F.N., S.S., B.W. and D.L.; writing—original draft preparation, E.G.G.; writing, E.G.G.; reviewing and editing, all authors; supervision, I.K. and O.K.; funding acquisition, I.K. and O.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by MITACS grant #33301.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Bensellam, M.; Jonas, J.-C.; Laybutt, D.R. Mechanisms of β-cell dedifferentiation in diabetes: Recent findings and future research directions. J. Endocrinol. 2018, 236, R109–R143. [Google Scholar] [CrossRef]
  2. Son, J.; Accili, D. Reversing pancreatic β-cell dedifferentiation in the treatment of type 2 diabetes. Exp. Mol. Med. 2023, 55, 1652–1658. [Google Scholar] [CrossRef] [PubMed]
  3. Ghasemi-Gojani, E.; Kovalchuk, I.; Kovalchuk, O. Cannabinoids and terpenes for diabetes mellitus and its complications: From mechanisms to new therapies. Trends Endocrinol. Metab. 2022, 33, 828–849. [Google Scholar] [CrossRef] [PubMed]
  4. Tanday, N.; Tarasov, A.I.; Moffett, R.C.; Flatt, P.R.; Irwin, N. Pancreatic islet cell plasticity: Pathogenic or therapeutically exploitable? Diabetes Obes. Metab. 2024, 26, 16–31. [Google Scholar] [CrossRef] [PubMed]
  5. Honzawa, N.; Fujimoto, K. The Plasticity of Pancreatic β-Cells. Metabolites 2021, 11, 218. [Google Scholar] [CrossRef]
  6. Khin, P.-P.; Lee, J.-H.; Jun, H.-S. A Brief Review of the Mechanisms of β-Cell Dedifferentiation in Type 2 Diabetes. Nutrients 2021, 13, 1593. [Google Scholar] [CrossRef] [PubMed]
  7. Cinti, F.; Bouchi, R.; Kim-Muller, J.Y.; Ohmura, Y.; Sandoval, P.R.; Masini, M.; Marselli, L.; Suleiman, M.; Ratner, L.E.; Marchetti, P.; et al. Evidence of β-Cell Dedifferentiation in Human Type 2 Diabetes. J. Clin. Endocrinol. Metab. 2016, 101, 1044–1054. [Google Scholar] [CrossRef]
  8. Gao, T.; McKenna, B.; Li, C.; Reichert, M.; Nguyen, J.; Singh, T.; Yang, C.; Pannikar, A.; Doliba, N.; Zhang, T.; et al. Pdx1 Maintains β Cell Identity and Function by Repressing an α Cell Program. Cell Metab. 2014, 19, 259–271. [Google Scholar] [CrossRef]
  9. Zhou, Q.; Brown, J.; Kanarek, A.; Rajagopal, J.; Melton, D.A. In vivo reprogramming of adult pancreatic exocrine cells to β-cells. Nature 2008, 455, 627–632. [Google Scholar] [CrossRef]
  10. Eizirik, D.L.; Sammeth, M.; Bouckenooghe, T.; Bottu, G.; Sisino, G.; Igoillo-Esteve, M.; Ortis, F.; Santin, I.; Colli, M.L.; Barthson, J.; et al. The Human Pancreatic Islet Transcriptome: Expression of Candidate Genes for Type 1 Diabetes and the Impact of Pro-Inflammatory Cytokines. PLoS Genet. 2012, 8, e1002552. [Google Scholar] [CrossRef]
  11. Efrat, S. Beta-cell dedifferentiation in type 2 diabetes: Concise review. Stem Cells 2019, 37, 1267–1272. [Google Scholar] [CrossRef] [PubMed]
  12. Chikodili, I.M.; Chioma, I.I.; Chinwendu, N.M.; IfedibaluChukwu, E.I. In-silico study for African plants with possible beta-cell regeneration effect through inhibition of DYRK1A. Sci. Phytochem. 2022, 1, 13–28. [Google Scholar] [CrossRef]
  13. Hædersdal, S.; Andersen, A.; Knop, F.K.; Vilsbøll, T. Revisiting the role of glucagon in health, diabetes mellitus and other metabolic diseases. Nat. Rev. Endocrinol. 2023, 19, 321–335. [Google Scholar] [CrossRef] [PubMed]
  14. Wei, R.; Gu, L.; Yang, J.; Yang, K.; Liu, J.; Le, Y.; Lang, S.; Wang, H.; Thai, D.; Yan, H.; et al. Antagonistic Glucagon Receptor Antibody Promotes α-Cell Proliferation and Increases β-Cell Mass in Diabetic Mice. iScience 2019, 16, 326–339. [Google Scholar] [CrossRef]
  15. Wei, T.; Cui, X.; Jiang, Y.; Wang, K.; Wang, D.; Li, F.; Lin, X.; Gu, L.; Yang, K.; Yang, J.; et al. Glucagon Acting at the GLP-1 Receptor Contributes to β-Cell Regeneration Induced by Glucagon Receptor Antagonism in Diabetic Mice. Diabetes 2023, 72, 599–610. [Google Scholar] [CrossRef]
  16. Xi, Y.; Song, B.; Ngan, I.; Solloway, M.J.; Humphrey, M.; Wang, Y.; Mondal, K.; Wu, H.; Liu, W.; Lindhout, D.A.; et al. Glucagon-receptor-antagonism-mediated β-cell regeneration as an effective anti-diabetic therapy. Cell Rep. 2022, 39, 110872. [Google Scholar] [CrossRef]
  17. Robertson, M.A.; Padgett, L.R.; Fine, J.A.; Chopra, G.; Mastracci, T.L. Targeting polyamine biosynthesis to stimulate beta cell regeneration in zebrafish. Islets 2020, 12, 99–107. [Google Scholar] [CrossRef] [PubMed]
  18. Mezza, T.; Cinti, F.; Cefalo, C.M.A.; Pontecorvi, A.; Kulkarni, R.N.; Giaccari, A. β-Cell Fate in Human Insulin Resistance and Type 2 Diabetes: A Perspective on Islet Plasticity. Diabetes 2019, 68, 1121–1129. [Google Scholar] [CrossRef]
  19. Zhou, Q.; Melton, D.A. Pancreas regeneration. Nature 2018, 557, 351–358. [Google Scholar] [CrossRef]
  20. DiMeglio, L.A.; Evans-Molina, C.; Oram, R.A. Type 1 diabetes. Lancet 2018, 391, 2449–2462. [Google Scholar] [CrossRef]
  21. DeFronzo, R.A.; Ferrannini, E.; Groop, L.; Henry, R.R.; Herman, W.H.; Holst, J.J.; Hu, F.B.; Kahn, C.R.; Raz, I.; Shulman, G.I. Type 2 diabetes mellitus. Nat. Rev. Dis. Prim. 2015, 1, 1–22. [Google Scholar] [CrossRef]
  22. Ahlqvist, E.; Prasad, R.B.; Groop, L. Subtypes of Type 2 Diabetes Determined From Clinical Parameters. Diabetes 2020, 69, 2086–2093. [Google Scholar] [CrossRef]
  23. McIntyre, H.D.; Catalano, P.; Zhang, C.; Desoye, G.; Mathiesen, E.R.; Damm, P. Gestational diabetes mellitus. Nat. Rev. Dis. Prim. 2019, 5, 47. [Google Scholar] [CrossRef]
  24. Liang, R.; Liu, N.; Wang, G.; Sun, P.; Liu, Y.; Zou, J.; Wang, L.; Ding, X.; Zhang, B.; Shen, Z.; et al. Cytohistologic analyses of β cell dedifferentiation induced by inflammation in human islets. Eur. J. Inflamm. 2021, 19. [Google Scholar] [CrossRef]
  25. Nordmann, T.M.; Dror, E.; Schulze, F.; Traub, S.; Berishvili, E.; Barbieux, C.; Böni-Schnetzler, M.; Donath, M.Y. The Role of Inflammation in β-cell Dedifferentiation. Sci. Rep. 2017, 7, 6285. [Google Scholar] [CrossRef]
  26. Urizar, A.I.; Prause, M.; Ingerslev, L.R.; Wortham, M.; Sui, Y.; Sander, M.; Williams, K.; Barrès, R.; Larsen, M.R.; Christensen, G.L.; et al. Beta cell dysfunction induced by bone morphogenetic protein (BMP)-2 is associated with histone modifications and decreased NeuroD1 chromatin binding. Cell Death Dis. 2023, 14, 399. [Google Scholar] [CrossRef]
  27. Wang, H.-L.; Wang, L.; Zhao, C.-Y.; Lan, H.-Y. Role of TGF-Beta Signaling in Beta Cell Proliferation and Function in Diabetes. Biomolecules 2022, 12, 373. [Google Scholar] [CrossRef]
  28. Blum, B.; Roose, A.N.; Barrandon, O.; Maehr, R.; Arvanites, A.C.; Davidow, L.S.; Davis, J.C.; Peterson, Q.P.; Rubin, L.L.; Melton, D.A.; et al. Reversal of β cell de-differentiation by a small molecule inhibitor of the TGFβ pathway. eLife 2014, 3, e02809. [Google Scholar] [CrossRef]
  29. Qian, J.; Tao, D.; Shan, X.; Xiao, X.; Chen, C. Role of angiogenesis in beta-cell epithelial–mesenchymal transition in chronic pancreatitis-induced diabetes. Mod. Pathol. 2022, 102, 290–297. [Google Scholar] [CrossRef]
  30. Brown, M.L.; Andrzejewski, D.; Burnside, A.; Schneyer, A.L. Activin enhances α-to β-cell transdifferentiation as a source for β-cells in male FSTL3 knockout mice. Endocrinology 2016, 157, 1043–1054. [Google Scholar] [CrossRef]
  31. Chen, X.Y.; Shi, Y.X.; Huang, Y.P.; Ding, M.; Shen, Q.L.; Li, C.J.; Lin, J.N. SDF-1 inhibits the dedifferentiation of islet β cells in hyperglycaemia by up-regulating FoxO1 via binding to CXCR4. J. Cell. Mol. Med. 2022, 26, 750–763. [Google Scholar] [CrossRef]
  32. Bosma, K.J.; Andrei, S.R.; Katz, L.S.; Smith, A.A.; Dunn, J.C.; Ricciardi, V.F.; Ramirez, M.A.; Baumel-Alterzon, S.; Pace, W.A.; Carroll, D.T.; et al. Pharmacological blockade of the EP3 prostaglandin E2 receptor in the setting of type 2 diabetes enhances β-cell proliferation and identity and relieves oxidative damage. Mol. Metab. 2021, 54, 101347. [Google Scholar] [CrossRef]
  33. Moin, A.S.M.; Dhawan, S.; Shieh, C.; Butler, P.C.; Cory, M.; Butler, A.E. Increased Hormone-Negative Endocrine Cells in the Pancreas in Type 1 Diabetes. J. Clin. Endocrinol. Metab. 2016, 101, 3487–3496. [Google Scholar] [CrossRef]
  34. Michels, A.W.; Landry, L.G.; McDaniel, K.A.; Yu, L.; Campbell-Thompson, M.; Kwok, W.W.; Jones, K.L.; Gottlieb, P.A.; Kappler, J.W.; Tang, Q.; et al. Islet-Derived CD4 T Cells Targeting Proinsulin in Human Autoimmune Diabetes. Diabetes 2016, 66, 722–734. [Google Scholar] [CrossRef]
  35. Diedisheim, M.; Oshima, M.; Albagli, O.; Huldt, C.W.; Ahlstedt, I.; Clausen, M.; Menon, S.; Aivazidis, A.; Andreasson, A.-C.; Haynes, W.G.; et al. Modeling human pancreatic beta cell dedifferentiation. Mol. Metab. 2018, 10, 74–86. [Google Scholar] [CrossRef]
  36. Oshima, M.; Knoch, K.-P.; Diedisheim, M.; Petzold, A.; Cattan, P.; Bugliani, M.; Marchetti, P.; Choudhary, P.; Huang, G.-C.; Bornstein, S.R.; et al. Virus-like infection induces human β cell dedifferentiation. J. Clin. Investig. 2018, 3, e97732. [Google Scholar] [CrossRef]
  37. Thorel, F.; Népote, V.; Avril, I.; Kohno, K.; Desgraz, R.; Chera, S.; Herrera, P.L. Conversion of adult pancreatic α-cells to β-cells after extreme β-cell loss. Nature 2010, 464, 1149–1154. [Google Scholar] [CrossRef]
  38. Brissova, M.; Haliyur, R.; Saunders, D.; Shrestha, S.; Dai, C.; Blodgett, D.M.; Bottino, R.; Campbell-Thompson, M.; Aramandla, R.; Poffenberger, G.; et al. α Cell Function and Gene Expression Are Compromised in Type 1 Diabetes. Cell Rep. 2018, 22, 2667–2676. [Google Scholar] [CrossRef]
  39. Furth-Lavi, J.; Hija, A.; Tornovsky-Babeay, S.; Mazouz, A.; Dahan, T.; Stolovich-Rain, M.; Klochendler, A.; Dor, Y.; Avrahami, D.; Glaser, B. Glycemic control releases regenerative potential of pancreatic beta cells blocked by severe hyperglycemia. Cell Rep. 2022, 41, 111719. [Google Scholar] [CrossRef]
  40. Hahn, M.; van Krieken, P.P.; Nord, C.; Alanentalo, T.; Morini, F.; Xiong, Y.; Eriksson, M.; Mayer, J.; Kostromina, E.; Ruas, J.L.; et al. Topologically selective islet vulnerability and self-sustained downregulation of markers for β-cell maturity in streptozotocin-induced diabetes. Commun. Biol. 2020, 3, 541. [Google Scholar] [CrossRef]
  41. Katz, L.S.; Brill, G.; Zhang, P.; Kumar, A.; Baumel-Alterzon, S.; Honig, L.B.; Gómez-Banoy, N.; Karakose, E.; Tanase, M.; Doridot, L.; et al. Maladaptive positive feedback production of ChREBPβ underlies glucotoxic β-cell failure. Nat. Commun. 2022, 13, 4423. [Google Scholar] [CrossRef]
  42. Oshima, M.; Pechberty, S.; Bellini, L.; Göpel, S.O.; Campana, M.; Rouch, C.; Dairou, J.; Cosentino, C.; Fantuzzi, F.; Toivonen, S.; et al. Stearoyl CoA desaturase is a gatekeeper that protects human beta cells against lipotoxicity and maintains their identity. Diabetologia 2019, 63, 395–409. [Google Scholar] [CrossRef]
  43. Dobosz, A.M.; Janikiewicz, J.; Krogulec, E.; Dziewulska, A.; Ajduk, A.; Szpila, M.; Nieznańska, H.; Szczepankiewicz, A.A.; Wypych, D.; Dobrzyn, A. Inhibition of stearoyl-CoA desaturase 1 in the mouse impairs pancreatic islet morphogenesis and promotes loss of β-cell identity and α-cell expansion in the mature pancreas. Mol. Metab. 2023, 67, 101659. [Google Scholar] [CrossRef]
  44. Leenders, F.; Groen, N.; de Graaf, N.; Engelse, M.A.; Rabelink, T.J.; de Koning, E.J.P.; Carlotti, F. Oxidative Stress Leads to β-Cell Dysfunction Through Loss of β-Cell Identity. Front. Immunol. 2021, 12, 690379. [Google Scholar] [CrossRef]
  45. Motomura, K.; Matsuzaka, T.; Shichino, S.; Ogawa, T.; Pan, H.; Nakajima, T.; Asano, Y.; Okayama, T.; Takeuchi, T.; Ohno, H.; et al. Single-Cell Transcriptome Profiling of Pancreatic Islets From Early Diabetic Mice Identifies Anxa10 for Ca2+ Allostasis Toward β-Cell Failure. Diabetes 2023, 73, 75–92. [Google Scholar] [CrossRef]
  46. Dadheech, N.; Srivastava, A.; Shah, R.G.; Shah, G.M.; Gupta, S. Role of poly(ADP-ribose) polymerase-1 in regulating human islet cell differentiation. Sci. Rep. 2022, 12, 21496. [Google Scholar] [CrossRef]
  47. Liu, B.; Hua, D.; Shen, L.; Li, T.; Tao, Z.; Fu, C.; Tang, Z.; Yang, J.; Zhang, L.; Nie, A.; et al. NPC1 is required for postnatal islet β cell differentiation by maintaining mitochondria turnover. Theranostics 2024, 14, 2058–2074. [Google Scholar] [CrossRef]
  48. Yeh, Y.-T.; Sona, C.; Yan, X.; Li, Y.; Pathak, A.; McDermott, M.I.; Xie, Z.; Liu, L.; Arunagiri, A.; Wang, Y.; et al. Restoration of PITPNA in Type 2 diabetic human islets reverses pancreatic beta-cell dysfunction. Nat. Commun. 2023, 14, 4250. [Google Scholar] [CrossRef]
  49. Song, J.; Ni, Q.; Sun, J.; Xie, J.; Liu, J.; Ning, G.; Wang, W.; Wang, Q. Aging Impairs Adaptive Unfolded Protein Response and Drives Beta Cell Dedifferentiation in Humans. J. Clin. Endocrinol. Metab. 2022, 107, 3231–3241. [Google Scholar] [CrossRef] [PubMed]
  50. Téllez, N.; Vilaseca, M.; Martí, Y.; Pla, A.; Montanya, E. β-Cell dedifferentiation, reduced duct cell plasticity, and impaired β-cell mass regeneration in middle-aged rats. Am. J. Physiol. Endocrinol. Metab. 2016, 311, E554–E563. [Google Scholar] [CrossRef] [PubMed]
  51. Murao, N.; Yokoi, N.; Takahashi, H.; Hayami, T.; Minami, Y.; Seino, S. Increased glycolysis affects β-cell function and identity in aging and diabetes. Mol. Metab. 2021, 55, 101414. [Google Scholar] [CrossRef] [PubMed]
  52. Tanday, N.; Coulter-Parkhill, A.; Moffett, R.C.; Suruli, K.; Dubey, V.; Flatt, P.R.; Irwin, N. Sex-based impact of pancreatic islet stressors in GluCreERT2/Rosa26-eYFP mice. J. Endocrinol. 2023, 259, e230174. [Google Scholar] [CrossRef] [PubMed]
  53. Yang, B.C.; Wu, S.Y.; Leung, P.S. Alcohol ingestion induces pancreatic islet dysfunction and apoptosis via mediation of FGF21 resistance. Ann. Transl. Med. 2020, 8, 310. [Google Scholar] [CrossRef] [PubMed]
  54. Rasineni, K.; Kubik, J.L.; Knight, K.L.; Hall, L.; Casey, C.A.; Kharbanda, K.K. Ghrelin regulates adipose tissue metabolism: Role in hepatic steatosis. Chem. Interactions 2020, 322, 109059. [Google Scholar] [CrossRef] [PubMed]
  55. Lee, Y.; Ravazzola, M.; Park, B.-H.; Bashmakov, Y.K.; Orci, L.; Unger, R.H. Metabolic mechanisms of failure of intraportally transplanted pancreatic β-cells in rats: Role of lipotoxicity and prevention by leptin. Diabetes 2007, 56, 2295–2301. [Google Scholar] [CrossRef] [PubMed]
  56. Holland, W.L.; Miller, R.A.; Wang, Z.V.; Sun, K.; Barth, B.M.; Bui, H.H.; Davis, K.E.; Bikman, B.T.; Halberg, N.; Rutkowski, J.M.; et al. Receptor-mediated activation of ceramidase activity initiates the pleiotropic actions of adiponectin. Nat. Med. 2011, 17, 55–63. [Google Scholar] [CrossRef]
  57. Dufrane, D.; van Steenberghe, M.; Guiot, Y.; Goebbels, R.-M.; Saliez, A.; Gianello, P. Streptozotocin-Induced Diabetes in Large Animals (Pigs/Primates): Role of GLUT2 Transporter and β-cell Plasticity. Transplantation 2006, 81, 36–45. [Google Scholar] [CrossRef]
  58. Feng, Y.; Qiu, W.-L.; Yu, X.-X.; Zhang, Y.; He, M.-Y.; Li, L.-C.; Yang, L.; Zhang, W.; Franti, M.; Ye, J. Characterizing pancreatic β-cell heterogeneity in the streptozotocin model by single-cell transcriptomic analysis. Mol. Metab. 2020, 37, 100982. [Google Scholar] [CrossRef] [PubMed]
  59. Cheng, Y.; Kang, H.; Shen, J.; Hao, H.; Liu, J.; Guo, Y.; Mu, Y.; Han, W. Beta-cell regeneration from vimentin+/MafB+ cells after STZ-induced extreme beta-cell ablation. Sci. Rep. 2015, 5, 11703. [Google Scholar] [CrossRef]
  60. Rafacho, A.; Ortsäter, H.; Nadal, A.; Quesada, I. Glucocorticoid treatment and endocrine pancreas function: Implications for glucose homeostasis, insulin resistance and diabetes. J. Endocrinol. 2014, 223, R49–R62. [Google Scholar] [CrossRef]
  61. Esguerra, J.L.; Ofori, J.K.; Nagao, M.; Shuto, Y.; Karagiannopoulos, A.; Fadista, J.; Sugihara, H.; Groop, L.; Eliasson, L. Glucocorticoid induces human beta cell dysfunction by involving riborepressor GAS5 LincRNA. Mol. Metab. 2020, 32, 160–167. [Google Scholar] [CrossRef] [PubMed]
  62. Beaupere, C.; Liboz, A.; Fève, B.; Blondeau, B.; Guillemain, G. Molecular Mechanisms of Glucocorticoid-Induced Insulin Resistance. Int. J. Mol. Sci. 2021, 22, 623. [Google Scholar] [CrossRef] [PubMed]
  63. Valtat, B.; Dupuis, C.; Zenaty, D.; Singh-Estivalet, A.; Tronche, F.; Bréant, B.; Blondeau, B. Genetic evidence of the programming of beta cell mass and function by glucocorticoids in mice. Diabetologia 2010, 54, 350–359. [Google Scholar] [CrossRef] [PubMed]
  64. Martínez-Pinna, J.; Sempere-Navarro, R.; Medina-Gali, R.M.; Fuentes, E.; Quesada, I.; Sargis, R.M.; Trasande, L.; Nadal, A. Endocrine disruptors in plastics alter β-cell physiology and increase the risk of diabetes mellitus. Am. J. Physiol.-Endocrinol. Metab. 2023, 324, E488–E505. [Google Scholar] [CrossRef] [PubMed]
  65. Dos Santos, R.S.; Medina-Gali, R.M.; Babiloni-Chust, I.; Marroqui, L.; Nadal, A. In Vitro Assays to Identify Metabolism-Disrupting Chemicals with Diabetogenic Activity in a Human Pancreatic β-Cell Model. Int. J. Mol. Sci. 2022, 23, 5040. [Google Scholar] [CrossRef] [PubMed]
  66. Sidarala, V.; Zhu, J.; Levi-D’ancona, E.; Pearson, G.L.; Reck, E.C.; Walker, E.M.; Kaufman, B.A.; Soleimanpour, S.A. Mitofusin 1 and 2 regulation of mitochondrial DNA content is a critical determinant of glucose homeostasis. Nat. Commun. 2022, 13, 2340. [Google Scholar] [CrossRef] [PubMed]
  67. Barazzuol, L.; Giamogante, F.; Calì, T. Mitochondria Associated Membranes (MAMs): Architecture and physiopathological role. Cell Calcium 2021, 94, 102343. [Google Scholar] [CrossRef] [PubMed]
  68. Boronat-Belda, T.; Ferrero, H.; Al-Abdulla, R.; Quesada, I.; Gustafsson, J.-A.; Nadal, Á.; Alonso-Magdalena, P. Bisphenol-A exposure during pregnancy alters pancreatic β-cell division and mass in male mice offspring: A role for ERβ. Food Chem. Toxicol. 2020, 145, 111681. [Google Scholar] [CrossRef] [PubMed]
  69. Azevedo, L.F.; Masiero, M.M.; Cherkaoui, S.; Carneiro, M.H.; Barbosa Jr, F.; Zamboni, N. The alternative analog plasticizer BPS displays similar phenotypic and metabolomic responses to BPA in HepG2 and INS-1E cells. Food Chem. Toxicol. 2022, 167, 113266. [Google Scholar] [CrossRef]
  70. García-Arévalo, M.; Alonso-Magdalena, P.; Servitja, J.-M.; Boronat-Belda, T.; Merino, B.; Villar-Pazos, S.; Medina-Gómez, G.; Novials, A.; Quesada, I.; Nadal, A. Maternal Exposure to Bisphenol-A During Pregnancy Increases Pancreatic β-Cell Growth During Early Life in Male Mice Offspring. Endocrinology 2016, 157, 4158–4171. [Google Scholar] [CrossRef]
  71. Bansal, A.; Rashid, C.; Xin, F.; Li, C.; Polyak, E.; Duemler, A.; van der Meer, T.; Stefaniak, M.; Wajid, S.; Doliba, N.; et al. Sex- and Dose-Specific Effects of Maternal Bisphenol A Exposure on Pancreatic Islets of First- and Second-Generation Adult Mice Offspring. Environ. Heal. Perspect. 2017, 125, 097022. [Google Scholar] [CrossRef]
  72. Yang, R.; Zheng, J.; Qin, J.; Liu, S.; Liu, X.; Gu, Y.; Yang, S.; Du, J.; Li, S.; Chen, B.; et al. Dibutyl phthalate affects insulin synthesis and secretion by regulating the mitochondrial apoptotic pathway and oxidative stress in rat insulinoma cells. Ecotoxicol. Environ. Saf. 2023, 249, 114396. [Google Scholar] [CrossRef]
  73. Brusco, N.; Sebastiani, G.; Di Giuseppe, G.; Licata, G.; Grieco, G.E.; Fignani, D.; Nigi, L.; Formichi, C.; Aiello, E.; Auddino, S.; et al. Intra-islet insulin synthesis defects are associated with endoplasmic reticulum stress and loss of beta cell identity in human diabetes. Diabetologia 2022, 66, 354–366. [Google Scholar] [CrossRef] [PubMed]
  74. Rewers, M.; Ludvigsson, J. Environmental risk factors for type 1 diabetes. Lancet 2016, 387, 2340–2348. [Google Scholar] [CrossRef]
  75. Almanza, A.; Carlesso, A.; Chintha, C.; Creedican, S.; Doultsinos, D.; Leuzzi, B.; Luís, A.; McCarthy, N.; Montibeller, L.; More, S.; et al. Endoplasmic reticulum stress signalling—From basic mechanisms to clinical applications. FEBS J. 2018, 286, 241–278. [Google Scholar] [CrossRef]
  76. Chen, C.-W.; Guan, B.-J.; Alzahrani, M.R.; Gao, Z.; Gao, L.; Bracey, S.; Wu, J.; Mbow, C.A.; Jobava, R.; Haataja, L.; et al. Adaptation to chronic ER stress enforces pancreatic β-cell plasticity. Nat. Commun. 2022, 13, 4621. [Google Scholar] [CrossRef]
  77. Lee, H.; Lee, Y.-S.; Harenda, Q.; Pietrzak, S.; Oktay, H.Z.; Schreiber, S.; Liao, Y.; Sonthalia, S.; Ciecko, A.E.; Chen, Y.-G.; et al. Beta Cell Dedifferentiation Induced by IRE1α Deletion Prevents Type 1 Diabetes. Cell Metab. 2020, 31, 822–836.e5. [Google Scholar] [CrossRef]
  78. Lee, K.; Chan, J.Y.; Liang, C.; Ip, C.K.; Shi, Y.-C.; Herzog, H.; Hughes, W.E.; Bensellam, M.; Delghingaro-Augusto, V.; Koina, M.E.; et al. XBP1 maintains beta cell identity, represses beta-to-alpha cell transdifferentiation and protects against diabetic beta cell failure during metabolic stress in mice. Diabetologia 2022, 65, 984–996. [Google Scholar] [CrossRef] [PubMed]
  79. Kitakaze, K.; Oyadomari, M.; Zhang, J.; Hamada, Y.; Takenouchi, Y.; Tsuboi, K.; Inagaki, M.; Tachikawa, M.; Fujitani, Y.; Okamoto, Y.; et al. ATF4-mediated transcriptional regulation protects against β-cell loss during endoplasmic reticulum stress in a mouse model. Mol. Metab. 2021, 54, 101338. [Google Scholar] [CrossRef] [PubMed]
  80. Lee, J.-H.; Ryu, H.; Lee, H.; Yu, H.R.; Gao, Y.; Lee, K.-M.; Kim, Y.-J.; Lee, J. Endoplasmic reticulum stress in pancreatic β cells induces incretin desensitization and β-cell dysfunction via ATF4-mediated PDE4D expression. Am. J. Physiol. Endocrinol. Metab. 2023, 325, E448–E465. [Google Scholar] [CrossRef]
  81. Kulkarni, A.; Muralidharan, C.; May, S.C.; Tersey, S.A.; Mirmira, R.G. Inside the β Cell: Molecular Stress Response Pathways in Diabetes Pathogenesis. Endocrinology 2022, 164, bqac184. [Google Scholar] [CrossRef] [PubMed]
  82. Pakos-Zebrucka, K.; Koryga, I.; Mnich, K.; Ljujic, M.; Samali, A.; Gorman, A.M. The integrated stress response. EMBO Rep. 2016, 17, 1374–1395. [Google Scholar] [CrossRef] [PubMed]
  83. Koromilas, A.E. M(en)TORship lessons on life and death by the integrated stress response. Biochim. Biophys. Acta (BBA) Gen. Subj. 2019, 1863, 644–649. [Google Scholar] [CrossRef] [PubMed]
  84. Zaborske, J.M.; Narasimhan, J.; Jiang, L.; Wek, S.A.; Dittmar, K.A.; Freimoser, F.; Pan, T.; Wek, R.C. Genome-wide Analysis of tRNA Charging and Activation of the eIF2 Kinase Gcn2p. J. Biol. Chem. 2009, 284, 25254–25267. [Google Scholar] [CrossRef]
  85. Kilberg, M.S.; Shan, J.; Su, N. ATF4-dependent transcription mediates signaling of amino acid limitation. Trends Endocrinol. Metab. 2009, 20, 436–443. [Google Scholar] [CrossRef] [PubMed]
  86. Kanno, A.; Asahara, S.-I.; Furubayashi, A.; Masuda, K.; Yoshitomi, R.; Suzuki, E.; Takai, T.; Kimura-Koyanagi, M.; Matsuda, T.; Bartolome, A.; et al. GCN2 regulates pancreatic β cell mass by sensing intracellular amino acid levels. J. Clin. Investig. 2020, 5, e128820. [Google Scholar] [CrossRef] [PubMed]
  87. Darden, C.M.; Vasu, S.; Mattke, J.; Liu, Y.; Rhodes, C.J.; Naziruddin, B.; Lawrence, M.C. Calcineurin/NFATc2 and PI3K/AKT signaling maintains β-cell identity and function during metabolic and inflammatory stress. iScience 2022, 25, 104125. [Google Scholar] [CrossRef]
  88. Ramalingam, L.; Sopontammarak, B.; Menikdiwela, K.R.; Moustaid-Moussa, N. Endoplasmic Reticulum (ER) Stress in Part Mediates Effects of Angiotensin II in Pancreatic Beta Cells. Diabetes Metab. Syndr. Obes. Targets Ther. 2020, 13, 2843–2853. [Google Scholar] [CrossRef]
  89. Chen, H.; Zhou, W.; Ruan, Y.; Yang, L.; Xu, N.; Chen, R.; Yang, R.; Sun, J.; Zhang, Z. Reversal of angiotensin ll-induced β-cell dedifferentiation via inhibition of NF-κb signaling. Mol. Med. 2018, 24, 43. [Google Scholar] [CrossRef]
  90. Li, M.-Y.; Liu, L.-Z.; Xin, Q.; Zhou, J.; Zhang, X.; Zhang, R.; Wu, Z.; Yi, J.; Dong, M. Downregulation of mTORC1 and Mcl-1 by lipid-oversupply contributes to islet β-cell apoptosis and dysfunction. Biochim. Biophys. Acta (BBA) Mol. Cell Biol. Lipids 2023, 1868, 159332. [Google Scholar] [CrossRef]
  91. Cai, Z.; Liu, F.; Yang, Y.; Li, D.; Hu, S.; Song, L.; Yu, S.; Li, T.; Liu, B.; Luo, H. GRB10 regulates β-cell mass by inhibiting β-cell proliferation and stimulating β-cell dedifferentiation. J. Genet. Genom. 2022, 49, 208–216. [Google Scholar] [CrossRef] [PubMed]
  92. Yin, Q.; Ni, Q.; Wang, Y.; Zhang, H.; Li, W.; Nie, A.; Wang, S.; Gu, Y.; Wang, Q.; Ning, G. Raptor determines β-cell identity and plasticity independent of hyperglycemia in mice. Nat. Commun. 2020, 11, 2538. [Google Scholar] [CrossRef] [PubMed]
  93. Ni, Q.; Sun, J.; Wang, Y.; Wang, Y.; Liu, J.; Ning, G.; Wang, W.; Wang, Q. mTORC1 is required for epigenetic silencing during β-cell functional maturation. Mol. Metab. 2022, 64, 101559. [Google Scholar] [CrossRef] [PubMed]
  94. Cui, C.; Li, T.; Xie, Y.; Yang, J.; Fu, C.; Qiu, Y.; Shen, L.; Ni, Q.; Wang, Q.; Nie, A. Enhancing Acsl4 in absence of mTORC2/Rictor drove β-cell dedifferentiation via inhibiting FoxO1 and promoting ROS production. Biochim. Biophys. Acta (BBA) Mol. Basis Dis. 2021, 1867, 166261. [Google Scholar] [CrossRef] [PubMed]
  95. Lupse, B.; Annamalai, K.; Ibrahim, H.; Kaur, S.; Geravandi, S.; Sarma, B.; Pal, A.; Awal, S.; Joshi, A.; Rafizadeh, S. Inhibition of PHLPP1/2 phosphatases rescues pancreatic β-cells in diabetes. Cell Rep. 2021, 36, 109490. [Google Scholar] [CrossRef] [PubMed]
  96. Liu, Z.; Wang, P.; Zhao, Y.; Lai, K.P.; Li, R. Biomedical importance of the ubiquitin–proteasome system in diabetes and metabolic transdifferentiation of pancreatic duct epithelial cells into β-cells. Gene 2023, 858, 147191. [Google Scholar] [CrossRef] [PubMed]
  97. Kumar, A.; Francis, M.; Bhaskar, S.; Vishnuvajhala, S.; Prasanna, J. Dynamics of Ubiquitination in Differentiation and Dedifferentiation of Pancreatic β-cells: Putative Target for Diabetes. Curr. Protein Pept. Sci. 2022, 23, 602–618. [Google Scholar] [CrossRef]
  98. Francis, M.; Bhaskar, S.; Komanduri, S.; Sheshadri, P.; Prasanna, J.; Kumar, A. Deubiquitinase USP1 influences the dedifferentiation of mouse pancreatic β-cells. iScience 2023, 26, 106771. [Google Scholar] [CrossRef]
  99. Manea, T.; Nelson, J.K.; Garrone, C.M.; Hansson, K.; Evans, I.; Behrens, A.; Sancho, R. USP7 controls NGN3 stability and pancreatic endocrine lineage development. Nat. Commun. 2023, 14, 2457. [Google Scholar] [CrossRef]
  100. Cho, G.; Hyun, K.; Choi, J.; Shin, E.; Kim, B.; Kim, H.; Kim, J.; Han, Y.-M. Arginine 65 methylation of Neurogenin 3 by PRMT1 is required for pancreatic endocrine development of hESCs. Exp. Mol. Med. 2023, 55, 1506–1519. [Google Scholar] [CrossRef]
  101. Zhang, T.; Wang, H.; Wang, T.; Wei, C.; Jiang, H.; Jiang, S.; Yang, J.; Shao, J.; Ma, L. Pax4 synergistically acts with Pdx1, Ngn3 and MafA to induce HuMSCs to differentiate into functional pancreatic β-cells. Exp. Ther. Med. 2019, 18, 2592–2598. [Google Scholar] [CrossRef]
  102. Lorenzo, P.I.; Juárez-Vicente, F.; Cobo-Vuilleumier, N.; García-Domínguez, M.; Gauthier, B.R. The Diabetes-Linked Transcription Factor PAX4: From Gene to Functional Consequences. Genes 2017, 8, 101. [Google Scholar] [CrossRef] [PubMed]
  103. Brun, T.; Franklin, I.; St-Onge, L.; Biason-Lauber, A.; Schoenle, E.J.; Wollheim, C.B.; Gauthier, B.R. The diabetes-linked transcription factor PAX4 promotes β-cell proliferation and survival in rat and human islets. J. Cell Biol. 2004, 167, 1123–1135. [Google Scholar] [CrossRef]
  104. Parajuli, K.R.; Zhang, Y.; Cao, A.M.; Wang, H.; Fonseca, V.A.; Wu, H. Pax4 Gene Delivery Improves Islet Transplantation Efficacy by Promoting β Cell Survival and α-to-β Cell Transdifferentiation. Cell Transplant. 2020, 29. [Google Scholar] [CrossRef]
  105. Auerbach, A.; Cohen, A.; Shlomai, N.O.; Weinberg-Shukron, A.; Gulsuner, S.; King, M.-C.; Hemi, R.; Levy-Lahad, E.; Abulibdeh, A.; Zangen, D. NKX2-2 Mutation Causes Congenital Diabetes and Infantile Obesity With Paradoxical Glucose-Induced Ghrelin Secretion. J. Clin. Endocrinol. Metab. 2020, 105, 3486–3495. [Google Scholar] [CrossRef]
  106. Doyle, M.J.; Sussel, L. Nkx2. 2 regulates β-cell function in the mature islet. Diabetes 2007, 56, 1999–2007. [Google Scholar] [CrossRef]
  107. Papizan, J.B.; Singer, R.A.; Tschen, S.-I.; Dhawan, S.; Friel, J.M.; Hipkens, S.B.; Magnuson, M.A.; Bhushan, A.; Sussel, L. Nkx2. 2 repressor complex regulates islet β-cell specification and prevents β-to-α-cell reprogramming. Genes Dev. 2011, 25, 2291–2305. [Google Scholar] [CrossRef] [PubMed]
  108. Anderson, K.R.; Torres, C.A.; Solomon, K.; Becker, T.C.; Newgard, C.B.; Wright, C.V.; Hagman, J.; Sussel, L. Cooperative Transcriptional Regulation of the Essential Pancreatic Islet Gene NeuroD1 (Beta2) by Nkx2.2 and Neurogenin 3. J. Biol. Chem. 2009, 284, 31236–31248. [Google Scholar] [CrossRef] [PubMed]
  109. Gutiérrez, G.D.; Bender, A.S.; Cirulli, V.; Mastracci, T.L.; Kelly, S.M.; Tsirigos, A.; Kaestner, K.H.; Sussel, L. Pancreatic β cell identity requires continual repression of non–β cell programs. J. Clin. Investig. 2017, 127, 244–259. [Google Scholar] [CrossRef]
  110. Fontcuberta-PiSunyer, M.; García-Alamán, A.; Prades, È.; Téllez, N.; Alves-Figueiredo, H.; Ramos-Rodríguez, M.; Enrich, C.; Fernandez-Ruiz, R.; Cervantes, S.; Clua, L.; et al. Direct reprogramming of human fibroblasts into insulin-producing cells using transcription factors. Commun. Biol. 2023, 6, 256. [Google Scholar] [CrossRef]
  111. Pan, F.C.; Brissova, M.; Powers, A.C.; Pfaff, S.; Wright, C.V.E. Inactivating the permanent neonatal diabetes gene Mnx1 switches insulin-producing β-cells to a δ-like fate and reveals a facultative proliferative capacity in aged β-cells. Development 2015, 142, 3637–3648. [Google Scholar] [CrossRef] [PubMed]
  112. Flanagan, S.E.; De Franco, E.; Allen, H.L.; Zerah, M.; Abdul-Rasoul, M.M.; Edge, J.A.; Stewart, H.; Alamiri, E.; Hussain, K.; Wallis, S.; et al. Analysis of Transcription Factors Key for Mouse Pancreatic Development Establishes NKX2-2 and MNX1 Mutations as Causes of Neonatal Diabetes in Man. Cell Metab. 2014, 19, 146–154. [Google Scholar] [CrossRef] [PubMed]
  113. Arkhipova, V.; Wendik, B.; Devos, N.; Ek, O.; Peers, B.; Meyer, D. Characterization and regulation of the hb9/mnx1 beta-cell progenitor specific enhancer in zebrafish. Dev. Biol. 2012, 365, 290–302. [Google Scholar] [CrossRef] [PubMed]
  114. Dalgin, G.; Ward, A.B.; Hao, L.T.; Beattie, C.E.; Nechiporuk, A.; Prince, V.E. Zebrafish mnx1 controls cell fate choice in the developing endocrine pancreas. Development 2011, 138, 4597–4608. [Google Scholar] [CrossRef] [PubMed]
  115. Dalgin, G.; Prince, V.E. Mnx1: A gatekeeper of β cell fate. Islets 2012, 4, 320–322. [Google Scholar] [CrossRef] [PubMed]
  116. Li, X.; Wan, T.; Li, Y. Role of FoxO1 in regulating autophagy in type 2 diabetes mellitus (Review). Exp. Ther. Med. 2021, 22, 707. [Google Scholar] [CrossRef] [PubMed]
  117. Kitamura, Y.I.; Kitamura, T.; Kruse, J.-P.; Raum, J.C.; Stein, R.; Gu, W.; Accili, D. FoxO1 protects against pancreatic β cell failure through NeuroD and MafA induction. Cell Metab. 2005, 2, 153–163. [Google Scholar] [CrossRef]
  118. Casteels, T.; Zhang, Y.; Frogne, T.; Sturtzel, C.; Lardeau, C.-H.; Sen, I.; Liu, X.; Hong, S.; Pauler, F.M.; Penz, T.; et al. An inhibitor-mediated beta-cell dedifferentiation model reveals distinct roles for FoxO1 in glucagon repression and insulin maturation. Mol. Metab. 2021, 54, 101329. [Google Scholar] [CrossRef]
  119. Khatri, R.; Mazurek, S.; Petry, S.F.; Linn, T. Mesenchymal stem cells promote pancreatic β-cell regeneration through downregulation of FoxO1 pathway. Stem Cell Res. Ther. 2020, 11, 497. [Google Scholar] [CrossRef]
  120. Nie, Y.; Zhang, Y.; Liu, S.; Xu, Z.; Xia, C.; Du, L.; Yin, X.; Wang, J. Downregulation of Sirt3 contributes to β-cell dedifferentiation via FoxO1 in type 2 diabetic mellitus. Acta Diabetol. 2023, 61, 485–494. [Google Scholar] [CrossRef]
  121. Li, Y.; Deng, S.; Peng, J.; Wang, X.; Essandoh, K.; Mu, X.; Peng, T.; Meng, Z.-X.; Fan, G.-C. MicroRNA-223 is essential for maintaining functional β-cell mass during diabetes through inhibiting both FOXO1 and SOX6 pathways. J. Biol. Chem. 2019, 294, 10438–10448. [Google Scholar] [CrossRef] [PubMed]
  122. Tang, S.; Xin, Y.; Yang, M.; Zhang, D.; Xu, C. Osteoprotegerin promotes islet β cell proliferation in intrauterine growth retardation rats through the PI3K/AKT/FoxO1 pathway. Int. J. Clin. Exp. Pathol. 2019, 12, 2324. [Google Scholar] [PubMed]
  123. Wang, K.; Cui, X.; Li, F.; Xia, L.; Wei, T.; Liu, J.; Fu, W.; Yang, J.; Hong, T.; Wei, R. Glucagon receptor blockage inhibits β-cell dedifferentiation through FoxO1. Am. J. Physiol. Metab. 2023, 324, E97–E113. [Google Scholar] [CrossRef] [PubMed]
  124. Ebrahim, N.; Shakirova, K.; Dashinimaev, E. PDX1 is the cornerstone of pancreatic β-cell functions and identity. Front. Mol. Biosci. 2022, 9, 1091757. [Google Scholar] [CrossRef]
  125. Chen, F.; Sha, M.; Wang, Y.; Wu, T.; Shan, W.; Liu, J.; Zhou, W.; Zhu, Y.; Sun, Y.; Shi, Y.; et al. Transcription factor Ets-1 links glucotoxicity to pancreatic beta cell dysfunction through inhibiting PDX-1 expression in rodent models. Diabetologia 2015, 59, 316–324. [Google Scholar] [CrossRef] [PubMed]
  126. Zhang, Y.; Fang, X.; Wei, J.; Miao, R.; Wu, H.; Ma, K.; Tian, J. PDX-1: A Promising Therapeutic Target to Reverse Diabetes. Biomolecules 2022, 12, 1785. [Google Scholar] [CrossRef] [PubMed]
  127. Weidemann, B.J.; Marcheva, B.; Kobayashi, M.; Omura, C.; Newman, M.V.; Kobayashi, Y.; Waldeck, N.J.; Perelis, M.; Lantier, L.; McGuinness, O.P. Repression of latent NF-κB enhancers by PDX1 regulates β cell functional heterogeneity. Cell Metab. 2024, 36, 90–102.e107. [Google Scholar] [CrossRef] [PubMed]
  128. Tsuyama, T.; Sato, Y.; Yoshizawa, T.; Matsuoka, T.; Yamagata, K. Hypoxia causes pancreatic β-cell dysfunction and impairs insulin secretion by activating the transcriptional repressor BHLHE40. EMBO Rep. 2023, 24, e56227. [Google Scholar] [CrossRef]
  129. Vanhoose, A.M.; Samaras, S.; Artner, I.; Henderson, E.; Hang, Y.; Stein, R. MafA and MafB Regulate Pdx1 Transcription through the Area II Control Region in Pancreatic β Cells. J. Biol. Chem. 2008, 283, 22612–22619. [Google Scholar] [CrossRef]
  130. Aigha, I.I.; Abdelalim, E.M. NKX6. 1 transcription factor: A crucial regulator of pancreatic β cell development, identity, and proliferation. Stem Cell Res. Ther. 2020, 11, 459. [Google Scholar] [CrossRef]
  131. Luan, C.; Ye, Y.; Singh, T.; Barghouth, M.; Eliasson, L.; Artner, I.; Zhang, E.; Renström, E. The calcium channel subunit gamma-4 is regulated by MafA and necessary for pancreatic beta-cell specification. Commun. Biol. 2019, 2, 106. [Google Scholar] [CrossRef] [PubMed]
  132. Liang, J.; Chirikjian, M.; Pajvani, U.B.; Bartolomé, A. MafA Regulation in β-Cells: From Transcriptional to Post-Translational Mechanisms. Biomolecules 2022, 12, 535. [Google Scholar] [CrossRef] [PubMed]
  133. Nishimura, W.; Iwasa, H.; Tumurkhuu, M. Role of the Transcription Factor MAFA in the Maintenance of Pancreatic β-Cells. Int. J. Mol. Sci. 2022, 23, 4478. [Google Scholar] [CrossRef] [PubMed]
  134. Hang, Y.; Yamamoto, T.; Benninger, R.K.; Brissova, M.; Guo, M.; Bush, W.; Piston, D.W.; Powers, A.C.; Magnuson, M.; Thurmond, D.C.; et al. The MafA Transcription Factor Becomes Essential to Islet β-Cells Soon After Birth. Diabetes 2014, 63, 1994–2005. [Google Scholar] [CrossRef] [PubMed]
  135. Eto, K.; Nishimura, W.; Oishi, H.; Udagawa, H.; Kawaguchi, M.; Hiramoto, M.; Fujiwara, T.; Takahashi, S.; Yasuda, K. MafA Is Required for Postnatal Proliferation of Pancreatic β-Cells. PLoS ONE 2014, 9, e104184. [Google Scholar] [CrossRef] [PubMed]
  136. Gradwohl, G.; Dierich, A.; LeMeur, M.; Guillemot, F. neurogenin3 is required for the development of the four endocrine cell lineages of the pancreas. Proc. Natl. Acad. Sci. USA 2000, 97, 1607–1611. [Google Scholar] [CrossRef] [PubMed]
  137. Zhu, Y.; Liu, Q.; Zhou, Z.; Ikeda, Y. PDX1, Neurogenin-3, and MAFA: Critical transcription regulators for beta cell development and regeneration. Stem Cell Res. Ther. 2017, 8, 240. [Google Scholar] [CrossRef] [PubMed]
  138. Schreiber, V.; Mercier, R.; Jiménez, S.; Ye, T.; García-Sánchez, E.; Klein, A.; Meunier, A.; Ghimire, S.; Birck, C.; Jost, B.; et al. Extensive NEUROG3 occupancy in the human pancreatic endocrine gene regulatory network. Mol. Metab. 2021, 53, 101313. [Google Scholar] [CrossRef]
  139. Miyatsuka, T.; Kosaka, Y.; Kim, H.; German, M.S. Neurogenin3 inhibits proliferation in endocrine progenitors by inducing Cdkn1a. Proc. Natl. Acad. Sci. USA 2010, 108, 185–190. [Google Scholar] [CrossRef]
  140. McDonald, E.; Li, J.; Krishnamurthy, M.; Fellows, G.F.; Goodyer, C.G.; Wang, R. SOX9 regulates endocrine cell differentiation during human fetal pancreas development. Int. J. Biochem. Cell Biol. 2012, 44, 72–83. [Google Scholar] [CrossRef]
  141. Dettmer, R.; Niwolik, I.; Mehmeti, I.; Jörns, A.; Naujok, O. New hPSC SOX9 and INS Reporter Cell Lines Facilitate the Observation and Optimization of Differentiation into Insulin-Producing Cells. Stem Cell Rev. Rep. 2021, 17, 2193–2209. [Google Scholar] [CrossRef] [PubMed]
  142. Wang, Q.; Chen, J.; Han, J.; Yang, Z.; Yang, Y.; Li, H.; Wang, S.; Hong, Y. Sox9 is required in regeneration of pancreatic β cells following injury. Exp. Cell Res. 2023, 422, 113406. [Google Scholar] [CrossRef] [PubMed]
  143. Zhang, M.; Lin, Q.; Qi, T.; Wang, T.; Chen, C.-C.; Riggs, A.D.; Zeng, D. Growth factors and medium hyperglycemia induce Sox9+ ductal cell differentiation into β cells in mice with reversal of diabetes. Proc. Natl. Acad. Sci. USA 2016, 113, 650–655. [Google Scholar] [CrossRef] [PubMed]
  144. Qian, M.F.; Bevacqua, R.J.; Coykendall, V.M.; Liu, X.; Zhao, W.; Chang, C.A.; Gu, X.; Dai, X.-Q.; MacDonald, P.E.; Kim, S.K. HNF1α maintains pancreatic α and β cell functions in primary human islets. JCI Insight 2023, 8, e170884. [Google Scholar] [CrossRef] [PubMed]
  145. Sato, Y.; Rahman, M.; Haneda, M.; Tsuyama, T.; Mizumoto, T.; Yoshizawa, T.; Kitamura, T.; Gonzalez, F.J.; Yamamura, K.-I.; Yamagata, K. HNF1α controls glucagon secretion in pancreatic α-cells through modulation of SGLT1. Biochim. Biophys. Acta (BBA) Mol. Basis Dis. 2020, 1866, 165898. [Google Scholar] [CrossRef] [PubMed]
  146. Low, B.S.J.; Lim, C.S.; Ding, S.S.L.; Tan, Y.S.; Ng, N.H.J.; Krishnan, V.G.; Ang, S.F.; Neo, C.W.Y.; Verma, C.S.; Hoon, S. Decreased GLUT2 and glucose uptake contribute to insulin secretion defects in MODY3/HNF1A hiPSC-derived mutant β cells. Nat. Commun. 2021, 12, 3133. [Google Scholar] [CrossRef] [PubMed]
  147. Chen, Y.; Jia, J.; Zhao, Q.; Zhang, Y.; Huang, B.; Wang, L.; Tian, J.; Huang, C.; Li, M.; Li, X. Novel Loss-of-Function Variant in HNF1a Induces β-Cell Dysfunction through Endoplasmic Reticulum Stress. Int. J. Mol. Sci. 2022, 23, 13022. [Google Scholar] [CrossRef] [PubMed]
  148. Cujba, A.-M.; Alvarez-Fallas, M.E.; Pedraza-Arevalo, S.; Laddach, A.; Shepherd, M.H.; Hattersley, A.T.; Watt, F.M.; Sancho, R. An HNF1α truncation associated with maturity-onset diabetes of the young impairs pancreatic progenitor differentiation by antagonizing HNF1β function. Cell Rep. 2022, 38, 110425. [Google Scholar] [CrossRef] [PubMed]
  149. Sujjitjoon, J.; Charoensuk, C.; Thanyaphon, T.; Kooptiwut, S.; Thamtarana, P.J.; Tangjittipokin, W.; Chongjaroen, N.; Chanprasert, C.; Abubakar, Z.; Lapbenjakul, S.; et al. Defective functions of HNF1A variants on BCL2L1 transactivation and beta-cell growth. Biochem. Biophys. Res. Commun. 2020, 529, 826–833. [Google Scholar] [CrossRef]
  150. Friedman-Mazursky, O.; Elkon, R.; Efrat, S. Redifferentiation of expanded human islet β cells by inhibition of ARX. Sci. Rep. 2016, 6, 20698. [Google Scholar] [CrossRef]
  151. Courtney, M.; Rabe, T.; Collombat, P.; Mansouri, A. Pax4 and Arx Represent Crucial Regulators of the Development of the Endocrine Pancreas. New J. Sci. 2014, 2014, 1–6. [Google Scholar] [CrossRef]
  152. Đorđević, M.; Stepper, P.; Feuerstein-Akgoz, C.; Gerhauser, C.; Paunović, V.; Tolić, A.; Rajić, J.; Dinić, S.; Uskoković, A.; Grdović, N.; et al. EpiCRISPR targeted methylation of Arx gene initiates transient switch of mouse pancreatic alpha to insulin-producing cells. Front. Endocrinol. 2023, 14, 1134478. [Google Scholar] [CrossRef] [PubMed]
  153. Courtney, M.; Gjernes, E.; Druelle, N.; Ravaud, C.; Vieira, A.; Ben-Othman, N.; Pfeifer, A.; Avolio, F.; Leuckx, G.; Lacas-Gervais, S.; et al. The Inactivation of Arx in Pancreatic α-Cells Triggers Their Neogenesis and Conversion into Functional β-like Cells. PLoS Genet. 2013, 9, e1003934. [Google Scholar] [CrossRef] [PubMed]
  154. Villamayor, L.; Rodríguez-Seguel, E.; Araujo, R.; Carrasco, M.; Bru-Tarí, E.; Mellado-Gil, J.M.; Gauthier, B.R.; Martinelli, P.; Quesada, I.; Soria, B.; et al. GATA6 Controls Insulin Biosynthesis and Secretion in Adult β-Cells. Diabetes 2017, 67, 448–460. [Google Scholar] [CrossRef]
  155. Chia, C.Y.; Madrigal, P.; Denil, S.L.; Martinez, I.; Garcia-Bernardo, J.; El-Khairi, R.; Chhatriwala, M.; Shepherd, M.H.; Hattersley, A.T.; Dunn, N.R. GATA6 cooperates with EOMES/SMAD2/3 to deploy the gene regulatory network governing human definitive endoderm and pancreas formation. Stem Cell Rep. 2019, 12, 57–70. [Google Scholar] [CrossRef] [PubMed]
  156. Tiyaboonchai, A.; Cardenas-Diaz, F.L.; Ying, L.; Maguire, J.A.; Sim, X.; Jobaliya, C.; Gagne, A.L.; Kishore, S.; Stanescu, D.E.; Hughes, N.; et al. GATA6 Plays an Important Role in the Induction of Human Definitive Endoderm, Development of the Pancreas, and Functionality of Pancreatic β Cells. Stem Cell Rep. 2017, 8, 589–604. [Google Scholar] [CrossRef] [PubMed]
  157. Sanyoura, M.; Jacobsen, L.; Carmody, D.; del Gaudio, D.; Alkorta-Aranburu, G.; Arndt, K.; Hu, Y.Y.; Kobiernicki, F.; Kusmartseva, I.; Atkinson, M.A.; et al. Pancreatic Histopathology of Human Monogenic Diabetes Due to Causal Variants in KCNJ11, HNF1A, GATA6, and LMNA. J. Clin. Endocrinol. Metab. 2017, 103, 35–45. [Google Scholar] [CrossRef] [PubMed]
  158. Yue, X.; Luo, Y.; Wang, J.; Huang, D. Monogenic Diabetes with GATA6 Mutations: Characterization of a Novel Family and a Comprehensive Analysis of the GATA6 Clinical and Genetics Traits. Mol. Biotechnol. 2023, 66, 467–474. [Google Scholar] [CrossRef] [PubMed]
  159. Kim, Y.-S.; Kang, H.S.; Takeda, Y.; Hom, L.; Song, H.-Y.; Jensen, J.; Jetten, A.M. Glis3 Regulates Neurogenin 3 Expression in Pancreatic β-Cells and Interacts with Its Activator, Hnf6. Mol. Cells 2012, 34, 193–200. [Google Scholar] [CrossRef]
  160. Scoville, D.W.; Lichti-Kaiser, K.; Grimm, S.A.; Jetten, A.M. GLIS3 binds pancreatic beta cell regulatory regions alongside other islet transcription factors. J. Endocrinol. 2019, 243, 1–14. [Google Scholar] [CrossRef]
  161. Amin, S.; Cook, B.; Zhou, T.; Ghazizadeh, Z.; Lis, R.; Zhang, T.; Khalaj, M.; Crespo, M.; Perera, M.; Xiang, J.Z.; et al. Discovery of a drug candidate for GLIS3-associated diabetes. Nat. Commun. 2018, 9, 2681. [Google Scholar] [CrossRef] [PubMed]
  162. Meulebrouck, S.; Scherrer, V.; Boutry, R.; Toussaint, B.; Vaillant, E.; Dechaume, A.; Loiselle, H.; Balkau, B.; Charpentier, G.; Franc, S.; et al. Pathogenic monoallelic variants in GLIS3 increase type 2 diabetes risk and identify a subgroup of patients sensitive to sulfonylureas. Diabetologia 2023, 67, 327–332. [Google Scholar] [CrossRef] [PubMed]
  163. Grieve, L.M.; Rani, A.; ZeRuth, G.T. Downregulation of Glis3 in INS1 cells exposed to chronically elevated glucose contributes to glucotoxicity-associated β cell dysfunction. Islets 2024, 16, 2344622. [Google Scholar] [CrossRef] [PubMed]
  164. Swisa, A.; Avrahami, D.; Eden, N.; Zhang, J.; Feleke, E.; Dahan, T.; Cohen-Tayar, Y.; Stolovich-Rain, M.; Kaestner, K.H.; Glaser, B.; et al. PAX6 maintains β cell identity by repressing genes of alternative islet cell types. J. Clin. Investig. 2016, 127, 230–243. [Google Scholar] [CrossRef] [PubMed]
  165. Gosmain, Y.; Katz, L.S.; Masson, M.H.; Cheyssac, C.; Poisson, C.; Philippe, J. Pax6 is crucial for β-cell function, insulin biosynthesis, and glucose-induced insulin secretion. Mol. Endocrinol. 2012, 26, 696–709. [Google Scholar] [CrossRef] [PubMed]
  166. Ashery-Padan, R.; Zhou, X.; Marquardt, T.; Herrera, P.; Toube, L.; Berry, A.; Gruss, P. Conditional inactivation of Pax6 in the pancreas causes early onset of diabetes. Dev. Biol. 2004, 269, 479–488. [Google Scholar] [CrossRef] [PubMed]
  167. Sheng, J.; Wang, L.; Tang, P.M.-K.; Wang, H.-L.; Li, J.-C.; Xu, B.-H.; Xue, V.W.; Tan, R.-Z.; Jin, N.; Chan, T.-F.; et al. Smad3 deficiency promotes beta cell proliferation and function in db/db mice via restoring Pax6 expression. Theranostics 2021, 11, 2845–2859. [Google Scholar] [CrossRef] [PubMed]
  168. Bsharat, S.; Monni, E.; Singh, T.; Johansson, J.K.; Achanta, K.; Bertonnier-Brouty, L.; Schmidt-Christensen, A.; Holmberg, D.; Kokaia, Z.; Prasad, R.B. MafB-dependent neurotransmitter signaling promotes β cell migration in the developing pancreas. Development 2023, 150, dev201009. [Google Scholar] [CrossRef] [PubMed]
  169. Cyphert, H.A.; Walker, E.M.; Hang, Y.; Dhawan, S.; Haliyur, R.; Bonatakis, L.; Avrahami, D.; Brissova, M.; Kaestner, K.H.; Bhushan, A.; et al. Examining How the MAFB Transcription Factor Affects Islet β-Cell Function Postnatally. Diabetes 2018, 68, 337–348. [Google Scholar] [CrossRef] [PubMed]
  170. Cha, J.; Tong, X.; Walker, E.M.; Dahan, T.; Cochrane, V.A.; Ashe, S.; Russell, R.; Osipovich, A.B.; Mawla, A.M.; Guo, M.; et al. Species-specific roles for the MAFA and MAFB transcription factors in regulating islet β cell identity. J. Clin. Investig. 2023, 8, e166386. [Google Scholar] [CrossRef]
  171. Russell, R.; Carnese, P.P.; Hennings, T.G.; Walker, E.M.; Russ, H.A.; Liu, J.S.; Giacometti, S.; Stein, R.; Hebrok, M. Loss of the transcription factor MAFB limits β-cell derivation from human PSCs. Nat. Commun. 2020, 11, 2742. [Google Scholar] [CrossRef] [PubMed]
  172. Bar, Y.; Russ, H.A.; Sintov, E.; Anker-Kitai, L.; Knoller, S.; Efrat, S. Redifferentiation of Expanded Human Pancreatic β-Cell-derived Cells by Inhibition of the NOTCH Pathway. J. Biol. Chem. 2012, 287, 17269–17280. [Google Scholar] [CrossRef]
  173. Bar, Y.; Russ, H.A.; Knoller, S.; Ouziel-Yahalom, L.; Efrat, S. HES-1 Is Involved in Adaptation of Adult Human β-Cells to Proliferation In Vitro. Diabetes 2008, 57, 2413–2420. [Google Scholar] [CrossRef] [PubMed]
  174. Marui, S.; Nishikawa, Y.; Shiokawa, M.; Yokode, M.; Matsumoto, S.; Muramoto, Y.; Ota, S.; Nakamura, T.; Yoshida, H.; Okada, H.; et al. Context-Dependent Roles of Hes1 in the Adult Pancreas and Pancreatic Tumor Formation. Gastroenterology 2022, 163, 1613–1629.e12. [Google Scholar] [CrossRef]
  175. Xu, Y.; Mao, S.; Fan, H.; Wan, J.; Wang, L.; Zhang, M.; Zhu, S.; Yuan, J.; Lu, Y.; Wang, Z.; et al. LINC MIR503HG Controls SC-β Cell Differentiation and Insulin Production by Targeting CDH1 and HES1. Adv. Sci. 2024, 11, e2305631. [Google Scholar] [CrossRef] [PubMed]
  176. Barth, R.; Ruoso, C.; Ferreira, S.M.; de Ramos, F.C.; Lima, F.B.; Boschero, A.C.; Dos Santos, G.J. Hepatocyte Nuclear Factor 4-α (HNF4α) controls the insulin resistance-induced pancreatic β-cell mass expansion. Life Sci. 2022, 289, 120213. [Google Scholar] [CrossRef]
  177. Han, E.H.; Singh, P.; Lee, I.-K.; Urrutia, R.; Chi, Y.-I. ErbB3-binding protein 1 (EBP1) represses HNF4α-mediated transcription and insulin secretion in pancreatic β-cells. J. Biol. Chem. 2019, 294, 13983–13994. [Google Scholar] [CrossRef]
  178. Romer, A.I.; Singer, R.A.; Sui, L.; Egli, D.; Sussel, L. Murine Perinatal β-Cell Proliferation and the Differentiation of Human Stem Cell–Derived Insulin-Expressing Cells Require NEUROD1. Diabetes 2019, 68, 2259–2271. [Google Scholar] [CrossRef]
  179. Bohuslavova, R.; Fabriciova, V.; Smolik, O.; Lebrón-Mora, L.; Abaffy, P.; Benesova, S.; Zucha, D.; Valihrach, L.; Berkova, Z.; Saudek, F.; et al. NEUROD1 reinforces endocrine cell fate acquisition in pancreatic development. Nat. Commun. 2023, 14, 5554. [Google Scholar] [CrossRef]
  180. Gu, C.; Stein, G.H.; Pan, N.; Goebbels, S.; Hörnberg, H.; Nave, K.-A.; Herrera, P.; White, P.; Kaestner, K.H.; Sussel, L.; et al. Pancreatic β Cells Require NeuroD to Achieve and Maintain Functional Maturity. Cell Metab. 2010, 11, 298–310. [Google Scholar] [CrossRef]
  181. Schaffer, A.E.; Taylor, B.L.; Benthuysen, J.R.; Liu, J.; Thorel, F.; Yuan, W.; Jiao, Y.; Kaestner, K.H.; Herrera, P.L.; Magnuson, M.A.; et al. Nkx6.1 Controls a Gene Regulatory Network Required for Establishing and Maintaining Pancreatic Beta Cell Identity. PLoS Genet. 2013, 9, e1003274. [Google Scholar] [CrossRef]
  182. Memon, B.; Younis, I.; Abubaker, F.; Abdelalim, E.M. PDX1−/NKX6. 1+ progenitors derived from human pluripotent stem cells as a novel source of insulin-secreting cells. Diabetes/Metab. Res. Rev. 2021, 37, e3400. [Google Scholar] [CrossRef] [PubMed]
  183. Fujita, Y.; Kozawa, J.; Fukui, K.; Iwahashi, H.; Eguchi, H.; Shimomura, I. Increased NKX6.1 expression and decreased ARX expression in alpha cells accompany reduced beta-cell volume in human subjects. Sci. Rep. 2021, 11, 17796. [Google Scholar] [CrossRef]
  184. Tessem, J.S.; Moss, L.G.; Chao, L.C.; Arlotto, M.; Lu, D.; Jensen, M.V.; Stephens, S.B.; Tontonoz, P.; Hohmeier, H.E.; Newgard, C.B. Nkx6.1 regulates islet β-cell proliferation via Nr4a1 and Nr4a3 nuclear receptors. Proc. Natl. Acad. Sci. USA 2014, 111, 5242–5247. [Google Scholar] [CrossRef] [PubMed]
  185. Ratiu, J.; Southard, S.M.; Rust, W.L. The transcription factors ISL-1 and MAFA, but not NKX6-1 Characterize a Stem-cell Derived Population of Endocrine Pancreatic Cells Capable of Controlling blood Glucose in Rodent Models of Diabetes. Med. Res. Arch. 2023, 11. [Google Scholar] [CrossRef]
  186. Zhang, J.; Zhang, R.; Liu, C.; Ge, X.; Wang, Y.; Jiang, F.; Zhuang, L.; Li, T.; Zhu, Q.; Jiang, Y.; et al. Missense mutation of ISL1 (E283D) is associated with the development of type 2 diabetes. Diabetologia 2024. [Google Scholar] [CrossRef]
  187. Bohuslavova, R.; Fabriciova, V.; Lebrón-Mora, L.; Malfatti, J.; Smolik, O.; Valihrach, L.; Benesova, S.; Zucha, D.; Berkova, Z.; Saudek, F.; et al. ISL1 controls pancreatic alpha cell fate and beta cell maturation. Cell Biosci. 2023, 13, 53. [Google Scholar] [CrossRef] [PubMed]
  188. Guo, T.; Wang, W.; Zhang, H.; Liu, Y.; Chen, P.; Ma, K.; Zhou, C. ISL1 Promotes Pancreatic Islet Cell Proliferation. PLoS ONE 2011, 6, e22387. [Google Scholar] [CrossRef]
  189. Connors, C.T.; Villaca, C.B.; Anderson-Baucum, E.K.; Rosario, S.R.; Rutan, C.D.; Childress, P.J.; Padgett, L.R.; Robertson, M.A.; Mastracci, T.L. A Translational Regulatory Mechanism Mediated by Hypusinated Eukaryotic Initiation Factor 5A Facilitates β-Cell Identity and Function. Diabetes 2023, 73, 461–473. [Google Scholar] [CrossRef]
  190. Wang, W.; Zhang, C. Targeting β-cell dedifferentiation and transdifferentiation: Opportunities and challenges. Endocr. Connect. 2021, 10, R213–R228. [Google Scholar] [CrossRef]
  191. Wu, Q.-J.; Zhang, T.-N.; Chen, H.-H.; Yu, X.-F.; Lv, J.-L.; Liu, Y.-Y.; Liu, Y.-S.; Zheng, G.; Zhao, J.-Q.; Wei, Y.-F. The sirtuin family in health and disease. Signal Transduct. Target. Ther. 2022, 7, 402. [Google Scholar] [PubMed]
  192. Cao, H.; Chung, A.C.; Ming, X.; Mao, D.; Lee, H.M.; Cao, X.; Rutter, G.A.; Chan, J.C.; Tian, X.Y.; Kong, A.P. Autotaxin signaling facilitates β cell dedifferentiation and dysfunction induced by Sirtuin 3 deficiency. Mol. Metab. 2022, 60, 101493. [Google Scholar] [CrossRef] [PubMed]
  193. Zhang, L.; Sheng, C.; Zhou, F.; Zhu, K.; Wang, S.; Liu, Q.; Yuan, M.; Xu, Z.; Liu, Y.; Lu, J.; et al. CBP/p300 HAT maintains the gene network critical for β cell identity and functional maturity. Cell Death Dis. 2021, 12, 476. [Google Scholar] [CrossRef] [PubMed]
  194. Vanderkruk, B.; Maeshima, N.; Pasula, D.J.; An, M.; McDonald, C.L.; Suresh, P.; Luciani, D.S.; Lynn, F.C.; Hoffman, B.G. Methylation of histone H3 lysine 4 is required for maintenance of beta cell function in adult mice. Diabetologia 2023, 66, 1097–1115. [Google Scholar] [CrossRef] [PubMed]
  195. Al-Hasani, K.; Marikar, S.N.; Kaipananickal, H.; Maxwell, S.; Okabe, J.; Khurana, I.; Karagiannis, T.; Liang, J.J.; Mariana, L.; Loudovaris, T.; et al. EZH2 inhibitors promote β-like cell regeneration in young and adult type 1 diabetes donors. Signal Transduct. Target. Ther. 2024, 9, 2. [Google Scholar] [CrossRef] [PubMed]
  196. Parveen, N.; Dhawan, S. DNA Methylation Patterning and the Regulation of Beta Cell Homeostasis. Front. Endocrinol. 2021, 12, 651258. [Google Scholar] [CrossRef]
  197. Parveen, N.; Wang, J.K.; Bhattacharya, S.; Cuala, J.; Rajkumar, M.S.; Butler, A.E.; Wu, X.; Shih, H.-P.; Georgia, S.K.; Dhawan, S. DNA Methylation-Dependent Restriction of Tyrosine Hydroxylase Contributes to Pancreatic β-Cell Heterogeneity. Diabetes 2023, 72, 575–589. [Google Scholar] [CrossRef] [PubMed]
  198. Celen, C.; Chuang, J.-C.; Shen, S.; Li, L.; Maggiore, G.; Jia, Y.; Luo, X.; Moore, A.; Wang, Y.; Otto, J.E.; et al. Arid1a loss potentiates pancreatic β-cell regeneration through activation of EGF signaling. Cell Rep. 2022, 41, 111581. [Google Scholar] [CrossRef] [PubMed]
  199. Spaeth, J.M.; Liu, J.-H.; Peters, D.; Guo, M.; Osipovich, A.B.; Mohammadi, F.; Roy, N.; Bhushan, A.; Magnuson, M.A.; Hebrok, M.; et al. The Pdx1-Bound Swi/Snf Chromatin Remodeling Complex Regulates Pancreatic Progenitor Cell Proliferation and Mature Islet β-Cell Function. Diabetes 2019, 68, 1806–1818. [Google Scholar] [CrossRef] [PubMed]
  200. Davidson, R.K.; Kanojia, S.; Wu, W.; Kono, T.; Xu, J.; Osmulski, M.; Bone, R.N.; Casey, N.; Evans-Molina, C.; Sims, E.K.; et al. The Chd4 Helicase Regulates Chromatin Accessibility and Gene Expression Critical for β-Cell Function In Vivo. Diabetes 2023, 72, 746–757. [Google Scholar] [CrossRef]
  201. Davidson, R.K.; Weaver, S.A.; Casey, N.; Kanojia, S.; Hogarth, E.; Aguirre, R.S.; Sims, E.K.; Evans-Molina, C.; Spaeth, J.M. The Chd4 subunit of the NuRD complex regulates Pdx1-controlled genes involved in β-cell function. J. Mol. Endocrinol. 2022, 69, 329–341. [Google Scholar] [CrossRef] [PubMed]
  202. Soltani, A.; Jafarian, A.; Allameh, A. The Predominant microRNAs in β-cell Clusters for Insulin Regulation and Diabetic Control. Curr. Drug Targets 2020, 21, 722–734. [Google Scholar] [CrossRef] [PubMed]
  203. Sebastiani, G.; Grieco, G.E.; Brusco, N.; Ventriglia, G.; Formichi, C.; Marselli, L.; Marchetti, P.; Dotta, F. MicroRNA Expression Analysis of In Vitro Dedifferentiated Human Pancreatic Islet Cells Reveals the Activation of the Pluripotency-Related MicroRNA Cluster miR-302s. Int. J. Mol. Sci. 2018, 19, 1170. [Google Scholar] [CrossRef]
  204. Melnik, B.C.; Schmitz, G. Milk Exosomal microRNAs: Postnatal Promoters of β Cell Proliferation but Potential Inducers of β Cell De-Differentiation in Adult Life. Int. J. Mol. Sci. 2022, 23, 11503. [Google Scholar] [CrossRef]
  205. Melnik, B.C. Milk exosomal miRNAs: Potential drivers of AMPK-to-mTORC1 switching in β-cell de-differentiation of type 2 diabetes mellitus. Nutr. Metab. 2019, 16, 85. [Google Scholar] [CrossRef]
  206. Wang, Z.; Mohan, R.; Chen, X.; Matson, K.; Waugh, J.; Mao, Y.; Zhang, S.; Li, W.; Tang, X.; Satin, L.S.; et al. microRNA-483 Protects Pancreatic β-Cells by Targeting ALDH1A3. Endocrinology 2021, 162, bqab031. [Google Scholar] [CrossRef]
  207. Mohan, R.; Mao, Y.; Zhang, S.; Zhang, Y.-W.; Xu, C.-R.; Gradwohl, G.; Tang, X. Differentially Expressed MicroRNA-483 Confers Distinct Functions in Pancreatic β- and α-Cells. J. Biol. Chem. 2015, 290, 19955–19966. [Google Scholar] [CrossRef]
  208. Gannon, M. Micro-RNA-mediated Maintenance of Beta-cell Identity Reveals Targets for Reversing Beta-cell Dedifferentiation. Endocrinology 2021, 162, bqab067. [Google Scholar] [CrossRef]
  209. Sun, Y.; Zhou, Y.; Shi, Y.; Zhang, Y.; Liu, K.; Liang, R.; Sun, P.; Chang, X.; Tang, W.; Zhang, Y.; et al. Expression of miRNA-29 in Pancreatic β Cells Promotes Inflammation and Diabetes via TRAF3. Cell Rep. 2021, 34, 108576. [Google Scholar] [CrossRef] [PubMed]
  210. Li, J.; Zhang, Y.; Ye, Y.; Li, D.; Liu, Y.; Lee, E.; Zhang, M.; Dai, X.; Zhang, X.; Wang, S. Pancreatic β cells control glucose homeostasis via the secretion of exosomal miR-29 family. J. Extracell. Vesicles 2021, 10, e12055. [Google Scholar] [CrossRef]
  211. Lang, H.; Lin, N.; Chen, X.; Xiang, J.; Zhang, X.; Kang, C. Repressing miR-23a promotes the transdifferentiation of pancreatic α cells to β cells via negatively regulating the expression of SDF-1α. PLoS ONE 2024, 19, e0299821. [Google Scholar] [CrossRef] [PubMed]
  212. Zhu, Y.; Sun, Y.; Zhou, Y.; Zhang, Y.; Zhang, T.; Li, Y.; You, W.; Chang, X.; Yuan, L.; Han, X. MicroRNA-24 promotes pancreatic beta cells toward dedifferentiation to avoid endoplasmic reticulum stress-induced apoptosis. J. Mol. Cell Biol. 2019, 11, 747–760. [Google Scholar] [CrossRef] [PubMed]
  213. de Jesus, D.S.; Mak, T.C.; Wang, Y.-F.; von Ohlen, Y.; Bai, Y.; Kane, E.; Chabosseau, P.; Chahrour, C.M.; Distaso, W.; Salem, V. Dysregulation of the Pdx1/Ovol2/Zeb2 axis in dedifferentiated β-cells triggers the induction of genes associated with epithelial–mesenchymal transition in diabetes. Mol. Metab. 2021, 53, 101248. [Google Scholar] [CrossRef] [PubMed]
  214. Liu, H.; Zhang, D.; Zhou, Y.; Cui, S. MicroRNA-7a inhibits Isl1 expression to regulate insulin secretion by targeting Raf1 and Mapkap1 in NIT-1 cells. Vitr. Cell. Dev. Biol. Anim. 2021, 57, 817–824. [Google Scholar] [CrossRef] [PubMed]
  215. Shen, Z.; Yu, Y.; Yang, Y.; Xiao, X.; Sun, T.; Chang, X.; Tang, W.; Zhu, Y.; Han, X. miR-25 and miR-92b regulate insulin biosynthesis and pancreatic β-cell apoptosis. Endocrine 2022, 76, 526–535. [Google Scholar] [CrossRef] [PubMed]
  216. Hu, D.; Wang, Y.; Zhang, H.; Kong, D. Identification of miR-9 as a negative factor of insulin secretion from beta cells. J. Physiol. Biochem. 2018, 74, 291–299. [Google Scholar] [CrossRef] [PubMed]
  217. Li, L.; Wang, S.; Li, H.; Wan, J.; Zhou, Q.; Zhou, Y.; Zhang, C. microRNA-96 protects pancreatic β-cell function by targeting PAK1 in gestational diabetes mellitus. Biofactors 2018, 44, 539–547. [Google Scholar] [CrossRef] [PubMed]
  218. Feng, Y.; Qu, X.; Chen, Y.; Feng, Q.; Zhang, Y.; Hu, J.; Li, X. MicroRNA-33a-5p sponges to inhibit pancreatic β-cell function in gestational diabetes mellitus LncRNA DANCR. Reprod. Biol. Endocrinol. 2020, 18, 1–9. [Google Scholar] [CrossRef]
  219. Li, Y.; Yang, Y.; Sun, Y.; He, L.; Liu, K.; Zhang, Y.; Zhou, Y.; Chang, X.; Liang, R.; Wang, S. The miR-203/ZBTB20/MAFA Axis Orchestrates Pancreatic β-Cell Maturation and Identity During Weaning and Diabetes. SSRN 2023, 4467623. [Google Scholar] [CrossRef]
  220. Zhang, F.; Ma, D.; Zhao, W.; Wang, D.; Liu, T.; Liu, Y.; Yang, Y.; Liu, Y.; Mu, J.; Li, B.; et al. Obesity-induced overexpression of miR-802 impairs insulin transcription and secretion. Nat. Commun. 2020, 11, 1822. [Google Scholar] [CrossRef]
  221. Mao, Y.; Schoenborn, J.; Wang, Z.; Chen, X.; Matson, K.; Mohan, R.; Zhang, S.; Tang, X.; Arunagiri, A.; Arvan, P.; et al. Transgenic overexpression of microRNA-30d in pancreatic beta-cells progressively regulates beta-cell function and identity. Sci. Rep. 2022, 12, 11969. [Google Scholar] [CrossRef] [PubMed]
  222. Bai, C.; Ren, Q.; Liu, H.; Li, X.; Guan, W.; Gao, Y. miR-212/132-enriched extracellular vesicles promote differentiation of induced pluripotent stem cells into pancreatic beta cells. Front. Cell Dev. Biol. 2021, 9, 673231. [Google Scholar] [CrossRef] [PubMed]
  223. Xu, Y.; Huang, Y.; Guo, Y.; Xiong, Y.; Zhu, S.; Xu, L.; Lu, J.; Li, X.; Wan, J.; Lu, Y.; et al. microRNA-690 regulates induced pluripotent stem cells (iPSCs) differentiation into insulin-producing cells by targeting Sox9. Stem Cell Res. Ther. 2019, 10, 1–13. [Google Scholar] [CrossRef] [PubMed]
  224. Zhang, Y.; Cong, R.; Lv, T.; Liu, K.; Chang, X.; Li, Y.; Han, X.; Zhu, Y. Islet-resident macrophage-derived miR-155 promotes β cell decompensation via targeting PDX1. iScience 2024, 27, 109540. [Google Scholar] [CrossRef] [PubMed]
  225. Sun, Y.; Zhou, S.; Shi, Y.; Zhou, Y.; Zhang, Y.; Liu, K.; Zhu, Y.; Han, X. Inhibition of miR-153, an IL-1β-responsive miRNA, prevents beta cell failure and inflammation-associated diabetes. Metabolism 2020, 111, 154335. [Google Scholar] [CrossRef] [PubMed]
  226. Du, H.; Yin, Z.; Zhao, Y.; Li, H.; Dai, B.; Fan, J.; He, M.; Nie, X.; Wang, C.-Y.; Wang, D.W.; et al. miR-320a induces pancreatic β cells dysfunction in diabetes by inhibiting MafF. Mol. Ther. Nucleic Acids 2021, 26, 444–457. [Google Scholar] [CrossRef] [PubMed]
  227. Wan, S.; Zhang, J.; Chen, X.; Lang, J.; Li, L.; Chen, F.; Tian, L.; Meng, Y.; Yu, X. MicroRNA-17-92 Regulates Beta-Cell Restoration After Streptozotocin Treatment. Front. Endocrinol. 2020, 11, 9. [Google Scholar] [CrossRef] [PubMed]
  228. Ibrahim, S.M.; Mirmira, R.; Anderson, R.; Sims, E. MiR-21 Contributes to Cytokine-Induced Beta Cell Dysfunction via Inhibition of mRNAs Regulating Beta Cell Identity. FASEB J. 2019, 33, 694.13. [Google Scholar] [CrossRef]
  229. Ibrahim, S. miR-21 Exacerbates Cytokine Induced Beta Cell Dysfunction via Inhibition of mRNAs Regulating Beta Cell Identity. Ph.D. Thesis, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA, 2020. [Google Scholar]
  230. Grieco, G.E.; Brusco, N.; Fignani, D.; Nigi, L.; Formichi, C.; Licata, G.; Marselli, L.; Marchetti, P.; Salvini, L.; Tinti, L.; et al. Reduced miR-184-3p expression protects pancreatic β-cells from lipotoxic and proinflammatory apoptosis in type 2 diabetes via CRTC1 upregulation. Cell Death Discov. 2022, 8, 340. [Google Scholar] [CrossRef]
  231. Sato-Kunisada, R.; Yoshida, N.; Nakamura, S.; Uchiyama, H.; Matsumoto, H. Enhanced Expression of miR-199b-5p Promotes Proliferation of Pancreatic.β-Cells by Down-Regulation of MLK3. MicroRNA 2016, 5, 57–65. [Google Scholar] [CrossRef]
  232. Qian, B.; Yang, Y.; Tang, N.; Wang, J.; Sun, P.; Yang, N.; Chen, F.; Wu, T.; Sun, T.; Li, Y.; et al. M1 macrophage-derived exosomes impair beta cell insulin secretion via miR-212-5p by targeting SIRT2 and inhibiting Akt/GSK-3β/β-catenin pathway in mice. Diabetologia 2021, 64, 2037–2051. [Google Scholar] [CrossRef] [PubMed]
  233. Eliasson, L. The small RNA miR-375—A pancreatic islet abundant miRNA with multiple roles in endocrine beta cell function. Mol. Cell. Endocrinol. 2017, 456, 95–101. [Google Scholar] [CrossRef]
  234. Li, Y.; Xu, X.; Liang, Y.; Liu, S.; Xiao, H.; Li, F.; Cheng, H.; Fu, Z. miR-375 enhances palmitate-induced lipoapoptosis in insulin-secreting NIT-1 cells by repressing myotrophin (V1) protein expression. Int. J. Clin. Exp. Pathol. 2010, 3, 254–264. [Google Scholar]
  235. Guo, C.; Sun, Y.-Q.; Li, Q.; Zhang, J.-C. MiR-7, miR-9 and miR-375 contribute to effect of Exendin-4 on pancreatic β-cells in high-fat-diet-fed mice. Clin. Investig. Med. 2018, 41, E16–E24. [Google Scholar] [CrossRef]
  236. Lin, X.; Cheng, L.; Wan, Y.; Yan, Y.; Zhang, Z.; Li, X.; Wu, J.; Wang, X.; Xu, M. Ang II Controls the Expression of Mapkap1 by miR-375 and Affects the Function of Islet β Cells. Endocrine, Metab. Immune Disord. Drug Targets 2023, 23, 1186–1200. [Google Scholar] [CrossRef]
  237. Lu, Z.; Wang, J.; Wang, X.; Li, Z.; Niu, D.; Wang, M.; Xiang, J.; Yue, Y.; Xia, Y.; Li, X. miR-375 Promotes Pancreatic Differentiation In Vitro by Affecting Different Target Genes at Different Stages. Stem Cells Int. 2021, 2021, 1–13. [Google Scholar] [CrossRef] [PubMed]
  238. Keller, D.M.; Perez, I.G. Dual regulation of miR-375 and CREM genes in pancreatic beta cells. Islets 2022, 14, 139–148. [Google Scholar] [CrossRef] [PubMed]
  239. Nathan, G.; Kredo-Russo, S.; Geiger, T.; Lenz, A.; Kaspi, H.; Hornstein, E.; Efrat, S. MiR-375 Promotes Redifferentiation of Adult Human β Cells Expanded In Vitro. PLoS ONE 2015, 10, e0122108. [Google Scholar] [CrossRef]
  240. Title, A.C.; Silva, P.N.; Godbersen, S.; Hasenöhrl, L.; Stoffel, M. The miR-200–Zeb1 axis regulates key aspects of β-cell function and survival in vivo. Mol. Metab. 2021, 53, 101267. [Google Scholar] [CrossRef]
  241. He, Q.; Song, J.; Cui, C.; Wang, J.; Hu, H.; Guo, X.; Yang, M.; Wang, L.; Yan, F.; Liang, K. Mesenchymal stem cell-derived exosomal miR-146a reverses diabetic β-cell dedifferentiation. Stem Cell Res. Ther. 2021, 12, 1–16. [Google Scholar] [CrossRef]
  242. Xu, Y.; Tang, Z.; Dai, H.; Hou, J.; Li, F.; Tang, Z.; Zhang, D. MiR-195 promotes pancreatic β-cell dedifferentiation by targeting Mfn2 and impairing Pi3k/Akt signaling in type 2 diabetes. Obesity 2022, 30, 447–459. [Google Scholar] [CrossRef]
  243. Sisino, G.; Zhou, A.-X.; Dahr, N.; Sabirsh, A.; Soundarapandian, M.M.; Perera, R.; Larsson-Lekholm, E.; Magnone, M.C.; Althage, M.; Tyrberg, B. Long noncoding RNAs are dynamically regulated during β-cell mass expansion in mouse pregnancy and control β-cell proliferation in vitro. PLoS ONE 2017, 12, e0182371. [Google Scholar] [CrossRef]
  244. López–Noriega, L.; Rutter, G.A. Long Non-Coding RNAs as Key Modulators of Pancreatic β-Cell Mass and Function. Front. Endocrinol. 2021, 11, 610213. [Google Scholar] [CrossRef] [PubMed]
  245. Ding, G.-L.; Wang, F.-F.; Shu, J.; Tian, S.; Jiang, Y.; Zhang, D.; Wang, N.; Luo, Q.; Zhang, Y.; Jin, F. Transgenerational glucose intolerance with Igf2/H19 epigenetic alterations in mouse islet induced by intrauterine hyperglycemia. Diabetes 2012, 61, 1133–1142. [Google Scholar] [CrossRef]
  246. Sanchez-Parra, C.; Jacovetti, C.; Dumortier, O.; Lee, K.; Peyot, M.-L.; Guay, C.; Prentki, M.; Laybutt, D.R.; Van Obberghen, E.; Regazzi, R. Contribution of the Long Noncoding RNA H19 to β-Cell Mass Expansion in Neonatal and Adult Rodents. Diabetes 2018, 67, 2254–2267. [Google Scholar] [CrossRef]
  247. Alfaifi, M.; Verma, A.K.; Alshahrani, M.Y.; Joshi, P.C.; Alkhathami, A.G.; Ahmad, I.; Hakami, A.R.; Beg, M.M.A. Assessment of Cell-Free Long Non-Coding RNA-H19 and miRNA-29a, miRNA-29b Expression and Severity of Diabetes. Diabetes Metab. Syndr. Obes. Targets Ther. 2020, 13, 3727–3737. [Google Scholar] [CrossRef] [PubMed]
  248. Zhou, A.-X.; Mondal, T.; Tabish, A.M.; Abadpour, S.; Ericson, E.; Smith, D.M.; Knöll, R.; Scholz, H.; Kanduri, C.; Tyrberg, B.; et al. The long noncoding RNA TUNAR modulates Wnt signaling and regulates human β-cell proliferation. Am. J. Physiol. Metab. 2021, 320, E846–E857. [Google Scholar] [CrossRef]
  249. You, L.; Wang, N.; Yin, D.; Wang, L.; Jin, F.; Zhu, Y.; Yuan, Q.; De, W. Downregulation of Long Noncoding RNA Meg3 Affects Insulin Synthesis and Secretion in Mouse Pancreatic Beta Cells. J. Cell. Physiol. 2015, 231, 852–862. [Google Scholar] [CrossRef] [PubMed]
  250. Wang, N.; Zhu, Y.; Xie, M.; Wang, L.; Jin, F.; Li, Y.; Yuan, Q.; De, W. Long Noncoding RNA Meg3 Regulates Mafa Expression in Mouse Beta Cells by Inactivating Rad21, Smc3 or Sin3α. Cell. Physiol. Biochem. 2018, 45, 2031–2043. [Google Scholar] [CrossRef] [PubMed]
  251. Zhang, F.; Yang, Y.; Chen, X.; Liu, Y.; Hu, Q.; Huang, B.; Liu, Y.; Pan, Y.; Zhang, Y.; Liu, D.; et al. The long non-coding RNA βFaar regulates islet β-cell function and survival during obesity in mice. Nat. Commun. 2021, 12, 3997. [Google Scholar] [CrossRef]
  252. Rashidmayvan, M.; Sahebi, R.; Ghayour-Mobarhan, M. Long non-coding RNAs: A valuable biomarker for metabolic syndrome. Mol. Genet. Genom. 2022, 297, 1169–1183. [Google Scholar] [CrossRef] [PubMed]
  253. Huang, Y.; Xu, Y.; Lu, Y.; Zhu, S.; Guo, Y.; Sun, C.; Xu, L.; Chen, X.; Zhao, Y.; Yu, B. lncRNA Gm10451 regulates PTIP to facilitate iPSCs-derived β-like cell differentiation by targeting miR-338-3p as a ceRNA. Biomaterials 2019, 216, 119266. [Google Scholar] [CrossRef] [PubMed]
  254. Zhang, F.F.; Liu, Y.H.; Wang, D.W.; Liu, T.S.; Yang, Y.; Guo, J.M.; Pan, Y.; Zhang, Y.F.; Du, H.; Li, L.; et al. Obesity-induced reduced expression of the lncRNA ROIT impairs insulin transcription by downregulation of Nkx6.1 methylation. Diabetologia 2020, 63, 811–824. [Google Scholar] [CrossRef] [PubMed]
  255. Ruan, Y.; Lin, N.; Ma, Q.; Chen, R.; Zhang, Z.; Wen, W.; Chen, H.; Sun, J. Circulating LncRNAs Analysis in Patients with Type 2 Diabetes Reveals Novel Genes Influencing Glucose Metabolism and Islet β-Cell Function. Cell. Physiol. Biochem. 2018, 46, 335–350. [Google Scholar] [CrossRef] [PubMed]
  256. Cao, Z.; Yao, F.; Lang, Y.; Feng, X. Elevated Circulating LINC-P21 Serves as a Diagnostic Biomarker of Type 2 Diabetes Mellitus and Regulates Pancreatic β-cell Function by Sponging miR-766-3p to Upregulate NR3C2. Exp. Clin. Endocrinol. Diabetes 2020, 130, 156–164. [Google Scholar] [CrossRef] [PubMed]
  257. Wang, L.; Hu, L.; Wang, X.; Geng, Z.; Wan, M.; Hao, J.; Liu, H.; Fan, Y.; Xu, T.; Li, Z. Long non-coding RNA LncCplx2 regulates glucose homeostasis and pancreatic β cell function. Mol. Metab. 2024, 80, 101878. [Google Scholar] [CrossRef] [PubMed]
  258. Ding, H.; Wang, F.; Shi, X.; Ma, H.; Du, Y.; Hou, L.; Xing, N. LncRNA MALAT1 induces the dysfunction of β cells via reducing the histone acetylation of the PDX-1 promoter in type 1 diabetes. Exp. Mol. Pathol. 2020, 114, 104432. [Google Scholar] [CrossRef] [PubMed]
  259. Luo, Y.; Guo, J.; Xu, P.; Gui, R. Long non-coding RNA GAS5 maintains insulin secretion by regulating multiple miRNAs in INS-1 832/13 cells. Front. Mol. Biosci. 2020, 7, 559267. [Google Scholar] [CrossRef] [PubMed]
  260. Jin, F.; Wang, N.; Zhu, Y.; You, L.; Wang, L.; De, W.; Tang, W. Downregulation of Long Noncoding RNA Gas5 Affects Cell Cycle and Insulin Secretion in Mouse Pancreatic β Cells. Cell. Physiol. Biochem. 2017, 43, 2062–2073. [Google Scholar] [CrossRef]
  261. Liu, L.; Li, Y.; Zhang, X. LncRNA LINC01018 Screens Type 2 Diabetes Mellitus and Regulates β Cell Function Through Modulating miR-499a-5p. Horm. Metab. Res. 2023, 55, 642–648. [Google Scholar] [CrossRef]
  262. Pardo, C.A.C.; Massoz, L.; Dupont, M.A.; Bergemann, D.; Bourdouxhe, J.; Lavergne, A.; Tarifeño-Saldivia, E.; Helker, C.S.; Stainier, D.Y.; Peers, B.; et al. A δ-cell subpopulation with a pro-β-cell identity contributes to efficient age-independent recovery in a zebrafish model of diabetes. eLife 2022, 11, e67576. [Google Scholar] [CrossRef] [PubMed]
  263. Singh, S.P.; Chawla, P.; Hnatiuk, A.; Kamel, M.; Silva, L.D.; Spanjaard, B.; Eski, S.E.; Janjuha, S.; Olivares-Chauvet, P.; Kayisoglu, O. A single-cell atlas of de novo β-cell regeneration reveals the contribution of hybrid β/δ-cells to diabetes recovery in zebrafish. Development 2022, 149, dev199853. [Google Scholar] [CrossRef] [PubMed]
  264. Zhao, H.; Huang, X.; Liu, Z.; Pu, W.; Lv, Z.; He, L.; Li, Y.; Zhou, Q.; Lui, K.O.; Zhou, B. Pre-existing beta cells but not progenitors contribute to new beta cells in the adult pancreas. Nat. Metab. 2021, 3, 352–365. [Google Scholar] [CrossRef]
  265. Piran, R.; Lee, S.-H.; Li, C.-R.; Charbono, A.; Bradley, L.M.; Levine, F. Pharmacological induction of pancreatic islet cell transdifferentiation: Relevance to type I diabetes. Cell Death Dis. 2014, 5, e1357. [Google Scholar] [CrossRef] [PubMed]
  266. Chera, S.; Baronnier, D.; Ghila, L.; Cigliola, V.; Jensen, J.N.; Gu, G.; Furuyama, K.; Thorel, F.; Gribble, F.M.; Reimann, F.; et al. Diabetes recovery by age-dependent conversion of pancreatic δ-cells into insulin producers. Nature 2014, 514, 503–507. [Google Scholar] [CrossRef]
  267. Bru-Tari, E.; Cobo-Vuilleumier, N.; Alonso-Magdalena, P.; Dos Santos, R.S.; Marroqui, L.; Nadal, A.; Gauthier, B.R.; Quesada, I. Pancreatic alpha-cell mass in the early-onset and advanced stage of a mouse model of experimental autoimmune diabetes. Sci. Rep. 2019, 9, 9515. [Google Scholar] [CrossRef] [PubMed]
  268. Hang, Y.; Stein, R. MafA and MafB activity in pancreatic β cells. Trends Endocrinol. Metab. 2011, 22, 364–373. [Google Scholar] [CrossRef] [PubMed]
  269. Furuyama, K.; Chera, S.; van Gurp, L.; Oropeza, D.; Ghila, L.; Damond, N.; Vethe, H.; Paulo, J.A.; Joosten, A.M.; Berney, T. Diabetes relief in mice by glucose-sensing insulin-secreting human α-cells. Nature 2019, 567, 43–48. [Google Scholar] [CrossRef] [PubMed]
  270. Yu, J.; Ma, J.; Li, Y.; Zhou, Y.; Luo, L.; Yang, Y. Pax4-Ghrelin mediates the conversion of pancreatic ε-cells to β-cells after extreme β-cell loss in zebrafish. Development 2023, 150, dev201306. [Google Scholar] [CrossRef]
  271. Sasaki, S.; Lee, M.Y.Y.; Wakabayashi, Y.; Suzuki, L.; Winata, H.; Himuro, M.; Matsuoka, T.-A.; Shimomura, I.; Watada, H.; Lynn, F.C.; et al. Spatial and transcriptional heterogeneity of pancreatic beta cell neogenesis revealed by a time-resolved reporter system. Diabetologia 2022, 65, 811–828. [Google Scholar] [CrossRef]
  272. Huang, H.; Yang, K.; Wang, R.; Han, W.H.; Kuny, S.; Zelmanovitz, P.H.; Sauvé, Y.; Chan, C.B. β-Cell compensation concomitant with adaptive endoplasmic reticulum stress and β-cell neogenesis in a diet-induced type 2 diabetes model. Appl. Physiol. Nutr. Metab. 2019, 44, 1355–1366. [Google Scholar] [CrossRef]
  273. Gribben, C.; Lambert, C.; Messal, H.A.; Hubber, E.-L.; Rackham, C.; Evans, I.; Heimberg, H.; Jones, P.; Sancho, R.; Behrens, A. Ductal Ngn3-expressing progenitors contribute to adult β cell neogenesis in the pancreas. Cell Stem Cell 2021, 28, 2000–2008.e4. [Google Scholar] [CrossRef] [PubMed]
  274. Karampelias, C.; Watt, K.; Mattsson, C.L.; Ruiz, F.; Rezanejad, H.; Mi, J.; Liu, X.; Chu, L.; Locasale, J.W.; Korbutt, G.S.; et al. MNK2 deficiency potentiates β-cell regeneration via translational regulation. Nat. Chem. Biol. 2022, 18, 942–953. [Google Scholar] [CrossRef] [PubMed]
  275. Zhang, J.; An, H.; Ni, K.; Chen, B.; Li, H.; Li, Y.; Sheng, G.; Zhou, C.; Xie, M.; Chen, S.; et al. Glutathione prevents chronic oscillating glucose intake-induced β-cell dedifferentiation and failure. Cell Death Dis. 2019, 10, 321. [Google Scholar] [CrossRef]
  276. John, A.N.; Iqbal, Z.; Colley, S.; Morahan, G.; Makishima, M.; Jiang, F.-X. Vitamin D receptor-targeted treatment to prevent pathological dedifferentiation of pancreatic β cells under hyperglycaemic stress. Diabetes Metab. 2018, 44, 269–280. [Google Scholar] [CrossRef]
  277. Xing, C.; Tang, M.; Yang, J.; Wang, S.; Xu, Q.; Feng, W.; Mu, Y.; Li, F.; Zhao, A.Z. Eicosapentaenoic acid metabolites promotes the trans-differentiation of pancreatic α cells to β cells. Biochem. Pharmacol. 2023, 216, 115775. [Google Scholar] [CrossRef] [PubMed]
  278. Karampelias, C.; Rezanejad, H.; Rosko, M.; Duan, L.; Lu, J.; Pazzagli, L.; Bertolino, P.; Cesta, C.E.; Liu, X.; Korbutt, G.S.; et al. Reinforcing one-carbon metabolism via folic acid/Folr1 promotes β-cell differentiation. Nat. Commun. 2021, 12, 3362. [Google Scholar] [CrossRef]
  279. Kaya-Dagistanli, F.; Ozturk, M. Transdifferentiation of both intra- and extra-islet cells into beta cells in nicotinamide treated neonatal diabetic rats: An in situ hybridization and double immunohistochemical study. Acta Histochem. 2020, 122, 151612. [Google Scholar] [CrossRef] [PubMed]
  280. Cristelo, C.; Nunes, R.; Pinto, S.; Marques, J.M.; Gama, F.M.; Sarmento, B. Targeting β Cells with Cathelicidin Nanomedicines Improves Insulin Function and Pancreas Regeneration in Type 1 Diabetic Rats. ACS Pharmacol. Transl. Sci. 2023, 6, 1544–1560. [Google Scholar] [CrossRef]
  281. Patel, S.; Yan, Z.; Remedi, M.S. Intermittent fasting protects β-cell identity and function in a type-2 diabetes model. Metabolism 2024, 153, 155813. [Google Scholar] [CrossRef]
  282. Li, C.; Zhu, J.; Wei, S.; Ye, X.; Yang, L.; Wang, Z.; Chen, Y. Intermittent protein restriction improves glucose homeostasis in Zucker diabetic fatty rats and single-cell sequencing reveals distinct changes in β cells. J. Nutr. Biochem. 2023, 114, 109275. [Google Scholar] [CrossRef]
  283. Zhang, M.; Chen, Y.; Sun, S.; Zhang, S.; Yuan, L.; Xu, Y.; Li, X.; Chen, G.; Wei, X.; Liu, C. Ketogenic diet alleviates β-cell dedifferentiation but aggravates hepatic lipid accumulation in db/db mice. Nutrition 2024, 119, 112284. [Google Scholar] [CrossRef] [PubMed]
  284. Luo, W.; Zhou, J.; Yang, X.; Wu, R.; Liu, H.; Shao, H.; Huang, B.; Kang, X.; Yang, L.; Liu, D. A Chinese medical nutrition therapy diet accompanied by intermittent energy restriction alleviates type 2 diabetes by enhancing pancreatic islet function and regulating gut microbiota composition. Food Res. Int. 2022, 161, 111744. [Google Scholar] [CrossRef]
  285. Lv, C.; Sun, Y.; Zhang, Z.Y.; Aboelela, Z.; Qiu, X.; Meng, Z.-X. β-cell dynamics in type 2 diabetes and in dietary and exercise interventions. J. Mol. Cell Biol. 2022, 14, mjac046. [Google Scholar] [CrossRef]
  286. Lyngbaek, M.P.P.; Legaard, G.E.; Bennetsen, S.L.; Feineis, C.S.; Rasmussen, V.; Moegelberg, N.; Brinkløv, C.F.; Nielsen, A.B.; Kofoed, K.S.; Lauridsen, C.A.; et al. The effects of different doses of exercise on pancreatic β-cell function in patients with newly diagnosed type 2 diabetes: Study protocol for and rationale behind the "DOSE-EX" multi-arm parallel-group randomised clinical trial. Trials 2021, 22, 244. [Google Scholar] [CrossRef]
  287. Chow, L.S.; Gerszten, R.E.; Taylor, J.M.; Pedersen, B.K.; Van Praag, H.; Trappe, S.; Febbraio, M.A.; Galis, Z.S.; Gao, Y.; Haus, J.M. Exerkines in health, resilience and disease. Nat. Rev. Endocrinol. 2022, 18, 273–289. [Google Scholar] [CrossRef] [PubMed]
  288. Wedell-Neergaard, A.S.; Lehrskov, L.L.; Christensen, R.H.; Legaard, G.E.; Dorph, E.; Larsen, M.K.; Launbo, N.; Fagerlind, S.R.; Seide, S.K.; Nymand, S.; et al. Exercise-induced changes in visceral adipose tissue mass are regulated by IL-6 signaling: A randomized controlled trial. Yearb. Paediatr. Endocrinol. 2019, 29, 844–855. [Google Scholar] [CrossRef]
  289. Lehrskov, L.L.; Lyngbaek, M.P.; Soederlund, L.; Legaard, G.E.; Ehses, J.A.; Heywood, S.E.; Albrechtsen, N.J.W.; Holst, J.J.; Karstoft, K.; Pedersen, B.K.; et al. Interleukin-6 Delays Gastric Emptying in Humans with Direct Effects on Glycemic Control. Cell Metab. 2018, 27, 1201–1211.e3. [Google Scholar] [CrossRef]
  290. Rutti, S.; Dusaulcy, R.; Hansen, J.S.; Howald, C.; Dermitzakis, E.T.; Pedersen, B.K.; Pinget, M.; Plomgaard, P.; Bouzakri, K. Angiogenin and Osteoprotegerin are type II muscle specific myokines protecting pancreatic beta-cells against proinflammatory cytokines. Sci. Rep. 2018, 8, 10072. [Google Scholar] [CrossRef] [PubMed]
  291. Becic, T.; Studenik, C.; Hoffmann, G. Exercise Increases Adiponectin and Reduces Leptin Levels in Prediabetic and Diabetic Individuals: Systematic Review and Meta-Analysis of Randomized Controlled Trials. Med. Sci. 2018, 6, 97. [Google Scholar] [CrossRef]
  292. Ohashi, K.; Parker, J.L.; Ouchi, N.; Higuchi, A.; Vita, J.A.; Gokce, N.; Pedersen, A.A.; Kalthoff, C.; Tullin, S.; Sams, A.; et al. Adiponectin Promotes Macrophage Polarization toward an Anti-inflammatory Phenotype. J. Biol. Chem. 2010, 285, 6153–6160. [Google Scholar] [CrossRef] [PubMed]
  293. Fulgenzi, G.; Hong, Z.; Tomassoni-Ardori, F.; Barella, L.F.; Becker, J.; Barrick, C.; Swing, D.; Yanpallewar, S.; Croix, B.S.; Wess, J.; et al. Novel metabolic role for BDNF in pancreatic β-cell insulin secretion. Nat. Commun. 2020, 11, 1950. [Google Scholar] [CrossRef] [PubMed]
  294. Strömberg, A.; Olsson, K.; Dijksterhuis, J.P.; Rullman, E.; Schulte, G.; Gustafsson, T. CX3CL1—A macrophage chemoattractant induced by a single bout of exercise in human skeletal muscle. Am. J. Physiol. Integr. Comp. Physiol. 2016, 310, R297–R304. [Google Scholar] [CrossRef] [PubMed]
  295. Liu, S.; Du, F.; Li, X.; Wang, M.; Duan, R.; Zhang, J.; Wu, Y.; Zhang, Q. Effects and underlying mechanisms of irisin on the proliferation and apoptosis of pancreatic β cells. PLoS ONE 2017, 12, e0175498. [Google Scholar] [CrossRef] [PubMed]
  296. Natalicchio, A.; Marrano, N.; Biondi, G.; Spagnuolo, R.; Labarbuta, R.; Porreca, I.; Cignarelli, A.; Bugliani, M.; Marchetti, P.; Perrini, S.; et al. The Myokine Irisin Is Released in Response to Saturated Fatty Acids and Promotes Pancreatic β-Cell Survival and Insulin Secretion. Diabetes 2017, 66, 2849–2856. [Google Scholar] [CrossRef] [PubMed]
  297. Wei, J.; Hanna, T.; Suda, N.; Karsenty, G.; Ducy, P. Osteocalcin Promotes β-Cell Proliferation During Development and Adulthood Through Gprc6a. Diabetes 2014, 63, 1021–1031. [Google Scholar] [CrossRef] [PubMed]
  298. Kover, K.; Yan, Y.; Tong, P.Y.; Watkins, D.; Li, X.; Tasch, J.; Hager, M.; Clements, M.; Moore, W.V. Osteocalcin protects pancreatic beta cell function and survival under high glucose conditions. Biochem. Biophys. Res. Commun. 2015, 462, 21–26. [Google Scholar] [CrossRef] [PubMed]
  299. Cui, X.; Feng, J.; Wei, T.; Zhang, L.; Lang, S.; Yang, K.; Yang, J.; Liu, J.; Sterr, M.; Lickert, H.; et al. Pancreatic alpha cell glucagon–liver FGF21 axis regulates beta cell regeneration in a mouse model of type 2 diabetes. Diabetologia 2022, 66, 535–550. [Google Scholar] [CrossRef] [PubMed]
  300. Davey, J.R.; Estevez, E.; Thomson, R.E.; Whitham, M.; Watt, K.I.; Hagg, A.; Qian, H.; Henstridge, D.C.; Ludlow, H.; Hedger, M.P.; et al. Intravascular Follistatin gene delivery improves glycemic control in a mouse model of type 2 diabetes. FASEB J. 2020, 34, 5697–5714. [Google Scholar] [CrossRef]
  301. Curran, M.; Drayson, M.T.; Andrews, R.C.; Zoppi, C.; Barlow, J.P.; Solomon, T.P.; Narendran, P. The benefits of physical exercise for the health of the pancreatic β-cell: A review of the evidence. Exp. Physiol. 2020, 105, 579–589. [Google Scholar] [CrossRef]
  302. Tanday, N.; Irwin, N.; Moffett, R.C.; Flatt, P.R.; O’Harte, F.P.M. Beneficial actions of a long-acting apelin analogue in diabetes are related to positive effects on islet cell turnover and transdifferentiation. Diabetes Obes. Metab. 2020, 22, 2468–2478. [Google Scholar] [CrossRef]
  303. Li, H.-Q.; Spitzer, N.C. Exercise enhances motor skill learning by neurotransmitter switching in the adult midbrain. Nat. Commun. 2020, 11, 2195. [Google Scholar] [CrossRef] [PubMed]
  304. Ben-Othman, N.; Vieira, A.; Courtney, M.; Record, F.; Gjernes, E.; Avolio, F.; Hadzic, B.; Druelle, N.; Napolitano, T.; Navarro-Sanz, S.; et al. Long-Term GABA Administration Induces Alpha Cell-Mediated Beta-like Cell Neogenesis. Cell 2016, 168, 73–85.e11. [Google Scholar] [CrossRef]
  305. Li, J.; Casteels, T.; Frogne, T.; Ingvorsen, C.; Honoré, C.; Courtney, M.; Huber, K.V.; Schmitner, N.; Kimmel, R.A.; Romanov, R.A.; et al. Artemisinins Target GABAA Receptor Signaling and Impair α Cell Identity. Cell 2016, 168, 86–100.e15. [Google Scholar] [CrossRef]
  306. Maykish, A.; Sikalidis, A.K. Utilization of Hydroxyl-Methyl Butyrate, Leucine, Glutamine and Arginine Supplementation in Nutritional Management of Sarcopenia—Implications and Clinical Considerations for Type 2 Diabetes Mellitus Risk Modulation. J. Pers. Med. 2020, 10, 19. [Google Scholar] [CrossRef]
  307. Tanday, N.; Flatt, P.R.; Irwin, N.; Moffett, R.C. Liraglutide and sitagliptin counter beta- to alpha-cell transdifferentiation in diabetes. J. Endocrinol. 2020, 245, 53–64. [Google Scholar] [CrossRef]
  308. Tanday, N.; Irwin, N.; Flatt, P.R.; Moffett, R.C. Dapagliflozin exerts positive effects on beta cells, decreases glucagon and does not alter beta-to alpha-cell transdifferentiation in mouse models of diabetes and insulin resistance. Biochem. Pharmacol. 2020, 177, 114009. [Google Scholar] [CrossRef]
  309. Takahashi, K.; Nakamura, A.; Miyoshi, H.; Nomoto, H.; Kitao, N.; Omori, K.; Yamamoto, K.; Cho, K.Y.; Terauchi, Y.; Atsumi, T. Effect of the sodium–glucose cotransporter 2 inhibitor luseogliflozin on pancreatic beta cell mass in db/db mice of different ages. Sci. Rep. 2018, 8, 6864. [Google Scholar] [CrossRef] [PubMed]
  310. Patel, R.; Parmar, N.; Rathwa, N.; Palit, S.P.; Li, Y.; Garcia-Ocaña, A.; Begum, R. A novel therapeutic combination of sitagliptin and melatonin regenerates pancreatic β-cells in mouse and human islets. Biochim. Biophys. Acta (BBA) Mol. Cell Res. 2022, 1869, 119263. [Google Scholar] [CrossRef]
  311. Moin, A.S.M.; Butler, A.E. Alterations in Beta Cell Identity in Type 1 and Type 2 Diabetes. Curr. Diabetes Rep. 2019, 19, 83. [Google Scholar] [CrossRef] [PubMed]
  312. Téllez, N.; Montanya, E. Gastrin induces ductal cell dedifferentiation and b-cell neogenesis after 90% pancreatectomy. J. Endocrinol. 2014, 223, 67–78. [Google Scholar] [CrossRef] [PubMed]
  313. Kucukbas, G.N.; Komuroglu, A.U.; Dirik, D.; Korpe, B.; Kose, C.; Karaaslan, O.; Dirik, Y.; Sahin, H.G. Maternal serum xenin-25 levels in gestational diabetes mellitus. Eur. Rev. Med. Pharmacol. Sci. 2023, 27, 9902–9907. [Google Scholar] [CrossRef] [PubMed]
  314. Michele, L.A.; Mantzouratou, P.; Cerullo, D.; Sangalli, F.; Corna, D.; Zoja, C.; Remuzzi, G.; Xinaris, C. Thyroid hormone signalling regulates beta-cell pathological growth, dedifferentiation and transdifferentiation in diabetic pancreas. Endocr. Abstr. 2023, 92, OP13-03. [Google Scholar] [CrossRef]
  315. Guo, D.; He, J.; Guo, H.; Song, G.; Peng, L.; An, M.; Wang, C. Angiotensin (1–7) reverses glucose-induced islet β cell dedifferentiation by Wnt/β-catenin/FoxO1 signalling pathway. Endokrynol. Pol. 2023, 74, 544–552. [Google Scholar] [CrossRef] [PubMed]
  316. Gómez-Banoy, N.; Guseh, J.S.; Li, G.; Rubio-Navarro, A.; Chen, T.; Poirier, B.; Putzel, G.; Rosselot, C.; Pabón, M.A.; Camporez, J.P.; et al. Adipsin preserves beta cells in diabetic mice and associates with protection from type 2 diabetes in humans. Nat. Med. 2019, 25, 1739–1747. [Google Scholar] [CrossRef] [PubMed]
  317. Lei, L.; Huan, Y.; Liu, Q.; Li, C.; Cao, H.; Ji, W.; Gao, X.; Fu, Y.; Li, P.; Zhang, R. Morus alba L.(Sangzhi) alkaloids promote insulin secretion, restore diabetic β-cell function by preventing dedifferentiation and apoptosis. Front. Pharmacol. 2022, 13, 841981. [Google Scholar] [CrossRef] [PubMed]
  318. Gojani, E.G.; Wang, B.; Li, D.-P.; Kovalchuk, O.; Kovalchuk, I. Anti-Inflammatory Properties of Eugenol in Lipopolysaccharide-Induced Macrophages and Its Role in Preventing β-Cell Dedifferentiation and Loss Induced by High Glucose-High Lipid Conditions. Molecules 2023, 28, 7619. [Google Scholar] [CrossRef] [PubMed]
  319. Yuan, Y.; Zhou, J.; Hu, R.; Zou, L.; Ji, L.; Jiang, G. Piperine protects against pancreatic β-cell dysfunction by alleviating macrophage inflammation in obese mice. Life Sci. 2021, 274, 119312. [Google Scholar] [CrossRef] [PubMed]
  320. Gojani, E.G.; Wang, B.; Li, D.-P.; Kovalchuk, O.; Kovalchuk, I. The Impact of Psilocybin on High Glucose/Lipid-Induced Changes in INS-1 Cell Viability and Dedifferentiation. Genes 2024, 15, 183. [Google Scholar] [CrossRef]
  321. Gojani, E.G.; Wang, B.; Li, D.-P.; Kovalchuk, O.; Kovalchuk, I. Effect of THC, CBD, THCV, CBC and CBN Cannabinoids on β-Cells Exposed to High Glucose—High Lipids. Preprints 2023, 2023090973. [Google Scholar] [CrossRef]
  322. Kannan, P.; Raghunathan, M.; Mohan, T.; Palanivelu, S.; Periandavan, K. Gymnemic Acid Ameliorates Pancreatic β-Cell Dysfunction by Modulating Pdx1 Expression: A Possible Strategy for β-Cell Regeneration. Tissue Eng. Regen. Med. 2022, 19, 603–616. [Google Scholar] [CrossRef]
  323. Moosavi, A.; Yazdanparast, R. Beta cell regeneration upon magainin and growth hormone treatment as a possible alternative to insulin therapy. FEBS Open Bio. 2023, 13, 447–458. [Google Scholar] [CrossRef] [PubMed]
  324. Zhong, Q.; Yin, J.; Wang, K.; Chen, X.; Wang, H.; Hu, X.; Wang, W.; Wang, L.; Bei, W.; Guo, J. FTZ promotes islet β-cell regeneration in T1DM mice via the regulation of nuclear proliferation factors. J. Ethnopharmacol. 2023, 315, 116564. [Google Scholar] [CrossRef]
  325. Martínez-Montoro, J.I.; Damas-Fuentes, M.; Fernández-García, J.C.; Tinahones, F.J. Role of the Gut Microbiome in Beta Cell and Adipose Tissue Crosstalk: A Review. Front. Endocrinol. 2022, 13, 869951. [Google Scholar] [CrossRef]
  326. Wang, S.; Yuan, M.; Zhang, L.; Zhu, K.; Sheng, C.; Zhou, F.; Xu, Z.; Liu, Q.; Liu, Y.; Lu, J.; et al. Sodium butyrate potentiates insulin secretion from rat islets at the expense of compromised expression of β cell identity genes. Cell Death Dis. 2022, 13, 67. [Google Scholar] [CrossRef] [PubMed]
  327. Pedersen, S.S.; Prause, M.; Williams, K.; Barrès, R.; Billestrup, N. Butyrate inhibits IL-1β-induced inflammatory gene expression by suppression of NF-κB activity in pancreatic beta cells. J. Biol. Chem. 2022, 298, 102312. [Google Scholar] [CrossRef]
  328. Vandenbempt, V.; Eski, S.E.; Brahma, M.K.; Li, A.; Negueruela, J.; Bruggeman, Y.; Demine, S.; Xiao, P.; Cardozo, A.K.; Baeyens, N.; et al. HAMSAB diet ameliorates dysfunctional signaling in pancreatic islets in autoimmune diabetes. iScience 2024, 27, 108694. [Google Scholar] [CrossRef] [PubMed]
  329. Liang, Y.; Widjaja, J.; Sun, J.; Li, M.; Qiao, Z.; Cao, T.; Wang, Y.; Zhang, X.; Zhang, Z.; Gu, Y.; et al. Bariatric surgery induces pancreatic cell transdifferentiation as indicated by single-cell transcriptomics in Zucker diabetic rats. J. Diabetes 2023. [Google Scholar] [CrossRef]
  330. Koduru, S.V.; Leberfinger, A.N.; Ozbolat, I.T.; Ravnic, D.J. Navigating the Genomic Landscape of Human Adipose Stem Cell-Derived β-Cells. Stem Cells Dev. 2021, 30, 1153–1170. [Google Scholar] [CrossRef]
  331. Kamal, M.M.; Ammar, R.A.; Kassem, D.H. Silencing of forkhead box protein O-1 (FOXO-1) enhances insulin-producing cell generation from adipose mesenchymal stem cells for diabetes therapy. Life Sci. 2024, 344, 122579. [Google Scholar] [CrossRef]
  332. Zaeifi, D.; Azarnia, M. Promoting β-cells function by the recapitulation of in vivo microenvironmental differentiation signals. Cell Tissue Res. 2023, 393, 37–46. [Google Scholar] [CrossRef] [PubMed]
  333. Zhu, S.; Russ, H.A.; Wang, X.; Zhang, M.; Ma, T.; Xu, T.; Tang, S.; Hebrok, M.; Ding, S. Human pancreatic beta-like cells converted from fibroblasts. Nat. Commun. 2016, 7, 10080. [Google Scholar] [CrossRef] [PubMed]
  334. Wang, L.; Liu, T.; Liang, R.; Wang, G.; Liu, Y.; Zou, J.; Liu, N.; Zhang, B.; Liu, Y.; Ding, X.; et al. Mesenchymal stem cells ameliorate β cell dysfunction of human type 2 diabetic islets by reversing β cell dedifferentiation. EBioMedicine 2020, 51, 102615. [Google Scholar] [CrossRef]
  335. Isildar, B.; Ozkan, S.; Ercin, M.; Gezginci-Oktayoglu, S.; Oncul, M.; Koyuturk, M. 2D and 3D cultured human umbilical cord-derived mesenchymal stem cell-conditioned medium has a dual effect in type 1 diabetes model in rats: Immunomodulation and beta-cell regeneration. Inflamm. Regen. 2022, 42, 55. [Google Scholar] [CrossRef] [PubMed]
  336. Goode, R.A.; Hum, J.M.; Kalwat, M.A. Therapeutic Strategies Targeting Pancreatic Islet β-Cell Proliferation, Regeneration, and Replacement. Endocrinology 2023, 164, bqac193. [Google Scholar] [CrossRef] [PubMed]
  337. Basile, G.; Qadir, M.; Mauvais-Jarvis, F.; Vetere, A.; Shoba, V.; Modell, A.; Pastori, R.; Russ, H.; Wagner, B.; Dominguez-Bendala, J. Emerging diabetes therapies: Bringing back the β-cells. Mol. Metab. 2022, 60, 101477. [Google Scholar] [CrossRef] [PubMed]
  338. Wang, P.; Karakose, E.; Argmann, C.; Wang, H.; Balev, M.; Brody, R.I.; Rivas, H.G.; Liu, X.; Wood, O.; Liu, H.; et al. Disrupting the DREAM complex enables proliferation of adult human pancreatic β cells. J. Clin. Investig. 2022, 132, e157086. [Google Scholar] [CrossRef] [PubMed]
  339. Barzowska, A.; Pucelik, B.; Pustelny, K.; Matsuda, A.; Martyniak, A.; Stępniewski, J.; Maksymiuk, A.; Dawidowski, M.; Rothweiler, U.; Dulak, J.; et al. DYRK1A Kinase Inhibitors Promote β-Cell Survival and Insulin Homeostasis. Cells 2021, 10, 2263. [Google Scholar] [CrossRef]
  340. Guo, Y.; Li, L.; Yao, Y.; Li, H. Regeneration of Pancreatic β-Cells for Diabetes Therapeutics by Natural DYRK1A Inhibitors. Metabolites 2022, 13, 51. [Google Scholar] [CrossRef]
  341. Czarna, A.; Wang, J.; Zelencova, D.; Liu, Y.; Deng, X.; Choi, H.G.; Zhang, T.; Zhou, W.; Chang, J.W.; Kildalsen, H.; et al. Novel Scaffolds for Dual Specificity Tyrosine-Phosphorylation-Regulated Kinase (DYRK1A) Inhibitors. J. Med. Chem. 2018, 61, 7560–7572. [Google Scholar] [CrossRef]
  342. Ackeifi, C.; Wang, P.; Karakose, E.; Fox, J.E.M.; González, B.J.; Liu, H.; Wilson, J.; Swartz, E.; Berrouet, C.; Li, Y.; et al. GLP-1 receptor agonists synergize with DYRK1A inhibitors to potentiate functional human β cell regeneration. Sci. Transl. Med. 2020, 12. [Google Scholar] [CrossRef] [PubMed]
  343. Kimani, C.N.; Reuter, H.; Kotzé, S.H.; Venter, P.; Ramharack, P.; Muller, C.J.F. Pancreatic beta cell regenerative potential of Zanthoxylum chalybeum Engl. Aqueous stem bark extract. J. Ethnopharmacol. 2024, 320, 117374. [Google Scholar] [CrossRef]
  344. Gu, L.; Cui, X.; Lang, S.; Wang, H.; Hong, T.; Wei, R. Glucagon receptor antagonism increases mouse pancreatic δ-cell mass through cell proliferation and duct-derived neogenesis. Biochem. Biophys. Res. Commun. 2019, 512, 864–870. [Google Scholar] [CrossRef] [PubMed]
  345. Cui, X.; Feng, J.; Wei, T.; Gu, L.; Wang, D.; Lang, S.; Yang, K.; Yang, J.; Yan, H.; Wei, R.; et al. Pro-α-cell-derived β-cells contribute to β-cell neogenesis induced by antagonistic glucagon receptor antibody in type 2 diabetic mice. iScience 2022, 25, 104567. [Google Scholar] [CrossRef]
  346. Wang, M.-Y.; Dean, E.D.; Quittner-Strom, E.; Zhu, Y.; Chowdhury, K.H.; Zhang, Z.; Zhao, S.; Li, N.; Ye, R.; Lee, Y.; et al. Glucagon blockade restores functional β-cell mass in type 1 diabetic mice and enhances function of human islets. Proc. Natl. Acad. Sci. USA 2021, 118, e2022142118. [Google Scholar] [CrossRef]
  347. Lang, S.; Yang, J.; Yang, K.; Gu, L.; Cui, X.; Wei, T.; Liu, J.; Le, Y.; Wang, H.; Wei, R.; et al. Glucagon receptor antagonist upregulates circulating GLP-1 level by promoting intestinal L-cell proliferation and GLP-1 production in type 2 diabetes. BMJ Open Diabetes Res. Care 2020, 8, e001025. [Google Scholar] [CrossRef]
  348. Lang, S.; Wei, R.; Wei, T.; Gu, L.; Feng, J.; Yan, H.; Yang, J.; Hong, T. Glucagon receptor antagonism promotes the production of gut proglucagon-derived peptides in diabetic mice. Peptides 2020, 131, 170349. [Google Scholar] [CrossRef]
  349. Zhang, Z.; Hu, Y.; Xu, N.; Zhou, W.; Yang, L.; Chen, R.; Yang, R.; Sun, J.; Chen, H. A New Way for Beta Cell Neogenesis: Transdifferentiation from Alpha Cells Induced by Glucagon-Like Peptide 1. J. Diabetes Res. 2019, 2019, 1–11. [Google Scholar] [CrossRef] [PubMed]
  350. Gu, L.; Wang, D.; Cui, X.; Wei, T.; Yang, K.; Yang, J.; Wei, R.; Hong, T. Combination of GLP-1 Receptor Activation and Glucagon Blockage Promotes Pancreatic β-Cell Regeneration In Situ in Type 1 Diabetic Mice. J. Diabetes Res. 2021, 2021, 1–10. [Google Scholar] [CrossRef]
  351. Wei, R.; Cui, X.; Feng, J.; Gu, L.; Lang, S.; Wei, T.; Yang, J.; Liu, J.; Le, Y.; Wang, H.; et al. Dapagliflozin promotes beta cell regeneration by inducing pancreatic endocrine cell phenotype conversion in type 2 diabetic mice. Metabolism 2020, 111, 154324. [Google Scholar] [CrossRef]
  352. Jiang, Y.; Yang, J.; Xia, L.; Wei, T.; Cui, X.; Wang, D.; Jin, Z.; Lin, X.; Li, F.; Yang, K. Gut microbiota–tryptophan metabolism–GLP-1 axis participates in β-cell regeneration induced by dapagliflozin. Diabetes 2024, 73, 926–940. [Google Scholar] [CrossRef] [PubMed]
  353. Sachs, S.; Bastidas-Ponce, A.; Tritschler, S.; Bakhti, M.; Böttcher, A.; Sánchez-Garrido, M.A.; Tarquis-Medina, M.; Kleinert, M.; Fischer, K.; Jall, S.; et al. Targeted pharmacological therapy restores β-cell function for diabetes remission. Nat. Metab. 2020, 2, 192–209. [Google Scholar] [CrossRef] [PubMed]
  354. Gu, L.; Cui, X.; Lin, X.; Yang, J.; Wei, R.; Hong, T.; Yang, K. γ-aminobutyric acid modulates α-cell hyperplasia but not β-cell regeneration induced by glucagon receptor antagonism in type 1 diabetic mice. Acta Diabetol. 2022, 60, 19–28. [Google Scholar] [CrossRef] [PubMed]
  355. Karacay, C.; Prietl, B.; Harer, C.; Ehall, B.; Haudum, C.W.; Bounab, K.; Franz, J.; Eisenberg, T.; Madeo, F.; Kolb, D.; et al. The effect of spermidine on autoimmunity and beta cell function in NOD mice. Sci. Rep. 2022, 12, 4502. [Google Scholar] [CrossRef] [PubMed]
  356. Sims, E.K.; Kulkarni, A.; Hull, A.; Woerner, S.E.; Cabrera, S.; Mastrandrea, L.D.; Hammoud, B.; Sarkar, S.; Nakayasu, E.S.; Mastracci, T.L.; et al. Inhibition of polyamine biosynthesis preserves β cell function in type 1 diabetes. Cell Rep. Med. 2023, 4, 101261. [Google Scholar] [CrossRef] [PubMed]
  357. Xu, J.; Jia, Y.-F.; Tapadar, S.; Weaver, J.D.; Raji, I.O.; Pithadia, D.J.; Javeed, N.; García, A.J.; Choi, D.-S.; Matveyenko, A.V. Inhibition of TBK1/IKKε promotes regeneration of pancreatic β-cells. Sci. Rep. 2018, 8, 15587. [Google Scholar] [CrossRef]
  358. Charbord, J.; Ren, L.; Sharma, R.B.; Johansson, A.; Ågren, R.; Chu, L.; Tworus, D.; Schulz, N.; Charbord, P.; Stewart, A.F.; et al. In vivo screen identifies a SIK inhibitor that induces β cell proliferation through a transient UPR. Nat. Metab. 2021, 3, 682–700. [Google Scholar] [CrossRef]
Figure 1. The mechanism of β-cell dedifferentiation and transdifferentiation. The process of β-cell dedifferentiation and transdifferentiation involves metabolic stresses such as glucotoxicity, lipotoxicity, viral infections, and aging. These stresses induce oxidative stress and inflammation, disrupting pathways such as ER stress, the mTOR pathway, and the UPS. This disruption affects the activities of transcription factors and epigenetic factors, including various miRNAs, leading to β-cell dedifferentiation and transdifferentiation. Abbreviations: endoplasmic reticulum: ER; ubiquitin–proteasome system: UPS; transcription factors: TFs. The information included in this figure is based on the research findings cited in this review paper.
Figure 1. The mechanism of β-cell dedifferentiation and transdifferentiation. The process of β-cell dedifferentiation and transdifferentiation involves metabolic stresses such as glucotoxicity, lipotoxicity, viral infections, and aging. These stresses induce oxidative stress and inflammation, disrupting pathways such as ER stress, the mTOR pathway, and the UPS. This disruption affects the activities of transcription factors and epigenetic factors, including various miRNAs, leading to β-cell dedifferentiation and transdifferentiation. Abbreviations: endoplasmic reticulum: ER; ubiquitin–proteasome system: UPS; transcription factors: TFs. The information included in this figure is based on the research findings cited in this review paper.
Cimb 46 00453 g001
Table 1. Important transcription factors involved in β-cell dedifferentiation, transdifferentiation, neogenesis, or proliferation (De/Tra/Neo/Pro).
Table 1. Important transcription factors involved in β-cell dedifferentiation, transdifferentiation, neogenesis, or proliferation (De/Tra/Neo/Pro).
Transcription FactorFull NameTargets GenesImpact on the GenesDe/Tra/Neo/ProImpact on β-Cell Plasticity/RegenerationReferences
Pax4Paired Box 4GCGRDeN[101,102,103,104]
ArxR
InsITraN
Bcl-xLI
GherlinRNeoP
SSTR
IAPPRProP
C-mycI
Nkx2.2NK2 homeobox 2InsIDeN[105,106,107,108,109,110]
Pax6I
Nkx6-1ITraN
MafAI
NeuroD1INeoP
ArxR
SSTRProNM
GherlinR
Mnx1 (also known as HLXB9 or HB9)Motor neuron and pancreas homeobox 1Nkx6.1IDeN[111,112,113,114,115]
TraN
InsINeoNM
ProP
FoxO1Forkhead box O1Ins1R/IDeN and P[116,117,118,119,120,121,122,123]
Pdx1RTraN and P
NeuroDI
Superoxide dismutase 1 (Sod1)INeoN
Catalase (Cat)I
MafAIProP
Glutathione peroxidase 1I
Pdx1Pancreatic and Duodenal Homeobox 1InsIDeN[124,125,126,127,128]
Glut2I
GckITraN
IAPPI
SSTINeoP
Pdx1I
Ngn3IProP
NFκBR
MafAI
MafAMusculoaponeurotic Fibrosarcoma Oncogene Family, Protein AInsIDeN[129,130,131,132,133,134,135]
Glut2I
GckITraN
Cacng4I
NanogRNeoP
Pdx1I
NeuroD1IProP
Nkx6.1I
Glis3I
Ngn3Neurogenin 3NeuroD1IDeN[99,136,137,138,139]
Pax4I
Pax6ITraN
Isl1I
Cdkn1aINeoP
Nkx2-2I
Insm1IProN
Kcnj11I
GckI
Slc30a8I
Slc18a2I
SOX9Sex-determining region Y (SRY)-box 9Ngn3IDeN[140,141,142,143]
Nkx6.1ITraN
Pax6INeoP
InsIProP
HNF1AHepatocyte Nuclear Factor 1 AHNF4AIDeN[144,145,146,147,148,149]
AS1I
Tmem27I
Kcnj11I
Glut2ITraN
InsINeP
NeuroDIIProP
GckI
MafBI
GiprI
Glpr1I
G6pc2I
Slc5a1I
Pdx1I
Bcl2li(Bcl-xL)I
ARXAristaless-Related HomeoboxGcgIDep[150,151,152,153]
Pax4R
SSTRTrap
NeoN
Pro
GATA6GATA Binding Protein 6iNOSRDeN[154,155,156,157,158]
InsI
Glut2ITraN
Abcc8I
Snap-25INeoP
Pdx1I
Nkx6.1IProP
MafAI
GLIS3GLI Similar 3InsIDeN[159,160,161,162,163]
Ngn3ITraN
MafAINeoP
Glut2IProP
PAX6Paired Box 6InsIDeN/P[164,165,166,167]
GCGR
Glut2I
GiprI
Nkx2.2I
PC1/3I
Glp1rI
SSTRTraN/P
GherlinR
IsiRNeoP
Foxa2R/I
Ngn3R/IProP
Pdx1R/I
NeuroD1R/I
MafAR/I
Nkx6.1R/I
MafBMusculoaponeurotic Fibrosarcoma Oncogene Homolog BGASTRDeN[168,169,170,171]
SSTR
P2ry1ITraN
Adra2AI
Tph1INeoP
InsI
ChrnA3, ChrnA4, ChrnB4IProP
Robo1, Robo2, Nrp1, Nrp2I
Hes1Hairy and Enhancer of Split-1Ngn3RDeP[172,173,174,175]
NeuroDRTraP
Ptf1aRNeoN
p27Kip1 and p57Kip2RProP
HNF4 AHepatocyte Nuclear Factor 4 AAldolase BIDeN[176,177]
Glut2I
GckITraN
L-pyruvate kinaseI
Kir6.2INeoP
InsI
Pdx1IProP
Pax4I
Ngn3I
Ki67I
NeuroD1Neurogenic Differentiation 1InsIDeN[178,179,180]
GckI
Abcc8/Sur1, Kcnj11I
Cacna1aITraN
MafaI
Nkx6-1I
Pdx1INeoP
Nkx2-2I
Insm1I
Isl1IProP
Pax6I
Rfx6I
Cdh4R
OclnR
MecomR
ldhaR
Nkx6.1NK6 homeobox 1Glut2IDeN[130,181,182,183,184]
G6pc2I
ArxRTraN
Pdx1I
MafAINeoP
Ero1lbIProP
Slc30a8I
Nr4a1 and Nr4a3I
ISL1ISL LIM homeobox 1MafAIDeN[185,186,187,188]
Pdx1I
Glut2I
InsI
NeuroD1ITraN
Pax6I
MafB,I
Nkx6.2I
Insm1INeoP
Sox9I
ArxI
Foxa2I
Nkx6.1IProP
GCGI
SSTI
G6pc2I
Table 3. LncRNAs involved in β-cell dedifferentiation, transdifferentiation, neogenesis, or proliferation (De/Tra/Pro/Neo).
Table 3. LncRNAs involved in β-cell dedifferentiation, transdifferentiation, neogenesis, or proliferation (De/Tra/Pro/Neo).
LncRNAsDirect and Indirect TargetsDe/Tra/Pro/NeoType of EffectReferences
Gm16308 ProP[243]
DANCRmiR-33a-5pProP[218,244]
H19miRNA let-7- miRNA-29aProP[245,246,247]
TUNAREZH2ProP[248]
Meg3EZH2, Pdx1, MafADeN[249,250]
βFaarmiR-138-5pDeN[251]
PLUTOPdx1DeN[124]
HILNC25Glis3DeN[252]
Gm10451miR-338-3pDeN[253]
810019D21Rik (RIOT)Nkx6.1DeN[254]
p3134Pdx1DeN[124,255]
linc-p21miR-766-3pProN[256]
HOTAIRPdx1DeN[124]
LncCplx2EZH2ProN[257]
MALAT1Pdx1DeP[258]
MIR503HGCtBP1NeoN[175]
GAS5miR-29a-3p, miR-96-3p, miR-208a-3pDeN[259,260]
ProP
LINC01018miR-499a-5pDeP[261]
ProN
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Ghasemi Gojani, E.; Rai, S.; Norouzkhani, F.; Shujat, S.; Wang, B.; Li, D.; Kovalchuk, O.; Kovalchuk, I. Targeting β-Cell Plasticity: A Promising Approach for Diabetes Treatment. Curr. Issues Mol. Biol. 2024, 46, 7621-7667. https://doi.org/10.3390/cimb46070453

AMA Style

Ghasemi Gojani E, Rai S, Norouzkhani F, Shujat S, Wang B, Li D, Kovalchuk O, Kovalchuk I. Targeting β-Cell Plasticity: A Promising Approach for Diabetes Treatment. Current Issues in Molecular Biology. 2024; 46(7):7621-7667. https://doi.org/10.3390/cimb46070453

Chicago/Turabian Style

Ghasemi Gojani, Esmaeel, Sweta Rai, Farzaneh Norouzkhani, Salma Shujat, Bo Wang, Dongping Li, Olga Kovalchuk, and Igor Kovalchuk. 2024. "Targeting β-Cell Plasticity: A Promising Approach for Diabetes Treatment" Current Issues in Molecular Biology 46, no. 7: 7621-7667. https://doi.org/10.3390/cimb46070453

Article Metrics

Back to TopTop