Home
JournalsCollections
For Authors For Reviewers For Editorial Board Members
Article Processing Charges Open Access
Ethics Advertising Policy
Editorial Policy Resource Center
Company Information Contact Us Membership Collaborators Partners
OPEN ACCESS

The Future of Type 1 Diabetes: Can Stem Cells Provide a Cure?

  • Swarup K. Chakrabarti1,*  and
  • Dhrubajyoti Chattopadhyay1,2
Exploratory Research and Hypothesis in Medicine   2025;10(4):e00029

doi: 10.14218/ERHM.2025.00029

Received:

Revised:

Accepted:

Published online:

 Author information

Citation: Chakrabarti SK, Chattopadhyay D. The Future of Type 1 Diabetes: Can Stem Cells Provide a Cure?Explor Res Hypothesis Med. 2025;10(4):e00029. doi: 10.14218/ERHM.2025.00029.

Abstract

Type 1 diabetes (T1D) develops when the immune system targets and destroys pancreatic β-cells responsible for insulin production, ultimately resulting in reduced insulin levels. Islet transplantation has garnered significant attention as a potential treatment, but it presents numerous challenges that hinder its effectiveness for T1D patients. A primary issue is the immune system’s tendency to reject transplanted islets, leading to a gradual decline in their functionality. Furthermore, many individuals remain reliant on additional insulin therapy. These challenges are exacerbated by the global shortage of organ donors, which limits the availability of pancreata for transplantation. This review outlines several innovative strategies to regenerate insulin-producing β-cells for the treatment of T1D, with a primary focus on pancreatic progenitor and stem cells. The strategy of converting non-β cells, particularly pancreatic α-cells, into functional β-cells continues to show promise. Moreover, α-cells, which are less vulnerable to autoimmune attacks, present a distinct opportunity for β-cell regeneration in individuals with T1D. While the use of progenitor or stem cells for β-cell regeneration appears encouraging, various hurdles, such as immune rejection, suboptimal differentiation, and other challenges, still impede the implementation of this strategy. Nonetheless, this approach may ultimately pave the way for long-lasting treatment and potential cures for T1D.

Keywords

Type 1 diabetes, β-cell regeneration, Pancreatic progenitor cells, Stem cells, Autoimmune disease, NGN3 signaling

Introduction

Type 1 diabetes (T1D) is an autoimmune disorder in which the immune system mistakenly attacks and destroys the β-cells in the pancreas that are responsible for producing insulin, resulting in inadequate insulin production.1–3 The immune system often targets insulin itself or components that are essential for its synthesis, like pre-proinsulin, the insulin precursor.4,5 Moreover, a combination of environmental and metabolic factors, along with genetic and epigenetic factors, contributes to the initiation and progression of this autoimmune response.6–8 Its distinctive feature is this immune-mediated destruction of pancreatic β-cells.1–3 Therefore, a person diagnosed with T1D requires continuous insulin therapy.9 Hyperglycemia, or an increase in glucose in the blood without the conjunction of insulin, can result in a lethal condition known as diabetic ketoacidosis.10,11 Although T1D is traditionally considered a childhood disease, more than half of the new cases now occur in adults, with diagnoses spanning from early childhood to well into middle age.12,13 It is crucial to distinguish between T1D and type 2 diabetes, since type 2 diabetes primarily affects older individuals and is characterized by an initial insulin resistance instead of a complete absence of insulin.14,15 Interestingly, in T1D, the immune system attacks β-cells only, leaving glucagon-producing α-cells unaffected.16,17 Non-β cells may have innate resistance to autoimmune attack, making them suitable for transformation into β-cells by innovative techniques to promote evasion from autoimmunity.18–20 In simple terms, the immune response affects specific types of cells, namely β-cells, confirming that the autoimmune mechanism in T1D has a degree of selectivity that can be utilized for future treatment approaches. In this context, the immune system’s rejection of the recipient of transplanted cells poses a significant challenge to established treatments such as islet transplantation (IT).21,22 This immune response can result in a gradual reduction of islet function over the subsequent years, and it may also prevent certain individuals from fully reducing their reliance on exogenous insulin therapy.23,24

Moreover, the worldwide shortage of organs is a major challenge that hampers progress in pancreatic IT. Annually, around 8,000 organ donations take place, yet fewer than one-third of the pancreases are considered suitable for transplantation.25,26 Although IT can provide a provisional solution for T1D, it necessitates the administration of immunosuppressive drugs to prevent graft rejection.27,28 However, these immunosuppressants can cause lymphopenia, which is associated with increased serum levels of the cytokines interleukin (IL)-7 and IL-15, contributing to the in vivo proliferation of autoreactive clusters of differentiation (CD) 8+ T cells.29,30 The resurgence of autoreactivity is likely a significant factor behind the long-term failure of islet transplants. Graft rejections, under normal circumstances, typically involve an immune response directed against donor antigens. However, in the case of T1D, the resurgence of autoimmunity is fundamentally different from these occurrences and indicates the reactivation of autoreactive T cells targeting self-antigens in the graft, thereby signifying another resurgence of the disease’s original pathogenic mechanisms. In other words, rather than the typical rejection of foreign tissue, the immune system could activate the dormant autoimmune process that originally caused β-cell destruction in T1D. This revival of autoreactive immune responses likely plays a role in the eventual failure of the islet grafts.31,32 It also underscores the complex immunological landscape that needs to be managed to achieve successful and lasting IT outcomes.

In light of these challenges, researchers are increasingly investigating the potential of pancreatic stem or progenitor cells to produce immune-tolerant β-cells, making them a logical option for T1D patients with autoimmunity. To elaborate, multiple experimental findings indicate that, under specific culture conditions or when triggered by key transcription factors, these progenitor cells can produce functional β-like cells that release insulin in response to glucose.33–35 Moreover, there is a growing focus on whether stem cell-derived β-cells are more resilient to immune attacks or can be designed to promote immune tolerance.36–38 Thus, immune modulation through stem cell-based approaches could help alleviate immune attacks, aiming to restore β-cell function and address the fundamental autoimmune response seen in T1D. Although identifying and isolating pancreatic stem cells remains technically challenging and they are available in limited quantities for research, these cells hold potential for modulating immune responses and preventing autoimmune destruction of β-cells in T1D. Pancreatic stem or progenitor cells are typically obtained from donated organs. Unlike transplanting entire organs, these cells can be expanded outside the body and may be modified for immune invisibility. Therefore, they offer a scalable and less resource-intensive solution that could alleviate the challenges associated with organ shortages.39–42 Mesenchymal stem cells release anti-inflammatory cytokines such as IL-10, transforming growth factor-beta (TGF-β), and prostaglandin E2, which may slow effector T cell growth and promote the expansion of regulatory T cells (Tregs), thereby reducing immune responses.43,44 These generate a protective environment for the neogenesis of β-cells formed from pancreatic progenitor cells to boost immunity against the immune attack.45,46 A highly small number of pancreatic stem cells in the islets can differentiate into insulin-producing β-cells. They provide the possibility to restore endogenous insulin production through immune modulation and regeneration, with minimal rejection risk.47–49

However, a significant barrier is the ambiguity regarding the existence of specific pancreatic stem cells in adults. Pancreatic regeneration mainly occurs through β-cell division rather than through stem cells that self-renew.50,51 Pancreatic progenitor cells play a critical role in building both exocrine and endocrine cells during embryonic development, but seem to lose this ability with age.52,53 Recent studies suggest that progenitor-like cells in ductal and acinar regions may generate new β-cells, especially under injury or metabolic stress.54,55 The mature pancreas lacks a dedicated stem cell niche like the intestines and bone marrow. Advances in regenerative medicine and stem cell research show that cells can be reprogrammed to act like pancreatic progenitors or stem cells from other tissues, generating functional β-cells.56–59 This has led to new diabetes treatments, such as activating progenitor cells or using patient-derived stem cells to restore insulin production.60–63

Recent studies have also highlighted the potential of senescent or damaged β-cells—cells that endure an autoimmune attack yet lose their ability to replicate or produce insulin.64,65 Although typically nonfunctional, these cells may possess significant flexibility in terms of differentiation and could potentially be reprogrammed under certain conditions. Genetic or drug-based strategies may help dysfunctional β-cells revert to a progenitor-like state, enabling them to grow and produce insulin.66–70 This concept of cellular adaptability provides optimism for restoring insulin production in individuals with T1D. Interestingly, senescent β-cells may acquire progenitor-like characteristics in the presence of appropriate signaling cues.71–74 Genetic or regulatory factors may drive the cells back to a more stem-like state and allow for the replacement and expansion of insulin-secreting cell mass.75–77 Under certain conditions, studies on animals indicate that α-cells might transdifferentiate into a cell type reminiscent of a β-cell.78,79 This suggests that, under appropriate signals, both α-cells and aged β-cells could be reprogrammed to perform the roles of insulin-producing cells. Some reprogrammed cells may arise from surviving β-cells, suggesting that senescent ones could regain a flexible, progenitor-like state to support regeneration.80,81 Hence, reprogramming senescent β-cells into functioning insulin-producing cells represents a significant breakthrough in diabetes care.82,83 This approach aims to restore β-cell function in T1D, moving beyond insulin replacement to regenerative therapies that address the disease’s root cause. If successful, reprogramming senescent β-cells into functional β-cells, either directly or after turning them into progenitor β-cells, could provide a permanent solution for restoring insulin production.84–86 This concept envisions a future in which persons with T1D will no longer rely on insulin injections.

Against this backdrop, this article aims to expand T1D research by critically analyzing past studies and forming hypothesis-driven insights. It explores alternative sources of insulin-producing cells, focusing on whether pancreatic stem or progenitor cells exist in islets, as adult stem cells are found in other organs. It also investigates why α-cells resist autoimmune attacks in T1D. A critical question is whether the distinctive property of their resistance to autoimmunity can help convert non-β-cell counterparts into ‘normally’ resistant β-cells. This research aims to uncover new approaches for T1D management and explore their broader societal impact.

Hence, the article opens with the search for elusive pancreatic stem cells and the inquiry into whether β-cell replication suffices for insulin regeneration. It subsequently explores the regenerative role of ductal epithelium and insights gained from single-cell transcriptomics.87,88 It focuses on the immune resistance of α-cells, their role in β-cell protection, and approaches to replicate this immunity.89,90 The final sections briefly examine important transcriptional and signaling pathways that could be utilized to combat T1D and regenerate β-cells: neurogenin-3 (NGN3), Hippo signaling pathway (Hippo)/Yes-associated protein (YAP), and glucagon-like peptide-1 (GLP-1).34,91–94

The hidden treasure island: The search for elusive pancreatic stem cells

Recent research shows that turning stem cells, like embryonic stem cells (ESCs) or induced pluripotent stem cells, into β-cells could help with cell replacement therapies.95,96 For instance, studies have indicated that researchers successfully produced glucose-responsive β-like cells from ESCs that managed blood glucose levels in diabetic mice.97–99 Initial clinical trials conducted by ViaCyte using encapsulated pancreatic progenitors derived from ESCs showed their potential for application, although some challenges related to immune responses were encountered.100 Building on this initial research, ViaCyte progressed its proprietary pancreatic endocrine cell (PEC)-01 cell line, sourced from human pluripotent stem cells, into Phase 1 and Phase 2 clinical trials (e.g., NCT02239354, NCT03163511). These clinical studies involved subcutaneous delivery of PEC-01 cells through macroencapsulation devices, such as Encaptra. While the results were encouraging regarding safety and partial engraftment, several problems arose, including limited oxygen and nutrient diffusion, excessive fibrosis growth, and insufficient insulin secretion, presenting major hurdles to achieving long-term functionality and immune compatibility. These findings underscore the need for ongoing enhancement in cell encapsulation and device innovation to optimize the survival and efficacy of transplanted cells. Vertex Pharmaceuticals, which acquired ViaCyte in 2022, recently announced favorable outcomes for its VX-880 treatment, inspired by Douglas Melton’s foundational research, indicating a revival of spontaneous insulin production and reduced reliance on insulin in patients.101,102 This approach offers major benefits, like scalability and the ability to modify cells so the immune system doesn’t attack them after transplantation, which is crucial for treating T1D. It also suggests that we can trigger pancreatic stem cells, possibly in the islets, to become insulin-producing β-cells. By changing key biological signals and the pancreatic environment, we may activate this regeneration and restore β-cell function in people with T1D.103,104 The main challenge is figuring out how to change these signals and control stem cells to encourage β-cell production while preventing immune system rejection. As a result, the quest for endogenous pancreatic stem or progenitor cells could significantly impact T1D management. If scientists could identify these cells and determine whether they have regenerative ability, there may then be the possibility of inducing the direct conversion of these cells into insulin-secreting β-cells in the pancreas of the subjects. This would usher in a new mode of replacement for β-cell functioning, which would be far more sustainable and personalized and would considerably reduce reliance on an external cell source as well as the immunorejection risk, which is currently a major hurdle for treatment.105,106 Understanding the major mechanisms governing this important process will be crucial in devising efficient therapies for T1D and ultimately restoring normal insulin production.

Replicating β-cells: A limited solution or the future of insulin regeneration

Looking back at the history of scientific inquiry, groundbreaking research by Susan Bonner-Weir and colleagues showed that the number of β-cells in the pancreas progressively increases during normal development and continues throughout adulthood.107 This growth links the formation of new, small islets to ductal progenitor cells in a process called neogenesis. Another finding was that while pancreatic cells, like β-cells, acinar cells, and ductal cells, can replicate, mature β-cells may have limited ability to do so due to two types: one that can replicate and another that has aged. It’s noteworthy that while replicating β-cells may undergo slight dedifferentiation, they do not revert to progenitor or stem cell states. During replication, these β-cells might temporarily lose their functional capabilities. This suggests that using existing β-cells to produce insulin may be less effective at regulating blood glucose than converting pancreatic progenitor cells into stable, long-lasting insulin-producing β-cells.

Subsequent studies have confirmed the concept of neogenesis, particularly during physiological conditions such as pregnancy or stress from injury, where ductal cells are implicated in the regeneration of β-cells.108–110 Notably, studies by Bonner-Weir and associates have provided histological and lineage-tracing proof demonstrating the plasticity of ductal epithelial cells in creating endocrine progenitors.111–114 Similarly, studies have shown that the activation of the Notch pathway can influence the differentiation of ductal cells into cells that produce insulin.115,116 Furthermore, research has shown that while mature β-cells are capable of limited proliferation, this process is often followed by temporary dedifferentiation.117,118 During this phase, there is a decrease in the expression of insulin and key genes associated with β-cell identity, such as pancreatic and duodenal homeobox 1 (Pdx1) and V-maf musculoaponeurotic fibrosarcoma oncogene homolog A.119–121 These findings emphasize the challenges of relying only on the replication of mature β-cells to regain function in diabetes and instead support the therapeutic strategy of employing progenitor cell populations or neogenesis to create robust, functional β-cells.

Uncovering hidden stem cells: The potential of ductal epithelium in pancreatic regeneration

Studies show that the duct epithelium in adults can still produce all the cell types in the pancreas, offering a potential source for pancreatic stem and progenitor cells.105,111–113,122 Researchers have successfully extracted potential intra-islet stem cells, marked by nestin, from both rat and human islets.123,124 One alternative concept suggests the existence of facultative or functional stem cells.125,126 This implies that a fully differentiated epithelial cell can momentarily behave like a stem cell by undergoing multiple divisions, despite not being an actual stem cell. Animal studies show that after partial pancreas removal, mature ductal cells in rats can replicate and become less differentiated, forming islet, acinar, or duct cells.127,128 Similarly, transgenic mouse models with overproduced interferon (IFN)-γ regulated by the insulin promoter have been shown to enhance neogenesis in adulthood.129,130 These experiments cause insulitis by promoting IFN γ expression through insulin, leading to continuous ductal cell growth and new islet formation.131,132 Most importantly, the new islets extend toward the ductal lumen, suggesting that pancreatic progenitor cells exist in the duct.133,134

Building on these findings, later research has further strengthened the regenerative abilities of adult pancreatic ductal epithelium. For example, studies indicate that ductal cells can transform into functional β-cells under specific conditions, such as the activation of key signaling pathways like Notch and Wingless-related integration site (Wnt).135–137 Importantly, research in transgenic mice revealed that stimulating these pathways can convert mature ductal cells into insulin-producing β-like cells, offering a possible treatment method for regenerating β-cells.138,139 Later research has also examined the role of inflammation and stress in promoting the growth of ductal progenitor cells. Research has indicated that enhancing β-cell regeneration following injury can be achieved by blocking key elements that limit progenitor cell activation, such as the TGF-β signaling pathway.140,141 Additionally, studies revealed that after partial pancreatic damage, ductal cells exhibit greater plasticity and can regenerate functional insulin-secreting cells.142,143 Recently, it has been shown that metabolic stress, such as insulin resistance caused by a high-fat diet, can stimulate the proliferation of ductal progenitor cells and enhance β-cell regeneration in diabetic models.144,145 Similarly, it was recently discovered that altering the Hippo signaling pathway in adult mice enhanced the plasticity of ductal cells and prompted the neogenesis of β-cells after pancreatic damage.146–148 These findings support the idea that the ductal epithelium serves as an essential source of progenitor cells for β-cell regeneration and suggest that adjusting specific signaling pathways might improve neogenesis and offer a possible therapy for T1D.

Unlocking pancreatic regeneration: The power of single-cell transcriptomics

Put together, while the therapeutic potential of pluripotent stem cell-derived β-like cells has been demonstrated, the existence of a dedicated endogenous pancreatic stem cell population in adult humans remains under investigation. Nevertheless, the search for such cells remains crucial, particularly in the context of T1D, where inflammation-driven β-cell loss highlights the need for regeneration and replacement strategies. A comprehensive identification of plausible cell types could ultimately lead to the discovery of true pancreatic progenitor stem cells. In this context, single-cell transcriptomics serves as an effective technology for in-depth analysis. By exploring the gene expression profiles of individual cells within the ductal epithelium and newly formed islets, single-cell RNA sequencing can effectively identify and isolate potential progenitor cells, even those exhibiting minor or transient characteristics.149,150 This method could identify markers specific to progenitor states, differentiate pathways, and assess the process of dedifferentiation or transdifferentiation occurring in pancreatic tissues.151,152 In addition, the single-cell RNA sequencing technique can deconvolute the variegated cell types existing in the ductal epithelium and help discover subgroups with stem-like properties or plasticity, especially when injury is present or inflammation is triggered in transgenic models.153,154 This ability to distinguish progenitor populations by their gene expression provides an avenue for their isolation and further study.155,156 In essence, single-cell transcriptomics helps uncover more about pancreatic progenitor cells and provides the foundation for advancing regenerative medicine.157,158 Critically, to assist readers in navigating the diverse techniques commonly employed in pancreatic β-cell regeneration, Table 1 provides a comprehensive summary of advanced experimental and translational techniques designed for the regeneration and preservation of β-cells in a T1D setting.38,149–152,157–185 Such information is crucial to the development of next-generation cell therapies for diabetes.38,157–185

Table 1

Emerging techniques in β-cell regeneration and therapeutic applications in type 1 diabetes

TechniquePurposeDescriptionTherapeutic implications
Single-cell RNA sequencing (scRNA-seq)Identification of progenitor/stem-like cellsExamines gene expression at the single-cell level to investigate rare or transient progenitor cell states within the pancreatic ductal epithelium149151Enables the identification and isolation of potential regenerative cell types, which is an essential step in advancing targeted cell-based therapies157,158
Cellular reprogrammingRegeneration of β-cellsTransforms non-β cells, such as α-cells or ductal cells, into insulin-secreting β-cells through the use of transcription factors and the modulation of signaling pathways159,160Provides a method to restore missing β-cells without depending on external stem cell sources161
Transgenic animal modelsUnderstanding neogenesis and immune mechanismsGenetic modification, such as the overexpression of IFN-γ, causes damage or inflammation in the pancreas to investigate regeneration and immune reactions162Replicates human diseases, facilitating the identification of regeneration pathways and immune regulators 163
Epigenetic profilingStudying chromatin states and immune resilienceInvestigates DNA methylation, histone modifications, and chromatin accessibility to understand the mechanisms behind α-cells’ resistance to immune attacks164,165May assist in reprogramming β-cells to reduce their immunogenicity and enhance their resistance to immune destruction166
Molecular mimicryβ-cell protection via immune evasionEngineered β-cells produce immune-protective molecules similar to those found in α-cells, thereby minimizing immune targeting167,168Decreases autoimmunity and improves the survival of β-cells following transplantation or during endogenous regeneration169
Immune checkpoint modulationImmunosuppression and β-cell protectionIntroduction or activation of checkpoint inhibitors (e.g., PD-1, CTLA-4) on β-cells to prevent T-cell mediated killing170,171It could enhance β-cell tolerance and prolong survival in autoimmune conditions172
Anti-inflammatory cytokine therapyModulating immune environmentUtilization of cytokines like IL-10 or TGF-β to mitigate localized inflammation and safeguard β-cells173,174Creates a favorable niche for β-cell regeneration and survival in T1D175
Lineage tracingTracking cell origin and fateEmploys genetic labeling techniques to monitor the conversion of ductal or α-cells into β-cells in vivo. The labeled cells are then tracked over time in vivo to monitor their potential conversion into insulin-producing β-cells176Verifies the origins of neogenesis; substantiates regenerative processes in living tissues177
Organoid culture systemsIn vitro modeling of regenerationThree-dimensional (3D) cultures developed from pancreatic tissue or stem cells to replicate the structural and functional characteristics of the pancreas178,179Facilitates the evaluation of regeneration protocols and the screening of pharmaceuticals within a regulated setting180
Gene editing (e.g., CRISPR/Cas9)Modulating gene expression in pancreatic cellsEmployed to alter the expression of autoantigens, improve β-cell functionality, or replicate the protective characteristics of α-cells181,182Enables the development of tailored therapies and the generation of immune-resistant β-cells183
Beta cell encapsulationEncapsulation of β-cells or progenitors in a protective barrier to shield them from immune attackInvolves enclosing insulin-producing cells in a semi-permeable membrane to protect them from immune attack while allowing nutrient and insulin exchange184,185Long-term β-cell protection in an immune-privileged environment38

What α-cells know: Lessons learned to safeguard β-cells in T1D

As previously stated, T1D is an autoimmune disorder that destroys the β-cells in the pancreas, whereas the α-cells that generate glucagon typically remain unharmed. The selective preservation of β- and α-cells during immune attacks is shaped by a variety of molecular, immunological, and physiological factors that contribute to the protection of α-cells. Gaining insight into these mechanisms is crucial for understanding the onset of T1D and investigating potential treatment alternatives. A primary cause for the specific destruction of β-cells is the diverse expression of autoantigens.186,187 β-cells are responsible for producing insulin and other proteins such as glutamic acid decarboxylase and insulinoma-associated antigen-2, which are targeted by autoreactive T cells in T1D.188,189 Cytotoxic T lymphocytes (CTLs) kill the β-cells upon being activated by stimulation from major histocompatibility complex molecules.190 Notably, in contrast to the β-cells, the α-cells seem to express considerably less of these autoantigens and are, therefore, not recognizable by the immune system.191 As a result, α-cells frequently manage to evade the harmful effects that are primarily directed at the β-cells.

α-cell resilience: A key to β-cell regeneration and protection in T1D

Immunological stresses are not confined to the expression of antigens; rather, they inhibit normal β-cell function.192 In addition, activated T cells produce inflammatory cytokines like IFN-γ and tumor necrosis factor alpha during any autoimmune response or condition.193,194 This eventually leads to the endoplasmic reticulum inability to withstand oxidative stresses and subsequent cell death.195,196 Given their substantial metabolic needs and high levels of insulin production, β-cells face a greater risk.197,198 In contrast, α-cells demonstrate greater resilience to these cytokines, largely due to their different gene expression profiles and reduced reliance on insulin production.199 Additionally, α-cells boast a stronger anti-apoptotic signaling route that balances oxidative stress and lowers responses to endoplasmic reticulum stress, thereby allowing these cells to sustain the inflammatory atmosphere typical of T1D.200 Under certain conditions, α-cells can transdifferentiate into β-cells, enabling them to adapt and compensate for the loss of β-cells.201,202 Though presumably not to the extent found in humans, this points toward the relatively plastic characteristics of α-cells compared to the more inflexible nature of β-cells. Differences might also arise in the vascular system as well as in the microenvironment within pancreatic islets, affecting immune responses.203,204 α-cells are generally located at the periphery of islets and may be exposed to different levels of cytokines and immune cells than β-cells, which are situated centrally and are more directly involved in glucose regulation, making them more susceptible to immune attacks.205 It is possible that the entire immune system is directed against the β-cells, considering their important function, insulin, in overall glucose regulation. As a major hormone regulating various functions in the body, insulin has strong immunogenic properties, making β-cells apparent immunological targets.206 Glucagon secreted by α-cells is also a regulator of glucose, although its effects are secondary to those of insulin, and thus immune responses against α-cells are toned down.207 Additionally, research indicates that α-cells might influence immune reactions within the islet microenvironment, which could aid in their survival.208

Immune resistance in pancreatic β-cells could potentially be enhanced by adopting strategies employed by α-cells in T1D, which have evolved mechanisms to evade immune-mediated destruction. One option could be molecular mimicry: modifying β-cells to express the protective molecules expressed by α-cells, thereby decreasing their immunogenicity.209–211 In addition, α-cells employ immune checkpoint regulators, such as programmed cell death protein 1 (PD-1) and cytotoxic T-lymphocyte antigen 4 (CTLA-4), to shield themselves from T-cell attacks; therefore, encouraging similar regulatory molecules in β-like cells may fortify them against autoimmunity.212,213 Recognizing and increasing the levels of anti-inflammatory cytokines like IL-10 and TGF-β could also enable β-cells to withstand immune challenges, akin to α-cells.214,215 For instance, in the non-obese diabetic mouse model for T1D, it has been noted that IL-10 therapy reduces β-cell damage and the infiltration of immune cells into the pancreas.216,217 Furthermore, IL-10 enhances the Treg response, which is crucial for maintaining immune tolerance.218 Preclinical studies, particularly in non-obese diabetic mice, have shown that IL-10–mediated enhancement of Tregs can delay or prevent diabetes onset, underscoring its potential as a therapeutic target for preserving β-cell function in T1D.219,220 In contrast, a lack of IL-10 accelerates the onset of diabetes and alters intestinal immunity.221

Another promising avenue is the downregulation or modified presentation of major β-cell autoantigens, such as insulin or glutamic acid decarboxylase-65, to reduce their visibility to autoreactive T cells.222,223 Furthermore, epigenetic profiling of α-cells has revealed a more compact and immune-inactive chromatin state around key inflammatory gene loci, which may underlie their reduced immunogenicity.224–227 Understanding and recreating such epigenetic states in β-cells could serve as a strategy to reprogram their immune profile.228,229 These are strategies that can be applied in principle but require large-scale experimental verification before being turned into clinical processes.

In summary, during T1D, α-cells resist immune stress based on a low autoantigen expression profile, higher resistance against inflammatory stimuli, and activation of pathways that inhibit cell death. The possibility that α-cells may transdifferentiate into β-cells could allow for certain regenerative strategies. Strategies such as immune checkpoint modulation, molecular mimicry, and the use of anti-inflammatory cytokines could help protect or restore β-cell function, though further research is required. The immune-mediated destruction of β-cells in T1D involves intricate interactions concerning antigen presentation, immune targeting, cellular stress, and renewal. In this context of autoimmunity, α-cells demonstrate resilience, presenting opportunities to minimize β-cell loss through their adaptability and informing future T1D therapies.

Reversing T1D: Transcriptional and NGN3 signaling pathways in β-cell regeneration

As mentioned earlier, although current T1D management includes insulin replacement, new regenerative therapies aim to replenish endogenous β-cell mass through cellular reprogramming. They rely on an accurate understanding of the molecular mechanisms that govern pancreatic development, lineage determination, and β-cell maturation. Key transcription factors, initially identified during embryonic development and currently utilized to guide stem or progenitor cells toward a β-cell identity, continue to be vital for the future of this area.230,231 Simultaneously, pathways such as Wnt/β-catenin and Hippo/YAP provide crucial signals for the proliferation, survival, and differentiation of progenitor cells.232,233 By adjusting these signaling and transcriptional networks, scientists aim to reprogram non-β-cell types or increase β-cell numbers, ultimately offering a route to insulin independence in T1D. In addition to the aforementioned transcription factors, Table 2 includes a comprehensive list of key transcription factors and signaling pathways that play a role in β-cell reprogramming and regeneration.234–271 This table will serve as a reference throughout the article, highlighting their roles in promoting β-cell regeneration, protecting against β-cell impairment, and facilitating the conversion of non-β-cells into insulin-producing cells. These elements hold significant therapeutic potential for diabetes treatment. In essence, Table 2 outlines the essential transcription factors and pathways involved in this regenerative process, along with their therapeutic significance. This introduction lays the foundation for the subsequent discussion on key signaling pathways, including NGN3, Hippo/YAP, and GLP-1.

Table 2

Key transcription factors and signaling pathways in β-cell reprogramming and regeneration

Transcription factor/signaling pathwayFunction in β-cell reprogramming/regenerationTherapeutic implications
Pdx1 (pancreatic and duodenal homeobox 1)Master regulator of β-cell differentiation and maintenance. Promotes insulin secretion and β-cell identity234Potential therapeutic target for β-cell regeneration in T1D. Can enhance endogenous β-cell regeneration or reprogram non-β-cells into functional β-cells235
NGN3 (neurogenin 3)Induces pancreatic endocrine differentiation and promotes β-cell lineage specification236Targeting NGN3 could induce the generation of new β-cells from pancreatic progenitors or ductal cells, offering promising treatments for T1D237
MAFA (V-maf avian musculoaponeurotic fibrosarcoma oncogene homolog A)Regulates insulin expression in β-cells and aids in maintaining their function. Important for maintaining β-cell function during stem cell differentiation238Therapeutic application in augmenting insulin production or improving β-cell survival in diabetes239
NEUROD1 (neurogenic differentiation 1)It is a basic helix-loop-helix (bHLH) transcription factor. Works downstream of NGN3; critical for terminal differentiation and functional maintenance of β-cells240Utilization in cell-based therapies or reprogramming protocols may help restore β-cell mass in diabetes, especially when used in combination with Pdx1, MAFA, or NGN3241
PAX4 (paired box gene 4)Promotes β-cell fate over α-cell fate during endocrine progenitor differentiation; supports β-cell survival242Conversion of pancreatic stem cells or non-β-cells into insulin-producing β-cells: a potential strategy for β-cell restoration in T1D244
ARX (aristaless-related homeobox)Antagonistic to PAX4; promotes α-cell fate—needs to be suppressed to favor β-cell lineage244Downregulation or inhibition of ARX in pancreatic stem cells or progenitors can enhance β-cell differentiation by shifting the endocrine fate toward insulin-producing β-cells as opposed to glucagon-producing α-cells245
ISL1 (ISL LIM homeobox 1)Supports pancreatic endocrine development and insulin gene transcription246Promotes endocrine lineage commitment and β-cell maturation; has the potential to enhance functional β-cell generation from stem cells for T1D therapy247
FOXO1 (forkhead box O1)A transcription factor that regulates β-cell fate by balancing proliferation, maintenance, and differentiation from progenitor or stem cells248Modulating FOXO1 activity may enhance β-cell regeneration and function, offering a potential strategy for restoring β-cell mass in T1D249
Sox9 (SRY-box transcription factor 9)Regulates pancreatic progenitor cell fate and is critical in ductal cell differentiation250Targeting Sox9 could aid in reprogramming ductal cells into β-cells, a potential therapeutic approach for restoring β-cell mass in T1D250,251
GATA4Critical for β-cell survival and differentiation, it contributes to maintaining β-cell identity252Therapeutic modulation of GATA4 can potentially enhance stem cell-derived β-cell generation in T1D253
HNF1β & HNF4α (hepatocyte nuclear factors)Key in early pancreas development and glucose regulation; mutations associated with maturity-onset diabetes of the young (MODY)254Potential therapeutic modulation of HNF1β and HNF4α can promote β-cell neogenesis from stem cells by regulating pancreatic development and insulin expression255
YAP/TAZ (Yes-associated protein/transcriptional coactivator with PDZ-binding motif)Part of the Hippo signaling pathway, it regulates β-cell proliferation and regeneration256Therapeutic modulation of YAP/TAZ signaling potentially promotes β-cell proliferation, regeneration, and differentiation from stem cells, offering a strategy for restoring β-cell mass in diabetes257
Wnt (Wingless/Integrated)/β-catenin pathwayRegulates β-cell differentiation and proliferation. Important for the expansion of the β-cell population258Therapeutic activation of Wnt/β-catenin signaling may enhance β-cell regeneration and promote stem cell differentiation into β-cells for T1D treatment259
Tcf7l2 (T-cell factor 7-like 2)Involved in Wnt/β-catenin signaling and promotes β-cell proliferation and survival260Activating Tcf7l2 signaling may offer potential for stimulating β-cell regeneration from stem cells in T1D therapy261
Notch signaling pathwayModulates endocrine differentiation, influencing β-cell identity and function262Manipulating Notch signaling could enhance β-cell regeneration and reprogramming of progenitor cells263
JAK/STAT (Janus kinase/signal transducer and activator of transcription) pathwayInvolved in cell survival and immune response regulation. Plays a role in β-cell regeneration264Targeting JAK/STAT could enhance β-cell survival and regeneration, particularly in inflammation-driven β-cell loss in T1D265
TGF-β (transforming growth factor beta) pathwayInhibits β-cell proliferation and differentiation but also regulates their function266TGF-β pathway inhibition may enhance the differentiation of pancreatic progenitor cells into β-cells, overcoming differentiation barriers267
SMAD pathwayRegulates β-cell differentiation and expansion. Plays a role in TGF-β signaling in β-cells268Targeting SMAD signaling may overcome β-cell growth inhibition and enhance differentiation of pancreatic stem cells into functional β-cells for T1D therapy269
GLP-1 (Glucagon-like peptide-1)Enhances β-cell proliferation, survival, and function270Therapeutic GLP-1 analogs can promote β-cell regeneration and support stem cell-derived β-cell maturation in diabetes271

In principle, T1D reversal needs to be poised to undertake therapeutic intervention directed at the repair of pancreatic insulin-producing β-cells that were destroyed in T1D patients.272 This notion forms an important part of current research attempting to understand the underlying mechanisms of T1D pathology, thereby offering hope for new therapeutic avenues and even a cure. The regeneration of β-cells is considered the ultimate objective of T1D therapy, which could transform the approach to managing the disease or possibly eliminate it altogether. Achieving β-cell regeneration presents significant possibilities for novel therapies.273 If scientists succeed in finding ways to regenerate or substitute these lost cells, it could result in groundbreaking treatments for T1D. A noteworthy strategy for generating functional β-cells involves transforming pancreatic stem cells into insulin-producing cells.274,275 Various crucial signaling pathways play a vital role in guiding these stem cells to differentiate into β-cells, providing researchers with several targets for therapeutic approaches.266,276 NGN3 is one of the major signaling pathways involved in this process, and it is important in the development of pancreatic endocrine tissue.277,278 NGN3 is a transcription factor that directs progenitor cells to differentiate into hormone-secreting cells, including β-cells. Pancreatic stem or progenitor cells are rendered insulin-secreting β-cells by NGN3 expression, which can bring about the restoration of insulin production in T1D patients.236,237

To further elaborate, recent studies have highlighted the essential function of NGN3 in reinstating endocrine differentiation in the adult pancreas during instances of injury or regenerative cues.279,280 Models of pancreatic damage or diabetes suggest that brief activation of NGN3 in ductal or acinar cells can reactivate an endocrine differentiation pathway and produce insulin-secreting cells.281,282 Induced NGN3 expression in pancreatic duct cells has been shown to lead to the formation of glucose-responsive β-like cells in mouse models.283,284 Moreover, lineage-tracing investigations have confirmed that reactivated adult ductal epithelium serves as a source of endocrine progenitors when stimulated with NGN3.285,286 Importantly, NGN3 has been integrated with other key transcription factors, such as Pdx1 and V-maf musculoaponeurotic fibrosarcoma oncogene homolog A, in reprogramming strategies to convert non-β pancreatic cells or even intestinal cells into functional β-cells.287,288 Moreover, organoid and stem cell-derived pancreatic progenitor models have demonstrated that NGN3 induction is a vital step for committing to the endocrine lineage, and its appropriate expression significantly enhances the effectiveness of generating insulin-positive cells in vitro.289,290 Collectively, this information highlights that NGN3 is essential not only in embryonic development but can also be utilized in adult regenerative contexts to restore β-cell mass and function, suggesting potential treatments for diabetes.

Importantly, the Wnt/β-catenin signaling pathway, along with NGN3, affects cell fate, growth, and differentiation levels throughout pancreatic development.291,292 When activated, this pathway promotes the self-renewal and proliferation of pancreatic progenitor cells, resulting in a greater number of potential β-cell precursors. Furthermore, this pathway is essential for maintaining stem cells in a progenitor-like state, which is crucial for their transformation into β-cells.293 Inducing Wnt signaling in β-cells is crucial for their continued proliferation and differentiation into functional β-cells, as it has been shown to be effective in promoting regeneration. Recent studies indicate that activating canonical Wnt/β-catenin signaling enhances the levels of key β-cell transcription factors such as Pdx1 and NK6 homeobox 1, which are vital for maintaining β-cell identity and facilitating insulin secretion.294,295 Additionally, modifying Wnt signaling in pancreatic organoid cultures has significantly improved the effectiveness of endocrine lineage specification, indicating its potential therapeutic benefit in β-cell replacement therapy.296,297

Key pathways in β-cell regeneration: Hippo/YAP, GLP-1, growth differentiation factor 11 (GDF11), and beyond

Another important pathway in β-cell regeneration is the Hippo/YAP signaling pathway, which plays a crucial role in pancreatic tissue repair.298,299 Typically, the Hippo pathway limits cell growth to maintain organ size. However, when this pathway is inhibited, it activates YAP, promoting the growth, survival, and advancement of β-cells.298,300 The triggering of YAP via the inhibition of the Hippo pathway underlines the core aspect of pancreatic stem cell differentiation and maturation to β-cells for tissue repair and regeneration through accelerated development.[299,301] Hormones, in addition to these signaling pathways, also trigger β-cell regeneration. The action of GLP-1 not only augments the secretion of insulin from β-cells and favors their growth and survival,302,303 it has been reported that GLP-1 is capable of increasing the mass and functionality of β-cells, particularly when those cells are under stress in the milieu of T1D.304,305 For instance, numerous studies indicate that GLP-1 improves β-cell mass and functionality, especially in T1D situations where β-cells face immune-related damage.306,307 The use of GLP-1 or GLP-1R agonists (GLP-1RAs) in T1D animal models has led to a significant increase in β-cell mass and improved insulin secretion, even with ongoing immune challenges.308,309 Additionally, it has been shown that GLP-1 enhances β-cell survival in diabetes by reducing inflammation and oxidative stress, key factors behind β-cell impairment and loss in T1D.310,311

In addition to benefiting mature β-cells, GLP-1 stimulates pancreatic progenitor cells to develop into insulin-producing β-cells.312 These regenerative processes are thought to be partly influenced by GLP-1’s regulation of central signaling routes, such as the phosphatidylinositol 3-kinase/protein kinase B pathway, which is crucial for cell survival and proliferation.313,314 The protective role of GLP-1 against β-cell apoptosis has been further substantiated by evidence showing that GLP-1RAs can counteract the negative effects of inflammatory cytokines commonly increased in the diabetic setting, such as tumor necrosis factor alpha and IL-1β, which are recognized as inducers of β-cell apoptosis.315,316 Scientists are thus using GLP-1 or GLP-1RAs to aid in β-cell regeneration from stem cells, potentially leading to improved insulin secretion and the development of new β-cells.317,318 Alongside these effects on β-cell growth and longevity, recent findings have also suggested that GLP-1 plays a role in enhancing the functional efficacy of β-cells, particularly under metabolic stress conditions. The administration of GLP-1 in animal models exhibiting insulin resistance has resulted in elevated insulin secretion and enhanced glucose tolerance, indicating that GLP-1 preserves β-cell function even during periods of metabolic stress on β-cells.319,320 This is particularly significant in T1D, where β-cell function is often compromised due to autoimmune damage and where the regeneration or maintenance of functional β-cells is essential for reinstating normal glucose balance. Moreover, GLP-1’s effect on β-cell regeneration extends beyond β-cell proliferation and survival to also enhance β-cell plasticity.295,321 Studies have shown that GLP-1RAs can convert α-cells into insulin-producing β-like cells in diabetic mice, thereby reinforcing the therapeutic promise of GLP-1 in promoting the regeneration of β-cells in diabetes.295 These findings suggest that GLP-1 could be a very potent therapeutic agent for increasing β-cell mass as well as improving the functional incorporation of newly generated β-cells into the pancreatic islet. In summary, these investigations highlight the multifaceted role of GLP-1 in enhancing β-cell regeneration and functionality, particularly in stressful environments such as T1D, and support its use as a potential treatment strategy for β-cell replacement and diabetes management.

On the other hand, GDF11, a probable candidate for β-cell regeneration, is yet another TGF-β family member.322 It stimulates pancreatic stem cells to become active β-cells.323,273 Several studies have shown that GDF11 increased both β-cell mass and glucose regulation in animals, suggesting that it might have a role in therapy for T1D.324 Furthermore, the tissue-rejuvenating properties of GDF11 may be especially advantageous for elderly individuals, as their capacity for pancreatic tissue regeneration usually declines with age.325,326

Together, these pathways, NGN3 activation, Wnt/β-catenin signaling, the Hippo/YAP pathway, GLP-1, and GDF11, create an important framework for regenerative medicine to reverse T1D effects. Figure 1 provides an overview of the integrated signaling pathways that regulate pancreatic β-cell regeneration and maintain their functional homeostasis. By manipulating these pathways, we may trigger β-cell regeneration in the pancreas and restore normal insulin production. These strategies offer hope for T1D patients and could change how diabetes is treated. However, alongside these regenerative approaches, it is essential to address the ongoing risk of autoimmune relapse, which remains an important obstacle. Various mechanisms have been proposed to either prevent or control such relapses, but this topic still requires more detailed investigation, which is beyond the scope of this narrative review. These strategies include inducing immune tolerance, such as using antigen-specific therapies aimed at re-educating the immune system rather than suppressing it entirely, and immune modulation with agents like anti-CD3 monoclonal antibodies, CTLA-4-Ig, and anti-CD20 treatments that target autoreactive T cells or boost Treg activity.327,328 Also, β-cell encapsulation techniques provide a physical barrier that shields transplanted β-cells from immune attack while allowing the exchange of nutrients and insulin.329,330 Advances in β-cell genetic engineering, employing methods like clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (hereinafter referred to as Cas9) to modify genes, aim to reduce their immunogenicity or produce local immunomodulatory factors, offering hope for longer-lasting β-cell survival.331,332 What’s more, combining immune modulation and β-cell replacement therapies may offer the most promising approach to achieve a cure and prevent relapses, eventually leading to sustained remission from T1D.333,334

Integrated signaling pathways governing pancreatic β-cell regeneration and functional homeostasis.
Fig. 1  Integrated signaling pathways governing pancreatic β-cell regeneration and functional homeostasis.

Schematic representation of interconnected molecular pathways that control the regeneration of pancreatic beta (β)-cells and maintain functional balance. Orange boxes indicate significant upstream signaling molecules (neurogenin 3 (NGN3), Wingless/Integrated (Wnt)/β-catenin pathway, glucagon-like peptide-1 (GLP-1), and GDF11 (growth differentiation factor 11)) that function at distinct yet comparable stages in β-cell regeneration. NGN3 is a fundamental helix-loop-helix transcription factor well-known for determining endocrine lineage during pancreatic development and facilitating the activation of pancreatic progenitor cells. Wnt/β-catenin signaling is a well-preserved cell-cell communication pathway that manages self-renewal and the growth of endocrine progenitors. GLP-1 is released following nutrient intake and is an incretin hormone that aids in the proliferation and survival of β-cells; GDF11 contributes to maintaining functional β-cell plasticity. All these routes lead to the creation of an endocrine lineage (bright blue) that develops into functional β-cells. The supportive intermediate processes are illustrated as progenitor cell differentiation, new β-cell formation, and stem cell stimulation (light blue boxes). These mechanisms ultimately aid in the growth and development of β-cells. The functional maturity of the β-cell then yields physiological results from a rise in β-cell mass, improved insulin secretion, and preservation of glucose homeostasis (orange boxes), contributing to the restoration of functional homeostasis. Color labels also signify functional classifications: orange denotes upstream signals and downstream physiological endpoints; light blue boxes illustrate transitional cellular events, whereas bright blue indicates critical lineage transitions.

Finally, besides stem cell–based approaches for β-cell replacement, a novel and highly promising strategy is emerging that aims to stimulate endogenous pancreatic cell regeneration.94,335 Therapies employing glucagon receptor (GCGR) monoclonal antibodies, such as evogliptin, leverage the glucagon signaling pathway—aligning with the classical view of diabetes as a bi-hormonal disease.336–338 GCGR monoclonal antibodies exert their effects by inhibiting glucagon action, leading to physiological changes characterized by elevated circulating amino acids and increased levels of gut-derived hormones such as GLP-1.339–341 These changes promote α-cell proliferation and facilitate the transdifferentiation of α-cells into insulin-producing β-like cells. Preclinical studies in rodent and non-human primate models have demonstrated substantial improvements in glycemic control and increases in functional β-cell mass, even in the absence of exogenous stem cell input.342,343 Early-phase clinical trials in individuals with T1D have reported reduced insulin requirements and improved glycemic regulation following administration of GCGR monoclonal antibodies.341 While further large-scale and long-term studies are necessary to confirm these findings, GCGR-targeted therapy represents a compelling endogenous regeneration strategy that could complement or potentially substitute for stem cell transplantation, particularly in patients retaining a functional α-cell population. Notably, Table 3 offers a comprehensive overview of current strategies for pancreatic β-cell regeneration, outlining their progression from basic research to clinical application.122,341,344–360 It summarizes available approaches—such as stem cell-derived islet cells, autologous and allogeneic transplantation, endogenous regeneration, gene editing, and encapsulation technologies—and organizes them by source, developmental stage, significant clinical trials or references, main benefits, and key challenges. The goal is to help readers understand the full translational pipeline and the clinical potential and challenges of these therapies for the treatment of T1D.

Table 3

Bench-to-bedside strategies for pancreatic β-cell regeneration

StrategyApproach/cell sourceDevelopment stagePivotal trials/referencesKey advantagesMajor challenges
Stem cell–derived β-cellsDifferentiation from hESCs/iPSCs (e.g., VX-880)Phase 1/2 Clinical TrialsThe FORWARD study (VX-880) accomplished insulin independence within a year, with many participants experiencing a reduction in severe hypoglycemia344,345Unlimited availability; expandable; significant glucose regulationImmune rejection: transplantation requires immunosuppression
Islet allotransplantationDonor-derived islets (Donislecel/Lantidra)FDA-approved since June 2023Donislecel received FDA approval with 21 out of 30 patients insulin independent for over a year; serious adverse events occurred in about 90%346,347Immediate glycemic restoration; no cell engineering neededLimited donors, necessity for continuous immunosuppression, and adverse events
Autologous iPSC-derived β-cellsPatient-derived iPSCs to β-cellsPreclinical / Early clinicalProof-of-concept studies are ongoing; it is not yet in late-stage trials348,349Personalized; lower immune rejection riskHigh cost, time-consuming, and QC and maturation challenges
Endogenous regeneration (drug-induced)GCGR (glucagon receptor antagonist) mAb (volagidemab), GLP-1 analogs, GABA (gamma-aminobutyric acid)Phase 1/2 Clinical TrialsVolagidemab Phase 2 study: Decrease in HbA1c, reduced insulin usage, yet primary endpoint not achieved341Non-invasive; stimulates α-to-β conversionEffects limited; safety issues with off-target outcomes; primary goals not achieved
Direct lineage reprogrammingα-cells/ductal cells → β-cells via TFsPreclinicalClassic studies122,350Uses endogenous plasticityIn vivo efficiency low; off-target reprogramming
MSC/HSC therapiesImmunomodulation, β-cell mass preservationPhase 1/2 Clinical TrialsMSC trials in new-onset T1D show immune regulation351,352Delay autoimmune degradationLimited β-cell regeneration
Encapsulation technologiesImmune-shielding of transplanted β-cellsClinical Trials (ViaCyte PEC-Encap)NCT02239354 trials are ongoing353,354Protects grafts without global immunosuppressionFibrosis, diffusion limits, vascularization
Gene editing (CRISPR/Cas9)β-cells engineered for hypoimmunityPreclinical / Early trialsVCTX210 for universal stem-cell islets355,356Precision modifications; potential to avoid rejectionRegulatory and ethical challenges
Organoids & 3D bioprintingScaffold-based islet cluster constructsPreclinicalStudies by Sun Lab and Kaminiski Lab357,358Improved maturation and insulin responseComplex fabrication; vascularization issues
Combined β-cell + immunotherapyβ-cell therapy plus immunomodulatorsPreclinical / Early trialsOngoing combo trials like Teplizumab + islet therapy359,360Targets regeneration and autoimmunityRisk of immune adverse events

Major technical hurdles hindering the clinical translation of stem cell therapy for T1D

Stem cell–based therapies represent a promising avenue for a long-term cure for T1D. However, their clinical translation faces several technical and biological hurdles. A major challenge lies in scaling up the production of insulin-producing beta-like cells from pluripotent stem cells.361,362 This process requires complex, multi-step differentiation protocols that are difficult to standardize and scale under Good Manufacturing Practice conditions. Additional issues include batch-to-batch variability, high reagent costs, and the need for tightly controlled culture environments, all of which contribute to inconsistent outcomes and elevated production costs, ultimately impacting therapy accessibility and affordability.363–365

Another significant obstacle is immune rejection. Since T1D is an autoimmune disease, even autologous or human leukocyte antigen (HLA)-matched allogeneic cells may be susceptible to immune attack.366,367 Encapsulation strategies have been developed to offer immunoprotection. Microencapsulation uses semi-permeable biomaterials such as alginate, chitosan hydrogel, or agarose to allow the exchange of essential molecules while preventing immune cell infiltration.61,368 However, inconsistencies in biomaterial composition can affect their mechanical properties and stability, leading to variable foreign body responses and potential graft failure.369,370 Macroencapsulation devices like the PEC-Direct enable direct vascularization, supporting graft survival and functional integration, but they do not provide immune shielding, necessitating lifelong immunosuppression.329,371 Moreover, macroencapsulation is associated with limited nutrient and oxygen diffusion, risk of cell aggregation and central necrosis, and suboptimal vascularization, all of which impair islet viability and function.372,373 Gene-editing strategies such as CRISPR have also been explored to reduce immune recognition by deleting HLA molecules or immune-activating surface proteins.374,375 While promising, these approaches raise valid concerns about off-target effects and long-term genomic stability.

Importantly, a persistent limitation in the field is the functional immaturity of stem cell–derived beta-like cells.376,377 These cells often exhibit fetal-like phenotypes characterized by poor glucose responsiveness and inadequate insulin secretion. Contributing factors may include incomplete differentiation, altered ion channel expression, and insufficient vascularization at transplantation sites, particularly ectopic locations lacking the appropriate pancreatic microenvironment.378,379 This suboptimal glucose-stimulated insulin secretion limits their ability to tightly regulate glycemia, undermining their therapeutic efficacy.380,381

Furthermore, differentiation protocols often generate heterogeneous cell populations, including off-target or incompletely differentiated cells. These may express markers of non-β-cell pancreatic or extrapancreatic lineages, raising concerns about ectopic hormone production, inappropriate signaling, or unintended paracrine interactions.382,383 Residual undifferentiated pluripotent stem cells within the graft pose a significant tumorigenic risk, such as teratoma formation, especially in long-term applications.384,385 These concerns highlight the necessity for rigorous purification and safety validation protocols before clinical use.

Although the study of immune checkpoint pathways, such as PD-1 and CTLA-4 ligands, offers promising avenues for immunomodulation in T1D, a critical appraisal reveals that their clinical translation is considerably more complex than initially anticipated. These pathways have been extensively studied in the context of cancer immunotherapy, primarily to reinvigorate exhausted T cells.386,387 However, in T1D, they may be more appropriately applied to restrain autoreactive immune responses targeting pancreatic β-cells. Despite this potential, checkpoint blockade carries significant risks, including off-target immune activation and systemic immune dysregulation, which could exacerbate existing autoimmunity or precipitate immune-related adverse events in other organ systems.388,389 Moreover, their efficacy in preserving or restoring endogenous β-cell mass in T1D remains unproven, particularly when used in isolation.

Parallel investigations into β-cell regeneration have focused on transcription factors such as NGN3, a key regulator of endocrine lineage specification during pancreatic development. Reactivation of NGN3 in adult pancreatic tissue holds theoretical promise for the de novo generation of insulin-producing cells.51,390 However, the therapeutic modulation of NGN3 and associated pathways (e.g., Notch, Wnt/β-catenin) is inherently challenging. NGN3 expression is tightly regulated in both temporal and spatial dimensions, and its ectopic or sustained activation can lead to aberrant differentiation or tumorigenesis.391,392 Additionally, successful regeneration requires a careful balance between progenitor cell proliferation and proper β-cell maturation and function, an outcome not yet reliably achieved by current approaches.393,394 Therefore, while these signaling pathways represent compelling therapeutic targets, their clinical implementation must prioritize safety and necessitate a nuanced understanding of their context-dependent activity in the diabetic pancreas.

Taken together, these intersecting biological, engineering, and regulatory challenges underscore the complexity of translating stem cell–based therapies into safe, scalable, and effective treatments for T1D.395,396 Addressing these issues will require sustained interdisciplinary innovation across both research and clinical practice.

Limitations

While advancements in regenerative medicine for T1D have led to some beneficial therapeutic solutions, there are still challenges in translating recent progress into clinical practice. A significant challenge is that β-like cells derived from stem cells continue to show functional immaturity; they typically display a fetal-like characteristic with restricted glucose responsiveness and compromised insulin secretion. Producing these cells on a large scale while adhering to Good Manufacturing Practice standards is technically challenging, frequently hindered by inconsistencies in differentiation efficiency, elevated costs, and prolonged protocols. Immune rejection continues to be a significant obstacle—autologous or HLA-matched cells can still face the risk of autoimmune reactivation. Although encapsulation technologies and gene-editing strategies have demonstrated potential, they bring about further complications, including limited nutrient diffusion, fibrotic overgrowth, and unintended genetic consequences. Additionally, existing differentiation protocols tend to result in mixed cell populations, which raises the likelihood of inappropriate hormone release and the potential for tumor formation. Reprogramming approaches encounter challenges related to stability, effectiveness, and safety, as excessive expression of lineage-defining transcription factors may result in partial conversion or oncogenic transformation. Immunotherapy techniques, like checkpoint blockade, are limited by their systemic immune system effects and their restricted ability to prevent recurrent β-cell autoimmunity. Additionally, one of the most significant obstacles is the insufficient integration of transplanted or reprogrammed β-like cells within the host pancreatic environment. Adequate vascularization, nerve supply, and cell-to-cell communication are vital for the long-term survival and functionality of these cells, yet these features are not adequately replicated in existing models.

Future directions

In the future, strategies aimed at regeneration for T1D will need to move beyond traditional models by embracing a more comprehensive, systems-level approach that integrates stem cell biology, immunoengineering, developmental endocrinology, and tissue bioengineering. A key aim in these regenerative efforts will be to elucidate the processes through which immature β-like cells develop into glucose-responsive, functionally effective insulin-producing cells. The developmental environment can be mimicked using biomimetic scaffolds alone, ECM hydrogels by themselves, or possibly through vascularized organ-on-chip systems, which may provide the necessary biochemical and mechanical cues for proper islet development and functionality.397,398 These culture systems might also have the capability to facilitate the co-differentiation of supporting islet-resident cell types, such as endothelial cells, pericytes, or pancreatic stellate cells, which play essential supportive roles by providing angiocrine signals, paracrine signals, and immune modulation.399,400

Concurrently, studies that utilize single-cell multi-omics and spatial transcriptomics for lineage tracing and fate mapping are necessary to gain a clearer understanding of the identity, plasticity, and differentiation capacity of adult pancreatic progenitor and non-β-cell lineages. This is important for developing more informed in situ regenerative therapy strategies, such as activating dormant progenitor-like states or orchestrating transdifferentiation pathways of either α-, δ-, or acinar cells into insulin-secreting cells through the application of transcription factor combinations or epigenetic modifiers. Moreover, reprogramming techniques should not only achieve lineage conversion but also guarantee that they encourage lasting phenotypic stability and controlled growth, as any improper growth could lead to hyperplasia or neoplasia.399,400

Understanding these strategies within the realm of immunology by combining tolerogenic and regenerative elements will be essential. There are emerging possibilities with tissue-engineered islets that are encapsulated with immunoregulatory ligands, local administration of checkpoint modulator-based methods to promote antigen-specific tolerance, and hybrid encapsulation systems that can release immunosuppressive cytokines or utilize agents like TGF-β or immune checkpoint agonists such as CTLA-4-Ig in optimal spatiotemporal release patterns. Furthermore, using neo-self signaling pathways to reprogram designed β-cells can help them evade immune recognition by showcasing “don’t-eat-me” signals like CD47 or create synthetic immunomodulatory networks to promote immune effector evasion, while simultaneously allowing host surveillance to overpower foreign pathogens.401,402

A critical yet underutilized area of research is the neuroendocrine regulation of regenerated β-cells.403,404 Successful biofunctionality requires the secretion of insulin triggered by hyperglycemia, along with autonomic nerve input and endocrine coordination with systemic hormones.405,406 There is an urgent need for research focused on the reorganization of neurovascular networks and how this may influence β-cell function and longevity. Similarly, it is essential to develop closed-loop systems that integrate biosensors to facilitate the smart delivery of various trophic and immunomodulatory factors capable of promoting regeneration in response to mechanical stimulation.407,408

Moreover, predictive modeling driven by artificial intelligence and based on individualized immunogenetic, transcriptomic, and metabolic information can be employed to categorize patients, anticipate the reactivation of autoimmunity, and customize regenerative strategies to improve their effectiveness and safety.409,410 Gene-editing technologies like base editors or prime editors could allow for precise modifications of loci associated with autoimmune susceptibility or even changes to β-cell immunogenicity while minimizing the risk of genotoxic effects.411,412 It will also be essential for ethical and regulatory frameworks to evolve in order to ensure that the ramifications of these personalized and potentially heritable treatments are safe, fair, and monitored for long-term safety.

Ultimately, future clinical trials need to implement adaptive and decentralized designs that allow for real-time biomarker analysis, immune profiling, and comprehensive multi-omics evaluations focusing not only on glycemic control but also on immune compatibility, graft integration, and the β-cell stress response.413,414 Collaborative global consortia and partnerships between the public and private sectors will be crucial for standardizing differentiation methods, potency evaluations, and long-term safety outcomes, which will help transition the field from a proof-of-concept stage to large-scale and equitable treatments. In conclusion, the path ahead necessitates collaboration among bioengineering, immunology, systems biology, and patient-centered precision medicine to create effective, safe, and sustainable cures for T1D.

Conclusions

Regenerative medicine aims to transform conventional T1D treatment by enabling the restoration of functional insulin-secreting β-cells. A particularly promising therapeutic strategy involves cellular reprogramming, which seeks to convert adult non-β pancreatic cells—such as α-cells or ductal epithelial cells—into insulin-producing β-like cells, potentially bypassing the need for exogenous stem cell sources. This approach leverages the inherent plasticity of pancreatic cell lineages and is driven by key developmental signaling pathways, including NGN3, Wnt/β-catenin, and Hippo/YAP. Recent advances, such as single-cell RNA sequencing and lineage tracing technologies, are critical for identifying rare progenitor subsets, mapping dynamic cell fate transitions, and confirming the fidelity and functionality of reprogrammed cells. Additionally, 3D pancreatic organoid models provide physiologically relevant platforms for assessing reprogramming efficiency, β-cell maturation, and disease modeling. Another major opportunity lies in engineering β-cells with immune-evasive properties, inspired by the natural immune privilege of α-cells. Epigenetic profiling has uncovered protective chromatin configurations and methylation landscapes in α-cells, which may be recapitulated in β-cells using CRISPR/Cas9 genome editing to downregulate autoantigen expression and enhance cell survival. Concurrently, immune-modulatory approaches, such as PD-1/CTLA-4 immune checkpoint inhibition and administration of anti-inflammatory cytokines (e.g., IL-10, TGF-β), are being developed to promote a localized immune-tolerant microenvironment. Nonetheless, several critical knowledge gaps impede clinical translation, including incomplete understanding of the molecular determinants of stable β-cell identity post-reprogramming; the long-term functionality and glucose responsiveness of reprogrammed cells under physiological stress; the mechanisms of immune escape; and the integration of engineered cells within native islet architecture, innervation, and vasculature. Moreover, issues of scalability, ethical considerations, and regulatory complexity must be systematically addressed to advance these innovations toward durable, clinically viable therapies for T1D.

Declarations

Acknowledgement

None.

Funding

This research was supported by Bandhan, Kolkata, India.

Conflict of interest

The authors have nothing to declare.

Authors’ contributions

Conceptualization, formal analysis, original draft preparation, project supervision, project administration, funding acquisition (SKC), and writing—review and editing (SKC, DC). Both authors have approved the final version and publication of the manuscript.

References

  1. Roy S, Pokharel P, Piganelli JD. Decoding the immune dance: Unraveling the interplay between beta cells and type 1 diabetes. Mol Metab 2024;88:101998 View Article PubMed/NCBI
  2. Roep BO, Thomaidou S, van Tienhoven R, Zaldumbide A. Type 1 diabetes mellitus as a disease of the β-cell (do not blame the immune system?). Nat Rev Endocrinol 2021;17(3):150-161 View Article PubMed/NCBI
  3. James EA, Joglekar AV, Linnemann AK, Russ HA, Kent SC. The beta cell-immune cell interface in type 1 diabetes (T1D). Mol Metab 2023;78:101809 View Article PubMed/NCBI
  4. Weiss M, Steiner DF, Philipson LH. Insulin Biosynthesis, Secretion, Structure, and Structure-Activity Relationships. In: Feingold KR, Anawalt B, Blackman MR, Chrousos G, de Herder WW, Dungan K, et al (eds). Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc; 2000 View Article PubMed/NCBI
  5. Liu M, Weiss MA, Arunagiri A, Yong J, Rege N, Sun J, et al. Biosynthesis, structure, and folding of the insulin precursor protein. Diabetes Obes Metab 2018;20(Suppl 2):28-50 View Article PubMed/NCBI
  6. Mittal R, Camick N, Lemos JRN, Hirani K. Gene-environment interaction in the pathophysiology of type 1 diabetes. Front Endocrinol (Lausanne) 2024;15:1335435 View Article PubMed/NCBI
  7. Minniakhmetov I, Yalaev B, Khusainova R, Bondarenko E, Melnichenko G, Dedov I, et al. Genetic and Epigenetic Aspects of Type 1 Diabetes Mellitus: Modern View on the Problem. Biomedicines 2024;12(2):399 View Article PubMed/NCBI
  8. Zajec A, Trebušak Podkrajšek K, Tesovnik T, Šket R, Čugalj Kern B, Jenko Bizjan B, et al. Pathogenesis of Type 1 Diabetes: Established Facts and New Insights. Genes (Basel) 2022;13(4):706 View Article PubMed/NCBI
  9. Shalitin S, Phillip M. New Curative Treatment Strategies and New Therapies for Type 1 Diabetes. Horm Res Paediatr 2025;98(4):369-370 View Article PubMed/NCBI
  10. Lizzo JM, Goyal A, Gupta V. Adult Diabetic Ketoacidosis. StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2025 View Article PubMed/NCBI
  11. Subramanian S, Baidal D. The Management of Type 1 Diabetes. In: Feingold KR, Anawalt B, Blackman MR, Chrousos G, de Herder WW, Dungan K, et al (eds). Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc; 2000 View Article PubMed/NCBI
  12. American Diabetes Association Professional Practice Committee. 14. Children and Adolescents: Standards of Care in Diabetes-2024. Diabetes Care 2024;47(Suppl 1):S258-S281 View Article PubMed/NCBI
  13. Leslie RD, Evans-Molina C, Freund-Brown J, Buzzetti R, Dabelea D, Gillespie KM, et al. Adult-Onset Type 1 Diabetes: Current Understanding and Challenges. Diabetes Care 2021;44(11):2449-2456 View Article PubMed/NCBI
  14. Lu X, Xie Q, Pan X, Zhang R, Zhang X, Peng G, et al. Type 2 diabetes mellitus in adults: pathogenesis, prevention and therapy. Signal Transduct Target Ther 2024;9(1):262 View Article PubMed/NCBI
  15. Wondmkun YT. Obesity, Insulin Resistance, and Type 2 Diabetes: Associations and Therapeutic Implications. Diabetes Metab Syndr Obes 2020;13:3611-3616 View Article PubMed/NCBI
  16. Eizirik DL, Szymczak F, Mallone R. Why does the immune system destroy pancreatic β-cells but not α-cells in type 1 diabetes?. Nat Rev Endocrinol 2023;19(7):425-434 View Article PubMed/NCBI
  17. Scherm MG, Wyatt RC, Serr I, Anz D, Richardson SJ, Daniel C. Beta cell and immune cell interactions in autoimmune type 1 diabetes: How they meet and talk to each other. Mol Metab 2022;64:101565 View Article PubMed/NCBI
  18. Carroll J, Chen J, Mittal R, Lemos JRN, Mittal M, Juneja S, et al. Decoding the Significance of Alpha Cell Function in the Pathophysiology of Type 1 Diabetes. Cells 2024;13(22):1914 View Article PubMed/NCBI
  19. Hilliard BK, Prendergast JE, Smith MJ. Dia-B-Ties: B Cells in the Islet-Immune-Cell Interface in T1D. Biomolecules 2025;15(3):332 View Article PubMed/NCBI
  20. Benkahla MA, Sabouri S, Kiosses WB, Rajendran S, Quesada-Masachs E, von Herrath MG. HLA class I hyper-expression unmasks beta cells but not alpha cells to the immune system in pre-diabetes. J Autoimmun 2021;119:102628 View Article PubMed/NCBI
  21. Kabakchieva P, Assyov Y, Gerasoudis S, Vasilev G, Peshevska-Sekulovska M, Sekulovski M, et al. Islet transplantation-immunological challenges and current perspectives. World J Transplant 2023;13(4):107-121 View Article PubMed/NCBI
  22. Ho BX, Teo AKK, Ng NHJ. Innovations in bio-engineering and cell-based approaches to address immunological challenges in islet transplantation. Front Immunol 2024;15:1375177 View Article PubMed/NCBI
  23. Wang Q, Huang YX, Liu L, Zhao XH, Sun Y, Mao X, et al. Pancreatic islet transplantation: current advances and challenges. Front Immunol 2024;15:1391504 View Article PubMed/NCBI
  24. Shi Y, Zhao YZ, Jiang Z, Wang Z, Wang Q, Kou L, et al. Immune-Protective Formulations and Process Strategies for Improved Survival and Function of Transplanted Islets. Front Immunol 2022;13:923241 View Article PubMed/NCBI
  25. Altabas V, Bulum T. Current Challenges in Pancreas and Islet Transplantation: A Scoping Review. Biomedicines 2024;12(12):2853 View Article PubMed/NCBI
  26. Bharadwaj HR, Tan JK, Ali SH, Shah MH, Nicholas A, Ahmed KAHM, et al. Exploring the current provision of pancreatic transplantations in low- and middle-income countries: Current landscape, challenges, future prospects. World J Gastrointest Pathophysiol 2024;15(6):94590 View Article PubMed/NCBI
  27. Wang X, Zeng Z, Li D, Wang K, Zhang W, Yu Y, et al. Advancements and Challenges in Immune Protection Strategies for Islet Transplantation. J Diabetes 2025;17(1):e70048 View Article PubMed/NCBI
  28. Paez-Mayorga J, Campa-Carranza JN, Capuani S, Hernandez N, Liu HC, Chua CYX, et al. Implantable niche with local immunosuppression for islet allotransplantation achieves type 1 diabetes reversal in rats. Nat Commun 2022;13(1):7951 View Article PubMed/NCBI
  29. Eldershaw S, Verma K, Croft W, Rai T, Kinsella FAM, Stephens C, et al. Erratum: Lymphopenia-induced lymphoproliferation drives activation of naive T cells and expansion of regulatory populations. iScience 2025;28(2):111958 View Article PubMed/NCBI
  30. Sheu TT, Chiang BL. Lymphopenia, Lymphopenia-Induced Proliferation, and Autoimmunity. Int J Mol Sci 2021;22(8):4152 View Article PubMed/NCBI
  31. Burrack AL, Martinov T, Fife BT. T Cell-Mediated Beta Cell Destruction: Autoimmunity and Alloimmunity in the Context of Type 1 Diabetes. Front Endocrinol (Lausanne) 2017;8:343 View Article PubMed/NCBI
  32. Langlois A, Pinget M, Kessler L, Bouzakri K. Islet Transplantation: Current Limitations and Challenges for Successful Outcomes. Cells 2024;13(21):1783 View Article PubMed/NCBI
  33. Song H, Li J, Yang H, Kong B, Xu Y, Li X, et al. Enhancement of functional insulin-producing cell differentiation from embryonic stem cells through MST1-silencing. Diabetol Metab Syndr 2025;17(1):93 View Article PubMed/NCBI
  34. Ghasemi Gojani E, Rai S, Norouzkhani F, Shujat S, Wang B, Li D, et al. Targeting β-Cell Plasticity: A Promising Approach for Diabetes Treatment. Curr Issues Mol Biol 2024;46(7):7621-7667 View Article PubMed/NCBI
  35. Al-Hasani K, Marikar SN, Kaipananickal H, Maxwell S, Okabe J, Khurana I, et al. EZH2 inhibitors promote β-like cell regeneration in young and adult type 1 diabetes donors. Signal Transduct Target Ther 2024;9(1):2 View Article PubMed/NCBI
  36. Mu-U-Min RBA, Diane A, Allouch A, Al-Siddiqi HH. Immune Evasion in Stem Cell-Based Diabetes Therapy-Current Strategies and Their Application in Clinical Trials. Biomedicines 2025;13(2):383 View Article PubMed/NCBI
  37. Hogrebe NJ, Ishahak M, Millman JR. Developments in stem cell-derived islet replacement therapy for treating type 1 diabetes. Cell Stem Cell 2023;30(5):530-548 View Article PubMed/NCBI
  38. Wang Y, McGarrigle J, Cook J, Rios P, Monica G, Chen Y, et al. The future of islet transplantation beyond the BLA approval: challenges and opportunities. Front Transplant 2025;4:1522409 View Article PubMed/NCBI
  39. Jacques K, Coles BLK, van der Kooy D. Pancreatic stem cells originate during the pancreatic progenitor developmental stage. Front Cell Dev Biol 2025;13:1521411 View Article PubMed/NCBI
  40. Jiang FX, Morahan G. Pancreatic stem cells remain unresolved. Stem Cells Dev 2014;23(23):2803-2812 View Article PubMed/NCBI
  41. Bonner-Weir S, Sharma A. Pancreatic stem cells. J Pathol 2002;197(4):519-526 View Article PubMed/NCBI
  42. Feng X, Zhang H, Yang S, Cui D, Wu Y, Qi X, et al. From stem cells to pancreatic β-cells: strategies, applications, and potential treatments for diabetes. Mol Cell Biochem 2025;480(1):173-190 View Article PubMed/NCBI
  43. Li P, Ou Q, Shi S, Shao C. Immunomodulatory properties of mesenchymal stem cells/dental stem cells and their therapeutic applications. Cell Mol Immunol 2023;20(6):558-569 View Article PubMed/NCBI
  44. Weiss ARR, Dahlke MH. Immunomodulation by Mesenchymal Stem Cells (MSCs): Mechanisms of Action of Living, Apoptotic, and Dead MSCs. Front Immunol 2019;10:1191 View Article PubMed/NCBI
  45. Song N, Scholtemeijer M, Shah K. Mesenchymal Stem Cell Immunomodulation: Mechanisms and Therapeutic Potential. Trends Pharmacol Sci 2020;41(9):653-664 View Article PubMed/NCBI
  46. Liu S, Liu F, Zhou Y, Jin B, Sun Q, Guo S. Immunosuppressive Property of MSCs Mediated by Cell Surface Receptors. Front Immunol 2020;11:1076 View Article PubMed/NCBI
  47. Silva IBB, Kimura CH, Colantoni VP, Sogayar MC. Stem cells differentiation into insulin-producing cells (IPCs): recent advances and current challenges. Stem Cell Res Ther 2022;13(1):309 View Article PubMed/NCBI
  48. Yang L, Hu ZM, Jiang FX, Wang W. Stem cell therapy for insulin-dependent diabetes: Are we still on the road?. World J Stem Cells 2022;14(7):503-512 View Article PubMed/NCBI
  49. Inoue R, Nishiyama K, Li J, Miyashita D, Ono M, Terauchi Y, et al. The Feasibility and Applicability of Stem Cell Therapy for the Cure of Type 1 Diabetes. Cells 2021;10(7):1589 View Article PubMed/NCBI
  50. Wang KL, Tao M, Wei TJ, Wei R. Pancreatic β cell regeneration induced by clinical and preclinical agents. World J Stem Cells 2021;13(1):64-77 View Article PubMed/NCBI
  51. Chernysheva МB, Ruchko ЕS, Karimova МV, Vorotelyak ЕA, Vasiliev АV. Development, regeneration, and physiological expansion of functional β-cells: Cellular sources and regulators. Front Cell Dev Biol 2024;12:1424278 View Article PubMed/NCBI
  52. Mansouri A. Development and regeneration in the endocrine pancreas. ISRN Endocrinol 2012;2012:640956 View Article PubMed/NCBI
  53. Høj K, Baldan J, Seymour PA, Rift CV, Hasselby JP, Sandelin A, et al. Age-Related Decline in Pancreas Regeneration Is Associated With an Increased Proinflammatory Response to Injury. Gastro Hep Adv 2024;3(7):973-985 View Article PubMed/NCBI
  54. Spears E, Serafimidis I, Powers AC, Gavalas A. Debates in Pancreatic Beta Cell Biology: Proliferation Versus Progenitor Differentiation and Transdifferentiation in Restoring β Cell Mass. Front Endocrinol (Lausanne) 2021;12:722250 View Article PubMed/NCBI
  55. Zook HN, Quijano JC, Ortiz JA, Donohue C, Lopez K, Li W, et al. Activation of ductal progenitor-like cells from adult human pancreas requires extracellular matrix protein signaling. iScience 2024;27(3):109237 View Article PubMed/NCBI
  56. Mahla RS. Stem Cells Applications in Regenerative Medicine and Disease Therapeutics. Int J Cell Biol 2016;2016:6940283 View Article PubMed/NCBI
  57. Hussen BM, Taheri M, Yashooa RK, Abdullah GH, Abdullah SR, Kheder RK, et al. Revolutionizing medicine: recent developments and future prospects in stem-cell therapy. Int J Surg 2024;110(12):8002-8024 View Article PubMed/NCBI
  58. Kuang J, Huang T, Pei D. The Art of Reprogramming for Regenerative Medicine. Front Cell Dev Biol 2022;10:927555 View Article PubMed/NCBI
  59. Kalo E, Read S, Ahlenstiel G. Reprogramming-Evolving Path to Functional Surrogate β-Cells. Cells 2022;11(18):2813 View Article PubMed/NCBI
  60. Rohban R, Martins CP, Esni F. Advanced therapy to cure diabetes: mission impossible is now possible?. Front Cell Dev Biol 2024;12:1484859 View Article PubMed/NCBI
  61. Abdalla MMI. Advancing diabetes management: Exploring pancreatic beta-cell restoration’s potential and challenges. World J Gastroenterol 2024;30(40):4339-4353 View Article PubMed/NCBI
  62. Kumar D, Tanwar R. World’s first: stem cell therapy reverses diabetes. Stem Cell Res Ther 2024;15(1):487 View Article PubMed/NCBI
  63. Abusalah MAH, Abd Rahman ENSE, Choudhary OP, Priyanka. Evolving trends in stem cell therapy: an emerging and promising approach against various diseases. Int J Surg 2024;110(11):6862-6868 View Article PubMed/NCBI
  64. Rubin de Celis MF, Bonner-Weir S. Reversing and modulating cellular senescence in beta cells, a new field of opportunities to treat diabetes. Front Endocrinol (Lausanne) 2023;14:1217729 View Article PubMed/NCBI
  65. Varghese SS, Dhawan S. Senescence: a double-edged sword in beta-cell health and failure?. Front Endocrinol (Lausanne) 2023;14:1196460 View Article PubMed/NCBI
  66. Webster KL, Mirmira RG. Beta cell dedifferentiation in type 1 diabetes: sacrificing function for survival?. Front Endocrinol (Lausanne) 2024;15:1427723 View Article PubMed/NCBI
  67. Wang W, Zhang C. Targeting β-cell dedifferentiation and transdifferentiation: opportunities and challenges. Endocr Connect 2021;10(8):R213-R228 View Article PubMed/NCBI
  68. Cerf ME. Beta cell dysfunction and insulin resistance. Front Endocrinol (Lausanne) 2013;4:37 View Article PubMed/NCBI
  69. Basile G, Qadir MMF, Mauvais-Jarvis F, Vetere A, Shoba V, Modell AE, et al. Emerging diabetes therapies: Bringing back the β-cells. Mol Metab 2022;60:101477 View Article PubMed/NCBI
  70. Niu F, Liu W, Ren Y, Tian Y, Shi W, Li M, et al. β-cell neogenesis: A rising star to rescue diabetes mellitus. J Adv Res 2024;62:71-89 View Article PubMed/NCBI
  71. Li N, Liu F, Yang P, Xiong F, Yu Q, Li J, et al. Aging and stress induced β cell senescence and its implication in diabetes development. Aging (Albany NY) 2019;11(21):9947-9959 View Article PubMed/NCBI
  72. Patra M, Klochendler A, Condiotti R, Kaffe B, Elgavish S, Drawshy Z, et al. Senescence of human pancreatic beta cells enhances functional maturation through chromatin reorganization and promotes interferon responsiveness. Nucleic Acids Res 2024;24 ;52(11):6298-6316 View Article PubMed/NCBI
  73. Motlagh RA, Pipella J, Thompson PJ. Exploring senescence as a modifier of β cell extracellular vesicles in type 1 diabetes. Front Endocrinol (Lausanne) 2024;15:1422279 View Article PubMed/NCBI
  74. Cha J, Aguayo-Mazzucato C, Thompson PJ. Pancreatic β-cell senescence in diabetes: mechanisms, markers and therapies. Front Endocrinol (Lausanne) 2023;14:1212716 View Article PubMed/NCBI
  75. Moon JE, Lee YN, Jeong S, Jun HR, Hoang MH, Jo Y, et al. Enhancing differentiation and functionality of insulin-producing cells derived from iPSCs using esterified collagen hydrogel for cell therapy in diabetes mellitus. Stem Cell Res Ther 2024;15(1):374 View Article PubMed/NCBI
  76. Piemonti L. The Last Mile in Beta-Cell Replacement Therapy for Type 1 Diabetes: Time to Grow Up. Transpl Int 2025;38:14565 View Article PubMed/NCBI
  77. Choi J, Cayabyab F, Perez H, Yoshihara E. Scaling Insulin-Producing Cells by Multiple Strategies. Endocrinol Metab (Seoul) 2024;39(2):191-205 View Article PubMed/NCBI
  78. Habener JF, Stanojevic V. α-cell role in β-cell generation and regeneration. Islets 2012;4(3):188-198 View Article PubMed/NCBI
  79. Xiao X, Guo P, Shiota C, Zhang T, Coudriet GM, Fischbach S, et al. Endogenous Reprogramming of Alpha Cells into Beta Cells, Induced by Viral Gene Therapy, Reverses Autoimmune Diabetes. Cell Stem Cell 2018;22(1):78-90.e4 View Article PubMed/NCBI
  80. Chera S, Herrera PL. Regeneration of pancreatic insulin-producing cells by in situ adaptive cell conversion. Curr Opin Genet Dev 2016;40:1-10 View Article PubMed/NCBI
  81. Zhang L, Pitcher LE, Yousefzadeh MJ, Niedernhofer LJ, Robbins PD, Zhu Y. Cellular senescence: a key therapeutic target in aging and diseases. J Clin Invest 2022;132(15):e158450 View Article PubMed/NCBI
  82. Passerini L, Forlani A, Gregori S. Advances in Regulatory Cell Therapy for Type 1 Diabetes: Emerging Strategies and Future Directions. Eur J Immunol 2025;55(5):e202451722 View Article PubMed/NCBI
  83. Berishvili E, Peloso A, Tomei AA, Pepper AR. The Future of Beta Cells Replacement in the Era of Regenerative Medicine and Organ Bioengineering. Transpl Int 2024;37:12885 View Article PubMed/NCBI
  84. Cavelti-Weder C, Li W, Zumsteg A, Stemann M, Yamada T, Bonner-Weir S, et al. Direct Reprogramming for Pancreatic Beta-Cells Using Key Developmental Genes. Curr Pathobiol Rep 2015;3(1):57-65 View Article PubMed/NCBI
  85. Nair GG, Tzanakakis ES, Hebrok M. Emerging routes to the generation of functional β-cells for diabetes mellitus cell therapy. Nat Rev Endocrinol 2020;16(9):506-518 View Article PubMed/NCBI
  86. Foster TP, Bruggeman BS, Haller MJ. Emerging Immunotherapies for Disease Modification of Type 1 Diabetes. Drugs 2025;85(4):457-473 View Article PubMed/NCBI
  87. Zook HN, Quijano JC, Ortiz JA, Donohue C, Erdem N, Ku HT. Protocol to study ductal progenitor-like cells from the adult human pancreas using 3D suspension and methylcellulose-based culture systems. STAR Protoc 2025;6(2):103847 View Article PubMed/NCBI
  88. Bhattachan P, Jeschke MG. SINGLE-CELL TRANSCRIPTOME ANALYSIS IN HEALTH AND DISEASE. Shock 2024;61(1):19-27 View Article PubMed/NCBI
  89. Guo J, Fu W. Immune regulation of islet homeostasis and adaptation. J Mol Cell Biol 2020;12(10):764-774 View Article PubMed/NCBI
  90. Stanojevic V, Habener JF. Evolving function and potential of pancreatic alpha cells. Best Pract Res Clin Endocrinol Metab 2015;29(6):859-871 View Article PubMed/NCBI
  91. Cao R, Tian H, Zhang Y, Liu G, Xu H, Rao G, et al. Signaling pathways and intervention for therapy of type 2 diabetes mellitus. MedComm (2020) 2023;4(3):e283 View Article PubMed/NCBI
  92. Wei L, Gao J, Wang L, Tao Q, Tu C. Hippo/YAP signaling pathway: a new therapeutic target for diabetes mellitus and vascular complications. Ther Adv Endocrinol Metab 2023;14:20420188231220134 View Article PubMed/NCBI
  93. Kim HS, Lee MK. β-Cell regeneration through the transdifferentiation of pancreatic cells: Pancreatic progenitor cells in the pancreas. J Diabetes Investig 2016;7(3):286-296 View Article PubMed/NCBI
  94. Vasavada RC, Dhawan S. Harnessing beta-cell replication: advancing molecular insights to regenerative therapies in diabetes. Front Endocrinol (Lausanne) 2025;16:1612576 View Article PubMed/NCBI
  95. Liu G, David BT, Trawczynski M, Fessler RG. Advances in Pluripotent Stem Cells: History, Mechanisms, Technologies, and Applications. Stem Cell Rev Rep 2020;16(1):3-32 View Article PubMed/NCBI
  96. Cerneckis J, Cai H, Shi Y. Induced pluripotent stem cells (iPSCs): molecular mechanisms of induction and applications. Signal Transduct Target Ther 2024;9(1):112 View Article PubMed/NCBI
  97. Alipio Z, Liao W, Roemer EJ, Waner M, Fink LM, Ward DC, et al. Reversal of hyperglycemia in diabetic mouse models using induced-pluripotent stem (iPS)-derived pancreatic beta-like cells. Proc Natl Acad Sci U S A 2010;107(30):13426-13431 View Article PubMed/NCBI
  98. Ghoneim MA, Gabr MM, El-Halawani SM, Refaie AF. Current status of stem cell therapy for type 1 diabetes: a critique and a prospective consideration. Stem Cell Res Ther 2024;15(1):23 View Article PubMed/NCBI
  99. Ren M, Shang C, Zhong X, Guo R, Lao G, Wang X, et al. Insulin-producing cells from embryonic stem cells rescues hyperglycemia via intra-spleen migration. Sci Rep 2014;4:7586 View Article PubMed/NCBI
  100. Nemati M, Ranjbar Omrani G, Ebrahimi B, Alizadeh A. Efficiency of Stem Cell (SC) Differentiation into Insulin-Producing Cells for Treating Diabetes: a Systematic Review. Stem Cells Int 2021;2021:6652915 View Article PubMed/NCBI
  101. Butler PC, Gale EA. Reversing type 1 diabetes with stem cell-derived islets: a step closer to the dream?. J Clin Invest 2022;132(3):e158305 View Article PubMed/NCBI
  102. Melton D. The promise of stem cell-derived islet replacement therapy. Diabetologia 2021;64(5):1030-1036 View Article PubMed/NCBI
  103. Dong S, Wu H. Regenerating β cells of the pancreas - potential developments in diabetes treatment. Expert Opin Biol Ther 2018;18(2):175-185 View Article PubMed/NCBI
  104. Basile G, Kulkarni RN, Morgan NG. How, When, and Where Do Human β-Cells Regenerate?. Curr Diab Rep 2019;19(8):48 View Article PubMed/NCBI
  105. Ogieuhi IJ, Agbo CE, Ajekiigbe VO, Anthony CS, Onyehalu JC, Nwankwo CK, et al. Stem cell-derived pancreatic beta cells: a step closer to functional diabetes treatment?. BMC Endocr Disord 2025;25(1):181 View Article PubMed/NCBI
  106. Brusko TM, Russ HA, Stabler CL. Strategies for durable β cell replacement in type 1 diabetes. Science 2021;373(6554):516-522 View Article PubMed/NCBI
  107. Aguayo-Mazzucato C, Bonner-Weir S. Pancreatic β Cell Regeneration as a Possible Therapy for Diabetes. Cell Metab 2018;27(1):57-67 View Article PubMed/NCBI
  108. Guney MA, Lorberbaum DS, Sussel L. Pancreatic β cell regeneration: To β or not to β. Curr Opin Physiol 2020;14:13-20 View Article PubMed/NCBI
  109. Rieck S, Kaestner KH. Expansion of beta-cell mass in response to pregnancy. Trends Endocrinol Metab 2010;21(3):151-158 View Article PubMed/NCBI
  110. Li Y, Zhu J, Yue C, Song S, Tian L, Wang Y. Recent advances in pancreatic α-cell transdifferentiation for diabetes therapy. Front Immunol 2025;16:1551372 View Article PubMed/NCBI
  111. Bonner-Weir S, Inada A, Yatoh S, Li WC, Aye T, Toschi E, et al. Transdifferentiation of pancreatic ductal cells to endocrine beta-cells. Biochem Soc Trans 2008;36(Pt 3):353-356 View Article PubMed/NCBI
  112. Li WC, Rukstalis JM, Nishimura W, Tchipashvili V, Habener JF, Sharma A, et al. Activation of pancreatic-duct-derived progenitor cells during pancreas regeneration in adult rats. J Cell Sci 2010;123(Pt 16):2792-2802 View Article PubMed/NCBI
  113. Bonner-Weir S, Toschi E, Inada A, Reitz P, Fonseca SY, Aye T, et al. The pancreatic ductal epithelium serves as a potential pool of progenitor cells. Pediatr Diabetes 2004;5(Suppl 2):16-22 View Article PubMed/NCBI
  114. Alvarez Fallas ME, Pedraza-Arevalo S, Cujba AM, Manea T, Lambert C, Morrugares R, et al. Stem/progenitor cells in normal physiology and disease of the pancreas. Mol Cell Endocrinol 2021;538:111459 View Article PubMed/NCBI
  115. Kim W, Shin YK, Kim BJ, Egan JM. Notch signaling in pancreatic endocrine cell and diabetes. Biochem Biophys Res Commun 2010;392(3):247-251 View Article PubMed/NCBI
  116. Hu C, Chen Y, Yin X, Xu R, Yin C, Wang C, et al. Pancreatic endocrine and exocrine signaling and crosstalk in physiological and pathological status. Signal Transduct Target Ther 2025;10(1):39 View Article PubMed/NCBI
  117. Talchai C, Xuan S, Lin HV, Sussel L, Accili D. Pancreatic β cell dedifferentiation as a mechanism of diabetic β cell failure. Cell 2012;150(6):1223-1234 View Article PubMed/NCBI
  118. Wang Z, York NW, Nichols CG, Remedi MS. Pancreatic β cell dedifferentiation in diabetes and redifferentiation following insulin therapy. Cell Metab 2014;19(5):872-882 View Article PubMed/NCBI
  119. Chakrabarti SK, Mirmira RG. Transcription factors direct the development and function of pancreatic beta cells. Trends Endocrinol Metab 2003;14(2):78-84 View Article PubMed/NCBI
  120. 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(1):240 View Article PubMed/NCBI
  121. Hang Y, Yamamoto T, Benninger RK, Brissova M, Guo M, Bush W, et al. The MafA transcription factor becomes essential to islet β-cells soon after birth. Diabetes 2014;63(6):1994-2005 View Article PubMed/NCBI
  122. Domínguez-Bendala J, Bonner-Weir S. Bend It Like Occam: Ductal Origin of New Islet Cells in Human Pancreas After Injury. Diabetes 2025;74(5):682-684 View Article PubMed/NCBI
  123. Dorisetty RK, Kiran SG, Umrani MR, Boindala S, Bhonde RR, Venkatesan V. Immuolocalization of nestin in pancreatic tissue of mice at different ages. World J Gastroenterol 2008;14(46):7112-7116 View Article PubMed/NCBI
  124. Ernst C, Christie BR. The putative neural stem cell marker, nestin, is expressed in heterogeneous cell types in the adult rat neocortex. Neuroscience 2006;138(1):183-188 View Article PubMed/NCBI
  125. Yanger K, Stanger BZ. Facultative stem cells in liver and pancreas: fact and fancy. Dev Dyn 2011;240(3):521-529 View Article PubMed/NCBI
  126. Parham LR, Johnson NM, Lengner CJ, Hamilton KE. High autophagic vesicle content marks facultative stem cells of the gut. Autophagy 2023;19(9):2611-2612 View Article PubMed/NCBI
  127. Arutyunyan IV, Fatkhudinov TK, Makarov AV, Elchaninov AV, Sukhikh GT. Regenerative medicine of pancreatic islets. World J Gastroenterol 2020;26(22):2948-2966 View Article PubMed/NCBI
  128. Liu T, Wang C, Wan C, Xiong J, Zhou F. Proliferation and differentiation of duct epithelial cells after partial pancreatectomy in rats. J Huazhong Univ Sci Technolog Med Sci 2006;26(5):567-569 View Article PubMed/NCBI
  129. Gu D, Sarvetnick N. A transgenic model for studying islet development. Recent Prog Horm Res 1994;49:161-165 View Article PubMed/NCBI
  130. Semerci F, Maletic-Savatic M. Transgenic mouse models for studying adult neurogenesis. Front Biol (Beijing) 2016;11(3):151-167 View Article PubMed/NCBI
  131. In’t Veld P. Insulitis in human type 1 diabetes: a comparison between patients and animal models. Semin Immunopathol 2014;36(5):569-579 View Article PubMed/NCBI
  132. Atkinson MA, Mirmira RG. The pathogenic “symphony” in type 1 diabetes: A disorder of the immune system, β cells, and exocrine pancreas. Cell Metab 2023;35(9):1500-1518 View Article PubMed/NCBI
  133. Domínguez-Bendala J, Qadir MMF, Pastori RL. Pancreatic Progenitors: There and Back Again. Trends Endocrinol Metab 2019;30(1):4-11 View Article PubMed/NCBI
  134. Soltani SM, O’Brien TD, Loganathan G, Bellin MD, Anazawa T, Tiwari M, et al. Severely fibrotic pancreases from young patients with chronic pancreatitis: evidence for a ductal origin of islet neogenesis. Acta Diabetol 2013;50(5):807-814 View Article PubMed/NCBI
  135. Napolitano T, Silvano S, Ayachi C, Plaisant M, Sousa-Da-Veiga A, Fofo H, et al. Wnt Pathway in Pancreatic Development and Pathophysiology. Cells 2023;12(4):565 View Article PubMed/NCBI
  136. Corritore E, Lee YS, Sokal EM, Lysy PA. β-cell replacement sources for type 1 diabetes: a focus on pancreatic ductal cells. Ther Adv Endocrinol Metab 2016;7(4):182-199 View Article PubMed/NCBI
  137. Gasa R, Mrejen C, Leachman N, Otten M, Barnes M, Wang J, et al. Proendocrine genes coordinate the pancreatic islet differentiation program in vitro. Proc Natl Acad Sci U S A 2004;101(36):13245-13250 View Article PubMed/NCBI
  138. Shamsi F, Parlato R, Collombat P, Mansouri A. A genetic mouse model for progressive ablation and regeneration of insulin producing beta-cells. Cell Cycle 2014;13(24):3948-3957 View Article PubMed/NCBI
  139. Karadimos MJ, Kapoor A, El Khattabi I, Sharma A. β-cell preservation and regeneration for diabetes treatment: where are we now?. Diabetes Manag (Lond) 2012;2(3):213-222 View Article PubMed/NCBI
  140. Dhawan S, Dirice E, Kulkarni RN, Bhushan A. Inhibition of TGF-β Signaling Promotes Human Pancreatic β-Cell Replication. Diabetes 2016;65(5):1208-1218 View Article PubMed/NCBI
  141. Lee JH, Lee JH, Rane SG. TGF-β Signaling in Pancreatic Islet β Cell Development and Function. Endocrinology 2021;162(3):bqaa233 View Article PubMed/NCBI
  142. Soldovieri L, Di Giuseppe G, Ciccarelli G, Quero G, Cinti F, Brunetti M, et al. An update on pancreatic regeneration mechanisms: Searching for paths to a cure for type 2 diabetes. Mol Metab 2023;74:101754 View Article PubMed/NCBI
  143. Reichert M, Rustgi AK. Pancreatic ductal cells in development, regeneration, and neoplasia. J Clin Invest 2011;121(12):4572-4578 View Article PubMed/NCBI
  144. Park IR, Chung YG, Won KC. Overcoming β-Cell Dysfunction in Type 2 Diabetes Mellitus: CD36 Inhibition and Antioxidant System. Diabetes Metab J 2025;49(1):1-12 View Article PubMed/NCBI
  145. Szablewski L. Changes in Cells Associated with Insulin Resistance. Int J Mol Sci 2024;25(4):2397 View Article PubMed/NCBI
  146. Wu Y, Aegerter P, Nipper M, Ramjit L, Liu J, Wang P. Hippo Signaling Pathway in Pancreas Development. Front Cell Dev Biol 2021;9:663906 View Article PubMed/NCBI
  147. Ardestani A, Maedler K. The Hippo Signaling Pathway in Pancreatic β-Cells: Functions and Regulations. Endocr Rev 2018;39(1):21-35 View Article PubMed/NCBI
  148. Ardestani A, Maedler K. Deathly triangle for pancreatic β-cells: Hippo pathway-MTORC1-autophagy. Autophagy 2021;17(12):4494-4496 View Article PubMed/NCBI
  149. Kang RB, Li Y, Rosselot C, Zhang T, Siddiq M, Rajbhandari P, et al. Single-nucleus RNA sequencing of human pancreatic islets identifies novel gene sets and distinguishes β-cell subpopulations with dynamic transcriptome profiles. Genome Med 2023;15(1):30 View Article PubMed/NCBI
  150. Fu H, Sun H, Kong H, Lou B, Chen H, Zhou Y, et al. Discoveries in Pancreatic Physiology and Disease Biology Using Single-Cell RNA Sequencing. Front Cell Dev Biol 2021;9:732776 View Article PubMed/NCBI
  151. Puri S, Folias AE, Hebrok M. Plasticity and dedifferentiation within the pancreas: development, homeostasis, and disease. Cell Stem Cell 2015;16(1):18-31 View Article PubMed/NCBI
  152. Tang X, Kuo T, Wei Z. Editorial: Pancreatic beta-cell dedifferentiation. Front Endocrinol (Lausanne) 2024;15:1524001 View Article PubMed/NCBI
  153. Ivich A, Davidson NR, Grieshober L, Li W, Hicks SC, Doherty JA, et al. Missing cell types in single-cell references impact deconvolution of bulk data but are detectable. Genome Biol 2025;26(1):86 View Article PubMed/NCBI
  154. Wruck W, Adjaye J. Single Cell Data Enables Dissecting Cell Types Present in Bulk Transcriptome Data. Stem Cells Dev 2025;34(1-2):17-25 View Article PubMed/NCBI
  155. Virk MS, Luo W, Sikes KJ, Li J, Plaas A, Cole BJ. Gene expression profiling of progenitor cells isolated from rat rotator cuff musculotendinous junction. BMC Musculoskelet Disord 2020;21(1):194 View Article PubMed/NCBI
  156. Miranda MA, Macias-Velasco JF, Lawson HA. Pancreatic β-cell heterogeneity in health and diabetes: classes, sources, and subtypes. Am J Physiol Endocrinol Metab 2021;320(4):E716-E731 View Article PubMed/NCBI
  157. Domínguez-Bendala J, Qadir MMF, Pastori RL. Temporal single-cell regeneration studies: the greatest thing since sliced pancreas?. Trends Endocrinol Metab 2021;32(7):433-443 View Article PubMed/NCBI
  158. Stanescu DE, Yu R, Won KJ, Stoffers DA. Single cell transcriptomic profiling of mouse pancreatic progenitors. Physiol Genomics 2017;49(2):105-114 View Article PubMed/NCBI
  159. McKimpson WM, Accili D. Reprogramming Cells to Make Insulin. J Endocr Soc 2019;3(6):1214-1226 View Article PubMed/NCBI
  160. Patel A, Rajgopal B, Jaiswal M. Various strategies to induce beta cell neogenesis: a comprehensive review for unravelling the potential future therapy for curing diabetes. Growth Factors 2025;43(2):69-96 View Article PubMed/NCBI
  161. Nakayama M, Moriya Y, Ueno H, Watanabe T, Hirabayashi H, Yamamoto S. Considerations in biodistribution evaluation of iPSC-derived cell therapy: A pancreatic islet cell case study. Mol Ther Methods Clin Dev 2025;33(3):101538 View Article PubMed/NCBI
  162. Habtezion A. Inflammation in acute and chronic pancreatitis. Curr Opin Gastroenterol 2015;31(5):395-399 View Article PubMed/NCBI
  163. Lange S, Inal JM. Animal Models of Human Disease 2.0. Int J Mol Sci 2024;25(24):13743 View Article PubMed/NCBI
  164. Acharjee S, Chauhan S, Pal R, Tomar RS. Mechanisms of DNA methylation and histone modifications. Prog Mol Biol Transl Sci 2023;197:51-92 View Article PubMed/NCBI
  165. Chen Y, Liang R, Li Y, Jiang L, Ma D, Luo Q, et al. Chromatin accessibility: biological functions, molecular mechanisms and therapeutic application. Signal Transduct Target Ther 2024;9(1):340 View Article PubMed/NCBI
  166. Diane A, Mu-U-Min RBA, Al-Siddiqi HH. Epigenetic memory as crucial contributing factor in directing the differentiation of human iPSC into pancreatic β-cells in vitro. Cell Tissue Res 2025;399(3):267-276 View Article PubMed/NCBI
  167. Suliman BA. Potential clinical implications of molecular mimicry-induced autoimmunity. Immun Inflamm Dis 2024;12(2):e1178 View Article PubMed/NCBI
  168. Lemos JRN, Hirani K, von Herrath M. Immunological and virological triggers of type 1 diabetes: insights and implications. Front Immunol 2023;14:1326711 View Article PubMed/NCBI
  169. Zhong F, Jiang Y. Endogenous Pancreatic β Cell Regeneration: A Potential Strategy for the Recovery of β Cell Deficiency in Diabetes. Front Endocrinol (Lausanne) 2019;10:101 View Article PubMed/NCBI
  170. Meng L, Wu H, Wu J, Ding P, He J, Sang M, et al. Mechanisms of immune checkpoint inhibitors: insights into the regulation of circular RNAS involved in cancer hallmarks. Cell Death Dis 2024;15(1):3 View Article PubMed/NCBI
  171. Kiaie SH, Sanaei MJ, Heshmati M, Asadzadeh Z, Azimi I, Hadidi S, et al. Immune checkpoints in targeted-immunotherapy of pancreatic cancer: New hope for clinical development. Acta Pharm Sin B 2021;11(5):1083-1097 View Article PubMed/NCBI
  172. Paluch C, Santos AM, Anzilotti C, Cornall RJ, Davis SJ. Immune Checkpoints as Therapeutic Targets in Autoimmunity. Front Immunol 2018;9:2306 View Article PubMed/NCBI
  173. Al-Qahtani AA, Alhamlan FS, Al-Qahtani AA. Pro-Inflammatory and Anti-Inflammatory Interleukins in Infectious Diseases: A Comprehensive Review. Trop Med Infect Dis 2024;9(1):13 View Article PubMed/NCBI
  174. Russell MA, Morgan NG. The impact of anti-inflammatory cytokines on the pancreatic β-cell. Islets 2014;6(3):e950547 View Article PubMed/NCBI
  175. Nepom GT, Ehlers M, Mandrup-Poulsen T. Anti-cytokine therapies in T1D: Concepts and strategies. Clin Immunol 2013;149(3):279-285 View Article PubMed/NCBI
  176. Zhao H, Huang X, Liu Z, Pu W, Lv Z, He L, et al. Pre-existing beta cells but not progenitors contribute to new beta cells in the adult pancreas. Nat Metab 2021;3(3):352-365 View Article PubMed/NCBI
  177. Hsu YC. Theory and Practice of Lineage Tracing. Stem Cells 2015;33(11):3197-3204 View Article PubMed/NCBI
  178. Grapin-Botton A. Three-dimensional pancreas organogenesis models. Diabetes Obes Metab 2016;18(Suppl 1):33-40 View Article PubMed/NCBI
  179. Petrauskas V, Damaseviciute R, Gulla A. Pancreatic 3D Organoids and Microfluidic Systems-Applicability and Utilization in Surgery: A Literature Review. Medicina (Kaunas) 2025;61(4):623 View Article PubMed/NCBI
  180. Tao B, Li X, Hao M, Tian T, Li Y, Li X, et al. Organoid-Guided Precision Medicine: From Bench to Bedside. MedComm (2020) 2025;6(5):e70195 View Article PubMed/NCBI
  181. El Nahas R, Al-Aghbar MA, Herrero L, van Panhuys N, Espino-Guarch M. Applications of Genome-Editing Technologies for Type 1 Diabetes. Int J Mol Sci 2023;25(1):344 View Article PubMed/NCBI
  182. Ansori AN, Antonius Y, Susilo RJ, Hayaza S, Kharisma VD, Parikesit AA, et al. Application of CRISPR-Cas9 genome editing technology in various fields: A review. Narra J 2023;3(2):e184 View Article PubMed/NCBI
  183. Qiu HY, Ji RJ, Zhang Y. Current advances of CRISPR-Cas technology in cell therapy. Cell Insight 2022;1(6):100067 View Article PubMed/NCBI
  184. Cuscino N, Castelbuono S, Centi C, Tinnirello R, Cimino M, Zito G, et al. A Bioartificial Device for the Encapsulation of Pancreatic β-Cells Using a Semipermeable Biocompatible Porous Membrane. J Clin Med 2025;14(5):1631 View Article PubMed/NCBI
  185. Kioulaphides S, García AJ. Encapsulation and immune protection for type 1 diabetes cell therapy. Adv Drug Deliv Rev 2024;207:115205 View Article PubMed/NCBI
  186. Popoviciu MS, Kaka N, Sethi Y, Patel N, Chopra H, Cavalu S. Type 1 Diabetes Mellitus and Autoimmune Diseases: A Critical Review of the Association and the Application of Personalized Medicine. J Pers Med 2023;13(3):422 View Article PubMed/NCBI
  187. Yue M, He X, Min X, Yang H, Xu H, Wu W, et al. The role of islet autoantigen-specific T cells in the onset and treatment of type 1 diabetes mellitus. Front Immunol 2024;15:1462384 View Article PubMed/NCBI
  188. Kawasaki E. Anti-Islet Autoantibodies in Type 1 Diabetes. Int J Mol Sci 2023;24(12):10012 View Article PubMed/NCBI
  189. Mitchell AM, Michels AW. Self-Antigens Targeted by Regulatory T Cells in Type 1 Diabetes. Int J Mol Sci 2022;23(6):3155 View Article PubMed/NCBI
  190. Lin TC, Lacorcia M, Mannering SI. Current and Emerging Assays for Measuring Human T-Cell Responses Against Beta-Cell Antigens in Type 1 Diabetes. Biomolecules 2025;15(3):384 View Article PubMed/NCBI
  191. Arvan P, Pietropaolo M, Ostrov D, Rhodes CJ. Islet autoantigens: structure, function, localization, and regulation. Cold Spring Harb Perspect Med 2012;2(8):a007658 View Article PubMed/NCBI
  192. Herold KC, Delong T, Perdigoto AL, Biru N, Brusko TM, Walker LSK. The immunology of type 1 diabetes. Nat Rev Immunol 2024;24(6):435-451 View Article PubMed/NCBI
  193. Jang DI, Lee AH, Shin HY, Song HR, Park JH, Kang TB, et al. The Role of Tumor Necrosis Factor Alpha (TNF-α) in Autoimmune Disease and Current TNF-α Inhibitors in Therapeutics. Int J Mol Sci 2021;22(5):2719 View Article PubMed/NCBI
  194. Sun L, Su Y, Jiao A, Wang X, Zhang B. T cells in health and disease. Signal Transduct Target Ther 2023;8(1):235 View Article PubMed/NCBI
  195. Junjappa RP, Patil P, Bhattarai KR, Kim HR, Chae HJ. IRE1α Implications in Endoplasmic Reticulum Stress-Mediated Development and Pathogenesis of Autoimmune Diseases. Front Immunol 2018;9:1289 View Article PubMed/NCBI
  196. Yuan S, She D, Jiang S, Deng N, Peng J, Ma L. Endoplasmic reticulum stress and therapeutic strategies in metabolic, neurodegenerative diseases and cancer. Mol Med 2024;30(1):40 View Article PubMed/NCBI
  197. Diane A, Allouch A, Mu-U-Min RBA, Al-Siddiqi HH. Endoplasmic reticulum stress in pancreatic β-cell dysfunctionality and diabetes mellitus: a promising target for generation of functional hPSC-derived β-cells in vitro. Front Endocrinol (Lausanne) 2024;15:1386471 View Article PubMed/NCBI
  198. Chen CW, Guan BJ, Alzahrani MR, Gao Z, Gao L, Bracey S, et al. Adaptation to chronic ER stress enforces pancreatic β-cell plasticity. Nat Commun 2022;13(1):4621 View Article PubMed/NCBI
  199. Brissova M, Haliyur R, Saunders D, Shrestha S, Dai C, Blodgett DM, et al. α Cell Function and Gene Expression Are Compromised in Type 1 Diabetes. Cell Rep 2018;22(10):2667-2676 View Article PubMed/NCBI
  200. Marroqui L, Masini M, Merino B, Grieco FA, Millard I, Dubois C, et al. Pancreatic α Cells are Resistant to Metabolic Stress-induced Apoptosis in Type 2 Diabetes. EBioMedicine 2015;2(5):378-385 View Article PubMed/NCBI
  201. Liu N, Liu T, Alim N, Zou J, Hu X, Zhang B, et al. 5-HT promotes pancreatic α-to-β cell transdifferentiation. Biochim Biophys Acta Mol Cell Res 2025;1872(5):119958 View Article PubMed/NCBI
  202. Docherty FM, Sussel L. Islet Regeneration: Endogenous and Exogenous Approaches. Int J Mol Sci 2021;22(7):3306 View Article PubMed/NCBI
  203. Aamodt KI, Powers AC. Signals in the pancreatic islet microenvironment influence β-cell proliferation. Diabetes Obes Metab 2017;19(Suppl 1):124-136 View Article PubMed/NCBI
  204. Yan LL, Ye LP, Chen YH, He SQ, Zhang CY, Mao XL, et al. The Influence of Microenvironment on Survival of Intraportal Transplanted Islets. Front Immunol 2022;13:849580 View Article PubMed/NCBI
  205. Bosco D, Armanet M, Morel P, Niclauss N, Sgroi A, Muller YD, et al. Unique arrangement of alpha- and beta-cells in human islets of Langerhans. Diabetes 2010;59(5):1202-1210 View Article PubMed/NCBI
  206. Donnor T, Sarkar S. Insulin- Pharmacology, Therapeutic Regimens and Principles of Intensive Insulin Therapy. In: Feingold KR, Anawalt B, Blackman MR, Chrousos G, de Herder WW, Dungan K, et al (eds). Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc; 2000 View Article PubMed/NCBI
  207. Martínez MS, Manzano A, Olivar LC, Nava M, Salazar J, D’Marco L, et al. The Role of the α Cell in the Pathogenesis of Diabetes: A World beyond the Mirror. Int J Mol Sci 2021;22(17):9504 View Article PubMed/NCBI
  208. Kale A, Azar M, Cheng V, Robertson H, Coulter S, Mehta PM, et al. Regulating islet stress responses through CD47 activation. Diabetologia 2025;68(6):1279-1297 View Article PubMed/NCBI
  209. Cusick MF, Libbey JE, Fujinami RS. Molecular mimicry as a mechanism of autoimmune disease. Clin Rev Allergy Immunol 2012;42(1):102-111 View Article PubMed/NCBI
  210. Thompson PJ, Pipella J, Rutter GA, Gaisano HY, Santamaria P. Islet autoimmunity in human type 1 diabetes: initiation and progression from the perspective of the beta cell. Diabetologia 2023;66(11):1971-1982 View Article PubMed/NCBI
  211. Adiguzel Y, Bogdanos DP, Shoenfeld Y. Molecular/antigenic mimicry and immunological cross-reactivity explains SARS-CoV-2-induced autoimmunity. Autoimmun Rev 2025;24(7):103811 View Article PubMed/NCBI
  212. Wong SK, Beckermann KE, Johnson DB, Das S. Combining anti-cytotoxic T-lymphocyte antigen 4 (CTLA-4) and -programmed cell death protein 1 (PD-1) agents for cancer immunotherapy. Expert Opin Biol Ther 2021;21(12):1623-1634 View Article PubMed/NCBI
  213. Gutic B, Bozanovic T, Mandic A, Dugalic S, Todorovic J, Stanisavljevic D, et al. Programmed cell death-1 and its ligands: Current knowledge and possibilities in immunotherapy. Clinics (Sao Paulo) 2023;78:100177 View Article PubMed/NCBI
  214. Hansen MS, Pokharel P, Piganelli J, Sussel L. The Chicken or the Egg Dilemma: Understanding the Interplay between the Immune System and the β Cell in Type 1 Diabetes. Cold Spring Harb Perspect Med 2025;15(4):a041591 View Article PubMed/NCBI
  215. Massagué J, Sheppard D. TGF-β signaling in health and disease. Cell 2023;186(19):4007-4037 View Article PubMed/NCBI
  216. Pearson JA, Wong FS, Wen L. The importance of the Non Obese Diabetic (NOD) mouse model in autoimmune diabetes. J Autoimmun 2016;66:76-88 View Article PubMed/NCBI
  217. Ebrahimpour Y, Khatami S, Saffar M, Fereidouni A, Biniaz Z, Erfanian N, et al. A Comprehensive Review of Novel Advances in Type 1 Diabetes Mellitus. J Diabetes 2025;17(8):e70120 View Article PubMed/NCBI
  218. Chaudhry A, Samstein RM, Treuting P, Liang Y, Pils MC, Heinrich JM, et al. Interleukin-10 signaling in regulatory T cells is required for suppression of Th17 cell-mediated inflammation. Immunity 2011;34(4):566-578 View Article PubMed/NCBI
  219. Bettini M, Bettini ML. Function, Failure, and the Future Potential of Tregs in Type 1 Diabetes. Diabetes 2021;70(6):1211-1219 View Article PubMed/NCBI
  220. Hardtke-Wolenski M, Landwehr-Kenzel S. Tipping the balance in autoimmunity: are regulatory t cells the cause, the cure, or both?. Mol Cell Pediatr 2024;11(1):3 View Article PubMed/NCBI
  221. Huang J, Tan Q, Tai N, Pearson JA, Li Y, Chao C, et al. IL-10 Deficiency Accelerates Type 1 Diabetes Development via Modulation of Innate and Adaptive Immune Cells and Gut Microbiota in BDC2.5 NOD Mice. Front Immunol 2021;12:702955 View Article PubMed/NCBI
  222. Bass LE, Bonami RH. Factors Governing B Cell Recognition of Autoantigen and Function in Type 1 Diabetes. Antibodies (Basel) 2024;13(2):27 View Article PubMed/NCBI
  223. Towns R, Pietropaolo M. GAD65 autoantibodies and its role as biomarker of Type 1 diabetes and Latent Autoimmune Diabetes in Adults (LADA). Drugs Future 2011;36(11):847 View Article PubMed/NCBI
  224. Chakrabarti SK, Francis J, Ziesmann SM, Garmey JC, Mirmira RG. Covalent histone modifications underlie the developmental regulation of insulin gene transcription in pancreatic beta cells. J Biol Chem 2003;278(26):23617-23623 View Article PubMed/NCBI
  225. Chakrabarti SK, James JC, Mirmira RG. Quantitative assessment of gene targeting in vitro and in vivo by the pancreatic transcription factor, Pdx1. Importance of chromatin structure in directing promoter binding. J Biol Chem 2002;277(15):13286-13293 View Article PubMed/NCBI
  226. Pollin G, Mathison AJ, de Assuncao TM, Thomas A, Zeighami A, Salmonson A, et al. Ehmt2 inactivation in pancreatic epithelial cells shapes the transcriptional landscape and inflammation response of the whole pancreas. Front Genet 2024;15:1412767 View Article PubMed/NCBI
  227. Kaimala S, Kumar CA, Allouh MZ, Ansari SA, Emerald BS. Epigenetic modifications in pancreas development, diabetes, and therapeutics. Med Res Rev 2022;42(3):1343-1371 View Article PubMed/NCBI
  228. Tan SYX, Zhang J, Tee WW. Epigenetic Regulation of Inflammatory Signaling and Inflammation-Induced Cancer. Front Cell Dev Biol 2022;10:931493 View Article PubMed/NCBI
  229. Pierre TH, Toren E, Kepple J, Hunter CS. Epigenetic Regulation of Pancreas Development and Function. Adv Anat Embryol Cell Biol 2024;239:1-30 View Article PubMed/NCBI
  230. Conrad E, Stein R, Hunter CS. Revealing transcription factors during human pancreatic β cell development. Trends Endocrinol Metab 2014;25(8):407-414 View Article PubMed/NCBI
  231. Darwish R, Alcibahy Y, Dhawan S, Butler AE, Moin ASM. Pancreatic β-cell remodeling in health and aging: Lessons from rodents and humans. Ageing Res Rev 2025;110:102815 View Article PubMed/NCBI
  232. Liu J, Xiao Q, Xiao J, Niu C, Li Y, Zhang X, et al. Wnt/β-catenin signalling: function, biological mechanisms, and therapeutic opportunities. Signal Transduct Target Ther 2022;7(1):3 View Article PubMed/NCBI
  233. Sileo P, Simonin C, Melnyk P, Chartier-Harlin MC, Cotelle P. Crosstalk between the Hippo Pathway and the Wnt Pathway in Huntington’s Disease and Other Neurodegenerative Disorders. Cells 2022;11(22):3631 View Article PubMed/NCBI
  234. Ebrahim N, Shakirova K, Dashinimaev E. PDX1 is the cornerstone of pancreatic β-cell functions and identity. Front Mol Biosci 2022;9:1091757 View Article PubMed/NCBI
  235. Zhang Y, Fang X, Wei J, Miao R, Wu H, Ma K, et al. PDX-1: A Promising Therapeutic Target to Reverse Diabetes. Biomolecules 2022;12(12):1785 View Article PubMed/NCBI
  236. Juksar J, Mijdam R, Bosman S, van Oudenaarden A, Carlotti F, de Koning EJP. Effects of Neurogenin 3 Induction on Endocrine Differentiation and Delamination in Adult Human Pancreatic Ductal Organoids. Transpl Int 2025;38:13422 View Article PubMed/NCBI
  237. Shilleh AH, Russ HA. Cell Replacement Therapy for Type 1 Diabetes Patients: Potential Mechanisms Leading to Stem-Cell-Derived Pancreatic β-Cell Loss upon Transplant. Cells 2023;12(5):698 View Article PubMed/NCBI
  238. 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(9):4478 View Article PubMed/NCBI
  239. Liang J, Chirikjian M, Pajvani UB, Bartolomé A. MafA Regulation in β-Cells: From Transcriptional to Post-Translational Mechanisms. Biomolecules 2022;12(4):535 View Article PubMed/NCBI
  240. Bohuslavova R, Fabriciova V, Smolik O, Lebrón-Mora L, Abaffy P, Benesova S, et al. NEUROD1 reinforces endocrine cell fate acquisition in pancreatic development. Nat Commun 2023;14(1):5554 View Article PubMed/NCBI
  241. Pavlinkova G, Smolik O. NEUROD1: transcriptional and epigenetic regulator of human and mouse neuronal and endocrine cell lineage programs. Front Cell Dev Biol 2024;12:1435546 View Article PubMed/NCBI
  242. Zhang Y, Parajuli KR, Fonseca VA, Wu H. PAX4 gene delivery improves β-cell function in human islets of Type II diabetes. Regen Med 2024;19(5):239-246 View Article PubMed/NCBI
  243. Liew CG, Shah NN, Briston SJ, Shepherd RM, Khoo CP, Dunne MJ, et al. PAX4 enhances beta-cell differentiation of human embryonic stem cells. PLoS One 2008;3(3):e1783 View Article PubMed/NCBI
  244. Wong A, Alejandro EU. Post translational modification regulation of transcription factors governing pancreatic β-cell identity and functional mass. Front Endocrinol (Lausanne) 2025;16:1562646 View Article PubMed/NCBI
  245. Courtney M, Gjernes E, Druelle N, Ravaud C, Vieira A, Ben-Othman N, et al. The inactivation of Arx in pancreatic α-cells triggers their neogenesis and conversion into functional β-like cells. PLoS Genet 2013;9(10):e1003934 View Article PubMed/NCBI
  246. Bohuslavova R, Fabriciova V, Lebrón-Mora L, Malfatti J, Smolik O, Valihrach L, et al. ISL1 controls pancreatic alpha cell fate and beta cell maturation. Cell Biosci 2023;13(1):53 View Article PubMed/NCBI
  247. Jiang H, Jiang FX. Human pluripotent stem cell-derived β cells: Truly immature islet β cells for type 1 diabetes therapy?. World J Stem Cells 2023;15(4):182-195 View Article PubMed/NCBI
  248. Zhang T, Kim DH, Xiao X, Lee S, Gong Z, Muzumdar R, et al. FoxO1 Plays an Important Role in Regulating β-Cell Compensation for Insulin Resistance in Male Mice. Endocrinology 2016;157(3):1055-1070 View Article PubMed/NCBI
  249. Boland BB, Rhodes CJ, Grimsby JS. The dynamic plasticity of insulin production in β-cells. Mol Metab 2017;6(9):958-973 View Article PubMed/NCBI
  250. Puri S, Maachi H, Nair G, Russ HA, Chen R, Pulimeno P, et al. Sox9 regulates alternative splicing and pancreatic beta cell function. Nat Commun 2024;15(1):588 View Article PubMed/NCBI
  251. Kashbour M, Abdelmalik A, Yassin MNA, Abed M, Aldieb E, Abdullah DM, et al. Mesenchymal stem cell-based therapy for type 1 & 2 diabetes mellitus patients: a systematic review and meta-analysis of randomized controlled trials. Diabetol Metab Syndr 2025;17(1):189 View Article PubMed/NCBI
  252. Ding L, Cai M, Chen L, Yan H, Lu S, Pang S, et al. Identification and functional study of GATA4 gene regulatory variants in type 2 diabetes mellitus. BMC Endocr Disord 2021;21(1):73 View Article PubMed/NCBI
  253. Millman JR, Xie C, Van Dervort A, Gürtler M, Pagliuca FW, Melton DA. Corrigendum: Generation of stem cell-derived β-cells from patients with type 1 diabetes. Nat Commun 2016;7:12379 View Article PubMed/NCBI
  254. Zhao Q, Ding L, Yang Y, Sun J, Wang M, Li X, et al. Clinical Characteristics of Patients With HNF1-alpha MODY: A Literature Review and Retrospective Chart Review. Front Endocrinol (Lausanne) 2022;13:900489 View Article PubMed/NCBI
  255. Cui D, Feng X, Lei S, Zhang H, Hu W, Yang S, et al. Pancreatic β-cell failure, clinical implications, and therapeutic strategies in type 2 diabetes. Chin Med J (Engl) 2024;137(7):791-805 View Article PubMed/NCBI
  256. Heng BC, Zhang X, Aubel D, Bai Y, Li X, Wei Y, et al. An overview of signaling pathways regulating YAP/TAZ activity. Cell Mol Life Sci 2021;78(2):497-512 View Article PubMed/NCBI
  257. Jeong MG, Kim HK, Lee G, Won HY, Yoon DH, Hwang ES. TAZ promotes PDX1-mediated insulinogenesis. Cell Mol Life Sci 2022;79(3):186 View Article PubMed/NCBI
  258. Xue C, Chu Q, Shi Q, Zeng Y, Lu J, Li L. Wnt signaling pathways in biology and disease: mechanisms and therapeutic advances. Signal Transduct Target Ther 2025;10(1):106 View Article PubMed/NCBI
  259. Goode RA, Hum JM, Kalwat MA. Therapeutic Strategies Targeting Pancreatic Islet β-Cell Proliferation, Regeneration, and Replacement. Endocrinology 2022;164(1):bqac193 View Article PubMed/NCBI
  260. Wu HH, Li YL, Liu NJ, Yang Z, Tao XM, Du YP, et al. TCF7L2 regulates pancreatic β-cell function through PI3K/AKT signal pathway. Diabetol Metab Syndr 2019;11:55 View Article PubMed/NCBI
  261. Migliorini A, Lickert H. Beyond association: A functional role for Tcf7l2 in β-cell development. Mol Metab 2015;4(5):365-366 View Article PubMed/NCBI
  262. Bartolome A, Zhu C, Sussel L, Pajvani UB. Notch signaling dynamically regulates adult β cell proliferation and maturity. J Clin Invest 2019;129(1):268-280 View Article PubMed/NCBI
  263. Colarusso JL, Zhou Q. Direct Reprogramming of Different Cell Lineages into Pancreatic β-Like Cells. Cell Reprogram 2022;24(5):252-258 View Article PubMed/NCBI
  264. Mortlock RD, Georgia SK, Finley SD. Dynamic Regulation of JAK-STAT Signaling Through the Prolactin Receptor Predicted by Computational Modeling. Cell Mol Bioeng 2021;14(1):15-30 View Article PubMed/NCBI
  265. Dludla PV, Mabhida SE, Ziqubu K, Nkambule BB, Mazibuko-Mbeje SE, Hanser S, et al. Pancreatic β-cell dysfunction in type 2 diabetes: Implications of inflammation and oxidative stress. World J Diabetes 2023;14(3):130-146 View Article PubMed/NCBI
  266. Jiang WJ, Peng YC, Yang KM. Cellular signaling pathways regulating β-cell proliferation as a promising therapeutic target in the treatment of diabetes. Exp Ther Med 2018;16(4):3275-3285 View Article PubMed/NCBI
  267. Wang HL, Wang L, Zhao CY, Lan HY. Role of TGF-Beta Signaling in Beta Cell Proliferation and Function in Diabetes. Biomolecules 2022;12(3):373 View Article PubMed/NCBI
  268. Gao Y, Zhang R, Dai S, Zhang X, Li X, Bai C. Corrigendum: Role of TGF-β/Smad Pathway in the Transcription of Pancreas-Specific Genes During Beta Cell Differentiation. Front Cell Dev Biol 2020;8:614840 View Article PubMed/NCBI
  269. Starr L, Dutta S, Danne T, Karpen SR, Hutton C, Kowalski A. The Urgent Need for Breakthrough Therapies and a World Without Type 1 Diabetes. Diabetes Ther 2025;16(6):1063-1076 View Article PubMed/NCBI
  270. Zheng Z, Zong Y, Ma Y, Tian Y, Pang Y, Zhang C, et al. Glucagon-like peptide-1 receptor: mechanisms and advances in therapy. Signal Transduct Target Ther 2024;9(1):234 View Article PubMed/NCBI
  271. Wei T, Wei R, Hong T. Regeneration of β cells from cell phenotype conversion among the pancreatic endocrine cells. Chronic Dis Transl Med 2022;8(1):1-4 View Article PubMed/NCBI
  272. Fields S, Johnston M. When Cells Are the Cure: Diabetes and Stem Cells. Genetic Twists of Fate [Internet]. Cambridge (MA): MIT Press; 2010 View Article PubMed/NCBI
  273. Bartolomé A. The Pancreatic Beta Cell: Editorial. Biomolecules 2023;13(3):495 View Article PubMed/NCBI
  274. Arroyave F, Uscátegui Y, Lizcano F. From iPSCs to Pancreatic β Cells: Unveiling Molecular Pathways and Enhancements with Vitamin C and Retinoic Acid in Diabetes Research. Int J Mol Sci 2024;25(17):9654 View Article PubMed/NCBI
  275. Dahiya S, Saleh M, Rodriguez UA, Rajasundaram DR, Arbujas J, Hajihassani A, et al. Acinar to β-like cell conversion through inhibition of focal adhesion kinase. Nat Commun 2024;15(1):3740 View Article PubMed/NCBI
  276. Shirakawa J. Signaling pathways that regulate adaptive β-cell proliferation for the treatment of diabetes. J Diabetes Investig 2023;14(6):735-740 View Article PubMed/NCBI
  277. Manea T, Nelson JK, Garrone CM, Hansson K, Evans I, Behrens A, et al. USP7 controls NGN3 stability and pancreatic endocrine lineage development. Nat Commun 2023;14(1):2457 View Article PubMed/NCBI
  278. Wang S, Yan J, Anderson DA, Xu Y, Kanal MC, Cao Z, et al. Neurog3 gene dosage regulates allocation of endocrine and exocrine cell fates in the developing mouse pancreas. Dev Biol 2010;339(1):26-37 View Article PubMed/NCBI
  279. Yang L, Yu XX, Wang X, Jin CT, Xu CR. The expression order determines the pioneer functions of NGN3 and NEUROD1 in pancreatic endocrine differentiation. Sci Adv 2025;11(13):eadt4770 View Article PubMed/NCBI
  280. Zhang X, McGrath PS, Salomone J, Rahal M, McCauley HA, Schweitzer J, et al. A Comprehensive Structure-Function Study of Neurogenin3 Disease-Causing Alleles during Human Pancreas and Intestinal Organoid Development. Dev Cell 2019;50(3):367-380.e7 View Article PubMed/NCBI
  281. Okuno M, Minami K, Okumachi A, Miyawaki K, Yokoi N, Toyokuni S, et al. Generation of insulin-secreting cells from pancreatic acinar cells of animal models of type 1 diabetes. Am J Physiol Endocrinol Metab 2007;292(1):E158-E165 View Article PubMed/NCBI
  282. Lee J, Sugiyama T, Liu Y, Wang J, Gu X, Lei J, et al. Expansion and conversion of human pancreatic ductal cells into insulin-secreting endocrine cells. Elife 2013;2:e00940 View Article PubMed/NCBI
  283. Li L, Pandol SJ. Editorial: Interaction between endocrine and exocrine pancreas. Front Endocrinol (Lausanne) 2022;13:967066 View Article PubMed/NCBI
  284. Sheets TP, Park KE, Park CH, Swift SM, Powell A, Donovan DM, et al. Targeted Mutation of NGN3 Gene Disrupts Pancreatic Endocrine Cell Development in Pigs. Sci Rep 2018;8(1):3582 View Article PubMed/NCBI
  285. Gribben C, Lambert C, Messal HA, Hubber EL, Rackham C, Evans I, et al. Ductal Ngn3-expressing progenitors contribute to adult β cell neogenesis in the pancreas. Cell Stem Cell 2021;28(11):2000-2008.e4 View Article PubMed/NCBI
  286. Kerper N, Ashe S, Hebrok M. Pancreatic β-Cell Development and Regeneration. Cold Spring Harb Perspect Biol 2022;14(5):a040741 View Article PubMed/NCBI
  287. Jeyagaran A, Urbanczyk M, Layland SL, Weise F, Schenke-Layland K. Forward programming of hiPSCs towards beta-like cells using Ngn3, Pdx1, and MafA. Sci Rep 2024;14(1):13608 View Article PubMed/NCBI
  288. Jara MA, Werneck-De-Castro JP, Lubaczeuski C, Johnson JD, Bernal-Mizrachi E. Pancreatic and duodenal homeobox-1 (PDX1) contributes to β-cell mass expansion and proliferation induced by Akt/PKB pathway. Islets 2020;12(2):32-40 View Article PubMed/NCBI
  289. Greggio C, De Franceschi F, Grapin-Botton A. Concise reviews: In vitro-produced pancreas organogenesis models in three dimensions: self-organization from few stem cells or progenitors. Stem Cells 2015;33(1):8-14 View Article PubMed/NCBI
  290. Sandilya S, Singh S. Development of islet organoids from human induced pluripotent stem cells in a cross-linked collagen scaffold. Cell Regen 2021;10(1):38 View Article PubMed/NCBI
  291. Adeerjiang Y, Sidike A, Gan XX, Li QT, Jiang S. The Role of Wnt3a/β-Catenin/TCF7L2 Pathway in Diabetes and Cardiorenal Complications. Cardiorenal Med 2025;15(1):72-82 View Article PubMed/NCBI
  292. Maurice MM, Angers S. Mechanistic insights into Wnt-β-catenin pathway activation and signal transduction. Nat Rev Mol Cell Biol 2025;26(5):371-388 View Article PubMed/NCBI
  293. Sarabia-Sánchez MA, Robles-Flores M. WNT Signaling in Stem Cells: A Look into the Non-Canonical Pathway. Stem Cell Rev Rep 2024;20(1):52-66 View Article PubMed/NCBI
  294. Aigha II, Abdelalim EM. NKX6.1 transcription factor: a crucial regulator of pancreatic β cell development, identity, and proliferation. Stem Cell Res Ther 2020;11(1):459 View Article PubMed/NCBI
  295. Sali S, Azzam L, Jaro T, Ali AAG, Mardini A, Al-Dajani O, et al. A perfect islet: reviewing recent protocol developments and proposing strategies for stem cell derived functional pancreatic islets. Stem Cell Res Ther 2025;16(1):160 View Article PubMed/NCBI
  296. Bittenglova K, Habart D, Saudek F, Koblas T. The Potential of Pancreatic Organoids for Diabetes Research and Therapy. Islets 2021;13(5-6):85-105 View Article PubMed/NCBI
  297. Chmielowiec J, Szlachcic WJ, Yang D, Scavuzzo MA, Wamble K, Sarrion-Perdigones A, et al. Human pancreatic microenvironment promotes β-cell differentiation via non-canonical WNT5A/JNK and BMP signaling. Nat Commun 2022;13(1):1952 View Article PubMed/NCBI
  298. Wang X, Du J, Li H, Cao Z, Cheng Z, Wang Z. The Hippo signaling pathway modulates pancreatic tissue homeostasis. Cell Death Discov 2025;11(1):343 View Article PubMed/NCBI
  299. Fu M, Hu Y, Lan T, Guan KL, Luo T, Luo M. The Hippo signalling pathway and its implications in human health and diseases. Signal Transduct Target Ther 2022;7(1):376 View Article PubMed/NCBI
  300. Yu FX, Zhao B, Guan KL. Hippo Pathway in Organ Size Control, Tissue Homeostasis, and Cancer. Cell 2015;163(4):811-828 View Article PubMed/NCBI
  301. Rosado-Olivieri EA, Anderson K, Kenty JH, Melton DA. YAP inhibition enhances the differentiation of functional stem cell-derived insulin-producing β cells. Nat Commun 2019;10(1):1464 View Article PubMed/NCBI
  302. Olukorode JO, Orimoloye DA, Nwachukwu NO, Onwuzo CN, Oloyede PO, Fayemi T, et al. Recent Advances and Therapeutic Benefits of Glucagon-Like Peptide-1 (GLP-1) Agonists in the Management of Type 2 Diabetes and Associated Metabolic Disorders. Cureus 2024;16(10):e72080 View Article PubMed/NCBI
  303. Lymperopoulos A, Altsman VL, Stoicovy RA. Glucagon-like Peptide-1 Receptor (GLP-1R) Signaling: Making the Case for a Functionally G(s) Protein-Selective GPCR. Int J Mol Sci 2025;26(15):7239 View Article PubMed/NCBI
  304. Delrue C, Speeckaert MM. Mechanistic Pathways and Clinical Implications of GLP-1 Receptor Agonists in Type 1 Diabetes Management. Int J Mol Sci 2024;25(17):9351 View Article PubMed/NCBI
  305. Kaneto H, Kimura T, Shimoda M, Obata A, Sanada J, Fushimi Y, et al. Favorable Effects of GLP-1 Receptor Agonist against Pancreatic β-Cell Glucose Toxicity and the Development of Arteriosclerosis: “The Earlier, the Better” in Therapy with Incretin-Based Medicine. Int J Mol Sci 2021;22(15):7917 View Article PubMed/NCBI
  306. Müller TD, Finan B, Bloom SR, D’Alessio D, Drucker DJ, Flatt PR, et al. Glucagon-like peptide 1 (GLP-1). Mol Metab 2019;30:72-130 View Article PubMed/NCBI
  307. Al-Noshokaty TM, Abdelhamid R, Abdelmaksoud NM, Khaled A, Hossam M, Ahmed R, et al. Unlocking the multifaceted roles of GLP-1: Physiological functions and therapeutic potential. Toxicol Rep 2025;14:101895 View Article PubMed/NCBI
  308. Ma X, Liu Z, Ilyas I, Little PJ, Kamato D, Sahebka A, et al. GLP-1 receptor agonists (GLP-1RAs): cardiovascular actions and therapeutic potential. Int J Biol Sci 2021;17(8):2050-2068 View Article PubMed/NCBI
  309. Ellenbroek JH, Töns HA, Westerouen van Meeteren MJ, de Graaf N, Hanegraaf MA, Rabelink TJ, et al. Glucagon-like peptide-1 receptor agonist treatment reduces beta cell mass in normoglycaemic mice. Diabetologia 2013;56(9):1980-1986 View Article PubMed/NCBI
  310. Mehdi SF, Pusapati S, Anwar MS, Lohana D, Kumar P, Nandula SA, et al. Glucagon-like peptide-1: a multi-faceted anti-inflammatory agent. Front Immunol 2023;14:1148209 View Article PubMed/NCBI
  311. Petersen KE, Rakipovski G, Raun K, Lykkesfeldt J. Does Glucagon-like Peptide-1 Ameliorate Oxidative Stress in Diabetes? Evidence Based on Experimental and Clinical Studies. Curr Diabetes Rev 2016;12(4):331-358 View Article PubMed/NCBI
  312. Abraham EJ, Leech CA, Lin JC, Zulewski H, Habener JF. Insulinotropic hormone glucagon-like peptide-1 differentiation of human pancreatic islet-derived progenitor cells into insulin-producing cells. Endocrinology 2002;143(8):3152-3161 View Article PubMed/NCBI
  313. Marzook A, Tomas A, Jones B. The Interplay of Glucagon-Like Peptide-1 Receptor Trafficking and Signalling in Pancreatic Beta Cells. Front Endocrinol (Lausanne) 2021;12:678055 View Article PubMed/NCBI
  314. Yang Y, Choi PP, Smith WW, Xu W, Ma D, Cordner ZA, et al. Exendin-4 reduces food intake via the PI3K/AKT signaling pathway in the hypothalamus. Sci Rep 2017;7(1):6936 View Article PubMed/NCBI
  315. Pang J, Feng JN, Ling W, Jin T. The anti-inflammatory feature of glucagon-like peptide-1 and its based diabetes drugs-Therapeutic potential exploration in lung injury. Acta Pharm Sin B 2022;12(11):4040-4055 View Article PubMed/NCBI
  316. Alharbi SH. Anti-inflammatory role of glucagon-like peptide 1 receptor agonists and its clinical implications. Ther Adv Endocrinol Metab 2024;15:20420188231222367 View Article PubMed/NCBI
  317. Zhao X, Wang M, Wen Z, Lu Z, Cui L, Fu C, et al. GLP-1 Receptor Agonists: Beyond Their Pancreatic Effects. Front Endocrinol (Lausanne) 2021;12:721135 View Article PubMed/NCBI
  318. Andreasen CR, Andersen A, Knop FK, Vilsbøll T. How glucagon-like peptide 1 receptor agonists work. Endocr Connect 2021;10(7):R200-R212 View Article PubMed/NCBI
  319. Rosenstock J, Robins DA, Duffin KL, Wilson JM, Lin Y, Banerjee H, et al. Orforglipron, an oral non-peptide glucagon-like peptide-1 receptor agonist, improves markers of β-cell function and insulin sensitivity in type 2 diabetes. Diabetes Obes Metab 2025 View Article PubMed/NCBI
  320. Smith EP, An Z, Wagner C, Lewis AG, Cohen EB, Li B, et al. The role of β cell glucagon-like peptide-1 signaling in glucose regulation and response to diabetes drugs. Cell Metab 2014;19(6):1050-1057 View Article PubMed/NCBI
  321. Moon JH, Choe HJ, Lim S. Pancreatic beta-cell mass and function and therapeutic implications of using antidiabetic medications in type 2 diabetes. J Diabetes Investig 2024;15(6):669-683 View Article PubMed/NCBI
  322. Zhang C, Lin Y, Liu Q, He J, Xiang P, Wang D, et al. Growth differentiation factor 11 promotes differentiation of MSCs into endothelial-like cells for angiogenesis. J Cell Mol Med 2020;24(15):8703-8717 View Article PubMed/NCBI
  323. Zhang Y, Wei Y, Liu D, Liu F, Li X, Pan L, et al. Role of growth differentiation factor 11 in development, physiology and disease. Oncotarget 2017;8(46):81604-81616 View Article PubMed/NCBI
  324. Li H, Li Y, Xiang L, Zhang J, Zhu B, Xiang L, et al. GDF11 Attenuates Development of Type 2 Diabetes via Improvement of Islet β-Cell Function and Survival. Diabetes 2017;66(7):1914-1927 View Article PubMed/NCBI
  325. Ma Y, Liu Y, Han F, Qiu H, Shi J, Huang N, et al. Growth differentiation factor 11: a “rejuvenation factor” involved in regulation of age-related diseases?. Aging (Albany NY) 2021;13(8):12258-12272 View Article PubMed/NCBI
  326. Driss LB, Lian J, Walker RG, Howard JA, Thompson TB, Rubin LL, et al. GDF11 and aging biology - controversies resolved and pending. J Cardiovasc Aging 2023;3(4):42 View Article PubMed/NCBI
  327. Menon AP, Moreno B, Meraviglia-Crivelli D, Nonatelli F, Villanueva H, Barainka M, et al. Modulating T Cell Responses by Targeting CD3. Cancers (Basel) 2023;15(4):1189 View Article PubMed/NCBI
  328. Thakkar S, Chopra A, Nagendra L, Kalra S, Bhattacharya S. Teplizumab in Type 1 Diabetes Mellitus: An Updated Review. touchREV Endocrinol 2023;19(2):22-30 View Article PubMed/NCBI
  329. Photiadis SJ, Gologorsky RC, Sarode D. The Current Status of Bioartificial Pancreas Devices. ASAIO J 2021;67(4):370-381 View Article PubMed/NCBI
  330. Miller J, Perrier Q, Rengaraj A, Bowlby J, Byers L, Peveri E, et al. State of the Art of Bioengineering Approaches in Beta-Cell Replacement. Curr Transplant Rep 2025;12(1):17 View Article PubMed/NCBI
  331. Liu W, Li L, Jiang J, Wu M, Lin P. Applications and challenges of CRISPR-Cas gene-editing to disease treatment in clinics. Precis Clin Med 2021;4(3):179-191 View Article PubMed/NCBI
  332. Cetin B, Erendor F, Eksi YE, Sanlioglu AD, Sanlioglu S. Advancing CRISPR genome editing into gene therapy clinical trials: progress and future prospects. Expert Rev Mol Med 2025;27:e16 View Article PubMed/NCBI
  333. Jeun R. Immunotherapies for prevention and treatment of type 1 diabetes. Immunotherapy 2025;17(3):201-210 View Article PubMed/NCBI
  334. Bonilla-Bonilla JD, Chávez-Sánchez L, Legorreta-Haquet MV. Immunotherapies and their potential to preserve beta cells in type 1 diabetes: a review of current immunotherapy. Bol Med Hosp Infant Mex 2025;82(4):203-218 View Article PubMed/NCBI
  335. Powell CJ, Singer HD, Juarez AR, Kim RT, Kim E, Payzin-Dogru D, et al. Pancreatic injury induces β-cell regeneration in axolotl. Dev Dyn 2025 View Article PubMed/NCBI
  336. Jia Y, Liu Y, Feng L, Sun S, Sun G. Role of Glucagon and Its Receptor in the Pathogenesis of Diabetes. Front Endocrinol (Lausanne) 2022;13:928016 View Article PubMed/NCBI
  337. Yan H, Gu W, Yang J, Bi V, Shen Y, Lee E, et al. Fully human monoclonal antibodies antagonizing the glucagon receptor improve glucose homeostasis in mice and monkeys. J Pharmacol Exp Ther 2009;329(1):102-111 View Article PubMed/NCBI
  338. Sloop KW, Michael MD, Moyers JS. Glucagon as a target for the treatment of Type 2 diabetes. Expert Opin Ther Targets 2005;9(3):593-600 View Article PubMed/NCBI
  339. Novikoff A, Müller TD. The molecular pharmacology of glucagon agonists in diabetes and obesity. Peptides 2023;165:171003 View Article PubMed/NCBI
  340. Finan B, Capozzi ME, Campbell JE. Repositioning Glucagon Action in the Physiology and Pharmacology of Diabetes. Diabetes 2020;69(4):532-541 View Article PubMed/NCBI
  341. Pettus J, Boeder SC, Christiansen MP, Denham DS, Bailey TS, Akturk HK, et al. Glucagon receptor antagonist volagidemab in type 1 diabetes: a 12-week, randomized, double-blind, phase 2 trial. Nat Med 2022;28(10):2092-2099 View Article PubMed/NCBI
  342. Wei R, Gu L, Yang J, Yang K, Liu J, Le Y, et al. Antagonistic Glucagon Receptor Antibody Promotes α-Cell Proliferation and Increases β-Cell Mass in Diabetic Mice. iScience 2019;16:326-339 View Article PubMed/NCBI
  343. Wei T, Cui X, Jiang Y, Wang K, Wang D, Li F, 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(5):599-610 View Article PubMed/NCBI
  344. Yau AWN, Chia AYY. Advancements in diabetes research and stem cell therapy: a concise review. J Diabetes Metab Disord 2025;24(1):130 View Article PubMed/NCBI
  345. Al-Horani RA, Aliter KF, Aliter HF. Future is Brighter: New Potential Paradigm-Shifting Medications and Regimens for Diabetes and Obesity. Curr Diabetes Rev 2024;20(8):84-97 View Article PubMed/NCBI
  346. Giri O, Goldman JD. Donislecel: First Cellular Therapy to Treat Patients With Brittle Type 1 Diabetes. Clin Diabetes 2024;42(2):337-340 View Article PubMed/NCBI
  347. Parums DV. Editorial: First Regulatory Approval for Allogeneic Pancreatic Islet Beta Cell Infusion for Adult Patients with Type 1 Diabetes Mellitus. Med Sci Monit 2023;29:e941918 View Article PubMed/NCBI
  348. Marei HE. Stem cell therapy: a revolutionary cure or a pandora’s box. Stem Cell Res Ther 2025;16(1):255 View Article PubMed/NCBI
  349. Mohite P, Puri A, Dave R, Budar A, Munde S, Ghosh SB, et al. Unlocking the therapeutic potential: odyssey of induced pluripotent stem cells in precision cell therapies. Int J Surg 2024;110(10):6432-6455 View Article PubMed/NCBI
  350. Wang H, Yang Y, Liu J, Qian L. Direct cell reprogramming: approaches, mechanisms and progress. Nat Rev Mol Cell Biol 2021;22(6):410-424 View Article PubMed/NCBI
  351. Syed Khaja AS, Binsaleh NK, Qanash H, Alshetaiwi H, Ginawi IAM, Saleem M. Dysregulation and therapeutic prospects of regulatory T cells in type 1 diabetes. Acta Diabetol 2025;62(6):785-800 View Article PubMed/NCBI
  352. Izadi M, Sadr Hashemi Nejad A, Moazenchi M, Masoumi S, Rabbani A, Kompani F, et al. Mesenchymal stem cell transplantation in newly diagnosed type-1 diabetes patients: a phase I/II randomized placebo-controlled clinical trial. Stem Cell Res Ther 2022;13(1):264 View Article PubMed/NCBI
  353. Desai T, Shea LD. Advances in islet encapsulation technologies. Nat Rev Drug Discov 2017;16(5):367 View Article PubMed/NCBI
  354. Lee K, Aviles Vargas A, Bottino R, Wang Y. Islet Transplantation: Microencapsulation, Nanoencapsulation, and Hypoimmune Engineering. Wiley Interdiscip Rev Nanomed Nanobiotechnol 2025;17(3):e70016 View Article PubMed/NCBI
  355. Rech Tondin A, Lanzoni G. Islet Cell Replacement and Regeneration for Type 1 Diabetes: Current Developments and Future Prospects. BioDrugs 2025;39(2):261-280 View Article PubMed/NCBI
  356. Karpov DS, Sosnovtseva AO, Pylina SV, Bastrich AN, Petrova DA, Kovalev MA, et al. Challenges of CRISPR/Cas-Based Cell Therapy for Type 1 Diabetes: How Not to Engineer a “Trojan Horse”. Int J Mol Sci 2023;24(24):17320 View Article PubMed/NCBI
  357. Li Y, Xu M, Chen J, Huang J, Cao J, Chen H, et al. Ameliorating and refining islet organoids to illuminate treatment and pathogenesis of diabetes mellitus. Stem Cell Res Ther 2024;15(1):188 View Article PubMed/NCBI
  358. Klak M, Wszoła M, Berman A, Filip A, Kosowska A, Olkowska-Truchanowicz J, et al. Bioprinted 3D Bionic Scaffolds with Pancreatic Islets as a New Therapy for Type 1 Diabetes-Analysis of the Results of Preclinical Studies on a Mouse Model. J Funct Biomater 2023;14(7):371 View Article PubMed/NCBI
  359. Ajmal N, Bogart MC, Khan P, Max-Harry IM, Healy AM, Nunemaker CS. Identifying Promising Immunomodulators for Type 1 Diabetes (T1D) and Islet Transplantation. J Diabetes Res 2024;2024:5151171 View Article PubMed/NCBI
  360. Siddiqui K, Nawaz SS. Exploration of Immune Targets for Type 1 Diabetes and Latent Autoimmune Disease Immunotherapy. Immunotargets Ther 2023;12:91-103 View Article PubMed/NCBI
  361. Yu X, Liu C, Kuang Z, Song S, Tian L, Wang Y. Islet organoids: a new hope for islet transplantation in diabetes. Front Immunol 2024;15:1540209 View Article PubMed/NCBI
  362. Vähäkangas E, Saarimäki-Vire J, Montaser H, Lithovius V, Eurola S, Ibrahim H, et al. Stem-cell-derived beta cells mature metabolically upon murine engraftment. Diabetologia 2025;68(9):1997-2010 View Article PubMed/NCBI
  363. Jayaraman P, Lim R, Ng J, Vemuri MC. Acceleration of Translational Mesenchymal Stromal Cell Therapy Through Consistent Quality GMP Manufacturing. Front Cell Dev Biol 2021;9:648472 View Article PubMed/NCBI
  364. Česnik AB, Švajger U. The issue of heterogeneity of MSC-based advanced therapy medicinal products-a review. Front Cell Dev Biol 2024;12:1400347 View Article PubMed/NCBI
  365. Marcuzzi A, Maximova N. Editorial: Advances in stem cell therapy: new applications and innovative therapeutic approaches. Front Med (Lausanne) 2023;10:1225551 View Article PubMed/NCBI
  366. Kotsiri I, Xanthi M, Domazinaki CM, Magiorkinis E. The Role of Viral Infections in the Immunopathogenesis of Type 1 Diabetes Mellitus: A Narrative Review. Biology (Basel) 2025;14(8):981 View Article PubMed/NCBI
  367. Arivarasan VK, Diwakar D, Kamarudheen N, Loganathan K. Current approaches in CRISPR-Cas systems for diabetes. Prog Mol Biol Transl Sci 2025;210:95-125 View Article PubMed/NCBI
  368. Marikar SN, El-Osta A, Johnston A, Such G, Al-Hasani K. Microencapsulation-based cell therapies. Cell Mol Life Sci 2022;79(7):351 View Article PubMed/NCBI
  369. Mariani E, Lisignoli G, Borzì RM, Pulsatelli L. Biomaterials: Foreign Bodies or Tuners for the Immune Response?. Int J Mol Sci 2019;20(3):636 View Article PubMed/NCBI
  370. Gomes YVR, Tavares AA, Barbosa RC, Tomaz AF, Sousa WJB, Oliveira LCC, et al. Biological responses to biomaterials: a review. Braz J Med Biol Res 2025;58:e14599 View Article PubMed/NCBI
  371. Kumar D, Tanwar R, Gupta V. First-ever stem cell therapy restores insulin independence in type 1 diabetes: A medical milestone. World J Stem Cells 2025;17(7):106856 View Article PubMed/NCBI
  372. Fernandez SA, Champion KS, Danielczak L, Gasparrini M, Paraskevas S, Leask RL, et al. Engineering Vascularized Islet Macroencapsulation Devices: An in vitro Platform to Study Oxygen Transport in Perfused Immobilized Pancreatic Beta Cell Cultures. Front Bioeng Biotechnol 2022;10:884071 View Article PubMed/NCBI
  373. Shang KM, Suzuki T, Kato H, Toyoda T, Tai YC, Komatsu H. Oxygen dynamics and delivery strategies to enhance beta cell replacement therapy. Am J Physiol Cell Physiol 2025;328(5):C1667-C1684 View Article PubMed/NCBI
  374. Allemailem KS, Alsahli MA, Almatroudi A, Alrumaihi F, Al Abdulmonem W, Moawad AA, et al. Innovative Strategies of Reprogramming Immune System Cells by Targeting CRISPR/Cas9-Based Genome-Editing Tools: A New Era of Cancer Management. Int J Nanomedicine 2023;18:5531-5559 View Article PubMed/NCBI
  375. Pavlovic K, Tristán-Manzano M, Maldonado-Pérez N, Cortijo-Gutierrez M, Sánchez-Hernández S, Justicia-Lirio P, et al. Using Gene Editing Approaches to Fine-Tune the Immune System. Front Immunol 2020;11:570672 View Article PubMed/NCBI
  376. Sun ZY, Yu TY, Jiang FX, Wang W. Functional maturation of immature β cells: A roadblock for stem cell therapy for type 1 diabetes. World J Stem Cells 2021;13(3):193-207 View Article PubMed/NCBI
  377. Karimova MV, Gvazava IG, Vorotelyak EA. Overcoming the Limitations of Stem Cell-Derived Beta Cells. Biomolecules 2022;12(6):810 View Article PubMed/NCBI
  378. Anazawa T, Marubashi S, Kodama S, Goto M, Maruyama M, Eguchi H, et al. Efficacy and Safety of Allogeneic Islet Transplantation Demonstrated by a Multicenter Clinical Trial in Japan. Transplant Direct 2025;11(3):e1765 View Article PubMed/NCBI
  379. Thorngren J, Brboric A, Vasylovska S, Hjelmqvist D, Westermark GT, Saarimäki-Vire J, et al. Efficient Vascular and Neural Engraftment of Stem Cell-Derived Islets. Diabetes 2024;73(7):1127-1139 View Article PubMed/NCBI
  380. Deepa Maheshvare M, Raha S, König M, Pal D. A pathway model of glucose-stimulated insulin secretion in the pancreatic β-cell. Front Endocrinol (Lausanne) 2023;14:1185656 View Article PubMed/NCBI
  381. Molano RD, Pileggi A, Tse HM, Stabler CL, Fraker CA. A static glucose-stimulated insulin secretion (sGSIS) assay that is significantly predictive of time to diabetes reversal in the human islet bioassay. BMJ Open Diabetes Res Care 2024;12(2):e003897 View Article PubMed/NCBI
  382. Aldous N, Moin ASM, Abdelalim EM. Pancreatic β-cell heterogeneity in adult human islets and stem cell-derived islets. Cell Mol Life Sci 2023;80(6):176 View Article PubMed/NCBI
  383. Zikmund T, Fiorentino J, Penfold C, Stock M, Shpudeiko P, Agarwal G, et al. Differentiation success of reprogrammed cells is heterogeneous in vivo and modulated by somatic cell identity memory. Stem Cell Reports 2025;20(4):102447 View Article PubMed/NCBI
  384. Movahed AY, Bagheri R, Savatier P, Šarić T, Moradi S. Elimination of tumorigenic pluripotent stem cells from their differentiated cell therapy products: An important step toward ensuring safe cell therapy. Stem Cell Reports 2025;20(7):102543 View Article PubMed/NCBI
  385. Yasuda S, Bando K, Henry MP, Libertini S, Watanabe T, Bando H, et al. Detection of residual pluripotent stem cells in cell therapy products utilizing droplet digital PCR: an international multisite evaluation study. Stem Cells Transl Med 2024;13(10):1001-1014 View Article PubMed/NCBI
  386. Wojtukiewicz MZ, Rek MM, Karpowicz K, Górska M, Polityńska B, Wojtukiewicz AM, et al. Inhibitors of immune checkpoints-PD-1, PD-L1, CTLA-4-new opportunities for cancer patients and a new challenge for internists and general practitioners. Cancer Metastasis Rev 2021;40(3):949-982 View Article PubMed/NCBI
  387. Sun Q, Hong Z, Zhang C, Wang L, Han Z, Ma D. Immune checkpoint therapy for solid tumours: clinical dilemmas and future trends. Signal Transduct Target Ther 2023;8(1):320 View Article PubMed/NCBI
  388. Khan S, Gerber DE. Autoimmunity, checkpoint inhibitor therapy and immune-related adverse events: A review. Semin Cancer Biol 2020;64:93-101 View Article PubMed/NCBI
  389. Ramos-Casals M, Brahmer JR, Callahan MK, Flores-Chávez A, Keegan N, Khamashta MA, et al. Immune-related adverse events of checkpoint inhibitors. Nat Rev Dis Primers 2020;6(1):38 View Article PubMed/NCBI
  390. Britt M, Abdilmasih N, Rezanejad H. Pancreatic Ductal Cell Heterogeneity: Insights into the Potential for β-Cell Regeneration in Diabetes. Stem Cell Rev Rep 2025;21(4):953-963 View Article PubMed/NCBI
  391. Chugh RM, Bhanja P, Zitter R, Gunewardena S, Badkul R, Saha S. Modulation of β-Catenin promotes WNT expression in macrophages and mitigates intestinal injury. Cell Commun Signal 2025;23(1):78 View Article PubMed/NCBI
  392. Song P, Gao Z, Bao Y, Chen L, Huang Y, Liu Y, et al. Wnt/β-catenin signaling pathway in carcinogenesis and cancer therapy. J Hematol Oncol 2024;17(1):46 View Article PubMed/NCBI
  393. Massoz L, Bergemann D, Lavergne A, Reynders C, Désiront C, Goossens C, et al. Negative cell cycle regulation by calcineurin is necessary for proper beta cell regeneration in zebrafish. Elife 2024;12:RP88813 View Article PubMed/NCBI
  394. Vidman S, Ma YHE, Fullenkamp N, Plant GW. Human induced pluripotent stem cell-derived therapies for regeneration after central nervous system injury. Neural Regen Res 2025;20(11):3063-3075 View Article PubMed/NCBI
  395. Zuo DB, Wang CH, Sang M, Sun XD, Chen GP, Ji KK. Stem cell therapy for diabetes: Advances, prospects, and challenges. World J Diabetes 2025;16(7):107344 View Article PubMed/NCBI
  396. Raoufinia R, Rahimi HR, Saburi E, Moghbeli M. Advances and challenges of the cell-based therapies among diabetic patients. J Transl Med 2024;22(1):435 View Article PubMed/NCBI
  397. Smandri A, Al-Masawa ME, Hwei NM, Fauzi MB. ECM-derived biomaterials for regulating tissue multicellularity and maturation. iScience 2024;27(3):109141 View Article PubMed/NCBI
  398. Hsiung N, Ju Y, Yang K, Yang P, Zeng W, Zhao H, et al. Organoid-based tissue engineering for advanced tissue repair and reconstruction. Mater Today Bio 2025;33:102093 View Article PubMed/NCBI
  399. Aplin AC, Aghazadeh Y, Mohn OG, Hull-Meichle RL. Role of the Pancreatic Islet Microvasculature in Health and Disease. J Histochem Cytochem 2024;72(11-12):711-728 View Article PubMed/NCBI
  400. Craig-Schapiro R, Li G, Chen K, Gomez-Salinero JM, Nachman R, Kopacz A, et al. Single-cell atlas of human pancreatic islet and acinar endothelial cells in health and diabetes. Nat Commun 2025;16(1):1338 View Article PubMed/NCBI
  401. Gomes Alves Martins R, Tekin MM, Cragg MS, Roghanian A. Therapeutic targeting of tumour-associated macrophage receptors. Immunother Adv 2025;5(1):ltaf009 View Article PubMed/NCBI
  402. Di Spirito A, Balkhi S, Vivona V, Mortara L. Key immune cells and their crosstalk in the tumor microenvironment of bladder cancer: insights for innovative therapies. Explor Target Antitumor Ther 2025;6:1002304 View Article PubMed/NCBI
  403. Karampelias C, Liu KC, Tengholm A, Andersson O. Mechanistic insights and approaches for beta cell regeneration. Nat Chem Biol 2025;21(6):807-818 View Article PubMed/NCBI
  404. Steenblock C, Bornstein SR. GHRH in diabetes and metabolism. Rev Endocr Metab Disord 2025;26(3):413-426 View Article PubMed/NCBI
  405. Mirzadeh Z, Faber CL, Schwartz MW. Central Nervous System Control of Glucose Homeostasis: A Therapeutic Target for Type 2 Diabetes?. Annu Rev Pharmacol Toxicol 2022;62:55-84 View Article PubMed/NCBI
  406. Rahman MS, Hossain KS, Das S, Kundu S, Adegoke EO, Rahman MA, et al. Role of Insulin in Health and Disease: An Update. Int J Mol Sci 2021;22(12):6403 View Article PubMed/NCBI
  407. Cicha I, Priefer R, Severino P, Souto EB, Jain S. Biosensor-Integrated Drug Delivery Systems as New Materials for Biomedical Applications. Biomolecules 2022;12(9):1198 View Article PubMed/NCBI
  408. Basu P, Banerjee A, Okoro PD, Masoumi A, Kanjilal B, Akbari M, et al. Integration of Functional Polymers and Biosensors to Enhance Wound Healing. Adv Healthc Mater 2024;13(29):e2401461 View Article PubMed/NCBI
  409. Nosrati H, Nosrati M. Artificial Intelligence in Regenerative Medicine: Applications and Implications. Biomimetics (Basel) 2023;8(5):442 View Article PubMed/NCBI
  410. Dixon D, Sattar H, Moros N, Kesireddy SR, Ahsan H, Lakkimsetti M, et al. Unveiling the Influence of AI Predictive Analytics on Patient Outcomes: A Comprehensive Narrative Review. Cureus 2024;16(5):e59954 View Article PubMed/NCBI
  411. Laurent M, Geoffroy M, Pavani G, Guiraud S. CRISPR-Based Gene Therapies: From Preclinical to Clinical Treatments. Cells 2024;13(10):800 View Article PubMed/NCBI
  412. Xu W, Zhang S, Qin H, Yao K. From bench to bedside: cutting-edge applications of base editing and prime editing in precision medicine. J Transl Med 2024;22(1):1133 View Article PubMed/NCBI
  413. Guan H, Zhao S, Li J, Wang Y, Niu P, Zhang Y, et al. Exploring the design of clinical research studies on the efficacy mechanisms in type 2 diabetes mellitus. Front Endocrinol (Lausanne) 2024;15:1363877 View Article PubMed/NCBI
  414. Mohr AE, Ortega-Santos CP, Whisner CM, Klein-Seetharaman J, Jasbi P. Navigating Challenges and Opportunities in Multi-Omics Integration for Personalized Healthcare. Biomedicines 2024;12(7):1496 View Article PubMed/NCBI

About this Article

Cite this article
Chakrabarti SK, Chattopadhyay D. The Future of Type 1 Diabetes: Can Stem Cells Provide a Cure?Explor Res Hypothesis Med. 2025;10(4):e00029. doi: 10.14218/ERHM.2025.00029.
Copy Export to RIS Export to EndNote
Article History
Received Revised Accepted Published
June 20, 2025 July 29, 2025 August 8, 2025 September 16, 2025
DOI http://dx.doi.org/10.14218/ERHM.2025.00029