PERSPECTIVE article

Front. Immunol., 04 June 2025

Sec. Autoimmune and Autoinflammatory Disorders : Autoimmune Disorders

Volume 16 - 2025 | https://doi.org/10.3389/fimmu.2025.1599321

This article is part of the Research TopicExploring the Immune-Metabolic Network in DiabetesView all 4 articles

Interplay of hypoxia, immune dysregulation, and metabolic stress in pathophysiology of type 1 diabetes

Rahul Mittal,*Rahul Mittal1,2*Joana R. N. Lemos,Joana R. N. Lemos1,2Prem Chapagain,Prem Chapagain3,4Khemraj Hirani,*Khemraj Hirani1,2*
  • 1Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, United States
  • 2Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
  • 3Department of Physics, Florida International University, Miami, FL, United States
  • 4Biomolecular Sciences Institute, Florida International University, Miami, FL, United States

Type 1 diabetes (T1D) is an autoimmune disease characterized by the progressive destruction of pancreatic β-cells, leading to insulin deficiency and chronic hyperglycemia. While immune-mediated mechanisms of β-cell destruction are well-recognized, emerging evidence highlights hypoxia as a silent yet critical contributor to T1D pathogenesis. Hypoxia in the pancreatic islets arises from inflammation, vascular dysfunction, hyperglycemia, and immune cell infiltration, creating a microenvironment that exacerbates β-cell dysfunction and amplifies autoimmune responses. Hypoxia-inducible factors (HIFs) play a dual role in regulating adaptive and maladaptive responses to hypoxia, influencing β-cell survival, immune activation, and oxidative stress. Specifically, hypoxia promotes the polarization of macrophages toward a pro-inflammatory M1 phenotype, enhances the differentiation of Th17 cells, and impairs the function of regulatory T cells (Tregs), thereby shifting the immune landscape toward sustained autoimmunity. This perspective discusses the multifaceted role of hypoxia in driving immune dysregulation and β-cell vulnerability in T1D as well as highlights the need for innovative research approaches to target this pathway. We propose future directions that emphasize the development of advanced experimental models to mimic the interplay between hypoxia, hyperglycemia, and immune responses in clinically relevant conditions. Furthermore, we highlight the potential of therapeutic strategies that target hypoxia and its downstream effects to preserve β-cell function and modulate autoimmunity. Collaborative efforts across disciplines will be crucial to translating these insights into clinical innovations that improve outcomes for individuals with T1D.

Introduction

Type 1 diabetes (T1D) is an autoimmune disease characterized by the progressive destruction of insulin-producing β-cells within the pancreatic islets of Langerhans (13). This loss of β-cells results in an inability to produce insulin, a hormone critical for regulating blood glucose levels, necessitating lifelong insulin replacement therapy for affected individuals. While T1D is primarily recognized as an immune-mediated disorder driven by autoreactive T cells targeting β-cell antigens, the microenvironment within the islets plays an increasingly recognized role in modulating disease progression. Emerging evidence highlights hypoxia, a state of insufficient oxygen supply, as a key factor that contributes to β-cell dysfunction (4, 5). It also exacerbates immune activation, forming a vicious cycle that accelerates the destruction of β-cells (6).

Pancreatic islets are among the most vascularized tissues in the body, with a dense capillary network that ensures rapid oxygen and nutrient delivery to β-cells (7). This vascularization is essential for supporting the high metabolic demands of insulin production and secretion (8). However, in T1D, this intricate network is disrupted by multiple factors, including chronic inflammation, hyperglycemia, and immune cell infiltration (9). Inflammatory cytokines such as interleukin-1β (IL-1β) and tumor necrosis factor-alpha (TNF-α) damage endothelial cells as well as impair vascular function, while the recruitment of immune cells to the islets increases local oxygen consumption (10, 11). Hyperglycemia, a hallmark of T1D, further compounds these effects by inducing metabolic stress and promoting vascular dysfunction. Together, these factors create a hypoxic microenvironment within the islets (12).

The consequences of hypoxia in T1D extend beyond impaired oxygen supply. Hypoxia directly affects β-cell survival by inducing mitochondrial dysfunction, oxidative stress, and endoplasmic reticulum (ER) stress, all of which compromise insulin secretion and β-cell viability (13, 14) (Figure 1). Furthermore, hypoxia triggers maladaptive cellular responses mediated by hypoxia-inducible factors (HIFs), transcription factors that regulate genes involved in angiogenesis, metabolism, and inflammation (15) (Figure 1). While transient HIF activation can promote protective mechanisms, such as vascular remodeling and metabolic adaptation, chronic HIF signaling has been implicated in exacerbating β-cell apoptosis and inflammation (16, 17).

Figure 1
www.frontiersin.org

Figure 1. Hypoxia-Induced Pathways: Adaptive and Maladaptive Responses. A schematic representation regarding the role of Hypoxia-Inducible Factors (HIFs) in regulating cellular responses within a hypoxic microenvironment. Hypoxia stabilizes HIF-1α, leading to an adaptive response via upregulation of VEGF and GLUT1, promoting angiogenesis and glycolysis. However, chronic hypoxia induces maladaptive consequences, including ROS production, caspase activation, and apoptosis. Additionally, hypoxia triggers angiogenic imbalance (VEGF, PDGF, Ang1/Ang2) contributing to islet dysfunction. Inflammatory pathways (TNF-1α, IL-1β, IL-6, NF-κB) promote autoimmunity, further exacerbating tissue damage. HIF-1α – Hypoxia-inducible factor 1-alpha; VEGF – Vascular endothelial growth factor; GLUT1 – Glucose transporter 1; PDGF – Platelet-derived growth factor; Ang1/Ang2 – Angiopoietin-1 and Angiopoietin-2; ROS – Reactive oxygen species; TNF-1α – Tumor necrosis factor-alpha; IL-1β – Interleukin-1 beta; IL-6 – Interleukin-6; NF-κB – Nuclear factor-kappa B. Created with BioRender.com

In addition to its effects on β-cells, hypoxia significantly influences the immune microenvironment within the islets. Hypoxia-induced metabolic reprogramming of immune cells promotes a shift toward pro-inflammatory phenotypes, including Th17 cells and M1 macrophages, while impairing the function of regulatory T cells (Tregs) that are critical for maintaining immune tolerance (18). This pro-inflammatory milieu perpetuates autoimmune activity and amplifies β-cell destruction. Moreover, hypoxia-induced oxidative stress enhances the release of damage-associated molecular patterns (DAMPs) and the formation of neoantigens, further driving immune activation (19).

Despite advances in understanding the immune mechanisms of T1D, the role of hypoxia as a driver of both β-cell dysfunction and immune dysregulation has been underexplored. Addressing hypoxia in T1D represents a promising therapeutic avenue, as it offers the potential to not only preserve β-cell function but also modulate the autoimmune response. Understanding the role of hypoxia in the pathogenesis of T1D necessitates its integration into established immunopathological frameworks. Hypoxia does not act in isolation but rather potentiates immune dysregulation by intensifying pro-inflammatory cytokine signaling, reprogramming immune cell metabolism toward pathogenic phenotypes, and heightening the susceptibility of pancreatic β-cells to stress-induced apoptosis. This mechanistic interplay highlights the need for a systems biology perspective, wherein hypoxia is recognized not merely as a downstream consequence of islet inflammation, but as an active modulator of both immune and metabolic dysfunction in T1D. By elucidating the interplay between hypoxia, β-cell stress, and immune activation, researchers can identify novel targets for intervention, creating opportunities for therapies that address the underlying mechanisms of T1D and potentially prevent disease onset or progression. This perspective aims to highlight the multifaceted role of hypoxia in T1D and its implications for disease progression and therapeutic innovation. It synthesizes findings from peer-reviewed experimental and clinical literature to integrate current knowledge on hypoxia in T1D pathogenesis. Our analysis draws upon mechanistic studies, animal models, and single-cell transcriptomics to identify converging pathways of β-cell stress and immune dysfunction.

Sources of hypoxia in T1D

To understand how hypoxia contributes to β-cell failure and immune dysregulation in T1D, it is essential to examine the mechanisms that drive oxygen deprivation within the islet microenvironment. The pancreatic islets are uniquely vulnerable to hypoxia due to their reliance on a dense and highly specialized vascular network that facilitates efficient oxygen delivery to support insulin secretion and metabolic activity. However, in the context of T1D, multiple factors converge to disrupt this delicate balance, leading to the development of a hypoxic microenvironment (Table 1).

Table 1
www.frontiersin.org

Table 1. Sources of Hypoxia in Type 1 Diabetes (T1D).

Inflammation and vascular dysfunction

Inflammatory cytokines, particularly IL-1β, TNF-α, and interferon-gamma (IFN-γ), secreted by infiltrating immune cells, not only exert cytotoxic effects on β-cells but also disrupt endothelial cell function within the microvasculature (20). These cytokines impair the structural integrity of islet capillaries by promoting endothelial apoptosis and increasing vascular permeability. The islet endothelium, crucial for maintaining optimal oxygen and nutrient exchange, becomes compromised under inflammatory stress. This leads to increased vascular permeability, reduced nitric oxide bioavailability, and endothelial cell apoptosis. As the vasculature deteriorates, the effective delivery of oxygen and nutrients to β-cells diminishes, contributing to the onset of hypoxia (21). Additionally, chronic inflammation leads to the deposition of extracellular matrix proteins and fibrosis, which physically obstructs oxygen diffusion within the islets (22). This fibrotic remodeling occurs in parallel with increased leukocyte adhesion and transmigration, facilitated by the upregulation of adhesion molecules such as ICAM-1 and VCAM-1 on endothelial surfaces, further perpetuating local inflammation and vascular dysfunction (23). Beyond the islets, systemic vascular dysfunction is frequently observed in individuals with T1D, even in the early stages of the disease. This includes impaired flow-mediated dilation, increased arterial stiffness, and microvascular rarefaction. Such changes are not only markers of early cardiovascular risk but may also reflect persistent low-grade inflammation, endothelial activation, and impaired repair mechanisms. Importantly, alterations in vascular health may precede overt hyperglycemia and serve as early indicators of disease development in at-risk individuals.

Hyperglycemia-induced stress

Sustained hyperglycemia, a hallmark of T1D, exacerbates hypoxia through several mechanisms. High glucose levels induce oxidative stress in endothelial cells, reducing their ability to respond to angiogenic signals and repair damaged vasculature (24, 25). Furthermore, hyperglycemia increases the metabolic demand of β-cells, forcing them to consume more oxygen for insulin production and secretion (26, 27). This hypermetabolic state creates an oxygen imbalance, where demand surpasses supply, further deepening the hypoxic state. Hyperglycemia also induces advanced glycation end products (AGEs), which contribute to endothelial dysfunction and impair microvascular function (28).

Immune cell infiltration

The autoimmune nature of T1D results in the infiltration of immune cells, including T cells, B cells, and macrophages, into the pancreatic islets (29, 30). These cells are metabolically active and consume large amounts of oxygen to sustain their pro-inflammatory activity. For example, activated macrophages rely on aerobic glycolysis, a process driven by high oxygen consumption, to produce inflammatory mediators (3133). Similarly, autoreactive T cells utilize oxygen to proliferate and release cytokines, further straining the already limited oxygen availability within the islets (34). This increased oxygen demand by immune cells compounds the hypoxic burden on β-cells, creating a vicious cycle of oxygen deprivation and immune activation (35, 36).

β-cell overactivation and metabolic stress

As β-cells are destroyed by autoimmune attack, the remaining β-cells are forced to compensate by increasing their insulin production. This overactivation leads to higher metabolic activity and oxygen consumption. Simultaneously, the stress imposed by hyperglycemia and inflammatory cytokines amplifies oxidative phosphorylation within β-cells, leading to excessive mitochondrial oxygen utilization and the generation of reactive oxygen species (ROS). The combination of high oxygen consumption and ROS-induced mitochondrial dysfunction further exacerbates hypoxia and β-cell vulnerability (37). Unlike other tissues, β cells have relatively low antioxidant capacity, making them particularly susceptible to oxidative stress (38). This environment promotes endoplasmic reticulum stress, unfolded protein response (UPR) activation, and mitochondrial dysfunction, processes that can trigger pro-apoptotic signaling cascades and compromise β-cell viability.

Islet capillary dysfunction

In T1D, the capillary network within the islets becomes increasingly dysfunctional due to pericyte loss, basement membrane thickening, and impaired angiogenesis. This dysfunction reduces the ability of the capillaries to adequately supply oxygen and nutrients to β-cells. Studies have shown that islet endothelial cells are particularly sensitive to inflammatory and hyperglycemic stress, which accelerates their deterioration and diminishes capillary density over time (3941).

Role of hypoxia-inducible factors

Hypoxia-inducible factors (HIFs) are pivotal transcriptional regulators orchestrating cellular responses to oxygen deprivation (42) (Supplementary Table 1). Specifically, under normoxic conditions, HIF-1α and HIF-2α undergo prolyl hydroxylation by oxygen-dependent prolyl hydroxylase domain (PHD) enzymes, targeting them for ubiquitination and proteasomal degradation via the von Hippel–Lindau (VHL) E3 ligase (43, 44). In hypoxic conditions, this hydroxylation is inhibited, allowing HIF-α subunits to stabilize, accumulate in the cytoplasm, translocate to the nucleus, dimerize with HIF-1β, and drive the transcription of genes (45). In pancreatic islets, transient HIF activation during early hypoxia confers adaptive advantages. HIF-1α promotes the expression of vascular endothelial growth factor (VEGF), which facilitates angiogenesis to improve oxygen delivery to hypoxic islets (46). Concurrently, HIF-1α enhances the expression of glycolytic enzymes, enabling β-cells and surrounding immune cells to shift from oxidative phosphorylation to glycolysis, thereby supporting cellular energy production and viability despite reduced oxygen availability (47). This metabolic adaptation provides temporary protection to islets under acute hypoxic stress.

However, prolonged HIF activation in chronic hypoxia has maladaptive consequences, severely compromising β-cell function and immune homeostasis (4). Sustained HIF stabilization exacerbates ROS accumulation and oxidative stress, impairing mitochondrial integrity and β-cell insulin secretion. Chronic HIF activity decreases insulin signaling pathways and disrupts the expression of critical insulin-related transcription factors, including PDX1 and MAFA, undermining β-cell identity and functionality (5, 48). Moreover, in immune cells, HIF-1α drives macrophage polarization toward a pro-inflammatory M1 phenotype and enhances the differentiation of Th17 cells, both of which contribute to autoimmune β-cell destruction (49). Simultaneously, HIF-1α diminishes the functionality of regulatory T cells (Tregs) by promoting the proteasomal degradation of FOXP3, a master transcription factor critical for Treg identity and suppressive capacity (50, 51). Additionally, HIF-1α promotes glycolytic metabolism, which antagonizes the oxidative phosphorylation-dependent metabolic program necessary for Treg stability and function (50, 52). This intricate interplay between HIF-mediated immune dysregulation and β-cell dysfunction accelerates the progression of islet pathology (53, 54).

Chronic HIF activation also disrupts angiogenesis within islets, leading to a persistent imbalance. Despite the upregulated VEGF expression, the resultant blood vessels are structurally abnormal, leaky, and non-functional, failing to restore adequate oxygen delivery. This angiogenic dysregulation sustains hypoxia and perpetuates islet dysfunction. Additionally, HIF signaling amplifies inflammatory pathways by promoting the secretion of pro-inflammatory cytokines such as IL-6 and TNF-α from β-cells and infiltrating immune cells. These cytokines exacerbate immune cell recruitment and activation, intensifying islet inflammation. The combined effects of HIF-driven inflammation, oxidative stress, and angiogenic imbalance create a hostile microenvironment that accelerates β-cell apoptosis and disrupts islet homeostasis.

β-Cell stress pathways: mitochondrial, ER, and autophagic dysfunction

Concurrently with its effects on immune architecture, hypoxia imposes direct cellular stress on pancreatic β-cells. In the subsequent section, we delineate how hypoxia-induced perturbations in mitochondrial function, protein folding, and intracellular degradation pathways contribute to β-cell dysfunction.

Mitochondrial dysfunction

β-cells exhibit increased sensitivity to mitochondrial dysfunction, particularly when subjected to the simultaneous stressors of hypoxia and hyperglycemia (55, 56). Chronic hypoxia compromises the efficiency of oxidative phosphorylation by limiting oxygen availability, which is a critical substrate for the electron transport chain (57). This results in reduced ATP production and altering cellular energy homeostasis (58). Concurrently, hyperglycemia exacerbates the metabolic burden on mitochondria by driving an excessive flux of glucose-derived substrates through the tricarboxylic acid (TCA) cycle (59). This overloading of mitochondrial metabolism leads to an increased generation of ROS, creating a state of oxidative stress (60). The accumulation of ROS initiates widespread oxidative damage to lipids, proteins, and mitochondrial DNA, impairing the integrity and function of the mitochondria. This oxidative stress also disrupts the delicate balance of intracellular signaling pathways critical for β-cells survival, such as those regulating insulin secretion (56, 61). Additionally, the sustained oxidative environment activates apoptotic pathways, including mitochondrial outer membrane permeabilization and cytochrome c release, which accelerate β-cell death (62).

While mitochondrial dysfunction is well documented in type 2 diabetes, similar mechanisms may contribute to β-cell loss in T1D. However, the direct connection with T1D remains unclear, highlighting the need for future investigations to elucidate the role of mitochondrial impairment in T1D pathogenesis. Emerging evidence suggests that in T1D, immune mediated inflammation driven by cytokines such as IL-1β, TNF-α, and IFN-γ, along with local hypoxia, impairs mitochondrial respiration and elevates ROS production in pancreatic β-cells (63). This oxidative stress disrupts ATP synthesis, damages mitochondrial structures, and activates apoptotic pathways (6466). Hypoxia further exacerbates mitochondrial dysfunction by stabilizing HIF-1α, which promotes a shift to glycolysis and impairs insulin secretion (4, 67, 68). Additionally, chronic inflammation impairs mitophagy, leading to the accumulation of dysfunctional mitochondria (6971). These synergistic stressors accelerate β-cell dysfunction and death, highlighting potential therapeutic targets in oxidative stress regulation and immune modulation.

Endoplasmic reticulum stress

In addition to impairing mitochondrial bioenergetics, hypoxia and hyperglycemia synergistically activate endoplasmic reticulum stress pathways, thereby disrupting insulin biosynthesis and triggering apoptotic signaling cascades. Endoplasmic reticulum (ER) stress occurs when the protein-folding capacity of the ER is overwhelmed, leading to an accumulation of misfolded or unfolded proteins within its lumen. This disruption activates a highly conserved cellular mechanism known as the unfolded protein response (UPR). The UPR is designed to mitigate ER stress by halting general protein translation, increasing the production of molecular chaperones to aid in proper protein folding, and enhancing ER-associated degradation (ERAD) to clear misfolded proteins. However, when ER stress is prolonged or severe, the adaptive mechanisms of the UPR become maladaptive, triggering pathways that promote cell dysfunction and death, primarily through apoptosis (7274).

In the context of hyperglycemia, elevated glucose levels intensify ER stress. The β-cells are highly sensitive to ER stress due to their essential role in insulin biosynthesis, a process that places a significant burden on the ER. Hyperglycemia increases the demand for insulin production, leading to excessive protein synthesis. This heightened demand exacerbates the likelihood of protein misfolding and impairs the ER’s ability to manage the cellular workload. Consequently, persistent hyperglycemia and ER stress contribute to β-cell dysfunction and eventual apoptosis, undermining insulin production and promoting hyperglycemia in a vicious cycle. Moreover, in hypoxic conditions, often observed in inflamed or poorly vascularized tissues, ER stress is further intensified due to impaired oxygen availability, which disrupts protein folding processes that rely on oxidative environments. Proper formation of disulfide bonds in the ER requires oxygen-dependent enzymes such as ERO1 and protein disulfide isomerase (PDI) (7578). Hypoxia inhibits these redox reactions, leading to the accumulation of misfolded proinsulin, sustained activation of the unfolded protein response (UPR), and subsequent apoptotic signaling (79, 80). Combined with the effects of hyperglycemia, this dual stressor amplifies UPR activation, leading to chronic inflammation through the release of pro-inflammatory cytokines. This inflammatory state exacerbates systemic metabolic disturbances and cellular damage, contributing to the pathogenesis of T1D (8184).

Impaired autophagy

Hypoxia disrupts cellular homeostasis by impairing various metabolic and repair processes, including autophagy (85). Autophagy is a highly conserved cellular mechanism responsible for degrading and recycling damaged organelles, misfolded proteins, and other cytoplasmic debris (86, 87). In pancreatic β-cells, autophagy plays a critical role in maintaining cellular health, especially given their high metabolic activity and demand for protein synthesis to support insulin production. Under normal conditions, autophagy supports β-cell survival by mitigating oxidative stress, removing damaged mitochondria, and preserving mitochondrial quality, which is essential for ATP production and glucose-stimulated insulin secretion (43, 88, 89). During hypoxia, the ability of β-cells to sustain efficient autophagic activity is compromised. Hypoxia directly inhibits autophagic flux by interfering with key steps in the autophagy pathway. Specifically, the low oxygen levels can disrupt the activity of hypoxia-sensitive proteins and signaling pathways, such as AMP-activated protein kinase (AMPK) and mechanistic target of rapamycin (mTOR) (90). AMPK typically promotes autophagy by activating the ULK1 complex, while mTOR acts as a negative regulator of autophagy (91, 92). In hypoxic conditions, dysregulation of these pathways reduces the induction and progression of autophagy, leading to the accumulation of damaged organelles, such as dysfunctional mitochondria. This exacerbates oxidative stress, as impaired mitochondria produce ROS, creating a self-reinforcing cycle of cellular damage (9395).

Hyperglycemia compounds the autophagic impairment induced by hypoxia. Chronic high glucose levels generate excessive amounts of toxic metabolites, including AGEs and ROS (96). These metabolites further stress cellular systems and increase the demand for autophagic clearance (9799). However, when autophagic capacity is already compromised due to hypoxia, β-cells are unable to efficiently clear these harmful byproducts. This leads to cellular dysfunction and damage, contributing to β-cell apoptosis and exacerbating the loss of insulin production. Furthermore, hyperglycemia-driven activation of the hexosamine biosynthetic pathway and protein kinase C (PKC) signaling disrupts autophagy by altering post-translational modifications of key autophagy-related proteins (100102). These modifications reduce the efficiency of autophagosome formation and lysosomal degradation. Additionally, excess glucose exposure causes ER stress, which interacts with and inhibits autophagic pathways, further compounding the inability of β-cells to manage intracellular damage (103).

The combined effects of hypoxia and hyperglycemia create a cellular environment where repair and quality control mechanisms are overwhelmed. The progressive accumulation of damaged organelles, misfolded proteins, and oxidative stress amplifies β-cell dysfunction, contributing to the pathogenesis and progression of diabetes. These findings highlight the critical importance of maintaining autophagic activity for β-cell health and suggest potential therapeutic avenues aimed at restoring autophagic flux in hypoxic and hyperglycemic conditions. Given the multifaceted role of hypoxia in amplifying both immune dysregulation and β-cell intrinsic stress, therapeutic strategies that target hypoxia-adaptive pathways represent a novel and potentially transformative approach for disease modification in T1D.

Future directions

Recent studies have suggested the potential role of modulating hypoxia in T1D pathogenesis (4, 104107). A study demonstrated that human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) enhance the survival and function of pancreatic islets under hypoxic conditions via the HIF-1α/PFKFB3 pathway, suggesting a protective mechanism against hypoxia-induced β-cell dysfunction (108). Additionally, research by Kelly et al. revealed that intrauterine growth restriction leads to adaptive and immune responses in fetal sheep islets, including the activation of immune pathways and stress responses, which may predispose individuals to T1D (109). These findings highlight the necessity for advanced experimental models that accurately mimic the hypoxic and immunological milieu of T1D, thereby facilitating the development of targeted therapeutic strategies aimed at preserving β-cell function and modulating autoimmunity. Building on these findings, it becomes increasingly important to elucidate the cellular and molecular mechanisms that mediate the interplay between hypoxia, immune activation, and β-cell stress, thereby guiding the development of precise, mechanism-based interventions.

Mechanistic studies

Advancing our understanding of the molecular mechanisms linking hyperglycemia, hypoxia, and immune dysregulation in T1D is critical for developing targeted therapeutic strategies. Despite significant progress in characterizing the individual contributions of these factors, the precise cellular and molecular pathways through which they interact remain poorly defined. Future research should aim to dissect how hyperglycemia-induced metabolic stress and hypoxia collectively drive immune activation and β-cell dysfunction.

Organoid platforms that recapitulate the 3D architecture and cellular complexity of human islets are emerging as powerful tools for modeling these interactions under controlled conditions (110). By incorporating endothelial and immune components into islet organoids, researchers can more accurately simulate the hypoxic and inflammatory microenvironment characteristic of early T1D, allowing for longitudinal and mechanistic studies of disease initiation and progression.

Single-cell RNA sequencing (scRNA-seq) can play a pivotal role in elucidating the heterogeneity of cellular responses within the hypoxic islet microenvironment (111, 112). This approach enables the high-resolution mapping of transcriptomic changes in β-cells, endothelial cells, and infiltrating immune cells, revealing dynamic interactions that exacerbate hypoxia and inflammation. Spatial transcriptomics can complement scRNA-seq by providing spatial context to molecular changes, capturing the precise localization of hypoxia-related genes in relation to islet architecture and immune cell infiltration (35, 113, 114). Utilizing these techniques will provide novel insights regarding the role of hypoxia in the pathophysiology of T1D.

Therapeutic delivery platforms

While advanced in vitro and omics-based platforms are critical for decoding the molecular underpinnings of hypoxia in T1D, translating these insights into viable therapies requires parallel innovation in delivery strategies capable of targeting the islet microenvironment with precision. The development of innovative therapeutic delivery platforms offers significant promise in targeting hypoxia and immune dysregulation in T1D. Traditional systemic treatments are often limited by off-target effects and insufficient delivery to the pancreatic islet microenvironment. Encapsulation technologies for β-cell transplants, for instance, could provide localized protection from immune attack while supporting oxygenation and nutrient delivery. These encapsulated transplants can be engineered to enhance survival and function in the hypoxic and inflamed microenvironment characteristic of T1D (115, 116).

Localized drug delivery systems also hold potential for improving therapeutic precision. Nanoparticle-based carriers, for example, can be designed to deliver antioxidants, hypoxia modulators, or anti-inflammatory agents directly to the islet microenvironment, reducing systemic side effects while maximizing efficacy. Hydrogel-based systems that release therapeutic agents in response to specific triggers, such as low oxygen levels or high reactive oxygen species, could offer another level of control. Further exploration of these delivery systems is needed to refine their effectiveness and scalability, as well as to evaluate their long-term safety and durability in clinical trials (117, 118).

Data-driven approaches

As delivery technologies evolve to address site-specific therapeutic challenges, data-driven approaches offer a complementary framework to optimize intervention timing, predict therapeutic responses, and uncover non-obvious molecular targets within the hypoxia–autoimmunity axis. The integration of data-driven methodologies, particularly machine learning and computational modeling, has the potential to revolutionize the understanding and treatment of hypoxia-related mechanisms in T1D (119, 120). By leveraging machine learning techniques, researchers can extract meaningful insights from complex, large-scale datasets generated from multi-omics analyses, such as transcriptomics, proteomics, and metabolomics, as well as from clinical trials and patient registries. These algorithms can uncover intricate patterns, correlations, and predictive markers that might otherwise remain undetected (121123). For instance, specific molecular drivers or pathways linking hypoxia to immune dysregulation, oxidative stress, and β-cell dysfunction can be identified, which are critical in the pathogenesis of T1D (124).

In parallel, computational modeling provides a powerful tool to simulate the dynamic and multifaceted islet microenvironment under various physiological and pathological conditions, including hypoxia, hyperglycemia, and inflammatory stress (125). These models enable researchers to explore how β-cells interact with immune cells and their surrounding microenvironment, allowing for the in silico testing of hypotheses about disease progression and potential interventions. By incorporating data from experimental studies and clinical observations, computational simulations can predict how changes in oxygen availability, nutrient levels, and inflammatory mediators influence β-cell viability and function (126).

Developing therapeutic strategies

Building on the foundation of experimental and computational advances, therapeutic efforts now focus on mitigating hypoxia-driven β-cell dysfunction through molecular and tissue-level interventions. Hypoxia in T1D plays a pivotal role in the dysfunction and death of pancreatic β-cells, amplifying the progression of the disease. Therefore, targeting hypoxia will pave the way to develop novel therapeutic modalities for T1D (Supplementary Table 2). Restoring oxygen levels in the pancreatic microenvironment could alleviate hypoxic stress and improve β-cell survival (127129). Oxygenation-based approaches, such as hyperbaric oxygen therapy or oxygen-releasing nanoparticles, have been proposed as potential strategies to counteract hypoxia. By addressing the oxygen supply to β-cells, these strategies may mitigate cellular damage and improve metabolic outcomes. Modulation of HIFs is another promising approach for addressing hypoxia-related β-cell dysfunction. Dysregulated HIF-1α signaling during hypoxia contributes to inflammation, impaired autophagy, and β-cell apoptosis (130). Stabilizing HIF-2α or targeting maladaptive HIF-1α activity could enhance angiogenesis, improve islet vascularization, and support β-cell survival. These strategies highlight the therapeutic potential of manipulating hypoxia signaling pathways to mitigate cellular stress and maintain β-cell health (131, 132).

Islet vascularization is critically impaired in T1D, exacerbating hypoxic conditions and β-cell loss. Promoting angiogenesis through VEGF-based therapies or cell-based approaches, such as mesenchymal stem cells (MSCs), could enhance islet perfusion and oxygen delivery (133, 134). These interventions may also address inflammation-induced vascular damage, improving overall islet resilience and functionality in the context of T1D (135).

Hypoxia-induced autophagy impairment is another significant contributor to β-cell dysfunction in T1D. Autophagy plays a crucial role in removing damaged organelles and maintaining cellular health. Hypoxia disrupts this process, leading to the accumulation of damaged mitochondria, increased oxidative stress, and β-cell death (136, 137). Strategies to restore autophagic flux, such as mTOR inhibition or AMPK activation, could counteract hypoxia-induced cellular damage by reactivating autophagy and enhancing cellular stress resilience (138140). However, systemic administration of these modulators raises concerns due to their broad physiological effects, including metabolic dysregulation, immune suppression, or unintended impacts on proliferative signaling in non-target tissues (141, 142). To circumvent these risks, localized delivery approaches are being developed to restrict therapeutic activity to the pancreatic microenvironment. These include engineered nanoparticles that exploit local tissue markers or microenvironmental cues (such as pH or hypoxia) for targeted release, oxygen-sensitive or hypoxia-responsive hydrogels that release payloads in response to local oxygen tension and encapsulated islet or beta-cell devices incorporating drug reservoirs or responsive release systems (143148). Such strategies aim to minimize off-target exposure while ensuring sufficient local concentration to modulate autophagy-regulatory pathways within the islet graft or endogenous pancreas. In islet transplantation, hypoxia remains a major barrier to long-term graft survival and function. Transplanted islets often experience hypoxic conditions due to insufficient vascularization, leading to graft failure. Approaches to mitigate hypoxia, such as preconditioning islets with hypoxia-mimicking agents or incorporating oxygen-releasing materials into encapsulation technologies, have the potential to improve transplant outcomes. Additionally, strategies that reduce hypoxia-induced inflammation and oxidative stress could further enhance graft viability (149).

Conclusions

The intricate interplay between oxygen availability, immune dynamics, and β-cell resilience in T1D highlights a paradigm shift in our understanding of disease progression. Hypoxia, traditionally regarded as a secondary consequence of inflammatory and metabolic disturbances, emerges as a fundamental driver of β-cell vulnerability. It can act as a molecular amplifier of immune-mediated cytotoxicity, oxidative stress, and metabolic dysfunction, establishing a pathophysiological axis that extends beyond conventional immune-centric frameworks.

Deciphering the hypoxic landscape of pancreatic islets necessitates an integrated, systems-biology approach. The chronic stabilization of HIFs within β-cells and immune infiltrates orchestrates a cascade of transcriptional and metabolic perturbations, precipitating a shift toward maladaptive cellular states (150). This aberrant signaling interface disrupts immune homeostasis, subverts β-cell identity by downregulating essential transcriptional regulators including PDX1, MAFA, and NKX6.1, thereby impairing insulin gene expression and promoting a dedifferentiated or progenitor-like phenotype (151153). This loss of identity compromises glucose-stimulated insulin secretion and β-cell survival. Consequently, interventions targeting hypoxia-mediated dysregulation represent an unexplored frontier in modifying T1D trajectory.

Advancements in spatial transcriptomics, single-cell metabolic profiling, and in silico modeling hold the potential to deconvolute the heterogeneity of hypoxic responses within islet microenvironments. Precision-engineered therapeutic strategies, including hypoxia-responsive biomaterials, oxygenation-modulating nanoformulations, and selective HIF modulation, offer novel avenues for mitigating hypoxia-induced β-cell attrition. Furthermore, the integration of hypoxia-centric interventions with immune-modulatory frameworks may redefine therapeutic paradigms, bridging the gap between metabolic preservation and immunological tolerance.

Although targeting hypoxic signaling in T1D represents a compelling therapeutic avenue, translating these concepts into clinical practice presents considerable complexity. One of the foremost challenges is the ability to selectively modulate oxygen-sensitive pathways within the pancreatic islet microenvironment without perturbing systemic oxygen homeostasis. Pharmacologic agents that influence the activity of hypoxia-inducible factors or related metabolic sensors often exert pleiotropic effects in tissues such as the myocardium, central nervous system, and renal epithelium, where oxygen signaling plays fundamental physiological roles. This raises significant safety concerns, including the potential for disrupting angiogenesis, altering mitochondrial bioenergetics, or impairing adaptive cellular responses under normal oxygen conditions. Additionally, the anatomical and physiological inaccessibility of pancreatic islets complicates the targeted delivery of such interventions. Therapeutic compounds must traverse dense stromal tissue, evade rapid systemic clearance, and retain functional activity within the islet niche, which is often characterized by inflammatory and fibrotic remodeling. Overcoming these delivery and stability barriers necessitates the development of next-generation carrier systems that are both biocompatible and capable of site-specific drug release. To advance these strategies toward clinical utility, it is essential to conduct preclinical assessments in experimental models that accurately recapitulate human islet immunobiology, oxygen gradients, and disease kinetics. Moreover, longitudinal studies evaluating toxicity, pharmacokinetics, immune interactions, and off-target responses are critical for establishing a robust safety profile. Collectively, these efforts are foundational to ensuring that hypoxia-focused interventions are both biologically viable and clinically translatable in the context of T1D.

A comprehensive elucidation of the molecular interfaces between oxygen deprivation, β-cell stress responses, and immune-mediated cytotoxicity is essential for the rational development of targeted therapeutic interventions in T1D. This evolving paradigm positions hypoxia not solely as a passive biomarker of disease progression, but as an actionable pathogenic determinant amenable to intervention. By harnessing emerging insights into hypoxia-induced signaling cascades and their impact on β-cell function and immune dynamics, future therapeutic strategies may be optimized to preserve β-cell identity, suppress autoreactive immune effector mechanisms, and fundamentally alter the disease trajectory toward sustained glycemic control and immune tolerance.

Data availability statement

The original contributions presented in the study are included in the article/Supplementary Material. Further inquiries can be directed to the corresponding authors.

Author contributions

RM: Formal analysis, Resources, Data curation, Project administration, Validation, Methodology, Writing – review & editing, Supervision, Investigation, Writing – original draft, Conceptualization. JL: Writing – original draft, Investigation, Formal analysis, Project administration, Supervision, Validation, Methodology, Conceptualization, Data curation, Writing – review & editing. PC: Resources, Validation, Writing – original draft, Formal analysis, Supervision, Writing – review & editing, Data curation, Methodology, Investigation, Conceptualization. KH: Conceptualization, Investigation, Data curation, Software, Resources, Formal analysis, Writing – review & editing, Project administration, Supervision, Methodology, Writing – original draft, Validation.

Funding

The author(s) declare that no financial support was received for the research and/or publication of this article.

Acknowledgments

We are grateful to Dr. Valerie Gramling for critical reading of the manuscript.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Generative AI statement

The author(s) declare that no Generative AI was used in the creation of this manuscript.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Supplementary material

The Supplementary Material for this article can be found online at: https://www.frontiersin.org/articles/10.3389/fimmu.2025.1599321/full#supplementary-material

References

1. DiMeglio LA, Evans-Molina C, and Oram RA. Type 1 diabetes. Lancet. (2018) 391:2449–62. doi: 10.1016/S0140-6736(18)31320-5

PubMed Abstract | Crossref Full Text | Google Scholar

2. Mittal R, Camick N, Lemos JRN, and Hirani K. Gene-environment interaction in the pathophysiology of type 1 diabetes. Front Endocrinol (Lausanne). (2024) 15:1335435. doi: 10.3389/fendo.2024.1335435

PubMed Abstract | Crossref Full Text | Google Scholar

3. Abel ED, Gloyn AL, Evans-Molina C, Joseph JJ, Misra S, Pajvani UB, et al. Diabetes mellitus-Progress and opportunities in the evolving epidemic. Cell. (2024) 187:3789–820. doi: 10.1016/j.cell.2024.06.029

PubMed Abstract | Crossref Full Text | Google Scholar

4. Fagundes RR, Zaldumbide A, and Taylor CT. Role of hypoxia-inducible factor 1 in type 1 diabetes. Trends Pharmacol Sci. (2024) 45:798–810. doi: 10.1016/j.tips.2024.07.001

PubMed Abstract | Crossref Full Text | Google Scholar

5. Tsuyama T, Sato Y, Yoshizawa T, Matsuoka T, and Yamagata K. Hypoxia causes pancreatic β-cell dysfunction and impairs insulin secretion by activating the transcriptional repressor BHLHE40. EMBO Rep. (2023) 24:e56227. doi: 10.15252/embr.202256227

PubMed Abstract | Crossref Full Text | Google Scholar

6. Taylor CT and Scholz CC. The effect of HIF on metabolism and immunity. Nat Rev Nephrol. (2022) 18:573–87. doi: 10.1038/s41581-022-00587-8

PubMed Abstract | Crossref Full Text | Google Scholar

7. Muratore M, Santos C, and Rorsman P. The vascular architecture of the pancreatic islets: A homage to August Krogh. Comp Biochem Physiol A Mol Integr Physiol. (2021) 252:110846. doi: 10.1016/j.cbpa.2020.110846

PubMed Abstract | Crossref Full Text | Google Scholar

8. Tamayo A, Gonçalves LM, Rodriguez-Diaz R, Pereira E, Canales M, Caicedo A, et al. Pericyte control of blood flow in intraocular islet grafts impacts glucose homeostasis in mice. Diabetes. (2022) 71:1679–93. doi: 10.2337/db21-1104

PubMed Abstract | Crossref Full Text | Google Scholar

9. Mateus Gonçalves L, Fahd Qadir MM, Boulina M, Makhmutova M, Pereira E, and Almaça J. Pericyte dysfunction and impaired vasomotion are hallmarks of islets during the pathogenesis of type 1 diabetes. Cell Rep. (2023) 42:112913. doi: 10.1016/j.celrep.2023.112913

PubMed Abstract | Crossref Full Text | Google Scholar

10. Dri E, Lampas E, Lazaros G, Lazarou E, Theofilis P, Tsioufis C, et al. Inflammatory mediators of endothelial dysfunction. Life (Basel). (2023) 13. doi: 10.3390/life13061420

PubMed Abstract | Crossref Full Text | Google Scholar

11. Scherm MG, Wyatt RC, Serr I, Anz D, Richardson SJ, and 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. doi: 10.1016/j.molmet.2022.101565

PubMed Abstract | Crossref Full Text | Google Scholar

12. Tang YY, Wang DC, Wang YQ, Huang AF, and Xu WD. Emerging role of hypoxia-inducible factor-1α in inflammatory autoimmune diseases: A comprehensive review. Front Immunol. (2022) 13:1073971.

PubMed Abstract | Google Scholar

13. Gao J, Ding XS, Zhang YM, Dai DZ, Liu M, Zhang C, et al. Hypoxia/oxidative stress alters the pharmacokinetics of CPU86017-RS through mitochondrial dysfunction and NADPH oxidase activation. Acta Pharmacol Sin. (2013) 34:1575–84. doi: 10.1038/aps.2013.94

PubMed Abstract | Crossref Full Text | Google Scholar

14. Zhao Y, Gai C, Yu S, Song Y, Gu B, Luo Q, et al. Liposomes-loaded miR-9-5p alleviated hypoxia-ischemia-induced mitochondrial oxidative stress by targeting ZBTB20 to inhibiting nrf2/keap1 interaction in neonatal mice. Antioxid Redox Signal. (2025). doi: 10.1089/ars.2024.0640

PubMed Abstract | Crossref Full Text | Google Scholar

15. Lotsios NS, Keskinidou C, Karagiannis SP, Papavassiliou KA, Papavassiliou AG, Kotanidou A, et al. Expression and regulation of hypoxia-inducible factor signaling in acute lung inflammation. Cells. (2024) 14.

PubMed Abstract | Google Scholar

16. Morton L, Garza AP, Debska-Vielhaber G, Villafuerte LE, Henneicke S, Arndt P, et al. Pericytes and extracellular vesicle interactions in neurovascular adaptation to chronic arterial hypertension. J Am Heart Assoc. (2025) 14:e038457. doi: 10.1161/JAHA.124.038457

PubMed Abstract | Crossref Full Text | Google Scholar

17. Albendea-Gomez T, Mendoza-Tamajon S, Castro-Mecinas R, Escobar B, Rocha SF, Urra-Balduz S, et al. Vascular HIF2 signaling prevents cardiomegaly, alveolar congestion, and capillary remodeling during chronic hypoxia. Arterioscler Thromb Vasc Biol. (2025). doi: 10.1161/ATVBAHA.124.321780

PubMed Abstract | Crossref Full Text | Google Scholar

18. Zhang S, Gang X, Yang S, Cui M, Sun L, Li Z, et al. The alterations in and the role of the th17/treg balance in metabolic diseases. Front Immunol. (2021) 12:678355. doi: 10.3389/fimmu.2021.678355

PubMed Abstract | Crossref Full Text | Google Scholar

19. Zeng S, Wang Y, Ai L, Huang L, Liu Z, He C, et al. Chronic intermittent hypoxia-induced oxidative stress activates TRB3 and phosphorylated JNK to mediate insulin resistance and cell apoptosis in the pancreas. Clin Exp Pharmacol Physiol. (2024) 51:e13843. doi: 10.1111/1440-1681.13843

PubMed Abstract | Crossref Full Text | Google Scholar

20. Liu M, Chen R, Zheng Z, Xu S, Hou C, Ding Y, et al. Mechanisms of inflammatory microenvironment formation in cardiometabolic diseases: molecular and cellular perspectives. Front Cardiovasc Med. (2024) 11:1529903. doi: 10.3389/fcvm.2024.1529903

PubMed Abstract | Crossref Full Text | Google Scholar

21. Castillo JJ, Aplin AC, Hackney DJ, Hogan MF, Esser N, Templin AT, et al. Islet amyloid polypeptide aggregation exerts cytotoxic and proinflammatory effects on the islet vasculature in mice. Diabetologia. (2022) 65:1687–700. doi: 10.1007/s00125-022-05756-9

PubMed Abstract | Crossref Full Text | Google Scholar

22. Richards OC, Raines SM, and Attie AD. The role of blood vessels, endothelial cells, and vascular pericytes in insulin secretion and peripheral insulin action. Endocr Rev. (2010) 31:343–63. doi: 10.1210/er.2009-0035

PubMed Abstract | Crossref Full Text | Google Scholar

23. Song Y, Manson JE, Tinker L, Rifai N, Cook NR, Hu FB, et al. Circulating levels of endothelial adhesion molecules and risk of diabetes in an ethnically diverse cohort of women. Diabetes. (2007) 56:1898–904. doi: 10.2337/db07-0250

PubMed Abstract | Crossref Full Text | Google Scholar

24. An Y, Xu BT, Wan SR, Ma XM, Long Y, Xu Y, et al. The role of oxidative stress in diabetes mellitus-induced vascular endothelial dysfunction. Cardiovasc Diabetol. (2023) 22:237. doi: 10.1186/s12933-023-01965-7

PubMed Abstract | Crossref Full Text | Google Scholar

25. Binjawhar DN, Alhazmi AT, Bin Jawhar WN, MohammedSaeed W, and Safi SZ. Hyperglycemia-induced oxidative stress and epigenetic regulation of ET-1 gene in endothelial cells. Front Genet. (2023) 14:1167773. doi: 10.3389/fgene.2023.1167773

PubMed Abstract | Crossref Full Text | Google Scholar

26. Hauwanga WN, Abdalhamed TY, Ezike LA, Chukwulebe IS, Ko Oo A, Wilfred A, et al. The pathophysiology and vascular complications of diabetes in chronic kidney disease: A comprehensive review. Cureus. (2024) 16:e76498. doi: 10.7759/cureus.76498

PubMed Abstract | Crossref Full Text | Google Scholar

27. Kubota T, Kubota N, Kumagai H, Yamaguchi S, Kozono H, Takahashi T, et al. Impaired insulin signaling in endothelial cells reduces insulin-induced glucose uptake by skeletal muscle. Cell Metab. (2011) 13:294–307. doi: 10.1016/j.cmet.2011.01.018

PubMed Abstract | Crossref Full Text | Google Scholar

28. Du C, Whiddett RO, Buckle I, Chen C, Forbes JM, and Fotheringham AK. Advanced glycation end products and inflammation in type 1 diabetes development. Cells. (2022) 11. doi: 10.3390/cells11213503

PubMed Abstract | Crossref Full Text | Google Scholar

29. Toren E, Burnette KS, Banerjee RR, Hunter CS, and Tse HM. Partners in crime: beta-cells and autoimmune responses complicit in type 1 diabetes pathogenesis. Front Immunol. (2021) 12:756548. doi: 10.3389/fimmu.2021.756548

PubMed Abstract | Crossref Full Text | Google Scholar

30. Burrack AL, Martinov T, and Fife BT. T cell-mediated beta cell destruction: autoimmunity and alloimmunity in the context of type 1 diabetes. Front Endocrinol (Lausanne). (2017) 8:343. doi: 10.3389/fendo.2017.00343

PubMed Abstract | Crossref Full Text | Google Scholar

31. Viola A, Munari F, Sánchez-Rodríguez R, Scolaro T, and Castegna A. The metabolic signature of macrophage responses. Front Immunol. (2019) 10:1462. doi: 10.3389/fimmu.2019.01462

PubMed Abstract | Crossref Full Text | Google Scholar

32. Pavlou S, Wang L, Xu H, and Chen M. Higher phagocytic activity of thioglycollate-elicited peritoneal macrophages is related to metabolic status of the cells. J Inflammation (Lond). (2017) 14:4. doi: 10.1186/s12950-017-0151-x

PubMed Abstract | Crossref Full Text | Google Scholar

33. Freemerman AJ, Johnson AR, Sacks GN, Milner JJ, Kirk EL, Troester MA, et al. Metabolic reprogramming of macrophages: glucose transporter 1 (GLUT1)-mediated glucose metabolism drives a proinflammatory phenotype. J Biol Chem. (2014) 289:7884–96. doi: 10.1074/jbc.M113.522037

PubMed Abstract | Crossref Full Text | Google Scholar

34. Peng HY, Lucavs J, Ballard D, Das JK, Kumar A, Wang L, et al. Metabolic reprogramming and reactive oxygen species in T cell immunity. Front Immunol. (2021) 12:652687. doi: 10.3389/fimmu.2021.652687

PubMed Abstract | Crossref Full Text | Google Scholar

35. Chen P, Yao F, Lu Y, Peng Y, Zhu S, Deng J, et al. Single-cell landscape of mouse islet allograft and syngeneic graft. Front Immunol. (2022) 13:853349. doi: 10.3389/fimmu.2022.853349

PubMed Abstract | Crossref Full Text | Google Scholar

36. Denroche HC, Miard S, Sallé-Lefort S, Picard F, and Verchere CB. T cells accumulate in non-diabetic islets during ageing. Immun Ageing. (2021) 18:8. doi: 10.1186/s12979-021-00221-4

PubMed Abstract | Crossref Full Text | Google Scholar

37. Cuenco J and Dalmas E. Islet inflammation and β Cell dysfunction in type 2 diabetes. Handb Exp Pharmacol. (2022) 274:227–51.

Google Scholar

38. Ahmed Alfar E, Kirova D, Konantz J, Birke S, Mansfeld J, and Ninov N. Distinct levels of reactive oxygen species coordinate metabolic activity with beta-cell mass plasticity. Sci Rep. (2017) 7:3994. doi: 10.1038/s41598-017-03873-9

PubMed Abstract | Crossref Full Text | Google Scholar

39. Kapitulnik J, Benaim C, and Sasson S. Endothelial cells derived from the blood-brain barrier and islets of langerhans differ in their response to the effects of bilirubin on oxidative stress under hyperglycemic conditions. Front Pharmacol. (2012) 3:131. doi: 10.3389/fphar.2012.00131

PubMed Abstract | Crossref Full Text | Google Scholar

40. Martens GA and Van de Casteele M. Glycemic control of apoptosis in the pancreatic beta cell: danger of extremes? Antioxid Redox Signal. (2007) 9:309–17.

PubMed Abstract | Google Scholar

41. Robertson RP and Harmon JS. Diabetes, glucose toxicity, and oxidative stress: A case of double jeopardy for the pancreatic islet beta cell. Free Radic Biol Med. (2006) 41:177–84. doi: 10.1016/j.freeradbiomed.2005.04.030

PubMed Abstract | Crossref Full Text | Google Scholar

42. Takikawa A, Mahmood A, Nawaz A, Kado T, Okabe K, Yamamoto S, et al. HIF-1α in myeloid cells promotes adipose tissue remodeling toward insulin resistance. Diabetes. (2016) 65:3649–59. doi: 10.2337/db16-0012

PubMed Abstract | Crossref Full Text | Google Scholar

43. Abd El-Emam MM, Behairy A, Mostafa M, Khamis T, Osman NMS, Alsemeh AE, et al. Chrysin-loaded PEGylated liposomes protect against alloxan-induced diabetic neuropathy in rats: the interplay between endoplasmic reticulum stress and autophagy. Biol Res. (2024) 57:45. doi: 10.1186/s40659-024-00521-1

PubMed Abstract | Crossref Full Text | Google Scholar

44. Kaelin WG Jr. The von Hippel-Lindau tumor suppressor protein: O2 sensing and cancer. Nat Rev Cancer. (2008) 8:865–73.

Google Scholar

45. DiStefano MT, Roth Flach RJ, Senol-Cosar O, Danai LV, Virbasius JV, Nicoloro SM, et al. Adipocyte-specific Hypoxia-inducible gene 2 promotes fat deposition and diet-induced insulin resistance. Mol Metab. (2016) 5:1149–61. doi: 10.1016/j.molmet.2016.09.009

PubMed Abstract | Crossref Full Text | Google Scholar

46. Hartman GD, Muniyandi A, Sishtla K, Kpenu EK, Miller WP, Kaplan BA, et al. Ref-1 redox activity regulates retinal neovascularization by modulating transcriptional activation of HIF-1α. FASEB J. (2025) 39:e70348. doi: 10.1096/fj.202401989RR

PubMed Abstract | Crossref Full Text | Google Scholar

47. Zhang R, Chang R, Wang H, Chen J, Lu C, Fan K, et al. Untargeted metabolomic and proteomic analysis implicates SIRT2 as a novel therapeutic target for diabetic nephropathy. Sci Rep. (2025) 15:4236. doi: 10.1038/s41598-024-80492-1

PubMed Abstract | Crossref Full Text | Google Scholar

48. Regazzetti C, Peraldi P, Grémeaux T, Najem-Lendom R, Ben-Sahra I, Cormont M, et al. Hypoxia decreases insulin signaling pathways in adipocytes. Diabetes. (2009) 58:95–103. doi: 10.2337/db08-0457

PubMed Abstract | Crossref Full Text | Google Scholar

49. Dang EV, Barbi J, Yang HY, Jinasena D, Yu H, Zheng Y, et al. Control of T(H)17/T(reg) balance by hypoxia-inducible factor 1. Cell. (2011) 146:772–84. doi: 10.1016/j.cell.2011.07.033

PubMed Abstract | Crossref Full Text | Google Scholar

50. Hsu TS and Lai MZ. Hypoxia-inducible factor 1α plays a predominantly negative role in regulatory T cell functions. J Leukoc Biol. (2018) 104:911–8. doi: 10.1002/JLB.MR1217-481R

PubMed Abstract | Crossref Full Text | Google Scholar

51. Feldhoff LM, Rueda CM, Moreno-Fernandez ME, Sauer J, Jackson CM, Chougnet CA, et al. IL-1β induced HIF-1α inhibits the differentiation of human FOXP3(+) T cells. Sci Rep. (2017) 7:465. doi: 10.1038/s41598-017-00508-x

PubMed Abstract | Crossref Full Text | Google Scholar

52. Miska J, Lee-Chang C, Rashidi A, Muroski ME, Chang AL, Lopez-Rosas A, et al. HIF-1α Is a metabolic switch between glycolytic-driven migration and oxidative phosphorylation-driven immunosuppression of tregs in glioblastoma. Cell Rep. (2019) 27:226–37.e4. doi: 10.1016/j.celrep.2019.03.029

PubMed Abstract | Crossref Full Text | Google Scholar

53. Li Y, Zhu J, Yue C, Song S, Tian L, and Wang Y. Recent advances in pancreatic α-cell transdifferentiation for diabetes therapy. Front Immunol. (2025) 16:1551372. doi: 10.3389/fimmu.2025.1551372

PubMed Abstract | Crossref Full Text | Google Scholar

54. Deng J, Gan W, Hu C, Liu Z, Chen N, Jia C, et al. San Huang Xiao Yan recipe promoted wound healing in diabetic ulcer mice by inhibiting Th17 cell differentiation. J Ethnopharmacol. (2025) 341:119243. doi: 10.1016/j.jep.2024.119243

PubMed Abstract | Crossref Full Text | Google Scholar

55. Eguchi N, Vaziri ND, Dafoe DC, and Ichii H. The role of oxidative stress in pancreatic β Cell dysfunction in diabetes. Int J Mol Sci. (2021) 22. doi: 10.3390/ijms22041509

PubMed Abstract | Crossref Full Text | Google Scholar

56. Hasnain SZ, Prins JB, and McGuckin MA. Oxidative and endoplasmic reticulum stress in β-cell dysfunction in diabetes. J Mol Endocrinol. (2016) 56:R33–54. doi: 10.1530/JME-15-0232

PubMed Abstract | Crossref Full Text | Google Scholar

57. Flood D, Lee ES, and Taylor CT. Intracellular energy production and distribution in hypoxia. J Biol Chem. (2023) 299:105103. doi: 10.1016/j.jbc.2023.105103

PubMed Abstract | Crossref Full Text | Google Scholar

58. Wheaton WW and Chandel NS. Hypoxia. 2. Hypoxia regulates cellular metabolism. Am J Physiol Cell Physiol. (2011) 300:C385–93. doi: 10.1152/ajpcell.00485.2010

PubMed Abstract | Crossref Full Text | Google Scholar

59. Satapati S, Sunny NE, Kucejova B, Fu X, He TT, Méndez-Lucas A, et al. Elevated TCA cycle function in the pathology of diet-induced hepatic insulin resistance and fatty liver. J Lipid Res. (2012) 53:1080–92. doi: 10.1194/jlr.M023382

PubMed Abstract | Crossref Full Text | Google Scholar

60. Zhong L, Tran T, Baguley TD, Lee SJ, Henke A, To A, et al. A novel inhibitor of inducible NOS dimerization protects against cytokine-induced rat beta cell dysfunction. Br J Pharmacol. (2018) 175:3470–85. doi: 10.1111/bph.v175.17

PubMed Abstract | Crossref Full Text | Google Scholar

61. Robertson RP. Chronic oxidative stress as a central mechanism for glucose toxicity in pancreatic islet beta cells in diabetes. J Biol Chem. (2004) 279:42351–4. doi: 10.1074/jbc.R400019200

PubMed Abstract | Crossref Full Text | Google Scholar

62. Lee WD, Weilandt DR, Liang L, MacArthur MR, Jaiswal N, Ong O, et al. Lactate homeostasis is maintained through regulation of glycolysis and lipolysis. Cell Metab. (2025). doi: 10.1016/j.cmet.2024.12.009

PubMed Abstract | Crossref Full Text | Google Scholar

63. Cantley J, Grey ST, Maxwell PH, and Withers DJ. The hypoxia response pathway and β-cell function. Diabetes Obes Metab. (2010) 12 Suppl 2:159–67.

Google Scholar

64. Zhang Z, Huang Q, Zhao D, Lian F, Li X, and Qi W. The impact of oxidative stress-induced mitochondrial dysfunction on diabetic microvascular complications. Front Endocrinol (Lausanne). (2023) 14:1112363. doi: 10.3389/fendo.2023.1112363

PubMed Abstract | Crossref Full Text | Google Scholar

65. Guo C, Sun L, Chen X, and Zhang D. Oxidative stress, mitochondrial damage and neurodegenerative diseases. Neural Regener Res. (2013) 8:2003–14.

Google Scholar

66. Kowalczyk P, Sulejczak D, Kleczkowska P, Bukowska-Ośko I, Kucia M, Popiel M, et al. Mitochondrial oxidative stress-A causative factor and therapeutic target in many diseases. Int J Mol Sci. (2021) 22. doi: 10.3390/ijms222413384

PubMed Abstract | Crossref Full Text | Google Scholar

67. Luo Z, Tian M, Yang G, Tan Q, Chen Y, Li G, et al. Hypoxia signaling in human health and diseases: implications and prospects for therapeutics. Signal Transduct Target Ther. (2022) 7:218. doi: 10.1038/s41392-022-01080-1

PubMed Abstract | Crossref Full Text | Google Scholar

68. Lee YS, Kim JW, Osborne O, Oh DY, Sasik R, Schenk S, et al. Increased adipocyte O2 consumption triggers HIF-1α, causing inflammation and insulin resistance in obesity. Cell. (2014) 157:1339–52. doi: 10.1016/j.cell.2014.05.012

PubMed Abstract | Crossref Full Text | Google Scholar

69. Blagov AV, Summerhill VI, Sukhorukov VN, Popov MA, Grechko AV, and Orekhov AN. Type 1 diabetes mellitus: Inflammation, mitophagy, and mitochondrial function. Mitochondrion. (2023) 72:11–21. doi: 10.1016/j.mito.2023.07.002

PubMed Abstract | Crossref Full Text | Google Scholar

70. Sidarala V, Pearson GL, Parekh VS, Thompson B, Christen L, Gingerich MA, et al. Mitophagy protects β cells from inflammatory damage in diabetes. JCI Insight. (2020) 5. doi: 10.1172/jci.insight.141138

PubMed Abstract | Crossref Full Text | Google Scholar

71. Oh SJ, Park K, Sonn SK, Oh GT, and Lee MS. Pancreatic β-cell mitophagy as an adaptive response to metabolic stress and the underlying mechanism that involves lysosomal Ca(2+) release. Exp Mol Med. (2023) 55:1922–32. doi: 10.1038/s12276-023-01055-4

PubMed Abstract | Crossref Full Text | Google Scholar

72. Oh H, Kang MK, Park SH, Kim DY, Kim SI, Oh SY, et al. Asaronic acid inhibits ER stress sensors and boosts functionality of ubiquitin-proteasomal degradation in 7β-hydroxycholesterol-loaded macrophages. Phytomedicine. (2021) 92:153763. doi: 10.1016/j.phymed.2021.153763

PubMed Abstract | Crossref Full Text | Google Scholar

73. Shrestha N, Reinert RB, and Qi L. Endoplasmic reticulum protein quality control in β Cells. Semin Cell Dev Biol. (2020) 103:59–67. doi: 10.1016/j.semcdb.2020.04.006

PubMed Abstract | Crossref Full Text | Google Scholar

74. Thomaidou S, Zaldumbide A, and Roep BO. Islet stress, degradation and autoimmunity. Diabetes Obes Metab. (2018) 20 Suppl 2:88–94.

Google Scholar

75. Shergalis AG, Hu S, Bankhead A 3rd, and Neamati N. Role of the ERO1-PDI interaction in oxidative protein folding and disease. Pharmacol Ther. (2020) 210:107525. doi: 10.1016/j.pharmthera.2020.107525

PubMed Abstract | Crossref Full Text | Google Scholar

76. Jha V, Kumari T, Manickam V, Assar Z, Olson KL, Min JK, et al. ERO1-PDI redox signaling in health and disease. Antioxid Redox Signal. (2021) 35:1093–115. doi: 10.1089/ars.2021.0018

PubMed Abstract | Crossref Full Text | Google Scholar

77. Benham AM, van Lith M, Sitia R, and Braakman I. Ero1-PDI interactions, the response to redox flux and the implications for disulfide bond formation in the mammalian endoplasmic reticulum. Philos Trans R Soc Lond B Biol Sci. (2013) 368:20110403. doi: 10.1098/rstb.2011.0403

PubMed Abstract | Crossref Full Text | Google Scholar

78. Wang L and Wang CC. Oxidative protein folding fidelity and redoxtasis in the endoplasmic reticulum. Trends Biochem Sci. (2023) 48:40–52. doi: 10.1016/j.tibs.2022.06.011

PubMed Abstract | Crossref Full Text | Google Scholar

79. Bartoszewska S and Collawn JF. Unfolded protein response (UPR) integrated signaling networks determine cell fate during hypoxia. Cell Mol Biol Lett. (2020) 25:18. doi: 10.1186/s11658-020-00212-1

PubMed Abstract | Crossref Full Text | Google Scholar

80. Hetz C, Zhang K, and Kaufman RJ. Mechanisms, regulation and functions of the unfolded protein response. Nat Rev Mol Cell Biol. (2020) 21:421–38. doi: 10.1038/s41580-020-0250-z

PubMed Abstract | Crossref Full Text | Google Scholar

81. Mohan R, Jo S, Lockridge A, Ferrington DA, Murray K, Eschenlauer A, et al. OGT regulates mitochondrial biogenesis and function via diabetes susceptibility gene pdx1. Diabetes. (2021) 70:2608–25. doi: 10.2337/db21-0468

PubMed Abstract | Crossref Full Text | Google Scholar

82. Dhayalan B, Chen YS, Ni CL, and Weiss MA. Synthetic studies of the mutant proinsulin syndrome demonstrate correlation between folding efficiency and age of diabetes onset. Int J Pept Res Ther. (2025) 31.

PubMed Abstract | Google Scholar

83. Huber MK, Widener AE, Cuaycal AE, Smurlick D, Butterworth EA, Lenchik NI, et al. Beta cell dysfunction occurs independently of insulitis in type 1 diabetes pathogenesis. bioRxiv. (2024). doi: 10.1101/2024.12.29.630665

PubMed Abstract | Crossref Full Text | Google Scholar

84. Dekkers MC, Pu X, Enciso-Martinez A, and Zaldumbide A. Beta-cell-derived extracellular vesicles: mediators of intercellular communication in the islet microenvironment in type 1 diabetes. Cells. (2024) 13. doi: 10.3390/cells13231996

PubMed Abstract | Crossref Full Text | Google Scholar

85. Gao H, Nepovimova E, Heger Z, Valko M, Wu Q, Kuca K, et al. Role of hypoxia in cellular senescence. Pharmacol Res. (2023) 194:106841. doi: 10.1016/j.phrs.2023.106841

PubMed Abstract | Crossref Full Text | Google Scholar

86. Saha S, Panigrahi DP, Patil S, and Bhutia SK. Autophagy in health and disease: A comprehensive review. BioMed Pharmacother. (2018) 104:485–95. doi: 10.1016/j.biopha.2018.05.007

PubMed Abstract | Crossref Full Text | Google Scholar

87. Ren J and Zhang Y. Targeting autophagy in aging and aging-related cardiovascular diseases. Trends Pharmacol Sci. (2018) 39:1064–76. doi: 10.1016/j.tips.2018.10.005

PubMed Abstract | Crossref Full Text | Google Scholar

88. Khater SI, El-Emam MMA, Abdellatif H, Mostafa M, Khamis T, Soliman RHM, et al. Lipid nanoparticles of quercetin (QU-Lip) alleviated pancreatic microenvironment in diabetic male rats: The interplay between oxidative stress - unfolded protein response (UPR) - autophagy, and their regulatory miRNA. Life Sci. (2024) 344:122546. doi: 10.1016/j.lfs.2024.122546

PubMed Abstract | Crossref Full Text | Google Scholar

89. Ye S, Cheng Z, Zhuo D, and Liu S. Different types of cell death in diabetic neuropathy: A focus on mechanisms and therapeutic strategies. Int J Mol Sci. (2024) 25. doi: 10.3390/ijms25158126

PubMed Abstract | Crossref Full Text | Google Scholar

90. Kim J, Kundu M, Viollet B, and Guan KL. AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1. Nat Cell Biol. (2011) 13:132–41. doi: 10.1038/ncb2152

PubMed Abstract | Crossref Full Text | Google Scholar

91. Egan DF, Shackelford DB, Mihaylova MM, Gelino S, Kohnz RA, Mair W, et al. Phosphorylation of ULK1 (hATG1) by AMP-activated protein kinase connects energy sensing to mitophagy. Science. (2011) 331:456–61. doi: 10.1126/science.1196371

PubMed Abstract | Crossref Full Text | Google Scholar

92. Ge Y, Zhou M, Chen C, Wu X, and Wang X. Role of AMPK mediated pathways in autophagy and aging. Biochimie. (2022) 195:100–13. doi: 10.1016/j.biochi.2021.11.008

PubMed Abstract | Crossref Full Text | Google Scholar

93. Li J, Madsen AB, Knudsen JR, Henriquez-Olguin C, Persson KW, Li Z, et al. mTOR Ser1261 is an AMPK-dependent phosphosite in mouse and human skeletal muscle not required for mTORC2 activity. FASEB J. (2025) 39:e70277. doi: 10.1096/fj.202402064R

PubMed Abstract | Crossref Full Text | Google Scholar

94. Saha S, Fang X, Green CD, and Das A. mTORC1 and SGLT2 inhibitors-A therapeutic perspective for diabetic cardiomyopathy. Int J Mol Sci. (2023) 24. doi: 10.3390/ijms242015078

PubMed Abstract | Crossref Full Text | Google Scholar

95. Yao Y, Chen Y, Chen H, Pan X, Li X, Liu W, et al. Black mulberry extract inhibits hepatic adipogenesis through AMPK/mTOR signaling pathway in T2DM mice. J Ethnopharmacol. (2024) 319:117216. doi: 10.1016/j.jep.2023.117216

PubMed Abstract | Crossref Full Text | Google Scholar

96. Chen Y, Meng Z, Li Y, Liu S, Hu P, and Luo E. Advanced glycation end products and reactive oxygen species: uncovering the potential role of ferroptosis in diabetic complications. Mol Med. (2024) 30:141. doi: 10.1186/s10020-024-00905-9

PubMed Abstract | Crossref Full Text | Google Scholar

97. Amin R, Hidayat R, Maritska Z, and Putri TW. Activated growth factor from platelets as treatment for diabetic retinopathy through antioxidant-oxidative stress pathway. Diabetes Metab Syndr Obes. (2025) 18:305–13. doi: 10.2147/DMSO.S490055

PubMed Abstract | Crossref Full Text | Google Scholar

98. Liang J, Cheng S, Song Q, Tang Y, Wang Q, Chen H, et al. Effect of mesenchymal stem cell-derived extracellular vesicles induced by advanced glycation end products on energy metabolism in vascular endothelial cells. Kidney Int Rep. (2025) 10:227–46. doi: 10.1016/j.ekir.2024.10.015

PubMed Abstract | Crossref Full Text | Google Scholar

99. Liu S, Lu M, Zhang M, Sun X, Luo B, and Wu Y. Multifaceted catalytic glucose depletion and reactive oxygen species-scavenging nanoenzyme composite hydrogel for facilitating diabetic bone regeneration. ACS Nano. (2025) 19:2677–94. doi: 10.1021/acsnano.4c14596

PubMed Abstract | Crossref Full Text | Google Scholar

100. Rajamani U and Essop MF. Hyperglycemia-mediated activation of the hexosamine biosynthetic pathway results in myocardial apoptosis. Am J Physiol Cell Physiol. (2010) 299:C139–47. doi: 10.1152/ajpcell.00020.2010

PubMed Abstract | Crossref Full Text | Google Scholar

101. Wang F, Xu C, Reece EA, Li X, Wu Y, Harman C, et al. Protein kinase C-alpha suppresses autophagy and induces neural tube defects via miR-129–2 in diabetic pregnancy. Nat Commun. (2017) 8:15182. doi: 10.1038/ncomms15182

PubMed Abstract | Crossref Full Text | Google Scholar

102. Yang D, Livingston MJ, Liu Z, Dong G, Zhang M, Chen JK, et al. Autophagy in diabetic kidney disease: regulation, pathological role and therapeutic potential. Cell Mol Life Sci. (2018) 75:669–88. doi: 10.1007/s00018-017-2639-1

PubMed Abstract | Crossref Full Text | Google Scholar

103. Zhang C, Gu L, Xie H, Liu Y, Huang P, Zhang J, et al. Glucose transport, transporters and metabolism in diabetic retinopathy. Biochim Biophys Acta Mol Basis Dis. (2024) 1870:166995. doi: 10.1016/j.bbadis.2023.166995

PubMed Abstract | Crossref Full Text | Google Scholar

104. Huang M, Paglialunga S, Wong JM, Hoang M, Pillai R, and Joseph JW. Role of prolyl hydroxylase domain proteins in the regulation of insulin secretion. Physiol Rep. (2016) 4. doi: 10.14814/phy2.12722

PubMed Abstract | Crossref Full Text | Google Scholar

105. Hoang M, Jentz E, Janssen SM, Nasteska D, Cuozzo F, Hodson DJ, et al. Isoform-specific roles of prolyl hydroxylases in the regulation of pancreatic β-cell function. Endocrinology. (2022) 163. doi: 10.1210/endocr/bqab226

PubMed Abstract | Crossref Full Text | Google Scholar

106. Wróbel M, Rokicka D, Gołaś A, Drozd M, Nowowiejska-Wiewióra A, Pyka Ł, et al. Combined aerobic and resistance training performed under conditions of normobaric hypoxia and normoxia has the same impact on metabolic control in men with type 1 diabetes. Int J Environ Res Public Health. (2021) 18. doi: 10.3390/ijerph182413058

PubMed Abstract | Crossref Full Text | Google Scholar

107. Isildar B, Ozkan S, Sahin H, Ercin M, Gezginci-Oktayoglu S, and Koyuturk M. Preconditioning of human umbilical cord mesenchymal stem cells with deferoxamine potentiates the capacity of the secretome released from the cells and promotes immunomodulation and beta cell regeneration in a rat model of type 1 diabetes. Int Immunopharmacol. (2024) 129:111662. doi: 10.1016/j.intimp.2024.111662

PubMed Abstract | Crossref Full Text | Google Scholar

108. Wei L, Zhang L, Yang L, Wang X, Zhao C, and Zhao D. Protective effect of mesenchymal stem cells on isolated islets survival and against hypoxia associated with the HIF-1α/PFKFB3 pathway. Cell Transplant. (2022) 31:9636897211073127. doi: 10.1177/09636897211073127

PubMed Abstract | Crossref Full Text | Google Scholar

109. Kelly AC, Bidwell CA, McCarthy FM, Taska DJ, Anderson MJ, Camacho LE, et al. RNA sequencing exposes adaptive and immune responses to intrauterine growth restriction in fetal sheep islets. Endocrinology. (2017) 158:743–55. doi: 10.1210/en.2016-1901

PubMed Abstract | Crossref Full Text | Google Scholar

110. Zhao Z, Chen X, Dowbaj AM, Sljukic A, Bratlie K, Lin L, et al. Organoids. Nat Rev Methods Primers. (2022) 2. doi: 10.1038/s43586-022-00174-y

PubMed Abstract | Crossref Full Text | Google Scholar

111. Sun D, Guan X, Moran AE, Wu LY, Qian DZ, Schedin P, et al. Identifying phenotype-associated subpopulations by integrating bulk and single-cell sequencing data. Nat Biotechnol. (2022) 40:527–38. doi: 10.1038/s41587-021-01091-3

PubMed Abstract | Crossref Full Text | Google Scholar

112. Camp JG, Platt R, and Treutlein B. Mapping human cell phenotypes to genotypes with single-cell genomics. Science. (2019) 365:1401–5. doi: 10.1126/science.aax6648

PubMed Abstract | Crossref Full Text | Google Scholar

113. Qiu J, Zhu P, Shi X, Xia J, Dong S, and Chen L. Identification of a pancreatic stellate cell gene signature and lncRNA interactions associated with type 2 diabetes progression. Front Endocrinol (Lausanne). (2024) 15:1532609. doi: 10.3389/fendo.2024.1532609

PubMed Abstract | Crossref Full Text | Google Scholar

114. Song Z, Li S, Shang Z, Lv W, Cheng X, Meng X, et al. Integrating multi-omics data to analyze the potential pathogenic mechanism of CTSH gene involved in type 1 diabetes in the exocrine pancreas. Brief Funct Genomics. (2024) 23:406–17. doi: 10.1093/bfgp/elad052

PubMed Abstract | Crossref Full Text | Google Scholar

115. Gharatape A, Sadeghi-Abandansari H, Ghanbari H, Basiri M, and Faridi-Majidi R. Synthesis and characterization of poly (β-amino ester) polyplex nanocarrier with high encapsulation and uptake efficiency: impact of extracellular conditions. Nanomed (Lond). (2025) 20:125–39. doi: 10.1080/17435889.2024.2440307

PubMed Abstract | Crossref Full Text | Google Scholar

116. Ma H, Xu J, Fang H, Su Y, Lu Y, Shu Y, et al. A capsule-based scaffold incorporating decellularized extracellular matrix and curcumin for islet beta cell therapy in type 1 diabetes mellitus. Biofabrication. (2024) 16. doi: 10.1088/1758-5090/ad7907

PubMed Abstract | Crossref Full Text | Google Scholar

117. Tong MQ, Lu CT, Huang LT, Yang JJ, Yang ST, Chen HB, et al. Polyphenol-driven facile assembly of a nanosized acid fibroblast growth factor-containing coacervate accelerates the healing of diabetic wounds. Acta Biomater. (2023) 157:467–86. doi: 10.1016/j.actbio.2022.11.054

PubMed Abstract | Crossref Full Text | Google Scholar

118. Mahaling B, Srinivasarao DA, Raghu G, Kasam RK, Bhanuprakash Reddy G, and Katti DS. A non-invasive nanoparticle mediated delivery of triamcinolone acetonide ameliorates diabetic retinopathy in rats. Nanoscale. (2018) 10:16485–98. doi: 10.1039/C8NR00058A

PubMed Abstract | Crossref Full Text | Google Scholar

119. Bougnères P and Valleron AJ. Causes of early-onset type 1 diabetes: toward data-driven environmental approaches. J Exp Med. (2008) 205:2953–7. doi: 10.1084/jem.20082622

PubMed Abstract | Crossref Full Text | Google Scholar

120. Degen I, Robson Brown K, Reeve HWJ, and Abdallah ZS. Beyond expected patterns in insulin needs of people with type 1 diabetes: temporal analysis of automated insulin delivery data. JMIRx Med. (2024) 5:e44384. doi: 10.2196/44384

PubMed Abstract | Crossref Full Text | Google Scholar

121. Nemat H, Khadem H, Elliott J, and Benaissa M. Data-driven blood glucose level prediction in type 1 diabetes: a comprehensive comparative analysis. Sci Rep. (2024) 14:21863. doi: 10.1038/s41598-024-70277-x

PubMed Abstract | Crossref Full Text | Google Scholar

122. Kumari R, Anand PK, and Shin J. Improving the accuracy of continuous blood glucose measurement using personalized calibration and machine learning. Diagn (Basel). (2023) 13. doi: 10.3390/diagnostics13152514

PubMed Abstract | Crossref Full Text | Google Scholar

123. Mittal R WM, Rendon A, Shafazand S, Lemos JRN, and Hirani K. Harnessing machine learning, a subset of artificial intelligence, for early detection and diagnosis of type 1 diabetes: A systematic review. Int J Mol Sci. (2025) 26:3935. doi: 10.3390/ijms26093935

PubMed Abstract | Crossref Full Text | Google Scholar

124. Woldaregay AZ, Årsand E, Walderhaug S, Albers D, Mamykina L, Botsis T, et al. Data-driven modeling and prediction of blood glucose dynamics: Machine learning applications in type 1 diabetes. Artif Intell Med. (2019) 98:109–34. doi: 10.1016/j.artmed.2019.07.007

PubMed Abstract | Crossref Full Text | Google Scholar

125. Saleem O and Iqbal J. Blood-glucose regulator design for diabetics based on LQIR-driven Sliding-Mode-Controller with self-adaptive reaching law. PloS One. (2024) 19:e0314479. doi: 10.1371/journal.pone.0314479

PubMed Abstract | Crossref Full Text | Google Scholar

126. Diaz CJ, Colmegna P, and Pryor E. Breton MD. A performance-based adaptation index for automated insulin delivery systems. J Diabetes Sci Technol. (2025), 19322968251315499.

PubMed Abstract | Google Scholar

127. Yu J, Lalwani A, and Gunton JE. β-cell deletion of hypoxia-inducible factor 1α (HIF-1α) increases pancreatic β-cell susceptibility to streptozotocin. Int J Mol Sci. (2024) 25. doi: 10.3390/ijms252413451

PubMed Abstract | Crossref Full Text | Google Scholar

128. Canavesi I, Viswakarma N, Epel B, and Kotecha M. In vivo mouse abdominal oxygen imaging and assessment of subcutaneously implanted beta cell replacement devices. Mol Imaging Biol. (2024). doi: 10.1007/s11307-024-01963-5

PubMed Abstract | Crossref Full Text | Google Scholar

129. Paez-Mayorga J, Lukin I, Emerich D, de Vos P, Orive G, and Grattoni A. Emerging strategies for beta cell transplantation to treat diabetes. Trends Pharmacol Sci. (2022) 43:221–33. doi: 10.1016/j.tips.2021.11.007

PubMed Abstract | Crossref Full Text | Google Scholar

130. Ghahremani PT, BaniArdalan S, Alehossein P, Parveen A, Jorjani M, Brown CM, et al. Poststroke hyperglycemia dysregulates cap-dependent translation in neural cells. Life Sci. (2025) 361:123336. doi: 10.1016/j.lfs.2024.123336

PubMed Abstract | Crossref Full Text | Google Scholar

131. Seong J, Kim M, Yoo J, Mack DL, Lee JH, and Joo J. Sustained release of HIF-2α Inhibitors using biodegradable porous silicon carriers for enhanced immunogenic cell death of Malignant merkel cell carcinoma. ACS Appl Mater Interfaces. (2025) 17:7449–61. doi: 10.1021/acsami.4c19961

PubMed Abstract | Crossref Full Text | Google Scholar

132. Mendoza SV, Wells KV, Murugesh DK, Hum NR, Sebastian A, Gorman BM, et al. Osteocytic oxygen sensing: Distinct impacts of VHL and HIF-2alpha on bone integrity. Bone. (2025) 192:117339. doi: 10.1016/j.bone.2024.117339

PubMed Abstract | Crossref Full Text | Google Scholar

133. Cooper TT, Sherman SE, Bell GI, Dayarathna T, McRae DM, Ma J, et al. Ultrafiltration and injection of islet regenerative stimuli secreted by pancreatic mesenchymal stromal cells. Stem Cells Dev. (2021) 30:247–64. doi: 10.1089/scd.2020.0206

PubMed Abstract | Crossref Full Text | Google Scholar

134. Forkan CP, Shrestha A, Yu A, Chuang C, Pociot F, and Yarani R. Could hypoxic conditioning augment the potential of mesenchymal stromal cell-derived extracellular vesicles as a treatment for type 1 diabetes? Stem Cell Res Ther. (2025) 16:37. doi: 10.1186/s13287-025-04153-4

PubMed Abstract | Crossref Full Text | Google Scholar

135. Gálisová A, Fábryová E, Jirák D, Sticová E, Lodererová A, Herynek V, et al. Multimodal imaging reveals improvement of blood supply to an artificial cell transplant site induced by bioluminescent mesenchymal stem cells. Mol Imaging Biol. (2017) 19:15–23. doi: 10.1007/s11307-016-0986-1

PubMed Abstract | Crossref Full Text | Google Scholar

136. Dong X, Xu J, Du K, Chen X, Shu H, and Yu S. Plateau hypoxia-induced upregulation of reticulon 4 pathway mediates altered autophagic flux involved in blood-brain barrier disruption after traumatic brain injury. Neuroreport. (2025) 36:81–92. doi: 10.1097/WNR.0000000000002122

PubMed Abstract | Crossref Full Text | Google Scholar

137. Zhou X, Cai M, Yang F, Huang L, Ling Y, Zhang Y, et al. Hypoxia-induced autophagy in pancreatic cancer counteracts the cytotoxicity of CD8(+) T cells by inhibiting the expression of MHC-I. Genes Immun. (2025) 26:45–53. doi: 10.1038/s41435-024-00315-1

PubMed Abstract | Crossref Full Text | Google Scholar

138. Cheng W, Wang C, Ma M, and Zhou Y. Hydroxyurea mitigates diabetic kidney disease through mTOR-S6K signaling pathway in STZ-induced diabetic mice. Front Cell Dev Biol. (2025) 13:1529901. doi: 10.3389/fcell.2025.1529901

PubMed Abstract | Crossref Full Text | Google Scholar

139. Hardie DG, Ross FA, and Hawley SA. AMPK: a nutrient and energy sensor that maintains energy homeostasis. Nat Rev Mol Cell Biol. (2012) 13:251–62. doi: 10.1038/nrm3311

PubMed Abstract | Crossref Full Text | Google Scholar

140. Jung CH, Ro SH, Cao J, Otto NM, and Kim DH. mTOR regulation of autophagy. FEBS Lett. (2010) 584:1287–95. doi: 10.1016/j.febslet.2010.01.017

PubMed Abstract | Crossref Full Text | Google Scholar

141. Saxton RA and Sabatini DM. mTOR signaling in growth, metabolism, and disease. Cell. (2017) 168:960–76. doi: 10.1016/j.cell.2017.02.004

PubMed Abstract | Crossref Full Text | Google Scholar

142. Mihaylova MM and Shaw RJ. The AMPK signaling pathway coordinates cell growth, autophagy and metabolism. Nat Cell Biol. (2011) 13:1016–23. doi: 10.1038/ncb2329

PubMed Abstract | Crossref Full Text | Google Scholar

143. Dutta D, Ray P, De A, Ghosh A, Hazra RS, Ghosh P, et al. pH-responsive targeted nanoparticles release ERK-inhibitor in the hypoxic zone and sensitize free gemcitabine in mutant K-Ras-addicted pancreatic cancer cells and mouse model. PloS One. (2024) 19:e0297749. doi: 10.1371/journal.pone.0297749

PubMed Abstract | Crossref Full Text | Google Scholar

144. Gu X and Minko T. Targeted nanoparticle-based diagnostic and treatment options for pancreatic cancer. Cancers (Basel). (2024) 16. doi: 10.3390/cancers16081589

PubMed Abstract | Crossref Full Text | Google Scholar

145. Tomei AA, Villa C, and Ricordi C. Development of an encapsulated stem cell-based therapy for diabetes. Expert Opin Biol Ther. (2015) 15:1321–36. doi: 10.1517/14712598.2015.1055242

PubMed Abstract | Crossref Full Text | Google Scholar

146. Desai T and Shea LD. Advances in islet encapsulation technologies. Nat Rev Drug Discov. (2017) 16:338–50. doi: 10.1038/nrd.2016.232

PubMed Abstract | Crossref Full Text | Google Scholar

147. Li C, Huang Z, Gao N, Zheng J, and Guan J. Injectable, thermosensitive, fast gelation, bioeliminable, and oxygen sensitive hydrogels. Mater Sci Eng C Mater Biol Appl. (2019) 99:1191–8. doi: 10.1016/j.msec.2019.02.075

PubMed Abstract | Crossref Full Text | Google Scholar

148. Ashrafi K, Heaysman CL, Phillips GJ, Lloyd AW, and Lewis AL. Towards hypoxia-responsive drug-eluting embolization beads. Int J Pharm. (2017) 524:226–37. doi: 10.1016/j.ijpharm.2017.03.084

PubMed Abstract | Crossref Full Text | Google Scholar

149. Wang X, Gong L, Wei C, Zhao Y, Ran L, Li P, et al. Inhibition of NSUN6 protects against intermittent hypoxia-induced oxidative stress and inflammatory response in adipose tissue through suppressing macrophage ferroptosis and M1 polarization. Life Sci. (2025) 364:123433. doi: 10.1016/j.lfs.2025.123433

PubMed Abstract | Crossref Full Text | Google Scholar

150. Liu N, Cai X, Liu T, Zou J, Wang L, Wang G, et al. Hypoxia-inducible factor-1α mediates the expression of mature β cell-disallowed genes in hypoxia-induced β cell dedifferentiation. Biochem Biophys Res Commun. (2020) 523:382–8. doi: 10.1016/j.bbrc.2019.12.063

PubMed Abstract | Crossref Full Text | Google Scholar

151. Palazon A, Goldrath AW, Nizet V, and Johnson RS. HIF transcription factors, inflammation, and immunity. Immunity. (2014) 41:518–28. doi: 10.1016/j.immuni.2014.09.008

PubMed Abstract | Crossref Full Text | Google Scholar

152. Zhang C, Moriguchi T, Kajihara M, Esaki R, Harada A, Shimohata H, et al. MafA is a key regulator of glucose-stimulated insulin secretion. Mol Cell Biol. (2005) 25:4969–76. doi: 10.1128/MCB.25.12.4969-4976.2005

PubMed Abstract | Crossref Full Text | Google Scholar

153. Zhu Y, Liu Q, Zhou Z, and Ikeda Y. PDX1, Neurogenin-3, and MAFA: critical transcription regulators for beta cell development and regeneration. Stem Cell Res Ther. (2017) 8:240. doi: 10.1186/s13287-017-0694-z

PubMed Abstract | Crossref Full Text | Google Scholar

Keywords: hypoxia, type 1 diabetes, immune dysregulation, hypoxia-inducible factors, autoimmune response, regulatory T cells, hyperglycemia, inflammation

Citation: Mittal R, Lemos JRN, Chapagain P and Hirani K (2025) Interplay of hypoxia, immune dysregulation, and metabolic stress in pathophysiology of type 1 diabetes. Front. Immunol. 16:1599321. doi: 10.3389/fimmu.2025.1599321

Received: 24 March 2025; Accepted: 14 May 2025;
Published: 04 June 2025.

Edited by:

Beatrice Dufrusine, University of Teramo, Italy

Reviewed by:

Hans Dooms, National Jewish Health, United States
Tamer A. Addissouky, University of Menoufia, Shibin Al Kawm, Egypt

Copyright © 2025 Mittal, Lemos, Chapagain and Hirani. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Rahul Mittal, ci5taXR0YWwxMUBtZWQubWlhbWkuZWR1; Khemraj Hirani, a2hpcmFuaUBtZWQubWlhbWkuZWR1

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.