v
Search
Advanced

Publications > Journals > Exploratory Research and Hypothesis in Medicine> Article Full Text

  • OPEN ACCESS

The Link Between Immune Aging and Type 2 Diabetes: A Review of Mechanisms and Implications

  • Swarup K. Chakrabarti1,*  and
  • Dhrubajyoti Chattopadhyay1,2
 Author information 

Abstract

Type 2 diabetes (T2D) is a metabolic disorder characterized by insulin resistance (IR), inflammation, and dysregulation in glucose metabolism. The disease is spreading globally, partly due to aging, which can damage the immune system and speed up the progression of the metabolic disorder. This review primarily delves into the triggers for T2D within the framework of the ominous octet, which emphasizes 8 principal factors under the “ominous octet” framework that contribute to high blood glucose and associated metabolic disorders. The article studies the interplay of hyperinsulinemia, mitochondrial dysfunction (MD), and endoplasmic reticulum (ER) stress with immune aging in driving disease progression affecting each component of the octet. MD and ER stress can result in defects in insulin signaling, ultimately leading to β-cell death. Chronic inflammation associated with aging, also known as inflammaging, especially affects older adults by worsening IR and glucose regulation, which creates a continuous sequence of metabolic problems. Thus, the “ominous octet” framework provides fundamental knowledge to develop personalized treatment approaches that target metabolic dysfunction together with ER stress, MD, and immune system imbalances. These strategies show promising potential to improve treatments for T2D and may lead to better health outcomes for older adults dealing with this condition.

Keywords

Type 2 diabetes, Insulin resistance, Chronic inflammation, Ominous octet, Mitochondrial dysfunction, Endoplasmic reticulum stress, Immune aging, Glucose metabolism, β-cell dysfunction, Hyperinsulinemia, Metabolic syndrome

Introduction

Demographic shifts and the aging immune system

Demographic projections show a significant rise in the global aging population. According to the World Health Organization (WHO), by 2030, one out of every six people globally is projected to be aged 60 years or above. The number of people in this age bracket is projected to rise from 1 billion in 2020 to 1.4 billion by 2030 and to surpass 2 billion by the year 2050. Moreover, the population aged 80 and above is projected to increase threefold during this period, reaching 426 million by the middle of the century.1 The demographic shift is calling for immediate attention to the issues of health associated with the aging process, particularly cellular senescence, through which the immune system becomes less effective as people grow older.2,3 As we age, cellular dysfunctions like telomere shortening and increased oxidative stress activate key signaling pathways, such as nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB).4,5 This, in turn, triggers the senescence-associated secretory phenotype (SASP), a complex pro-inflammatory response marked by secretion of cytokines, chemokines, growth factors, and proteases from senescent cells. SASP is essential in promoting chronic low-grade inflammation, which in turn can interfere with tissue homeostasis and cause the pathogenesis of many age-related diseases, including type 2 diabetes (T2D).6,7 This phenotype is characterized by excessive production of proinflammatory cytokines, chemokines, growth factors, and lipids. These substances not only impair immune balance but also abrogate inflammation resolution, worsening diseases associated with aging. Immunosenescence accelerates chronic inflammation and disrupts the delicate balance between the innate and adaptive immune systems.8 The innate immune response, largely driven by innate immune cells like dendritic cells, remains active throughout life.9 However, aging weakens adaptive immunity, resulting in the hyperactivation of innate immunity. This imbalance substantially reduces vaccine efficacy, increases susceptibility to infections, and heightens the vulnerability of older adults to opportunistic pathogens.10,11

Thymic involution: A core driver of immune aging

A key driver of immune dysfunction is thymic involution, the age-associated decline in thymus size and function.12,13 The thymus is essential for the maturation of hematopoietic stem cells into functional T cells. As the thymic output of naïve T cells decreases, the diversity of the T-cell repertoire becomes severely limited.14 This reduction impairs the immune system’s ability to mount robust responses to new pathogens. T-cell dysfunction and reduced immune surveillance speed up immunosenescence and raise the risk of chronic diseases in older individuals. Although the thymus naturally shrinks over time, conditions like aging can cause thymic atrophy. Both processes result in a decline in T cell production and function.15 During thymus involution with aging, the immune repertoire shrinks, reducing the diversity of T cell receptors. This limits the ability to recognize and respond to new pathogens, a key feature of immunosenescence.16,17 Put another way, immune aging causes the immune cells that have been in the body for a long time to become less effective. In this context, “immune aging” pertains to the general decline of the immune system with increasing age, whereas “immunosenescence” focuses on the specific decline in effectiveness and increased susceptibility to dysfunction of certain immune cells due to aging. Throughout the article, the two terms are used interchangeably, depending on the context. This process is intensified by the decrease in both the number and efficacy of immune cells as people grow older, which ultimately compromises the body’s capacity to identify and eliminate pathogens.16,17 In certain instances, this decline results in a condition referred to as immune paralysis, marked by a substantial reduction in immune system responsiveness, making it unable to effectively ward off pathogens or other immune threats.3 In essence, immune aging is a multifaceted phenomenon influenced by a variety of interconnected elements that both contribute to and arise from the decline in immune function. As depicted in Figure 1, significant factors leading to immune aging include persistent inflammation, immune system dysregulation, thymic involution, and thymic atrophy. These alterations initiate a cascade of downstream effects such as immune paralysis, a diminished immune repertoire, compromised adaptive and innate immunity, and metabolic dysfunction within immune cells.14–17 Together, these changes give rise to the concept of immunosenescence. The bidirectional arrows in the figure highlight the cyclical nature of this process, where each factor can worsen or amplify the others, leading to a self-reinforcing cycle of immune decline.

Summary of the impact of immune aging on immune cell function.
Fig. 1  Summary of the impact of immune aging on immune cell function.

Immune function deteriorates progressively with age through a range of mechanisms, including chronic inflammation, immune dysregulation, thymic involution, thymic atrophy, immune paralysis, and a reduction in the diversity of the immune repertoire. Additionally, defects in immunosenescence and immunometabolism contribute to this decline. Immune aging exerts a detrimental effect on both innate and adaptive immunity. The (+) symbol denotes activation, while the (-) symbol indicates inhibition.

Inflammaging and metabolic disorders

Moreover, chronic systemic inflammation, or “inflammaging”, is a key feature of age-related immune dysfunction.17 It plays a crucial role in the development of metabolic disorders, particularly insulin resistance (IR) and T2D.18,19 High levels of pro-inflammatory cytokines, like interleukin (IL)-6 and tumor necrosis factor-alpha (TNF-α), are characteristic of inflammaging.11,16,17 Interestingly, recent studies have underscored the alterations in metabolism as one of the primary drivers of organism dysfunctions during aging, the deterioration of which with age lowers immune cell function.20 Hence, the interplay between metabolism and inflammaging is beginning to receive increasing recognition in the regulation of the development of T2D in recent years. Importantly, hyperinsulinemia, IR, and inflammation establish the cycle, exacerbating the metabolic complications in older adults.21,22 It starts with elevated glucose levels, leading to hyperinsulinemia, which causes IR and triggers more insulin production and inflammation. At first, pancreatic β-cells can withstand the increased workload, but over time, IR leads to β-cell dysfunction, loss of β-cell mass, and the onset of T2D.23,24 At the cellular level, the combination of hyperinsulinemia and age-related inflammation plays a key role in causing organelle dysfunction, mainly mitochondrial dysfunction (MD) and endoplasmic reticulum (ER) stress.25,26 Such impairment at the cellular level further aggravates metabolic dysregulations.

Hyperinsulinemia–inflammation axis and organelle dysfunction in T2D

In light of this, this review provides a detailed analysis of the intricate connections between immune aging, cellular stress mechanisms, and chronic inflammation, particularly in relation to age-associated metabolic issues such as IR and T2D. It seeks to clarify how age-related declines in immune function and ongoing low-grade inflammation, termed “inflammaging”, interact with organelle stress-especially MD and ER stress to sustain the detrimental cycle of hyperinsulinemia and inflammation. Distinctively, this review takes a fresh perspective by directly examining how cellular stress responses affect the various elements of the “ominous octet” of T2D, which includes impaired insulin secretion, diminished incretin effect, increased lipolysis, heightened hepatic glucose production (HGP), neurotransmitter dysfunction, augmented renal glucose reabsorption, reduced glucose uptake in muscle, and inflammation-driven IR in adipose tissue (AT).27–29 By doing this, a mechanistic framework is established that links immune aging, thymic atrophy, and critical signaling pathways—like NF-κB, c-Jun N-terminal Kinase (JNK), and Phosphoinositide 3-Kinase-Protein Kinase B (PI3K-Akt)—to the disruption of glucose homeostasis. What distinguishes this review from current literature is its thorough examination of how immune aging and organelle stress function as significant factors contributing to the pathological mechanisms of T2D, linking all components of the “ominous octet” associated with T2D. To further emphasize this, as illustrated in Figure 2, the hyperinsulinemia–inflammation axis plays a crucial role in the regulation of metabolic disturbances by compromising the functional integrity of mitochondria and the ER. Dysfunction in mitochondria leads to inefficient energy metabolism, while stress in the ER causes improper protein folding and triggers inflammatory signaling. The dysfunctions occurring in these organelles intersect, amplifying the core pathophysiological mechanisms described in the “Ominous Octet”, thereby promoting the onset and worsening of metabolic diseases. This underscores the interconnected relationship between insulin dysregulation, inflammation, and organelle stress in the development of metabolic disorders such as T2D. Furthermore, the article offers fresh insights and highlights essential aspects that require more in-depth exploration through empirical research as it unfolds.

Organelle dysfunction in insulin resistance (IR) and metabolic disease.
Fig. 2  Organelle dysfunction in insulin resistance (IR) and metabolic disease.

This figure captures the core narrative of this review, illustrating how mitochondrial dysfunction (MD) and endoplasmic reticulum (ER) stress act as central drivers of IR and metabolic disease progression. The reciprocal reinforcement between the dysfunction and hyperinsulinemia-inflammation cycle may ignite a self-propagating loop of metabolic disruption. Additionally, the diagram integrates MD and ER stress within the framework of the “Ominous Octet”, highlighting their pivotal role in metabolic dysregulation. Targeting both IR and its underlying cellular impairments is imperative for developing effective therapeutic interventions. Mitochondria and ER images were adopted from Pixabay.

A unified perspective

Together, the article essentially provides a unified and comprehensive perspective that links immunology, cell biology, and metabolic diseases. This holistic examination not only synthesizes current understanding but also delivers a deeper assessment of potential therapeutic targets designed to reverse or mitigate the immunometabolic deterioration associated with aging, exceeding traditional methods that concentrate solely on glycemic management.

Overview of structure

This review is structured into five sections. The first addresses the dual function of inflammation in metabolic control and aging in T2D. The second explores the interaction among hyperinsulinemia, inflammation, and insulin resistance. The third links these systemic factors to organelle impairment. The fourth examines how organelle stress influences the “ominous octet” associated with T2D. The concluding subsection emphasizes the interaction between mitochondria and the ER as an essential modulator of these pathological pathways.

Inflammation in T2D: A double-edged sword in metabolic regulation and aging

The dual role of inflammation in T2D onset

In the early stages of T2D, our body’s natural immune system, along with pro-inflammatory cytokines like IL-1β and TNF-α, plays a key role in maintaining metabolic balance.24 These immune responses help protect us by clearing out cellular waste and enhancing insulin sensitivity in vital metabolic tissues, such as skeletal muscle and the liver.18,30 Anti-inflammatory macrophages (M2) play a key role by releasing cytokines that improve insulin signaling, boost glucose uptake, and optimize metabolism.31,32 They also play a role in resolving inflammation, preserving tissue integrity, and ensuring ongoing functional capacity.33 As a result, the immune response helps control glucose levels and reduces metabolic strain, delaying IR and β-cell dysfunction.34 The immunometabolic progression of T2D begins with a transient activation of the innate immune system, marked by the release of IL-1β and TNF-α. This early response promotes the recruitment of anti-inflammatory M2 macrophages, which enhance insulin sensitivity and facilitate glucose uptake. In essence, this brings us to the forefront of innovative preventive strategies for mitigating T2D, where pro-inflammatory cytokines, traditionally viewed as harmful, may instead represent therapeutic opportunities when their release is precisely timed and tightly regulated. For example, the CANTOS (Canakinumab Anti-inflammatory Thrombosis Outcomes Study) trial showed that targeting IL-1β with the monoclonal antibody canakinumab decreased systemic inflammation and improved glycemic control in patients with both cardiovascular diseases (CVDs) and T2D, without raising the risk of infections.35 Likewise, Anakinra, which functions as an IL-1 receptor antagonist, improved insulin secretion and reduced HbA1c (hemoglobin A1c (glucose-bound hemoglobin)) levels in T2D patients across various randomized controlled trials, reinforcing the potential for modifying inflammation as a therapeutic approach.36 Hence, understanding the mechanisms that distinguish acute, beneficial inflammation from the chronic inflammation observed in established T2D is critical. Identifying specific cytokines and immune cell subsets involved in these early adaptive responses could guide strategies to fine-tune short-lived inflammation and promote metabolic resilience. Studies on pioglitazone and GLP-1 receptor agonists (GLP-1 RAs) like liraglutide have shown that these medications can indirectly promote M2 macrophage polarization and enhance anti-inflammatory responses, highlighting their dual benefit in enhancing both glycemic management and immunometabolic balance.37–39

Modulating immunity to delay T2D onset

Importantly, enhancing or maintaining M2 macrophage polarization, recognized for its ability to improve insulin signaling and reduce inflammation, through lifestyle changes like diet and exercise or through targeted medications may aid in preventing IR.40,41 Similarly, studying how temporary inflammatory signals make tissues more responsive to insulin, while extended exposure leads to resistance, could uncover biomarkers or signaling pathways essential for the regulation of immune and metabolic functions. Leveraging natural anti-inflammatory processes, such as regulatory T cells (Tregs) and specialized pro-resolving mediators (SPMs), also provides a way to re-establish immune balance and promote metabolic stability.42 In a pioneering phase 1 clinical trial involving humans, the synthetic Resolvin E1 analog RX-10045 demonstrated safety and good tolerance in both healthy participants and individuals with inflammatory eye conditions (NCT01639846), setting the stage for the exploration of SPM analogs in acute systemic inflammatory diseases such as T2D.43 Although trials specific to T2D are still in early development, preclinical studies have shown that SPMs like Resolvin D1 and Maresin 1 can improve insulin sensitivity, reduce the infiltration of macrophages into AT, and restore glucose tolerance in obese mice, indicating they could be valuable as metabolic immunomodulators in the future.44 With clinical-grade analogs approaching readiness, these compounds are set to undergo testing to assess their ability to mitigate chronic inflammation and reestablish immune-metabolic balance in humans. Together, these findings indicate a strategic shift: instead of just suppressing inflammation, therapeutic strategies could aim to modulate it at specific intervals, utilizing the immune system’s inherent flexibility to delay or avert the development of full-blown diabetes.

Inflammation’s shift from protection to progression in T2D

As T2D advances, the balance between the immune system and metabolism begins to break down. The immune system, which was once protective, starts to contribute to disease progression through chronic inflammation—a characteristic feature of the later stages of T2D.19,20,24 At first, inflammation helps manage metabolic stress and regulate glucose levels. However, with ongoing activation of pro-inflammatory pathways, this balance is disrupted, resulting in β-cell dysfunction and IR. A transition in macrophage type—from the anti-inflammatory M2 to the pro-inflammatory M1—intensifies tissue damage and boosts the production of inflammatory cytokines.31,32,45 These cytokines further hinder insulin signaling, accelerate β-cell death, and interfere with insulin release.46 Consequently, inflammation transforms from a protective response into a catalyst for metabolic impairment, highlighting its complex role in the progression of the disease. When inflammation becomes persistent or misregulated, it not only harms tissues but also undermines immune regulation, speeding up the deterioration of insulin function and exacerbating the effects of T2D.47,48 Recognizing this shift in immunometabolism opens up promising avenues for treatment.19 Employing approaches that focus on inflammation at optimal times, such as modifying macrophage activity or inhibiting specific cytokines, could safeguard β-cell well-being and improve insulin sensitivity.49,50 Tailored methods that correspond with a person’s inflammatory profile could enhance treatment effectiveness and decelerate disease advancement, signifying a significant shift in the management of T2D.51,52 In the later stages, hyperinsulinemia and aging immune cells propagate chronic inflammation through the SASP, leading to β-cell loss and worsening insulin resistance. Therapeutic strategies that target the SASP, restore M2 macrophages, and enhance β-cell survival may help halt the advancement of the disease.53,54

Senescence, inflammation, and β-cell dysfunction in T2D

Moreover, chronic hyperinsulinemia in T2D accelerates the aging process through intricate immune responses.18,55 During the early stages of T2D, elevated insulin levels, or hyperinsulinemia, attract senescent immune cells, such as aging macrophages, to the pancreatic islets. The sustained inflammatory environment, along with alterations in pro-inflammatory cytokines and reactive oxygen species (ROS) linked to the SASP in these senescent cells, triggers apoptosis of β cells, impairing their function and leading to reduced insulin production in the pancreas.56,57 As these senescent immune cells accumulate, they worsen inflammatory conditions, creating a self-perpetuating cycle that heightens IR and speeds up the decline of β-cells.58,59 Ultimately, the reduction in β-cell mass and the dysfunction in insulin signaling contribute to the ongoing persistence and advancement of the disease.60,61 Strategies for eliminating or diminishing senescent cells, such as senolytics, have demonstrated potential in early-stage studies and may be especially beneficial for managing the chronic inflammation associated with T2D.62,63 These senolytic drugs could be combined with immunomodulatory therapies that aim to counteract the detrimental immune response to hyperinsulinemia, such as those that affect macrophage polarization, to restore immune equilibrium and safeguard β-cell functionality.64,65 In the end, a comprehensive approach that simultaneously targets insulin sensitizers, immune-modulating therapies, and senolytics might disrupt the detrimental cycle of inflammation and β-cell impairment. This approach presents a possibility for a treatment strategy to reduce or possibly reverse the advancement of T2D. By addressing the immune-metabolic components of the disease, these findings could lead to an innovative approach in the prevention and treatment of T2D. For instance, a notable clinical trial (NCT02848131) involving the senolytic duo of dasatinib and quercetin in individuals with diabetic kidney disease, a common complication of T2D, led to reductions in senescence markers and inflammatory mediator levels, suggesting potential systemic health advantages of eliminating senescent cells in metabolic conditions.66 These senolytic treatments may also be paired with immunomodulatory therapies aimed at mitigating the detrimental immune response associated with hyperinsulinemia, including those that focus on altering macrophage polarization to restore immune equilibrium and safeguard β-cell function.

Preserving β-cell function: A key to T2D management

In essence, the interaction between immune cells and β-cell health is essential for comprehending the pathophysiology of T2D. As the mass of β-cells declines, insulin production reduces, exacerbating hyperinsulinemia and disrupting glucose control.67 This cycle emphasizes the importance of understanding how immune-mediated inflammation impacts β-cell function in the progression of T2D. Strategies that disrupt this cycle could help maintain β-cell health, enhance insulin sensitivity, and slow the progression of the disease, presenting potential therapeutic approaches for better management of T2D.68

The interplay of hyperinsulinemia, inflammation, and IR

The rising prevalence of ischemic heart disease during aging is closely linked to the growing incidences of T2D, CVDs, and metabolic syndrome (MS), underscoring the critical role that metabolic dysfunction plays in cardiovascular health.69,70 A critical factor driving the pathophysiology of IR is the interaction between hyperinsulinemia and systemic inflammation, which has gathered lots of attention in recent years. Ischemic heart disease shows increased incidence with rising prevalence of IR.71 Generally, this condition develops as a compensatory response to restore the glucose homeostatic effect by increasing the release of insulin when receptor sensitivity is decreased because of associated conditions or the overproduction of insulin by the substrates. This may initially be adaptive, but over time can damage the ability to regulate normal glucose and activate inflammatory pathways of metabolic dysfunction that further complicate IR. These molecules impair the signaling of insulin by promoting serine phosphorylation of insulin receptor substrates (IRS), thereby obstructing glucose uptake in the peripheral tissues.72 Obesity-induced metabolic disruption further amplifies this effect, as expanded AT releases inflammatory cytokines such as TNF-α and IL-6, along with adipokines like leptin and resistin.73,74 This leads to hyperglycemia by stimulating liver glucose production through increased gluconeogenesis and glycogenolysis. In the liver, inflammation-driven dysregulation of insulin signaling within hepatocytes results from the increased HGP through the enhancement of gluconeogenesis and glycogenolysis, thus adding to hyperglycemia.75,76 Similarly, local inflammatory processes in skeletal muscle decrease the activity of insulin receptor signaling, while inflammation in pancreatic islets restricts the secretion of insulin, therefore maintaining hyperglycemia.77,78 Hyperinsulinemia produces inflammation, which causes conditions that promote hyperinsulinemia, sustaining an evil cycle that leads to an accelerated progression of IR and T2D.79 This bidirectional feedback mechanism illustrates how hyperinsulinemia intensifies inflammation, which subsequently worsens IR. Increased insulin levels trigger inflammatory pathways such as NF-κB, JNK, and Suppressor of Cytokine Signaling 3, resulting in heightened serine phosphorylation of IRS-1, thereby disrupting insulin signaling and exacerbating IR.80,81 This feedback mechanism enhances inflammation, boosts HGP, and reduces peripheral glucose uptake, perpetuating a sustained state of hyperglycemia that further raises insulin levels. Moreover, chronic inflammation stimulated by pro-inflammatory cytokines leads to tissue remodeling, enlargement of adipocytes, and infiltration of immune cells, thereby aggravating metabolic dysfunction. Over time, this cycle hastens the development of T2D and MS. Consequently, the interaction between inflammation and hyperinsulinemia underscores the significant role of signaling pathways such as NF-κB, JNK, and PI3K-Akt in the onset of IR and T2D, presenting potential therapeutic avenues to break this cycle and enhance metabolic health.82 For instance, a double-blind, randomized clinical trial assessed the effects of metformin on inflammatory mediators in obese adolescents with IR.83 After 3 months of metformin treatment, there was a significant reduction in serum levels of TNF-α and stabilization of adiponectin levels, suggesting improved inflammatory activity and potentially cardiovascular implications. Additionally, in a randomized, double-blind, placebo-controlled study, subjects with MS received treatment with galantamine. The findings indicated that galantamine, an acetylcholinesterase inhibitor, significantly lowered plasma levels of proinflammatory mediators (TNF-α and leptin) while increasing anti-inflammatory markers (adiponectin and IL-10). There was also a noted improvement in measurements of IR, including plasma insulin and Homeostatic Model Assessment-IR values. Moreover, a randomized controlled trial assessed the impact of oleoylethanolamide, a naturally occurring lipid (fat-like molecule) produced in the small intestine, especially after eating, supplementation in individuals diagnosed with prediabetes. The findings indicated that oleoylethanolamide intake improved glycemic control, reduced IR, and diminished inflammation by modulating oxidative stress and the release of inflammatory cytokines. Also, in a randomized controlled trial, participants with acute hypertriglyceridemia and IR who received bezafibrate—a medication used to lower lipid levels and activate peroxisome proliferator-activated receptor alpha—experienced significant decreases in inflammatory markers like C-reactive protein (CRP) and interleukin-6, along with enhanced insulin sensitivity.84 Finally, the Whitehall II study, involving over 7,600 non-diabetic participants, found a positive link between elevated levels of inflammatory biomarkers (high sensitivity CRP and IL-6) and higher fasting insulin and IR over a 5-year follow-up.85 Conversely, greater levels of adiponectin were correlated with improved glycemic control and enhanced insulin sensitivity.

Importantly, recognizing individuals who are more susceptible to T2D through biomarkers linked to dysregulated signaling pathways like NF-κB, JNK, and PI3K-Akt presents a promising opportunity for early intervention. Instead of solely depending on traditional risk factors such as body mass index and fasting glucose levels, incorporating molecular markers that reflect inflammation and insulin signaling problems leads to a better categorization of patients.86,87 This approach facilitates a shift from general preventive strategies to specific, mechanism-driven interventions aimed at disrupting established pathogenic processes before significant metabolic damage occurring. For example, elevated levels of CRP, TNF-α, and IL-6 suggest active involvement of the NF-κB pathway and are increasingly recognized as early markers of inflammation related to IR.88 Similarly, the detection of serine-phosphorylated IRS-1 in muscle tissues and a reduction in Akt phosphorylation in peripheral tissues serve as biological indicators of disrupted insulin signaling via the PI3K-Akt pathway.89 With this knowledge, current clinical initiatives, such as the Diabetes Prevention Program and extensive studies like the UK Biobank, are beginning to integrate these biomarkers into their risk assessment frameworks to enhance the early identification of T2D risk.90 This emphasis on pathway-oriented risk profiling signifies a significant transition towards predictive, preventive, and personalized medicine for metabolic disorders, allowing healthcare professionals to respond more swiftly, customize treatments more efficiently, and alleviate the worldwide impact of T2D, especially in the aging demographic.

Connecting hyperinsulinemia, inflammation, and organelle dysfunction in T2D

At the cellular level, organelle dysfunction, particularly in mitochondria and the ER, serves as a significant mechanistic link between metabolic disturbances and inflammation in relation to aging.91,92 MD, which reduces oxidative phosphorylation (OXOPHOS) and leads to ROS buildup, causes cellular stress that raises inflammation.93,94 ER stress occurs when the ER, an essential cellular component that ensures proper protein folding and processing, becomes overloaded with an excess of unfolded or incorrectly folded proteins. In response, the cell activates the unfolded protein response (UPR), a sophisticated, adaptive signaling system aimed at reestablishing cellular equilibrium.95,96 This is achieved by reducing the overall protein load, enhancing protein-folding capacity, and promoting the degradation of misfolded proteins. The UPR operates primarily through three principal ER stress sensors: inositol-requiring enzyme 1 (IRE1), protein kinase RNA-like ER kinase, and activating transcription factor 6.97 These sensors initiate signaling pathways that alter gene expression and elicit protective measures to mitigate the stress. However, if ER stress persists or remains unresolved, continuous UPR activation can become harmful. In particular, it can interfere with insulin signaling pathways, potentially leading to IR and disrupting metabolic homeostasis.98 These organelles play an important role in keeping the cell in homeostasis; therefore, any dysfunction only creates metabolic-inflammation imbalances that spark IR onset. Pro-inflammatory cytokines, adipokines, and immune cells play key roles in driving these dysregulations, creating a harmful feedback loop.99,100 Investigating the relationships between hyperinsulinemia, chronic low-grade inflammation, and organelle dysfunction opens up promising possibilities for cutting-edge treatments that could prevent or possibly reverse IR before it develops into T2D or causes CVDs. Significantly, this cellular-level dysfunction contributes to a larger systemic context in which metabolic stress, immune reactions, and hormonal imbalances interact in an ongoing cycle.101 These findings are particularly crucial regarding the ominous octet theory. Organelle dysfunction can act as a unifying factor that exacerbates multiple pathways, either by directly impairing tissue function or by heightening systemic inflammation and IR.

To successfully break this harmful cycle, further investigation is needed to better understand the specific impact of organelle dysfunction in different tissues and how its timing is associated with metabolic issues. This includes examining innovative treatment strategies that focus on the health of mitochondria and the ER, such as promoting mitophagy, enhancing protein-folding capacity, and minimizing ROS build-up.102,103 Acknowledging the differences in these processes across diverse populations and stages of the disease could aid in developing more personalized methods for managing T2D.

Hence, the following sections offer a concise overview of recent research on organelle dysfunction, particularly in the mitochondria and ER, and its role in driving the development of IR. By examining how dysfunction in these organelles influences the key components of the “ominous octet”, we can gain a deeper understanding of the underlying mechanisms of T2D. This knowledge is crucial to furthering the progress of the development of targeted interventions beyond glucose-lowering and addressing the underlying causes of metabolic and cardiovascular complications in T2D. Such strategies have the potential for more effective, disease-modifying therapy.

Organelle dysfunction in T2D: the impact on the ominous octet pathways

The “ominous octet” in T2D highlights how dysfunction in pancreatic β-cells, liver, muscle, AT, GI tract, pancreatic α-cells, kidneys, and the central nervous system (CNS) contributes to hyperglycemia and metabolic dysregulation.24,27 To expand on this further, the model details the various factors that contribute to hyperglycemia in T2D. Impaired glucose regulation mainly results from disruptions in the functioning of several interconnected organ systems. Key roles in the “Ominous Octet” concept are played by the liver, pancreatic β-cells, and skeletal muscles, as they lead to IR, reduced insulin secretion, and alterations in glucose utilization. While the primary underlying factors are essential, additional elements such as pancreatic α-cells, AT, the kidneys, the digestive system, and the brain also worsen this metabolic imbalance. These systems engage in a detrimental feedback loop that amplifies hyperglycemia and accelerates the advancement of T2D. In essence, this model highlights the interconnectedness of these organ systems and underscores the importance of a comprehensive strategy for improving T2D management and slowing the advancement of the disease. These pathways are primarily driven by reduced insulin sensitivity and decreased insulin secretion. This section provides a brief overview of the individual elements of the Ominous Octet while adhering to the journal’s constraints. The main focus will be on how these factors interact, with a particular emphasis on how MD and ER stress link hyperinsulinemia and inflammation in the context of the ominous octet of T2D.

Decreased insulin secretion

According to the ominous octet of T2D, dysfunctional β-cells reduce insulin secretion.18,19,22,23 This worsens with lipotoxicity and amyloid deposits, along with the body’s resistance to normal blood glucose regulation, leading to hyperglycemia. As we grow older, advanced glycation end products (AGEs) accumulate due to prolonged exposure to high blood sugar levels and oxidative stress.104 AGEs are created when sugars non-enzymatically attach to proteins, lipids, or nucleic acids and interact with Receptor for Advanced Glycation End Products (RAGE), a receptor found on multiple cell types, including endothelial cells, immune cells, and pancreatic β-cells.105 The interaction between AGEs and RAGE triggers inflammation and oxidative stress, which significantly disrupts insulin signaling and raises IR in tissues such as the liver, muscle, and AT.106 In the aging process, this mechanism hastens the deterioration of pancreatic β-cells, resulting in decreased insulin secretion.107 The activation of RAGE triggers an inflammatory response that leads to vascular damage, disrupts blood flow, and further reduces insulin sensitivity.108 This creates a persistent cycle of glucolipotoxicity, where high levels of glucose and fatty acids cause additional damage to β-cells, exacerbating the decrease in insulin production and secretion.109 Over time, the body’s capacity to eliminate AGEs declines, worsening the situation and creating a glycotoxic environment.110 This not only hinders effective glucose regulation but also raises the risk of T2D-related complications such as CVDs and neuropathy, which further diminishes insulin secretion as people age.111 One of the primary effects of RAGE activation is the stimulation of the NOD-like Receptor Pyrin Domain Containing 3 (NLRP3) inflammasome.112 This inflammasome is a multi-subunit protein complex that is essential for the innate immune response, as it activates caspase-1 and facilitates the secretion of pro-inflammatory cytokines IL-1β and IL-18.113 RAGE-induced inflammasome activation contributes to chronic inflammation, which is often observed in aging and several age-associated conditions, like T2D.114 The increased levels of cytokines lead to IR and damage to pancreatic β-cells, exacerbating metabolic disorders. The activation of inflammasomes through RAGE amplifies the detrimental effects of AGEs, leading to increased inflammation and metabolic issues, especially in older adults.115 These interrelated factors contribute to the intricate nature of disease mechanisms. Continuous high blood glucose levels and persistent inflammation gradually result in IR and T2D, which, if left untreated, can lead to various complications over time. This underscores the critical need for treatments that address both chronic inflammation and hyperglycemia to manage and prevent disease progression.116 Additionally, MD impairs adenosine triphosphate (ATP) synthesis, reducing ATP levels and limiting insulin secretion.117 Excess stress from misfolded proteins like amyloids in the ER causes β-cell apoptosis, reducing insulin production.60,61 Such deposits further induce the ER stress due to misfolded proteins.118 An excess of free fatty acids (FFAs), referred to as lipotoxicity, disrupts the functionality of pancreatic β-cells due to the accumulation of FFAs in their mitochondria.119 This accumulation negatively impacts mitochondrial performance, resulting in increased oxidative stress. The elevated oxidative stress damages β-cell integrity and hinders their ability to efficiently release insulin.120 Consequently, the buildup of FFAs leads to β-cell dysfunction, which is a significant factor in the development of IR and T2D.121

Enhanced gluconeogenesis

Gluconeogenesis mainly affects the liver’s ability to generate glucose from non-carbohydrate materials, hence playing an important part in the regulation of metabolism.122 While this mechanism is essential during fasting or strenuous exercise, an exaggerated form of gluconeogenesis is a serious issue in some metabolic disorders such as T2D, together with IR and hyperinsulinemia. While high insulin is expected to lower blood glucose levels, it counterintuitively enhances gluconeogenesis by stimulating pathways that activate key enzymes like phosphoenolpyruvate carboxykinase (PEPCK) and glucose-6-phosphatase in the liver, impairing glucose tolerance.123 This process occurs alongside inflammation, which is commonly observed in obesity and metabolic syndrome, and together these factors hinder the insulin signaling pathway in the liver, worsening gluconeogenesis.124 These interrelations create a negative feedback loop associated with excess glucose, and blood glucose levels appear to rise even higher, increasing the body’s IR. Chronic inflammatory changes will also lead to metabolic derangements of lipids and an increased tendency toward nonalcoholic fatty liver disease (NAFLD).125 The link between increased gluconeogenesis and inflammation allows one to unravel some of the mechanisms underlying metabolic dysfunction, emphasizing the importance of integrated strategies that combine lifestyle changes with therapies that improve insulin sensitivity and reduce inflammation, breaking harmful cycles and promoting long-term health. Moreover, hepatic gluconeogenesis is curtailed by MD in hepatocytes, and unregulated gluconeogenesis leads to excessive glucose production.126 In addition, FFAs overload the peroxisomes involved in fatty acid oxidation, causing increased gluconeogenesis and oxidative stress due to their inability to handle the excess.127 Disruption of mitochondrial OXOPHOS generates increased ROS, thereby worsening IR in the liver and enhancing glucose production.128

Importantly, recent research has shifted focus toward how circadian rhythms and neuroendocrine signaling influence the regulation of gluconeogenesis. The liver’s gluconeogenic activity is closely governed by the circadian clock, and disruptions, like those caused by poor sleep or shift work, can lead to the desynchronization of metabolic cycles, which exacerbates IR and disrupts glucose regulation.129 Furthermore, the hypothalamus’s regulation through agouti-related peptide (AgRP)/ neuropeptide Y (NPY) neurons, alongside input from the sympathetic nervous system and vagal signals, plays a crucial role in managing hepatic glucose synthesis.130 These interconnected systems underscore the importance of communication between the gut, brain, and liver in maintaining bodily balance, suggesting that aligning these regulatory mechanisms may be beneficial in preventing abnormal gluconeogenesis, particularly in the context of T2D.131

One major obstacle in targeting gluconeogenesis for the treatment of T2D is the current restrictions of available therapies. Metformin, the primary drug used to suppress HGP, shows inconsistent results across different patients and carries risks of side effects, such as lactic acidosis.132 Similarly, while sodium-glucose cotransporter 2 (SGLT2) inhibitors boost the elimination of glucose through urine, they also unintentionally trigger the formation of ketone bodies and prompt hepatic gluconeogenesis as a side effect.133 These challenges highlight the necessity for more comprehensive treatment strategies. The combination of agents that focus on liver metabolism with those that improve peripheral insulin sensitivity and modulate gut hormones, like GLP-1 RAs or gastric inhibitory polypeptide (GIP) co-agonists, may offer a more effective approach to address gluconeogenic abnormalities in T2D.134,135

Increased lipolysis

Heightened lipolytic activity results in increased amounts of FFAs, which play a critical role in the development of T2D by disrupting insulin signaling and damaging pancreatic β-cells. Elevated FFA levels impede insulin signaling in muscle and liver cells, resulting in a reduced capacity for glucose uptake from the bloodstream, particularly in cases of obesity.136 FFAs also cause lipotoxic damage to β-cells, which triggers cell death and reduces insulin secretion. As a result, this creates a situation referred to as glucolipotoxicity, where increased glucose levels further intensify the harmful effects of FFAs, leading to increased oxidative stress and inflammation in β-cells.137 Research involving both human and animal subjects has identified a robust link between elevated FFA levels and IR, as well as impaired β-cell functionality.138,139 To further elaborate, animal models have provided valuable insights into the mechanisms by which elevated FFAs contribute to IR, including the roles of inflammation, oxidative stress, and dysregulation in lipid metabolism. These findings illuminate the cellular and molecular processes that contribute to IR in humans. On the other hand, clinical studies in humans have demonstrated similar relationships between increased FFA levels and reduced insulin sensitivity along with β-cell dysfunction, reinforcing the relevance of animal model findings to human physiology.140,141 Although human studies are crucial for understanding the clinical significance of these results, animal research yields essential mechanistic insights. Collectively, these investigations bridge the gap between fundamental science and clinical applications, highlighting the potential of targeting elevated FFAs as a therapeutic strategy for addressing IR and β-cell dysfunction in humans.142 A thorough discussion of many of these studies is beyond the scope of this article. Importantly, weight loss interventions aimed at decreasing FFA levels can improve insulin sensitivity and β-cell function, highlighting the crucial role of FFAs in the advancement of T2D. Furthermore, metabolic disorders in AT boost FFA levels, leading to their accumulation in the bloodstream, which can negatively impact other tissues such as muscle and liver.143 Higher FFA concentrations disrupt mitochondrial functioning in β-cells and muscle cells, contributing to oxidative stress that further aggravates IR and diminishes glucose uptake.144

Association between inflammation and lipid metabolism

The connection between lipid metabolism and inflammation plays a vital role in the onset of T2D.145 Elevated FFAs not only hinder insulin signaling and damage β-cells, but they also function as significant contributors to inflammation.146 FFAs initiate inflammatory pathways, resulting in a higher release of pro-inflammatory cytokines like TNF-α and IL-6. These cytokines further disrupt insulin signaling, establishing a cycle of ongoing low-grade inflammation that heightens IR and affects glucose metabolism. This inflammatory response exacerbates the metabolic issues triggered by increased lipolysis and is implicated in the development of T2D.124 Additionally, the interplay between inflammation and altered lipid metabolism leads to MD, ER stress, and further harm to tissues, particularly in muscle and liver cells, thereby worsening the progression of T2D.147,148

For example, NAFLD has become a major health concern and is on the rise globally. Once thought to be a benign condition, NAFLD can escalate to non-alcoholic steatohepatitis (NASH), a more serious stage of the disease marked by inflammation, hepatocyte damage, and fibrosis.149 While NAFLD is largely associated with metabolic disorders, such as IR, obesity, and dyslipidemia, immune system dysfunction becomes particularly important, especially as the disease progresses to NASH.150 In individuals with T2D, the inflammatory processes that contribute to IR and metabolic disturbances play a key role in the onset of NAFLD.151 As IR in AT increases, there is a recruitment of macrophages along with the release of cytokines. AT changes into an inflammatory organ, secreting pro-inflammatory cytokines like TNF-α, IL-6, and monocyte chemoattractant protein-1. These cytokines induce inflammation in various tissues, including the liver, leading to fat accumulation (steatosis) and progression to NASH.152 The rise in FFAs, resulting from enhanced lipolysis in obesity and IR, significantly aids in the development of NAFLD. FFAs activate Toll-like receptors (TLRs) on immune cells, initiating pro-inflammatory signaling pathways such as NF-κB and inflammasomes, which not only exacerbate liver injury but also increase IR, creating a detrimental cycle that exacerbates the disease progression.153 As NAFLD evolves into NASH, hepatic macrophages (Kupffer cells) become activated and release cytokines that attract more immune cells, resulting in hepatocyte apoptosis, inflammation, and fibrosis.154 The combination of metabolic issues and persistent inflammation eventually leads to liver cirrhosis and promotes the development of liver cancer, both of which are serious outcomes of NAFLD and T2D.155 In addition to their role in inflammation, immune cells in AT also have functions related to lipid metabolism that are not solely immune-related. These functions include balancing lipogenesis, which is the production of fatty acids and triglycerides, and lipolysis, the breakdown of stored fats, as well as managing how adipocytes react to changes in nutrient levels and body temperature.156 For example, immune cells contribute to the adaptive thermogenesis process in brown adipose tissue, where lipid oxidation initiates heat production in response to cold exposure.157 This highlights the complex interplay between the immune system, lipid metabolism, and energy expenditure. Notably, natural polysaccharides improve lipid and energy metabolism while alleviating inflammation by decreasing MD and ER stress.158 Given that immunity also impacts lipid and energy metabolism, natural polysaccharides facilitate these processes by modifying host immunity.159 This strengthens the positive reciprocal relationship between inflammation and lipid metabolism, as they can influence each other and exacerbate conditions, thereby contributing to the onset and advancement of metabolic disorders.

Nevertheless, the relationship between immune and metabolic functions presents a therapeutic challenge, as lowering inflammation might inadvertently disrupt vital metabolic processes. This intricacy necessitates that the immune-metabolic interactions related to pathology and homeostasis are differentiated at the molecular level. For instance, focusing on crucial immunometabolic points—like the states of macrophage polarization, thresholds of TLR signaling, or the function of adipose-resident Tregs—could provide chances to restore metabolic balance without compromising necessary immune responses.160,161 Such insights could pave the way for more precise and effective treatments for T2D, addressing both the metabolic and inflammatory components of the condition.

Decreased glucose uptake in muscle

Diminished glucose uptake in muscle tissues, in particular, initiates the development of IR.162 Muscle uptake of glucose from the bloodstream is essential; its derangement raises blood glucose to a degree that causes metabolism to run amok. The IR blocks glucose transporter type 4 (GLUT4) translocation to the plasma membrane, a prerequisite for glucose uptake, disrupting an insulin-mediated signaling pathway. The defect is aggravated in most cases by obesity.163 This prolonged inflammation due to excess AT further disrupts insulin signaling in a damaging cycle, thereby advancing IR.164 Excessive FFAs engender lipotoxicity and oxidative stress in muscle tissues, contributing to IR.165 MD, accompanied by development towards a higher ratio of insulin-resistant fast-twitch muscle fibers, accounts for inadequate glucose transport and consequent metabolic afflictions.166 MD in muscle cells would impair energy production and would thus, in turn, impede glucose utilization by muscle and promote IR. In addition, increased ER stress would inhibit the translocation of GLUT4 to the plasma membrane for insulin-mediated glucose uptake by muscle, thus further compromising insulin signaling.167 This information is particularly crucial for understanding the link between T2D and immune aging, as age-associated MD, increased ER stress, and chronic low-grade inflammation significantly enhance IR and hinder glucose metabolism, thereby influencing the pathogenesis and advancement of T2D.

In this regard, one significant challenge in utilizing GLP-1RAs for managing T2D is their potential to aggravate sarcopenia (age-related loss of muscle mass, strength, and function) through negative impacts on muscle functionality and metabolism.168 Although GLP-1RAs are very effective at improving insulin sensitivity and controlling blood glucose levels, their influence on muscle tissue may lead to unintended adverse consequences, particularly in vulnerable populations. This presents a crucial issue, as the long-term implications of GLP-1RAs on muscle health remain inadequately studied. Specifically, it is essential to elucidate how these medications influence mitochondrial function, ER stress, and muscle fiber structure. Addressing this knowledge deficiency is crucial for developing strategies that prevent or diminish muscle-related side effects. Identifying strategies to mitigate muscle-related side effects while optimizing the metabolic advantages of GLP-1RAs will be critical to ensuring their efficacy in T2D management without compromising muscle integrity.

Increased glucagon secretion

In T2D, pancreatic α-cells produce glucagon improperly, leading to elevated blood glucose levels. Typically, glucagon is secreted when blood glucose falls below a specific threshold, serving to stimulate glucose production in the liver. In T2D, excessive release of glucagon therefore worsens hyperglycemia.169 IR ameliorates the inhibitory effect of insulin on glucagon production, particularly at high levels of insulin; thus, glucagon levels result in elevated levels despite hyperinsulinemia.50 Chronic hyperinsulinemia further affects the signaling pathways controlling α- and β-cell function, promoting excessive glucagon secretion.170 This imbalance causes increased HGP, thereby further worsening hyperglycemia. All of these contribute to complications of T2D, for example, comorbidity with CVDs and neuropathy. ER stress disrupts the normal secretion of glucagon in pancreatic α-cells.171 The pathways triggered by ER stress led to the activation of the UPR response, which might influence the secretion of certain hormones, including glucagon. MD in α-cells results in inefficient energy production, consequently causing inappropriate glucagon secretion, aggravating IR in the liver, and increasing the production of glucose.172 These activities are further exacerbated by aging, as age-related declines in mitochondrial function and increased ER stress can impair α-cell function, intensifying the metabolic imbalances that contribute to the onset of T2D.

An innovative approach to improve the treatment for T2D involves investigating the transdifferentiation of alpha (α) cells into beta (β) cells, which could help reduce excessive glucagon secretion and enhance metabolic control.173 By transforming α-cells into insulin-producing β-cells, it might be feasible to lessen abnormal glucagon levels while boosting insulin production. Nevertheless, this strategy faces considerable challenges, particularly in achieving a proper balance between insulin release and glucagon suppression. Excessive insulin production may lead to hypoglycemia, while improperly functioning transdifferentiated β-cells could contribute to β-cell failure and exacerbate metabolic problems. Moreover, it is essential to tackle the underlying factors of cellular stress, including MD and ER stress, to ensure that the newly created β-cells operate effectively and prevent a resurgence of T2D-related issues. To progress in this field of study, research efforts need to concentrate on the molecular mechanisms that regulate α-cell reprogramming and methods to maintain the functionality of transdifferentiated β-cells over time.174 By clarifying the intricate relationships between glucagon, insulin, and cellular stress, we can develop more precise and effective methods to manage or potentially reverse the advancement of T2D.

Increased glucose reabsorption in the kidneys

In conditions of hyperinsulinemia and IR, the kidneys increase glucose reabsorption.175 Insulin causes sodium reabsorption in the proximal tubules, leading to glucose reabsorption through SGLTs and high blood glucose levels even after minor plasma glucose increases.176 Chronic low-grade inflammation, commonly observed in obesity and metabolic syndrome, worsens this condition as cytokines such as TNF-α and IL-6 disrupt insulin signaling, thereby increasing IR and kidney dysfunction.177 These processes induce a vicious cycle or feedback loop that elevates the risk for T2D. Thus, exploring the connection between hyperinsulinemia and inflammation is crucial to addressing downstream metabolic disturbances, including organelle dysfunction like MD and ER stress. Moreover, MD and ER stress within renal tubular cells hinder SGLT function, leading to impaired glucose reabsorption and worsening hyperglycemia.178,179 This emphasizes the importance of addressing organelle dysfunction to avert subsequent metabolic disruptions.

However, SGLT2 inhibitors increase glucose elimination in the urine, leading to fluid loss.180 This may cause dehydration and low blood pressure, particularly in older adults or those with impaired kidney function. The diuretic effect of SGLT2 inhibitors can exacerbate dehydration, potentially resulting in drops in blood pressure, dizziness, and, in severe cases, fainting. This situation poses a significant challenge for patients on antihypertensive medications, as their combined effects may heighten the risk of dehydration and electrolyte imbalances.181 To address these issues, certain pharmacological agents that target the UPR might help reduce ER stress by supporting normal protein folding and functionality. Tauroursodeoxycholic acid and 4-phenylbutyrate have demonstrated efficacy in alleviating ER stress and boosting insulin sensitivity.182 When used alongside SGLT2 inhibitors, these agents could help lessen some undesirable effects of the medication, contributing to a more balanced therapeutic approach.

Decreased incretin effect

Diminished incretin effect hastens the progression of T2D, which results from reduced secretion of GLP-1 and GIP, diminished receptor sensitivity within pancreatic β-cells, and altered dipeptidyl peptidase-4 activity, an enzyme that degrades incretin hormones.183 This leads to, in turn, insufficient insulin secretion, postprandial hyperglycemia, dysfunctional glucagon secretion, and eventually increased appetite, which typically worsens IR together with increased weight gain.184 Chronic hyperinsulinemia and inflammation further impair incretin action by blocking insulin signaling and secretion of incretin hormones.185 Pro-inflammatory cytokines secreted from the AT result in worsening β-cell dysfunction so that the insulinotropic effect fades gradually and converts into a cycle of inflammation, IR, and hyperglycemia.164 Enteroendocrine, GLP-1, and GIP-producing cells manifest ER stress; the increased production of insulinotropic incretin hormones, together with low glucose control.186 Chronic inflammation and oxidative stress can cause MD in β-cells, reducing their ability to respond to incretin hormones.187 This leads to impaired insulin production and hyperglycemia.

Moreover, emerging findings seem to indicate the possibility that IR may play a role in reducing the weight-loss effectiveness of GLP-1 RAs in individuals with T2D.188 While some individuals with obesity may retain insulin sensitivity, those diagnosed with T2D usually exhibit increased IR, which can disrupt GLP-1 receptor function, particularly within the brain, potentially due to unidentified desensitization mechanisms. Additionally, failures in mitochondrial function, ER stress, and the interplay between hyperinsulinemia and inflammation contribute to β-cell dysfunction and hinder incretin activity. Taken together, these factors may clarify why the weight-loss effects of GLP-1 RAs are diminished in people with diabetes, highlighting the necessity of comprehending the complex interactions between metabolic and inflammatory pathways to enhance treatment outcomes.

Neurotransmitter dysfunction

Neurotransmitter dysfunction is regarded as another essential component of the ominous octet in T2D, dysregulated dopaminergic and serotonergic signaling, and dysregulation of appetite control and reward pathways, leading to unhealthy eating habits and a gross increase in caloric intake.189 Alteration in dopaminergic function idealizes cravings and promotes the risk of obesity development; on the other hand, imbalance in serotonin reduces satiety and encourages emotional eating.190 Chronic low-grade inflammation also undermines neurotransmitter action, further aggravating it and metabolic dysregulation.191 Through increased oxidative stress and inflammation, MD and ER stress interfere with glucose homeostasis and worsen neurotransmitter dysregulation.192 These systemic interactions may aid in the effective and long-term management of T2D, including hyperglycemia, IR, inflammation, and organelle health. MD interferes with energy metabolism in neurons, affecting neurotransmitter function, especially dopamine and serotonin, which are important for regulating appetite and eating behaviors. With advancing age, MD becomes more common, resulting in compromised neurotransmitter management.193 Furthermore, ER stress in the brain intensifies this dysregulation, affecting reward pathways and further disrupting appetite regulation.194 This ongoing cycle exacerbates challenges such as IR and obesity, both prevalent in the cognitive decline associated with aging. Ultimately, these metabolic issues and dysfunctions in organelles may adversely affect cognitive capacities, thus complicating the development of neurodegenerative diseases and age-related disorders.195,196

The neuroimmune-metabolic axis in T2D

Building on this foundation, it is evident that an imbalance in neurotransmitters is fundamentally connected to the larger network of immune aging, systemic metabolic dysfunction, and neurological deterioration in T2D. Central to this pathological relationship are mechanisms like neuroinflammation, cellular senescence, glial dysfunction, and the infiltration of peripheral immune cells into the CNS, all of which are influenced by both aging and hyperglycemia. These processes interact to disrupt the equilibrium between neurons and glial cells, leading to a range of cognitive impairments, diminished synaptic plasticity, and heightened susceptibility to neurodegenerative conditions.

Neuronal changes in these conditions are characterized by a decrease in synaptic plasticity, especially the reduction of long-term potentiation in hippocampal neurons, which obstructs memory formation and the capacity to learn.197 At the same time, neurogenesis is significantly reduced, limiting the brain’s flexibility and its ability to reorganize when faced with metabolic stress. AGEs, which are often elevated in T2D and during aging, disrupt neuronal metabolism, hinder axonal transport, and contribute to ongoing neurodegeneration.198,199 The dysregulation of neuroglial cells is also notably important. In the brains of individuals with diabetes and aging, microglia adopt a pro-inflammatory M1 phenotype, which is marked by the secretion of cytokines such as IL-1β and TNF-α, leading to neuroinflammation and damage.200 Astrocytes undergo reactive astrogliosis, compromising their ability to regulate the extracellular environment, support synaptic function, and maintain the balance of neurotransmitters.201 This disturbance in neuroglial cells disrupts essential interactions between neurons and glia, impairs synaptic transmission, weakens the integrity of the blood-brain barrier (BBB), and reduces neurovascular coupling.202

The detrimental effects are further aggravated by the active interaction between the CNS and the immune system outside of the brain. Ongoing peripheral inflammation, originating from organs with metabolic imbalances, such as the liver and AT, results in increased levels of systemic cytokines like IL-6 and TNF-α.203,204 These cytokines can cross a compromised BBB, activating glial cells and initiating neuroinflammatory pathways. Furthermore, the breach of the BBB permits peripheral monocytes to infiltrate the brain, where they become inflammatory macrophages, intensifying neuroimmune responses.205 The vagus nerve is vital for this interaction, relaying immune signals from the gut and other organs to the brainstem while also regulating microglial function in response to peripheral metabolic signals.206 Ultimately, the aging of the blood cell-producing system leads to a systemic environment that favors inflammation. Age-related transformations in the properties of immune cells derived from bone marrow, such as monocytes and macrophages, lead to the secretion of inflammatory cytokines that affect glial activity and the brain’s immune landscape.207,208

Furthermore, the widespread effects of cellular senescence exacerbate both systemic and central dysfunction.209 Within the CNS, aging astrocytes and microglia sustain a chronic inflammatory condition, hinder the clearance of neurotoxic proteins, and inadequately support neuronal function.210 In summary, these systemic and localized senescent processes not only enhance the pathophysiological impact of T2D but also foster an environment that promotes cognitive decline and the advancement of neurodegenerative diseases in older adults.211 Therefore, the neuroimmune-metabolic axis in T2D and aging becomes an essential framework for comprehending the relationship between immune aging and T2D.115,212

Mitochondria-ER crosstalk in regulating the ominous octet of T2D

Mitochondria and the ER are connected at specific contact points known as mitochondria-associated membranes (MAMs), which serve as a platform for interaction and molecular exchange between the two organelles.213 Their interaction is essential in the regulation of calcium signaling, lipid metabolism, and ROS dynamics. Disruption of the communication between the two organelles can have serious implications, as it is involved in the development of diabetes, impacting the ominous octet of T2D. It is through this interaction between mitochondria and ER that pancreatic β-cells operate, facilitating the flow of calcium between them, which in turn leads to the synthesis of ATP essential for glucose-stimulated insulin secretion.214,215 When disrupted, this interface causes a calcium imbalance that hampers ATP production, which subsequently affects the glucose-stimulated insulin secretion. Besides, an increase in mitochondrial calcium can bring about apoptosis due to the release of cytochrome c, which is important for the electron transport chain and activation of apoptotic pathways.216 MD also enhances the production of ROS, which aggravates ER stress and affects β-cell viability and function.217 Moreover, the disruption of MAM has an impact not only on calcium regulation and mitochondrial function but also results in an imbalance in lipid metabolism, which leads to the accumulation of ceramides. This accumulation, along with elevated levels of ROS and ER stress, triggers the activation of the JNK signaling pathway. The activation of this stress-response pathway interferes with insulin signaling, ultimately resulting in IR and the onset of diabetes. Moreover, in peripheral tissues such as muscle and liver, MAMs are major modulators of insulin sensitivity.218 These intersectional contacts are at risk for catastrophe in lipid metabolism, allowing toxic intermediates, such as ceramides, to accumulate and interfere effectively with insulin signaling.219 In parallel, ROS and ER stress can activate inflammatory pathways, such as the JNK pathway, which inhibit critical components of insulin signaling and exacerbate IR.220,221 To elaborate further, JNK acts as a vital mediator that links cellular stress with inflammation and metabolic dysfunction when it is activated by ER and oxidative stress.222 Upon activation, JNK migrates to the nucleus and phosphorylates transcription factors such as c-Jun, enhancing the AP-1 (activator protein 1)-driven expression of pro-inflammatory cytokines like TNF-α, IL-6, and IL-1β, which are involved in the low-grade chronic inflammation seen in aging and metabolic disorders.223 At the same time, JNK hampers insulin action by attaching phosphate groups to IRS-1 on serine residues (including Ser307 in rodents), which interferes with its tyrosine phosphorylation, thereby impeding the subsequent PI3K-Akt activation necessary for glucose uptake.224 This alteration not only diminishes insulin sensitivity but also drives cells into a state that is both pro-inflammatory and metabolically inflexible. In tissues such as adipose and muscle, prolonged JNK activation leads to the infiltration of immune cells, changes in adipokine profiles, and a disruption in metabolic homeostasis.225 In the liver, even minor JNK hyperactivation can result in increased lipid accumulation and IR, which may contribute to the early stages of metabolic dysfunction in this organ.226 Furthermore, JNK engages with stress-responsive signaling pathways such as NF-κB, IRE1-XBP1 (X-box binding protein 1), and the NLRP3 inflammasome, exacerbating a harmful feedback loop.227 As a result, the JNK pathway functions as a key element that converts intracellular stress into widespread low-grade inflammation and IR, both of which are crucial to metabolic syndromes associated with aging.228

A significant area where ER stress and MD overlap is the activation of inflammatory signaling pathways associated with stress, particularly the NF-κB pathway, which is essential for disturbing metabolic homeostasis.229,230 The activation of NF-κB, which is often initiated by ROS, the UPR, and lipid-induced ER stress, intensifies inflammation in tissues sensitive to insulin.231 In AT, NF-κB boosts the generation of chemokines that attract pro-inflammatory M1 macrophages, creating a self-sustaining inflammatory cycle. In both skeletal muscle and liver tissues, cytokines activated by NF-κB disrupt insulin receptor signaling and impede OXOPHOS, further diminishing glucose uptake and utilization.232,233 Hepatocytes, in particular, experience a mild inflammatory state and cellular stress facilitated by NF-κB, creating a favorable environment for lipid accumulation and the development of hepatic IR.234 While these alterations might initially remain unnoticed, sustained NF-κB activation accelerates the metabolic decline typically associated with T2D. Additionally, NF-κB indirectly impairs insulin functionality by increasing levels of SOCS proteins and promoting the serine phosphorylation of IRS-1, which together interfere with the insulin signaling pathway and reduce downstream activation of PI3K-Akt.235 Therefore, given its crucial involvement in inflammation and IR, NF-κB acts as a molecular connection between mitochondrial-ER stress and various aspects of the “ominous octet” that drives the pathophysiology of T2D.

Furthermore, the important molecular elements involved in the anchoring junctions between mitochondria and ER include the mitochondrial calcium uniporter.236 This would allow entry of calcium into the mitochondria, the subunit essential for ATP formation. Malfunctions of inositol 1,4,5-trisphosphate receptor (IP3R) associated with calcium transport from the ER into mitochondria may cause excessive calcium overload into mitochondria, with increased ROS production.237 Moreover, proteins such as mitofusin-2 and voltage-dependent anion channel 1 are also critical for the structural integrity of MAM, including maintaining efficient transport of their metabolites, and their malfunction would lead to the onset of diabetes.238

The interaction between mitochondria and the ER primarily determines the liver’s ability to manage gluconeogenesis.239 Ineffective calcium signaling can activate enzymatic functions vital to gluconeogenesis, like PEPCK and glucose-6-phosphatase (G6Pase), ultimately resulting in excessive glucose production.240 Consequently, the liver’s ability to reduce glucose production will also decline due to decreased ATP generation from mitochondria, which indirectly elevates blood glucose levels.241 Incretin hormones such as GLP-1 depend, furthermore, on robust interactions between mitochondrial and ER functions in the body for their impact on insulin release.242 However, MD disrupts bioenergetics and also hampers the insulin-stimulating effect of GLP-1.243 Chronic and excessive strain on the ER leads to a diminished molecular affinity of incretin receptors, thereby severely undermining incretin’s role in glucose balance.244 Importantly, the communication between mitochondria and ERs is essential in AT, where it has an important regulatory role in fat metabolism. If this interaction is disrupted, it could lead to excessive breakdown of AT, resulting in the accumulation of FFAs that damage the β-cells, cause MD, and adversely affect overall IR.245 Similarly, α-cell metabolism and glucagon secretion are influenced by calcium concentrations at MAMs, where abnormal signaling may trigger excessive glucagon secretion and, as a result, unwarranted glucose production in the liver.246 As previously stated, disruptions in communication between mitochondria and ER affect renal glucose absorption since renal tubular cell stress has been linked to increased expression of SGLT2, which promotes glucose absorption and poses a burden on rising blood glucose levels.247 Interestingly, the dysfunction of neurotransmitters, specifically within the hypothalamus, represents yet another aspect that changes the interaction of mitochondria and the ER.248 The hypothalamus uses this connection to control food intake, maintain energy equilibrium, and support the overall function of insulin.249 Disruption of calcium transport hinders mitochondrial ATP production and fails to signal the effects of insulin and leptin, with leptin being the hormone that regulates appetite in hypothalamic neurons.250 Besides, the buildup of ROS intensifies the disruption of neuronal activity, which impacts the central regulation of glucose and lipid metabolism in the body.251

In summary, the interaction of mitochondria and the ER is critical given the various obstacles posed by diabetes. Disruption in communication from the mitochondria to the ER may reinforce β-cell failure, IR, hyperglycemia, and lipid mismanagement, creating a vicious cycle of metabolic deterioration by affecting individual components of the “ominous octet” of T2D. Any additional knowledge that may be uncovered about this relationship can serve as a guiding light in the search for new therapeutic tools aimed at restoring cellular homeostasis, as well as forestalling and/or preventing diabetes.

Limitations of the study

This review primarily focuses on the cellular and molecular mechanisms underlying T2D, highlighting essential processes such as metabolic dysfunction, ER stress, ROS, maintenance of calcium levels, and the activation of stress kinases like JNK and NF-κB. These mechanisms offer valuable insights into the pathophysiology of T2D and serve as a foundation for understanding the disease at the cellular level. However, our current knowledge primarily stems from preclinical and mechanistic studies, which do not necessarily accurately capture the clinical diversity found in human populations. Factors such as diet, obesity prevalence, and environmental influences all contribute to this variability, making it difficult for mechanistic research to fully address. While we emphasize the key components of the “ominous octet”, a thorough exploration of how these molecular changes translate into clinical outcomes and treatment strategies is not included in this paper. A significant portion of the mechanistic data is derived from in vitro studies or animal models, particularly mice, in the context of IR and organelle dysfunctions. However, these models are constrained by their failure to account for metabolic, immune response, and physiological differences between species. Demographic aspects like gender, age, and ethnicity are important but are less emphasized in this review. Variations in mitochondrial and ER functions with age, metabolic differences across genders, and the ethnic disparities in T2D prevalence and prognosis are vital, but they are beyond the focus of this study. Although we talk about well-known therapies such as GLP-1RAs and anti-inflammatory treatments, we do not explore emerging fields of study like the gut microbiome, circadian rhythms, and epigenetic modifications that may affect mitochondrial-endoplasmic reticulum interactions, which could be vital topics for future review articles. These areas hold potential for developing more targeted research into the pathophysiology of T2D. Taken together, many of the study’s limitations arise from the inherent difficulty of covering all aspects of T2D in a review that primarily focuses on mechanisms. Essentially, our primary goal is to establish a foundational understanding of mitochondrial-ER interactions in T2D, focusing on the pathological mechanisms that arise across multiple organ systems. This includes exploring the central role of the hyperinsulinemia–inflammation axis in disease progression. This underscores the need for forthcoming research to consider clinical, demographic, and environmental factors. Such studies will be crucial for making T2D treatment more individualized and for devising therapies that are more targeted and efficient.

Future directions

The rising global incidence of T2D in developing countries highlights the urgent necessity for novel and personalized treatment strategies. Predictions indicate that by 2045, around 783 million people are expected to be diagnosed with T2D, making the condition increasingly complicated. The T2D framework, which is based on metabolic imbalances marked by IR and inflammatory mechanisms, termed the hyperinsulinemia-inflammation axis, provides a thorough understanding of the need for customized therapeutic approaches. There are notable gaps in our knowledge regarding which specific factors are most negatively impacted in individuals with T2D, which is critical for implementing targeted treatment options. For instance, when MD becomes a key causal factor, it is essential to prioritize treatment strategies that aim to restore mitochondrial performance. Likewise, intervention methods that concentrate on reducing ER stress or lowering inflammation may yield better results. A comprehensive, multi-dimensional approach that simultaneously targets mitochondrial activity, ER stress, and inflammation is likely to improve the effectiveness of T2D treatment.

An area of research worth exploring is MAMs, which play a role in maintaining cellular balance and are linked to insulin resistance and inflammation in T2D. Treatments aimed at MAMs could restore cellular function and enhance insulin sensitivity by increasing interactions of MAM proteins, such as mitofusin-2, which improves communication between mitochondria and the ER while decreasing oxidative stress. Substances like resveratrol and spermidine promote the generation of new mitochondria, whereas calcium transport proteins at the MAM interface may be leveraged to restore calcium equilibrium, which is crucial for insulin signaling.252,253 Furthermore, modifying lipid metabolism at MAMs could help to decrease lipid buildup and enhance insulin sensitivity, presenting a new strategy to tackle the metabolic problems central to T2D.

Exploring the oxidative processes that contribute to IR and high blood glucose may create new opportunities for developing treatments that enhance insulin sensitivity, reduce chronic inflammation, and better regulate metabolism. When paired with lifestyle changes, such as physical activity and dietary adjustments, these approaches could present a more integrated strategy for managing T2D. Future studies should also examine drug therapies that impact mitochondrial function or promote insulin secretion. Tailored treatments involving gut hormones like GLP-1RAs, recognized for their anti-inflammatory properties, could enhance glucose management and cognitive functions, especially in older adults.

Ultimately, the role of immune aging in the development of T2D, particularly among older adults, warrants further investigation. Age-related immune changes, such as chronic inflammation and the buildup of senescent cells, can disrupt glucose balance, yet these mechanisms remain inadequately understood. Strategies aimed at mitigating immune aging, like senolytic therapies, immune modulators, and anti-inflammatory agents, show promise but require further refinement and evaluation.254,255 At the same time, epigenetic clocks that distinguish biological age from chronological age provide a valuable tool for identifying individuals at risk of accelerated immune decline.256,257 These resources can facilitate early interventions to mitigate immune dysfunction and lower the likelihood of developing T2D. Enhancing mitochondrial function, reducing ER stress, and preserving immune resilience are crucial not only for managing T2D but also for promoting healthy aging. A mechanistic and personalized approach will be essential in shaping future treatments and optimizing long-term care for elderly populations.

Conclusions

The Ominous Octet model offers a comprehensive perspective on the interconnected mechanisms through which T2D develops. The processes that drive T2D include hyperinsulinemia, widespread inflammation, MD, and ER stress, all of which contribute to the gradual decline of glucose metabolism. Compromised insulin signaling, increased gluconeogenesis, and impaired β-cell function led to hyperglycemia and IR. T2D in the elderly should be treated as such, as age-related difficulties complicate the condition due to a generally compromised immune system and, as a result, chronic inflammation, which can emerge from metabolic tissues. Developing personalized and integrated treatment strategies for mitochondrial health, inflammation, and organelle dysfunction is crucial for managing T2D. Key factors include pro-inflammatory cytokines (IL-1β, IL-6, TNF-α, IFN-γ) that impair β-cell functionality, along with chemokines such as C-C motif chemokine ligand 2 (CCL2) and C-X-C motif chemokine ligand 10 (CXCL10), which recruit inflammatory monocytes to insulin-sensitive tissues. Adipokines such as leptin and adiponectin play a role in modulating immune responses within adipose tissue, while AGEs activate RAGE receptors, triggering NF-κB signaling and persistent inflammation. Additionally, dysfunction is worsened by senescent cells through the SASP. Future research should prioritize personalized therapies to address T2D in developing nations and improve outcomes for aging populations.

Declarations

Acknowledgement

None.

Funding

This research was supported by Bandhan, Kolkata, India.

Conflict of interest

The authors have no conflicts of interest to declare.

Authors’ contributions

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

References

  1. Noto S. Perspectives on Aging and Quality of Life. Healthcare (Basel) 2023;11(15):2131 View Article PubMed/NCBI
  2. Theodorakis N, Feretzakis G, Hitas C, Kreouzi M, Kalantzi S, Spyridaki A, et al. Immunosenescence: How Aging Increases Susceptibility to Bacterial Infections and Virulence Factors. Microorganisms 2024;12(10):2052 View Article PubMed/NCBI
  3. Goyani P, Christodoulou R, Vassiliou E. Immunosenescence: Aging and Immune System Decline. Vaccines (Basel) 2024;12(12):1314 View Article PubMed/NCBI
  4. Boccardi V, Marano L. Aging, Cancer, and Inflammation: The Telomerase Connection. Int J Mol Sci 2024;25(15):8542 View Article PubMed/NCBI
  5. Nassour J, Przetocka S, Karlseder J. Telomeres as hotspots for innate immunity and inflammation. DNA Repair (Amst) 2024;133:103591 View Article PubMed/NCBI
  6. Fan H, Qiao Z, Li J, Shang G, Shang C, Chen S, et al. Recent advances in senescence-associated secretory phenotype and osteoporosis. Heliyon 2024;10(4):e25538 View Article PubMed/NCBI
  7. Kumari R, Jat P. Mechanisms of Cellular Senescence: Cell Cycle Arrest and Senescence Associated Secretory Phenotype. Front Cell Dev Biol 2021;9:645593 View Article PubMed/NCBI
  8. Li X, Li C, Zhang W, Wang Y, Qian P, Huang H. Inflammation and aging: signaling pathways and intervention therapies. Signal Transduct Target Ther 2023;8(1):239 View Article PubMed/NCBI
  9. Wang R, Lan C, Benlagha K, Camara NOS, Miller H, Kubo M, et al. The interaction of innate immune and adaptive immune system. MedComm (2020) 2024;5(10):e714 View Article PubMed/NCBI
  10. Chen J, Deng JC, Goldstein DR. How aging impacts vaccine efficacy: known molecular and cellular mechanisms and future directions. Trends Mol Med 2022;28(12):1100-1111 View Article PubMed/NCBI
  11. Bajaj V, Gadi N, Spihlman AP, Wu SC, Choi CH, Moulton VR. Aging, Immunity, and COVID-19: How Age Influences the Host Immune Response to Coronavirus Infections?. Front Physiol 2020;11:571416 View Article PubMed/NCBI
  12. Liang Z, Dong X, Zhang Z, Zhang Q, Zhao Y. Age-related thymic involution: Mechanisms and functional impact. Aging Cell 2022;21(8):e13671 View Article PubMed/NCBI
  13. Shirafkan F, Hensel L, Rattay K. Immune tolerance and the prevention of autoimmune diseases essentially depend on thymic tissue homeostasis. Front Immunol 2024;15:1339714 View Article PubMed/NCBI
  14. Cardinale A, De Luca CD, Locatelli F, Velardi E. Thymic Function and T-Cell Receptor Repertoire Diversity: Implications for Patient Response to Checkpoint Blockade Immunotherapy. Front Immunol 2021;12:752042 View Article PubMed/NCBI
  15. Duah M, Li L, Shen J, Lan Q, Pan B, Xu K. Thymus Degeneration and Regeneration. Front Immunol 2021;12:706244 View Article PubMed/NCBI
  16. Müller L, Di Benedetto S. Inflammaging, immunosenescence, and cardiovascular aging: insights into long COVID implications. Front Cardiovasc Med 2024;11:1384996 View Article PubMed/NCBI
  17. Wrona MV, Ghosh R, Coll K, Chun C, Yousefzadeh MJ. The 3 I’s of immunity and aging: immunosenescence, inflammaging, and immune resilience. Front Aging 2024;5:1490302 View Article PubMed/NCBI
  18. Berbudi A, Khairani S, Tjahjadi AI. Interplay Between Insulin Resistance and Immune Dysregulation in Type 2 Diabetes Mellitus: Implications for Therapeutic Interventions. Immunotargets Ther 2025;14:359-382 View Article PubMed/NCBI
  19. Li H, Zou L, Long Z, Zhan J. Immunometabolic alterations in type 2 diabetes mellitus revealed by single-cell RNA sequencing: insights into subtypes and therapeutic targets. Front Immunol 2024;15:1537909 View Article PubMed/NCBI
  20. Ginefra P, Hope HC, Lorusso G, D’Amelio P, Vannini N. The immunometabolic roots of aging. Curr Opin Immunol 2024;91:102498 View Article PubMed/NCBI
  21. Rizvi AA, Rizzo M. Age-Related Changes in Insulin Resistance and Muscle Mass: Clinical Implications in Obese Older Adults. Medicina (Kaunas) 2024;60(10):1648 View Article PubMed/NCBI
  22. Fazio S, Bellavite P, Affuso F. Chronically Increased Levels of Circulating Insulin Secondary to Insulin Resistance: A Silent Killer. Biomedicines 2024;12(10):2416 View Article PubMed/NCBI
  23. 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
  24. Galicia-Garcia U, Benito-Vicente A, Jebari S, Larrea-Sebal A, Siddiqi H, Uribe KB, et al. Pathophysiology of Type 2 Diabetes Mellitus. Int J Mol Sci 2020;21(17):6275 View Article PubMed/NCBI
  25. Zong Y, Li H, Liao P, Chen L, Pan Y, Zheng Y, et al. Mitochondrial dysfunction: mechanisms and advances in therapy. Signal Transduct Target Ther 2024;9(1):124 View Article PubMed/NCBI
  26. Bilbao-Malavé V, González-Zamora J, de la Puente M, Recalde S, Fernandez-Robredo P, Hernandez M, et al. Mitochondrial Dysfunction and Endoplasmic Reticulum Stress in Age Related Macular Degeneration, Role in Pathophysiology, and Possible New Therapeutic Strategies. Antioxidants (Basel) 2021;10(8):1170 View Article PubMed/NCBI
  27. Defronzo RA. Banting Lecture. From the triumvirate to the ominous octet: a new paradigm for the treatment of type 2 diabetes mellitus. Diabetes 2009;58(4):773-795 View Article PubMed/NCBI
  28. Papatheodorou K, Shubrook JH. Beta-cell preservation in T2DM using a pathophysiologic approach. Postgrad Med 2025;137(3-4):235-242 View Article PubMed/NCBI
  29. Nunkoo VS, Cristian A, Jurcau A, Diaconu RG, Jurcau MC. The Quest for Eternal Youth: Hallmarks of Aging and Rejuvenating Therapeutic Strategies. Biomedicines 2024;12(11):2540 View Article PubMed/NCBI
  30. Zhao X, An X, Yang C, Sun W, Ji H, Lian F. The crucial role and mechanism of insulin resistance in metabolic disease. Front Endocrinol (Lausanne) 2023;14:1149239 View Article PubMed/NCBI
  31. Orliaguet L, Dalmas E, Drareni K, Venteclef N, Alzaid F. Mechanisms of Macrophage Polarization in Insulin Signaling and Sensitivity. Front Endocrinol (Lausanne) 2020;11:62 View Article PubMed/NCBI
  32. Püschel GP, Klauder J, Henkel J. Macrophages, Low-Grade Inflammation, Insulin Resistance and Hyperinsulinemia: A Mutual Ambiguous Relationship in the Development of Metabolic Diseases. J Clin Med 2022;11(15):4358 View Article PubMed/NCBI
  33. Rodríguez-Morales P, Franklin RA. Macrophage phenotypes and functions: resolving inflammation and restoring homeostasis. Trends Immunol 2023;44(12):986-998 View Article PubMed/NCBI
  34. Witcoski Junior L, de Lima JD, Somensi AG, de Souza Santos LB, Paschoal GL, Uada TS, et al. Metabolic reprogramming of macrophages in the context of type 2 diabetes. Eur J Med Res 2024;29(1):497 View Article PubMed/NCBI
  35. d’Aiello A, Filomia S, Brecciaroli M, Sanna T, Pedicino D, Liuzzo G. Targeting Inflammatory Pathways in Atherosclerosis: Exploring New Opportunities for Treatment. Curr Atheroscler Rep 2024;26(12):707-719 View Article PubMed/NCBI
  36. Mahmoud LM, Mageed AAAA, Saadallah JM, Youssef MF, Rashed LA, Ammar HI. Interleukin 1β receptor blocker (Anakinra) and regenerative stem cell therapy: two novel approaches effectively ameliorating diabetic cardiomyopathy. Naunyn Schmiedebergs Arch Pharmacol 2024;397(10):8023-8041 View Article PubMed/NCBI
  37. 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
  38. 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
  39. Chen J, Mei A, Wei Y, Li C, Qian H, Min X, et al. GLP-1 receptor agonist as a modulator of innate immunity. Front Immunol 2022;13:997578 View Article PubMed/NCBI
  40. Zhang C, Shi Y, Liu C, Sudesh SM, Hu Z, Li P, et al. Therapeutic strategies targeting mechanisms of macrophages in diabetic heart disease. Cardiovasc Diabetol 2024;23(1):169 View Article PubMed/NCBI
  41. Zhao L, Hu H, Zhang L, Liu Z, Huang Y, Liu Q, et al. Inflammation in diabetes complications: molecular mechanisms and therapeutic interventions. MedComm (2020) 2024;5(4):e516 View Article PubMed/NCBI
  42. Julliard WA, Myo YPA, Perelas A, Jackson PD, Thatcher TH, Sime PJ. Specialized pro-resolving mediators as modulators of immune responses. Semin Immunol 2022;59:101605 View Article PubMed/NCBI
  43. Cholkar K, Gilger BC, Mitra AK. Topical delivery of aqueous micellar resolvin E1 analog (RX-10045). Int J Pharm 2016;498(1-2):326-334 View Article PubMed/NCBI
  44. Beyer MP, Videla LA, Farías C, Valenzuela R. Potential Clinical Applications of Pro-Resolving Lipids Mediators from Docosahexaenoic Acid. Nutrients 2023;15(15):3317 View Article PubMed/NCBI
  45. Sinha SK, Carpio MB, Nicholas SB. Fiery Connections: Macrophage-Mediated Inflammation, the Journey from Obesity to Type 2 Diabetes Mellitus and Diabetic Kidney Disease. Biomedicines 2024;12(10):2209 View Article PubMed/NCBI
  46. Song S, Ni J, Sun Y, Pu Q, Zhang L, Yan Q, et al. Association of inflammatory cytokines with type 2 diabetes mellitus and diabetic nephropathy: a bidirectional Mendelian randomization study. Front Med (Lausanne) 2024;11:1459752 View Article PubMed/NCBI
  47. Amo-Aparicio J, Dinarello CA, Lopez-Vales R. Metabolic reprogramming of the inflammatory response in the nervous system: the crossover between inflammation and metabolism. Neural Regen Res 2024;19(10):2189-2201 View Article PubMed/NCBI
  48. Liu AB, Tan B, Yang P, Tian N, Li JK, Wang SC, et al. The role of inflammatory response and metabolic reprogramming in sepsis-associated acute kidney injury: mechanistic insights and therapeutic potential. Front Immunol 2024;15:1487576 View Article PubMed/NCBI
  49. Li H, Meng Y, He S, Tan X, Zhang Y, Zhang X, et al. Macrophages, Chronic Inflammation, and Insulin Resistance. Cells 2022;11(19):3001 View Article PubMed/NCBI
  50. Li M, Chi X, Wang Y, Setrerrahmane S, Xie W, Xu H. Trends in insulin resistance: insights into mechanisms and therapeutic strategy. Signal Transduct Target Ther 2022;7(1):216 View Article PubMed/NCBI
  51. Sugandh F, Chandio M, Raveena F, Kumar L, Karishma F, Khuwaja S, et al. Advances in the Management of Diabetes Mellitus: A Focus on Personalized Medicine. Cureus 2023;15(8):e43697 View Article PubMed/NCBI
  52. Williams DM, Jones H, Stephens JW. Personalized Type 2 Diabetes Management: An Update on Recent Advances and Recommendations. Diabetes Metab Syndr Obes 2022;15:281-295 View Article PubMed/NCBI
  53. Murakami T, Inagaki N, Kondoh H. Cellular Senescence in Diabetes Mellitus: Distinct Senotherapeutic Strategies for Adipose Tissue and Pancreatic β Cells. Front Endocrinol (Lausanne) 2022;13:869414 View Article PubMed/NCBI
  54. Iwasaki K, Abarca C, Aguayo-Mazzucato C. Regulation of Cellular Senescence in Type 2 Diabetes Mellitus: From Mechanisms to Clinical Applications. Diabetes Metab J 2023;47(4):441-453 View Article PubMed/NCBI
  55. Fazio S, Affuso F, Cesaro A, Tibullo L, Fazio V, Calabrò P. Insulin Resistance/Hyperinsulinemia as an Independent Risk Factor That Has Been Overlooked for Too Long. Biomedicines 2024;12(7):1417 View Article PubMed/NCBI
  56. 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
  57. Narasimhan A, Flores RR, Robbins PD, Niedernhofer LJ. Role of Cellular Senescence in Type II Diabetes. Endocrinology 2021;162(10):bqab136 View Article PubMed/NCBI
  58. Teissier T, Boulanger E, Cox LS. Interconnections between Inflammageing and Immunosenescence during Ageing. Cells 2022;11(3):359 View Article PubMed/NCBI
  59. Pangrazzi L, Meryk A. Molecular and Cellular Mechanisms of Immunosenescence: Modulation Through Interventions and Lifestyle Changes. Biology (Basel) 2024;14(1):17 View Article PubMed/NCBI
  60. Sayyed Kassem L, Rajpal A, Barreiro MV, Ismail-Beigi F. Beta-cell function in type 2 diabetes (T2DM): Can it be preserved or enhanced?. J Diabetes 2023;15(10):817-837 View Article PubMed/NCBI
  61. Weir GC, Gaglia J, Bonner-Weir S. Inadequate β-cell mass is essential for the pathogenesis of type 2 diabetes. Lancet Diabetes Endocrinol 2020;8(3):249-256 View Article PubMed/NCBI
  62. Palmer AK, Tchkonia T, Kirkland JL. Senolytics: Potential for Alleviating Diabetes and Its Complications. Endocrinology 2021;162(8):bqab058 View Article PubMed/NCBI
  63. Islam MT, Tuday E, Allen S, Kim J, Trott DW, Holland WL, et al. Senolytic drugs, dasatinib and quercetin, attenuate adipose tissue inflammation, and ameliorate metabolic function in old age. Aging Cell 2023;22(2):e13767 View Article PubMed/NCBI
  64. Mannarino M, Wu-Martinez O, Sheng K, Li L, Navarro-Ramirez R, Jarzem P, et al. Senolytic Combination Treatment Is More Potent Than Single Drugs in Reducing Inflammatory and Senescence Burden in Cells from Painful Degenerating IVDs. Biomolecules 2023;13(8):1257 View Article PubMed/NCBI
  65. Lorenzo EC, Torrance BL, Haynes L. Impact of senolytic treatment on immunity, aging, and disease. Front Aging 2023;4:1161799 View Article PubMed/NCBI
  66. Hickson LJ, Langhi Prata LGP, Bobart SA, Evans TK, Giorgadze N, Hashmi SK, et al. Senolytics decrease senescent cells in humans: Preliminary report from a clinical trial of Dasatinib plus Quercetin in individuals with diabetic kidney disease. EBioMedicine 2019;47:446-456 View Article PubMed/NCBI
  67. 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
  68. Kuryłowicz A, Koźniewski K. Anti-Inflammatory Strategies Targeting Metaflammation in Type 2 Diabetes. Molecules 2020;25(9):2224 View Article PubMed/NCBI
  69. Ding Y, Deng A, Qi TF, Yu H, Wu LP, Zhang H. The burden and trend prediction of ischemic heart disease associated with lead exposure: Insights from the Global Burden of Disease study 2021. Environ Health 2025;24(1):23 View Article PubMed/NCBI
  70. Abdissa SG, Deressa W, Shah AJ. Incidence of heart failure among diabetic patients with ischemic heart disease: a cohort study. BMC Cardiovasc Disord 2020;20(1):181 View Article PubMed/NCBI
  71. Yoon J, Jung D, Lee Y, Park B. The Metabolic Score for Insulin Resistance (METS-IR) as a Predictor of Incident Ischemic Heart Disease: A Longitudinal Study among Korean without Diabetes. J Pers Med 2021;11(8):742 View Article PubMed/NCBI
  72. Martínez Báez A, Ayala G, Pedroza-Saavedra A, González-Sánchez HM, Chihu Amparan L. Phosphorylation Codes in IRS-1 and IRS-2 Are Associated with the Activation/Inhibition of Insulin Canonical Signaling Pathways. Curr Issues Mol Biol 2024;46(1):634-649 View Article PubMed/NCBI
  73. Kirichenko TV, Markina YV, Bogatyreva AI, Tolstik TV, Varaeva YR, Starodubova AV. The Role of Adipokines in Inflammatory Mechanisms of Obesity. Int J Mol Sci 2022;23(23):14982 View Article PubMed/NCBI
  74. Ouchi N, Parker JL, Lugus JJ, Walsh K. Adipokines in inflammation and metabolic disease. Nat Rev Immunol 2011;11(2):85-97 View Article PubMed/NCBI
  75. Jiang S, Young JL, Wang K, Qian Y, Cai L. Diabetic-induced alterations in hepatic glucose and lipid metabolism: The role of type 1 and type 2 diabetes mellitus (Review). Mol Med Rep 2020;22(2):603-611 View Article PubMed/NCBI
  76. Santoleri D, Titchenell PM. Resolving the Paradox of Hepatic Insulin Resistance. Cell Mol Gastroenterol Hepatol 2019;7(2):447-456 View Article PubMed/NCBI
  77. 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
  78. Langlois A, Cherfan J, Meugnier E, Rida A, Arous C, Peronet C, et al. DECORIN, a triceps-derived myokine, protects sorted β-cells and human islets against chronic inflammation associated with type 2 diabetes. Acta Physiol (Oxf) 2025;241(2):e14267 View Article PubMed/NCBI
  79. 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
  80. Zatterale F, Longo M, Naderi J, Raciti GA, Desiderio A, Miele C, et al. Chronic Adipose Tissue Inflammation Linking Obesity to Insulin Resistance and Type 2 Diabetes. Front Physiol 2019;10:1607 View Article PubMed/NCBI
  81. Le TKC, Dao XD, Nguyen DV, Luu DH, Bui TMH, Le TH, et al. Insulin signaling and its application. Front Endocrinol (Lausanne) 2023;14:1226655 View Article PubMed/NCBI
  82. Małkowska P. Positive Effects of Physical Activity on Insulin Signaling. Curr Issues Mol Biol 2024;46(6):5467-5487 View Article PubMed/NCBI
  83. Evia-Viscarra ML, Rodea-Montero ER, Apolinar-Jiménez E, Muñoz-Noriega N, García-Morales LM, Leaños-Pérez C, et al. The effects of metformin on inflammatory mediators in obese adolescents with insulin resistance: controlled randomized clinical trial. J Pediatr Endocrinol Metab 2012;25(1-2):41-49 View Article PubMed/NCBI
  84. Jonkers IJ, Mohrschladt MF, Westendorp RG, van der Laarse A, Smelt AH. Severe hypertriglyceridemia with insulin resistance is associated with systemic inflammation: reversal with bezafibrate therapy in a randomized controlled trial. Am J Med 2002;112(4):275-280 View Article PubMed/NCBI
  85. Herder C, Færch K, Carstensen-Kirberg M, Lowe GD, Haapakoski R, Witte DR, et al. Biomarkers of subclinical inflammation and increases in glycaemia, insulin resistance and beta-cell function in non-diabetic individuals: the Whitehall II study. Eur J Endocrinol 2016;175(5):367-377 View Article PubMed/NCBI
  86. Chen YK, Liu TT, Teia FKF, Xie MZ. Exploring the underlying mechanisms of obesity and diabetes and the potential of Traditional Chinese Medicine: an overview of the literature. Front Endocrinol (Lausanne) 2023;14:1218880 View Article PubMed/NCBI
  87. Amisi CA. Markers of insulin resistance in Polycystic ovary syndrome women: An update. World J Diabetes 2022;13(3):129-149 View Article PubMed/NCBI
  88. Bashir H, Ahmad Bhat S, Majid S, Hamid R, Koul RK, Rehman MU, et al. Role of inflammatory mediators (TNF-α, IL-6, CRP), biochemical and hematological parameters in type 2 diabetes mellitus patients of Kashmir, India. Med J Islam Repub Iran 2020;34:5 View Article PubMed/NCBI
  89. Zheng M, Wang P. Role of insulin receptor substance-1 modulating PI3K/Akt insulin signaling pathway in Alzheimer’s disease. 3 Biotech 2021;11(4):179 View Article PubMed/NCBI
  90. Andreae SJ, Reeves H, Casey T, Lindberg A, Pickett KA. A systematic review of diabetes prevention programs adapted to include family members. Prev Med Rep 2024;39:102655 View Article PubMed/NCBI
  91. Chen Q, Kovilakath A, Allegood J, Thompson J, Hu Y, Cowart LA, et al. Endoplasmic reticulum stress and mitochondrial dysfunction during aging: Role of sphingolipids. Biochim Biophys Acta Mol Cell Biol Lipids 2023;1868(10):159366 View Article PubMed/NCBI
  92. Patergnani S, Bouhamida E, Leo S, Pinton P, Rimessi A. Mitochondrial Oxidative Stress and “Mito-Inflammation”: Actors in the Diseases. Biomedicines 2021;9(2):216 View Article PubMed/NCBI
  93. 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(24):13384 View Article PubMed/NCBI
  94. Guo J, Huang X, Dou L, Yan M, Shen T, Tang W, et al. Aging and aging-related diseases: from molecular mechanisms to interventions and treatments. Signal Transduct Target Ther 2022;7(1):391 View Article PubMed/NCBI
  95. Yang J, Luo J, Tian X, Zhao Y, Li Y, Wu X. Progress in Understanding Oxidative Stress, Aging, and Aging-Related Diseases. Antioxidants (Basel) 2024;13(4):394 View Article PubMed/NCBI
  96. Lizák B, Kapuy O. Advances in Endoplasmic Reticulum Stress Research-Insights from the Special Issue “Endoplasmic Reticulum Stress and Apoptosis”. Int J Mol Sci 2025;26(6):2487 View Article PubMed/NCBI
  97. Kim P. Understanding the Unfolded Protein Response (UPR) Pathway: Insights into Neuropsychiatric Disorders and Therapeutic Potentials. Biomol Ther (Seoul) 2024;32(2):183-191 View Article PubMed/NCBI
  98. 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
  99. Yang ZH, Chen FZ, Zhang YX, Ou MY, Tan PC, Xu XW, et al. Therapeutic targeting of white adipose tissue metabolic dysfunction in obesity: mechanisms and opportunities. MedComm (2020) 2024;5(6):e560 View Article PubMed/NCBI
  100. Zadian SS, Jahanbin K, Nikooei S, Rostaminejad M, Rahimi A, Abdizadeh P, et al. View Article PubMed/NCBI
  101. Inoue S. [Clinical description of dementia. 5. Symptoms of dementia (4)]. Kangogaku Zasshi 1985;49(9):1053-1056 View Article PubMed/NCBI
  102. Chen W, Zhao H, Li Y. Mitochondrial dynamics in health and disease: mechanisms and potential targets. Signal Transduct Target Ther 2023;8(1):333 View Article PubMed/NCBI
  103. Wang S, Long H, Hou L, Feng B, Ma Z, Wu Y, et al. The mitophagy pathway and its implications in human diseases. Signal Transduct Target Ther 2023;8(1):304 View Article PubMed/NCBI
  104. Chen Y, Meng Z, Li Y, Liu S, Hu P, Luo E. Advanced glycation end products and reactive oxygen species: uncovering the potential role of ferroptosis in diabetic complications. Mol Med 2024;30(1):141 View Article PubMed/NCBI
  105. Twarda-Clapa A, Olczak A, Białkowska AM, Koziołkiewicz M. Advanced Glycation End-Products (AGEs): Formation, Chemistry, Classification, Receptors, and Diseases Related to AGEs. Cells 2022;11(8):1312 View Article PubMed/NCBI
  106. Vianello E, Beltrami AP, Aleksova A, Janjusevic M, Fluca AL, Corsi Romanelli MM, et al. The Advanced Glycation End-Products (AGE)-Receptor for AGE System (RAGE): An Inflammatory Pathway Linking Obesity and Cardiovascular Diseases. Int J Mol Sci 2025;26(8):3707 View Article PubMed/NCBI
  107. Tudurí E, Soriano S, Almagro L, Montanya E, Alonso-Magdalena P, Nadal Á, et al. The pancreatic β-cell in ageing: Implications in age-related diabetes. Ageing Res Rev 2022;80:101674 View Article PubMed/NCBI
  108. Rojas A, Lindner C, Schneider I, Gonzalez I, Uribarri J. The RAGE Axis: A Relevant Inflammatory Hub in Human Diseases. Biomolecules 2024;14(4):412 View Article PubMed/NCBI
  109. Weir GC. Glucolipotoxicity, β-Cells, and Diabetes: The Emperor Has No Clothes. Diabetes 2020;69(3):273-278 View Article PubMed/NCBI
  110. Zawada A, Machowiak A, Rychter AM, Ratajczak AE, Szymczak-Tomczak A, Dobrowolska A, et al. Accumulation of Advanced Glycation End-Products in the Body and Dietary Habits. Nutrients 2022;14(19):3982 View Article PubMed/NCBI
  111. Shen CY, Lu CH, Cheng CF, Li KJ, Kuo YM, Wu CH, et al. Advanced Glycation End-Products Acting as Immunomodulators for Chronic Inflammation, Inflammaging and Carcinogenesis in Patients with Diabetes and Immune-Related Diseases. Biomedicines 2024;12(8):1699 View Article PubMed/NCBI
  112. Papantoniou K, Aggeletopoulou I, Michailides C, Pastras P, Triantos C. Understanding the Role of NLRP3 Inflammasome in Acute Pancreatitis. Biology (Basel) 2024;13(11):945 View Article PubMed/NCBI
  113. Yao J, Sterling K, Wang Z, Zhang Y, Song W. The role of inflammasomes in human diseases and their potential as therapeutic targets. Signal Transduct Target Ther 2024;9(1):10 View Article PubMed/NCBI
  114. Zgutka K, Tkacz M, Tomasiak P, Tarnowski M. A Role for Advanced Glycation End Products in Molecular Ageing. Int J Mol Sci 2023;24(12):9881 View Article PubMed/NCBI
  115. García-Domínguez M. Pathological and Inflammatory Consequences of Aging. Biomolecules 2025;15(3):404 View Article PubMed/NCBI
  116. Ranbhise JS, Ju S, Singh MK, Han S, Akter S, Ha J, et al. Chronic Inflammation and Glycemic Control: Exploring the Bidirectional Link Between Periodontitis and Diabetes. Dent J (Basel) 2025;13(3):100 View Article PubMed/NCBI
  117. K S PK, Jyothi MN, Prashant A. Mitochondrial DNA variants in the pathogenesis and metabolic alterations of diabetes mellitus. Mol Genet Metab Rep 2025;42:101183 View Article PubMed/NCBI
  118. Back SH, Kaufman RJ. Endoplasmic reticulum stress and type 2 diabetes. Annu Rev Biochem 2012;81:767-793 View Article PubMed/NCBI
  119. Chen B, Li T, Wu Y, Song L, Wang Y, Bian Y, et al. Lipotoxicity: A New Perspective in Type 2 Diabetes Mellitus. Diabetes Metab Syndr Obes 2025;18:1223-1237 View Article PubMed/NCBI
  120. Dawi J, Misakyan Y, Affa S, Kades S, Narasimhan A, Hajjar F, et al. Oxidative Stress, Glutathione Insufficiency, and Inflammatory Pathways in Type 2 Diabetes Mellitus: Implications for Therapeutic Interventions. Biomedicines 2024;13(1):18 View Article PubMed/NCBI
  121. Dhokte S, Czaja K. Visceral Adipose Tissue: The Hidden Culprit for Type 2 Diabetes. Nutrients 2024;16(7):1015 View Article PubMed/NCBI
  122. Holeček M. Origin and Roles of Alanine and Glutamine in Gluconeogenesis in the Liver, Kidneys, and Small Intestine under Physiological and Pathological Conditions. Int J Mol Sci 2024;25(13):7037 View Article PubMed/NCBI
  123. Onyango AN. Excessive gluconeogenesis causes the hepatic insulin resistance paradox and its sequelae. Heliyon 2022;8(12):e12294 View Article PubMed/NCBI
  124. Ruze R, Liu T, Zou X, Song J, Chen Y, Xu R, et al. Obesity and type 2 diabetes mellitus: connections in epidemiology, pathogenesis, and treatments. Front Endocrinol (Lausanne) 2023;14:1161521 View Article PubMed/NCBI
  125. Deprince A, Haas JT, Staels B. Dysregulated lipid metabolism links NAFLD to cardiovascular disease. Mol Metab 2020;42:101092 View Article PubMed/NCBI
  126. Barroso E, Jurado-Aguilar J, Wahli W, Palomer X, Vázquez-Carrera M. Increased hepatic gluconeogenesis and type 2 diabetes mellitus. Trends Endocrinol Metab 2024;35(12):1062-1077 View Article PubMed/NCBI
  127. Liu H, Wang S, Wang J, Guo X, Song Y, Fu K, et al. Energy metabolism in health and diseases. Signal Transduct Target Ther 2025;10(1):69 View Article PubMed/NCBI
  128. Mooli RGR, Mukhi D, Ramakrishnan SK. Oxidative Stress and Redox Signaling in the Pathophysiology of Liver Diseases. Compr Physiol 2022;12(2):3167-3192 View Article PubMed/NCBI
  129. Zhang Z, Wang S, Gao L. Circadian rhythm, glucose metabolism and diabetic complications: the role of glucokinase and the enlightenment on future treatment. Front Physiol 2025;16:1537231 View Article PubMed/NCBI
  130. Díaz-Castro F, Morselli E, Claret M. Interplay between the brain and adipose tissue: a metabolic conversation. EMBO Rep 2024;25(12):5277-5293 View Article PubMed/NCBI
  131. Wachsmuth HR, Weninger SN, Duca FA. Role of the gut-brain axis in energy and glucose metabolism. Exp Mol Med 2022;54(4):377-392 View Article PubMed/NCBI
  132. Mahmood R, Maccourtney D, Vashi M, Mohamed A. A Case of Metformin-Associated Lactic Acidosis. Cureus 2023;15(4):e38222 View Article PubMed/NCBI
  133. Perry RJ, Shulman GI. Sodium-glucose cotransporter-2 inhibitors: Understanding the mechanisms for therapeutic promise and persisting risks. J Biol Chem 2020;295(42):14379-14390 View Article PubMed/NCBI
  134. Feng JN, Jin T. Hepatic function of glucagon-like peptide-1 and its based diabetes drugs. Med Rev (2021) 2024;4(4):312-325 View Article PubMed/NCBI
  135. 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
  136. Nicholas DA, Mbongue JC, Garcia-Pérez D, Sorensen D, Ferguson Bennit H, De Leon M, et al. Exploring the Interplay between Fatty Acids, Inflammation, and Type 2 Diabetes. Immuno 2024;4(1):91-107 View Article PubMed/NCBI
  137. Vilas-Boas EA, Almeida DC, Roma LP, Ortis F, Carpinelli AR. Lipotoxicity and β-Cell Failure in Type 2 Diabetes: Oxidative Stress Linked to NADPH Oxidase and ER Stress. Cells 2021;10(12):3328 View Article PubMed/NCBI
  138. Du H, Li D, Molive LM, Wu N. Advances in free fatty acid profiles in gestational diabetes mellitus. J Transl Med 2024;22(1):180 View Article PubMed/NCBI
  139. Lv C, Sun Y, Zhang ZY, Aboelela Z, Qiu X, Meng ZX. β-cell dynamics in type 2 diabetes and in dietary and exercise interventions. J Mol Cell Biol 2022;14(7):mjac046 View Article PubMed/NCBI
  140. Elkanawati RY, Sumiwi SA, Levita J. Impact of Lipids on Insulin Resistance: Insights from Human and Animal Studies. Drug Des Devel Ther 2024;18:3337-3360 View Article PubMed/NCBI
  141. Shiri H, Fallah H, Abolhassani M, Fooladi S, Ramezani Karim Z, Danesh B, et al. Relationship between types and levels of free fatty acids, peripheral insulin resistance, and oxidative stress in T2DM: A case-control study. PLoS One 2024;19(8):e0306977 View Article PubMed/NCBI
  142. Lee SH, Park SY, Choi CS. Insulin Resistance: From Mechanisms to Therapeutic Strategies. Diabetes Metab J 2022;46(1):15-37 View Article PubMed/NCBI
  143. An SM, Cho SH, Yoon JC. Adipose Tissue and Metabolic Health. Diabetes Metab J 2023;47(5):595-611 View Article PubMed/NCBI
  144. Veluthakal R, Esparza D, Hoolachan JM, Balakrishnan R, Ahn M, Oh E, et al. Mitochondrial Dysfunction, Oxidative Stress, and Inter-Organ Miscommunications in T2D Progression. Int J Mol Sci 2024;25(3):1504 View Article PubMed/NCBI
  145. Xu L, Yang Q, Zhou J. Mechanisms of Abnormal Lipid Metabolism in the Pathogenesis of Disease. Int J Mol Sci 2024;25(15):8465 View Article PubMed/NCBI
  146. Chueire VB, Muscelli E. Effect of free fatty acids on insulin secretion, insulin sensitivity and incretin effect - a narrative review. Arch Endocrinol Metab 2021;65(1):24-31 View Article PubMed/NCBI
  147. Dilworth L, Facey A, Omoruyi F. Diabetes Mellitus and Its Metabolic Complications: The Role of Adipose Tissues. Int J Mol Sci 2021;22(14):7644 View Article PubMed/NCBI
  148. Congur I, Mingrone G, Guan K. Targeting endoplasmic reticulum stress as a potential therapeutic strategy for diabetic cardiomyopathy. Metabolism 2025;162:156062 View Article PubMed/NCBI
  149. Berardo C, Di Pasqua LG, Cagna M, Richelmi P, Vairetti M, Ferrigno A. Nonalcoholic Fatty Liver Disease and Non-Alcoholic Steatohepatitis: Current Issues and Future Perspectives in Preclinical and Clinical Research. Int J Mol Sci 2020;21(24):9646 View Article PubMed/NCBI
  150. Petagine L, Zariwala MG, Patel VB. Non-alcoholic fatty liver disease: Immunological mechanisms and current treatments. World J Gastroenterol 2023;29(32):4831-4850 View Article PubMed/NCBI
  151. Marušić M, Paić M, Knobloch M, Liberati Pršo AM. NAFLD, Insulin Resistance, and Diabetes Mellitus Type 2. Can J Gastroenterol Hepatol 2021;2021:6613827 View Article PubMed/NCBI
  152. Rafaqat S, Gluscevic S, Mercantepe F, Rafaqat S, Klisic A. Interleukins: Pathogenesis in Non-Alcoholic Fatty Liver Disease. Metabolites 2024;14(3):153 View Article PubMed/NCBI
  153. Wang K, Huang H, Zhan Q, Ding H, Li Y. Toll-like receptors in health and disease. MedComm (2020) 2024;5(5):e549 View Article PubMed/NCBI
  154. Xu GX, Wei S, Yu C, Zhao SQ, Yang WJ, Feng YH, et al. Activation of Kupffer cells in NAFLD and NASH: mechanisms and therapeutic interventions. Front Cell Dev Biol 2023;11:1199519 View Article PubMed/NCBI
  155. Habibullah M, Jemmieh K, Ouda A, Haider MZ, Malki MI, Elzouki AN. Metabolic-associated fatty liver disease: a selective review of pathogenesis, diagnostic approaches, and therapeutic strategies. Front Med (Lausanne) 2024;11:1291501 View Article PubMed/NCBI
  156. Sakers A, De Siqueira MK, Seale P, Villanueva CJ. Adipose-tissue plasticity in health and disease. Cell 2022;185(3):419-446 View Article PubMed/NCBI
  157. Zhang X, Xiao J, Jiang M, Phillips CJC, Shi B. Thermogenesis and Energy Metabolism in Brown Adipose Tissue in Animals Experiencing Cold Stress. Int J Mol Sci 2025;26(7):3233 View Article PubMed/NCBI
  158. Tang C, Wang Y, Chen D, Zhang M, Xu J, Xu C, et al. Natural polysaccharides protect against diet-induced obesity by improving lipid metabolism and regulating the immune system. Food Res Int 2023;172:113192 View Article PubMed/NCBI
  159. Zhao T, Wang C, Liu Y, Li B, Shao M, Zhao W, et al. The role of polysaccharides in immune regulation through gut microbiota: mechanisms and implications. Front Immunol 2025;16:1555414 View Article PubMed/NCBI
  160. Hu T, Liu CH, Lei M, Zeng Q, Li L, Tang H, et al. Metabolic regulation of the immune system in health and diseases: mechanisms and interventions. Signal Transduct Target Ther 2024;9(1):268 View Article PubMed/NCBI
  161. Schleh MW, Caslin HL, Garcia JN, Mashayekhi M, Srivastava G, Bradley AB, et al. Metaflammation in obesity and its therapeutic targeting. Sci Transl Med 2023;15(723):eadf9382 View Article PubMed/NCBI
  162. Merz KE, Thurmond DC. Role of Skeletal Muscle in Insulin Resistance and Glucose Uptake. Compr Physiol 2020;10(3):785-809 View Article PubMed/NCBI
  163. van Gerwen J, Shun-Shion AS, Fazakerley DJ. Insulin signalling and GLUT4 trafficking in insulin resistance. Biochem Soc Trans 2023;51(3):1057-1069 View Article PubMed/NCBI
  164. Chandrasekaran P, Weiskirchen R. The Role of Obesity in Type 2 Diabetes Mellitus-An Overview. Int J Mol Sci 2024;25(3):1882 View Article PubMed/NCBI
  165. Yazıcı D, Demir SÇ, Sezer H. Insulin Resistance, Obesity, and Lipotoxicity. Adv Exp Med Biol 2024;1460:391-430 View Article PubMed/NCBI
  166. Jun L, Tao YX, Geetha T, Babu JR. Mitochondrial Adaptation in Skeletal Muscle: Impact of Obesity, Caloric Restriction, and Dietary Compounds. Curr Nutr Rep 2024;13(3):500-515 View Article PubMed/NCBI
  167. Brown M, Dainty S, Strudwick N, Mihai AD, Watson JN, Dendooven R, et al. Endoplasmic reticulum stress causes insulin resistance by inhibiting delivery of newly synthesized insulin receptors to the cell surface. Mol Biol Cell 2020;31(23):2597-2629 View Article PubMed/NCBI
  168. Massimino E, Izzo A, Riccardi G, Della Pepa G. The Impact of Glucose-Lowering Drugs on Sarcopenia in Type 2 Diabetes: Current Evidence and Underlying Mechanisms. Cells 2021;10(8):1958 View Article PubMed/NCBI
  169. Capozzi ME, D’Alessio DA, Campbell JE. The past, present, and future physiology and pharmacology of glucagon. Cell Metab 2022;34(11):1654-1674 View Article PubMed/NCBI
  170. 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
  171. He Z, Liu Q, Wang Y, Zhao B, Zhang L, Yang X, et al. The role of endoplasmic reticulum stress in type 2 diabetes mellitus mechanisms and impact on islet function. PeerJ 2025;13:e19192 View Article PubMed/NCBI
  172. Grubelnik V, Zmazek J, Markovič R, Gosak M, Marhl M. Mitochondrial Dysfunction in Pancreatic Alpha and Beta Cells Associated with Type 2 Diabetes Mellitus. Life (Basel) 2020;10(12):348 View Article PubMed/NCBI
  173. 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
  174. Saleh M, Gittes GK, Prasadan K. Alpha-to-beta cell trans-differentiation for treatment of diabetes. Biochem Soc Trans 2021;49(6):2539-2548 View Article PubMed/NCBI
  175. Parvathareddy VP, Wu J, Thomas SS. Insulin Resistance and Insulin Handling in Chronic Kidney Disease. Compr Physiol 2023;13(4):5069-5076 View Article PubMed/NCBI
  176. Ahwin P, Martinez D. The relationship between SGLT2 and systemic blood pressure regulation. Hypertens Res 2024;47(8):2094-2103 View Article PubMed/NCBI
  177. Varra FN, Varras M, Varra VK, Theodosis-Nobelos P. Molecular and pathophysiological relationship between obesity and chronic inflammation in the manifestation of metabolic dysfunctions and their inflammation-mediating treatment options (Review). Mol Med Rep 2024;29(6):95 View Article PubMed/NCBI
  178. Yang Y, Liu J, Shi Q, Guo B, Jia H, Yang Y, et al. Roles of Mitochondrial Dysfunction in Diabetic Kidney Disease: New Perspectives from Mechanism to Therapy. Biomolecules 2024;14(6):733 View Article PubMed/NCBI
  179. Feng A, Yin R, Xu R, Zhang B, Yang L. An update on renal tubular injury as related to glycolipid metabolism in diabetic kidney disease. Front Pharmacol 2025;16:1559026 View Article PubMed/NCBI
  180. Di Costanzo A, Esposito G, Indolfi C, Spaccarotella CAM. SGLT2 Inhibitors: A New Therapeutical Strategy to Improve Clinical Outcomes in Patients with Chronic Kidney Diseases. Int J Mol Sci 2023;24(10):8732 View Article PubMed/NCBI
  181. Elendu C, Amaechi DC, Elendu TC, Amaechi EC, Elendu ID. Dependable approaches to hypertension management: A review. Medicine (Baltimore) 2024;103(24):e38560 View Article PubMed/NCBI
  182. Xing D, Zhou Q, Wang Y, Xu J. Effects of Tauroursodeoxycholic Acid and 4-Phenylbutyric Acid on Selenium Distribution in Mice Model with Type 1 Diabetes. Biol Trace Elem Res 2023;201(3):1205-1213 View Article PubMed/NCBI
  183. Boer GA, Holst JJ. Incretin Hormones and Type 2 Diabetes-Mechanistic Insights and Therapeutic Approaches. Biology (Basel) 2020;9(12):473 View Article PubMed/NCBI
  184. Alcaino C, Reimann F, Gribble FM. Incretin hormones and obesity. J Physiol 2024 View Article PubMed/NCBI
  185. Drucker DJ, Holst JJ. The expanding incretin universe: from basic biology to clinical translation. Diabetologia 2023;66(10):1765-1779 View Article PubMed/NCBI
  186. Reed J, Bain SC, Kanamarlapudi V. The Regulation of Metabolic Homeostasis by Incretins and the Metabolic Hormones Produced by Pancreatic Islets. Diabetes Metab Syndr Obes 2024;17:2419-2456 View Article PubMed/NCBI
  187. Xu X, Pang Y, Fan X. Mitochondria in oxidative stress, inflammation and aging: from mechanisms to therapeutic advances. Signal Transduct Target Ther 2025;10(1):190 View Article PubMed/NCBI
  188. Dai M, Dai S, Gu L, Xiang Z, Xu A, Lu S, et al. Efficacy of Glucagon-like Peptide-1 Receptor Agonists in Overweight/Obese and/or T2DM Adolescents: A Meta-analysis Based on Randomized Controlled Trials. J Clin Res Pediatr Endocrinol 2024;16(3):323-333 View Article PubMed/NCBI
  189. Al-Sayyar A, Hammad MM, Williams MR, Al-Onaizi M, Abubaker J, Alzaid F. Neurotransmitters in Type 2 Diabetes and the Control of Systemic and Central Energy Balance. Metabolites 2023;13(3):384 View Article PubMed/NCBI
  190. van Galen KA, Ter Horst KW, Serlie MJ. Serotonin, food intake, and obesity. Obes Rev 2021;22(7):e13210 View Article PubMed/NCBI
  191. Bao Y, Chen X, Li Y, Yuan S, Han L, Deng X, et al. Chronic Low-Grade Inflammation and Brain Structure in the Middle-Aged and Elderly Adults. Nutrients 2024;16(14):2313 View Article PubMed/NCBI
  192. Dash UC, Bhol NK, Swain SK, Samal RR, Nayak PK, Raina V, et al. Oxidative stress and inflammation in the pathogenesis of neurological disorders: Mechanisms and implications. Acta Pharm Sin B 2025;15(1):15-34 View Article PubMed/NCBI
  193. Freyberg Z, Andreazza AC, McClung CA, Phillips ML. Linking Mitochondrial Dysfunction, Neurotransmitter, and Neural Network Abnormalities and Mania: Elucidating Neurobiological Mechanisms of the Therapeutic Effect of the Ketogenic Diet in Bipolar Disorder. Biol Psychiatry Cogn Neurosci Neuroimaging 2025;10(3):267-277 View Article PubMed/NCBI
  194. Shchaslyvyi AY, Antonenko SV, Telegeev GD. Comprehensive Review of Chronic Stress Pathways and the Efficacy of Behavioral Stress Reduction Programs (BSRPs) in Managing Diseases. Int J Environ Res Public Health 2024;21(8):1077 View Article PubMed/NCBI
  195. Adamu A, Li S, Gao F, Xue G. The role of neuroinflammation in neurodegenerative diseases: current understanding and future therapeutic targets. Front Aging Neurosci 2024;16:1347987 View Article PubMed/NCBI
  196. Klemmensen MM, Borrowman SH, Pearce C, Pyles B, Chandra B. Mitochondrial dysfunction in neurodegenerative disorders. Neurotherapeutics 2024;21(1):e00292 View Article PubMed/NCBI
  197. Navakkode S, Kennedy BK. Neural ageing and synaptic plasticity: prioritizing brain health in healthy longevity. Front Aging Neurosci 2024;16:1428244 View Article PubMed/NCBI
  198. Marzola P, Melzer T, Pavesi E, Gil-Mohapel J, Brocardo PS. Exploring the Role of Neuroplasticity in Development, Aging, and Neurodegeneration. Brain Sci 2023;13(12):1610 View Article PubMed/NCBI
  199. Lazarov O, Minshall RD, Bonini MG. Harnessing neurogenesis in the adult brain-A role in type 2 diabetes mellitus and Alzheimer’s disease. Int Rev Neurobiol 2020;155:235-269 View Article PubMed/NCBI
  200. Wendimu MY, Hooks SB. Microglia Phenotypes in Aging and Neurodegenerative Diseases. Cells 2022;11(13):2091 View Article PubMed/NCBI
  201. Cieri MB, Ramos AJ. Astrocytes, reactive astrogliosis, and glial scar formation in traumatic brain injury. Neural Regen Res 2025;20(4):973-989 View Article PubMed/NCBI
  202. Chen T, Dai Y, Hu C, Lin Z, Wang S, Yang J, et al. Cellular and molecular mechanisms of the blood-brain barrier dysfunction in neurodegenerative diseases. Fluids Barriers CNS 2024;21(1):60 View Article PubMed/NCBI
  203. Cáceres E, Olivella JC, Di Napoli M, Raihane AS, Divani AA. Immune Response in Traumatic Brain Injury. Curr Neurol Neurosci Rep 2024;24(12):593-609 View Article PubMed/NCBI
  204. Alexaki VI. The Impact of Obesity on Microglial Function: Immune, Metabolic and Endocrine Perspectives. Cells 2021;10(7):1584 View Article PubMed/NCBI
  205. Mayer MG, Fischer T. Microglia at the blood brain barrier in health and disease. Front Cell Neurosci 2024;18:1360195 View Article PubMed/NCBI
  206. Hwang YK, Oh JS. Interaction of the Vagus Nerve and Serotonin in the Gut-Brain Axis. Int J Mol Sci 2025;26(3):1160 View Article PubMed/NCBI
  207. Chen Q, Wu M, Tang Q, Yan P, Zhu L. Age-Related Alterations in Immune Function and Inflammation: Focus on Ischemic Stroke. Aging Dis 2024;15(3):1046-1074 View Article PubMed/NCBI
  208. Weyand CM, Goronzy JJ. Aging of the Immune System. Mechanisms and Therapeutic Targets. Ann Am Thorac Soc 2016;13(Suppl 5):S422-S428 View Article PubMed/NCBI
  209. Sun R, Feng J, Wang J. Underlying Mechanisms and Treatment of Cellular Senescence-Induced Biological Barrier Interruption and Related Diseases. Aging Dis 2024;15(2):612-639 View Article PubMed/NCBI
  210. Kumar M, Yan P, Kuchel GA, Xu M. Cellular Senescence as a Targetable Risk Factor for Cardiovascular Diseases: Therapeutic Implications: JACC Family Series. JACC Basic Transl Sci 2024;9(4):522-534 View Article PubMed/NCBI
  211. Qin Y, Liu H, Wu H. Cellular Senescence in Health, Disease, and Lens Aging. Pharmaceuticals (Basel) 2025;18(2):244 View Article PubMed/NCBI
  212. Rahimpour S, Clary BL, Nasoohi S, Berhanu YS, Brown CM. Immunometabolism In Brain Aging and Neurodegeneration: Bridging Metabolic Pathways and Immune Responses. Aging Dis 2024 View Article PubMed/NCBI
  213. Chen C, Dong X, Zhang W, Chang X, Gao W. Dialogue between mitochondria and endoplasmic reticulum-potential therapeutic targets for age-related cardiovascular diseases. Front Pharmacol 2024;15:1389202 View Article PubMed/NCBI
  214. Sha W, Hu F, Bu S. Mitochondrial dysfunction and pancreatic islet β-cell failure (Review). Exp Ther Med 2020;20(6):266 View Article PubMed/NCBI
  215. Weiser A, Feige JN, De Marchi U. Mitochondrial Calcium Signaling in Pancreatic β-Cell. Int J Mol Sci 2021;22(5):2515 View Article PubMed/NCBI
  216. Zhou Z, Arroum T, Luo X, Kang R, Lee YJ, Tang D, et al. Diverse functions of cytochrome c in cell death and disease. Cell Death Differ 2024;31(4):387-404 View Article PubMed/NCBI
  217. Wang L, Li L, Liu J, Sheng C, Yang M, Hu Z, et al. Associated factors and principal pathophysiological mechanisms of type 2 diabetes mellitus. Front Endocrinol (Lausanne) 2025;16:1499565 View Article PubMed/NCBI
  218. Rhea EM, Banks WA, Raber J. Insulin Resistance in Peripheral Tissues and the Brain: A Tale of Two Sites. Biomedicines 2022;10(7):1582 View Article PubMed/NCBI
  219. Chaurasia B, Summers SA. Ceramides in Metabolism: Key Lipotoxic Players. Annu Rev Physiol 2021;83:303-330 View Article PubMed/NCBI
  220. Ma K, Zhang Y, Zhao J, Zhou L, Li M. Endoplasmic reticulum stress: bridging inflammation and obesity-associated adipose tissue. Front Immunol 2024;15:1381227 View Article PubMed/NCBI
  221. Tsalamandris S, Antonopoulos AS, Oikonomou E, Papamikroulis GA, Vogiatzi G, Papaioannou S, et al. The Role of Inflammation in Diabetes: Current Concepts and Future Perspectives. Eur Cardiol 2019;14(1):50-59 View Article PubMed/NCBI
  222. Zhao X, Zhang G, Wu L, Tang Y, Guo C. Inhibition of ER stress-activated JNK pathway attenuates TNF-α-induced inflammatory response in bone marrow mesenchymal stem cells. Biochem Biophys Res Commun 2021;541:8-14 View Article PubMed/NCBI
  223. Yung JHM, Giacca A. Role of c-Jun N-terminal Kinase (JNK) in Obesity and Type 2 Diabetes. Cells 2020;9(3):706 View Article PubMed/NCBI
  224. de la Monte SM. Conquering Insulin Network Dysfunctions in Alzheimer’s Disease: Where Are We Today?. J Alzheimers Dis 2024;101(s1):S317-S343 View Article PubMed/NCBI
  225. Hemat Jouy S, Mohan S, Scichilone G, Mostafa A, Mahmoud AM. Adipokines in the Crosstalk between Adipose Tissues and Other Organs: Implications in Cardiometabolic Diseases. Biomedicines 2024;12(9):2129 View Article PubMed/NCBI
  226. Cicuéndez B, Ruiz-Garrido I, Mora A, Sabio G. Stress kinases in the development of liver steatosis and hepatocellular carcinoma. Mol Metab 2021;50:101190 View Article PubMed/NCBI
  227. Fu F, Doroudgar S. IRE1/XBP1 and endoplasmic reticulum signaling - from basic to translational research for cardiovascular disease. Curr Opin Physiol 2022;28:100552 View Article PubMed/NCBI
  228. Yan H, He L, Lv D, Yang J, Yuan Z. The Role of the Dysregulated JNK Signaling Pathway in the Pathogenesis of Human Diseases and Its Potential Therapeutic Strategies: A Comprehensive Review. Biomolecules 2024;14(2):243 View Article PubMed/NCBI
  229. Ni L, Yang L, Lin Y. Recent progress of endoplasmic reticulum stress in the mechanism of atherosclerosis. Front Cardiovasc Med 2024;11:1413441 View Article PubMed/NCBI
  230. Mustapha S, Mohammed M, Azemi AK, Jatau AI, Shehu A, Mustapha L, et al. Current Status of Endoplasmic Reticulum Stress in Type II Diabetes. Molecules 2021;26(14):4362 View Article PubMed/NCBI
  231. Guo Z, Chi R, Peng Y, Sun K, Liu H, Guo F, et al. The Role and Interactive Mechanism of Endoplasmic Reticulum Stress and Ferroptosis in Musculoskeletal Disorders. Biomolecules 2024;14(11):1369 View Article PubMed/NCBI
  232. Guria S, Hoory A, Das S, Chattopadhyay D, Mukherjee S. Adipose tissue macrophages and their role in obesity-associated insulin resistance: an overview of the complex dynamics at play. Biosci Rep 2023;43(3):BSR20220200 View Article PubMed/NCBI
  233. Guo Q, Jin Y, Chen X, Ye X, Shen X, Lin M, et al. NF-κB in biology and targeted therapy: new insights and translational implications. Signal Transduct Target Ther 2024;9(1):53 View Article PubMed/NCBI
  234. Heida A, Gruben N, Catrysse L, Koehorst M, Koster M, Kloosterhuis NJ, et al. The hepatocyte IKK:NF-κB axis promotes liver steatosis by stimulating de novo lipogenesis and cholesterol synthesis. Mol Metab 2021;54:101349 View Article PubMed/NCBI
  235. Pei J, Wang B, Wang D. Current Studies on Molecular Mechanisms of Insulin Resistance. J Diabetes Res 2022;2022:1863429 View Article PubMed/NCBI
  236. D’Angelo D, Rizzuto R. The Mitochondrial Calcium Uniporter (MCU): Molecular Identity and Role in Human Diseases. Biomolecules 2023;13(9):1304 View Article PubMed/NCBI
  237. Muthu S, Tran Z, Thilagavathi J, Bolarum T, Azzam EI, Suzuki CK, et al. Aging triggers mitochondrial, endoplasmic reticulum, and metabolic stress responses in the heart. J Cardiovasc Aging 2025;5(1):4 View Article PubMed/NCBI
  238. Varughese JT, Buchanan SK, Pitt AS. The Role of Voltage-Dependent Anion Channel in Mitochondrial Dysfunction and Human Disease. Cells 2021;10(7):1737 View Article PubMed/NCBI
  239. Rui L. Energy metabolism in the liver. Compr Physiol 2014;4(1):177-197 View Article PubMed/NCBI
  240. Dejos C, Gkika D, Cantelmo AR. The Two-Way Relationship Between Calcium and Metabolism in Cancer. Front Cell Dev Biol 2020;8:573747 View Article PubMed/NCBI
  241. Tsuno S, Harada K, Horikoshi M, Mita M, Kitaguchi T, Hirai MY, et al. Mitochondrial ATP concentration decreases immediately after glucose administration to glucose-deprived hepatocytes. FEBS Open Bio 2024;14(1):79-95 View Article PubMed/NCBI
  242. Liu QK. Mechanisms of action and therapeutic applications of GLP-1 and dual GIP/GLP-1 receptor agonists. Front Endocrinol (Lausanne) 2024;15:1431292 View Article PubMed/NCBI
  243. Luna-Marco C, de Marañon AM, Hermo-Argibay A, Rodriguez-Hernandez Y, Hermenejildo J, Fernandez-Reyes M, et al. Effects of GLP-1 receptor agonists on mitochondrial function, inflammatory markers and leukocyte-endothelium interactions in type 2 diabetes. Redox Biol 2023;66:102849 View Article PubMed/NCBI
  244. Holst JJ, Gasbjerg LS, Rosenkilde MM. The Role of Incretins on Insulin Function and Glucose Homeostasis. Endocrinology 2021;162(7):bqab065 View Article PubMed/NCBI
  245. Młynarska E, Czarnik W, Dzieża N, Jędraszak W, Majchrowicz G, Prusinowski F, et al. Type 2 Diabetes Mellitus: New Pathogenetic Mechanisms, Treatment and the Most Important Complications. Int J Mol Sci 2025;26(3):1094 View Article PubMed/NCBI
  246. Acreman S, Ma J, Denwood G, Gao R, Tarasov A, Rorsman P, et al. The endoplasmic reticulum plays a key role in α-cell intracellular Ca(2+) dynamics and glucose-regulated glucagon secretion in mouse islets. iScience 2024;27(5):109665 View Article PubMed/NCBI
  247. Vallon V, Nakagawa T. Renal Tubular Handling of Glucose and Fructose in Health and Disease. Compr Physiol 2021;12(1):2995-3044 View Article PubMed/NCBI
  248. Teleanu RI, Niculescu AG, Roza E, Vladâcenco O, Grumezescu AM, Teleanu DM. Neurotransmitters-Key Factors in Neurological and Neurodegenerative Disorders of the Central Nervous System. Int J Mol Sci 2022;23(11):5954 View Article PubMed/NCBI
  249. Pan S, Worker CJ, Feng Earley Y. The hypothalamus as a key regulator of glucose homeostasis: emerging roles of the brain renin-angiotensin system. Am J Physiol Cell Physiol 2023;325(1):C141-C154 View Article PubMed/NCBI
  250. Casado ME, Collado-Pérez R, Frago LM, Barrios V. Recent Advances in the Knowledge of the Mechanisms of Leptin Physiology and Actions in Neurological and Metabolic Pathologies. Int J Mol Sci 2023;24(2):1422 View Article PubMed/NCBI
  251. Caturano A, D’Angelo M, Mormone A, Russo V, Mollica MP, Salvatore T, et al. Oxidative Stress in Type 2 Diabetes: Impacts from Pathogenesis to Lifestyle Modifications. Curr Issues Mol Biol 2023;45(8):6651-6666 View Article PubMed/NCBI
  252. Szabo L, Lejri I, Grimm A, Eckert A. Spermidine Enhances Mitochondrial Bioenergetics in Young and Aged Human-Induced Pluripotent Stem Cell-Derived Neurons. Antioxidants (Basel) 2024;13(12):1482 View Article PubMed/NCBI
  253. Shaito A, Al-Mansoob M, Ahmad SMS, Haider MZ, Eid AH, Posadino AM, et al. Resveratrol-Mediated Regulation of Mitochondria Biogenesis-associated Pathways in Neurodegenerative Diseases: Molecular Insights and Potential Therapeutic Applications. Curr Neuropharmacol 2023;21(5):1184-1201 View Article PubMed/NCBI
  254. Yu W, Yu Y, Sun S, Lu C, Zhai J, Lei Y, et al. Immune Alterations with Aging: Mechanisms and Intervention Strategies. Nutrients 2024;16(22):3830 View Article PubMed/NCBI
  255. Wang S, Huo T, Lu M, Zhao Y, Zhang J, He W, et al. Recent Advances in Aging and Immunosenescence: Mechanisms and Therapeutic Strategies. Cells 2025;14(7):499 View Article PubMed/NCBI
  256. Cheishvili D, Do Carmo S, Caraci F, Grasso M, Cuello AC, Szyf M. EpiAge: a next-generation sequencing-based ELOVL2 epigenetic clock for biological age assessment in saliva and blood across health and disease. Aging (Albany NY) 2025;17(1):131-160 View Article PubMed/NCBI
  257. Tomusiak A, Floro A, Tiwari R, Riley R, Matsui H, Andrews N, et al. Development of an epigenetic clock resistant to changes in immune cell composition. Commun Biol 2024;7(1):934 View Article PubMed/NCBI

About this Article

Cite this article
Chakrabarti SK, Chattopadhyay D. The Link Between Immune Aging and Type 2 Diabetes: A Review of Mechanisms and Implications. Explor Res Hypothesis Med. Published online: Jul 1, 2025. doi: 10.14218/ERHM.2025.00018.
Copy        Export to RIS        Export to EndNote
Article History
Received Revised Accepted Published
March 26, 2025 May 5, 2025 May 20, 2025 July 1, 2025
DOI http://dx.doi.org/10.14218/ERHM.2025.00018
  • Exploratory Research and Hypothesis in Medicine
  • pISSN 2993-5113
  • eISSN 2472-0712
Back to Top

The Link Between Immune Aging and Type 2 Diabetes: A Review of Mechanisms and Implications

Swarup K. Chakrabarti, Dhrubajyoti Chattopadhyay
  • Reset Zoom
  • Download TIFF