Back to Journals » ImmunoTargets and Therapy » Volume 14
Interplay Between Insulin Resistance and Immune Dysregulation in Type 2 Diabetes Mellitus: Implications for Therapeutic Interventions
Authors Berbudi A , Khairani S, Tjahjadi AI
Received 7 October 2024
Accepted for publication 20 February 2025
Published 3 April 2025 Volume 2025:14 Pages 359—382
DOI https://doi.org/10.2147/ITT.S499605
Checked for plagiarism Yes
Review by Single anonymous peer review
Peer reviewer comments 2
Editor who approved publication: Professor Michael Shurin
Afiat Berbudi,1– 3 Shafia Khairani,4 Adi Imam Tjahjadi2,3,5
1Department of Biomedical Sciences, Parasitology Division, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia; 2Research Center for Care and Control of Infectious Diseases (RC3ID), Universitas Padjadjaran, Bandung, Indonesia; 3Universitas Padjadjaran Hospital, Sumedang, Indonesia; 4Department of Biomedical Sciences, Cell Biology Division, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia; 5Department of Biomedical Sciences, Microbiology Division, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
Correspondence: Afiat Berbudi, Department of Biomedical Sciences, Parasitology Division, Faculty of Medicine, Universitas Padjadjaran, Jl. Raya Bandung Sumedang Km. 21, Bandung, West Java, 45363, Indonesia, Tel +62 82297033616, Email [email protected]
Abstract: Type 2 Diabetes Mellitus (T2DM) is a rapidly growing global health issue characterized by insulin resistance and chronic inflammation. Beyond regulating glucose homeostasis, insulin plays a pivotal role in modulating immune cell function, linking metabolic dysregulation with immune responses. This review examines the intricate relationship between insulin resistance and immune dysfunction in T2DM, focusing on how impaired insulin signaling pathways, particularly PI3K/Akt and MAPK, contribute to immune cell activation, proliferation, and chronic inflammation. Insulin resistance impacts immune cells such as T cells, B cells, macrophages, and neutrophils, leading to an imbalance between pro-inflammatory and anti-inflammatory responses. Elevated pro-inflammatory cytokines (eg, TNF-α, IL-6) and adipokines (eg, leptin, resistin) exacerbate insulin resistance, promoting a vicious cycle of metabolic and immune dysregulation. This interplay contributes to the chronic low-grade inflammation that underlies T2DM pathogenesis, further impairing insulin signaling and glucose metabolism. Restoration of insulin sensitivity is, therefore, a critical step toward correcting immune imbalance in insulin-resistant states like T2DM. Therapeutic approaches that reduce inflammation could also support improvements in insulin sensitivity, addressing both metabolic and immune disturbances simultaneously. The review also explores therapeutic strategies, including insulin therapy, targeting insulin signaling pathways, and lifestyle interventions. Insulin therapy can reduce pro-inflammatory cytokine production and enhance anti-inflammatory responses, although challenges such as potential immune suppression and hyperinsulinemia remain. Targeting key signaling pathways and transcription factors offers promising avenues for modulating immune responses, while lifestyle interventions, such as dietary modifications, physical activity, and weight management, can improve insulin sensitivity and reduce inflammation. By understanding the dual role of insulin in regulating both metabolic and immune functions, this review underscores the importance of addressing immune dysfunction as part of comprehensive T2DM management. Targeting the interconnected pathways of insulin signaling and immune regulation could lead to more effective therapeutic approaches, ultimately improving patient outcomes and reducing disease complications.
Keywords: insulin resistance, type 2 diabetes mellitus, immune system dysregulation, inflammation, signal transduction, immunomodulation
Introduction
Type 2 Diabetes Mellitus (T2DM) has emerged as a major global health crisis, with its prevalence rising at an alarming rate worldwide. According to the World Health Organization (WHO) and the International Diabetes Federation (IDF), approximately 537 million individuals were affected by diabetes in 2021, with projections indicating that this figure could escalate to 783 million by 2045.1,2 This dramatic surge is particularly evident in developing countries, where lifestyle shifts, including sedentary behaviors and poor dietary choices, have accelerated the spread of T2DM.3,4 Consequently, T2DM has transitioned from being a disease of affluence to one of the most pressing public health challenges in low- and middle-income nations.
The escalating prevalence of T2DM is closely linked to the rise in obesity rates, which serve as a major risk factor for insulin resistance and the subsequent development of diabetes.5,6 As obesity reaches epidemic proportions worldwide, it contributes significantly to the burgeoning rates of T2DM. For instance, data indicate that the number of people living with T2DM increased dramatically from 382 million in 2013 to 592 million by 2035, highlighting the interplay between obesity and diabetes.2,7 This correlation underscores the importance of addressing obesity through targeted public health interventions to mitigate the rising incidence of T2DM.
Compounding the complexity of T2DM are its associated complications, which include cardiovascular disease, kidney disease, and neuropathy, substantially worsening the disease’s prognosis and increasing healthcare burdens.8–10 The economic implications of T2DM are significant, with global diabetes-related healthcare expenditures estimated to have reached $376 billion in 2010 alone.11 This financial burden emphasizes the necessity for comprehensive strategies focusing on both prevention and effective management of T2DM to curb its impact on healthcare systems.
Therefore, the rising prevalence of T2DM represents not only a clinical concern but also an urgent global public health challenge that demands a multifaceted response. Addressing this issue requires lifestyle interventions, public health policies, and increased awareness to effectively manage and reduce the risk factors and complications associated with T2DM.
Insulin plays a pivotal role in the regulation of glucose homeostasis, acting as a key hormone synthesized by pancreatic β-cells that facilitates glucose uptake by tissues such as skeletal muscle and adipose tissue.12,13 In T2DM, however, insulin resistance impairs this regulatory mechanism, resulting in chronic hyperglycemia and further metabolic dysregulation. Beyond its classical role in glucose metabolism, insulin has been identified as a modulator of immune cell function, linking metabolic dysregulation with inflammation.14 The intricate interplay between insulin signaling and immune cell activation suggests a bidirectional relationship between metabolic and immune pathways, which contributes to the pathogenesis of T2DM.
Recent studies have elucidated that insulin resistance is closely intertwined with immune dysfunction, which plays a significant role in the chronic low-grade inflammation observed in T2DM.15 This inflammation is mediated by immune cells such as macrophages, which adopt a pro-inflammatory phenotype and secrete cytokines that further impair insulin signaling.16,17 Additionally, insulin resistance in immune cells themselves perpetuates the inflammatory state, emphasizing the complex interactions between insulin action and immune responses in T2DM.10,18
The immune dysfunction associated with T2DM involves both innate and adaptive immune responses, with macrophages, T helper 17 (Th17) cells, and regulatory T cells (Tregs) playing prominent roles. Macrophages in adipose tissue contribute to insulin resistance by releasing pro-inflammatory cytokines, while the imbalance between pro-inflammatory Th17 cells and anti-inflammatory Tregs exacerbates chronic inflammation.19 This suggests that targeting immune cell activity and their modulation by insulin could offer promising therapeutic strategies for T2DM.
Understanding the role of insulin in modulating immune cell function in T2DM is crucial for comprehending the multifaceted nature of this disease. This narrative review seeks to explore the interrelationship between insulin signaling and immune cell function in T2DM, focusing on how insulin resistance contributes to immune dysregulation and chronic inflammation. By elucidating these pathways, we aim to highlight potential therapeutic targets that could enhance the management of T2DM and its associated complications.
Inflammation, Insulin Resistance and Beta-Cell Failure
Excess adipose tissue in metabolically unhealthy obesity triggers chronic low-grade inflammation, leading to insulin resistance and beta-cell dysfunction. Visceral fat releases pro-inflammatory cytokines like TNF-α, IL-6, and MCP-1, fueling systemic inflammation that disrupts metabolic processes and impairs insulin signaling.20 Specifically, TNF-α interferes with insulin receptor substrate (IRS) signaling, reducing glucose uptake, while elevated free fatty acids in obesity exacerbate inflammation and further impair insulin sensitivity.20
This persistent inflammation damages pancreatic beta cells, with inflammatory cytokines inducing apoptosis and reducing insulin secretion. Immune cell infiltration, particularly macrophages in the islets, elevates pro-inflammatory cytokine levels, aggravating beta-cell dysfunction and contributing to insulin resistance and hyperglycemia.10
Endoplasmic reticulum (ER) stress also plays a critical role. The increased demand for insulin in obesity strains the ER, initiating the unfolded protein response (UPR). Prolonged ER stress, when the UPR fails to adapt, leads to beta-cell apoptosis and progresses toward type 2 diabetes.21 Additionally, oxidative stress in obesity exacerbates inflammation and insulin resistance, fueled by reactive oxygen species (ROS) and dysregulated adipokine secretion, creating a vicious cycle of cellular damage.22,23
Mechanisms of Insulin Signaling in Immune Cells
Insulin Receptor Expression on Immune Cells
Insulin receptors (IRs) are present on various immune cells, such as T cells, B cells, macrophages, and dendritic cells, and play a crucial role in modulating immune responses.24 These receptors facilitate insulin signaling, impacting immune cell proliferation, differentiation, and cytokine production. For instance, T cells rely on insulin signaling for activation and glucose uptake, while macrophages enhance inflammatory responses via insulin receptor engagement.25 Dendritic cells use insulin receptors to influence antigen presentation, underscoring insulin’s role in immune homeostasis.
In T2DM, insulin receptor expression and functionality on immune cells are impaired, contributing to chronic inflammation and metabolic dysregulation.26 This impairment is driven by factors such as elevated pro-inflammatory cytokines (eg, TNF-α, IL-6) that downregulate insulin receptor expression, promoting insulin resistance.27 Consequently, immune cells, like macrophages, shift toward a pro-inflammatory M1 phenotype, perpetuating the inflammatory state typical of T2DM.24
Metabolic factors, including obesity and hyperglycemia, also impact insulin receptor levels. Excess fatty acids activate stress pathways (eg, JNK, IKKβ), impairing insulin receptor expression and signaling.28 Conversely, insulin itself can upregulate its receptor on immune cells, suggesting adaptability to metabolic challenges.29 Transcription factors, such as FOXO, further regulate insulin receptor expression, with activation leading to downregulation, contributing to insulin resistance.30
The expression of insulin receptors on immune cells is clearly shaped by various factors, including cytokines, metabolic signals, and transcriptional activity. Understanding these dynamics offers valuable insight into how immune dysfunction and insulin resistance interplay in T2DM.10,26,31
Insulin Signaling Pathways in Immune Cells
Insulin signaling plays a crucial role in immune cell activation, proliferation, and inflammation regulation, primarily through the PI3K/Akt and MAPK pathways32 (Figure 1). In macrophages, insulin binding activates the IRS/PI3K/Akt pathway, which is essential for glucose uptake and inflammatory response modulation. This pathway promotes the anti-inflammatory M2 phenotype, but in T2DM, insulin resistance disrupts it, leading to an increase in the pro-inflammatory M1 macrophages, contributing to chronic inflammation.33
![]() |
Figure 1 Mechanisms of Adipose Tissue Inflammation and Insulin Resistance Mediated by TLR4 Activation and Adipokines. Adipose tissue in obesity secretes pro-inflammatory cytokines (eg, TNF-α, IL-6, IL-1β) and adipokines (eg, leptin, resistin, chemerin), which activate Toll-like receptor 4 (TLR4) signaling. TLR4 is also activated by damage-associated molecular patterns (DAMPs), hypoxia-inducible factor 1α (HIF-1α), and excess free fatty acids (FFAs), leading to downstream activation of JNK1 and IκB kinase β (IKKβ). This signaling pathway activates nuclear factor kappa B (NF-κB) in immune cells, inducing pro-inflammatory cytokine synthesis. Simultaneously, reactive oxygen species (ROS) generated by these processes amplify the inflammatory response. Chronic inflammation interferes with insulin receptor signaling by suppressing IRS1/IRS2 via the induction of suppressor of cytokine signaling (SOCS) proteins. This disruption inhibits the PI3K/Akt pathway, impairing GLUT-1 translocation and glucose uptake. These processes collectively contribute to immune cell activation, proliferation, ROS production, and inflammation, exacerbating insulin resistance and metabolic dysfunction. Created in BioRender. Berbudi, (A) (2024) https://BioRender.com/p64z950. |
Similarly, insulin signaling is vital for T cell activation and proliferation, facilitating glucose uptake and glycolysis, which are essential for T cell function.34 In T2DM, disrupted insulin signaling impairs T cell activation, contributing to immune dysfunction. For dendritic cells, insulin influences maturation and antigen presentation, impacting T cell activation and adaptive immune responses.32
The MAPK pathway, another insulin signaling route, regulates cytokine production and cell proliferation. In T2DM, insulin resistance impairs this pathway, leading to excessive inflammatory cytokine production, worsening insulin resistance and inflammation.35
Insulin signaling in immune cells differs from metabolic tissues, as it focuses more on inflammatory response regulation than glucose uptake. Immune cells have receptors like Toll-like receptor 4 (TLR4), which interact with insulin signaling pathways, linking immune activation with metabolic regulation, especially in obesity-induced insulin resistance.36 Inflammatory cytokines such as TNF-α and IL-6, elevated in T2DM, can downregulate insulin receptor signaling, promoting a pro-inflammatory state and exacerbating insulin resistance34 (Figure 1).
It is evident that the PI3K/Akt and MAPK pathways are central to immune cell regulation, influencing processes like activation, proliferation, and cytokine production. This insight underscores the importance of these pathways in developing targeted therapies for T2DM.32,35
Insulin Resistance in Immune Cells
Insulin resistance in immune cells plays a vital role in the progression of Type 2 Diabetes Mellitus (T2DM), resulting from complex interactions between inflammatory signals, metabolic changes, and cellular adaptations.37 Chronic low-grade inflammation, often associated with obesity, is a key driver. As obesity progresses, immune cells like macrophages and T cells infiltrate adipose tissue, releasing pro-inflammatory cytokines such as TNF-α and IL-6, which impair insulin signaling by disrupting insulin receptor substrates (IRS) phosphorylation, leading to insulin resistance38 (Figure 1).
In macrophages, insulin resistance promotes a shift towards a pro-inflammatory M1 phenotype, increasing the production of cytokines and reactive oxygen species (ROS), further aggravating inflammation.39 Elevated free fatty acids and lipopolysaccharides (LPS), common in obesity, activate Toll-like receptors (TLRs) on macrophages, initiating inflammatory pathways like NF-κB, which further impair insulin signaling.40 This process creates a cycle where inflammation and insulin resistance reinforce each other.
Metabolic reprogramming also plays a significant role, with immune cells undergoing increased glycolysis in response to inflammation, sustaining their pro-inflammatory activity and contributing to insulin resistance.41 CD11c+ macrophages, which produce inflammatory mediators like TNF-α and IL-1β, are particularly influential in promoting insulin resistance in surrounding tissues, linking immune dysregulation to metabolic dysfunction.42
The chronic inflammatory state driven by insulin-resistant immune cells, particularly macrophages, significantly impacts adipose, liver, and muscle tissues, leading to impaired insulin signaling and systemic inflammation. In obesity, macrophages infiltrate adipose tissue, where they adopt a pro-inflammatory M1 phenotype, secreting cytokines such as TNF-α and IL-1β that disrupt insulin receptor signaling pathways.43,44 This inflammatory milieu not only promotes insulin resistance in adipose tissue but also affects the liver and muscle, contributing to a systemic inflammatory response that exacerbates insulin resistance across these tissues.45–47 In the liver, inflammatory cytokines from activated macrophages can enhance hepatic insulin resistance by promoting gluconeogenesis and lipid accumulation, further complicating metabolic control.46,48 The interplay between adipose tissue inflammation and liver dysfunction creates a vicious cycle, where increased fatty acid release from adipose tissue exacerbates liver inflammation and insulin resistance.43,45
Macrophages also infiltrate pancreatic islets and secrete pro-inflammatory cytokines such as TNF-α and IL-1β, which impair insulin signaling pathways and promote β-cell apoptosis.49 This inflammatory milieu not only disrupts the normal function of β-cells but also leads to a reduction in β-cell mass, further exacerbating insulin resistance and hyperglycemia.50 Moreover, the accumulation of free fatty acids (FFAs) in the context of obesity contributes to oxidative stress and endoplasmic reticulum (ER) stress in β-cells, which are critical factors in the pathogenesis of β-cell dysfunction.51,52 The interplay between chronic inflammation and metabolic dysregulation creates a vicious cycle where β-cell dysfunction leads to inadequate insulin secretion, worsening hyperglycemia and promoting further inflammation.20 Thus, the chronic inflammatory response initiated by insulin-resistant macrophages is a central mechanism linking obesity to β-cell dysfunction and the progression of T2D.
Addressing insulin resistance in immune cells remains a key factor in understanding the inflammatory processes and metabolic dysregulation characteristic of T2DM. Interventions focusing on these mechanisms could be pivotal in managing the disease’s progression.53
Insulin and Immune Cell Metabolism
Insulin is vital in regulating immune cell metabolism, impacting their energy utilization and function. It modulates glucose uptake, lipid metabolism, and amino acid transport, which are essential for immune cell activation, proliferation, and differentiation.54 However, in Type 2 Diabetes Mellitus (T2DM), this regulation is disrupted, contributing to chronic inflammation and disease progression.
Insulin signaling activates the PI3K/Akt pathway in immune cells, facilitating glucose uptake via GLUT1 and enhancing glycolytic activity.14 This process provides energy for immune cell activation. In macrophages, insulin signaling boosts glucose metabolism and promotes the production of pro-inflammatory cytokines like IL-1β. In T cells, insulin signaling supports activation, proliferation, and differentiation, linking energy availability to immune response.15
In T2DM, insulin resistance impairs glucose uptake in immune cells, such as macrophages and T cells, forcing them to rely on fatty acids as an energy source.55 The shift from glucose to free fatty acids (FFAs) as a primary energy source in immune cells, particularly macrophages, promotes a pro-inflammatory state that exacerbates insulin resistance in peripheral tissues. When macrophages utilize FFAs, especially saturated fatty acids like palmitic acid, they undergo metabolic reprogramming that enhances the activation of inflammatory pathways, including the NLRP3 inflammasome. This activation leads to the production of pro-inflammatory cytokines such as IL-1β, which has been shown to inhibit insulin signaling in target tissues.45
Moreover, the accumulation of FFAs within macrophages can generate reactive oxygen species (ROS), further fueling inflammation and contributing to cellular stress responses.56 This inflammatory environment not only impairs insulin signaling in adipose tissue, liver, and muscle but also promotes a cycle of chronic inflammation that perpetuates insulin resistance.57 Additionally, the metabolic byproducts of FFA oxidation can serve as signaling molecules that modulate immune responses, further entrenching the pro-inflammatory state.58 Thus, the metabolic shift towards FFAs in immune cells is a critical mechanism linking obesity, chronic inflammation, and insulin resistance.
The imbalance of adipokines, like increased leptin and decreased adiponectin, also affects immune cell metabolism. Elevated leptin promotes pro-inflammatory responses, while reduced adiponectin impairs anti-inflammatory effects, contributing to immune dysfunction in T2DM.59 Additionally, the activation of the NLRP3 inflammasome in immune cells, triggered by metabolic stress (eg, high glucose or fatty acids), leads to increased production of IL-1β, further worsening insulin resistance and inflammation.60
Metabolic changes in hepatocytes and myocytes, particularly the shift towards increased free fatty acid (FFA) utilization, significantly worsen insulin resistance through the promotion of an inflammatory state. In hepatocytes, elevated FFAs lead to the accumulation of diacylglycerols (DAGs), which activate protein kinase C (PKC) isoforms such as PKCε. This activation inhibits insulin receptor substrate (IRS) proteins, impairing insulin signaling and promoting hepatic insulin resistance.61,62 Additionally, FFAs can induce endoplasmic reticulum (ER) stress and activate inflammatory pathways, further exacerbating insulin resistance in the liver.63
In skeletal muscle, the presence of FFAs similarly activates inflammatory signaling pathways, including the phosphorylation of STAT3, which is associated with insulin resistance.64,65 The pro-inflammatory cytokines released from both adipose tissue and activated immune cells can also contribute to muscle insulin resistance by disrupting insulin signaling pathways.64,66 This inflammatory response not only impairs glucose uptake in muscle cells but also perpetuates a cycle of metabolic dysfunction, as the inflammatory mediators can further stimulate lipid accumulation and inflammation in both liver and muscle tissues.67,68
The intricate relationship between insulin resistance and altered metabolic pathways in immune cells is a driving force in the chronic inflammation observed in T2DM. Targeting these metabolic shifts may offer a promising avenue for therapeutic strategies.69
The Impact of Insulin on Specific Immune Cell Types in T2DM
The immune system’s complexity in T2DM is significantly influenced by insulin’s regulatory role across various immune cell types. Insulin resistance disrupts normal immune cell signaling, leading to an altered immune landscape characterized by heightened inflammation. Each immune cell type—T lymphocytes, B lymphocytes, macrophages, and others—responds differently to insulin signaling, contributing uniquely to the progression of T2DM. This interplay between insulin signaling and immune responses not only exacerbates systemic inflammation but also perpetuates insulin resistance (Table 1). Understanding how insulin impacts specific immune cell types is crucial for developing targeted interventions that can break this cycle of chronic inflammation and insulin resistance, which are central to T2DM’s pathogenesis.
![]() |
Table 1 The Role of Insulin on Specific Immune Cell Types and Its Dysfunction Impacts in T2DM |
T Lymphocytes
Insulin plays a crucial role in regulating T lymphocyte function, impacting their activation, differentiation, and cytokine production, which is particularly relevant in T2DM. T cells express insulin receptors, and insulin signaling via the PI3K/Akt pathway is essential for glucose uptake and glycolysis during T cell activation, providing energy for proliferation and effector functions.70
In T2DM, insulin resistance disrupts this pathway, leading to altered T cell metabolism and function. Insulin-resistant T cells shift from glucose metabolism to fatty acid oxidation, resulting in a pro-inflammatory state characterized by increased production of cytokines that exacerbate systemic inflammation and insulin resistance.71,72 This shift impairs T cell activation and contributes to the chronic inflammation seen in T2DM.
Insulin also influences the balance of T cell subsets, such as Th1, Th2, Th17, and regulatory T cells (Tregs). In T2DM, there’s a notable increase in pro-inflammatory Th1 and Th17 cells, which produce interferon-gamma (IFN-γ) and interleukin-17 (IL-17), respectively. These cytokines contribute to insulin resistance and tissue inflammation, worsening metabolic dysfunction.73 Conversely, Tregs, which suppress inflammation and maintain immune tolerance, are reduced in T2DM due to insulin resistance, leading to an imbalance between pro-inflammatory and regulatory immune responses.74
This imbalance is further exacerbated by elevated levels of IL-6, which correlate with decreased Treg populations in T2DM patients, allowing for unchecked activity of pro-inflammatory Th1 and Th17 cells.89 Additionally, increased expression of immune checkpoint molecules like PD-1 on T cells indicates T cell exhaustion, impairing their ability to regulate inflammation effectively.90
The influence of insulin on T cell function plays a crucial role in maintaining immune balance, with insulin resistance contributing significantly to the pro-inflammatory environment in T2DM. Expanding our comprehension of this relationship could lead to new therapeutic approaches.73,84
B Lymphocytes
Insulin significantly influences B lymphocyte development, function, and antibody production, playing a vital role in immune regulation and metabolic homeostasis. Insulin signaling through the PI3K/Akt pathway is crucial for B cell maturation, proliferation, and antibody production, as it facilitates glucose uptake needed for energy and antibody. However, in T2DM, insulin resistance impairs B cell responsiveness, leading to reduced glucose uptake and compromised antibody production, which contributes to overall immune dysfunction.75
In T2DM, B cells also exhibit a shift towards pro-inflammatory cytokine production, with increased levels of IL-6 and IL-8 and reduced anti-inflammatory cytokine IL-10.76 This imbalance promotes chronic inflammation, exacerbating insulin resistance and disrupting metabolic regulation. Pro-inflammatory cytokines not only impair insulin signaling but also recruit additional immune cells, intensifying inflammation in peripheral tissues.
B cells interact with T cells, influencing their differentiation and promoting pro-inflammatory subsets such as Th1 and Th17 cells, which produce cytokines like IFN-γ and IL-17, further impairing insulin signaling.77 This interaction perpetuates a feedback loop of inflammation and insulin resistance. Additionally, in T2DM, B cells may lose tolerance, leading to autoantibody production that negatively affects pancreatic β-cells, contributing to dysfunction and apoptosis.77
The presence of activated B cells in adipose tissue, common in obesity and T2DM, contributes to the inflammatory environment, recruiting other immune cells like macrophages and further impairing insulin sensitivity.78 This involvement of B cells in promoting inflammation and metabolic disruption underscores their role in T2DM progression.
The role of insulin in B cell function is vital, but its dysregulation in T2DM leads to significant immune disturbances. These findings highlight the potential of targeting B cell pathways to modulate inflammation and insulin resistance in diabetes management.76,79
Macrophages
Insulin plays a crucial role in regulating macrophage polarization between pro-inflammatory (M1) and anti-inflammatory (M2) phenotypes. In a healthy state, insulin promotes M2 macrophages, which support tissue repair and maintain metabolic balance through the PI3K/Akt pathway, enhancing anti-inflammatory cytokines like IL-10.80,91 However, in T2DM, insulin resistance disrupts this balance, leading to a predominance of M1 macrophages that secrete pro-inflammatory cytokines like TNF-α and IL-6, worsening chronic inflammation and insulin resistance.81
This shift towards M1 macrophages creates a feedback loop that further impairs insulin signaling in macrophages and insulin-sensitive tissues, such as adipose tissue and liver. M1 macrophages infiltrate these tissues, releasing cytokines that interfere with insulin signaling, thus exacerbating insulin resistance.82 Additionally, obesity-related free fatty acids and lipopolysaccharides (LPS) activate Toll-like receptors (TLRs) on macrophages, driving M1 polarization and inflammation.92
The influence of insulin on macrophages extends to the NLRP3 inflammasome, which, when activated in T2DM, produces interleukin-1β (IL-1β), further driving inflammation and insulin resistance.83 Conversely, M2 macrophages, which are typically reduced in T2DM, limit anti-inflammatory cytokine production, like IL-10, resulting in unchecked inflammation.93
The imbalance between M1 and M2 macrophages contributes to adipose tissue inflammation, a key factor in T2DM.72,94 The abundance of M1 macrophages secreting TNF-α, IL-6, and IL-1β creates a chronic inflammatory environment that disrupts insulin signaling in adipocytes, reducing glucose uptake and worsening hyperglycemia.14 This process also contributes to pancreatic β-cell dysfunction, impairing insulin secretion.95
Moreover, M1 macrophages contribute to inflammation in other metabolic organs, such as the liver and muscle, reinforcing systemic inflammation and insulin resistance in T2DM. This chronic inflammation leads to progressive β-cell deterioration, accelerating T2DM progression.96
Insulin’s regulatory impact on macrophage polarization directly affects the balance between inflammatory and anti-inflammatory states. Addressing this imbalance could be central to developing treatments that mitigate chronic inflammation in T2DM.81,97
Neutrophils, NK Cells, and Other Immune Cells
Neutrophils, natural killer (NK) cells, and other immune cells are crucial in maintaining immune homeostasis, but their functions are influenced by insulin, particularly in T2DM. In T2DM, chronic inflammation and insulin resistance significantly impair the activity and efficiency of these immune cells, contributing to immune dysfunction and disease progression.94
Insulin typically enhances neutrophil function by promoting chemotaxis, phagocytosis, and ROS production, which are essential for pathogen elimination.84 However, in T2DM, insulin resistance reduces neutrophil activity, leading to a diminished capacity to fight infections. This dysfunction, along with the formation of neutrophil extracellular traps (NETs), sustains chronic inflammation and tissue damage.10 Impaired chemotaxis and phagocytosis in neutrophils increase susceptibility to infections in T2DM patients.10,98
NK cells, which target infected or malignant cells, are also affected by insulin signaling. In healthy conditions, insulin supports NK cell proliferation and cytotoxicity, but in T2DM, chronic inflammation impairs their cytotoxic function and alters cytokine production, contributing to immune imbalance.85 The reduced activity of NK cells in T2DM hampers their ability to regulate other immune cells, aggravating inflammation and metabolic disruption.86
The impairment of neutrophil and NK cell functions due to insulin resistance sheds light on how immune dysregulation contributes to T2DM complications. Understanding these dynamics is crucial for developing comprehensive interventions.87,88
In a healthy state, insulin plays a regulatory role across diverse immune cell types, ensuring immune homeostasis and reducing inflammatory responses. Normal insulin signaling in T lymphocytes, for example, supports balanced cytokine production and differentiation, favoring a controlled immune response. Similarly, insulin enables B lymphocytes to maintain antibody production and supports macrophage polarization toward the anti-inflammatory M2 phenotype. Macrophages under insulin’s influence help to limit inflammation, aiding tissue repair and metabolic balance. Insulin’s regulatory influence extends to neutrophils and NK cells, enhancing chemotaxis, phagocytosis, and cytotoxic functions critical for pathogen clearance.
In contrast, insulin resistance in T2DM disrupts these regulatory pathways, resulting in immune dysregulation. T lymphocytes shift toward pro-inflammatory subsets (Th1, Th17), while B cells increase production of pro-inflammatory cytokines, contributing to systemic inflammation. Macrophages, under insulin resistance, predominantly polarize to the pro-inflammatory M1 phenotype, perpetuating chronic inflammation and exacerbating insulin resistance. Neutrophils and NK cells also lose functionality, diminishing their infection-fighting capabilities. This immune imbalance under insulin resistance fosters a self-perpetuating cycle of inflammation and metabolic dysfunction that drives T2DM progression. Understanding these contrasting roles of insulin in immune cell regulation could pave the way for targeted therapies to alleviate inflammation and improve metabolic outcomes in T2DM.
Inflammatory Mediators and Insulin Signaling in T2DM
Pro-Inflammatory Cytokines and Insulin Resistance
Chronic inflammation is closely linked to insulin resistance in T2DM, with pro-inflammatory cytokines like TNF-α, IL-6, and IL-1β playing a pivotal role. These cytokines interfere with insulin signaling, exacerbating metabolic dysfunction and promoting insulin resistance.
TNF-α is a well-studied cytokine that contributes to insulin resistance by promoting the serine phosphorylation of insulin receptor substrates (IRS), particularly IRS-1, which inhibits effective insulin signaling and reduces insulin sensitivity.99 Elevated TNF-α levels, often produced by adipose tissue macrophages, are associated with increased insulin resistance and perpetuate the chronic inflammatory state in T2DM.100
IL-6 is another key cytokine involved in insulin resistance, secreted by adipocytes and macrophages. IL-6 induces the expression of suppressor of cytokine signaling (SOCS) proteins, which degrade IRS proteins, disrupting insulin signaling pathways.101 Chronic elevation of IL-6 is linked to insulin resistance and increased inflammation in tissues such as the liver, muscle, and adipose tissue, contributing to glucose metabolism impairment.102
IL-1β, produced by activated macrophages, plays a significant role in the inflammatory response in T2DM. It promotes β-cell dysfunction, impairs insulin secretion, and induces apoptosis in pancreatic β-cells, exacerbating insulin resistance and hyperglycemia.103 Specifically, IL-1β can inhibit insulin secretion by interfering with the signaling pathways essential for glucose-stimulated insulin release, primarily through the suppression of insulin receptor substrate-1 (IRS-1) signaling.14,104 This inhibition not only reduces insulin output but also contributes to the overall decline in β-cell mass and function. Moreover, chronic exposure to IL-1β can induce β-cell apoptosis through mechanisms involving endoplasmic reticulum (ER) stress and the activation of pro-apoptotic pathways.105,106 The sustained inflammatory environment created by IL-1β leads to the generation of reactive oxygen species (ROS), further exacerbating cellular stress and promoting cell death.107 This cycle of inflammation and cell death not only diminishes the functional β-cell mass but also contributes to systemic insulin resistance, as the loss of insulin-producing cells impairs the body’s ability to regulate blood glucose levels effectively.108,109
C-reactive Protein (CRP), though not a cytokine, serves as a marker of systemic inflammation in T2DM. High CRP levels reflect chronic inflammation and correlate with insulin resistance, highlighting its role in metabolic dysfunction.110 Similarly, IL-8 is elevated in T2DM and contributes to insulin resistance by promoting inflammation and altering insulin-sensitive tissue function.101
Pro-inflammatory cytokines like TNF-α, IL-6, and IL-1β impair insulin signaling by promoting serine phosphorylation of IRS proteins, reducing insulin receptor sensitivity and affecting glucose uptake in target tissues. Additionally, cytokine activity activates Toll-like receptors (TLRs), especially TLR4, which respond to inflammatory stimuli and further disrupt insulin signaling, reinforcing insulin resistance.111
Pro-inflammatory cytokines are key players in the progression of insulin resistance in T2DM. Targeting these inflammatory pathways offers a promising direction for therapeutic interventions aimed at improving insulin sensitivity.
Adipokines and Immune Modulation
Adipokines, bioactive molecules secreted by adipose tissue, significantly impact insulin signaling and immune cell function in T2DM, playing roles in both pro-inflammatory and anti-inflammatory responses (Table 2).
![]() |
Table 2 Roles of Adipokines in Modulating Insulin Resistance and Inflammation in Type 2 Diabetes |
Adiponectin is an anti-inflammatory adipokine with insulin-sensitizing properties that enhances insulin signaling via the AMP-activated protein kinase (AMPK) pathway, improving glucose uptake and fatty acid oxidation.112 Adiponectin exerts its effects primarily through adiponectin receptor 1 (AdipoR1) and adiponectin receptor 2 (AdipoR2), which activate AMPK as well as peroxisome proliferator-activated receptor alpha (PPAR-α), contributing to its metabolic benefits. In T2DM, adiponectin levels are often reduced, contributing to insulin resistance and inflammation, as its protective effects on insulin sensitivity diminish.113,119,120
In contrast, leptin is a pro-inflammatory adipokine that is elevated in obesity and T2DM. It promotes inflammatory cytokine secretion from immune cells, enhancing inflammation and contributing to insulin resistance.114,115 This increase in leptin levels perpetuates the chronic low-grade inflammation typical of T2DM.
Resistin is another adipokine linked to insulin resistance and is often elevated in T2DM. Though its receptor is not fully elucidated, resistin is believed to interact with Toll-like receptor 4 (TLR4), promoting inflammation through the nuclear factor-kappa B (NF-κB) pathway, which increases the expression of cytokines like TNF-α and IL-6.121 It promotes inflammation by increasing the expression of cytokines like TNF-α and IL-6, reinforcing the connection between adipose tissue inflammation and metabolic dysfunction.116 Similarly, chemerin is associated with inflammation and insulin resistance. It signals through the chemokine-like receptor 1 (CMKLR1), facilitating macrophage recruitment to adipose tissue, which contributes to the chronic inflammatory environment characteristic of T2DM. This receptor interaction links chemerin to the immune-modulatory pathways that exacerbate inflammation and insulin resistance in metabolic tissues.117
Fetuin A is implicated in insulin resistance through its effects on insulin signaling and inflammation. Fetuin A inhibits insulin receptor tyrosine kinase activity, disrupting insulin signaling pathways in tissues like the liver and skeletal muscle.113 By serving as an adaptor protein for TLR4, fetuin A enhances lipid-induced insulin resistance, promoting inflammation within insulin-sensitive tissues.122,123 Elevated fetuin A levels are linked to a higher risk of T2DM, exacerbating metabolic dysfunction by intensifying inflammatory responses in adipose and other insulin-sensitive tissues.
In contrast, adipsin has a complex and still-debated role in metabolic health. As an adipokine produced by adipose tissue, adipsin has been observed at lower circulating levels in individuals with T2DM, suggesting a potential protective role in β-cell function and insulin sensitivity.118,124,125 Although some evidence indicates that adipsin may positively influence insulin secretion, its precise mechanisms of action and its interactions with other adipokines and inflammatory mediators remain insufficiently understood.126,127 Further research is needed to clarify how adipsin interacts within the complex network of metabolic and inflammatory pathways to influence insulin sensitivity.
The dysregulation of these adipokines in T2DM promotes a pro-inflammatory environment, influencing immune cells such as macrophages and T cells. Elevated levels of leptin, resistin, and chemerin promote macrophage polarization towards the M1 pro-inflammatory phenotype, increasing the secretion of cytokines like TNF-α and IL-6, which further contribute to insulin resistance.128 This pro-inflammatory state also affects T cells, shifting them towards a Th1-dominant immune response, perpetuating inflammation and metabolic dysfunction.129
The imbalance in adipokine levels contributes significantly to the inflammatory landscape of T2DM. Recognizing the impact of these molecules could guide the development of strategies to restore metabolic and immune equilibrium.
Myokines and Hepatokines in Insulin Sensitivity and Metabolic Health
Myokines and hepatokines play key roles in metabolic health, particularly in the regulation of insulin sensitivity and inflammation. Myokines like irisin and myonectin are released by skeletal muscles during exercise, enhancing insulin sensitivity, glucose uptake, and promoting the browning of white adipose tissue (WAT), which boosts energy expenditure.130–132 Irisin specifically activates uncoupling protein 1 (UCP1) in adipocytes, facilitating thermogenesis and aiding in weight regulation.133,134 Additionally, irisin has been shown to improve insulin receptor signaling in muscle and liver, enhance glucose metabolism, and protect pancreatic β-cells, essential for insulin secretion.135–137 Its anti-inflammatory properties further contribute to metabolic stability, counteracting the chronic low-grade inflammation often associated with obesity and insulin resistance.138,139
In contrast, hepatokines such as fibroblast growth factor 21 (FGF21) and fetuin A, secreted by the liver, have complex effects on insulin sensitivity. FGF21 improves insulin sensitivity by modulating lipid metabolism and reducing inflammation in muscle and liver.140 Conversely, fetuin A disrupts insulin receptor signaling and promotes inflammation, contributing to insulin resistance, especially in obesity.141
The interplay between myokines and hepatokines underscores the importance of muscle-liver communication in maintaining metabolic health. Dysregulation in these proteins can lead to insulin resistance, highlighting their potential as therapeutic targets for metabolic disorders.130,131
Insulin and Oxidative Stress in Immune Cells
Oxidative stress plays a multifaceted role in the immune system, influencing both immune responses and the overall health of immune cells. Under normal physiological conditions, reactive oxygen species (ROS) generated during metabolic processes serve as signaling molecules that help regulate immune functions, including the activation and proliferation of immune cells. For instance, ROS are crucial for the oxidative burst in phagocytes, which is essential for killing pathogens and initiating inflammatory responses.142,143 This controlled production of ROS can enhance the immune response by promoting the expression of pro-inflammatory cytokines and facilitating communication between immune cells.144
However, excessive oxidative stress can lead to detrimental effects on immune function, particularly in states of chronic inflammation or disease. Elevated levels of ROS can cause cellular damage, impair immune cell function, and induce apoptosis, thereby diminishing the efficacy of the immune response.145 For example, in conditions such as obesity and diabetes, oxidative stress contributes to the dysfunction of T cells and macrophages, promoting a pro-inflammatory state that exacerbates insulin resistance.146
One primary mechanism is the oxidative modification of insulin signaling proteins. ROS can oxidize key proteins, such as IRS-1, leading to its serine phosphorylation, which inhibits effective insulin signaling and disrupts pathways like PI3K/Akt.147 This impairment is a critical factor in insulin resistance, as insulin signaling is essential for glucose uptake and metabolism in immune cells.
Oxidative stress also activates stress-sensitive kinases such as JNK and p38 MAPK, which further phosphorylate IRS proteins, exacerbating insulin resistance.148 These kinases respond to increased inflammatory cytokines in T2DM, linking inflammation to impaired insulin signaling.
The alteration of the cellular redox state is another consequence, disrupting insulin signaling and promoting inflammatory responses.149 This altered state affects glucose transporter GLUT4’s expression and translocation, reducing glucose uptake and contributing to insulin resistance.150
Oxidative stress also impacts immune cell function, impairing neutrophil and macrophage activity, which promotes a pro-inflammatory state. It enhances cytokine production from macrophages, perpetuating inflammation and disrupting insulin signaling.151 Additionally, mitochondrial dysfunction due to oxidative stress results in impaired ATP synthesis and increased ROS production, creating a feedback loop that worsens insulin resistance.152
Oxidative stress influences macrophage polarization towards the M1 phenotype, leading to increased secretion of inflammatory cytokines like TNF-α and IL-6, which further impair insulin signaling and β-cell function.153 Elevated ROS levels also impair T cell function by inducing a pro-inflammatory state and promoting T cell exhaustion, contributing to immune dysregulation in T2DM.154
The impact of oxidative stress on insulin signaling and immune cell function is a critical factor in the development of insulin resistance in T2DM. Addressing this could help break the cycle of chronic inflammation and metabolic dysfunction.
Anti-Inflammatory Effects of Insulin
Insulin plays a critical role in modulating inflammation, with significant implications for managing T2DM. Its anti-inflammatory effects extend beyond glycemic control, influencing immune cell function and the overall inflammatory state.
One of the primary ways insulin exerts its anti-inflammatory effects is through the suppression of pro-inflammatory cytokines. Insulin administration decreases levels of cytokines like TNF-α, IL-1β, and IL-6, which are often elevated in T2DM.155 Insulin inhibits nuclear factor kappa B (NF-κB) activation, a transcription factor that drives these cytokines’ expression, thus reducing inflammation and mitigating insulin resistance.
Insulin also promotes the production of anti-inflammatory cytokines, particularly IL-10, which helps restore immune homeostasis and improve insulin sensitivity.155 This shift from a pro-inflammatory to an anti-inflammatory state reduces the inflammatory burden in T2DM.
Another crucial effect of insulin is its role in macrophage polarization. Insulin promotes the shift of macrophages from the pro-inflammatory M1 phenotype to the anti-inflammatory M2 phenotype, aiding in tissue repair and reducing inflammation.156 This shift improves insulin sensitivity, counteracting one of the primary drivers of insulin resistance in T2DM.
However, insulin resistance can impair the insulin receptor’s signaling efficacy, diminishing its ability to carry out anti-inflammatory actions, including suppressing pro-inflammatory cytokine production and encouraging anti-inflammatory responses. Consequently, immune cells in an insulin-resistant state may not fully experience insulin’s anti-inflammatory benefits through exogenous insulin alone. To overcome this limitation, enhancing insulin sensitivity prior to or alongside insulin therapy is often necessary. Insulin sensitizers—such as physical activity, metformin, and thiazolidinediones (TZDs)—help improve insulin receptor functionality by reducing systemic inflammation, promoting glucose uptake, and bolstering cellular insulin sensitivity. For instance, physical activity enhances glucose uptake in skeletal muscle, while metformin and TZDs regulate hepatic glucose production and improve overall cellular responsiveness to insulin. These mechanisms enable immune cells to effectively respond to insulin, allowing them to leverage its full metabolic and anti-inflammatory benefits.
In addition to modulating cytokine production, insulin enhances immune cell function by supporting glucose uptake and metabolism in T cells and NK cells, balancing immune responses (Xia et al, 2017). This is particularly relevant in T2DM, where immune cell dysfunction contributes to chronic inflammation and heightened susceptibility to infections. Insulin also reduces oxidative stress by lowering reactive oxygen species (ROS) levels, further aiding in the maintenance of normal insulin signaling in immune cells and helping to alleviate inflammation (Rehman & Akash, 2016).
The therapeutic implications of insulin’s anti-inflammatory effects, especially when paired with insulin sensitizers, are substantial. By targeting both glucose regulation and inflammation, this dual approach offers a comprehensive treatment strategy for T2DM (L. Su et al, 2018). In summary, utilizing insulin sensitizers to restore insulin responsiveness may be essential for achieving the full range of insulin’s metabolic and immune-modulating effects. This strategy could be more effective in managing both glycemic control and chronic inflammation in T2DM, as insulin exerts anti-inflammatory effects through the suppression of pro-inflammatory cytokines, promotion of anti-inflammatory cytokines, macrophage polarization, immune cell function enhancement, and reduction of oxidative stress.
Dual Roles of Insulin
The role of insulin in the immune response against pathogens can be conceptualized as a “double-edged sword”, where it exerts both beneficial and detrimental effects depending on the context of its action. On one hand, insulin plays a crucial role in enhancing immune responses, while on the other hand, it can contribute to immune dysfunction, particularly in states of insulin resistance and metabolic disease.
Insulin’s Role in Enhancing Immune Responses
Insulin is known to promote the proliferation and activation of immune cells, particularly T cells and macrophages, which are essential for mounting an effective immune response against pathogens. For instance, insulin signaling has been shown to enhance the differentiation of T cells into effector cells that produce cytokines such as IL-2 and IFN-γ, which are critical for fighting infections.157 This enhancement of T cell function is particularly important in the context of acute infections, where a robust immune response is necessary for pathogen clearance.
Moreover, insulin facilitates glucose uptake in immune cells, providing the necessary energy for their activation and function. This metabolic support is vital during an immune response, as activated immune cells require increased energy to proliferate and produce cytokines.158 Insulin also promotes the polarization of macrophages towards an M2 phenotype, which is associated with anti-inflammatory responses and tissue repair, further aiding in the resolution of inflammation following an infection.159,160
Insulin’s Detrimental Effects in Immune Dysfunction
Conversely, in states of insulin resistance, which is commonly observed in obesity and type 2 diabetes, insulin can have detrimental effects on the immune system. Chronic hyperinsulinemia can lead to a state of immune dysfunction characterized by an overactive inflammatory response. For example, elevated insulin levels can promote the activation of pro-inflammatory pathways in macrophages, leading to increased production of cytokines such as TNF-α and IL-6, which are associated with chronic inflammation and insulin resistance161,162. This inflammatory milieu can impair the ability of immune cells to respond effectively to pathogens, thereby increasing susceptibility to infections.
Furthermore, the shift in macrophage polarization from M2 to M1 in the context of insulin resistance contributes to a chronic inflammatory state that exacerbates metabolic dysfunction.84,163,164 This maladaptive immune response not only hinders the clearance of pathogens but also perpetuates a cycle of inflammation that can lead to further insulin resistance and metabolic disease.163,164
The Balance of Insulin’s Effects
The dual role of insulin in the immune response underscores the importance of maintaining insulin sensitivity for optimal immune function. In healthy individuals, insulin promotes a balanced immune response that is capable of effectively combating pathogens while also resolving inflammation. However, in individuals with insulin resistance, the beneficial effects of insulin are overshadowed by its pro-inflammatory actions, leading to a compromised immune system.
Therapeutic strategies aimed at improving insulin sensitivity, such as lifestyle interventions, pharmacological agents, and dietary modifications, may help restore the beneficial effects of insulin on the immune system.165,166 For instance, weight loss and physical activity have been shown to improve insulin sensitivity and reduce chronic inflammation, thereby enhancing immune function.167,168 Additionally, medications that target insulin signaling pathways may also provide a means to modulate immune responses in individuals with metabolic disorders.
The dual nature of insulin in regulating immune responses emphasizes the delicate balance required for optimal immune function. Therapeutic strategies that enhance insulin sensitivity while managing inflammation could be transformative for T2DM care.
Therapeutic Implications and Interventions
Insulin Therapy and Immune Modulation in T2DM
Insulin therapy plays a crucial role in managing hyperglycemia in T2DM and has significant effects on immune cell function and inflammation. Beyond regulating glucose metabolism, insulin offers therapeutic benefits by modulating immune responses and reducing inflammation.
Insulin therapy reduces pro-inflammatory cytokine production by inhibiting NF-κB pathways, which decreases cytokines like TNF-α and IL-6, helping to alleviate chronic inflammation and improve insulin sensitivity.54 Additionally, insulin promotes the secretion of anti-inflammatory cytokines, such as IL-10, contributing to a balanced immune response.169
Insulin also influences macrophage polarization, promoting a shift from the pro-inflammatory M1 phenotype to the anti-inflammatory M2 phenotype, reducing inflammation and enhancing insulin sensitivity.170 This shift aids tissue repair and inflammation resolution, contributing to insulin’s anti-inflammatory effects.
The therapy impacts T cell function by promoting glucose uptake, which is essential for T cell activation and proliferation, leading to a more balanced immune response and reducing chronic inflammation in T2DM.24 Moreover, insulin therapy helps reduce oxidative stress by lowering reactive oxygen species (ROS) levels, which restores normal insulin signaling pathways in immune cells, alleviating inflammation and improving insulin sensitivity.171
However, there are limitations and challenges associated with insulin therapy in T2DM. One concern is the potential for immune suppression, which can increase susceptibility to infections since insulin therapy might impair the overall immune response.172 Additionally, insulin therapy may result in altered immune cell function, such as impaired bactericidal activity in neutrophils, potentially leading to a higher risk of infections.173
Hyperinsulinemia presents another challenge, as chronically high insulin levels can promote inflammatory pathways and shift macrophage polarization towards a pro-inflammatory M1 phenotype, exacerbating insulin resistance and inflammation.172 This feedback loop presents a critical barrier to the long-term immunomodulatory effectiveness of insulin therapy in T2DM, underscoring the complexity of achieving sustained metabolic and immune benefits through insulin alone.
The onset of T2DM therapy typically involves insulin-sensitizing agents like metformin, which emphasizes the importance of improving insulin sensitivity as an initial therapeutic goal. This step is essential to restore physiological insulin responses, including within immune cells, to counter chronic inflammation. For patients with insulin resistance, initiating insulin therapy can aggravate hyperinsulinemia, contributing to adverse effects on immune cells.172 Over time, the efficacy of insulin in modulating immune responses may diminish, as increased insulin resistance often necessitates higher insulin doses, complicating immune regulation and limiting long-term benefits.172
Overall, insulin therapy offers considerable anti-inflammatory benefits in T2DM management by reducing pro-inflammatory cytokines, supporting anti-inflammatory cytokine production, influencing macrophage polarization, enhancing T-cell function, and mitigating oxidative stress. However, limitations such as immune suppression, increased infection risk, and hyperinsulinemia underscore the need for a more comprehensive approach. Integrating insulin therapy with lifestyle modifications, anti-inflammatory agents, and adjunctive therapies can more effectively address both the metabolic and immune challenges inherent to T2DM, fostering improved patient outcomes.
Targeting Insulin Signaling Pathways for Immune Modulation
Targeting insulin signaling pathways offers promising therapeutic options for modulating immune responses in T2DM. These pathways regulate insulin sensitivity and immune function, making them valuable targets for reducing inflammation and improving immune activity in T2DM (Table 3).
![]() |
Table 3 Therapeutic Targets and Mechanisms for Modulating Insulin Signaling and Immune Responses in Type 2 Diabetes Mellitus |
A key therapeutic target is the Insulin Receptor (IR). Enhancing the expression or sensitivity of IR on immune cells can improve insulin signaling, promoting glucose uptake and reducing inflammation.54 Improving IR function could enhance immune cell activity and decrease inflammation in T2DM.
The PI3K/Akt pathway, a crucial component of insulin signaling, regulates glucose metabolism and immune function. Activation of this pathway plays a significant role in mediating anti-inflammatory responses, particularly in macrophages. Targeting the PI3K/Akt pathway could regulate immune responses and improve insulin sensitivity in T2DM.174
The mTOR pathway is another key regulator of immune cell metabolism and function. Insulin activates mTOR, influencing T cell and macrophage activity. Modulating mTOR could improve immune responses and insulin sensitivity, making it a valuable therapeutic target in T2DM.175
Suppressor of Cytokine Signaling (SOCS) proteins play a critical role in negatively regulating insulin signaling and contributing to insulin resistance in immune cells. Inhibiting SOCS expression enhances insulin signaling and reduces inflammation, offering a potential therapeutic strategy for improving insulin sensitivity in type 2 diabetes mellitus (T2DM).176 Targeting cytokine receptors, particularly for pro-inflammatory cytokines like TNF-α and IL-6, has also been effective in reducing inflammation and improving insulin sensitivity, as demonstrated with TNF-α inhibition in adipose tissue.177
Recent studies have highlighted the therapeutic potential of anti-inflammatory agents such as IL-1 receptor blockers (anakinra), IL-1β antagonists (canakinumab), TNF antagonists (etanercept), and salicylates in improving insulin sensitivity. Anakinra improves β-cell function and insulin sensitivity by inhibiting IL-1 and suppressing other pro-inflammatory cytokines.180 Etanercept, targeting TNF-α, also reduces insulin resistance, and its combination with anakinra shows synergistic effects, restoring insulin sensitivity.180,181 Salicylates act by inhibiting IκB kinase, reducing NF-κB activation and inflammatory cytokines, key contributors to insulin resistance.182 These therapies demonstrate significant promise in mitigating inflammation and enhancing insulin action in T2DM.183
Transcription factors such as FoxO and NF-κB regulate insulin signaling and inflammatory responses in immune cells. Modulating their activity can influence the balance between pro- and anti-inflammatory responses, enhancing insulin sensitivity.184
The endocannabinoid system also regulates insulin sensitivity and immune responses. Activating cannabinoid receptors can influence insulin signaling and reduce inflammation, suggesting potential therapeutic benefits.178,179
Other promising molecules include Insulin-like Growth Factor-1 (IGF-1) and sphingosine 1-phosphate (S1P). IGF-1 stimulates regulatory T cells, reducing inflammation and improving insulin sensitivity, while S1P influences immune cell function, making it a potential target for enhancing insulin sensitivity in T2DM.185
Lastly, nutritional interventions, like omega-3 fatty acid intake, can modulate insulin signaling pathways and immune responses, impacting adipokine and cytokine production.186
Therapies that target insulin signaling pathways hold great potential in modulating immune responses in T2DM. Such interventions could redefine treatment paradigms by addressing both metabolic and immune dysfunctions.
Lifestyle Interventions and Insulin Sensitivity
Lifestyle interventions, including dietary modifications, physical activity, and weight management, play a fundamental role in enhancing insulin sensitivity and improving immune function in T2DM.
Dietary Modifications are crucial for managing insulin sensitivity. Diets low in saturated fats and high in fiber, whole grains, fruits, and vegetables can improve insulin sensitivity and reduce inflammation.187 Such diets decrease pro-inflammatory cytokines and adipokines, which contribute to insulin resistance. Additionally, omega-3 fatty acids, antioxidants, and polyphenols promote anti-inflammatory responses and modulate immune function, further enhancing insulin sensitivity.
Physical Activity is a cornerstone of T2DM management. Regular exercise improves glucose uptake, insulin receptor sensitivity, and the translocation of glucose transporters (GLUT4) to cell membranes, enhancing glycemic control.188 Moderate-intensity aerobic exercise, such as brisk walking for at least 150 minutes per week, significantly improves insulin sensitivity.189 Exercise also reduces pro-inflammatory cytokines and promotes macrophage polarization towards the anti-inflammatory M2 phenotype, benefiting both metabolic control and immune modulation.190
Weight Management is another key factor. Modest weight loss (5–10% of body weight) leads to substantial improvements in insulin sensitivity and glycemic control by reducing adipose tissue mass.191 This reduction lowers pro-inflammatory adipokine and cytokine secretion, alleviating the inflammatory burden and restoring normal insulin signaling.
Gut Microbiota Modulation can impact insulin sensitivity. Dietary changes, probiotics, and prebiotics positively influence gut microbiota composition, enhancing insulin sensitivity by modulating inflammatory responses and metabolic pathways.192 A healthier gut microbiome increases the production of short-chain fatty acids (SCFAs) like butyrate, which have anti-inflammatory effects and improve glucose metabolism.
Behavioral Changes, such as self-monitoring of blood glucose, dietary intake, and physical activity, empower individuals to make healthier choices, leading to better insulin sensitivity.193 Educational programs that promote these changes improve glycemic control and metabolic outcomes.
Pharmacotherapy Synergy enhances the benefits of lifestyle interventions. Combining medications like metformin with lifestyle changes improves insulin sensitivity and glycemic control more effectively than pharmacotherapy alone.194 This combined approach addresses both metabolic and immune aspects of T2DM.
Lifestyle changes offer a foundational approach to improving insulin sensitivity and immune function in T2DM. These interventions serve as a cornerstone for reducing inflammation and enhancing overall metabolic health.
Future Perspectives and Challenges
Emerging therapies targeting insulin’s role in immune modulation offer promising avenues for managing T2DM but also present significant challenges. Understanding the complex interactions between insulin signaling and immune function is crucial for developing effective treatments that address both metabolic and immune dysfunction in T2DM.
Targeting Insulin Resistance in immune cells is a major focus, as insulin resistance in these cells contributes to chronic inflammation in T2DM.91 Restoring insulin sensitivity in immune cells could achieve desired anti-inflammatory effects, but this remains challenging, particularly as insulin resistance can worsen inflammation rather than reduce it.
Addressing Chronic Inflammation is another promising approach since elevated pro-inflammatory cytokines like TNF-α and IL-6 exacerbate insulin resistance. Therapies that reduce inflammation could improve insulin sensitivity; however, achieving this without causing immune suppression is a delicate balance.
Diverse Immune Cell Responses to Insulin present a challenge, as macrophages and T cells exhibit variable reactions based on the individual’s inflammatory state. Insulin may reduce inflammation in some cells but exacerbate it in others, making it difficult to design targeted therapies that effectively modulate insulin signaling across all immune cell types.195
The Interaction with Competing Pathways is another hurdle. Pro-inflammatory cytokines activate pathways like NF-κB, which inhibit insulin signaling, creating a complex network that complicates therapeutic targeting.196 Developing treatments that selectively target insulin-related pathways without disrupting others is a significant challenge.
Potential Side Effects are also a concern. While enhancing insulin sensitivity might improve metabolic outcomes, it could suppress immune responses and increase infection risk.197 Balancing the anti-inflammatory effects of insulin with maintaining immune defenses is essential.
Individual Variability adds complexity, as T2DM patients have diverse genetic backgrounds, lifestyles, and comorbidities, making one-size-fits-all therapies impractical.198 Personalized treatment approaches could be more effective but require extensive research.
The Long-term Efficacy of Insulin Therapy remains uncertain. Patients may develop tolerance over time, requiring higher doses and reducing effectiveness.199 This underscores the need for sustainable treatment strategies.
For future research, mechanistic studies of insulin signaling in immune cells, focusing on pathways like PI3K/Akt and mTOR, can identify therapeutic targets.54 Investigating the role of adipokines (eg, leptin, adiponectin, and resistin) in insulin signaling could further elucidate the link between obesity, metabolic dysfunction, and immune responses.200
The gut microbiota is another promising area for research, as the gut-immune axis plays a significant role in metabolic health. Understanding how lifestyle interventions alter gut microbiota could lead to innovative strategies for improving insulin sensitivity and immune function.29
Research on immune cell plasticity, especially macrophage and T cell polarization in response to insulin, could guide new approaches to managing chronic inflammation in T2DM.10 Understanding how these cells transition between pro- and anti-inflammatory states could aid in developing therapies that promote immune balance.
Exploration of Novel Therapeutics targeting insulin signaling in immune cells, such as agents that enhance insulin sensitivity or modulate inflammatory pathways, shows potential for addressing both metabolic and immune dysfunctions in T2DM.30 Long-term studies assessing these interventions would be valuable.
Lastly, the role of oxidative stress in modulating insulin signaling and immune responses warrants further investigation. Understanding its impact on immune cell function could reveal new therapeutic targets to reduce inflammation and improve metabolic health.201
In summary, while targeting insulin signaling pathways for immune modulation in T2DM offers promising opportunities, challenges related to insulin resistance, inflammation, immune variability, and potential side effects remain. Future research should focus on elucidating insulin-immune interactions, exploring novel treatments, and developing personalized strategies to manage T2DM’s metabolic and immune dysfunctions effectively.
Concluding Remarks
Insulin is a key regulator of both glucose metabolism and immune function. In type 2 diabetes mellitus (T2DM), insulin resistance disrupts normal immune cell regulation, shifting T cells, B cells, macrophages, neutrophils, and natural killer (NK) cells toward a pro-inflammatory state. This chronic inflammation perpetuates a cycle of insulin resistance and β-cell dysfunction, exacerbating hyperglycemia and systemic inflammation. These interactions highlight T2DM as a disorder involving significant immune dysregulation in addition to metabolic dysfunction.
Insulin therapy offers substantial anti-inflammatory benefits, including suppression of pro-inflammatory cytokines, promotion of anti-inflammatory cytokines, macrophage polarization, improved T-cell function, and reduced oxidative stress. However, in insulin-resistant states, hyperinsulinemia may exacerbate systemic inflammation and impair immune responses. This underscores the importance of restoring insulin sensitivity as a foundational step. Insulin sensitizers, such as physical activity, metformin, and thiazolidinediones (TZDs), enhance insulin receptor functionality, reduce inflammation, and improve immune cell responses to insulin. When paired with insulin therapy, this dual strategy addresses both metabolic control and immune regulation, breaking the cycle of chronic inflammation and insulin resistance.
Future therapies should focus on the interplay between metabolic and immune pathways, integrating lifestyle interventions, anti-inflammatory agents, and adjunctive therapies. However, challenges such as individual variability, the long-term effects of insulin therapy, and the balance between immune modulation and insulin sensitivity must be addressed.
In conclusion, targeting immune dysfunction in T2DM is critical for comprehensive disease management. Insulin’s dual role as a metabolic regulator and immune modulator provides unique therapeutic opportunities. By addressing the underlying immune and inflammatory mechanisms, we can improve insulin sensitivity, prevent complications, and enhance outcomes for individuals with T2DM.
Acknowledgment
We express our sincere gratitude to the Directorate General of Higher Education, Research and Technology the Ministry of Education, Culture, Research and Technology for their support in the completion of this article. We also extend our appreciation to the Research Center for Regional Development and Community Empowerment at Universitas Padjadjaran for their invaluable assistance throughout the literature review process.
Disclosure
The authors report no conflicts of interest in this work.
References
1. Ikwuka AO, Omoju DI, Mahanera OK. Profiling of clinical dynamics of type 2 diabetes mellitus in patients: a perspective review. World J Curr Med Pharm Res. 2023;210–218. doi:10.37022/wjcmpr.v5i5.294
2. Aljabri KS, Bokhari SA, Alshareef MA, Khan PM, Aljabri BK. Associated risk factors in pre-diabetes and type 2 diabetes in Saudi community. Arch Diabetes Obes. 2018;1(2).
3. Thipsawat S. Intervention for prevention of type 2 diabetes mellitus among prediabetes: a review of the literature. Sage Open Nurs. 2023;9:237796082311755.
4. Kaveh MH, Noori K, Nazari M, Khademi K. Quality of life and metabolic indicators of patients with type 2 diabetes: a cross-sectional study in Iran. Int J Endocrinol. 2022;2022:1–8. doi:10.1155/2022/4046012
5. Raj R, Navis S, Bhatti JS, Bhadada SK, Ramteke PW. Analysis of Complicating Risk Factors of Type 2 Diabetes Mellitus (T2DM). Integr Obes Diabetes. 2018;4(1).
6. Pepa GD, Vetrani C, Vitale M, Riccardi G. Wholegrain intake and risk of type 2 diabetes: evidence from epidemiological and intervention studies. Nutrients. 2018;10(9):1288. doi:10.3390/nu10091288
7. Zaheer S, Zeb TF, Baloch A, Amin R, Amjad Z, Sheikh A. The multifactorial burden of type 2 diabetes mellitus: a study from clinical settings of university hospital, Karachi, Pakistan. Int J Endorsing Heal Sci Res. 2020;8(4):275–287. doi:10.29052/IJEHSR.v8.i4.2020.275-287
8. Asghar S, Asghar S, Mahmood T, Bukhari SMH, Mumtaz MA, Rasheed A. Microalbuminuria as the tip of iceberg in type 2 diabetes mellitus: prevalence, risk factors, and associated diabetic complications. Cureus. 2023;15.
9. Rey-Reñones C, Martínez-Torres S, Martín-Luján F, et al. Type 2 diabetes mellitus and COVID-19: a narrative review. Biomedicines. 2022;10(9):2089. doi:10.3390/biomedicines10092089
10. Berbudi A, Rahmadika N, Tjahjadi AI, Ruslami R, Cahyadi AI, Ruslami R. Type 2 diabetes and its impact on the immune system. Curr Diabetes Rev. 2020;15(5):442–449.
11. Villasanta-Gonzalez A, Alcala-Diaz JF, Vals-Delgado C, et al. A plasma fatty acid profile associated to type 2 diabetes development: from the CORDIOPREV study. Eur J Nutr. 2021;61(2):843–857. doi:10.1007/s00394-021-02676-z
12. Johnson AM, Olefsky JM. The origins and drivers of insulin resistance. Cell. 2013;152(4):673–684. doi:10.1016/j.cell.2013.01.041
13. Parmar M. Obesity and type 2 diabetes mellitus. Integr Obes Diabetes. 2018;4(4):1–2.
14. Dror E, Dalmas É, Meier DT, et al. Postprandial macrophage-derived IL-1β stimulates insulin, and both synergistically promote glucose disposal and inflammation. Nat Immunol. 2017;18(3):283–292. doi:10.1038/ni.3659
15. Tsai S, Clemente-Casares X, Zhou AC, Watts TH, Winer S, Winer DA. Insulin receptor-mediated stimulation boosts T cell immunity during inflammation and infection. Cell Metabolism. 2018;28(6):922–934.e4. doi:10.1016/j.cmet.2018.08.003
16. Wang C, Li F, Guo J, Li C, Xu D, Wang B. Insulin resistance, blood glucose and inflammatory cytokine levels are risk factors for cardiovascular events in diabetic patients complicated with coronary heart disease. Exp Ther Med. 2017. doi:10.3892/etm.2017.5584
17. Berbudi A, Anshori H, Andromeda A, Kwarteng A, Putri AC, Atik N. Topical administration of curcuma longa L. extract gel increases M2 macrophage polarization and collagen density in skin excision. J Appl Pharm Sci. 2021;11(1):095–100.
18. van Niekerk G, van der Merwe M, Engelbrecht A-M. Diabetes and susceptibility to infections: implication for COVID‐19. Immunology. 2021;164(3):467–475. doi:10.1111/imm.13383
19. Ip BC, Cilfone NA, Belkina AC, et al. Th17 cytokines differentiate obesity from obesity‐associated type 2 diabetes and promote TNFα production. Obesity. 2015;24(1):102–112. doi:10.1002/oby.21243
20. Solinas G, Karin M. JNK1 and IKKβ: molecular links between obesity and metabolic dysfunction. FASEB J. 2010;24(8):2596–2611. doi:10.1096/fj.09-151340
21. Harding HP, Ron D. Endoplasmic reticulum stress and the development of diabetes: a review. Diabetes. 2002;51(suppl 3):S455–61. doi:10.2337/diabetes.51.2007.S455
22. Chao WC, Yen CL, Wu YH, et al. Increased resistin may suppress reactive oxygen species production and inflammasome activation in type 2 diabetic patients with pulmonary tuberculosis infection. Microbes Infect. 2015;17(3):195–204. doi:10.1016/j.micinf.2014.11.009
23. Sukmawati IR, Donoseputro M, Lukito W. Association between Free Fatty Acid (FFA) and insulin resistance: the role of inflammation (Adiponectin and high sensivity C-reactive Protein/hs-CRP) and stress oxidative (Superoxide Dismutase/SOD) in obese non-diabetic individual. Indones Biomed J. 2009;1(3):71. doi:10.18585/inabj.v1i3.102
24. Nekoua MP, Fachinan R, Atchamou AK, et al. Modulation of immune cells and Th1/Th2 cytokines in insulin-treated type 2 diabetes mellitus. Afr Health Sci. 2016;16(3):712. doi:10.4314/ahs.v16i3.11
25. Xuân NT, Hoàng NH, Nhung VP, Dương NT, Hà NH, Hải NV. Regulation of dendritic cell function by Insulin/Igf-1/Pi3k/Akt signaling through klotho expression. J Recept Signal Transduct. 2016;37(3):297–303. doi:10.1080/10799893.2016.1247862
26. Eberle C, Merki E, Yamashita T, et al. Maternal immunization affects in utero programming of insulin resistance and type 2 diabetes. PLoS One. 2012;7(9):e45361. doi:10.1371/journal.pone.0045361
27. Gerrits AJ, Gitz E, Koekman CA, Visseren FL, van Haeften TW, Akkerman JWN. Induction of insulin resistance by the adipokines resistin, leptin, plasminogen activator inhibitor-1 and retinol binding protein 4 in human megakaryocytes. Haematologica. 2012;97(8):1149–1157. doi:10.3324/haematol.2011.054916
28. Kubota T, Kubota N, Kumagai H, et al. Impaired insulin signaling in endothelial cells reduces insulin-induced glucose uptake by skeletal muscle. Cell Metab. 2011;13(3):294–307. doi:10.1016/j.cmet.2011.01.018
29. Musselman LP, Fink JL, Grant AR, Gatto JA, Tuthill BF, Baranski TJ. A complex relationship between immunity and metabolism in drosophila diet-induced insulin resistance. mol Cell Biol. 2018;38(2). doi:10.1128/MCB.00259-17
30. Guo X, Asthana P, Gurung S, et al. Regulation of age-associated insulin resistance by MT1-MMP-mediated cleavage of insulin receptor. Nat Commun. 2022;13(1):3749.
31. Ivanov SM. Transcriptome-based analysis of human peripheral blood reveals regulators of immune response in different viral infections. Front Immunol. 2023;14.
32. Tian C, Chang H, La X, Li J. Wushenziye formula improves skeletal muscle insulin resistance in type 2 diabetes mellitus via PTP1B‐IRS1‐Akt‐GLUT4 signaling pathway. Evid Based Complement Altern Med. 2017;2017(1). doi:10.1155/2017/4393529
33. Yu N, Fang X, Zhao D, et al. Anti-diabetic effects of Jiang Tang Xiao Ke granule via PI3K/Akt signalling pathway in type 2 diabetes KKAy mice. PLoS One. 2017;12(1):e0168980. doi:10.1371/journal.pone.0168980
34. Sun Y, Shi H, Yin S, et al. Human mesenchymal stem cell derived exosomes alleviate type 2 diabetes mellitus by reversing peripheral insulin resistance and relieving Β-cell destruction. ACS Nano. 2018;12(8):7613–7628. doi:10.1021/acsnano.7b07643
35. Chen G, Yang XX, Yang XX, et al. Jia-Wei-Jiao-Tai-Wan ameliorates type 2 diabetes by improving Β cell function and reducing insulin resistance in diabetic rats. BMC Complement Altern Med. 2017;17(1). doi:10.1186/s12906-017-2016-5
36. Taha IM, Abdu Allah AM, Abd El Gayed EM. Expression of toll-like receptor 4 and its connection with type 2 diabetes mellitus. Cell mol Biol. 2018;64(13):15–20. doi:10.14715/cmb/2018.64.13.4
37. Qatanani M, Tan Y, Dobrin R, et al. Inverse regulation of inflammation and mitochondrial function in adipose tissue defines extreme insulin sensitivity in morbidly obese patients. Diabetes. 2013;62(3):855–863. doi:10.2337/db12-0399
38. Lee YS, Li PP, Huh JY, et al. Inflammation is necessary for long-term but not short-term high-fat diet–induced insulin resistance. Diabetes. 2011;60(10):2474–2483. doi:10.2337/db11-0194
39. Ieronymaki E, Theodorakis EM, Lyroni K, et al. Insulin resistance in macrophages alters their metabolism and promotes an M2-like phenotype. J Immunol. 2019;202(6):1786–1797. doi:10.4049/jimmunol.1800065
40. Zhou W, Sailani MR, Contrepois K, et al. Longitudinal multi-omics of host–microbe dynamics in prediabetes. Nature. 2019;569(7758):663–71. doi:10.1038/s41586-019-1236-x
41. Bronsart LL, Contag CH. A role of the adaptive immune system in glucose homeostasis. BMJ Open Diabetes Res Care. 2016;4(1):e000136. doi:10.1136/bmjdrc-2015-000136
42. Wu H, Perrard XD, Wang Q, et al. CD11c expression in adipose tissue and blood and its role in diet-induced obesity. Arterioscler Thromb Vasc Biol. 2010;30(2):186–192. doi:10.1161/ATVBAHA.109.198044
43. Saltiel AR, Olefsky JM. Inflammatory mechanisms linking obesity and metabolic disease. J Clin Invest. 2017;127(1):1–4. doi:10.1172/JCI92035
44. Gao D, Madi M, Ding C, et al. Interleukin-1β mediates macrophage-induced impairment of insulin signaling in human primary adipocytes. Ajp Endocrinol Metab. 2014;307(3):E289–304. doi:10.1152/ajpendo.00430.2013
45. Wen H, Gris D, Lei Y, et al. Fatty acid–induced NLRP3-ASC inflammasome activation interferes with insulin signaling. Nat Immunol. 2011;12(5):408–415. doi:10.1038/ni.2022
46. Zeng T, Liu F, Zhou J, Shixiu P, Xia W, Chen L. Depletion of Kupffer cells attenuates systemic insulin resistance, inflammation and improves liver autophagy in high-fat diet fed mice. Endocr J. 2015;62(7):615–626. doi:10.1507/endocrj.EJ15-0046
47. Morita Y, Senokuchi T, Yamada S, et al. Impact of tissue macrophage proliferation on peripheral and systemic insulin resistance in obese mice with diabetes. BMJ Open Diabetes Res Care. 2020;8(1):e001578. doi:10.1136/bmjdrc-2020-001578
48. Lanthier N, Molendi‐Coste O, Cani PD, Rooijen N, Horsmans Y, Leclercq IA. Kupffer cell depletion prevents but has no therapeutic effect on metabolic and inflammatory changes induced by a high‐fat diet. FASEB J. 2011;25(12):4301–4311. doi:10.1096/fj.11-189472
49. Eguchi K, Nagai R. Islet inflammation in type 2 diabetes and physiology. J Clin Invest. 2017;127(1):14–23. doi:10.1172/JCI88877
50. Melloul D. The role of obesity-induced inflammation in pancreatic -cell dysfunction and death. Interv Obes Diabetes. 2020;3(5).
51. Ivovic A, Yung JHM, Oprescu AI. Β-cell insulin resistance plays a causal role in fat-induced β-cell dysfunction in vitro and in vivo. Endocrinology. 2024;165(5). doi:10.1210/endocr/bqae044
52. Dludla PV, Mabhida SE, Ziqubu K, et al. Pancreatic Β-cell dysfunction in type 2 diabetes: implications of inflammation and oxidative stress. World J Diabetes. 2023;14(3):130–146. doi:10.4239/wjd.v14.i3.130
53. Vong CT, Tseng HHL, Kwan YW, Lee SM, Hoi MPM. Antrodia camphorata increases insulin secretion and protects from apoptosis in MIN6 cells. Front Pharmacol. 2016;7. doi:10.3389/fphar.2016.00067
54. Ratter JM, van Heck JIP, Rooijackers HMM. Insulin acutely activates metabolism of primary human monocytes and promotes a proinflammatory phenotype. J Leukoc Biol. 2021;110(5):885–891. doi:10.1002/JLB.3AB0120-019RR
55. Ji Z, Zhang X, Ji L, Jiang G. Identification of potential biomarkers of type 2 diabetes mellitus-related immune infiltration using weighted gene coexpression network analysis. Biomed Res Int. 2022;2022:1–14.
56. Torres‐Hernandez A, Wang W, Nikiforov YE, et al. Γδ T cells promote steatohepatitis by orchestrating innate and adaptive immune programming. Hepatology. 2019;71(2):477–494. doi:10.1002/hep.30952
57. Radzikowska U, Rinaldi A, Sözener ZÇ, et al. The influence of dietary fatty acids on immune responses. Nutrients. 2019;11(12):2990. doi:10.3390/nu11122990
58. Hidalgo MA, Carretta MD, Burgos RA. Long chain fatty acids as modulators of immune cells function: contribution of FFA1 and FFA4 receptors. Front Physiol. 2021;12.
59. Villarreal-Pérez JZ, Villarreal-Martínez JZ, Lavalle-González FJ, et al. Plasma and urine metabolic profiles are reflective of altered beta-oxidation in non-diabetic obese subjects and patients with type 2 diabetes mellitus. Diabetol Metab Syndr. 2014;6(1).
60. Reijrink M, van Ark J, Lexis CP, et al. increased frequency of proangiogenic tunica intima endothelial kinase 2 (Tie2) expressing monocytes in individuals with type 2 diabetes mellitus. Cardiovasc Diabetol. 2022;21(1).
61. Li P, Oh DY, Bandyopadhyay G, et al. LTB4 promotes insulin resistance in obese mice by acting on macrophages, hepatocytes and myocytes. Nat Med. 2015;21(3):239–247. doi:10.1038/nm.3800
62. ter Horst KW, Gilijamse PW, Versteeg RI. Hepatic diacylglycerol-associated protein kinase cε translocation links hepatic steatosis to hepatic insulin resistance in humans. Cell Rep. 2017;19(10):1997–2004. doi:10.1016/j.celrep.2017.05.035
63. Han CY, Rho HS, Kim A, et al. FXR inhibits endoplasmic reticulum stress-induced NLRP3 inflammasome in hepatocytes and ameliorates liver injury. Cell Rep. 2018;24(11):2985–2999. doi:10.1016/j.celrep.2018.07.068
64. Wu H, Ballantyne CM. Skeletal muscle inflammation and insulin resistance in obesity. J Clin Invest. 2017;127(1):43–54. doi:10.1172/JCI88880
65. Mashili F, Chibalin AV, Krook A, Zierath JR. Constitutive STAT3 phosphorylation contributes to skeletal muscle insulin resistance in type 2 diabetes. Diabetes. 2013;62(2):457–465. doi:10.2337/db12-0337
66. Spite M, Hellmann J, Tang Y, et al. Deficiency of the leukotriene B4 receptor, BLT-1, protects against systemic insulin resistance in diet-induced obesity. J Immunol. 2011;187(4):1942–1949. doi:10.4049/jimmunol.1100196
67. Li P, Liu S, Lü M, et al. Hematopoietic-derived galectin-3 causes cellular and systemic insulin resistance. Cell. 2016;167(4):973–984.e12. doi:10.1016/j.cell.2016.10.025
68. Yang B, Yang X, Tan X, et al. Regulatory networks, management approaches, and emerging treatments of nonalcoholic fatty liver disease. Can J Gastroenterol Hepatol. 2022;2022:1–18.
69. Koc M, Šiklová M, Šrámková V, et al. Signs of deregulated gene expression are present in both CD14+ and CD14- PBMC from non-obese men with family history of T2DM. Front Endocrinol. 2021;11.
70. Eller K, Kirsch AH, Wolf AM, et al. Potential role of regulatory t cells in reversing obesity-linked insulin resistance and diabetic nephropathy. Diabetes. 2011;60(11):2954–2962. doi:10.2337/db11-0358
71. Wensveen FM, Jelenčić V, Valentić S, et al. NK cells link obesity-induced adipose stress to inflammation and insulin resistance. Nat Immunol. 2015;16(4):376–385. doi:10.1038/ni.3120
72. Berbudi A, Surendar J, Ajendra J, et al. Filarial infection or antigen administration improves glucose tolerance in diet-induced obese mice. J Innate Immun. 2016;8(6):601. doi:10.1159/000448401
73. Mahmoud F, Haines D, Dashti AA, El-Shazly S, Al-najjar FA. Correlation between heat shock proteins, adiponectin, and T lymphocyte cytokine expression in type 2 diabetics. Cell Stress Chaperones. 2018;23(5):955–965. doi:10.1007/s12192-018-0903-4
74. Femminella GD, Livingston NR, Raza S, et al. Does insulin resistance influence neurodegeneration in non-diabetic alzheimer’s subjects? Alzheimer S Res Ther. 2021;13(1).
75. Weir GC. Glucolipotoxicity, Β-cells, and diabetes: the emperor has no clothes. Diabetes. 2020;69(3):273–278. doi:10.2337/db19-0138
76. Butcher M, Hallinger DR, Garcia E, et al. Association of proinflammatory cytokines and islet resident leucocytes with islet dysfunction in type 2 diabetes. Diabetologia. 2014;57(3):491–501. doi:10.1007/s00125-013-3116-5
77. DeFuria J, Belkina AC, Jagannathan-Bogdan M, et al. B cells promote inflammation in obesity and type 2 diabetes through regulation of T-cell function and an inflammatory cytokine profile. Proc Natl Acad Sci. 2013;110(13):5133–5138. doi:10.1073/pnas.1215840110
78. Wu L, Parekh VV, Hsiao J, Kitamura D, Kaer LV. Spleen supports a pool of innate-like b cells in white adipose tissue that protects against obesity-associated insulin resistance. Proc Natl Acad Sci. 2014;111(43). doi:10.1073/pnas.1324052111
79. Macdougall CE, Wood EG, Solomou A, et al. Constitutive activation of β-catenin in conventional dendritic cells increases the insulin reserve to ameliorate the development of type 2 diabetes in mice. Diabetes. 2019;68(7):1473–1484. doi:10.2337/db18-1243
80. Oliveira AG, Araújo TG, Carvalho BM, et al. Acute exercise induces a phenotypic switch in adipose tissue macrophage polarization in diet‐induced obese rats. Obesity. 2013;21(12):2545–2556. doi:10.1002/oby.20402
81. Yasui-Kato M, Patlada S, Yokode M, Kamei K, Minami M. EP4 signalling is essential for controlling islet inflammation by causing a shift in macrophage polarization in obesity/type 2 diabetes. Diab Vasc Dis Res. 2020;17(4):147916412094567. doi:10.1177/1479164120945675
82. Ma S, Wang J, Wang Y, et al. Diabetes mellitus impairs white matter repair and long-term functional deficits after cerebral ischemia. Stroke. 2018;49(10):2453–2463. doi:10.1161/STROKEAHA.118.021452
83. Pardali E, Makowski L, Leffers M, Borgscheiper A, Waltenberger J. BMP‐2 induces human mononuclear cell chemotaxis and adhesion and modulates monocyte‐to‐macrophage differentiation. J Cell Mol Med. 2018;22(11):5429–5438. doi:10.1111/jcmm.13814
84. Jagannathan-Bogdan M, McDonnell ME, Shin H, et al. Elevated proinflammatory cytokine production by a skewed t cell compartment requires monocytes and promotes inflammation in type 2 diabetes. J Immunol. 2011;186(2):1162–1172. doi:10.4049/jimmunol.1002615
85. Šiklová M, Krauzová E, Svobodová B, et al. Circulating monocyte and lymphocyte populations in healthy first-degree relatives of type 2 diabetic patients at fasting and during short-term hyperinsulinemia. Mediators Inflamm. 2019;2019:1–7. doi:10.1155/2019/1491083
86. Zhu Y, Xu M, Li J. Peripheral white blood cell subtypes and the development/progression of diabetic macular edema in type 2 diabetic patients: a comparative study. Ann Palliat Med. 2022;11(9):2887–2896. doi:10.21037/apm-22-962
87. Kula A. Retrospective analysis of the relationship between neutrophil-to-lymphocyte ratio, platelet-to-lymphocyte ratio, and glycemic regulation in patients with type 2 diabetes mellitus followed up at an internal medicine outpatient clinic. J Surg Med. 2023;7(7):404–408. doi:10.28982/josam.7795
88. Adane T, Melku M, Worku YB. The association between neutrophil-to-lymphocyte ratio and glycemic control in type 2 diabetes mellitus: a systematic review and meta-analysis. J Diabetes Res. 2023;2023:1–11. doi:10.1155/2023/3117396
89. Qiao Y-C, Shen J, He L, et al. Changes of regulatory t cells and of proinflammatory and immunosuppressive cytokines in patients with type 2 diabetes mellitus: a systematic review and meta-analysis. J Diabetes Res. 2016;2016:1–19. doi:10.1155/2016/3694957
90. Jia Y, Zhao Y, Li C, Shao R. The expression of programmed death-1 on CD4+ and CD8+ T lymphocytes in patients with type 2 diabetes and severe sepsis. PLoS One. 2016;11(7):e0159383. doi:10.1371/journal.pone.0159383
91. Li Y, Dong X, He W. Ube2L6 promotes M1 macrophage polarization in high-fat diet-fed obese mice via ISGylation of STAT1 to trigger STAT1 activation. Obes Facts. 2023;17(1):24–36. doi:10.1159/000533966
92. Wang H, Cao K, Liu S, Xu Y, Tang L. Tim-3 expression causes NK cell dysfunction in type 2 diabetes patients. Front Immunol. 2022;13.
93. Saika F, Kiguchi N, Matsuzaki S, Kobayashi D, Kishioka S. Inflammatory macrophages in the sciatic nerves facilitate neuropathic pain associated with type 2 diabetes mellitus. J Pharmacol Exp Ther. 2019;368(3):535–544. doi:10.1124/jpet.118.252668
94. Berbudi A, Ajendra J, Wardani AP, Hoerauf A, Hübner MP. Parasitic helminths and their beneficial impact on type 1 and type 2 diabetes. Diabetes Metab Res Rev. 2016;32(3):238–250. doi:10.1002/dmrr.2673
95. Saraswathi V, Ramnanan CJ, Wilks AW, et al. Impact of hematopoietic cyclooxygenase-1 deficiency on obesity-linked adipose tissue inflammation and metabolic disorders in mice. Metabolism. 2013;62(11):1673–1685. doi:10.1016/j.metabol.2013.07.007
96. Jourdan T, Godlewski G, Çınar R, et al. Activation of the Nlrp3 inflammasome in infiltrating macrophages by endocannabinoids mediates beta cell loss in type 2 diabetes. Nat Med. 2013;19(9):1132–1140. doi:10.1038/nm.3265
97. Lee WJ, Tateya S, Cheng AM. M2 macrophage polarization mediates anti-inflammatory effects of endothelial nitric oxide signaling. Diabetes. 2015;64(8):2836–2846. doi:10.2337/db14-1668
98. Margaryan S, Witkowicz A, Arakelyan A, Partyka A, Karabon L, Manukyan G. sFasL-mediated induction of neutrophil activation in patients with type 2 diabetes mellitus. PLoS One. 2018;13(7):e0201087. doi:10.1371/journal.pone.0201087
99. Kwak SJ, Kim CS, Choi MS, et al. The soy peptide phe–leu–val reduces TNFα-induced inflammatory response and insulin resistance in adipocytes. J Med Food. 2016;19(7):678–685. doi:10.1089/jmf.2016.3685
100. Swaroop JJ, Rajarajeswari D, Naidu JN. Association of TNF-α with insulin resistance in type 2 diabetes mellitus. Indian J Med Res. 2012;135(1):127. doi:10.4103/0971-5916.93435
101. Zheng X, Ke Y, Feng A, et al. The mechanism by which amentoflavone improves insulin resistance in HepG2 cells. Molecules. 2016;21(5):624. doi:10.3390/molecules21050624
102. Fan L, Ding L, Lan J, Niu J, He Y, Song L. Fibroblast growth factor-1 improves insulin resistance via repression of JNK-mediated inflammation. Front Pharmacol. 2019;10. doi:10.3389/fphar.2019.01478
103. Maris M, Overbergh L, Gysemans C, et al. Deletion of C/Ebp homologous protein (Chop) in C57Bl/6 mice dissociates obesity from insulin resistance. Diabetologia. 2012;55(4):1167–1178. doi:10.1007/s00125-011-2427-7
104. Hajmrle C, Smith N, Spigelman AF, et al. Interleukin-1 signaling contributes to acute islet compensation. Jci Insight. 2016;1(4). doi:10.1172/jci.insight.86055
105. Böni-Schnetzler M, Méreau H, Rachid L, et al. IL-1beta promotes the age-associated decline of beta cell function. Iscience. 2021;24(11):103250. doi:10.1016/j.isci.2021.103250
106. Santos RSD, Guzman-Llorens D, Perez-Serna AA, Nadal A, Marroquí L. Deucravacitinib, a tyrosine kinase 2 pseudokinase inhibitor, protects human beta cells against proinflammatory insults. bioRxiv. 2022;2022–12.
107. Koefoed MM, Larsen CM, Faulenbach M, et al. Serum proteome pool changes in type 2 diabetic patients treated with anakinra. Clin Proteomics. 2010;6(4):153–161. doi:10.1007/s12014-010-9056-x
108. Benner C, van der Meulen T, Cacéres E, Tigyi K, Donaldson CJ, Huising MO. The transcriptional landscape of mouse beta cells compared to human beta cells reveals notable species differences in long non-coding RNA and protein-coding gene expression. BMC Genomics. 2014;15(1).
109. Farooq N, Chuan B, Mahmud H, et al. Association of the systemic host immune response with acute hyperglycemia in mechanically ventilated septic patients. PLoS One. 2021;16(3):e0248853. doi:10.1371/journal.pone.0248853
110. Kim DH, Lee B, Lee JJSJ, et al. FoxO6-mediated IL-1β induces hepatic insulin resistance and age-related inflammation via the TF/PAR2 pathway in aging and diabetic mice. Redox Biol. 2019;24:101184. doi:10.1016/j.redox.2019.101184
111. Yu Q, Xia Z, Liong EC, Tipoe GL. Chronic aerobic exercise improves insulin sensitivity and modulates Nrf2 and NF‑κB/IκBα pathways in the skeletal muscle of rats fed with a high fat diet. Mol Med Rep. 2019. doi:10.3892/mmr.2019.10787
112. Eissa LA, Elsherbiny NM, Maghmomeh AO. Effect of 2-hydroxychalcone on adiponectin level in type 2 diabetes induced experimentally in rats. Egypt J Basic Appl Sci. 2017;4(1):1–8.
113. Karajibani M, Montazerifar F, Sadeghi M, Keikhaie MA, Dashipour A. Serum fetuin-a and adipsin levels in type II diabetes patients. Int J High Risk Behav Addict. 2019;8(3). doi:10.5812/ijhrba.91963
114. Shahnawaz W, Suhail N, Siddiqui MAI, Yasmeen S, Fatima SS. Does lipocalin-2 affect metabolic syndrome in hepatic infections? Cureus. 2020. doi:10.7759/cureus.10040
115. Neuparth MJ, Proença JB, Santos-Silva A, Coimbra S. Adipokines, oxidized low-density lipoprotein, and c-reactive protein levels in lean, overweight, and obese Portuguese patients with type 2 diabetes. Isrn Obes. 2013;2013:1–7. doi:10.1155/2013/142097
116. Tönjes A, Faßhauer M, Kratzsch J, Stümvoll M, Blüher M. Adipokine pattern in subjects with impaired fasting glucose and impaired glucose tolerance in comparison to normal glucose tolerance and diabetes. PLoS One. 2010;5(11):e13911. doi:10.1371/journal.pone.0013911
117. Karczewska-Kupczewska M, Nikołajuk A, Stefanowicz M, Matulewicz N, Kowalska I, Strączkowski M. Serum and adipose tissue chemerin is differentially related to insulin sensitivity. Endocr Connect. 2020;9(5):360–369. doi:10.1530/EC-20-0084
118. Guo D, Yuan Y, Huang R, et al. Association between plasma adipsin level and mild cognitive impairment in Chinese patients with type 2 diabetes: a cross-sectional study. BMC Endocr Disord. 2019;19(1). doi:10.1186/s12902-019-0431-y
119. Batista TM, García‐Martín R, Cai W, et al. Multi-dimensional transcriptional remodeling by physiological insulin in vivo. Cell Rep. 2019;26(12):3429–3443.e3. doi:10.1016/j.celrep.2019.02.081
120. Macia L, Yulyaningsih E, Pangon L, et al. Neuropeptide Y1 receptor in immune cells regulates inflammation and insulin resistance associated with diet-induced obesity. Diabetes. 2012;61(12):3228–3238. doi:10.2337/db12-0156
121. Matsuzawa T, Kim B-H, Shenoy AR, Kamitani S, Miyake M, MacMicking JD. IFN-γ elicits macrophage autophagy via the P38 MAPK signaling pathway. J Immunol. 2012;189(2):813–818. doi:10.4049/jimmunol.1102041
122. Song A, Xu M, Bi Y, et al. Serum fetuin-a associates with type 2 diabetes and insulin resistance in Chinese adults. PLoS One. 2011;6(4):e19228. doi:10.1371/journal.pone.0019228
123. Pamuk BÖ, Yilmaz H, Topcuoglu T, et al. Fetuin-a levels in hyperthyroidism. Clinics. 2013;68(3):379–383. doi:10.6061/clinics/2013(03)OA15
124. Kyohara M, Shirakawa J, Okuyama T, et al. Soluble EGFR, a hepatokine, and adipsin, an adipokine, are biomarkers correlated with distinct aspects of insulin resistance in type 2 diabetes subjects. Diabetol Metab Syndr. 2020;12(1). doi:10.1186/s13098-020-00591-7
125. Wang J, Lee W, Lee I-T, et al. Association between serum adipsin levels and insulin resistance in subjects with various degrees of glucose intolerance. J Endocr Soc. 2018;3(2):403–410. doi:10.1210/js.2018-00359
126. Zhou Q, Ge Q, Ding Y, et al. Relationship between serum adipsin and the first phase of glucose‐stimulated insulin secretion in individuals with different glucose tolerance. J Diabetes Investig. 2018;9(5):1128–1134. doi:10.1111/jdi.12819
127. Adela R, Reddy PNC, Ghosh TS, et al. Serum protein signature of coronary artery disease in type 2 diabetes mellitus. J Transl Med. 2019;17(1). doi:10.1186/s12967-018-1755-5
128. Fang Y, Wang X, Li W, et al. Screening of circular RNAs and validation of circANKRD36 associated with inflammation in patients with type 2 diabetes mellitus. Int J Mol Med. 2018. doi:10.3892/ijmm.2018.3783
129. Venuraju S, Lahiri A, Jeevarethinam A, Rakhit R, Shah PK, Nilsson J. Association of epicardial fat volume with the extent of coronary atherosclerosis and cardiovascular adverse events in asymptomatic patients with diabetes. Angiology. 2021;72(5):442–450. doi:10.1177/0003319720984607
130. Gamas L, Matafome P, Seiça R. Irisin and myonectin regulation in the insulin resistant muscle: implications to adipose tissue: muscle crosstalk. J Diabetes Res. 2015;2015:1–8. doi:10.1155/2015/359159
131. Eckel J. Myokines in metabolic homeostasis and diabetes. Diabetologia. 2019;62(9):1523–1528. doi:10.1007/s00125-019-4927-9
132. Shi X, Lin M, Liu C, et al. Elevated circulating Irisin is associated with lower risk of insulin resistance: association and path analyses of obese Chinese adults. BMC Endocr Disord. 2016;16(1). doi:10.1186/s12902-016-0123-9
133. Villarroya F. Turning up the heat. Cell Metab. 2012;15(3):277–278. doi:10.1016/j.cmet.2012.02.010
134. Yan B, Shi X, Zhang H, et al. Association of serum irisin with metabolic syndrome in obese Chinese adults. PLoS One. 2014;9(4):e94235. doi:10.1371/journal.pone.0094235
135. Saleh AA, Amin E, Elfallah AA, Hamed AM. Insulin resistance and idiopathic infertility: a potential possible link. Andrologia. 2020;52(11). doi:10.1111/and.13773
136. Yano N, Zhang L, Wei D, et al. Irisin counteracts high glucose and fatty acid-induced cytotoxicity by preserving the AMPK-insulin receptor signaling axis in C2C12 myoblasts. Ajp Endocrinol Metab. 2020;318(5):E791–805. doi:10.1152/ajpendo.00219.2019
137. Mai S, Fintini D, Mele C, et al. Circulating Irisin in children and adolescents with Prader-Willi syndrome: relation with glucose metabolism. Front Endocrinol. 2022;13.
138. Kulthinee S, Wang L, Yano N, et al. Irisin preserves cardiac performance and insulin sensitivity in response to hemorrhage. Pharmaceuticals. 2022;15(10):1193. doi:10.3390/ph15101193
139. Wu CZ, Chu N-F, Chang LC, et al. The relationship of Irisin with metabolic syndrome components and insulin secretion and resistance in schoolchildren. Medicine. 2021;100(5):e24061. doi:10.1097/MD.0000000000024061
140. Camporez JP, Jornayvaz FR, Petersen MC, et al. Cellular mechanisms by which FGF21 improves insulin sensitivity in male mice. Endocrinology. 2013;154(9):3099–3109. doi:10.1210/en.2013-1191
141. Pal D, Dasgupta S, Kundu R, et al. Fetuin-a acts as an endogenous ligand of TLR4 to promote lipid-induced insulin resistance. Nat Med. 2012;18(8):1279–1285. doi:10.1038/nm.2851
142. Costantini D. A meta-analysis of impacts of immune response and infection on oxidative status in vertebrates. Conserv Physiol. 2022;10(1). doi:10.1093/conphys/coac018
143. Zhang W, Zhong R, Qu X, Xiang Y, Ji M. Effect of 8-hydroxyguanine DNA Glycosylase 1 on the function of immune cells. Antioxidants. 2023;12(6):1300. doi:10.3390/antiox12061300
144. Sies H. Oxidative stress: a concept in redox biology and medicine. Redox Biol. 2015;4:180–183. doi:10.1016/j.redox.2015.01.002
145. Rong D. Experimentally validated oxidative stress -associated prognostic signatures describe the immune landscape and predict the drug response and prognosis of SKCM. Front Immunol. 2024;15.
146. Hersperger F, Meyring T, Weber P, et al. DNA damage signaling in drosophila macrophages modulates systemic cytokine levels in response to oxidative stress. Elife. 2024;12.
147. Chaudhuri R, Krycer JR, Fazakerley DJ, et al. The transcriptional response to oxidative stress is part of, but not sufficient for, insulin resistance in adipocytes. Sci Rep. 2018;8(1). doi:10.1038/s41598-018-20104-x
148. Bahnemiri MG, Nouri HR, Zabihi E, Sadeghi F, Pouramir M. Effects of arbutin on glucose uptake by glucose transporter 4 (GLUT4) and its cytoprotective properties in l6 skeletal muscle cell line. Cell Biochem Funct. 2022;40(4):417–425. doi:10.1002/cbf.3706
149. Katsura T, Kawamori D, Aida E, Matsuoka T, Shimomura I. Glucotoxicity induces abnormal glucagon secretion through impaired insulin signaling in InR1G cells. PLoS One. 2017;12(4):e0176271. doi:10.1371/journal.pone.0176271
150. Boden G, Homko CJ, Barrero C, et al. Excessive caloric intake acutely causes oxidative stress, GLUT4 carbonylation, and insulin resistance in healthy men. Sci Transl Med. 2015;7(304). doi:10.1126/scitranslmed.aac4765
151. Kaewlert W, Sakonsinsiri C, Lert-itthiporn W, et al. Overexpression of Insulin Receptor Substrate 1 (IRS1) relates to poor prognosis and promotes proliferation, stemness, migration, and oxidative stress resistance in cholangiocarcinoma. Int J mol Sci. 2023;24(3):2428. doi:10.3390/ijms24032428
152. Fazakerley DJ, Minard AY, Krycer JR, et al. Mitochondrial oxidative stress causes insulin resistance without disrupting oxidative phosphorylation. J Biol Chem. 2018;293(19):7315–7328. doi:10.1074/jbc.RA117.001254
153. Dregan A, Charlton JL, Chowienczyk P, Gulliford M. Chronic inflammatory disorders and risk of type 2 diabetes mellitus, coronary heart disease, and stroke. Circulation. 2014;130(10):837–844. doi:10.1161/CIRCULATIONAHA.114.009990
154. Mahdi A, Kövamees O, Checa A. Arginase inhibition improves endothelial function in patients with type 2 diabetes mellitus despite intensive glucose‐lowering therapy. J Intern Med. 2018;284(4):388–398. doi:10.1111/joim.12785
155. Kuryłowicz A, Koźniewski K. Anti-inflammatory strategies targeting metaflammation in type 2 diabetes. Molecules. 2020;25(9):2224. doi:10.3390/molecules25092224
156. Alshahrani A, Aljada A, Masood A. Proteomic profiling identifies distinct regulation of proteins in obese diabetic patients treated with metformin. Pharmaceuticals. 2023;16(10):1345. doi:10.3390/ph16101345
157. Winer S, Winer DA. The adaptive immune system as a fundamental regulator of adipose tissue inflammation and insulin resistance. Immunol Cell Biol. 2012;90(8):755–762.
158. Perry R, Camporez JP, Kursawe R, et al. Hepatic acetyl coa links adipose tissue inflammation to hepatic insulin resistance and type 2 diabetes. Cell. 2015;160(4):745–758. doi:10.1016/j.cell.2015.01.012
159. Sun C, Sun L, Ma H, et al. The phenotype and functional alterations of macrophages in mice with hyperglycemia for long term. J Cell Physiol. 2012;227(4):1670–1679. doi:10.1002/jcp.22891
160. Bhargava P, Lee C. Role and function of macrophages in the metabolic syndrome. Biochem J. 2012;442(2):253–262. doi:10.1042/BJ20111708
161. Gallagher IJ, Schéele C, Keller P, et al. Integration of microRNA changes in vivo identifies novel molecular features of muscle insulin resistance in type 2 diabetes. Genome Med. 2010;2(2). doi:10.1186/gm130
162. Zhang H, Dellsperger KC, Zhang C. The link between metabolic abnormalities and endothelial dysfunction in type 2 diabetes: an update. Basic Res Cardiol. 2011;107(1). doi:10.1007/s00395-011-0237-1
163. Dai M, Wu L, Wang P, Wen Z, Xu X, Wang DW. CYP2J2 and its metabolites EETs attenuate insulin resistance via regulating macrophage polarization in adipose tissue. Sci Rep. 2017;7(1):46743.
164. Xu L, Yan X, Zhao Y, et al. Macrophage polarization mediated by mitochondrial dysfunction induces adipose tissue inflammation in obesity. Int J mol Sci. 2022;23(16):9252. doi:10.3390/ijms23169252
165. Viardot A, Heilbronn LK, Samocha-Bonet D, Mackay F, Campbell LV, Samaras K. Obesity is associated with activated and insulin resistant immune cells. Diabetes Metab Res Rev. 2012;28(5):447–454. doi:10.1002/dmrr.2302
166. Hong X, Zhang X, You L, et al. Original research: association between adiponectin and newly diagnosed type 2 diabetes in population with the clustering of obesity, dyslipidaemia and hypertension: a cross-sectional study. BMJ Open. 2023;13(2):e060377. doi:10.1136/bmjopen-2021-060377
167. Kamei N, Tanaka M, Choi H, et al. Effect of an enhanced nose-to-brain delivery of insulin on mild and progressive memory loss in the senescence-accelerated mouse. Mol Pharm. 2017;14(3):916–927. doi:10.1021/acs.molpharmaceut.6b01134
168. Kamei N, Okada N, Ikeda T, et al. Effective nose-to-brain delivery of exendin-4 via coadministration with cell-penetrating peptides for improving progressive cognitive dysfunction. Sci Rep. 2018;8(1). doi:10.1038/s41598-018-36210-9
169. Fagninou A, Nekoua MP, Sossou D, Moutaïrou K, Fiévet N, Yessoufou A. Th2-immune polarizing and anti-inflammatory properties of insulin are not effective in type 2 diabetic pregnancy. J Immunol Res. 2020;2020:1–12. doi:10.1155/2020/2038746
170. Belle TLV, Pagni PP, Liao J, et al. Beta-cell specific production of IL6 in conjunction with a mainly intracellular but not mainly surface viral protein causes diabetes. J Autoimmun. 2014;55:24–32. doi:10.1016/j.jaut.2014.02.002
171. Mauer J, Chaurasia B, Plum L, et al. Myeloid cell-restricted insulin receptor deficiency protects against obesity-induced inflammation and systemic insulin resistance. PLoS Genet. 2010;6(5):e1000938. doi:10.1371/journal.pgen.1000938
172. Marín-Juez R, Jong-Raadsen S, Yang S, Spaink HP. Hyperinsulinemia induces insulin resistance and immune suppression via Ptpn6/Shp1 in zebrafish. J Endocrinol. 2014;222(2):229–241. doi:10.1530/JOE-14-0178
173. Watters C, Everett J, Haley CL, Clinton A, Rumbaugh KP. Insulin treatment modulates the host immune system to enhance pseudomonas aeruginosa wound biofilms. Infect Immun. 2014;82(1):92–100. doi:10.1128/IAI.00651-13
174. Humphrey SJ, Yang G, Yang P, et al. Dynamic adipocyte phosphoproteome reveals that akt directly regulates mTORC2. Cell Metab. 2013;17(6):1009–1020. doi:10.1016/j.cmet.2013.04.010
175. Amer J, Salhab A, Noureddin M, et al. Insulin signaling as a potential natural killer cell checkpoint in fatty liver disease. Hepatol Commun. 2018;2(3):285–298. doi:10.1002/hep4.1146
176. Damond N, Thorel F, Moyers JS, et al. Blockade of glucagon signaling prevents or reverses diabetes onset only if residual Β-cells persist. Elife. 2016;5. doi:10.7554/eLife.13828
177. Watson LS, Wilken-Resman B, Williams A, et al. Hyperinsulinemia alters insulin receptor presentation and internalization in brain microvascular endothelial cells. Diab Vasc Dis Res. 2022;19(4):147916412211186. doi:10.1177/14791641221118626
178. Siddle K. Signalling by insulin and IGF receptors: supporting acts and new players. J Mol Endocrinol. 2011;47(1):R1–10. doi:10.1530/JME-11-0022
179. Kra G, Daddam JR, Moallem U, et al. Alpha-linolenic acid modulates systemic and adipose tissue-specific insulin sensitivity, inflammation, and the endocannabinoid system in dairy cows. Sci Rep. 2023;13(1). doi:10.1038/s41598-023-32433-7
180. Dik B, Bahcivan E, Faki HE, Üney K. Combined treatment with interleukin-1 and tumor necrosis factor-alpha antagonists improve type 2 diabetes in rats. Can J Physiol Pharmacol. 2018;96(8):751–756. doi:10.1139/cjpp-2017-0769
181. Biswas S. Association of circulating fasting TNF-Α with hyperglycemia is stronger than with body mass index in newly diagnosed bangladeshi type 2 diabetic subjects. J Diabetes Metab. 2014;05(09). doi:10.4172/2155-6156.1000429
182. Ezzat O, Ibrahim SS, Gaber HA. The effect of periodontal treatment on circulating tumor necrosis factor-alpha and insulin resistance in patients with type 2 diabetes mellitus. J King Abdulaziz Univ Sci. 2012;19(4):25–45.
183. Pfeifer E, Saxon D, Janson RW. Etanercept-induced hypoglycemia in a patient with psoriatic arthritis and diabetes. J Investig Med High Impact Case Reports. 2017;5(3).
184. Kim W, Lao Q, Shin Y-K, et al. Cannabinoids induce pancreatic β-cell death by directly inhibiting insulin receptor activation. Sci Signal. 2012;5(216). doi:10.1126/scisignal.2002519
185. Bilbao D, Luciani L, Jóhannesson B, Piszczek A, Rosenthal N. Insulin‐like growth factor‐1 stimulates regulatory <scp>t</scp> cells and suppresses autoimmune disease. Embo Mol Med. 2014;6(11):1423–1435. doi:10.15252/emmm.201303376
186. McNitt DH, Joosse BA, Thomas JW, Bonami RH. Productive germinal center responses depend on the nature of stimuli received by anti-insulin B cells in type 1 diabetes–prone mice. Immunohorizons. 2023;7(6):384–397. doi:10.4049/immunohorizons.2300036
187. Mariyam SB, Muthubeevi SB, Vasantha SC. Serum C-peptide level in obese and non-obese patients with type 2 diabetes mellitus. J Evol Med Dent Sci. 2017;6(05):350–353. doi:10.14260/Jemds/2017/79
188. Gujral UP, Pradeepa R, Weber MB, Narayan KMV, Mohan V. Type 2 diabetes in south asians: similarities and differences with white caucasian and other populations. Ann N Y Acad Sci. 2013;1281(1):51–63. doi:10.1111/j.1749-6632.2012.06838.x
189. Luo W, Zhang J, Luo Y. Factors associated with glycemic control in patients with T2DM: evidence from a cross-sectional study in China. BMC Endocr Disord. 2024;24(1). doi:10.1186/s12902-024-01605-5
190. da Costa Fernandes CJ, da Cruz Rodrigues KC, de Melo DG, et al. Short-term strength exercise reduces the macrophage M1/M2 ratio in white adipose tissue of obese animals. Life Sci. 2023;329:121916. doi:10.1016/j.lfs.2023.121916
191. Yuen AKL, Sugeng Y, Weiland TJ, Jelinek GA. Lifestyle and medication interventions for the prevention or delay of type 2 diabetes mellitus in prediabetes: a systematic review of randomised controlled trials. Aust N Z J Public Health. 2010;34(2):172–178. doi:10.1111/j.1753-6405.2010.00503.x
192. Nunes TCC. Microbial partners in disease: understanding the role of gut microbiota in diabetes. Int J Heal Sci. 2024;4(27):2–14.
193. Walker CG, Solis-Trapala I, Holzapfel C, et al. Modelling the interplay between lifestyle factors and genetic predisposition on markers of type 2 diabetes mellitus risk. PLoS One. 2015;10(7):e0131681. doi:10.1371/journal.pone.0131681
194. Simões BES, Muniz MRR, Araujo TD, Magalhães Carneiro E, Simionato AVC. Evaluation of tauroursodeoxycholic acid in liver cells’ cultures by MEKC: initial hints to comprehend its role in diabetes mellitus of obese individuals. Electrophoresis. 2024;45(13–14):1252–1264. doi:10.1002/elps.202300223
195. Liu Y, Zhang J, An C. Identification of potential mechanisms of Rk1 combination with Rg5 in the treatment of type II diabetes mellitus by integrating network pharmacology and experimental validation. Int J mol Sci. 2023;24(19):14828. doi:10.3390/ijms241914828
196. Aierken A, Li B, Liu P, et al. Melatonin treatment improves human umbilical cord mesenchymal stem cell therapy in a mouse model of type ii diabetes mellitus via the PI3K/AKT signaling pathway. Stem Cell Res Ther. 2022;13(1). doi:10.1186/s13287-022-02832-0
197. Su C, Cheng Q, Wang L. Roux-en-Y gastric bypass improves hepatic glucose metabolism involving upregulation of Sirt1 in type 2 diabetes mellitus. Diabetes, Metab Syndr Obes Targets Ther. 2021;Volume 14:2269–2280. doi:10.2147/DMSO.S298897
198. Khoshnejat M, Banaei-Moghaddam AM, Moosavi-Movahedi AA, Kavousi K. A holistic view of muscle metabolic reprogramming through personalized metabolic modeling in newly diagnosed diabetic patients. PLoS One. 2023;18(6):e0287325. doi:10.1371/journal.pone.0287325
199. Chen C, Xiang Q, Li W, Liang S, Yang M, Jing T. Co-expression network revealed roles of RNA m6a methylation in human β-cell of type 2 diabetes mellitus. Front Cell Dev Biol. 2021;9.
200. Yang W. An estrogen receptor Α-derived peptide improves glucose homeostasis during obesity. Nat Commun. 2024;15(1).
201. Reidy PT, Yonemura NM, Madsen JH, et al. An accumulation of muscle macrophages is accompanied by altered insulin sensitivity after reduced activity and recovery. Acta Physiol. 2019;226(2). doi:10.1111/apha.13251
© 2025 The Author(s). This work is published and licensed by Dove Medical Press Limited. The
full terms of this license are available at https://www.dovepress.com/terms.php
and incorporate the Creative Commons Attribution
- Non Commercial (unported, 4.0) License.
By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted
without any further permission from Dove Medical Press Limited, provided the work is properly
attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.
Recommended articles

Platelet-Activating Factor Promotes the Development of Non-Alcoholic Fatty Liver Disease
Yin H, Shi A, Wu J
Diabetes, Metabolic Syndrome and Obesity 2022, 15:2003-2030
Published Date: 8 July 2022

Tang-Ping-San Decoction Remodel Intestinal Flora and Barrier to Ameliorate Type 2 Diabetes Mellitus in Rodent Model
Yin W, Zhang SQ, Pang WL, Chen XJ, Wen J, Hou J, Wang C, Song LY, Qiu ZM, Liang PT, Yuan JL, Yang ZS, Bian Y
Diabetes, Metabolic Syndrome and Obesity 2022, 15:2563-2581
Published Date: 20 August 2022

Relationship Between Acyl and Desacyl Ghrelin Levels with Insulin Resistance and Body Fat Mass in Type 2 Diabetes Mellitus
Zang P, Yang CH, Liu J, Lei HY, Wang W, Guo QY, Lu B, Shao JQ
Diabetes, Metabolic Syndrome and Obesity 2022, 15:2763-2770
Published Date: 7 September 2022

Association Between Triglyceride-Glucose Index and Serum Uric Acid Levels: A Biochemical Study on Anthropometry in Non-Obese Type 2 Diabetes Mellitus Patients
Luo Y, Hao J, He X, Wang C, Zhao H, Zhang Z, Yang L, Ren L
Diabetes, Metabolic Syndrome and Obesity 2022, 15:3447-3458
Published Date: 3 November 2022

The Association Between Sarcopenia and Diabetes: From Pathophysiology Mechanism to Therapeutic Strategy
Chen H, Huang X, Dong M, Wen S, Zhou L, Yuan X
Diabetes, Metabolic Syndrome and Obesity 2023, 16:1541-1554
Published Date: 30 May 2023