Open access peer-reviewed chapter - ONLINE FIRST

Exploring the Function of Inflammatory Routes in Insulin Resistance: Interpreting the Inflammatory Veil of Medusa

Written By

Anchala Kumari

Submitted: 22 April 2024 Reviewed: 29 April 2024 Published: 24 June 2024

DOI: 10.5772/intechopen.1005568

Hypoglycemia - New Insights IntechOpen
Hypoglycemia - New Insights Edited by Alok Raghav

From the Edited Volume

Hypoglycemia - New Insights [Working Title]

Dr. Alok Raghav and M.D. Rimma Shaginian

Chapter metrics overview

14 Chapter Downloads

View Full Metrics

Abstract

A common component of metabolic diseases including metabolic syndrome and type 2 diabetes, insulin resistance is now known to be closely linked to persistent low-grade inflammation. This chapter explores the intricate connection between insulin resistance and inflammatory pathways, clarifying the molecular processes that underlie inflammation-induced insulin resistance. We examine the part that important inflammatory mediators play in upsetting insulin signalling pathways and encouraging insulin resistance, including cytokines, chemokines, and adipokines. We also go over how inflammatory signalling cascades, like the JNK and NF-κB pathways, affect insulin sensitivity and cellular metabolism. Understanding the interaction between insulin resistance and inflammation can help to better understand the pathogenesis of metabolic diseases and identify possible treatment targets. In people who are at risk of developing issues associated with insulin resistance, strategies targeted at reducing inflammatory responses may be able to reduce insulin resistance and enhance metabolic health.

Keywords

  • insulin resistance
  • metabolic syndrome
  • type 2 diabetes
  • Medusa
  • inflammation

1. Introduction

Insulin resistance (IR) is a major contributor to the pathophysiology of metabolic diseases, such as metabolic syndrome and type 2 diabetes mellitus (T2DM), and it has a substantial impact on global health. According to newly available data, insulin resistance develops and progresses as a result of a complex interplay between inflammatory and metabolic pathways and persistent low-grade inflammation. Insulin resistance has many facets, and the idea that it is the “Medusa of the 21st Century” effectively conveys this. One of its veils is inflammation, which obscures its underlying mechanisms.

Several studies have demonstrated the connection between inflammation and insulin resistance, linking inflammatory mediators to insulin signalling pathway disruption and glucose homeostasis impairment [1, 2]. These mediators include cytokines, chemokines, and adipokines. Among them, interleukin-6 (IL-6), adiponectin, and tumour necrosis factor-alpha (TNF-α) have become important participants in the inflammatory cascade, influencing insulin sensitivity both locally and systemically [3, 4].

Furthermore, complex signalling pathways, such as c-Jun N-terminal kinase (JNK) and nuclear factor kappa B (NF-κB), facilitate the interaction between insulin resistance and inflammation, causing metabolic processes to become dysregulated and accelerating the development of metabolic diseases [5, 6]. Insulin receptor substrate (IRS) proteins get phosphorylated when these pro-inflammatory pathways are activated, upsetting insulin signalling cascades and ultimately resulting in insulin resistance [7].

To reduce inflammation and restore metabolic balance, specific therapeutic approaches that comprehend the molecular pathways behind inflammation-induced insulin resistance are necessary. We can create individualised strategies for treating insulin resistance and its related problems by dissecting the intricacies of this complicated interaction.

We explore the role of inflammatory pathways in insulin resistance in this chapter, looking at the molecular mechanisms through which inflammation fuels insulin resistance and the consequences for metabolic health. We seek to clarify the inflammatory veil that covers Medusa and provide insight into prospective targets and tactics for treating insulin resistance-related illnesses by conducting a thorough analysis of recent literature and emerging research discoveries.

Advertisement

2. Literature review

The pathophysiology of insulin resistance and related metabolic problems is largely dependent on persistent low-grade inflammation, as demonstrated by a large number of studies. Clarifying the processes behind inflammation-induced insulin resistance and discovering possible treatment targets require an understanding of the complex interactions between inflammatory mediators and signalling pathways.

TNF-α has been identified as a crucial mediator between inflammation and insulin resistance. Through the activation of serine kinases including c-Jun N-terminal kinase (JNK) and IκB kinase (IKK), which phosphorylate insulin receptor substrate (IRS) proteins and interfere with downstream insulin signalling, TNF-α has been shown in experimental experiments to block insulin signalling pathways [1, 2]. Clinical research has demonstrated increased TNF-α levels in patients with type 2 diabetes mellitus (T2DM) and obesity, further linking TNF-α to the aetiology of insulin resistance [2, 8].

Another pro-inflammatory cytokine linked to the emergence of insulin resistance is interleukin-6 (IL-6). Research has indicated that persistent increase in IL-6 levels impedes insulin signalling and glucose absorption in skeletal muscle, which in turn leads to insulin resistance [9, 10]. Acute-phase proteins like C-reactive protein (CRP), which worsen insulin resistance and prolong the inflammatory response, are also secreted in greater amounts when IL-6 is present [11].

Adiponectin is a hormone generated from adipocytes that has anti-inflammatory characteristics. It is essential for regulating insulin sensitivity. Adiponectin levels and insulin resistance are inversely correlated, according to clinical research, with patients with metabolic diseases and obese people having lower adiponectin levels [412]. Mechanistic investigations have demonstrated that adiponectin improves insulin sensitivity by triggering fatty acid oxidation and activating AMP-activated protein kinase (AMPK), which counteracts the inflammatory effects of IL-6 and TNF-α [4, 13].

Adipokines including resistin and leptin, in addition to cytokines, have been linked to the pathophysiology of insulin resistance. The hormone leptin, which is mostly released by adipocytes, controls hunger and energy balance. By influencing hypothalamic signalling pathways, it can also affect insulin sensitivity [14, 15]. Another adipokine called resistin has been demonstrated to increase insulin resistance in skeletal muscle and adipose tissue by obstructing insulin signalling and glucose uptake [16, 17].

One key mechanism connecting inflammation and insulin resistance is the activation of inflammatory signalling pathways, such as c-Jun N-terminal kinase (JNK) and nuclear factor kappa B (NF-κB). Experiments have shown that activation of NF-κB causes the expression of chemokines and pro-inflammatory cytokines, which exacerbates insulin resistance and accelerates the development of metabolic diseases [1, 2]. Similarly, by phosphorylating IRS proteins and upsetting insulin signalling pathways, JNK activation leads to insulin resistance [6, 7].

Overall, the review of the literature emphasises how important it is to target inflammatory pathways to prevent and treat metabolic illnesses, as well as how important inflammation is in the pathophysiology of insulin resistance.

Advertisement

3. Mechanisms of insulin resistance caused by inflammation

Insulin resistance is defined by a reduced ability of target tissues, including adipose, liver, and skeletal muscle, to respond to the effects of insulin. This leads to a decrease in the absorption and metabolism of glucose. As insulin resistance develops and progresses, chronic low-grade inflammation has been shown to play a significant role. Inflammatory mediators interfere with insulin signalling pathways on several levels [1, 2].

3.1 Insulin signalling pathway dysregulation

3.1.1 IRS protein serine phosphorylation

Serine kinases, such as IκB kinase (IKK) and c-Jun N-terminal kinase (JNK), are activated by inflammatory cytokines like TNF-α and IL-6. These kinases phosphorylate insulin receptor substrate (IRS) proteins on serine residues [2, 8]. By preventing the tyrosine phosphorylation of IRS proteins, this serine phosphorylation reduces insulin sensitivity and messes with downstream insulin signalling cascades [9, 10].

3.1.2 Blocking the PI3K/Akt pathway

By phosphorylating Akt (protein kinase B), phosphatidylinositol 3-kinase (PI3K) is essential for modulating the metabolic effects of insulin. Impaired glucose uptake and glycogen production can result from direct or indirect suppression of PI3K/Akt signalling by inflammatory signalling pathways such as NF-κB and JNK [18, 19].

3.1.3 Protein kinase C (PKC) isoform activation

PKC isoforms are activated by elevated amounts of the lipid second messenger diacylglycerol (DAG), which phosphorylates insulin receptor substrate-1 (IRS-1) and inhibits insulin-stimulated glucose uptake. These actions disrupt insulin signalling [20, 21].

3.2 Mitochondrial dysfunction and oxidative stress

3.2.1 Impairment in oxidative metabolism

Prolonged inflammation can cause disturbances in the synthesis and operation of mitochondria, resulting in impaired oxidative metabolism and mitochondrial dysfunction [22]. Insulin resistance is a result of decreased mitochondrial ATP synthesis and elevated reactive oxygen species (ROS) production, which impede insulin signalling and trigger cellular stress responses [23].

3.2.2 ROS-generated inflammatory pathways

Inflammatory signalling pathways, including NF-κB and JNK, are activated by ROS produced during mitochondrial failure, aggravating inflammation and insulin resistance [24].

3.3 Lipid accretion and lipotoxicity

3.3.1 Improved lipolysis and release of free fatty acids (FFA)

TNF-α and IL-6 are examples of inflammatory cytokines that promote lipolysis in adipose tissue, which increases the amount of free fatty acids released into the bloodstream [25]. Increased circulating FFA levels encourage the buildup of lipids in non-adipose tissues such as the liver and skeletal muscle, which exacerbates insulin resistance [26].

3.3.2 Lipid intermediate formation

Overindulgence in free fatty acids (FFAs) leads to the metabolism of lipid intermediates, including DAG and ceramides, which impede insulin signalling and enhance cellular stress responses [27]. For example, ceramides activate protein phosphatase 2A (PP2A), which inhibits insulin signalling and dephosphorylates Akt [28].

3.4 Dysregulation of adipokines and inflammation of adipose tissue

3.4.1 Macrophages in adipose tissue infiltration

Adipose tissue experiences dynamic remodelling in obesity and insulin resistance, which is typified by an increase in the infiltration of immune cells and pro-inflammatory macrophages [29]. TNF-α and IL-6 are among the inflammatory cytokines secreted by these activated immune cells, which lead to both local inflammation and systemic inflammation [30].

3.4.2 Dysregulated adipokine secretion

Insulin sensitivity and energy balance are regulated by adipokines, which are secreted by adipose tissue and include adiponectin, leptin, and resistin [31]. Dysregulated insulin signalling and metabolic dysfunction might result from adipocyte malfunction and inflammatory cytokines upsetting the balance of adipokine production [32].

Comprehending the complex processes via which inflammation leads to insulin resistance offers valuable perspectives on possible treatment targets and intervention tactics.

Advertisement

4. Clinical implications and associations

The development and progression of several metabolic disorders, such as type 2 diabetes mellitus (T2DM), cardiovascular disease (CVD), and non-alcoholic fatty liver disease (NAFLD), are closely linked to insulin resistance, which is characterised by impaired target tissue responsiveness to insulin action. Insulin resistance has significant clinical implications. The pathophysiology of insulin resistance is largely influenced by chronic low-grade inflammation, which also contributes to the clinical symptoms and related consequences of the condition.

4.1 Correlation to T2DM (type 2 diabetes mellitus)

One of the main characteristics of type 2 diabetes is insulin resistance, which is characterised by peripheral organs such as the liver, adipose tissue, and skeletal muscle having decreased sensitivity to insulin action [1, 2].

Pro-inflammatory cytokines including TNF-α and IL-6, which are high in chronic inflammation, lead to insulin resistance and β-cell dysfunction, which in turn accelerates the development and progression of type 2 diabetes [2, 8].

High-sensitivity C-reactive protein (hs-CRP) and the risk of acquiring type 2 diabetes (T2DM) have been linked to inflammation through epidemiological research, indicating the clinical importance of inflammation-induced insulin resistance in the pathophysiology of diabetes [9, 10].

4.2 Correlation with cardiovascular disease (CVD)

Atherosclerosis, hypertension, and coronary artery disease are all made more likely in people who are insulin-resistant [33]. Insulin resistance is a major factor in determining cardiovascular risk.

By encouraging endothelial dysfunction, oxidative stress, and vascular inflammation, chronic inflammation adds to the pathophysiology of CVD by hastening the development of atherosclerosis and raising the risk of cardiovascular events [34].

The clinical relevance of inflammation-induced insulin resistance in cardiovascular health is highlighted by the greatly increased risk of cardiovascular disease (CVD) in patients with metabolic syndrome and insulin resistance [35].

4.3 Correlation with non-alcoholic fatty liver disease (NAFLD)

The emergence of NAFLD, a group of liver diseases that includes cirrhosis, non-alcoholic steatohepatitis (NASH), and simple steatosis, is directly linked to insulin resistance [36].

The aetiology of non-alcoholic fatty liver disease (NAFLD) is aided by hepatic insulin resistance, which raises hepatic glucose synthesis, de novo lipogenesis, and triglyceride accumulation [37].

Pro-inflammatory cytokines stimulate hepatic inflammation, fibrosis, and hepatocyte death, all of which are important factors in the development of non-alcoholic fatty liver disease (NAFLD) [38].

4.4 Clinical relevance for risk evaluation and control

Evaluation of inflammatory markers, including hs-CRP, interleukins, and adipokines, might give important information on the underlying inflammatory state and cardiovascular risk profile of insulin-resistant people [39].

It has been demonstrated that dietary changes, physical activity, and weight loss are among the lifestyle therapies that target inflammation and can enhance insulin sensitivity and lower cardiovascular risk in people with metabolic disorders [40].

Additional advantages in lowering cardiovascular risk and enhancing clinical outcomes for people with insulin resistance may come from pharmacological therapies like anti-inflammatory drugs and statins [41].

In order to reduce the burden of metabolic disorders and cardiovascular consequences, it is imperative to understand the clinical implications and connections of inflammation-induced insulin resistance. This will help with risk assessment, early detection, and focused intervention options.

Advertisement

5. Diagnostic tools and biomarkers

Determining the risk of the disease, directing therapeutic approaches, and gauging the effectiveness of treatment all depend on the accurate diagnosis and monitoring of inflammation-induced insulin resistance. In clinical practice and research settings, a variety of diagnostic instruments and biomarkers have been developed to measure insulin sensitivity and identify inflammation.

5.1 Inflammatory markers

5.1.1 High-sensitivity C-reactive protein (hs-CRP)

hs-CRP is a commonly used biomarker of cardiovascular risk and systemic inflammation. Insulin resistance, metabolic syndrome, and an increased risk of cardiovascular events are all linked to elevated levels of hs-CRP [42, 43].

5.1.2 Interleukins (ILs)

The pro-inflammatory cytokines IL-6, IL-1β, and IL-18 are linked to the aetiology of metabolic diseases and insulin resistance. When circulating IL levels are measured in insulin-resistant individuals, it offers valuable information about their level of inflammation and the severity of their condition [9, 10].

5.1.3 Tumour necrosis factor-alpha (TNF-α)

TNF-α is an important modulator of insulin resistance brought on by inflammation, causing IRS protein serine phosphorylation and disrupting insulin signalling. Obesity, type 2 diabetes, and other metabolic diseases are associated with elevated levels of TNF-α [3, 8].

5.2 Adipokines

5.2.1 Adiponectin

This hormone, which is produced from adipocytes, has anti-inflammatory and insulin-sensitising characteristics. Obesity, insulin resistance, and cardiovascular risk are linked to decreased adiponectin levels [4, 12].

5.2.2 Leptin

Adipocytes secrete the hormone leptin, which controls hunger and energy balance. Obesity and insulin resistance are associated with elevated leptin levels, which are indicative of leptin resistance and dysfunctional adipose tissue [14, 15].

5.3 Lipid biomarkers

5.3.1 Free fatty acids (FFAs)

By encouraging fat accumulation in non-adipose tissues and disrupting insulin signalling, elevated circulating FFAs contribute to insulin resistance. In those with metabolic diseases, measuring FFAs offers information on insulin sensitivity and lipid metabolism [26].

5.3.2 Lipid intermediates

Insulin resistance and metabolic dysfunction have been linked to ceramides, diacylglycerols (DAGs), and other lipid intermediates. Type 2 diabetes, NAFLD, and obesity are all associated with elevated levels of lipid intermediates [44].

5.4 Imaging techniques

5.4.1 Magnetic resonance imaging (MRI) and positron emission tomography (PET)

These methods provide non-invasive evaluation of metabolic activity, insulin sensitivity, and tissue-specific glucose uptake. The pathogenesis of insulin resistance and metabolic diseases can be better understood by using these imaging modalities [45].

5.4.2 Dual-energy X-ray absorptiometry (DEXA) and computed tomography (CT)

These scans are used to measure visceral adiposity and ectopic fat deposition. Insulin resistance and an elevated risk of cardiovascular disease are linked to ectopic fat deposition and abdominal obesity [46].

5.5 Functional tests

5.5.1 Hyperinsulinemic euglycemic clamp and oral glucose tolerance test (OGTT)

Both are the gold standard tests for determining insulin sensitivity and glucose tolerance, respectively. In those with insulin resistance, these functional tests offer quantitative assessments of insulin action and metabolic function [47].

5.6 Molecular and genetic biomarkers

5.6.1 Single-nucleotide polymorphisms (SNPs)

The pathophysiology of insulin resistance and metabolic diseases has been linked to genetic variants linked to genes related to inflammation, such as TNF-α and IL-6. People who are more likely to develop insulin resistance due to inflammation may benefit from genetic testing [48].

5.6.2 Gene expression profiling

This technique enables a thorough examination of the gene expression patterns linked to insulin resistance and inflammation. The development of tailored therapeutics may be aided by molecular profiling approaches, which offer insights into the molecular pathways underlying disease pathogenesis [49].

Advertisement

6. Therapeutic strategies

Targeting both inflammatory pathways and metabolic dysfunction is necessary for the effective therapy of inflammation-induced insulin resistance. Numerous medication and lifestyle strategies have been created to enhance insulin sensitivity, lower inflammation, and lessen the chance of related metabolic problems.

6.1 Pharmacological interventions

6.1.1 Anti-inflammatory agents

The potential of nonsteroidal anti-inflammatory medicines (NSAIDs) like aspirin and ibuprofen, as well as selective inhibitors of inflammatory pathways like TNF-α antagonists and IL-1β blockers, to reduce systemic inflammation and improve insulin sensitivity has been studied [50, 51].

6.1.2 PPAR-γ agonists

The nuclear receptor known as peroxisome proliferator-activated receptor gamma (PPAR-γ), which is involved in the control of adipogenesis and insulin sensitivity, is agonistic to thiazolidinediones (TZDs), which include pioglitazone and rosiglitazone. By regulating adipokine production and reducing the expression of inflammatory genes, TZDs enhance insulin sensitivity and decrease inflammation [52, 53].

6.1.3 AMPK activators

A major modulator of insulin sensitivity and cellular energy metabolism is AMP-activated protein kinase (AMPK). Metformin and thiazolidinediones are examples of AMPK activators that improve insulin signalling and glucose absorption in target tissues by blocking mTOR signalling and activating AMPK [54, 55].

6.1.4 SGLT2 inhibitors

In people with type 2 diabetes mellitus, sodium-glucose cotransporter 2 (SGLT2) inhibitors, such as dapagliflozin and empagliflozin, decrease renal glucose reabsorption and increase urine glucose excretion, improving glycemic control and insulin sensitivity [56, 57].

6.1.5 GLP-1 receptor agonists

GLP-1 receptor agonists (GLP-1RAs) have surfaced as promising therapeutic options, boasting potent anti-inflammatory characteristics and a wide range of clinical applications [58].

6.2 Combination therapies

6.2.1 Dual PPAR-α/γ agonists

Preclinical and clinical investigations have demonstrated the potential of combination treatments that target various metabolic pathways to improve insulin sensitivity and reduce inflammation [59, 60].

6.2.2 Multimodal interventions

Behavioural counselling, exercise, and diet combined with pharmaceutical medications may provide synergistic effects in the management of metabolic diseases and inflammation-induced insulin resistance [61, 62].

6.3 Emerging therapeutic targets

6.3.1 Microbiome modulation

There may be a connection between insulin resistance, inflammation, and the gut microbiota composition, as per emerging research. Probiotics, prebiotics, and faecal microbiota transplantation are examples of strategies that target the gut microbiome and show promise as innovative therapeutic approaches for enhancing metabolic health [63, 64].

6.3.2 Epigenetic modification

Patterns of gene expression linked to inflammation and insulin resistance are regulated by epigenetic changes, including DNA methylation and histone acetylation. Histone deacetylase inhibitors and DNA methyltransferase inhibitors are examples of epigenetic modulators that may be used as therapeutic targets [65, 66].

Advertisement

7. Lifestyle interventions

7.1 Dietary changes

Embracing a nutritious diet high in fruits, vegetables, whole grains, and lean meats can help lower inflammation and enhance insulin sensitivity. Low-glycemic index and Mediterranean-style diets have been linked to better metabolic health and decreased systemic inflammation [67, 68].

7.2 Frequent exercise

Exercise is essential for lowering inflammation, encouraging weight loss, and enhancing insulin sensitivity. In those with insulin resistance, aerobic exercise and resistance training have both been demonstrated to improve glucose uptake, insulin action, and metabolic function [69, 70].

7.3 Handling weight

Insulin resistance and systemic inflammation are closely linked to obesity. Insulin sensitivity and metabolic parameters can be significantly improved by weight loss attained through calorie restriction, behavioural therapies, and bariatric surgery [71, 72].

Advertisement

8. Prospects for future research and directions

Progress in comprehending the intricate relationship among inflammation, insulin resistance, and metabolic dysfunction has created opportunities for novel approaches to treatment and investigation. It is highly promising that future research will clarify the molecular processes of inflammation-induced insulin resistance and find new targets for treatment, as these efforts will help to improve metabolic health and lessen the burden of related disorders.

8.1 Targeted therapies

8.1.1 Precision medicine methods

Individualised treatment plans based on genetic predispositions, inflammatory status, and metabolic profiles may enhance therapeutic efficacy and minimise side effects. Targeted therapeutics for inflammation-induced insulin resistance can be developed more easily when genomic, proteomic, and metabolomic data are integrated into clinical practice [73, 74].

8.1.2 Novel drug targets

The discovery and confirmation of new pharmacological targets related to the control of insulin signalling, metabolic homeostasis, and inflammatory pathways present promising prospects for the creation of next-generation treatments. The potential of emerging targets, like cytokine receptors, Toll-like receptors (TLRs), and inflammasomes, to reduce inflammation and enhance insulin sensitivity calls for more research [75, 76].

8.2 Metabolic crosstalk and immunometabolism

8.2.1 Immunometabolic pathways

Immunometabolism, the integration of immunological and metabolic signalling pathways, offers new perspectives on the aetiology of metabolic diseases including insulin resistance. Finding new treatment targets for inflammation-induced insulin resistance may be possible by clarifying the molecular mechanisms underlying immune cell metabolism, cytokine signalling, and metabolic reprogramming [77, 78].

8.2.2 Metabolic crosstalk

The control of systemic inflammation and insulin sensitivity is greatly influenced by communication between several metabolic organs and tissues, including adipose tissue, the liver, muscle, and the gut microbiota. Novel treatments that target metabolic crosstalk and dysregulation may be developed as a result of a better understanding of the reciprocal connections between immune cells and metabolic organs [78, 79].

8.3 Integrated omics methods

8.3.1 Multi-omics profiling

Integrating data from multiple fields, including proteomics, metabolomics, transcriptomics, and genomes, holds potential for a thorough understanding of the molecular mechanisms driving inflammation-induced insulin resistance. Network analysis and pathway modelling are two examples of systems biology techniques that can offer comprehensive insights into the pathophysiology of disease and suggest possible targets for treatment [80, 81].

8.3.2 Artificial intelligence and machine learning

Identification of predictive biomarkers, disease subtypes, and therapeutic response profiles are made possible by the use of artificial intelligence (AI) techniques and machine learning algorithms to large-scale omics datasets. The development of innovative therapeutic approaches and individualised treatment plans for inflammation-induced insulin resistance may proceed more quickly if AI-driven analytics is fully utilised [82, 83].

8.4 Behaviour interventions and lifestyle changes

8.4.1 Digital health technologies

By incorporating wearables, smartphone apps, and remote monitoring platforms with lifestyle treatments and behavioural change programmes, digital health technologies can improve patient outcomes, adherence, and engagement. Digital health platforms that enable social support networks, real-time feedback, and personalised coaching may enhance metabolic results and long-term adherence to healthy behaviours [84, 85].

8.4.2 Behavioural economics

The development of successful interventions targeted at encouraging healthy lifestyle choices and reducing the risk of inflammation-induced insulin resistance can be aided by behavioural economics insights such as choice architecture and nudge theory. Behavioural interventions that apply behavioural economics concepts—like default options, framing effects, and incentive structures—may help modify behaviour in a sustainable way and enhance metabolic health outcomes [86, 87].

Advertisement

9. Conclusion

Insulin resistance brought on by inflammation is a complicated metabolic disease with wide-ranging effects on human health. Despite tremendous advancements in our comprehension of the underlying pathophysiological pathways, effective therapeutic approaches for insulin resistance caused by inflammation are still unattainable.

In order to fully understand the complex interactions between inflammation and insulin signalling pathways, a multidisciplinary approach integrating knowledge from immunology, metabolism, genetics, and systems biology is essential going ahead. Personalised treatment plans, lifestyle changes, and targeted pharmaceutical interventions have the potential to enhance metabolic health and lessen the impact of comorbidities linked to inflammation-induced insulin resistance.

Furthermore, improving our knowledge of inflammation-induced insulin resistance and integrating scientific findings into clinical practice depend on ongoing research efforts focused on identifying novel therapeutic targets, clarifying the molecular mechanisms underlying disease pathogenesis, and creating cutting-edge diagnostic tools and biomarkers.

We can overcome the obstacles presented by inflammation-induced insulin resistance and open the door to the creation of efficient preventive and treatment plans that enhance the lives of those impacted by this common and crippling metabolic illness by adopting a collaborative and interdisciplinary research paradigm.

References

  1. 1. Hotamisligil GS. Inflammation and metabolic disorders. Nature. 2006;444(7121):860-867
  2. 2. Shoelson SE, Lee J, Goldfine AB. Inflammation and insulin resistance. Journal of Clinical Investigation. 2006;116(7):1793-1801
  3. 3. Pickup JC, Crook MA, Ismail IH. TNF-α, interleukin-1, and insulin resistance in obesity. Biochemical Society Transactions. 2000;28(2):522-526
  4. 4. Yamauchi T, Kamon J, Waki H, Terauchi Y, Kubota N, Hara K, et al. The fat-derived hormone adiponectin reverses insulin resistance associated with both lipoatrophy and obesity. Nature Medicine. 2001;7(8):941-946
  5. 5. Hotamisligil GS. Endoplasmic reticulum stress and the inflammatory basis of metabolic disease. Cell. 2010;140(6):900-917
  6. 6. Hirosumi J, Tuncman G, Chang L, Görgün CZ, Uysal KT, Maeda K, et al. A central role for JNK in obesity and insulin resistance. Nature. 2002;420(6913):333-336
  7. 7. Gual P, Le Marchand-Brustel Y, Tanti JF. Positive and negative regulation of insulin signaling through IRS-1 phosphorylation. Biochimie. 2007;89(6-7):1-9
  8. 8. Hotamisligil GS. Mechanisms of TNF-alpha-induced insulin resistance. Experimental and Clinical Endocrinology & Diabetes. 1999;107(2):119-125
  9. 9. Pedersen BK, Febbraio MA. Muscle as an endocrine organ: Focus on muscle-derived interleukin-6. Physiological Reviews. 2008;88(4):1379-1406
  10. 10. Carey AL, Steinberg GR, Macaulay SL, Thomas WG, Holmes AG, Ramm G, et al. Interleukin-6 increases insulin-stimulated glucose disposal in humans and glucose uptake and fatty acid oxidation in vitro via AMP-activated protein kinase. Diabetes. 2006;55(10):2688-2697
  11. 11. Senn JJ, Klover PJ, Nowak IA, Mooney RA. Interleukin-6 induces cellular insulin resistance in hepatocytes. Diabetes. 2002;51(12):3391-3399
  12. 12. Arita Y, Kihara S, Ouchi N, Takahashi M, Maeda K, Miyagawa J, et al. Paradoxical decrease of an adipose-specific protein, adiponectin, in obesity. Biochemical and Biophysical Research Communications. 1999;257(1):79-83
  13. 13. Berg AH, Combs TP, Du X, Brownlee M, Scherer PE. The adipocyte-secreted protein Acrp30 enhances hepatic insulin action. Nature Medicine. 2001;7(8):947-953
  14. 14. Maffei M, Halaas J, Ravussin E, Pratley RE, Lee GH, Zhang Y, et al. Leptin levels in human and rodent: Measurement of plasma leptin and Ob RNA in obese and weight-reduced subjects. Nature Medicine. 1995;1(11):1155-1161
  15. 15. Mantzoros CS, Magkos F, Brinkoetter M, Sienkiewicz E, Dardeno TA, Kim SY, et al. Leptin in human physiology and pathophysiology. American Journal of Physiology. Endocrinology and Metabolism. 2011;301(4):E567-E584
  16. 16. Steppan CM, Bailey ST, Bhat S, Brown EJ, Banerjee RR, Wright CM, et al. The hormone resistin links obesity to diabetes. Nature. 2001;409(6818):307-312
  17. 17. Satoh H, Nguyen MT, Miles PD, Imamura T, Usui I, Olefsky JM, et al. Adenovirus-mediated chronic “hyper-resistinemia” leads to in vivo insulin resistance in normal rats. Journal of Clinical Investigation. 2004;114(2):224-231
  18. 18. Ramasubbu K, Devi RV. Impairment of insulin signaling pathway PI3K/Akt/mTOR and insulin resistance induced AGEs on diabetes mellitus and neurodegenerative diseases: A perspective review. Molecular and Cellular Biochemistry. 2023;478(6):1307-1324
  19. 19. ElMahdy MK, Zaki MO, Al-Karmalawy AA, Abdo W, Alnasser SM, Antar SA. Glimepiride ameliorates renal toxicity induced by cadmium in mice: Modulation of Jun N terminal kinase (JNK)/nuclear factor kappa B (NF-κB) and phosphatidylinositol 3-kinases (PI3K)/protein kinase (AKT) pathways. Life Sciences. 2022;311:121184
  20. 20. Kolczynska K, Loza-Valdes A, Hawro I, Sumara G. Diacylglycerol-evoked activation of PKC and PKD isoforms in regulation of glucose and lipid metabolism: A review. Lipids in Health and Disease. 2020;19:1-5
  21. 21. Turban S, Hajduch E. Protein kinase C isoforms: Mediators of reactive lipid metabolites in the development of insulin resistance. FEBS Letters. 2011;585(2):269-274
  22. 22. Prasun P. Mitochondrial dysfunction in metabolic syndrome. Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease. 2020;1866(10):165838
  23. 23. Ayer A, Fazakerley DJ, James DE, Stocker R. The role of mitochondrial reactive oxygen species in insulin resistance. Free Radical Biology and Medicine. 2022;179:339-362
  24. 24. Choudhury S, Ghosh S, Gupta P, Mukherjee S, Chattopadhyay S. Inflammation-induced ROS generation causes pancreatic cell death through modulation of Nrf2/NF-κB and SAPK/JNK pathway. Free Radical Research. 2015;49(11):1371-1383
  25. 25. Kojta I, Chacińska M, Błachnio-Zabielska A. Obesity, bioactive lipids, and adipose tissue inflammation in insulin resistance. Nutrients. 2020;12(5):1305
  26. 26. Ahmed B, Sultana R, Greene MW. Adipose tissue and insulin resistance in obese. Biomedicine and Pharmacotherapy. 2021;137:111315
  27. 27. Sinha S, Haque M. Obesity, diabetes mellitus, and vascular impediment as consequences of excess processed food consumption. Cureus. 2022;14(9):1-24
  28. 28. Bharath LP, Ruan T, Li Y, Ravindran A, Wan X, Nhan JK, et al. Ceramide-initiated protein phosphatase 2A activation contributes to arterial dysfunction in vivo. Diabetes. 2015;64(11):3914-3926
  29. 29. Vieira-Potter VJ. Inflammation and macrophage modulation in adipose tissues. Cellular Microbiology. 2014;16(10):1484-1492
  30. 30. Wang T, He C. TNF-α and IL-6: The link between immune and bone system. Current Drug Targets. 2020;21(3):213-227
  31. 31. Ahima RS, Lazar MA. Adipokines and the peripheral and neural control of energy balance. Molecular Endocrinology. 2008;22(5):1023-1031
  32. 32. Lafontan M. Adipose tissue and adipocyte dysregulation. Diabetes & Metabolism. 2014;40(1):16-28
  33. 33. DeFronzo RA, Ferrannini E. Insulin resistance: A multifaceted syndrome responsible for NIDDM, obesity, hypertension, dyslipidemia, and atherosclerotic cardiovascular disease. Diabetes Care. 1991;14(3):173-194
  34. 34. Mason JC, Libby P. Cardiovascular disease in patients with chronic inflammation: Mechanisms underlying premature cardiovascular events in rheumatologic conditions. European Heart Journal. 2015;36(8):482-489
  35. 35. Romeo GR, Lee J, Shoelson SE. Metabolic syndrome, insulin resistance, and roles of inflammation–mechanisms and therapeutic targets. Arteriosclerosis, Thrombosis, and Vascular Biology. 2012;32(8):1771-1776
  36. 36. Liu Q , Bengmark S, Qu S. The role of hepatic fat accumulation in pathogenesis of non-alcoholic fatty liver disease (NAFLD). Lipids in Health and Disease. 2010;9:1-9
  37. 37. Mu W, Cheng XF, Liu Y, Lv QZ, Liu GL, Zhang JG, et al. Potential nexus of non-alcoholic fatty liver disease and type 2 diabetes mellitus: Insulin resistance between hepatic and peripheral tissues. Frontiers in Pharmacology. 2019;9:1566
  38. 38. Duan Y, Pan X, Luo J, Xiao X, Li J, Bestman PL, et al. Association of inflammatory cytokines with non-alcoholic fatty liver disease. Frontiers in Immunology. 2022;13:880298
  39. 39. Sutherland JP, McKinley B, Eckel RH. The metabolic syndrome and inflammation. Metabolic Syndrome and Related Disorders. 2004;2(2):82-104
  40. 40. Andersen CJ, Fernandez ML. Dietary strategies to reduce metabolic syndrome. Reviews in Endocrine and Metabolic Disorders. 2013;14:241-254
  41. 41. Ruscica M, Corsini A, Ferri N, Banach M, Sirtori CR. Clinical approach to the inflammatory etiology of cardiovascular diseases. Pharmacological Research. 2020;159:104916
  42. 42. Ridker PM. Clinical application of C-reactive protein for cardiovascular disease detection and prevention. Circulation. 2003;107(3):363-369
  43. 43. Pradhan AD, Manson JE, Rifai N, Buring JE, Ridker PM. C-reactive protein, interleukin 6, and risk of developing type 2 diabetes mellitus. JAMA. 2001;286(3):327-334
  44. 44. Kitessa SM, Abeywardena MY. Lipid-induced insulin resistance in skeletal muscle: The chase for the culprit goes from total intramuscular fat to lipid intermediates, and finally to species of lipid intermediates. Nutrients. 2016;8(8):466
  45. 45. Johansson E, Lubberink M, Heurling K, Eriksson JW, Skrtic S, Ahlström H, et al. Whole-body imaging of tissue-specific insulin sensitivity and body composition by using an integrated PET/MR system: A feasibility study. Radiology. 2018;286(1):271-278
  46. 46. Kaul S, Rothney MP, Peters DM, Wacker WK, Davis CE, Shapiro MD, et al. Dual-energy X-ray absorptiometry for quantification of visceral fat. Obesity. Jun 2012;20(6):1313-1318
  47. 47. Pratt-Phillips SE, Geor RJ, McCutcheon LJ. Comparison among the euglycemic-hyperinsulinemic clamp, insulin-modified frequently sampled intravenous glucose tolerance test, and oral glucose tolerance test for assessment of insulin sensitivity in healthy Standardbreds. American Journal of Veterinary Research. 2015;76(1):84-91
  48. 48. Malodobra M, Pilecka A, Gworys B, Adamiec R. Single nucleotide polymorphisms within functional regions of genes implicated in insulin action and association with the insulin resistant phenotype. Molecular and Cellular Biochemistry. 2011;349:187-193
  49. 49. Konstantopoulos N, Foletta VC, Segal DH, Shields KA, Sanigorski A, Windmill K, et al. A gene expression signature for insulin resistance. Physiological Genomics. 2011;43(3):110-120
  50. 50. Ridker PM, Lüscher TF. Anti-inflammatory therapies for cardiovascular disease. European Heart Journal. 2014;35(27):1782-1791
  51. 51. Gómez-García A, Hernández-Campos ME. An update on the use of TNF-α inhibitors in the treatment of psoriasis. Drug Design, Development and Therapy. 2016;10:1581-1591
  52. 52. Yki-Järvinen H. Thiazolidinediones. New England Journal of Medicine. 2004;351(11):1106-1118
  53. 53. Zinman B, Wanner C, Lachin JM, Fitchett D, Bluhmki E, Hantel S, et al. Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes. New England Journal of Medicine. 2015;373(22):2117-2128
  54. 54. Hardie DG. AMPK—Sensing energy while talking to other signaling pathways. Cell Metabolism. 2014;20(6):939-952
  55. 55. Viollet B, Guigas B, Sanz Garcia N, Leclerc J, Foretz M, Andreelli F. Cellular and molecular mechanisms of metformin: An overview. Clinical Science. 2012;122(6):253-270
  56. 56. DeFronzo RA, Norton L, Abdul-Ghani M. Renal, metabolic and cardiovascular considerations of SGLT2 inhibition. Nature Reviews Nephrology. 2017;13(1):11-26
  57. 57. Abdul-Ghani M, Del Prato S, Chilton R, DeFronzo RA. SGLT2 inhibitors and cardiovascular risk: Lessons learned from the EMPA-REG OUTCOME Study. Diabetes Care. 2016;39(5):717-725
  58. 58. Alharbi SH. Anti-inflammatory role of glucagon-like peptide 1 receptor agonists and its clinical implications. Therapeutic Advances in Endocrinology and Metabolism. 2024;15:20420188231222367
  59. 59. Marx N, Duez H. Peroxisome proliferator-activated receptors and atherogenesis: Regulators of gene expression in vascular cells. Circulation Research. 2007;100(6):803-817
  60. 60. Fougerat A, Gayral S, Gourdy P, Schambourg A. Dual PPARα/γ agonists: Promising new targets in the treatment of type 2 diabetes and associated complications. Current Opinion in Pharmacology. 2019;49:106-115
  61. 61. Look AHEAD Research Group. Reduction in weight and cardiovascular disease risk factors in individuals with type 2 diabetes: One-year results of the Look AHEAD trial. Diabetes Care. 2007;30(6):1374-1383
  62. 62. Knowler WC, Barrett-Connor E, Fowler SE, Hamman RF, Lachin JM, Walker EA, et al. Reduction in the incidence of type 2 diabetes with lifestyle intervention or metformin. New England Journal of Medicine. 2002;346(6):393-403
  63. 63. Cani PD, Amar J, Iglesias MA, Poggi M, Knauf C, Bastelica D, et al. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes. 2007;56(7):1761-1772
  64. 64. Vrieze A, Van Nood E, Holleman F, Salojärvi J, Kootte RS, Bartelsman JFWM, et al. Transfer of intestinal microbiota from lean donors increases insulin sensitivity in individuals with metabolic syndrome. Gastroenterology. 2012;143(4):913-916.e7
  65. 65. Ling C, Rönn T. Epigenetics in human obesity and type 2 diabetes. Cell Metabolism. 2019;29(5):1028-1044
  66. 66. Barres R, Zierath JR. DNA methylation in metabolic disorders. American Journal of Clinical Nutrition. 2011;93(4):897S-900S
  67. 67. Esposito K, Marfella R, Ciotola M, Di Palo C, Giugliano F, Giugliano G, et al. Effect of a Mediterranean-style diet on endothelial dysfunction and markers of vascular inflammation in the metabolic syndrome: A randomized trial. JAMA. 2004;292(12):1440-1446
  68. 68. Jenkins DJA, Kendall CWC, Augustin LSA, Franceschi S, Hamidi M, Marchie A, et al. Glycemic index: Overview of implications in health and disease. American Journal of Clinical Nutrition. 2002;76(1):266S-273S
  69. 69. Hawley JA, Lessard SJ. Exercise training-induced improvements in insulin action. Acta Physiologica. 2008;192(1):127-135
  70. 70. Church TS, Blair SN, Cocreham S, Johannsen N, Johnson W, Kramer K, et al. Effects of aerobic and resistance training on hemoglobin A1c levels in patients with type 2 diabetes. JAMA. 2010;304(20):2253-2262
  71. 71. Lean MEJ, Malkova D. Altered gut and adipose tissue hormones in overweight and obese individuals: Cause or consequence? International Journal of Obesity. 2016;40(4):622-632
  72. 72. Pi-Sunyer FX. The epidemiology of central fat distribution in relation to disease. Nutrition Reviews. 2009;67(suppl_2):S81-S84
  73. 73. Collins FS, Varmus H. A new initiative on precision medicine. New England Journal of Medicine. 2015;372(9):793-795
  74. 74. Khera AV, Chaffin M, Aragam KG, Haas ME, Roselli C, Choi SH, et al. Genome-wide polygenic scores for common diseases identify individuals with risk equivalent to monogenic mutations. Nature Genetics. 2018;50(9):1219-1224
  75. 75. Swanson KV, Deng M, Ting JPY. The NLRP3 inflammasome: Molecular activation and regulation to therapeutics. Nature Reviews Immunology. 2019;19(8):477-489
  76. 76. O'Neill LAJ, Netea MG. BCG-induced trained immunity: Can it offer protection against COVID-19? Nature Reviews Immunology. 2020;20(6):335-337
  77. 77. Odegaard JI, Chawla A. Connecting type 1 and type 2 diabetes through innate immunity. Cold Spring Harbor Perspectives in Medicine. 2013;3(3):a007724
  78. 78. Hotamisligil GS. Inflammation, metaflammation and immunometabolic disorders. Nature. 2017;542(7640):177-185
  79. 79. Tilg H, Moschen AR. Microbiota and diabetes: An evolving relationship. Gut. 2014;63(9):1513-1521
  80. 80. Hasin Y, Seldin M, Lusis A. Multi-omics approaches to disease. Genome Biology. 2017;18(1):83
  81. 81. Mardinoglu A, Nielsen J. Systems medicine and metabolic modelling. Journal of Internal Medicine. 2015;278(6):587-596
  82. 82. Obermeyer Z, Emanuel EJ. Predicting the future—Big data, machine learning, and clinical medicine. New England Journal of Medicine. 2016;375(13):1216-1219
  83. 83. Topol EJ. High-performance medicine: The convergence of human and artificial intelligence. Nature Medicine. 2019;25(1):44-56
  84. 84. Huckvale K, Morrison C, Ouyang J, Ghaghda A, Car J. The evolution of mobile apps for asthma: An updated systematic assessment of content and tools. BMC Medicine. 2019;17(1):58
  85. 85. Patel MS, Volpp KG, Rosin R, Bellamy SL, Small DS, Heuer J, et al. A randomized trial of social comparison feedback and financial incentives to increase physical activity. American Journal of Health Promotion. 2018;32(4):935-942
  86. 86. Thaler RH, Sunstein CR. Nudge: Improving Decisions about Health, Wealth, and Happiness. Penguin; 24 Feb 2009
  87. 87. Loewenstein G, Brennan T, Volpp KG. Asymmetric paternalism to improve health behaviors. JAMA. 2007;298(20):2415-2417

Written By

Anchala Kumari

Submitted: 22 April 2024 Reviewed: 29 April 2024 Published: 24 June 2024