Skip to main content
Bioengineering & Translational Medicine logoLink to Bioengineering & Translational Medicine
. 2019 Dec 13;5(2):e10150. doi: 10.1002/btm2.10150

Remodeling adipose tissue inflammasome for type 2 diabetes mellitus treatment: Current perspective and translational strategies

Amrita Banerjee 1,, Jagdish Singh 1
PMCID: PMC7237149  PMID: 32440558

Abstract

Obesity‐associated type 2 diabetes mellitus (T2DM) is characterized by low‐grade chronic systemic inflammation that arises primarily from the white adipose tissue. The interplay between various adipose tissue‐derived chemokines drives insulin resistance in T2DM and has therefore become a subject of rigorous investigation. The adipocytokines strongly associated with glucose homeostasis include tumor necrosis factor‐α, various interleukins, monocyte chemoattractant protein‐1, adiponectin, and leptin, among others. Remodeling the adipose tissue inflammasome in obesity‐associated T2DM is likely to treat the underlying cause of the disease and bring significant therapeutic benefit. Various strategies have been adopted or are being investigated to modulate the serum/tissue levels of pro‐ and anti‐inflammatory adipocytokines to improve glucose homeostasis in T2DM. These include use of small molecule agonists/inhibitors, mimetics, antibodies, gene therapy, and other novel formulations. Here, we discuss adipocytokines that are strongly associated with insulin activity and therapies that are under investigation for modulation of their levels in the treatment of T2DM.

Keywords: adipokines, chronic inflammation, cytokines, formulations, obesity, type 2 diabetes mellitus

1. INTRODUCTION

Type 2 diabetes mellitus (T2DM) is currently considered a global epidemic and projected to rise exponentially by more than 50% worldwide in the next 20 years.1, 2 Particularly, few central African and middle‐eastern countries will see a projected more than 100% rise in diabetes cases from 2015 to 2040.1 Life style changes in the modern era, food habits, and environmental pollutants are primarily responsible for the rapid global rise in T2DM cases. Additionally, in developing or low‐income countries where infectious diseases present a more pressing problem, there can be lack of proper resources and/or knowledge among primary caregivers for management of this slow but silent killer. According to a World Health Organization report, there were an estimated 1.6 million fatalities in 2016 due to the disease, making diabetes the seventh leading cause of deaths and the major cause of blindness, renal failure, cardiac arrests, stroke, and amputations of the lower limbs.3 Given these alarming facts, it is paramount to develop efficient therapies that not only halt the progression but also treat the underlying cause of the disease.

T2DM occurs primarily due to insulin resistance in adipose tissue, muscles, and liver leading to hyperglycemia. Obesity, age, family history of diabetes, hypertension, hypercholesterolemia, and presence of cardiovascular diseases are considered some of the biggest risk factors for T2DM.4 Therefore, in addition to diabetes pharmacotherapy, care often requires management of the aforementioned co‐conspirator diseases, consumption of healthy foods, and regular exercise to alleviate diabetic conditions. Among the various antidiabetic medications, the most commonly prescribed class of medications include biguanides (metformin), glucagon‐like peptide‐1 agonists/receptor agonists, sodium glucose transport protein‐2 inhibitors, dipeptidyl peptidase 4 inhibitors, thiazolidinediones (TZDs), sulfonylureas, and insulin.5

In obesity, adipocyte hypertrophy can cause secretion of proinflammatory adipokines such as monocyte chemoattractant protein 1 (MCP‐1), which recruits circulating monocytes from the blood circulation to the adipose tissue, where they undergo differentiation into macrophages that are predominantly polarized to proinflammatory M1 phenotype.6, 7 Over time, the white adipose tissue of obese individuals can comprise up to 40–50% macrophages that are M1 polarized, which is in clear contrast to lean individuals who have only about 5% macrophages in their adipose tissue and they are primarily polarized to noninflammatory M2 phenotype.8 The M1 macrophages secrete proinflammatory cytokines into the systemic circulation such as tumor necrosis factor‐α (TNF‐α), interleukin 1β (IL‐1β), IL‐6, IL‐18, and C‐X‐C motif chemokine 5 (CXCL5), among others.9 Additionally, dysregulation of other immune cells in the adipose tissue such as T cells, B cells, and dendritic cells is observed in obesity‐associated T2DM. An increased accumulation of conventional dendritic cells, CD8+ cytotoxic T lymphocytes, and proinflammatory B cells in the adipose tissue exacerbates inflammation and insulin resistance.10 Conversely, population of M2 macrophages, regulatory B or T cells, type 2 innate lymphoid cells, eosinophils, perforin+ dendritic cells, and invariant natural killer T cells that are responsible for regulation of immunity and metabolic homeostasis in the adipose tissue are substantially reduced in obesity11 (Figure 1).

Figure 1.

Figure 1

Adipose tissue modulation in obesity. Adipose tissue hypertrophy in obesity results in significant immunomodulation that skews adipose tissue microenvironment toward proinflammatory phenotype. Several immune cells normally present in adipose tissue of lean individuals are downregulated in obesity (not shown in the figure). For more details about immune cell changes in adipose tissue with obesity and type 2 diabetes, refer to publication by Lu et al10

Likewise, the adipocytes in obese individuals secrete several adipokines implicated in insulin resistance such as resistin, retinol‐binding protein 4 (RBP‐4), MCP‐1, progranulin, chemerin, and angiopoietin‐like protein 2 (ANGPTL2).9, 12 Similar pathophysiology is observed in other key insulin target organs such as muscles and liver. In muscles, increased accumulation of extramyocellular adipocytes lead to macrophage recruitment and activation, while in liver, the resident macrophage Kupffer cells are activated, which increase inflammatory cytokine production.13 Chronic presence of these inflammatory adipocytokines is widely implicated to cause insulin resistance in insulin‐sensitive organs, resulting in T2DM development.14, 15, 16, 17 A concomitant decrease in insulin‐sensitizing adipokines (adiponectin, leptin, apelin, omentin, and secreted frizzled protein‐5) and anti‐inflammatory cytokines (IL‐10 and IL‐22) is also observed, which exacerbates inflammatory conditions and subsequent insulin resistance.12, 18 Therefore, remodeling of adipose tissue inflammasome through modulation of adipocytokine production is a rational step toward efficient treatment of obesity‐associated T2DM (Figure 2).

Figure 2.

Figure 2

Adipose tissue inflammation in obesity‐associated type 2 diabetes mellitus and its treatment perspective. For simplicity only M1 macrophages are shown in the figure with the adipocytes

Many antidiabetic medications‐based therapies are symptomatic and do not address the underlying cause of the diseases such as chronic inflammation in the white adipose tissue of obese/overweight individuals. Metformin was recently shown to decrease adipose tissue inflammation while TZDs have been reported to decrease adipose tissue macrophage content and improve insulin sensitivity.19, 20, 21 TZDs are however associated with several significant risks including heart failure, bladder cancer, diabetic macular edema, among others.22, 23, 24 Anti‐inflammatory therapies that target adipose tissue inflammation can significantly improve insulin sensitivity and β‐cell function, and therefore have great potential in the treatment of T2DM.25, 26 This review discusses some of the major adipocytokines involved in T2DM development and few notable formulations/drugs that are being developed to halt the vicious cycle of chronic inflammation in the adipose tissue and development of systemic insulin resistance.

2. ADIPOKINES AND THERAPIES THAT MODULATE THEIR ACTIVITY

Adipocyte hypertrophy can cause secretion of proinflammatory adipokines and initiation of adipose tissue inflammation. The various mechanisms involved in activation of inflammation in adipocytes leading to insulin resistance are presented in Figure 3. These molecules have therefore garnered immense interest among researchers who have attempted to either increase serum levels of insulin‐sensitizing adipokines or decrease concentration of insulin resistance imparting adipokines. Major adipokines involved in glucose homeostasis and notable drugs/formulations under investigation for modulation of their activities are discussed in this section.

Figure 3.

Figure 3

Mechanism of inflammation activation in an adipocyte leading to insulin resistance. Adipocyte stress due to hypertrophy, adipose tissue hypoxia, autophagy, apoptosis, ROS (reactive oxygen species) production, ER (endoplasmic reticulum) stress, lipolysis leading to production, and increased flux of FFA (free fatty acids) result in secretion of proinflammatory adipokines and downregulation of anti‐inflammatory adipokines.27 The proinflammatory adipokines recruit immune cells in the adipose tissue that differentiate to proinflammatory phenotype and release proinflammatory cytokines that further promulgate inflammatory conditions. Chronic inflammatory conditions and high serum FFA concentration lead to impairment in insulin signaling and systemic insulin resistance. A decrease in GLUT4 (glucose transporter 4) expression and translocation to the cell membrane causing reduction in glucose uptake and glucose toxicity also occurs. For more details on mechanisms of adipose tissue inflammation and insulin resistance, refer to publication by Bluher27

2.1. Adipokines mediating insulin sensitization

Among various adipokines, adiponectin secreted exclusively by adipocytes plays a key role in glucose and lipid metabolism in insulin‐sensitive tissues and was shown to decrease blood glucose levels in many preclinical studies performed on diabetic animals. It improves insulin sensitivity and free fatty acid oxidation while decreases hepatic glucose output and vascular inflammation.28 In a recent study, higher adiponectin level was associated with decreased T2DM risk in Chinese and other populations.29 Proinflammatory cytokines, especially TNF‐α, are known to reduce adiponectin levels.30 A low serum adiponectin level is strongly associated with obesity and development of insulin resistance with a concurrent downregulation of adiponectin receptor expression.31, 32 Conversely, adiponectin administration leads to improvement in insulin sensitivity, decrease in plasma glucose levels, and reduced MCP‐1 production.33, 34, 35 Adiponectin formulations mainly include administration of recombinant adiponectin produced and isolated from mammalian or Escherichia coli cultures after transfection of plasmid encoding adiponectin. A single injection of recombinant adiponectin was found to transiently decrease hyperglycemia in ob/ob, NOD, and streptozotocin‐induced diabetic mice.36 In a different study, administration of globular head domain of adiponectin decreased free fatty acids in the serum and was postulated to be due to increased oxidation of fats by muscle.37 Similarly, intraperitoneal administration of recombinant adiponectin decreased visceral adiposity and weight gain in agouti yellow obese mice.38 However, Novo Nordisk obtained contradictory preclinical evidence on recombinant adiponectin therapy in lowering blood glucose levels in T2DM.39 The research group showed that intraperitoneal injection of human and murine adiponectin produced from CHOK1SV and HEK293 cells respectively were ineffective in reducing plasma glucose levels in db/db diabetic mice or sand rats. They posit that adiponectin may require processing by adipocytes to demonstrate bioactivity. In light of this contradictory evidence, researchers have successfully developed an orally bioavailable adiponectin mimetic small molecule drug, AdipoRon, that strongly binds to adiponectin receptors (AdipoR1 and AdipoR2) and ameliorates diabetic conditions while extending lifespan in diabetic db/db mice placed on high‐fat diet.40 Gene therapy for adiponectin supplementation has also been extensively studied for diabetes treatment. For example, when adiponectin cDNA subcloned in adenoviral vector containing a muscle specific promoter was delivered through electroporation and intramuscular injection, it resulted in significant improvement in insulin sensitivity in mice receiving high‐fat–high‐sucrose diet.41 Similarly, nonviral‐based adiponectin gene delivery using mini‐circle DNA in polyethyleneimine carrier resulted in optimum serum adiponectin levels and normalization of parameters pertaining to insulin resistance in obese C57BL/6J mice.42 Intravenous administration of adiponectin pDNA in streptozotocin‐induced diabetic mice resulted in 10–15‐fold enhancement in adiponectin serum levels as well as hepatic glucose uptake that led to reduction in serum glucose and triglyceride levels.43

Leptin is another important adipokine that acts through its receptor in the brain and is involved in glucose homeostasis, regulation of energy expenditure, and appetite.44 Serum levels of leptin are very low in obesity with leptin deficiency, lipodystrophy‐induced T2DM, HIV‐lipodystrophy‐induced T2DM, and in type 1 diabetes mellitus (T1DM) with lipodystrophy.45 In patients presenting these cases, leptin intervention can markedly improve glucose and lipid homeostasis.45 However, leptin levels are high in generalized obesity or obesity‐associated T2DM and such patients show poor efficacy to exogenous leptin administration.45 This may be attributed to leptin resistance in common obesity, which is mostly caused by disruption in leptin signaling, impairment in regulation of feeding/reward behavior, or reduced leptin transport across the blood–brain barrier (BBB).46 A synthetic human leptin analog, Metreleptin (Myalept™), has been FDA approved for treatment of lipodystrophy and complications arising due to leptin deficiency. To improve leptin delivery to the brain, intrathecal leptin administration to bypass the BBB, polyethylene glycol (PEG)‐grafted leptin to increase the circulation half‐life of the protein, oil‐based subcutaneous injection of leptin for slower release, and prolonged activity or leptin peptide agonists have been investigated, which demonstrated varying success.47 Adjuvant therapy using epinephrine and norepinephrine was found to modulate leptin receptor activity and significantly improve leptin transport.47 Gene delivery through intracranial injection to hypothalamus using adenoviral vector encoding human leptin to ob/ob mice led to body weight reduction in 4 weeks.48 Similarly, intraventricular administration of adenovirus encoding rat leptin DNA led to 17% lower weight and 80% lesser white adipose tissue in female rats compared to sham control.49 Intranasal delivery of pluronic P85 conjugated to N‐terminal portion of leptin led to higher accumulation in hippocampus and hypothalamus than native leptin administered intranasally.50 This resulted in activation of leptin receptors in hypothalamus at a lower dose than unmodified leptin. This technology is now being developed by NeuroNanoPharma Inc. (Raleigh, NC) for treatment of obesity.

Apelin, an adipokine that is also expressed in the brain, is postulated to possess anti‐inflammatory properties through inhibition of reactive oxygen species (ROS).51 Its expression in adipocytes increases with increase in insulin concentration such as in obesity–hyperinsulinemia animal models and upon insulin treatment in cultured adipocytes in vitro.51 Increase in hypoxia in adipose tissue that promulgates inflammatory conditions also induces apelin production.51 Existence of apelinemia (high apelin concentration) has been found in morbidly obese and patients without morbid obesity but exhibiting impaired glucose tolerance or T2DM.52 However, there are contradictions to the findings as apelin levels were found lower in obese T2DM patients not treated with antidiabetic drugs such as metformin and rosiglitazone.52 In the same vein, no correlation has been found between apelin levels and body weight, adiposity and insulin resistance in obese and lean children.53 An insulin‐sensitizing/insulin‐mimetic effect of apelin has been postulated after studies in high‐fat‐diet‐induced obese T2DM mice, where intravenous apelin injection significantly improved glucose tolerance and insulin sensitivity.54 Additionally, peripheral apelin administration was found to improve skeletal muscle glucose utilization, decrease triglycerides, free fatty acids, adiposity, body weight, and insulinemia.51 Two enzyme degradation‐resistant acylated analogs of apelin‐13 amide, (Lys8GluPAL)apelin‐13 amide and pGlu(Lys8GluPAL)apelin‐13 amide, were found to attenuate diabetic conditions, improve weight loss, and decrease circulating triglycerides and LDL cholesterol while increasing HDL cholesterol in high‐fat‐diet‐fed mice compared to saline control.55 A clinical trial on the influence of pyr1‐apelin‐13 (exopeptidase degradation resistant) on insulin sensitivity in healthy overweight men has been completed and shown to significantly improve insulin sensitivity in a hyperinsulinemic–euglycemic clamp study as well as cause no adverse effects due to apelin administration.56 A long circulating PEGylated variant of apelin‐36 was found to significantly lower blood glucose and improve glucose tolerance in diet‐induced obese mice.57

Omentin‐1 is a novel adipokine identified from omental fat pad and is preferentially secreted by the visceral rather than subcutaneous adipose tissue.58 It has anti‐inflammatory, anti‐atherosclerotic, and cardioprotective properties.59 In addition, it enhances insulin signal transduction, regulates blood glucose via insulin‐mediated glucose uptake in adipocytes, and is involved in lipid metabolism.60, 61 Compared to healthy individuals, the serum level of omentin‐1 is significantly lower in conditions that display insulin resistance such as in obesity, T2DM, gestational diabetes, and in women with polycystic ovarian syndrome, but increases with weight reduction.61, 62 However, absence of efficacy in basal glucose uptake suggests that omentin‐1 lacks inherent insulin‐like activity.60 When rat primary cardiomyocytes exposed to conditioned media from epicardial adipose tissue of T2DM patients were treated with recombinant omentin‐1, contractile dysfunction and insulin resistance were prevented, demonstrating a cardioprotective role of the adipokine in T2DM.63 However, omentin‐1 is not being actively pursued as a therapy for T2DM in any current clinical trial.

Another novel adipokine C1q/TNF‐related protein‐3 (CTRP3, also called cartonectin) is considered an adiponectin paralog and was shown to be involved in glucose homeostasis.64 Administration of recombinant CTRP3 protein resulted in blood glucose lowering in normal and insulin‐resistant ob/ob mice, without affecting the levels of adiponectin or insulin. In human, circulating CTRP3 level is lower in obesity and negatively correlated with insulin resistance.65 Glucagon‐like peptide‐1 receptor agonist Exendin‐4 was shown to increase CTRP3 expression in vitro.66 Metformin treatment was also found to elevate serum CTRP3 levels in women with polycystic ovarian disease.67

Secreted frizzled protein‐5 (SFP5) is secreted from various tissues including visceral, subcutaneous, pericardial adipose tissues, liver, heart, pancreatic islets, and mononuclear blood cells.68 Different cross‐sectional clinical studies have found that SFP5 serum levels are inversely related to diabetes and cardiovascular disease risk factors, and its concentration is lower in prediabetes or T2DM.68, 69 Treatment with Liraglutide increased SFP5 levels.69 However, confounding results have been presented in other studies where investigators either found no significant difference in serum SFP5 levels between obese and nonobese individuals or SFP5 overexpression decreased glucose metabolism, with the latter tested only in preclinical trials.70, 71

Vaspin, also known as visceral adipose tissue‐derived serine protease inhibitor or serpin A12, is an insulin‐sensitizing adipokine.72, 73 Recombinant vaspin administration to obese mice was shown to improve glucose tolerance as well as insulin sensitivity and demonstrated to be mediated through inhibition of inflammation caused by TNF‐α, IL‐1, and other inflammatory modulators.74, 75 In contradiction to its role, vaspin concentration is elevated in obesity and T2DM, but this is generally considered a compensatory mechanism to impaired glucose homeostasis in obesity and T2DM.75 However, confounding results were documented in newly diagnosed T2DM patients where circulating vaspin levels were significantly diminished compared with age‐matched healthy controls, and a positive correlation between serum vaspin and insulin sensitivity was obtained.76 In corroboration, when overweight women with polycystic ovary syndrome were treated with metformin, vaspin levels decreased with concomitant improvement in insulin sensitivity.77

2.2. Adipokines mediating insulin resistance

Adipokines that stimulate insulin resistance include MCP‐1, RBP‐4, progranulin, chemerin, and ANGPTL2. These adipokines and formulations/strategies that inhibit their activity are discussed below.

MCP‐1 is implicated in migration and infiltration of monocytes into the adipose tissue that transition to inflammatory macrophages (M1 polarized). Inhibition of MCP‐1 using NOX‐E36 (ematicap pegol) for the treatment of T2DM and albuminuria has successfully completed phase IIa clinical trials and showed reduction in HbA1c levels in albuminuric T2DM patients.78 The drug is a Spiegelmer® (l‐isomers of RNA oligonucleotides)‐based therapeutic agent that interacts specifically with MCP‐1 and inhibits its activity.79 Very recently, the drug showed preclinical success in the treatment of liver cancer.80 Bindarit is a small‐molecule inhibitor of MCP‐1 that has completed phase II clinical trials for the treatment of diabetic nephropathy. The results indicated reduction in albuminuria upon 12‐week‐long administration of the drug.81 A MCP‐1 receptor antagonist, CCX140‐B, improved glycemic parameters in T2DM patients upon oral administration and provided renal protection in T2DM patients with nephropathy.82, 83 Antidiabetic drug Rosiglitazone (a TZD) was found to inhibit MCP‐1 in human mesangial cells subjected to mechanical stretch.84 Numerous statin drugs have also been found to decrease MCP‐1 levels and therefore can be considered as promising therapy for T2DM patients presenting hyperlipidemia.85, 86, 87 For example, a phase IV clinical trial to assess safety and efficacy of rosuvastatin monotherapy or in combination with ezetimibe is currently recruiting T2DM patients with hypercholesteremia (identifier # NCT03217409). Rosuvastatin lowers LDL cholesterol in diabetic patients and a combination of rosuvastatin and ibesartan (a MCP‐1 receptor antagonist) was shown to interfere with MCP‐1 signal transduction synergistically in vascular injury mice model.88 Similarly, low‐dose atorvastatin (Lipitor®) decreased MCP‐1 levels in T2DM patients with hyperlipidemia.89 Administration of n‐3 polyunsaturated fatty acid (PUFA) in T2DM patients resulted in significant decrease in MCP‐1 levels compared with placebo‐treated group, and therefore the anti‐inflammatory effects of n‐3 PUFAs were postulated to be mediated through modulation of MCP‐1.90

RBP‐4 is a plasma retinol transporter that shuttles retinol from liver to the peripheral tissues and is secreted by adipocytes.91 Elevated RBP‐4 levels were linked to higher body mass index (BMI) in diabetic and nondiabetic patients.92 However, this information is debatable because in at least two separate clinical trials, no significant difference in circulating RBP‐4 levels was obtained between normal and obese individuals.65, 93 In diabetes, higher circulating RBP‐4 level was positively correlated in impaired glucose tolerance, T2DM, and insulin resistance.92 The adipokine has been shown to reduce expression of glucose transporters in skeletal muscles, and thus decrease insulin sensitivity.94 Additionally, RBP4 triggers adipose tissue inflammation through activation of ATMs to secrete inflammatory cytokines.95 Expectedly, RBP4 knockdown has resulted in improved insulin sensitivity.96, 97 Rosuvastatin intake decreased serum levels of RBP‐4 in T2DM patients with hyperlipidemia.98 Another drug, fenretinide, a synthetic retinoid, was found to decrease RBP‐4 levels and improve insulin sensitivity and its long‐term therapy showed efficacy in alleviating obesity, insulin resistance, and hepatic steatosis in mice fed with high‐fat diet.99, 100 The drug is currently in phase II clinical trial to investigate its insulin sensitization efficacy in obese insulin‐resistant patients and in liver inflammation (identifier # NCT00546455). The drug is also being investigated in clinical trials as a chemotherapeutic agent for various cancers.101 A nonretinoid RBP‐4 antagonist, A1120, was shown to decrease serum RBP‐4 levels by 75% in mice.102

Chemerin is another novel adipokine that functions as a chemoattractant for monocytes and macrophages, promotes differentiation of preadipocytes, and is postulated to promulgate monocyte infiltration in adipose tissue of obese individuals, leading to low‐grade inflammation.103 The adipokine is involved in glucose and lipid homeostasis and high serum chemerin expression is found in inflammatory fluids.103 A positive cross talk between chemerin levels and impairment of glucose tolerance has been observed.104 In patients with chronic periodontitis and T2DM, chemerin levels in gingival crevicular fluid (GCF) were elevated compared with chronic periodontitis patients, suggesting that GCF chemerin can be a proinflammatory marker for diabetes and periodontal disease.105 Chemerin receptor, ChemR23 (also called CMKLR1) antagonist CCX832, was shown to reduce chemerin‐stimulated arterial contraction ex vivo and very recently shown to improve vascular function in obese diabetic mice.12, 106 However, another preclinical study demonstrated that CMKLR1 agonist rather than antagonist may be more beneficial for T2DM therapy because of chemerin agonist‐mediated increased insulin‐stimulated glucose uptake by adipocytes.107

ANGPTL2 is a recently identified adipokine that shares structural similarity with angiopoietin and found elevated in the serum of obese diabetic women.108 ANGPTL2 production was shown to be stimulated in human adipocyte culture exposed to stresses that mimic adipose tissue in obesity, and therefore the adipokine is assumed to promulgate adipose tissue inflammation and insulin resistance.108 Indeed, TNF‐α was found to induce ANGPTL2 expression in adipocytes in vitro.109 This was corroborated in a separate study where administration of recombinant ANGPTL2 to db/db mice induced proinflammatory gene production, increased macrophage subpopulation in adipose tissue that was primarily M1 polarized, and enhanced lipid accrual in the liver as well as fatty acid synthesis.110 ANGPTL2 knockdown mice showed similar insulin sensitivity and weight gain profile as wild‐type mice that underwent intermittent fasting, suggesting that reduction in ANGPTL2 levels is a promising strategy to counter development of insulin resistance and obesity.111 However, contradictory result was obtained by Kitazawa et al, who showed that exogenous administration of ANGPTL2 led to decrease in serum glucose, insulin, and fatty acids and concomitant increase in adiponectin and insulin sensitivity.112 Despite this, ANGPTL2 is mostly acknowledged as a proinflammatory adipokine involved in many chronic disorders such as diabetes and cancer.113 The adipokine is not currently under investigation in a clinical trial for treatment of T2DM.

Another novel adipokine, wingless‐related integration site‐1 (WNT‐1)‐inducible signaling pathway protein‐1 (WISP1), also known as CCN4, was shown to inhibit adipogenesis by preventing adipocyte differentiation and block PPAR‐γ transcriptional activity, thus believed to contribute toward the development of obesity.114 The proinflammatory potential of the adipokine was exhibited in vitro, when macrophages treated with WISP1 caused increased secretion of proinflammatory cytokines in a dose‐dependent fashion and polarized them to M1 phenotype.115 Serum concentration of the adipokine is elevated in obesity, irrespective of glycemia or insulin resistance status.116 This observation was reinforced in a different study, where WISP1 levels were found elevated in obese men irrespective of glycemic status compared with nonobese men.117 Additionally, the group showed that insulin signaling was impaired when hepatocytes were incubated with WISP1 due to inhibition of insulin receptor phosphorylation, other downstream regulators, and suppression of glycogenesis as well as gluconeogenesis. However, although the adipokine is positively correlated with visceral fat mass and is possibly a marker for insulin resistance, further studies are warranted to validate its role in T2DM onset or progression, before being considered for diabetes therapy.115

On the other hand, progranulin has an established role in obesity‐associated T2DM pathogenesis. Specifically, this cysteine‐rich protein decreases insulin‐mediated glucose uptake, disrupts insulin signal pathway, increases adipocyte autophagy, and promotes monocyte recruitment in adipose tissue as well as IL‐6 secretion from adipocytes.118, 119, 120 Elevated progranulin levels are present in obesity‐associated T2DM, nonalcoholic fatty liver disease, nephropathy, and retinopathy.118, 121 However, despite the proinflammatory role of progranulin in T2DM, it exhibits anti‐inflammatory function in various other disorders including arthritis, psoriasis, wound repair, and acute ischemia–reperfusion injury.119 Modulation of the adipokine is not being currently investigated for T2DM therapy in a clinical trial.

Lipocalin‐2 (LCN2) is an adipokine whose production is rapidly increased after differentiation of preadipocytes into mature adipocytes or under inflammatory conditions such as upon stimulation with lipopolysaccharide or IL‐1β.122 Moreover, LCN2 levels are reported to be high in obesity and directly proportional to fat mass, fasting blood glucose, and insulin resistance.123 Treatment with rosiglitazone decreased LCN2 serum concentrations and led to amelioration of insulin resistance in T2DM patients.123 In preclinical studies, LCN2 knock out in obese mice showed marked improvement in insulin sensitivity and glycemia compared with wild‐type mice and was attributed to suppression of production/activity of TNF‐α and 12‐lipoxygenase in the LCN2‐deficient mice.122 However, in patients with long‐term T2DM, serum LCN2 levels were found lower than healthy controls, following the similar trend as observed with anti‐inflammatory adipokines such as adiponectin.124 Administration of LCN2 shRNA using retroviral vector improved insulin activity in 3T3‐L1 adipocyte cells while exposure to LCN2 decreased insulin sensitivity in hepatocytes.125 Inhibition of LCN2 using monoclonal antibody has been explored for breast cancer therapy.126

Visfatin, an adipokine discovered in 2004, was found elevated in obesity in various clinical trials, but showed no correlation with insulin resistance, and its role in diabetes risk or progression is controversial.127, 128 Cardiotrophin‐1 is also an adipokine with divergent information about its role in regulation of glucose metabolism.129, 130, 131 Another adipokine, plasminogen activator inhibitor‐1 (PAI‐1), has been linked with T2DM but a systematic review comparing results of various clinical trials supports moderate association between them.132 Therefore, in light of the conflicting or moderate association between these adipokines and T2DM, they are not discussed in detail in this review. A summary of various adipokines that have central roles in mediating inflammation and strategies to mitigate the underlying inflammation in obesity‐associated T2DM is presented in Table 1.

Table 1.

Adipokines involved in glucose homeostasis and notable therapies investigated for modulation of their activities for diabetes therapy

Adipokine Pro/anti‐inflammatory Drugs/formulations/strategies
Adiponectin Anti‐inflammatory

Recombinant protein33, 34, 35, 36, 37, 38

AdipoRon (small‐molecule mimetic)40

Gene therapy41, 42, 43

ANGPTL2 Proinflammatory Recombinant protein110
Apelin Anti‐inflammatory

Recombinant protein51, 54

Acylated analogs of apelin‐13 amide55

pyr1‐apelin‐13 [CT]56

PEGylated apelin‐3657

Cartonectin Anti‐inflammatory

Recombinant protein64

Exendin‐466

Metformin [M]67

Chemerin Proinflammatory

Receptor antagonist—CCX832106

CMKLR1 receptor agonist107

Leptin Anti‐inflammatory

Metreleptin (Myalept™) [M]

PEGylated leptin47

Gene therapy48, 49

Pluronic P85 conjugated leptin50

Lipocalin‐2 Anti‐inflammatory

Rosiglitazone [M]123

Gene therapy125

Monocyte chemoattractant protein‐1 Proinflammatory

NOX‐E36 (ematicap pegol) [CT]78

Small‐molecule inhibitor—Bindarit [CT]81

Receptor antagonists—CCX140‐B [CT], Ibesartan [M]82, 83, 88

Statins [M]85, 86, 87

Polyunsaturated fatty acids [CT]90

Retinol‐binding protein‐4 Proinflammatory

Retinoid‐based antagonist—Fenretinide [CT]99, 100

Nonretinoid antagonist—A1120102

Secreted frizzled protein‐5 Anti‐inflammatory Liraglutide [M]69
Vaspin Anti‐inflammatory Recombinant protein74, 75

Note: FDA‐approved marketed drugs for treatment of T2DM or other indications and those in clinical trials are represented as [M] or [CT], respectively.

3. CYTOKINES AND THERAPIES THAT MODULATE THEIR ACTIVITY

Alongside adipokines, cytokines also significantly influence health and disease. Few important cytokines that promulgate diabetes or those with antidiabetic properties are discussed below. Alongside, notable drugs/formulations under investigation for modulation of their activities are reviewed.

3.1. Cytokines mediating insulin sensitization

An important cytokine possessing potent anti‐inflammatory property is IL‐10. Secreted primarily by T helper cells, monocytes, macrophages, and dendritic cells, it has a central role in curtailing inflammatory and autoimmune response of the host toward a pathogen, thus limiting inflammation‐mediated damage to the host.133, 134 Deficiency or impaired signaling/expression of IL‐10 can cause robust inflammatory response to a pathogen and development of inflammatory bowel disease or various autoimmune pathologies such as multiple sclerosis and T1DM.134, 135 T2DM patients also have significantly lower serum concentrations of IL‐10 compared with healthy individuals.135 However, serum IL‐10 levels were reported to be elevated in obese women but were lower in obese/nonobese women with metabolic syndrome.136 Various studies show that exogenous administration of recombinant IL‐10, IL‐10‐Fc protein, or plasmid DNA encoding IL‐10 can prevent onset of diabetes while treatment with antibody against IL‐10 receptor can induce diabetes in mice.137, 138, 139, 140, 141, 142, 143 One of the mechanisms of protection afforded by IL‐10 against insulin resistance in obesity is documented to be its ability to safeguard skeletal muscles from macrophage infiltration, thus prevent exposure to inflammatory conditions.144, 145 In T2DM, resistance or hyporesponsiveness to IL‐10 in macrophages exposed to high glucose may lead to deleterious glucose metabolism and techniques to overcome the hyporesponsiveness can be useful for treatment.146 To this end, a small‐molecule SHIP‐1 agonist, AQX‐MN100, that mimics anti‐inflammatory activity of IL‐10 was shown to bypass IL‐10 hyporesponsiveness and inhibit inflammation in cells exposed to high glucose.146 However, IL‐10‐based diabetes therapy is not being currently investigated in a clinical trial. A human recombinant IL‐10 formulation, Prevascar, was being developed by Renovo PLC for scar reduction but did not complete all clinical trials.147 Currently, a phase III clinical trial is underway for PEGylated human IL‐10 (AM0010, peigilodecakin) as a chemotherapeutic agent for metastatic pancreatic cancer treatment.148, 149

IL‐22 is another cytokine strongly associated with T2DM and prediabetes.150 In Han Chinese population, serum IL‐22 levels were significantly and progressively lower in participants with impaired fasting glucose and T2DM compared with normal participants.150 IL‐22 receptor‐deficient mice fed with high‐fat diet were susceptible to development of metabolic syndrome while exogenous administration of the cytokine to high‐fat‐diet‐fed obese db/db mice improved insulin sensitivity and glucose metabolism.151 Additionally, IL‐22 therapy was found to promote diabetic wound healing in vivo.152 McGuckin and coworkers demonstrated that IL‐22 therapy attenuated oxidative stress in pancreatic β cells, which suppressed inflammation and restored insulin sensitivity.153 However, other studies have shown contradictory results, stating that IL‐22 levels are elevated in T2DM.154, 155 The cytokine was termed as a “double edged sword” because IL‐22 also protected endothelial cells from glucose‐mediated injury.154 In a separate study, IL‐22 was shown to increase release of IL‐1β, a key proinflammatory cytokine, from macrophages.156 Currently, IL‐22‐based therapy is not being assessed for T2DM treatment, which is not surprising given the unclear role of the cytokine in the disease pathophysiology.

Another cytokine IL‐13 was shown to have a protective role in obesity and T2DM in preclinical trials. Although anti‐inflammatory, its expression seems to be induced in the adipose tissue by TNF‐α and IL‐1β.157 Despite being elevated in patients with insulin resistance, its levels did not show correlation with inflammation parameters such as TNF‐α and IL‐10.158 However, exogenous administration of IL‐13 improved insulin sensitivity in high‐fat‐diet C57BL/6 mice while IL‐13 gene overexpression improved glucose homeostasis by repressing adipose tissue inflammation and gluconeogenesis.158, 159 The expression of another cytokine, IL‐37, is positively correlated with insulin sensitivity and it was shown to inhibit activation of various proinflammatory signaling cycles, increase serum adiponectin levels, and decrease adipose tissue macrophages, thus protect against diet‐induced T2DM.160, 161 However, this anti‐inflammatory cytokine is not being actively investigated as a therapy in clinical trials currently. Other cytokines such as IL‐2 and IL‐4 have been primarily investigated as therapeutic agents in the context of T1DM, and therefore not being discussed in this review.162, 163, 164, 165

3.2. Cytokines mediating insulin resistance

The pathophysiology of peripheral insulin resistance in insulin‐sensitive organs mediated by proinflammatory cytokines has been illustrated in Figure 4.

Figure 4.

Figure 4

Mechanism of induction of peripheral insulin resistance due to chronic systemic presence of proinflammatory cytokines in insulin‐sensitive organs. For simplicity proinflammatory cytokine production from only M1 macrophages is shown in the figure. Various other immune cells including conventional dendritic cells, CD8+ T cells, and B cells present in the adipose tissue are also known to secrete proinflammatory cytokines

A proinflammatory cytokine with well‐established role in insulin resistance is TNF‐α.166, 167 Elevated amount of this cytokine is secreted from the adipose tissue in obesity while its knockout or receptor‐deficient animals are protected from insulin resistance development.167, 168, 169 The detrimental effect of TNF‐α on insulin signaling is caused by three different pathways: (a) reduction in insulin receptor tyrosine phosphorylation while increasing serine or threonine phosphorylation, (b) suppression of glucose transporter 4, and (c) increasing fatty acid lipolysis.170 Given the central role of TNF‐α in insulin resistance, its downregulation is a promising treatment strategy for T2DM therapy. Six different TNF‐α inhibitors have been clinically approved for the treatment of arthritis (rheumatoid, juvenile, psoriatic), plaque psoriasis, ankylosing spondylitis, ulcerative colitis, and/or Crohn's disease. These include adalumumab (Humira®), Adalimumab‐atto (Amjevita™), Certolizumab pegol (Cimzia®), Etanercept (Enbrel®), Golimumab (Simponi®), and Infliximab (Remicade®). Infliximab was also found to improve insulin sensitivity and enhance adiponectin production in patients with rheumatoid arthritis.171, 172 Similarly, in a study on 40 obese patients exhibiting impaired glucose homeostasis, prolonged etanercept administration improved fasting blood glucose and concentration of high‐molecular‐weight adiponectin.173 A pilot study carried out on newly diagnosed T1DM pediatric patients showed that etanercept lowers HbA1c levels and increases insulin production.174 However, in psoriatic patients with risk for T2DM, etanercept did not modulate insulin secretion or sensitivity.175 Similarly, adalimumab given to rheumatoid arthritis patients with insulin resistance did not improve insulin sensitivity, despite overall reduction in systemic inflammation.176 Several small‐molecule inhibitors of TNF‐α are also under investigation for inflammation modulation.177, 178 Another study on eight patients with rheumatoid arthritis or Crohn's disease with T2DM showed that treatment with anti‐TNF‐α significantly decreased fasting blood glucose levels, triglycerides, and in some cases HbA1c values compared with initial levels.179 In a preclinical study involving obese Wistar rats on high‐fat/high‐sucrose diet, blockade of TNF‐α receptor 1 using peptide Fc fusion protein for 4 weeks greatly improved insulin sensitivity compared with control.180 Likewise, silencing TNF‐α using siRNA encapsulated in glucan shells that primarily consist of β‐1,3‐d‐glucan, a ligand for the receptor of dectin‐1 expressed in macrophages, improved glucose tolerance in obese mice.181 Antidiabetic drug TZDs also block TNF‐α‐mediated impairment in insulin signaling.182 Concomitant reduction in IL‐6 and increase in adiponectin production can also be achieved with TZD therapy.170 Additionally, metformin was shown to reduce production and mRNA levels of TNF‐α in high‐fat‐diet‐fed obese mice by downregulating scavenger receptors CD36 and SR‐A in macrophages.183 Furthermore, nonsteroidal anti‐inflammatory drugs such as salicylates have shown usefulness in T2DM therapy through modulation of TNF‐α‐mediated inflammation. In a cohort of 286 patients treated for 48 weeks with salsalate (salicylate prodrug), a decrease in TNF‐α levels was observed along with lower HbA1c, fasting glucose, uric acid, and triglycerides with concomitant increase in adiponectin levels, mean HbA1c, and glycemia improvement compared with placebo group.184 Currently, the American Diabetes Association recommends the use of low‐dose aspirin for prevention of T1DM and T2DM patients with high risk of cardiovascular diseases.185

Proinflammatory cytokines in the IL‐1 family include IL‐1α and IL‐1β, of which the latter is strongly associated with development of both T1DM and T2DM.186, 187 Activation of nod‐like receptor protein 3 (NLRP3) inflammasome in the macrophages in response to metabolic stress factors recognized as “danger signals” such as increased fatty acids, ceramides, ROS, endoplasmic reticulum stress, extracellular ATP, urate, and islet amyloid polypeptide is known to trigger posttranslational modification and subsequent release of IL‐1β and IL‐18.188, 189, 190 IL‐1β is implicated in pancreatic β‐cell functional impairment, apoptosis, and consequent decrease in β‐cell mass.186, 191, 192 Additionally, high blood glucose concentration leads to stimulation of its own release from the beta cells and recruitment of macrophages that further increase IL‐1β levels.186, 191, 192 Several IL‐1β knockdown strategies including IL‐1β receptor antagonist have been investigated for the treatment of T2DM. Out of these, the most notable one includes Anakinra (Kineret®), a marketed IL‐1β receptor antagonist used for the treatment of rheumatoid arthritis. A multicenter, randomized phase IV TRACK [Treatment of Rheumatoid Arthritis and Comorbidities with Kineret (anakinra)] study was being conducted to determine the effect of anakinra on lowering HbA1c levels in rheumatoid arthritis patients with T2DM (identifier # NCT02236481), but was terminated by the sponsor for early benefits. It was reported that treatment with anakinra on two patients with rheumatoid arthritis and T2DM was beneficial in reaching the therapeutic targets for both diseases.193 In patients with gout and T2DM, Anakinra treatment resulted in significant improvement in joint functions and glycemic control.194 A week‐long daily administration of 100 mg anakinra in insulin‐resistant T1DM patients resulted in significant improvement in insulin sensitivity for 4 weeks after treatment.195 Other clinically approved IL‐1β‐based treatments for crypopyrin‐associated periodic syndromes include canakinumab (Ilaris®, IL‐1β antagonist) and rilonacept (Arcalyst®, soluble decoy IL‐1 receptor).196 In the context of diabetes, treatment with canakinumab did not prevent progression from prediabetes to new onset diabetes nor decrease HbA1c and glucose levels over a long term.197 In this clinical trial, decrease in HbA1c was observed only during the first 6–9 months of treatment but not consistently over a longer period. Indeed, a decrease in HbA1c was noted in an earlier trial in T2DM patients on metformin upon using canakinumab for 4 months.198 On the other hand, rilonacept has been tested for T1DM therapy in a phase I study and showed good tolerance, but posited “unlikely to be efficacious as a single agent in preserving beta‐cell function.”199 Gevokizumab is another IL‐1β antagonist that enhanced insulin sensitivity and decreased inflammation in T2DM patients.200 A high‐affinity neutralizing antibody against IL‐1β, LY2189102, was also found to decrease inflammation and HbA1c.201 Similarly, a monoclonal antibody against IL‐1 β, XOMA052, was shown to prevent high‐fat‐diet‐induced impaired glucose tolerance and insulin production while decreasing β‐cell death in obese mice.202 Furthermore, Zha et al have developed an epitope peptide vaccine targeting IL‐1β with polylactic acid microsphere adjuvant to neutralize IL‐1β activity.203 When administered to diabetic KK‐Ay mice, the formulation was able to enhance glucose tolerance, insulin sensitivity while decrease free fatty acids in serum, cholesterol, β‐cell death, and IL‐1β production. Likewise, a different peptide antagonist of IL‐1β, SER140, was found to preserve β‐cell mass and delay the diabetes development in NOD mice.204

Another interleukin IL‐6, traditionally considered proinflammatory, is now believed to display both inflammatory and protective roles against inflammation, and its contribution toward T1DM or T2DM development remains debatable.205 However, Tocilizumab, an anti‐IL‐6 receptor antibody, was found to decrease HbA1c levels in patients to a greater extent than TNF‐α inhibitors.206 Similarly, there is conflicting evidence about the role of IL‐18 in T2DM.207, 208 While the serum levels of IL‐18 are elevated in obese and T2DM patients, therefore thought to mediate insulin resistance, studies have also shown glucose‐lowering potential of IL‐18.208, 209, 210 Other interleukins such as IL‐12 and IL‐17 have been reported elevated in T2DM.211, 212 However, in a preclinical study, IL‐17 was shown to negatively correlate with glucose homeostasis and adipogenesis.213 An antibody, ustekinumab (Stelara®), that binds to IL‐12 and IL‐23 and approved for the treatment of plaque psoriasis is currently undergoing phase II/III clinical trials for the treatment of T1DM (identifier # NCT03941132). Owing to contradictory evidence of these cytokines about their role in T2DM development, they are not discussed in detail in this review.

Resistin is another hormone secreted by adipocytes in mice but primarily expressed by macrophages and peripheral blood mononuclear cells (PBMCs) in humans.214 As the name suggests, the hormone is strongly associated with inflammation and insulin resistance. Heightened serum levels of resistin are present in diet‐induced obese mice and in diabetic mice.215 In humans, a significant positive correlation between fasting serum resistin levels, C‐reactive protein (inflammation biomarker), insulin resistance, fasting blood glucose levels, body mass index, and triglycerides was found and it was associated with T2DM‐induced retinopathy.216 The production of human resistin from macrophages and PBMCs is escalated upon treatment with lipopolysaccharide and proinflammatory cytokines while on the other hand, presence of human resistin also induces production of proinflammatory adipocytokines, leading to a vicious cycle of chronic inflammation.214 However, conflicting reports showing no positive correlation between serum resistin levels and insulin resistance markers as well as adiposity have also been documented in T2DM patients, probably making its role in human diabetes contentious.217 As a drug target, several antagonists of resistin have been developed, such as a mutant version of resistin (C6A), which was shown to improve glucose tolerance and insulin sensitivity in high‐fat‐diet‐fed insulin‐resistant mice.218 A resistin binding peptide was developed by Gu et al and shown to negate the function of resistin in vitro.219 In overweight women with polycystic ovarian disease, rosiglitazone decreased resistin levels and increased adiponectin levels after 4 months of treatment.220 A 12‐week‐long treatment comparing efficacy of pioglitazone and metformin in decreasing serum resistin levels, fasting blood glucose, and insulin concentration was completed in T2DM children, but results of the clinical trial have not been posted (identifier # NCT01396564). Antibodies against resistin and its putative receptors such as toll‐like receptor 4 and adenylyl cyclase‐associated protein 1 can be candidate therapeutic agents to antagonize resistin function.221, 222 Antioxidants may also be useful in lowering resistin levels. A clinical trial studying the effect of intake of alpha‐lipoic acid on serum resistin levels in T2DM patients with chronic periodontitis demonstrated that systemic administration of alpha‐lipoic acid as an adjunct therapy to scaling and root planning significantly decreased serum resistin and HbA1c in the patients.223

Transforming growth factor β (TGF‐β) secreted by macrophages and monocytes drives renal cell hypertrophy and buildup of extracellular matrix, and therefore plays a central role in diabetic nephropathy, a long‐term complication of T2DM.224, 225, 226, 227 The cytokine levels are elevated in serum and urine of T2DM individuals with diabetes nephropathy.228 An increased expression of the cytokine was also observed in adipose tissues of obese mice and attributed to upregulation of TNF‐α levels.229 Blockade of TGF‐β signaling with anti‐TGF‐β1 antibody protected mice from diet‐induced obesity and diabetes.230 An antibody against TGF‐β, fresolimumab (GC 1008), is being investigated for the treatment of glomerulosclerosis, systemic sclerosis, pulmonary fibrosis, and various cancer, but not in particular context of insulin resistance.230, 231, 232

CXCL5 secreted by white adipose tissue macrophages is also known to promote obesity‐induced insulin resistance and various other obesity‐related inflammatory disorders including atherosclerosis, retinopathy, and ulcerative colitis, among others.233, 234 Its expression is regulated by TNF‐α and significantly increased in obese subjects compared with lean individuals.233, 235 CXCL5 inhibition in insulin‐resistant obese mice with anti‐CXCL5 antibody or its specific receptor antagonist significantly improved glucose tolerance and insulin sensitivity.233, 234 Diabetes treatment based on CXCL5 neutralization is not being currently investigated in a clinical trial.

Macrophage migration inhibitory factor (MIF) is a proinflammatory cytokine secreted from a variety of cells including T‐lymphocytes, macrophages/monocytes, eosinophils, neutrophils, and endothelial cells, among others.236 Serum MIF level is elevated in prediabetes, T2DM, and gestational diabetes and a positive correlation was found between serum MIF concentration and impaired glucose tolerance, insulin resistance, BMI, and visceral fat mass in various clinical trials.237, 238 Although originally recognized as macrophage migration inhibitor, the cytokine is known to induce secretion of proinflammatory cytokines including TNF‐α, IL‐1, 6, 8, and 12, recruit various inflammatory cells, and upregulate expression of adhesion molecules on monocytes and endothelial cells.236 Several small‐molecule inhibitors of MIF have been investigated for cancer therapy, inflammatory, and immune disorders.239, 240, 241 An oral small‐molecule inhibitor of MIF, CSPI‐1306, was found to reduce blood glucose, TNF‐α, and IL‐6 levels in type 2 diabetic outbred Institute of Cancer Research (ICR) mice.242 Administration of another small‐molecule inhibitor of MIF, ISO‐1, to diabetic db/db mice reduced blood glucose and diabetic nephropathy conditions such as albuminuria, activation of macrophages, and extracellular matrix growth in diabetic kidney.243 Inhibition of MIF receptor, CD74, using specific shRNA decreased high‐fat‐diet‐induced polarization of macrophages to M1 phenotype and insulin resistance in mice.244 Additionally, RNAi therapy to inhibit cyclooxygenase‐2 (COX‐2) was found to reduce MIF levels in adipocytes exposed to high fat.244

A summary of various cytokines that have central roles in mediating inflammation and strategies to mitigate the underlying inflammation in obesity‐associated T2DM are presented in Table 2.

Table 2.

Cytokines involved in glucose homeostasis and notable therapies investigated for modulation of their activities for diabetes therapy

Cytokine Pro/anti‐inflammatory Drugs/formulations/strategies
CXCL5 Proinflammatory Antibodies233, 234
Interleukin‐1β Proinflammatory

Inhibitors—Anakinra [M], Canakinumab [M], Rilonacept [M], Gevokizumab [CT], LY2189102 [CT]193, 194, 195, 197, 198, 199, 200

Antibodies201, 202

Peptide antagonists203, 204

Interleukins 10, 13, 22, 37 Anti‐inflammatory

Recombinant protein138

Gene therapy139, 140, 141, 142

IL‐10‐Fc protein143

Small‐molecule mimetic—AQX‐MN100146

Macrophage inhibitory factor Proinflammatory

Small‐molecule inhibitors242, 243

Gene therapy244

Transforming growth factor‐β Proinflammatory Antibody230
Tumor necrosis factor‐α Proinflammatory

Inhibitors—Etanercept [M], Infliximab [M]171, 172, 173, 174

Small‐molecule inhibitors177, 178

Receptor blockade—Fc fusion protein180

Gene therapy181

Thiazolidinediones [M]170, 182

Metformin [M]183

Salicylates [M]184

Resistin Proinflammatory

Mutant resistin (C6A)218

Rosiglitazone [M]220

Antibodies against resistin and its receptor219, 221, 222

Alpha lipoic acid [CT]223

Note: FDA‐approved marketed drugs for treatment of T2DM or other indications and those in clinical trials are represented as [M] or [CT], respectively.

Taken together, a strong correlation exists between expression of several adipocytokines and systemic inflammation leading to insulin resistance in obesity‐associated T2DM. Therefore, these molecules present an appealing class of compounds that can be modulated to obtain significant therapeutic benefit in T2DM treatment. Consequently, several strategies are being developed to mitigate the underlying inflammation in obesity‐associated T2DM.

4. CONCLUSIONS

The adipose tissue is now considered an endocrine organ that secretes various molecules with paracrine or endocrine functions. Among them, adipocytokines secreted from adipocytes or adipose tissue immune cells are vital inflammatory modulators that regulate various physiological processes including metabolism and immunologic response. Increase in adiposity alters adipose tissue microenvironment and skews resident cells toward proinflammatory phenotype that significantly impairs glucose homeostasis and leads to development of obesity and T2DM. Hence, remodeling of adipose tissue inflammasome through immunoneutralization, small‐molecule inhibitors/agonists, exogenous administration of adipocytokines or mimetics, gene therapy, and novel formulations can bring significant benefit in T2DM therapy. Further development of these treatment strategies is essential to treat the underlying cause of insulin resistance in obesity‐associated T2DM, and thereby improve diabetes treatment outcome. The diversity of adipocytokines and immune cells involved in type 2 diabetes maintenance/progression opens prospect of disease modulation through various novel approaches. However, many adipocytokines are multifaceted and their physiological role is not well understood. Targeting adipose tissue inflammation for successful therapies would therefore first entail better understanding of the functions and crosstalk between various adipocytokines in the context of diabetes. Additionally, alleviation of inflammation in other tissues or organs that undergo fat cell and/or proinflammatory immune cell infiltration such as the muscle, liver, and pancreas should also be considered for developing effective T2DM therapy.

CONFLICT OF INTEREST

The authors declare no conflict of interest.

AUTHOR CONTRIBUTIONS

All authors have read and approved the final manuscript. A.B. conceived the review topic and wrote the manuscript. J.S. critically reviewed the manuscript.

ACKNOWLEDGMENTS

The authors thank North Dakota Established Program to Stimulate Competitive Research (ND EPSCoR) for its seed award funding and National Institutes of Health (NIH) grant R15GM114701. The figures were created using smart servier medical art templates licensed under a creative common attribution 3.0 unported license.

Banerjee A, Singh J. Remodeling adipose tissue inflammasome for type 2 diabetes mellitus treatment: Current perspective and translational strategies. Bioeng Transl Med. 2020;5:e10150 10.1002/btm2.10150

Peer Review The peer review history for this article is available at https://publons.com/publon/10.1002/btm2.10150.

Funding information National Institute of Health, Grant/Award Number: R15GM114701; North Dakota Established Program to Stimulate Competitive Research Seed Funds, Grant/Award Number: FAR0030636

REFERENCES

  • 1. Evans JL, Balkan B, Chuang E, Rushakoff RJ. Oral and injectable (non‐insulin) pharmacological agents for type 2 diabetes In: Feingold KR, Anawalt B, Boyce A, et al., eds. Endotext. South Dartmouth, MA: MDText.com, Inc; 2000, 1‐112. [Google Scholar]
  • 2. Rowley WR, Bezold C, Arikan Y, Byrne E, Krohe S. Diabetes 2030: insights from yesterday, today, and future trends. Popul Health Manag. 2017;20(1):6‐12. 10.1089/pop.2015.0181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. World Health Organization . Diabetes key facts. https://www.who.int/news-room/fact-sheets/detail/diabetes. Updated 2018. Accessed May 14, 2019.
  • 4. National Institute of Diabetes and Digestive and Kidney Diseases . Risk factors for type 2 diabetes. https://www.niddk.nih.gov/health-information/diabetes/overview/risk-factors-type-2-diabetes. Updated 2016. Accessed May 14, 2019.
  • 5. Garber AJ, Abrahamson MJ, Barzilay JI, et al. Consensus statement by the American association of clinical endocrinologists and American college of endocrinology on the comprehensive type 2 diabetes management algorithm—2018 executive summary. Endocr Pract. 2018;24(1):91‐120. [DOI] [PubMed] [Google Scholar]
  • 6. Kanda H, Tateya S, Tamori Y, et al. MCP‐1 contributes to macrophage infiltration into adipose tissue, insulin resistance, and hepatic steatosis in obesity. J Clin Invest. 2006;116(6):1494‐1505. 10.1172/JCI26498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Panee J. Monocyte chemoattractant protein 1 (MCP‐1) in obesity and diabetes. Cytokine. 2012;60(1):1‐12. 10.1016/j.cyto.2012.06.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Boutens L, Stienstra R. Adipose tissue macrophages: going off track during obesity. Diabetologia. 2016;59(5):879‐894. 10.1007/s00125-016-3904-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Makki K, Froguel P, Wolowczuk I. Adipose tissue in obesity‐related inflammation and insulin resistance: cells, cytokines, and chemokines. ISRN Inflamm. 2013;2013:139239 10.1155/2013/139239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Lu J, Zhao J, Meng H, Zhang X. Adipose tissue‐resident immune cells in obesity and type 2 diabetes. Front Immunol. 2019;10:1173 10.3389/fimmu.2019.01173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Wang Q, Wu H. T cells in adipose tissue: critical players in immunometabolism. Front Immunol. 2018;9:2509 10.3389/fimmu.2018.02509. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Bluher M. Adipokines—removing road blocks to obesity and diabetes therapy. Mol Metab. 2014;3(3):230‐240. 10.1016/j.molmet.2014.01.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Olefsky JM, Glass CK. Macrophages, inflammation, and insulin resistance. Annu Rev Physiol. 2010;72:219‐246. 10.1146/annurev-physiol-021909-135846. [DOI] [PubMed] [Google Scholar]
  • 14. Bastard JP, Maachi M, Lagathu C, et al. Recent advances in the relationship between obesity, inflammation, and insulin resistance. Eur Cytokine Netw. 2006;17(1):4‐12. [PubMed] [Google Scholar]
  • 15. Shoelson SE, Herrero L, Naaz A. Obesity, inflammation, and insulin resistance. Gastroenterology. 2007;132(6):2169‐2180. [DOI] [PubMed] [Google Scholar]
  • 16. Chen L, Chen R, Wang H, Liang F. Mechanisms linking inflammation to insulin resistance. Int J Endocrinol. 2015;2015:508409 10.1155/2015/508409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Stafeev IS, Vorotnikov AV, Ratner EI, Menshikov MY, Parfyonova YV. Latent inflammation and insulin resistance in adipose tissue. Int J Endocrinol. 2017;2017:5076732 10.1155/2017/5076732. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Xita N, Tsatsoulis A. Adiponectin in diabetes mellitus. Curr Med Chem. 2012;19(32):5451‐5458. [DOI] [PubMed] [Google Scholar]
  • 19. Jing Y, Wu F, Li D, Yang L, Li Q, Li R. Metformin improves obesity‐associated inflammation by altering macrophages polarization. Mol Cell Endocrinol. 2018;461:256‐264. [DOI] [PubMed] [Google Scholar]
  • 20. Koppaka S, Kehlenbrink S, Carey M, et al. Reduced adipose tissue macrophage content is associated with improved insulin sensitivity in thiazolidinedione‐treated diabetic humans. Diabetes. 2013;62(6):1843‐1854. 10.2337/db12-0868. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Esterson YB, Zhang K, Koppaka S, et al. Insulin sensitizing and anti‐inflammatory effects of thiazolidinediones are heightened in obese patients. J Invest Med. 2013;61(8):1152‐1160. 10.2310/JIM.0000000000000017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Colmers IN, Bowker SL, Majumdar SR, Johnson JA. Use of thiazolidinediones and the risk of bladder cancer among people with type 2 diabetes: a meta‐analysis. CMAJ. 2012;184(12):E675‐E683. 10.1503/cmaj.112102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Singh S, Loke YK, Furberg CD. Thiazolidinediones and heart failure: a teleo‐analysis. Diabetes Care. 2007;30(8):2148‐2153. [DOI] [PubMed] [Google Scholar]
  • 24. Idris I, Warren G, Donnelly R. Association between thiazolidinedione treatment and risk of macular edema among patients with type 2 diabetes. Arch Intern Med. 2012;172(13):1005‐1011. 10.1001/archinternmed.2012.1938. [DOI] [PubMed] [Google Scholar]
  • 25. Agrawal NK, Kant S. Targeting inflammation in diabetes: newer therapeutic options. World J Diabetes. 2014;5(5):697‐710. 10.4239/wjd.v5.i5.697. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Dinarello CA. Anti‐inflammatory agents: present and future. Cell. 2010;140(6):935‐950. 10.1016/j.cell.2010.02.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Bluher M. Adipose tissue inflammation: a cause or consequence of obesity‐related insulin resistance? Clin Sci (Lond). 2016;130(18):1603‐1614. 10.1042/CS20160005. [DOI] [PubMed] [Google Scholar]
  • 28. Chandran M, Phillips SA, Ciaraldi T, Henry RR. Adiponectin: more than just another fat cell hormone? Diabetes Care. 2003;26(8):2442‐2450. [DOI] [PubMed] [Google Scholar]
  • 29. Wang Y, Meng RW, Kunutsor SK, et al. Plasma adiponectin levels and type 2 diabetes risk: a nested case‐control study in a Chinese population and an updated meta‐analysis. Sci Rep. 2018;8(1):406 10.1038/s41598-017-18709-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Lihn AS, Pedersen SB, Richelsen B. Adiponectin: action, regulation and association to insulin sensitivity. Obes Rev. 2005;6(1):13‐21. [DOI] [PubMed] [Google Scholar]
  • 31. Aleidi S, Issa A, Bustanji H, Khalil M, Bustanji Y. Adiponectin serum levels correlate with insulin resistance in type 2 diabetic patients. Saudi Pharm J. 2015;23(3):250‐256. 10.1016/j.jsps.2014.11.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Kadowaki T, Yamauchi T, Kubota N, Hara K, Ueki K, Tobe K. Adiponectin and adiponectin receptors in insulin resistance, diabetes, and the metabolic syndrome. J Clin Invest. 2006;116(7):1784‐1792. 10.1172/JCI29126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Achari AE, Jain SK. Adiponectin, a therapeutic target for obesity, diabetes, and endothelial dysfunction. Int J Mol Sci. 2017;18(6):E1321 10.3390/ijms18061321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Ruan H, Dong LQ. Adiponectin signaling and function in insulin target tissues. J Mol Cell Biol. 2016;8(2):101‐109. 10.1093/jmcb/mjw014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Zoico E, Garbin U, Olioso D, et al. The effects of adiponectin on interleukin‐6 and MCP‐1 secretion in lipopolysaccharide‐treated 3T3‐L1 adipocytes: role of the NF‐kappaB pathway. Int J Mol Med. 2009;24(6):847‐851. [DOI] [PubMed] [Google Scholar]
  • 36. Berg AH, Combs TP, Du X, Brownlee M, Scherer PE. The adipocyte‐secreted protein Acrp30 enhances hepatic insulin action. Nat Med. 2001;7(8):947‐953. 10.1038/90992. [DOI] [PubMed] [Google Scholar]
  • 37. Fruebis J, Tsao TS, Javorschi S, et al. Proteolytic cleavage product of 30‐kDa adipocyte complement‐related protein increases fatty acid oxidation in muscle and causes weight loss in mice. Proc Natl Acad Sci U S A. 2001;98(4):2005‐2010. 10.1073/pnas.041591798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Masaki T, Chiba S, Yasuda T, et al. Peripheral, but not central, administration of adiponectin reduces visceral adiposity and upregulates the expression of uncoupling protein in agouti yellow (Ay/a) obese mice. Diabetes. 2003;52(9):2266‐2273. [DOI] [PubMed] [Google Scholar]
  • 39. Tullin S, Sams A, Brandt J, et al. Recombinant adiponectin does not lower plasma glucose in animal models of type 2 diabetes. PLoS One. 2012;7(10):e44270 10.1371/journal.pone.0044270. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Okada‐Iwabu M, Yamauchi T, Iwabu M, et al. A small‐molecule AdipoR agonist for type 2 diabetes and short life in obesity. Nature. 2013;503(7477):493‐499. 10.1038/nature12656. [DOI] [PubMed] [Google Scholar]
  • 41. Kandasamy AD, Sung MM, Boisvenue JJ, Barr AJ, Dyck JR. Adiponectin gene therapy ameliorates high‐fat, high‐sucrose diet‐induced metabolic perturbations in mice. Nutr Diabetes. 2012;2:e45 10.1038/nutd.2012.18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Park JH, Lee M, Kim SW. Non‐viral adiponectin gene therapy into obese type 2 diabetic mice ameliorates insulin resistance. J Control Release. 2006;114(1):118‐125. 10.1016/j.jconrel.2006.05.008. [DOI] [PubMed] [Google Scholar]
  • 43. Fukushima M, Hattori Y, Tsukada H, et al. Adiponectin gene therapy of streptozotocin‐induced diabetic mice using hydrodynamic injection. J Gene Med. 2007;9(11):976‐985. 10.1002/jgm.1104. [DOI] [PubMed] [Google Scholar]
  • 44. Denroche HC, Huynh FK, Kieffer TJ. The role of leptin in glucose homeostasis. J Diabetes Investig. 2012;3(2):115‐129. 10.1111/j.2040-1124.2012.00203.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Coppari R, Bjorbaek C. Leptin revisited: its mechanism of action and potential for treating diabetes. Nat Rev Drug Discov. 2012;11(9):692‐708. 10.1038/nrd3757. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Myers MG, Cowley MA, Munzberg H. Mechanisms of leptin action and leptin resistance. Annu Rev Physiol. 2008;70:537‐556. 10.1146/annurev.physiol.70.113006.100707. [DOI] [PubMed] [Google Scholar]
  • 47. Banks WA, Lebel CR. Strategies for the delivery of leptin to the CNS. J Drug Target. 2002;10(4):297‐308. 10.1080/10611860290031895. [DOI] [PubMed] [Google Scholar]
  • 48. Lundberg C, Jungles SJ, Mulligan RC. Direct delivery of leptin to the hypothalamus using recombinant adeno‐associated virus vectors results in increased therapeutic efficacy. Nat Biotechnol. 2001;19(2):169‐172. 10.1038/84448. [DOI] [PubMed] [Google Scholar]
  • 49. Turner RT, Dube M, Branscum AJ, et al. Hypothalamic leptin gene therapy reduces body weight without accelerating age‐related bone loss. J Endocrinol. 2015;227(3):129‐141. 10.1530/JOE-15-0280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Yuan D, Yi X, Zhao Y, et al. Intranasal delivery of N‐terminal modified leptin‐pluronic conjugate for treatment of obesity. J Control Release. 2017;263:172‐184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Castan‐Laurell I, Dray C, Attane C, Duparc T, Knauf C, Valet P. Apelin, diabetes, and obesity. Endocrine. 2011;40(1):1‐9. 10.1007/s12020-011-9507-9. [DOI] [PubMed] [Google Scholar]
  • 52. Li L, Yang G, Li Q, et al. Changes and relations of circulating visfatin, apelin, and resistin levels in normal, impaired glucose tolerance, and type 2 diabetic subjects. Exp Clin Endocrinol Diabetes. 2006;114(10):544‐548. 10.1055/s-2006-948309. [DOI] [PubMed] [Google Scholar]
  • 53. Reinehr T, Woelfle J, Roth CL. Lack of association between apelin, insulin resistance, cardiovascular risk factors, and obesity in children: a longitudinal analysis. Metabolism. 2011;60(9):1349‐1354. 10.1016/j.metabol.2011.02.005. [DOI] [PubMed] [Google Scholar]
  • 54. Dray C, Knauf C, Daviaud D, et al. Apelin stimulates glucose utilization in normal and obese insulin‐resistant mice. Cell Metab. 2008;8(5):437‐445. 10.1016/j.cmet.2008.10.003. [DOI] [PubMed] [Google Scholar]
  • 55. O'Harte FPM, Parthsarathy V, Hogg C, Flatt PR. Long‐term treatment with acylated analogues of apelin‐13 amide ameliorates diabetes and improves lipid profile of high‐fat fed mice. PLoS One. 2018;13(8):e0202350 10.1371/journal.pone.0202350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Gourdy P, Cazals L, Thalamas C, et al. Apelin administration improves insulin sensitivity in overweight men during hyperinsulinaemic‐euglycaemic clamp. Diabetes Obes Metab. 2018;20(1):157‐164. 10.1111/dom.13055. [DOI] [PubMed] [Google Scholar]
  • 57. Galon‐Tilleman H, Yang H, Bednarek MA, et al. Apelin‐36 modulates blood glucose and body weight independently of canonical APJ receptor signaling. J Biol Chem. 2017;292(5):1925‐1933. 10.1074/jbc.M116.748103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Tan BK, Adya R, Farhatullah S, et al. Omentin‐1, a novel adipokine, is decreased in overweight insulin‐resistant women with polycystic ovary syndrome: ex vivo and in vivo regulation of omentin‐1 by insulin and glucose. Diabetes. 2008;57(4):801‐808. 10.2337/db07-0990. [DOI] [PubMed] [Google Scholar]
  • 59. Watanabe T, Watanabe‐Kominato K, Takahashi Y, Kojima M, Watanabe R. Adipose tissue‐derived omentin‐1 function and regulation. Compr Physiol. 2017;7(3):765‐781. 10.1002/cphy.c160043. [DOI] [PubMed] [Google Scholar]
  • 60. Elsaid NH, Sadik NA, Ahmed NR, Fayez SE, Mohammed NAE. Serum omentin‐1 levels in type 2 diabetic obese women in relation to glycemic control, insulin resistance and metabolic parameters. J Clin Transl Endocrinol. 2018;13:14‐19. 10.1016/j.jcte.2018.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Lesna J, Ticha A, Hyspler R, et al. Omentin‐1 plasma levels and cholesterol metabolism in obese patients with diabetes mellitus type 1: impact of weight reduction. Nutr Diabetes. 2015;5:e183 10.1038/nutd.2015.33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Lis I, Pilarski L, Bogdanski P. Omentin—a newly‐discovered adipocytokine in insulin resistance pathogenesis. Pol Merkur Lekarski. 2015;39(229):56‐60. [PubMed] [Google Scholar]
  • 63. Greulich S, Chen WJ, Maxhera B, et al. Cardioprotective properties of omentin‐1 in type 2 diabetes: evidence from clinical and in vitro studies. PLoS One. 2013;8(3):e59697 10.1371/journal.pone.0059697. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Peterson JM, Wei Z, Wong GW. C1q/TNF‐related protein‐3 (CTRP3), a novel adipokine that regulates hepatic glucose output. J Biol Chem. 2010;285(51):39691‐39701. 10.1074/jbc.M110.180695. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Deng W, Li C, Zhang Y, et al. Serum C1q/TNF‐related protein‐3 (CTRP3) levels are decreased in obesity and hypertension and are negatively correlated with parameters of insulin resistance. Diabetol Metab Syndr. 2015;7:33 10.1186/s13098-015-0029-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Li X, Jiang L, Yang M, Wu Y, Sun S, Sun J. GLP‐1 receptor agonist increases the expression of CTRP3, a novel adipokine, in 3T3‐L1 adipocytes through PKA signal pathway. J Endocrinol Invest. 2015;38(1):73‐79. 10.1007/s40618-014-0156-8. [DOI] [PubMed] [Google Scholar]
  • 67. Tan BK, Chen J, Hu J, et al. Metformin increases the novel adipokine cartonectin/CTRP3 in women with polycystic ovary syndrome. J Clin Endocrinol Metab. 2013;98(12):E1891‐E1900. 10.1210/jc.2013-2227. [DOI] [PubMed] [Google Scholar]
  • 68. Carstensen‐Kirberg M, Kannenberg JM, Huth C, et al. Inverse associations between serum levels of secreted frizzled‐related protein‐5 (SFRP5) and multiple cardiometabolic risk factors: KORA F4 study. Cardiovasc Diabetol. 2017;16(1):109 10.1186/s12933-017-0591-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Hu W, Li L, Yang M, et al. Circulating Sfrp5 is a signature of obesity‐related metabolic disorders and is regulated by glucose and liraglutide in humans. J Clin Endocrinol Metab. 2013;98(1):290‐298. 10.1210/jc.2012-2466. [DOI] [PubMed] [Google Scholar]
  • 70. Rulifson IC, Majeti JZ, Xiong Y, et al. Inhibition of secreted frizzled‐related protein 5 improves glucose metabolism. Am J Physiol Endocrinol Metab. 2014;307(12):E1144‐E1152. 10.1152/ajpendo.00283.2014. [DOI] [PubMed] [Google Scholar]
  • 71. Wang R, Hong J, Liu R, et al. SFRP5 acts as a mature adipocyte marker but not as a regulator in adipogenesis. J Mol Endocrinol. 2014;53(3):405‐415. 10.1530/JME-14-0037. [DOI] [PubMed] [Google Scholar]
  • 72. Wada J. Vaspin: a novel serpin with insulin‐sensitizing effects. Expert Opin Investig Drugs. 2008;17(3):327‐333. 10.1517/13543784.17.3.327. [DOI] [PubMed] [Google Scholar]
  • 73. Li Q, Chen R, Moriya J, et al. A novel adipocytokine, visceral adipose tissue‐derived serine protease inhibitor (vaspin), and obesity. J Int Med Res. 2008;36(4):625‐629. 10.1177/147323000803600402. [DOI] [PubMed] [Google Scholar]
  • 74. Chadt A, Scherneck S, Joost HG, Al‐Hasani H. Molecular links between obesity and diabetes: “Diabesity” In: Feingold KR, Anawalt B, Boyce A, et al., eds. Endotext. South Dartmouth, MA: MDText.com, Inc; 2000. [Google Scholar]
  • 75. Dimova R, Tankova T. The role of vaspin in the development of metabolic and glucose tolerance disorders and atherosclerosis. Biomed Res Int. 2015;2015:823481 10.1155/2015/823481. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Tasnim F, Faruque MO, Hassan Z, Ali L. Serum vaspin levels are associated with decreased insulin sensitivity in newly diagnosed type 2 diabetes mellitus in Bangladesh. J Taibah Univ Med Sci. 2015;10(3):327‐332. 10.1016/j.jtumed.2015.02.010. [DOI] [Google Scholar]
  • 77. Tan BK, Heutling D, Chen J, et al. Metformin decreases the adipokine vaspin in overweight women with polycystic ovary syndrome concomitant with improvement in insulin sensitivity and a decrease in insulin resistance. Diabetes. 2008;57(6):1501‐1507. 10.2337/db08-0127. [DOI] [PubMed] [Google Scholar]
  • 78. Menne J, Eulberg D, Beyer D, et al. C‐C motif‐ligand 2 inhibition with emapticap pegol (NOX‐E36) in type 2 diabetic patients with albuminuria. Nephrol Dial Transplant. 2017;32(2):307‐315. 10.1093/ndt/gfv459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Vater A, Klussmann S. Turning mirror‐image oligonucleotides into drugs: the evolution of spiegelmer® therapeutics. Drug Discov Today. 2015;20(1):147‐155. 10.1016/j.drudis.2014.09.004. [DOI] [PubMed] [Google Scholar]
  • 80. Bartneck M, Schrammen PL, Mockel D, et al. The CCR2(+) macrophage subset promotes pathogenic angiogenesis for tumor vascularization in fibrotic livers. Cell Mol Gastroenterol Hepatol. 2019;7(2):371‐390. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Perez‐Gomez MV, Sanchez‐Nino MD, Sanz AB, et al. Horizon 2020 in diabetic kidney disease: the clinical trial pipeline for add‐on therapies on top of renin angiotensin system blockade. J Clin Med. 2015;4(6):1325‐1347. 10.3390/jcm4061325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Hanefeld M, Schell E, Gouni‐Berthold I, et al. Orally‐administered chemokine receptor CCR2 antagonist CCX140‐B in type 2 diabetes: a pilot double‐blind, randomized clinical trial. J Diab Metab. 2012;3(225):1‐8. [Google Scholar]
  • 83. de Zeeuw D, Bekker P, Henkel E, et al.; CCX140‐B Diabetic Nephropathy Study Group .The effect of CCR2 inhibitor CCX140‐B on residual albuminuria in patients with type 2 diabetes and nephropathy: a randomised trial. Lancet Diabetes Endocrinol. 2015;3(9):687‐696. 10.1016/S2213-8587(15)00261-2. [DOI] [PubMed] [Google Scholar]
  • 84. Gruden G, Setti G, Hayward A, et al. Mechanical stretch induces monocyte chemoattractant activity via an NF‐kappaB‐dependent monocyte chemoattractant protein‐1‐mediated pathway in human mesangial cells: inhibition by rosiglitazone. J Am Soc Nephrol. 2005;16(3):688‐696. [DOI] [PubMed] [Google Scholar]
  • 85. Eldor R, Raz I. American diabetes association indications for statins in diabetes: is there evidence? Diabetes Care. 2009;32(suppl 2):S384‐S391. 10.2337/dc09-S345. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Romano M, Diomede L, Sironi M, et al. Inhibition of monocyte chemotactic protein‐1 synthesis by statins. Lab Invest. 2000;80(7):1095‐1100. [DOI] [PubMed] [Google Scholar]
  • 87. Waiyaput W, Pumipichet S, Weerakiet S, Rattanasiri S, Sophonsritsuk A. Effect of simvastatin on monocyte chemoattractant protein‐1 expression in endometriosis patients: a randomized controlled trial. BMC Womens Health. 2017;17(1):89 10.1186/s12905-017-0446-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Ohshima K, Mogi M, Nakaoka H, et al. Inhibition of MCP‐1/CCR2 signaling pathway is involved in synergistic inhibitory effects of irbesartan with rosuvastatin on vascular remodeling. J Am Soc Hypertens. 2012;6(6):375‐384. 10.1016/j.jash.2012.10.002. [DOI] [PubMed] [Google Scholar]
  • 89. Takebayashi K, Matsumoto S, Wakabayashi S, et al. The effect of low‐dose atorvastatin on circulating monocyte chemoattractant protein‐1 in patients with type 2 diabetes complicated by hyperlipidemia. Metabolism. 2005;54(9):1225‐1229. [DOI] [PubMed] [Google Scholar]
  • 90. Mazaherioun M, Djalali M, Koohdani F, et al. Beneficial effects of n‐3 fatty acids on cardiometabolic and inflammatory markers in type 2 diabetes mellitus: a clinical trial. Med Princ Pract. 2017;26(6):535‐541. 10.1159/000484089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Zabetian‐Targhi F, Mahmoudi MJ, Rezaei N, Mahmoudi M. Retinol binding protein 4 in relation to diet, inflammation, immunity, and cardiovascular diseases. Adv Nutr. 2015;6(6):748‐762. 10.3945/an.115.008292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Kotnik P, Fischer‐Posovszky P, Wabitsch M. RBP4: a controversial adipokine. Eur J Endocrinol. 2011;165(5):703‐711. 10.1530/EJE-11-0431. [DOI] [PubMed] [Google Scholar]
  • 93. Janke J, Engeli S, Boschmann M, et al. Retinol‐binding protein 4 in human obesity. Diabetes. 2006;55(10):2805‐2810. [DOI] [PubMed] [Google Scholar]
  • 94. Polonsky KS. Retinol‐binding protein 4, insulin resistance, and type 2 diabetes. N Engl J Med. 2006;354(24):2596‐2598. [DOI] [PubMed] [Google Scholar]
  • 95. Moraes‐Vieira PM, Yore MM, Dwyer PM, Syed I, Aryal P, Kahn BB. RBP4 activates antigen‐presenting cells, leading to adipose tissue inflammation and systemic insulin resistance. Cell Metab. 2014;19(3):512‐526. 10.1016/j.cmet.2014.01.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Pu L, Cheng J, Wu G, et al. Effects of retinol binding protein 4 knockdown on the PI3K/akt pathways in porcine adipocytes. Sheng Wu Gong Cheng Xue Bao. 2013;29(4):447‐457. [PubMed] [Google Scholar]
  • 97. Rabe K, Lehrke M, Parhofer KG, Broedl UC. Adipokines and insulin resistance. Mol Med. 2008;14(11‐12):741‐751. 10.2119/2008-00058.Rabe. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Takebayashi K, Suetsugu M, Matsumoto S, Aso Y, Inukai T. Effects of rosuvastatin and colestimide on metabolic parameters and urinary monocyte chemoattractant protein‐1 in type 2 diabetic patients with hyperlipidemia. South Med J. 2009;102(4):361‐368. 10.1097/SMJ.0b013e31819bd023. [DOI] [PubMed] [Google Scholar]
  • 99. Preitner F, Mody N, Graham TE, Peroni OD, Kahn BB. Long‐term fenretinide treatment prevents high‐fat diet‐induced obesity, insulin resistance, and hepatic steatosis. Am J Physiol Endocrinol Metab. 2009;297(6):E1420‐E1429. 10.1152/ajpendo.00362.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Mcilroy GD, Delibegovic M, Owen C, et al. Fenretinide treatment prevents diet‐induced obesity in association with major alterations in retinoid homeostatic gene expression in adipose, liver, and hypothalamus. Diabetes. 2013;62(3):825‐836. 10.2337/db12-0458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Cooper JP, Reynolds CP, Cho H, Kang MH. Clinical development of fenretinide as an antineoplastic drug: pharmacology perspectives. Exp Biol Med (Maywood). 2017;242(11):1178‐1184. 10.1177/1535370217706952. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Dobri N, Qin Q, Kong J, et al. A1120, a nonretinoid RBP4 antagonist, inhibits formation of cytotoxic bisretinoids in the animal model of enhanced retinal lipofuscinogenesis. Invest Ophthalmol Vis Sci. 2013;54(1):85‐95. 10.1167/iovs.12-10050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Herova M, Schmid M, Gemperle C, Hersberger M. ChemR23, the receptor for chemerin and resolvin E1, is expressed and functional on M1 but not on M2 macrophages. J Immunol. 2015;194(5):2330‐2337. 10.4049/jimmunol.1402166. [DOI] [PubMed] [Google Scholar]
  • 104. Fatima SS, Butt Z, Bader N, Pathan AZ, Hussain S, Iqbal NT. Role of multifunctional chemerin in obesity and preclinical diabetes. Obes Res Clin Pract. 2015;9(5):507‐512. 10.1016/j.orcp.2015.01.004. [DOI] [PubMed] [Google Scholar]
  • 105. Dogan SB, Balli U, Dede FO, Sertoglu E, Tazegul K. Chemerin as a novel crevicular fluid marker of patients with periodontitis and type 2 diabetes mellitus. J Periodontol. 2016;87(8):923‐933. 10.1902/jop.2016.150657. [DOI] [PubMed] [Google Scholar]
  • 106. Neves KB, Nguyen Dinh Cat A, Alves‐Lopes R, et al. Chemerin receptor blockade improves vascular function in diabetic obese mice via redox‐sensitive and akt‐dependent pathways. Am J Physiol Heart Circ Physiol. 2018;315(6):H1851‐H1860. 10.1152/ajpheart.00285.2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Wargent ET, Zaibi MS, O'Dowd JF, et al. Evidence from studies in rodents and in isolated adipocytes that agonists of the chemerin receptor CMKLR1 may be beneficial in the treatment of type 2 diabetes. PeerJ. 2015;3:e753 10.7717/peerj.753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Kim J, Lee SK, Jang YJ, et al. Enhanced ANGPTL2 expression in adipose tissues and its association with insulin resistance in obese women. Sci Rep. 2018;8(1):13976 10.1038/s41598-018-32419-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Zheng J, Zou J, Wang W, et al. Tumor necrosis factor‐α increases angiopoietin‐like protein 2 gene expression by activating Foxo1 in 3T3‐L1 adipocytes. Mol Cell Endocrinol. 2011;339(1):120‐129. 10.1016/j.mce.2011.04.002. [DOI] [PubMed] [Google Scholar]
  • 110. Sasaki Y, Ohta M, Desai D, et al. Angiopoietin like protein 2 (ANGPTL2) promotes adipose tissue macrophage and T lymphocyte accumulation and leads to insulin resistance. PLoS One. 2015;10(7):e0131176 10.1371/journal.pone.0131176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Martel C, Pincon A, Belanger AM, et al. Knockdown of angiopoietin‐like 2 mimics the benefits of intermittent fasting on insulin responsiveness and weight loss. Exp Biol Med (Maywood). 2018;243(1):45‐49. 10.1177/1535370217745505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Kitazawa M, Nagano M, Masumoto KH, Shigeyoshi Y, Natsume T, Hashimoto S. Angiopoietin‐like 2, a circadian gene, improves type 2 diabetes through potentiation of insulin sensitivity in mice adipocytes. Endocrinology. 2011;152(7):2558‐2567. 10.1210/en.2010-1407. [DOI] [PubMed] [Google Scholar]
  • 113. Thorin‐Trescases N, Thorin E. Angiopoietin‐like‐2: a multifaceted protein with physiological and pathophysiological properties. Expert Rev Mol Med. 2014;16:e17 10.1017/erm.2014.19. [DOI] [PubMed] [Google Scholar]
  • 114. Ferrand N, Bereziat V, Moldes M, Zaoui M, Larsen AK, Sabbah M. WISP1/CCN4 inhibits adipocyte differentiation through repression of PPARgamma activity. Sci Rep. 2017;7(1):1749 10.1038/s41598-017-01866-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Murahovschi V, Pivovarova O, Ilkavets I, et al. WISP1 is a novel adipokine linked to inflammation in obesity. Diabetes. 2015;64(3):856‐866. 10.2337/db14-0444. [DOI] [PubMed] [Google Scholar]
  • 116. Barchetta I, Cimini FA, Capoccia D, et al. WISP1 is a marker of systemic and adipose tissue inflammation in dysmetabolic subjects with or without type 2 diabetes. J Endocr Soc. 2017;1(6):660‐670. 10.1210/js.2017-00108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Horbelt T, Tacke C, Markova M, et al. The novel adipokine WISP1 associates with insulin resistance and impairs insulin action in human myotubes and mouse hepatocytes. Diabetologia. 2018;61(9):2054‐2065. 10.1007/s00125-018-4636-9. [DOI] [PubMed] [Google Scholar]
  • 118. Korolczuk A, Beltowski J. Progranulin, a new adipokine at the crossroads of metabolic syndrome, diabetes, dyslipidemia and hypertension. Curr Pharm Des. 2017;23(10):1533‐1539. 10.2174/1381612823666170124114524. [DOI] [PubMed] [Google Scholar]
  • 119. Nicoletto BB, Canani LH. The role of progranulin in diabetes and kidney disease. Diabetol Metab Syndr. 2015;7:117 10.1186/s13098-015-0112-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. Guo Q, Xu L, Li H, et al. Progranulin causes adipose insulin resistance via increased autophagy resulting from activated oxidative stress and endoplasmic reticulum stress. Lipids Health Dis. 2017;16(1):25 10.1186/s12944-017-0425-6. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 121. Qu H, Deng H, Hu Z. Plasma progranulin concentrations are increased in patients with type 2 diabetes and obesity and correlated with insulin resistance. Mediators Inflamm. 2013;2013:360190 10.1155/2013/360190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Law IK, Xu A, Lam KS, et al. Lipocalin‐2 deficiency attenuates insulin resistance associated with aging and obesity. Diabetes. 2010;59(4):872‐882. 10.2337/db09-1541. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Wang Y, Lam KS, Kraegen EW, et al. Lipocalin‐2 is an inflammatory marker closely associated with obesity, insulin resistance, and hyperglycemia in humans. Clin Chem. 2007;53(1):34‐41. [DOI] [PubMed] [Google Scholar]
  • 124. De la Chesnaye E, Manuel‐Apolinar L, Zarate A, et al. Lipocalin‐2 plasmatic levels are reduced in patients with long‐term type 2 diabetes mellitus. Int J Clin Exp Med. 2015;8(2):2853‐2859. [PMC free article] [PubMed] [Google Scholar]
  • 125. Yan QW, Yang Q, Mody N, et al. The adipokine lipocalin 2 is regulated by obesity and promotes insulin resistance. Diabetes. 2007;56(10):2533‐2540. [DOI] [PubMed] [Google Scholar]
  • 126. Leng X, Ding T, Lin H, et al. Inhibition of lipocalin 2 impairs breast tumorigenesis and metastasis. Cancer Res. 2009;69(22):8579‐8584. 10.1158/0008-5472.CAN-09-1934. [DOI] [PubMed] [Google Scholar]
  • 127. Adeghate E. Visfatin: structure, function and relation to diabetes mellitus and other dysfunctions. Curr Med Chem. 2008;15(18):1851‐1862. [DOI] [PubMed] [Google Scholar]
  • 128. Saddi‐Rosa P, Oliveira CS, Giuffrida FM, Reis AF. Visfatin, glucose metabolism and vascular disease: a review of evidence. Diabetol Metab Syndr. 2010;2:21 10.1186/1758-5996-2-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Jimenez‐Gonzalez M, Jaques F, Rodriguez S, et al. Cardiotrophin 1 protects beta cells from apoptosis and prevents streptozotocin‐induced diabetes in a mouse model. Diabetologia. 2013;56(4):838‐846. 10.1007/s00125-012-2822-8. [DOI] [PubMed] [Google Scholar]
  • 130. Gamella‐Pozuelo L, Fuentes‐Calvo I, Gomez‐Marcos MA, et al. Plasma cardiotrophin‐1 as a marker of hypertension and diabetes‐induced target organ damage and cardiovascular risk. Medicine (Baltimore). 2015;94(30):e1218 10.1097/MD.0000000000001218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Moreno‐Aliaga MJ, Perez‐Echarri N, Marcos‐Gomez B, et al. Cardiotrophin‐1 is a key regulator of glucose and lipid metabolism. Cell Metab. 2011;14(2):242‐253. 10.1016/j.cmet.2011.05.013. [DOI] [PubMed] [Google Scholar]
  • 132. Yarmolinsky J, Bordin Barbieri N, Weinmann T, Ziegelmann PK, Duncan BB, Ines SM. Plasminogen activator inhibitor‐1 and type 2 diabetes: a systematic review and meta‐analysis of observational studies. Sci Rep. 2016;6:17714 10.1038/srep17714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133. Ma X, Yan W, Zheng H, et al. Regulation of IL‐10 and IL‐12 production and function in macrophages and dendritic cells. F1000Res. 2015;4:1‐13. 10.12688/f1000research.7010.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. Iyer SS, Cheng G. Role of interleukin 10 transcriptional regulation in inflammation and autoimmune disease. Crit Rev Immunol. 2012;32(1):23‐63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Yaghini N, Mahmoodi M, Asadikaram GR, Hassanshahi GH, Khoramdelazad H, Kazemi AM. Serum levels of interleukin 10 (IL‐10) in patients with type 2 diabetes. Iran Red Crescent Med J. 2011;13(10):752. [PMC free article] [PubMed] [Google Scholar]
  • 136. Esposito K, Pontillo A, Giugliano F, et al. Association of low interleukin‐10 levels with the metabolic syndrome in obese women. J Clin Endocrinol Metab. 2003;88(3):1055‐1058. 10.1210/jc.2002-021437. [DOI] [PubMed] [Google Scholar]
  • 137. Phillips JM, Parish NM, Drage M, Cooke A. Cutting edge: interactions through the IL‐10 receptor regulate autoimmune diabetes. J Immunol. 2001;167(11):6087‐6091. [DOI] [PubMed] [Google Scholar]
  • 138. Pennline KJ, Roque‐Gaffney E, Monahan M. Recombinant human IL‐10 prevents the onset of diabetes in the nonobese diabetic mouse. Clin Immunol Immunopathol. 1994;71(2):169‐175. 10.1006/clin.1994.1068. [DOI] [PubMed] [Google Scholar]
  • 139. Lee M, Park H, Youn J, et al. Interleukin‐10 plasmid construction and delivery for the prevention of type 1 diabetes. Ann N Y Acad Sci. 2006;1079:313‐319. [DOI] [PubMed] [Google Scholar]
  • 140. Mandke R, Singh J. Cationic nanomicelles for delivery of plasmids encoding interleukin‐4 and interleukin‐10 for prevention of autoimmune diabetes in mice. Pharm Res. 2012;29(3):883‐897. 10.1007/s11095-011-0616-1. [DOI] [PubMed] [Google Scholar]
  • 141. Kapturczak MH, Wasserfall CH, Scott Loiler S, et al. Systemic gene therapy with interleukin‐10 (IL‐10) for prevention of type 1 diabetes in NOD mice: comparison of mutant (187A substitution) to recombinant murine IL‐10. Mol Ther. 2003;7(5):S80. [Google Scholar]
  • 142. Zhang L, Chen Y, Li C, Lin X, Cheng X, Li T. Protective effects of combined intervention with adenovirus vector mediated IL‐10 and IGF‐1 genes on endogenous islet beta cells in nonobese diabetes mice with onset of type 1 diabetes mellitus. PLoS One. 2014;9(3):e92616 10.1371/journal.pone.0092616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143. Zheng XX, Steele AW, Hancock WW, et al. A noncytolytic IL‐10/fc fusion protein prevents diabetes, blocks autoimmunity, and promotes suppressor phenomena in NOD mice. J Immunol. 1997;158(9):4507‐4513. [PubMed] [Google Scholar]
  • 144. Hong EG, Ko HJ, Cho YR, et al. Interleukin‐10 prevents diet‐induced insulin resistance by attenuating macrophage and cytokine response in skeletal muscle. Diabetes. 2009;58(11):2525‐2535. 10.2337/db08-1261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145. Dagdeviren S, Jung DY, Lee E, et al. Altered interleukin‐10 signaling in skeletal muscle regulates obesity‐mediated inflammation and insulin resistance. Mol Cell Biol. 2016;36(23):2956‐2966. 10.1128/MCB.00181-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146. Barry JC, Shakibakho S, Durrer C, et al. Hyporesponsiveness to the anti‐inflammatory action of interleukin‐10 in type 2 diabetes. Sci Rep. 2016;6:21244 10.1038/srep21244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147. Genetic Engineering and Biotechnology News . Renovo stock plummets 74% as lead scar reducing candidate fails in EU phase III trial. https://www.genengnews.com/topics/drug-discovery/renovo-stock-plummets-74-as-lead-scar-reducing-candidate-fails-in-eu-phase-iii-trial/. Updated 2011. Accessed May 14, 2019.
  • 148. Hecht RJ, Naing A, Falchook GS, et al. Overall survival of PEGylated human IL‐10 (AM0010) with 5‐FU/LV oxaliplatin (FOLFOX) in metastatic pancreatic adenocarcinoma (PDAC). J Clin Oncol. 2018;36(4):374. [Google Scholar]
  • 149. Helwick C. Small study evaluates novel immunotherapeutic in metastatic pancreatic cancer. https://www.ascopost.com/issues/march-10-2018/small-study-evaluates-novel-immunotherapeutic-in-metastatic-pancreatic-cancer/. Updated 2018. Accessed May 14, 2019.
  • 150. Shen J, Fang Y, Zhu H, Ge W. Plasma interleukin‐22 levels are associated with prediabetes and type 2 diabetes in the Han Chinese population. J Diabetes Investig. 2018;9(1):33‐38. 10.1111/jdi.12640. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151. Wang X, Ota N, Manzanillo P, et al. Interleukin‐22 alleviates metabolic disorders and restores mucosal immunity in diabetes. Nature. 2014;514(7521):237‐241. 10.1038/nature13564. [DOI] [PubMed] [Google Scholar]
  • 152. Avitabile S, Odorisio T, Madonna S, et al. Interleukin‐22 promotes wound repair in diabetes by improving keratinocyte pro‐healing functions. J Invest Dermatol. 2015;135(11):2862‐2870. 10.1038/jid.2015.278. [DOI] [PubMed] [Google Scholar]
  • 153. Hasnain SZ, Borg DJ, Harcourt BE, et al. Glycemic control in diabetes is restored by therapeutic manipulation of cytokines that regulate beta cell stress. Nat Med. 2014;20(12):1417‐1426. 10.1038/nm.3705. [DOI] [PubMed] [Google Scholar]
  • 154. Gong F, Wu J, Zhou P, et al. Interleukin‐22 might act as a double‐edged sword in type 2 diabetes and coronary artery disease. Mediators Inflamm. 2016;2016:8254797 10.1155/2016/8254797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155. Herder C, Kannenberg JM, Carstensen‐Kirberg M, et al. Serum levels of interleukin‐22, cardiometabolic risk factors and incident type 2 diabetes: KORA F4/FF4 study. Cardiovasc Diabetol. 2017;16(1):17 10.1186/s12933-017-0498-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156. Dalmas E, Venteclef N, Caer C, et al. T cell‐derived IL‐22 amplifies IL‐1beta‐driven inflammation in human adipose tissue: relevance to obesity and type 2 diabetes. Diabetes. 2014;63(6):1966‐1977. 10.2337/db13-1511. [DOI] [PubMed] [Google Scholar]
  • 157. Kwon H, Laurent S, Tang Y, Zong H, Vemulapalli P, Pessin J. Adipocyte‐specific IKKβ signaling suppresses adipose tissue inflammation through an IL‐13‐dependent paracrine feedback pathway. Cell Rep. 2014;9(5):1574‐1583. 10.1016/j.celrep.2014.10.068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158. Martinez‐Reyes CP, Gomez‐Arauz AY, Torres‐Castro I, et al. Serum levels of interleukin‐13 increase in subjects with insulin resistance but do not correlate with markers of low‐grade systemic inflammation. J Diabetes Res. 2018;2018:7209872 10.1155/2018/7209872. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159. Darkhal P, Gao M, Ma Y, Liu D. Blocking high‐fat diet‐induced obesity, insulin resistance and fatty liver by overexpression of il‐13 gene in mice. Int J Obes (Lond). 2015;39(8):1292‐1299. 10.1038/ijo.2015.52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160. Ballak DB, van Diepen JA, Moschen AR, et al. IL‐37 protects against obesity‐induced inflammation and insulin resistance. Nat Commun. 2014;5:4711 10.1038/ncomms5711. [DOI] [PubMed] [Google Scholar]
  • 161. Jia H, Liu J, Han B. Reviews of interleukin‐37: functions, receptors, and roles in diseases. Biomed Res Int. 2018;2018:3058640 10.1155/2018/3058640. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162. Long SA, Buckner JH, Greenbaum CJ. IL‐2 therapy in type 1 diabetes: “trials” and tribulations. Clin Immunol. 2013;149(3, Part A):324‐331. 10.1016/j.clim.2013.02.005. [DOI] [PubMed] [Google Scholar]
  • 163. Rosenzwajg M, Churlaud G, Hartemann A, Klatzmann D. Interleukin 2 in the pathogenesis and therapy of type 1 diabetes. Curr Diab Rep. 2014;14(12):553 10.1007/s11892-014-0553-6. [DOI] [PubMed] [Google Scholar]
  • 164. Mi QS, Ly D, Zucker P, McGarry M, Delovitch TL. Interleukin‐4 but not interleukin‐10 protects against spontaneous and recurrent type 1 diabetes by activated CD1d‐restricted invariant natural killer T‐cells. Diabetes. 2004;53(5):1303‐1310. [DOI] [PubMed] [Google Scholar]
  • 165. Cameron MJ, Arreaza GA, Zucker P, et al. IL‐4 prevents insulitis and insulin‐dependent diabetes mellitus in nonobese diabetic mice by potentiation of regulatory T helper‐2 cell function. J Immunol. 1997;159(10):4686‐4692. [PubMed] [Google Scholar]
  • 166. Borst SE. The role of TNF‐alpha in insulin resistance. Endocrine. 2004;23(2‐3):177‐182. [DOI] [PubMed] [Google Scholar]
  • 167. Nieto‐Vazquez I, Fernandez‐Veledo S, Kramer DK, Vila‐Bedmar R, Garcia‐Guerra L, Lorenzo M. Insulin resistance associated to obesity: the link TNF‐alpha. Arch Physiol Biochem. 2008;114(3):183‐194. 10.1080/13813450802181047. [DOI] [PubMed] [Google Scholar]
  • 168. Hotamisligil GS. Mechanisms of TNF‐alpha‐induced insulin resistance. Exp Clin Endocrinol Diabetes. 1999;107(2):119‐125. 10.1055/s-0029-1212086. [DOI] [PubMed] [Google Scholar]
  • 169. Moller DE. Potential role of TNF‐alpha in the pathogenesis of insulin resistance and type 2 diabetes. Trends Endocrinol Metab. 2000;11(6):212‐217. [DOI] [PubMed] [Google Scholar]
  • 170. Quinn CE, Hamilton PK, Lockhart CJ, McVeigh GE. Thiazolidinediones: effects on insulin resistance and the cardiovascular system. Br J Pharmacol. 2008;153(4):636‐645. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171. Sayo Y, Dobashi H, Matubara S, Tada S, Taminato T, Ishida T. Infliximab (anti‐TNF‐alpha antibody) improved insulin resistance in rheumatoid arthritis patients. Diabetes. 2005;54(1):A149. [Google Scholar]
  • 172. Gonzalez‐Gay MA, De Matias JM, Gonzalez‐Juanatey C, et al. Anti‐tumor necrosis factor‐alpha blockade improves insulin resistance in patients with rheumatoid arthritis. Clin Exp Rheumatol. 2006;24(1):83‐86. [PubMed] [Google Scholar]
  • 173. Stanley TL, Zanni MV, Johnsen S, et al. TNF‐alpha antagonism with etanercept decreases glucose and increases the proportion of high molecular weight adiponectin in obese subjects with features of the metabolic syndrome. J Clin Endocrinol Metab. 2011;96(1):E146‐E150. 10.1210/jc.2010-1170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174. Mastrandrea L, Yu J, Behrens T, et al. Etanercept treatment in children with new‐onset type 1 diabetes: pilot randomized, placebo‐controlled, double‐blind study. Diabetes Care. 2009;32(7):1244‐1249. 10.2337/dc09-0054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175. Martinez‐Abundis E, Reynoso‐von Drateln C, Hernandez‐Salazar E, Gonzalez‐Ortiz M. Effect of etanercept on insulin secretion and insulin sensitivity in a randomized trial with psoriatic patients at risk for developing type 2 diabetes mellitus. Arch Dermatol Res. 2007;299(9):461‐465. 10.1007/s00403-007-0784-3. [DOI] [PubMed] [Google Scholar]
  • 176. Rosenvinge A, Krogh‐Madsen R, Baslund B, Pedersen BK. Insulin resistance in patients with rheumatoid arthritis: effect of anti‐TNFalpha therapy. Scand J Rheumatol. 2007;36(2):91‐96. [DOI] [PubMed] [Google Scholar]
  • 177. Richmond V, Michelini FM, Bueno CA, Alche LE, Ramirez JA. Small molecules as anti‐TNF drugs. Curr Med Chem. 2015;22(25):2920‐2942. [DOI] [PubMed] [Google Scholar]
  • 178. He MM, Smith AS, Oslob JD, et al. Small‐molecule inhibition of TNF‐alpha. Science. 2005;310(5750):1022‐1025. [DOI] [PubMed] [Google Scholar]
  • 179. Gupta‐Ganguli M, Cox K, Means B, Gerling I, Solomon SS. Does therapy with anti‐TNF‐alpha improve glucose tolerance and control in patients with type 2 diabetes? Diabetes Care. 2011;34(7):e121 10.2337/dc10-1334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180. Liang H, Yin B, Zhang H, et al. Blockade of tumor necrosis factor (TNF) receptor type 1‐mediated TNF‐alpha signaling protected wistar rats from diet‐induced obesity and insulin resistance. Endocrinology. 2008;149(6):2943‐2951. 10.1210/en.2007-0978. [DOI] [PubMed] [Google Scholar]
  • 181. Aouadi M, Tencerova M, Vangala P, et al. Gene silencing in adipose tissue macrophages regulates whole‐body metabolism in obese mice. Proc Natl Acad Sci U S A. 2013;110(20):8278‐8283. 10.1073/pnas.1300492110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182. Peraldi P, Xu M, Spiegelman BM. Thiazolidinediones block tumor necrosis factor‐alpha‐induced inhibition of insulin signaling. J Clin Invest. 1997;100(7):1863‐1869. 10.1172/JCI119715. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183. Hyun B, Shin S, Lee A, et al. Metformin down‐regulates TNF‐alpha secretion via suppression of scavenger receptors in macrophages. Immune Netw. 2013;13(4):123‐132. 10.4110/in.2013.13.4.123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184. Goldfine AB, Fonseca V, Jablonski KA, et al.; Targeting Inflammation Using Salsalate in Type 2 Diabetes Study Team .Salicylate (salsalate) in patients with type 2 diabetes: a randomized trial. Ann Intern Med. 2013;159(1):1‐12. 10.7326/0003-4819-159-1-201307020-00003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185. Neumiller JJ. Aspirin therapy in patients with diabetes: an update on current recommendations. Diabetes Spectrum. 2013;26(3):153‐155. [Google Scholar]
  • 186. Banerjee M, Saxena M. Interleukin‐1 (IL‐1) family of cytokines: role in type 2 diabetes. Clin Chim Acta. 2012;413(15):1163‐1170. 10.1016/j.cca.2012.03.021. [DOI] [PubMed] [Google Scholar]
  • 187. Ballak DB, Stienstra R, Tack CJ, Dinarello CA, van Diepen JA. IL‐1 family members in the pathogenesis and treatment of metabolic disease: focus on adipose tissue inflammation and insulin resistance. Cytokine. 2015;75(2):280‐290. 10.1016/j.cyto.2015.05.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188. Yang Y, Wang H, Kouadir M, Song H, Shi F. Recent advances in the mechanisms of NLRP3 inflammasome activation and its inhibitors. Cell Death Dis. 2019;10(2):128 10.1038/s41419-019-1413-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189. Dixit VD. Nlrp3 inflammasome activation in type 2 diabetes: is it clinically relevant? Diabetes. 2013;62(1):22‐24. 10.2337/db12-1115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190. Vandanmagsar B, Youm YH, Ravussin A, et al. The NLRP3 inflammasome instigates obesity‐induced inflammation and insulin resistance. Nat Med. 2011;17(2):179‐188. 10.1038/nm.2279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191. Zhao G, Dharmadhikari G, Maedler K, Meyer‐Hermann M. Possible role of interleukin‐1beta in type 2 diabetes onset and implications for anti‐inflammatory therapy strategies. PLoS Comput Biol. 2014;10(8):e1003798 10.1371/journal.pcbi.1003798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192. Dinarello CA, Donath MY, Mandrup‐Poulsen T. Role of IL‐1beta in type 2 diabetes. Curr Opin Endocrinol Diabetes Obes. 2010;17(4):314‐321. 10.1097/MED.0b013e32833bf6dc. [DOI] [PubMed] [Google Scholar]
  • 193. Ruscitti P, Cipriani P, Cantarini L, et al. Efficacy of inhibition of IL‐1 in patients with rheumatoid arthritis and type 2 diabetes mellitus: two case reports and review of the literature. J Med Case Reports. 2015;9:123 10.1186/s13256-015-0603-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194. Vitale A, Cantarini L, Rigante D, Bardelli M, Galeazzi M. Anakinra treatment in patients with gout and type 2 diabetes. Clin Rheumatol. 2015;34(5):981‐984. 10.1007/s10067-014-2601-7. [DOI] [PubMed] [Google Scholar]
  • 195. van Asseldonk EJ, van Poppel PC, Ballak DB, Stienstra R, Netea MG, Tack CJ. One week treatment with the IL‐1 receptor antagonist anakinra leads to a sustained improvement in insulin sensitivity in insulin resistant patients with type 1 diabetes mellitus. Clin Immunol. 2015;160(2):155‐162. 10.1016/j.clim.2015.06.003. [DOI] [PubMed] [Google Scholar]
  • 196. Dinarello CA, Simon A, van der Meer JW. Treating inflammation by blocking interleukin‐1 in a broad spectrum of diseases. Nat Rev Drug Discov. 2012;11(8):633‐652. 10.1038/nrd3800. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197. Everett BM, Donath MY, Pradhan AD, et al. Anti‐inflammatory therapy with canakinumab for the prevention and management of diabetes. J Am Coll Cardiol. 2018;71(21):2392‐2401. [DOI] [PubMed] [Google Scholar]
  • 198. Hensen J, Howard CP, Walter V, Thuren T. Impact of interleukin‐1beta antibody (canakinumab) on glycaemic indicators in patients with type 2 diabetes mellitus: results of secondary endpoints from a randomized, placebo‐controlled trial. Diabetes Metab. 2013;39(6):524‐531. 10.1016/j.diabet.2013.07.003. [DOI] [PubMed] [Google Scholar]
  • 199. White PC, Adhikari S, Grishman EK, Sumpter KM. A phase I study of anti‐inflammatory therapy with rilonacept in adolescents and adults with type 1 diabetes mellitus. Pediatr Diabetes. 2018;19(4):788‐793. 10.1111/pedi.12634. [DOI] [PubMed] [Google Scholar]
  • 200. Cavelti‐Weder C, Babians‐Brunner A, Keller C, et al. Effects of gevokizumab on glycemia and inflammatory markers in type 2 diabetes. Diabetes Care. 2012;35(8):1654‐1662. 10.2337/dc11-2219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201. Sloan‐Lancaster J, Abu‐Raddad E, Polzer J, et al. Double‐blind, randomized study evaluating the glycemic and anti‐inflammatory effects of subcutaneous LY2189102, a neutralizing IL‐1beta antibody, in patients with type 2 diabetes. Diabetes Care. 2013;36(8):2239‐2246. 10.2337/dc12-1835. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202. Owyang AM, Maedler K, Gross L, et al. XOMA 052, an anti‐IL‐1{beta} monoclonal antibody, improves glucose control and {beta}‐cell function in the diet‐induced obesity mouse model. Endocrinology. 2010;151(6):2515‐2527. 10.1210/en.2009-1124. [DOI] [PubMed] [Google Scholar]
  • 203. Zha J, Chi XW, Yu XL, et al. Interleukin‐1beta‐targeted vaccine improves glucose control and beta‐cell function in a diabetic KK‐ay mouse model. PLoS One. 2016;11(5):e0154298 10.1371/journal.pone.0154298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204. Cucak H, Hansen G, Vrang N, Skarsfeldt T, Steiness E, Jelsing J. The IL‐1beta receptor antagonist SER140 postpones the onset of diabetes in female nonobese diabetic mice. J Diabetes Res. 2016;2016:7484601 10.1155/2016/7484601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205. Kristiansen OP, Mandrup‐Poulsen T. Interleukin‐6 and diabetes: the good, the bad, or the indifferent? Diabetes. 2005;54(suppl 2):S114‐S124. [DOI] [PubMed] [Google Scholar]
  • 206. Otsuka Y, Kiyohara C, Kashiwado Y, et al. Effects of tumor necrosis factor inhibitors and tocilizumab on the glycosylated hemoglobin levels in patients with rheumatoid arthritis; an observational study. PLoS One. 2018;13(4):e0196368 10.1371/journal.pone.0196368. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207. Troseid M, Seljeflot I, Arnesen H. The role of interleukin‐18 in the metabolic syndrome. Cardiovasc Diabetol. 2010;9:11 10.1186/1475-2840-9-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208. Netea MG, Joosten LA, Lewis E, et al. Deficiency of interleukin‐18 in mice leads to hyperphagia, obesity and insulin resistance. Nat Med. 2006;12(6):650‐656. [DOI] [PubMed] [Google Scholar]
  • 209. Moriwaki Y, Yamamoto T, Shibutani Y, et al. Elevated levels of interleukin‐18 and tumor necrosis factor‐alpha in serum of patients with type 2 diabetes mellitus: relationship with diabetic nephropathy. Metabolism. 2003;52(5):605‐608. 10.1053/meta.2003.50096. [DOI] [PubMed] [Google Scholar]
  • 210. Escobar‐Morreale HF, Botella‐Carretero JI, Villuendas G, Sancho J, San Millan JL. Serum interleukin‐18 concentrations are increased in the polycystic ovary syndrome: relationship to insulin resistance and to obesity. J Clin Endocrinol Metab. 2004;89(2):806‐811. 10.1210/jc.2003-031365. [DOI] [PubMed] [Google Scholar]
  • 211. Mishra M, Kumar H, Bajpai S, Singh RK, Tripathi K. Level of serum IL‐12 and its correlation with endothelial dysfunction, insulin resistance, proinflammatory cytokines and lipid profile in newly diagnosed type 2 diabetes. Diabetes Res Clin Pract. 2011;94(2):255‐261. 10.1016/j.diabres.2011.07.037. [DOI] [PubMed] [Google Scholar]
  • 212. Abdel‐Moneim A, Bakery HH, Allam G. The potential pathogenic role of IL‐17/Th17 cells in both type 1 and type 2 diabetes mellitus. Biomed Pharmacother. 2018;101:287‐292. 10.1016/j.biopha.2018.02.103. [DOI] [PubMed] [Google Scholar]
  • 213. Zuniga LA, Shen WJ, Joyce‐Shaikh B, et al. IL‐17 regulates adipogenesis, glucose homeostasis, and obesity. J Immunol. 2010;185(11):6947‐6959. 10.4049/jimmunol.1001269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214. Park HK, Kwak MK, Kim HJ, Ahima RS. Linking resistin, inflammation, and cardiometabolic diseases. Korean J Intern Med. 2017;32(2):239‐247. 10.3904/kjim.2016.229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215. Berger A. Resistin: a new hormone that links obesity with type 2 diabetes. BMJ. 2001;322(7280):193. [Google Scholar]
  • 216. Azab N, Abdel‐Aziz T, Ahmed A, El‐deen IM. Correlation of serum resistin level with insulin resistance and severity of retinopathy in type 2 diabetes mellitus. J Saudi Chem Soc. 2016;20(3):272‐277. 10.1016/j.jscs.2012.07.003. [DOI] [Google Scholar]
  • 217. Kusminski CM, McTernan PG, Kumar S. Role of resistin in obesity, insulin resistance and type II diabetes. Clin Sci (Lond). 2005;109(3):243‐256. [DOI] [PubMed] [Google Scholar]
  • 218. Benomar Y, Solomon G, Amine H, Othmane A, Gertler A, Taouis M. Novel human resistin antagonist (monomeric C6A mutant) reduced body weight gain and restored insulin responsiveness in mice fed high fat diet. Endocr Abstr. 2015;37:GP15.04. [Google Scholar]
  • 219. Gu N, Han SP, Fei L, et al. Resistin‐binding peptide antagonizes role of resistin on white adipose tissue. Acta Pharmacol Sin. 2007;28(2):221‐226. 10.1111/j.1745-7254.2007.00480.x. [DOI] [PubMed] [Google Scholar]
  • 220. Majuri A, Santaniemi M, Rautio K, et al. Rosiglitazone treatment increases plasma levels of adiponectin and decreases levels of resistin in overweight women with PCOS: a randomized placebo‐controlled study. Eur J Endocrinol. 2007;156(2):263‐269. [DOI] [PubMed] [Google Scholar]
  • 221. Wang CH, Wang PJ, Hsieh YC, et al. Resistin facilitates breast cancer progression via TLR4‐mediated induction of mesenchymal phenotypes and stemness properties. Oncogene. 2018;37(5):589‐600. 10.1038/onc.2017.357. [DOI] [PubMed] [Google Scholar]
  • 222. Lee S, Lee HC, Kwon YW, et al. Adenylyl cyclase‐associated protein 1 is a receptor for human resistin and mediates inflammatory actions of human monocytes. Cell Metab. 2014;19(3):484‐497. 10.1016/j.cmet.2014.01.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223. Surapaneni K, Koduganti RR, Ganapthi SN, et al. Efficacy of systemic administration of alpha lipoic acid and scaling and root planning in patients with chronic peridontitis and type 2 diabetes mellitus—a randomised controlled trial. J Clin Diagn Res. 2018;12(4):1‐3. [Google Scholar]
  • 224. Chen S, Jim B, Ziyadeh FN. Diabetic nephropathy and transforming growth factor‐beta: transforming our view of glomerulosclerosis and fibrosis build‐up. Semin Nephrol. 2003;23(6):532‐543. [DOI] [PubMed] [Google Scholar]
  • 225. Chang AS, Hathaway CK, Smithies O, Kakoki M. Transforming growth factor‐beta1 and diabetic nephropathy. Am J Physiol Renal Physiol. 2016;310(8):F689‐F696. 10.1152/ajprenal.00502.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226. Pantsulaia T. Role of TGF‐beta in pathogenesis of diabetic nephropathy. Georgian Med News. 2006;(131):13‐18. [PubMed] [Google Scholar]
  • 227. Gomes KB, Karina B, Rodrigues KF, Fernandes AP. The role of transforming growth factor‐beta in diabetic nephropathy. Int J Med Genet. 2014;2014:180270. [Google Scholar]
  • 228. Mou X, Zhou DY, Zhou DY, et al. Serum TGF‐beta1 as a biomarker for type 2 diabetic nephropathy: a meta‐analysis of randomized controlled trials. PLoS One. 2016;11(2):e0149513 10.1371/journal.pone.0149513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229. Samad F, Yamamoto K, Pandey M, Loskutoff DJ. Elevated expression of transforming growth factor‐beta in adipose tissue from obese mice. Mol Med. 1997;3(1):37‐48. [PMC free article] [PubMed] [Google Scholar]
  • 230. Yadav H, Quijano C, Kamaraju AK, et al. Protection from obesity and diabetes by blockade of TGF‐beta/Smad3 signaling. Cell Metab. 2011;14(1):67‐79. 10.1016/j.cmet.2011.04.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231. Vincenti F, Fervenza FC, Campbell KN, et al.; Focal Segmental Glomerulosclerosis Study Group .A phase 2, double‐blind, placebo‐controlled, randomized study of fresolimumab in patients with steroid‐resistant primary focal segmental glomerulosclerosis. Kidney Int Rep. 2017;2(5):800‐810. 10.1016/j.ekir.2017.03.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232. Morris JC, Tan AR, Olencki TE, et al. Phase I study of GC1008 (fresolimumab): a human anti‐transforming growth factor‐beta (TGFbeta) monoclonal antibody in patients with advanced malignant melanoma or renal cell carcinoma. PLoS One. 2014;9(3):e90353 10.1371/journal.pone.0090353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233. Chavey C, Lazennec G, Lagarrigue S, et al. CXC ligand 5 is an adipose‐tissue derived factor that links obesity to insulin resistance. Cell Metab. 2009;9(4):339‐349. 10.1016/j.cmet.2009.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234. Chavey C, Fajas L. CXCL5 drives obesity to diabetes, and further. Aging (Albany, NY). 2009;1(7):674‐677. 10.18632/aging.100064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235. Nunemaker CS, Chung HG, Verrilli GM, Corbin KL, Upadhye A, Sharma PR. Increased serum CXCL1 and CXCL5 are linked to obesity, hyperglycemia, and impaired islet function. J Endocrinol. 2014;222(2):267‐276. 10.1530/JOE-14-0126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236. Kasama T, Ohtsuka K, Sato M, Takahashi R, Wakabayashi K, Kobayashi K. Macrophage migration inhibitory factor: a multifunctional cytokine in rheumatic diseases. Arthritis. 2010;2010:106202 10.1155/2010/106202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237. Morrison MC, Kleemann R. Role of macrophage migration inhibitory factor in obesity, insulin resistance, type 2 diabetes, and associated hepatic co‐morbidities: a comprehensive review of human and rodent studies. Front Immunol. 2015;6:308 10.3389/fimmu.2015.00308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238. Bilgir O, Gokcen B, Bilgir F, et al. Relationship between serum macrophage migration inhibitory factor level and insulin resistance, high‐sensitivity C‐reactive protein and visceral fat mass in prediabetes. Am J Med Sci. 2018;355(1):37‐43. [DOI] [PubMed] [Google Scholar]
  • 239. Kok T, Wasiel AA, Cool RH, Melgert BN, Poelarends GJ, Dekker FJ. Small‐molecule inhibitors of macrophage migration inhibitory factor (MIF) as an emerging class of therapeutics for immune disorders. Drug Discov Today. 2018;23(11):1910‐1918. 10.1016/j.drudis.2018.06.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240. Trivedi‐Parmar V, Jorgensen WL. Advances and insights for small molecule inhibition of macrophage migration inhibitory factor. J Med Chem. 2018;61(18):8104‐8119. 10.1021/acs.jmedchem.8b00589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241. Zhang Z, Zhang R, Li L, et al. Macrophage migration inhibitory factor (MIF) inhibitor, Z‐590 suppresses cartilage destruction in adjuvant‐induced arthritis via inhibition of macrophage inflammatory activation. Immunopharmacol Immunotoxicol. 2018;40(2):149‐157. 10.1080/08923973.2018.1424896. [DOI] [PubMed] [Google Scholar]
  • 242. Sanchez‐Zamora Y, Terrazas LI, Vilches‐Flores A, et al. Macrophage migration inhibitory factor is a therapeutic target in treatment of non‐insulin‐dependent diabetes mellitus. FASEB J. 2010;24(7):2583‐2590. 10.1096/fj.09-147066. [DOI] [PubMed] [Google Scholar]
  • 243. Wang Z, Wei M, Wang M, et al. Inhibition of macrophage migration inhibitory factor reduces diabetic nephropathy in type II diabetes mice. Inflammation. 2014;37(6):2020‐2029. 10.1007/s10753-014-9934-x. [DOI] [PubMed] [Google Scholar]
  • 244. Chan PC, Wu TN, Chen YC, et al. Targeted inhibition of CD74 attenuates adipose COX‐2‐MIF‐mediated M1 macrophage polarization and retards obesity‐related adipose tissue inflammation and insulin resistance. Clin Sci (Lond). 2018;132(14):1581‐1596. 10.1042/CS20180041. [DOI] [PubMed] [Google Scholar]

Articles from Bioengineering & Translational Medicine are provided here courtesy of Wiley

RESOURCES