Category: Children

Chronic hyperglycemia and inflammation

Chronic hyperglycemia and inflammation

Polyphenols can Detoxification Methods Explained found in a inflamkation of Chronic hyperglycemia and inflammation hypeglycemia spices, including berries, cinnamon, Thyroid Function Optimization chocolate, red onions and purple cabbage. Over time, chronic inflammation can cause permanent damage to cells and tissues. MCP-1 contributes to macrophage infiltration into adipose tissue, insulin resistance, and hepatic steatosis in obesity. Chronic hyperglycemia and inflammation

Video

Understanding the role of inflammation in type 2 diabetes

Chronic hyperglycemia and inflammation -

It not only prevents the metabolism of non-oxidative glucose [ 23 , 24 ], but also suppresses the lipoprotein lipase that consecutively increases the plasma levels of triglycerides [ 23 ].

Moreover, IL-6 also activates the suppressor of cytokine signaling SOCS proteins [ 6 , 25 ] which may block the cytokine-mediated transcriptional factor activation of insulin receptor [ 26 ].

Signal transducer and activator of transcription 5B STAT5B is a protein that belongs to the STAT family of transcription factors. STAT5B is aptly named for its unique ability to act as signal transducer and as transcription factor of insulin receptor [ 26 ].

In response to cytokines, STAT5B is phosphorylated by receptor associated kinases [ 27 ]. STAT5B activates insulin transcription factor through potentiating the tyrosine kinase by binding with phosphotyrosine of the insulin receptor. The activation of insulin transcription factor is blocked by SOCS proteins which suppresses the activity of tyrosine kinase by significantly competing with STAT5B [ 19 , 27 ].

SOCS proteins have negative effects on insulin action while IL-6 can activate these SOCS proteins. Therefore IL-6 is considered as an important biomarker for the development of IR [ 19 , 28 ].

Production of IL-6 is regulated by IL-1β via activation of interleukin-1 receptor type I IL-1RI [ 29 , 30 ]. Blocking the activity of IL-1RI with suitable anti-inflammatory agent like interleukin-1 receptor antagonist IL-1Ra antagonizes the agonistic effects of IL-1β that ultimately leads to the suppression of IL-6 production [ 4 , 31 ].

Anti-IL-6 receptor antibody and soluble receptor of IL-6 sIL-6R have proven to be effective by decreasing the development of IR [ 32 , 33 ], but this treatment strategy may not be very much effective as production of IL-6 is dependent on the activation of IL-1β and its role in the development of IR cannot be negelected.

Adipocytes secrete several pro-inflammatory mediators and among them, TNF-α has been proposed to develop a link between IR, obesity and T2DM [ 34 , 35 ]. Experimental studies conducted on obese animals indicate that the expression of TNF-α is increased in obese animals which modulates the insulin action [ 36 ].

TNF-α binds with its receptor and triggers a broad spectrum signaling cascade that results in the activation of various transcriptional pathways such as Nuclear factor kappa-B cells NF-κB and Jun NH2-terminal kinase JNK [ 37 , 38 ].

Once, NF-κB and JNK are activated, they phosphorylate serine in IRS-1 which result in the impairment of IR-mediated tyrosine phosphorylation of IRS-1 [ 37 ]. Recently, it has been found that serum level of TNF-α is positively correlated with the pathophysiology of IR [ 35 , 39 ] which exhibit that TNF-α is also a main causative factor that contributes the development of IR.

Therefore, it has been deliberated that adipose tissues are the major endocrine organ which have the ability to produce variety of adipose-derived mediators that are activitely involved to regulate the energy metabolism and insulin sensitivity [ 41 ].

The most important adipose-derived mediators are FFAs and adipokines. Adipokines include large number of pro-inflammatory mediators which include leptin, TNF-α, IL-6, tissue inhibitor of metalloproteinases TIMP-1 adiponectin, retinol-binding protein RBP-4 and monocyte chemotactic protein MCP-1 [ 42 , 43 ].

It has been evidenced from several experimental studies that there is a strong correlation between the mass of adipose tissues and development of IR Fig. IL-6, TNF-α, MCP-1, TIMP-1, RBP-4, and leptin are considered as pro-inflammatory cytokines which are responsible not only for the induction for local inflammation in adipocytes, but may also induce systemic inflammation after entering into the blood stream [ 4 , 47 , 48 ].

Adiponectin is the only adipokine that acts as anti-inflammatory cytokine and has the ability to ameliorate the deleterious effects of IL-6, TNF-α, MCP-1, TIMP-1, RBP-4, and leptin which are known to be produced in adipose tissues [ 11 ].

It has also been found that the level of adiponectin is downregulated in obesity and is positively associated with insulin sensitivity [ 49 , 50 ]. The imbalance between leptin and adiponectin may result in the development of systemic IR. Schematic representation of adipocytokines-induced IR. Glucolipotoxicity and induction of inflammation in adipocytes are responsible to make the adipocytes abnormal.

Once adipocytes are injured, glucose utilization is decreased in adipocytes and levels of FFAs are abnormally increased due to which metabolic stress in adipocytes is increased which ultimately leads to the abnormal secretion of various pro-inflammatory mediators and adipocytokines.

Chemokines are an important class of pro-inflammatory mediators. Their production is dependent on the activation IL-1β and various transcriptional pathways [ 4 ]. Up till now, various chemokines have been discovered, among which the most important are MCP-1, MCP-2, MCP-3, MCP-4, CCL2, MIP-1α and MIP-1β [ 51 ].

Several studies have reported that MCP-1 and CCL2 deficient mice prevented high fat diet-induced IR [ 52 , 53 ].

Moreover, overexpression of MCP-1 in adipose tissues was also observed to be responsible for the increase in adipose tissue macrophages and induction IR [ 52 , 54 ]. It has been found that chemokines play crucial role for the development of IR and T2DM Fig. Among various receptors for chemokines, CCR2 and CCR5 are the most important receptors that play decisive role in the pathogenesis of IR [ 56 ] in adipose tissues Fig.

It has been found that adipocytes secrete CCR2 in an inactive form. After activation, CCR2 induces the expression of various inflammatory genes and impaires the uptake of insulin-dependent glucose uptake.

Moreover, adipocytes can also secrete CCL2 and CCL3 which act as a potent signal for the recruitment of macrophages. The upregulation of CCL2 and CCL3 from adipocytes may contribute to the development of IR in adipose and peripheral tissues [ 57 ].

The above mentioned studies highlight the crucial role of various chemokines in the development of IR along with other pro-inflammatory mediators. Chemokines-induced IR. M2 macrophages in lean state, maintain the insulin sensitivity in adipose tissues whereas, due to overnutrition, adipose tissues initiates the secretion of MCP-1 which leads to the recruitment of circulating monocytes in adipocytes.

CCR2 macrophages are accumulated in obese adipocytes and presumably maintain the inflammation by recruiting M1 macrophages in obese adipocytes. While on the other side, CCR5-adipose tissue macrophages ATM also infiltrate from the obese adipocytes and promote the inflammatory responses by involving ATM recruitment and producing various pro-inflammatory mediators notably TNF-α, IL-6, and IL-1β in conjunction with other infiltrated immune cells and adipokines.

After production, these pro-inflammatory mediators induce IR in adipocytes and peripheral tissues through activation of several transcriptional pathways such as JNK and NF-κB. Adapted from Xu et al. C-C motif chemokine receptor 5 CCR5 promotes obesity-induced inflammation and IR.

Recently, it has been found that the expression of CCR5 and its ligand MCP-1 is significantly increased in white adipose tissues WAT and its accumulation is increased in adipose tissue macrophages ATM in WAT of obese mice and provides a novel link between inflammation and IR in adipocytes by stimulating the production of various pro-inflammatory cytokines and chemokines.

Adopted from Ota [ 51 ]. CRP has been considred as one of the most important human acute phase protein that correlates with development of IR [ 58 , 59 ].

CRP is a systemic inflammatory biomarker and has been considered as one of the major causative factor for the development of T2DM [ 60 ]. It has been evidenced that elevated levels of CRP not only reflect the induction of local inflammation, but also predict the pathogenesis of tissue-specific IR [ 61 ].

Overnutrition increases the cellular overload of glucose and FFAs which in turn increases the oxidative stress Fig. Peripheral and adipose tissues protect themselves from the damaging effects of oxidative stress producing resistance to the action of insulin by preventing the penetration of glucose and FFAs into the cells.

Oxidative stress is because of imbalance between the production of reactive oxygen species ROS and anti-oxidative defense mechanism against the production of ROS. β-cells of pancreatic islets, adipocytes and peripheral tissues are more vulnerable to the damaging effects of oxidative stress Fig. Several mechanisms are involved to influence the balance between ROS and anti-oxidant defense mechanisms including activation of stress-signaling pathways such as JNK pathway [ 64 ] and transcriptional mediated pathways such as NF-κB [ 65 ].

JNK and NF-κB pathways decrease the insulin-mediated glucose uptake by tissues and insuling signaling [ 66 — 68 ], that ultimately induces IR Fig.

Moreover, the activations of JNK and NF-κB pathways is also associated with the upregulation of various pro-inflammatory mediators such as TNF-α, IL-6, and CRP.

It has also been reported that oxidative stress-indcued activation of NF-κB pathway may also be associated with endothelial dysfunction that can lead to the induction of IR [ 69 , 70 ], but anti-oxidant therapy may act as a potential strategy to prevent the induction of IR-associated with endothelial dysfunction [ 71 ].

The growing body of evidence indicate that oxidative stress is a common pathogenic factor that leads to the development of tissues-specific IR. The results of experimental studies indicate that what happens in peripheral tissues also occur in the β-cells of pancreatic islets and endothelial cells to compensate the systemic oxidative stress.

Mechanism of oxidative stress-induced IR: Chronic exposure of hyperglycemia and hyperlipidemia due to over nutrition leads to the production of oxidative stress via activation of reactive oxygen species.

IKKβ also induces the activation of NF-κB. p38, JNK and IKKβ, further activates the serine phosphorylation of insulin receptor substrate-1 IRS While on the other side, NF-κB also activates the expression of iNOS which also induces the S-nitrosylation of IRS Both S-nitrosylation and serine phosphorylation of IRS-1 suppress the tyrosine phosphorylation of insulin signaling pathways which ultimately results into the induction of IR in liver, adipocytes and skeletal muscles.

Impact of oxidative stress on vital organs of the body. β-cells of pancreatic islets, adipocytes and peripheral tissues are more susceptible to the damaging effects of oxidative stress. Oxidative stress independently exhibit its hazardous effects on these organs due to which impaired insulin secretion occurs in β-cells of pancreatic islets and IR develops in adipocytes and peripheral tissues.

Impaired insulin secretion and IR lead to the development of post prandial hyperglycemia and overt T2DM both of which also acts as feedback mechanism for the development of oxidative stress.

Endoplasmic reticulum stress ERS is another mechanism that palys crucial role for the development of IR in adipocytes and peripheral tissues. ERS just like oxidative stress, is produced by the activation of JNK and inhibitory phosphorylation of IRS-1 in adipose tissues and liver [ 72 ] and induces the pathogenesis of IR in endothelial cells.

It has been found that ER is a major site for the production of various proteins such as insulin biosynthesis and act as a place for the lipid and sterol synthesis [ 73 ]. Any kind of abnormality that occurs in ER may lead to the development of ERS which also contribute to induce tissue-specific IR.

It has been revealved from experimental studies that some anti-diabetic agents alos modulate the ERS during the treatment of T2DM [ 74 ] which offer a new therapeutic target for the treatment of ERS-inducced IR and T2DM. NF-κB is a sequence-specific transcriptional mediated factor that primarily regulates various inflammatory responses [ 75 ] and IκB kinase β IKK-β is a central coordinator for these inflammatory responses through the activation of NF-κB [ 76 ].

IKK-β activates NF-κB through phosphorylation of IKK-β [ 77 , 78 ] and thereafter, NF-κB mediates the stimulation of numerous pro-inflammatory mediators such as IL-1β, IL-6, and TNF-α [ 76 , 78 ]. Once these pro-inflammatory cytokines are activated, they ultimately lead to cause IR [ 2 , 14 , 79 , 80 ].

Therefore, NF-κB and IKK-β are considered to be involved in the pathogenesis of IR [ 81 , 82 ]. IKK-β induces inflammatory responses in hepatocytes which massively increase the production of pro-inflammatory cytokines [ 83 ].

These pro-inflammatory cytokines then enter into the blood stream to cause IR in other tissues [ 81 ]. Various studies have investigated that nonsteroidal anti-inflammatory drugs NSAIDs such as cyclooxygenase inhibitors aspirin and salicylates can significantly inhibit the activation of NF-κB and IKK-β [ 84 ] in rodent models and humans [ 84 , 85 ].

These studies suggest that NSAIDs may exhibit their anti-inflammatory effects on myeloid cells rather than in muscle or fat. Expression of IKK-β in myeloid cells significantly suppresses the activation of pro-inflammatory cytokines that promote IR [ 81 ].

In the following sub-sections, role of various transcriptional pathways in the pathogenesis of IR has been briefly described. IR leads to the increased production of insulin from β-cells of pancreatic islets and as result, compensatory hyperinsulinemia within the body occurs.

Toll like receptors TLRs are the important modulators of IR and its comorbidities. Chronic inflammation plays a crucial role in variety of insulin resistant states [ 86 , 87 ] in which various signaling pathways are activated that directly interfere with the normal functioning of the key components of insulin signaling pathways [ 88 ].

Among various pathways, activation of TLRs imparts crucial role for the generation of inflammation. There are two main types of TLRs i. TLR2 and TLR4.

TLR4 is an extracellular cell surface receptor that is expressed in β-cells of pancreatic islets, brain, liver skeletal muscle and adipose tissues Fig.

In nomal conditions, TLR4 regulates insulin sensitivity in these tissues, but the activation of TLR4 directly dampen the insulin action through the activation of various pro-inflammatory mediators and ROS, indirectly generates the activation of various pro-inflammatory mediators by inducing various signaling cascades and transcriptional factors notably MyD88, TIRAP, TRIF, IKKs and JNKs that causes the activation of innate immune responses which ultimately leads to the development of IR Fig.

TLR4 plays this role primarly in coordination with the phosphorylation of IRS serine. Expression TLR4 in integrated tissues and organ systems of the body that regulate the insulin sensitivity.

Toll-like receptor 4 TLR4 present in adipocytes, initiates the inflammatory responses that release various pro-inflammatory mediators. Once, produced, these mediators are entred into the blood stream and thereby promote IR.

TLR4, expressed on Kupffer cells and other liver cell components, regulates the various inflammatory responses in liver. TLR4, expressed in skeletal muscles, has been shown to regulate the substrate metabolism in muscle, favoring glucose oxidation in the absence of insulin.

Hypothalamus and mesolimbic area are important sites that modulate the energy expenditure, pancreatic β-cell function and IR in peripheral tissue. Expression of TLR4 in hypothalamus potentiates various inflammatory responses that contribute to the pathogenesis of IR.

Adopted from Kim and Sears Schematic representation of TLR4 signaling cascades. Lipopolysaccharide LPS and its endotoxic moiety have been reported to be the potential activators of TLR4 Fig. LPS is composed of oligosaccharides and acylated saturated fatty acids SFAs.

Besides LPS, SFAs have also been reported to be the activator of TLR4. The expression and signaling of TLR4 are regulated mainly by the adiponectins. Several studies have reported that adiponectin can inhibit LPS-induced activation of TLR4 through the involvement of AMPK, IL, and heme oxygenase-1 [ 90 — 92 ].

Other regulators of TLR4 are peroxisome proliferators-activated receptor gamma PPARγ and sex hormones [ 93 , 94 ]. Taking together, TLR4 is a molecular link for pro-inflamatory mediators, different body organs, and several transcriptional pathways and cascades that modulate the innate immune system by regulating the insulin sensitivity.

In the proceeding sub-sections, role of TLR4 expression in various vital organs of the body for the pathogenesis of IR has been described. Despite of having the ability to act as storage depot for excess calories, adipose tissues secrete large number of hormones, pro-inflammatory cytokines and chemokines that directly influence the metabolism Fig.

Adipose tissues consist of adipocytes, preadipocytes, macrophages, lymphocytes and endothelial cells. Only adipocytes and macrophages are known to release various pro-inflammatory cytokines IL-1β, IL-6, and TNF-α and chemokines such as MCP-1 that potentiate inflammation in several tissues after being released into the systemic circulation [ 95 ].

Besides this, adipocytes are also a rich source of two important hormones namely leptin [ 96 , 97 ] and adiponectin [ 98 ]. Adiponectin, having anti-inflammatory properties, promotes insulin sensitivity whereas, leptin having inflammatory properties, impairs insulin sensitivity in adipocytes [ 87 ].

Several factors such as oxidative stress, increased FFAs flux and hypoxia that are associated with inflammation can induce IR in adipose tissues [ 87 ].

TLRs present in adipose tissues are directly activated by the nutrients [ 99 , ] which play a key role for the initiation of inflammatory responses which ultimately promotes IR in these tissues [ — ]. Nutritional fatty acids can activate the expression of TLR4 in adipocytes that play crucial role for the activation of various pro-inflammatory mediators and transcriptional mediated pathways which ultimately lead to the development of IR in adipocytes.

Skeletal muscles have marked significance to regulate the normal glucose homeostasis and development of IR as these are the primary site for insulin-induced glucose uptake and utilization in peripheral tissues.

Skeletal muscles contain myocytes and macrophages in which TLR4 receptors are expressed Fig. Signal transduction of TLR receptors is an underlying mechanism for the development of IR and chronic inflammation in skeletal muscles [ ]. TLR4 expression in skeletal muscle is associated with severity of IR and skeletal muscle metabolism.

The mechanis in the development of IR in skeletal muscles may include the direct effects of intramyocellular FFAs metabolites in skeletal muscles, macrophages and paracrine effects of adipocytes.

Recently, it has been experimentally confirmed that disruption of TLR4 expression prevents SFA-induced IR in TLR mutant mice and improves IRS-1 tyrosine phosphorylation and insulin-stimulated glucose uptake.

Moreover, disruption of TLR4 expression has also shown to decrease the JNK1 phosphorylation and IRS-1 serine phosphorylation [ , ]. Liver is the major and vital organ of the body which is composed of heterogenous types of cells notably hepatocytes, immune cells, kupffer cells and endothelial cells.

Due to their localization at sinusoids, kupffer cells are in close contact with circulating cytokines, lipids, hormones and postprandial LPS, and hence, kupffer cells are important mediators of inflammation within the liver. TLR4 expressed on kupffer cells in the liver Fig.

It has been found that activated levels of pro-inflammatory AP-1 and NF-κB in liver are directly correlated with IR and oxidative stress [ ]. TLR4 signaling pathway is strongly associated with IR as, it has been found that acute treatment of LPS inhibits the production of hepatic glucose via activation of TLR4 signaling pathway and induces IR in liver [ ].

Several TLRs such as TLR2, TLR3 and TLR4, are also expressed in β-cells of pancreatic islets [ ]. Signal transduction of TLRs in β-cells of pancreatic islets is mainly associated with inflammation in β-cells of pancreatic islets [ — ].

Distruction and malfunctioning of β-cells of pancreatic islets may lead to insufficient secretion of insulin in both types of DM. Expression of TLR4 in pancreatic islets may lead to impaired insulin secretion and promote β-cell apoptosis [ ].

Brain itself palys a central role to regulate glucose homeostasis and metabolism. In brain, hypothalamus and mesolimbic sites have been considered as important areas that are actively involved in the regulation of insulin sensitivity in peripheral tissues and β-cells secretory functions of pancreatic islets [ ].

TLR4 expression is widely distributed in the body Fig. Vascular endothelial dysfunction is a major complication for induction of IR and pathogenesis of T2DM. At molecular level, excess amount of nutrient is interlinked with IR through the activation of transcriptional mediated pathways such as IKKβ and NF-κB [ 83 , ].

Augmented levels of FFAs are associated with generation of inflammation and induction of IR in endothelial cells [ , ]. IKKβ and NF-κB are transcriptional mediators of inflammation and TLR4 is implicated as a mediator of IKKβ and NF-κB [ , ].

TLR4 receptors are also expressed in endothelial cells and expression of TLR4 via LPS-stimulated IKKβ and NF-κB activation contributes the dysfunctioning of endothelial cells [ ].

Activation of TLR4 via FFAs can trigger the cellular inflammatory responses in endothelial cells [ , ] whereas, whole body deletion of TLR4 expression has shown to prevent high-fat diet-induced vascular inflammation and IR in mice [ , ].

Similarly, activation of TLR4-dependent IKK and NF-κB indicated impaired insulin signaling and NO production in endothelial cells [ ].

The growing evidence implicates that TLR4 is the major causative factor to induce IR in endothelial cells via activation of various transcriptional mediated pathways and inflammation in endothelial cells.

AMP-activated protein kinase AMPK is an enzyme that is most commonly known as master regulator of energy metabolism [ ] and its activation is based on the energy level of the body.

Upon activation, AMPK resotres the energy levels of the body by stimulating various processes in different body organs Fig. AMPK plays a crucial role between adipose and peripheral tissues, and interferes various metabolic and secretory functions [ ] that are responsible for normoglycemia and glucose homeostasis Fig.

In adipocytes, adipokines exhibit their metabolic effects by activating AMPK which result in the increased β-oxidation in peripheral tissues. Activation of AMPK in peripheral tissues enables skeletal muscles to cope with elevated levels of FFAs.

Keeping in view the active role of AMPK in energy metabolim, it has been found that AMPK activation improves insulin sensitivity and glucose homeostasis.

IR is a major hallmark for the pathogenesis of T2DM however, AMPK activation can prevent the pathogenesis of IR and development of T2DM. Protein kinase C PKC and inhibitor kB kinase IKK are the two main important kinases that play crucial role in pro-inflammatory mediators-induced inflammatory processes in adipocytes and peripheral tissues underlying the development of systemic IR [ — ].

IKK induces IR in peripheral tissues by suppressing the insulin signaling and activating NF-κB [ , ]. Inhibition of IKK activation prevents the secretion of adipokines from adipocytes and improves insulin sensitivity in adipocytes and peripheral tissues [ 81 , , ].

NF-κB is a transcriptional mediated pathway that plays its crucial role in the transcription of signals for te production and release of various pro-inflammatory mediators. Most importantly, NF-κB plays active role to regulate IL-1β Fig. Once activated, NF-κB targets serval genes to potentiate the release of various pro-inflammatory mediators in adipose tissues and liver [ 81 , 83 , ].

These pro-inflammatory mediators that are produced in response to NF-κB activation induce tissue-specific IR. Glucolipotoxicity is a general term which is collectively used for the combination of glucotoxicity and lipotoxicity. These two terms are collectively responsible to activate the release of various pro-inflammatory mediators which lead to the development of tissue-specific IR and impaired insulin secretion from β-cells of pancreatic islets Fig.

Adipocytes are the main sites for the storage of fats and energy supplied to the body, is also regulated by the adipocytes. When accumulation of lipids exceeds the energy expenditure, then most of the excess amount is stored in the form of FFAs in adipose and other insulin-sensitive tissues.

When fat storage and energy supply is impaired in adipose tissues, elevation of FFAs levels in plasma occurs which is converted into the triglycerides and stores in non-adipose tissues [ ]. The ectopic storage of FFAs metabolites mostly triglycerides results in lipotoxic effects in peripheral tissues Fig.

In addition to this, elevated levels of FFAs in plasma may also interfere with insulin signaling pathways notably IRS-1 serine phosphorylation in peripheral tissues via activation of PKC and inhibition of IKK and JNK [ ]. Hence, it has been evidenced that glucolipotoxicity is one of the major contributor for the development of tissue-specific IR.

Mechanism of hyperglycemia- and dyslipidemia-induced inflammation for the development of IR and T2DM. Hyperglycemia and dyslipidemia collectively provoke the activation of pro-inflammatory mediators through the involvement of several metabolic pathways.

Once, these pro-inflammatory mediators are released, they induce tissue-specific inflammation due to which IR in peripheral tissues and impaired insulin secretion in pancreatic islets occur that ultimately lead to overt T2DM.

Adapted from Akash et al. Development of IR is one of the major hallmark for pathogenesis of T2DM. To control the propagation of IR is one of the most important targeted treatment.

For the development of IR, several factors are involved Fig. Several treatment strategies have been used to overcome the development of IR. The most important ones have been described here in the following sub-sections. Interleukin-1 receptor antagonist IL-1Ra is naturally occurring anti-inflammatory cytokine of interleukin-1 family.

It competitively binds with IL-1RI and prevent the binding of IL-1β and antagonizes its effects. It has been evidenced from several experimental studies that imbalance between IL-1Ra and IL-1β generates inflammation in various parts of the body where IL-1RI is present [ 4 , 12 ]. Moreover, it has also been found that expression of IL-1Ra is strongly correlated with the development of IR, impaired insulin secretion and T2DM [ 4 , ].

Treatment of human recombinant IL-1Ra improves normoglycemia, insulin sensitivity in adipose and peripheral tissues, and insulin secretion from β-cells of pancreatic islets impairs [ 31 , , ].

This is one of the most important treatment strategy that anti-inflammatory agent might indeed prevent the development of IR and improves glycemia.

One of the main shortcoming of IL-1Ra is its short biological half-life and to overcome this problem, high doses with frequent dosing intervals are required to achieve desired therapeutic effects. To overcome this problem, several treatment strategies have been applied to prolong the biological half-life and therapeutic effects of IL-1Ra [ 29 ].

Salicylates are an important class of anti-inflammatory agents. They are used in variety of inflammatory diseases and syndromes. Inflammation plays a crucial role for the development of IR and T2DM, therefore, by using salicylates as an alternate treatment strategy, it has been found that salicylates can imporve insulin sensitivity via inhibition of NF-κB and IKKβ [ 82 ] and glucose tolerance [ , ].

In the above sections, it has been briefly described that TNF-α is one of the most important pro-inflammatory mediator that is responsible to induce IR in adipocytes and peripheral tissues.

Inhibition of TNF-α production might be one of the choice to prevent the development of IR and pathogenesis of T2DM [ 4 ]. Recently, infliximab has been demonstrated to improve insulin signaling and inflammation especially in the liver in rodent model of diet-induced IR [ ].

Similarly, using anti-TNF-α antibodies also improve the insulin sensitivity in peripheral tissues [ ]. Lo et al. demonstrated that etanercept therapy can also improve total concentration of adiponectin which is anti-inflammatory adipokine and improved insulin sensitivity [ ].

Keeping in view the decisive role of TNF-α in pathogenesis of IR, several anti-TNF-α treatment strategies have been utilized to prevent the pathogeneis of IR and development of T2DM.

Similarly, anti-TNF-α treatment has also shown to prevent the IR in Sprague—Dawley rats [ ] while neutralization of TNF-α also prevented IR in hepatocytes [ ]. Few controversial studies have also demonstrated that using TNF-α blockade has no effect on IR [ ] which indicates that TNF-α blockade is not a treatment of choice as its production is dependent on the generation of IL-1β and activation of various transcriptional mediated pathways.

It has been thought that chemokines activately participate in the development of IR by potentiating the inflammation in adipocytes. Moreover, genetic inactivation of these chemokine signaling [ 52 , 53 , ] or inhibition of their axis [ , ] by pharmacological approaches have been shown to improve the insulin sensitivity in adipocytes and peripheral tissues.

ER stress, as mentioned in the above sections, is a key link between IR and T2DM [ ]. Blockade of ER stress is one of the treatment option to prevent the development of IR and pathogenesis of T2DM. In the recent years, various pharmaceutical chaperones, notably endogenous bile acids and the derivatives of these bile acids such as ursodeoxycholic acid UDCA , 4-phenyl butyric acid PBA have been investigated that have proven to have the ability to modulate the normal functioning of ER and its folding capacity [ 28 ].

Ozcan et al. The results of this study indicated that UDCA significantly improved insulin sensitivity and normoglycemia. Thiazolidinediones also known as glitazones, are one of the most important insulin sensitisers. They are the agonists of peroxisome proliferator-activated receptors-gamma PPARγ.

It has been found that thaizolidinediones have the ability to improve insulin action and decrease IR [ , ]. Inflammatory responses are induced through the activation of various pro-inflammatory and oxidative stress mediators via involment of various transcriptional mediated pathways.

To stop the inflammatory responses in IR development is one of the key treatment strategy. In this areticle, we have comprehensively highlighted the up-to-date scientific knowlesge of role of inflammatory responses in IR development and its treatment strategies.

IR plays a crucial role for the pathogenesis and development of T2DM and its associated complicaitons. Based on the findings mentioned in above sections, anti-inflammatory treatment strategies are one of the best choice to prevent the the pathogenesis of IR, but the studies conducted to investigate the role of anti-inflammatory strategies for the prevention of IR are still in their beginning stages and need to be focused further in future studies for more better and improved clinical outcomes.

Feve B, Bastard JP. The role of interleukins in insulin resistance and type 2 diabetes mellitus. Nat Rev Endocrinol. Article CAS PubMed Google Scholar. Hotamisligil GS. Inflammatory pathways and insulin action.

Int J Obes Relat Metab Disord. Moller DE. Potential role of TNF-alpha in the pathogenesis of insulin resistance and type 2 diabetes. Trends Endocrinol Metab. Akash MSH, Shen Q, Rehman K, Chen S. Interleukin-1 receptor antagonist: a new therapy for type 2 diabetes mellitus.

J Pharm Sci. Rehman K. and Akash M. Nutrition and Diabetes Mellitus: How are They Interlinked? Kawazoe Y, Naka T, Fujimoto M, Kohzaki H, Morita Y, Narazaki M, Okumura K, Saitoh H, Nakagawa R, Uchiyama Y, Akira S, Kishimoto T.

J Exp Med. Article CAS PubMed PubMed Central Google Scholar. Bruunsgaard H, Pedersen M, Pedersen BK. Aging and proinflammatory cytokines. Curr Opin Hematol.

Franceschi C, Bonafe M, Valensin S, Olivieri F, De Luca M, Ottaviani E, De Benedictis G. An evolutionary perspective on immunosenescence, Annals of the New York Academy of Sciences.

Google Scholar. Albright JW, Albright JF. Soluble receptors and other substances that regulate proinflammatory cytokines in young and aging humans. J Gerontol A Biol Sci Med Sci.

Akash MSH, Rehman K, Chen S. An overview of valuable scientific models for diabetes mellitus. Curr Diabetes Rev.

Article PubMed Google Scholar. Role of inflammatory mechanisms in pathogenesis of type 2 diabetes mellitus. J Cell Biochem. Donath MY, Shoelson SE. Type 2 diabetes as an inflammatory disease. Nat Rev Immunol. Hak AE, Pols HA, Stehouwer CD, Meijer J, Kiliaan AJ, Hofman A, Breteler MM, Witteman JC.

Markers of inflammation and cellular adhesion molecules in relation to insulin resistance in nondiabetic elderly: the Rotterdam study.

J Clin Endocrinol Metab. Elevated levels of acute-phase proteins and plasminogen activator inhibitor-1 predict the development of type 2 diabetes: the insulin resistance atherosclerosis study. Pickup JC, Crook MA. Is type II diabetes mellitus a disease of the innate immune system? Jager J, Gremeaux T, Cormont M, Le Marchand-Brustel Y, Tanti JF.

Interleukin-1beta-induced insulin resistance in adipocytes through down-regulation of insulin receptor substrate-1 expression. Donath MY, Boni-Schnetzler M, Ellingsgaard H, Ehses JA.

Islet inflammation impairs the pancreatic beta-cell in type 2 diabetes. Physiology Bethesda. Article CAS Google Scholar. Fagiolo U, Cossarizza A, Scala E, Fanales-Belasio E, Ortolani C, Cozzi E, Monti D, Franceschi C, Paganelli R.

Increased cytokine production in mononuclear cells of healthy elderly people. Eur J Immunol. Abbatecola AM, Ferrucci L, Grella R, Bandinelli S, Bonafe M, Barbieri M, Corsi AM, Lauretani F, Franceschi C, Paolisso G.

Diverse effect of inflammatory markers on insulin resistance and insulin-resistance syndrome in the elderly. J Am Geriatr Soc. Bastard JP, Maachi M, Van Nhieu JT, Jardel C, Bruckert E, Grimaldi A, Robert JJ, Capeau J, Hainque B. Adipose tissue IL-6 content correlates with resistance to insulin activation of glucose uptake both in vivo and in vitro.

Fernandez-Real JM, Vayreda M, Richart C, Gutierrez C, Broch M, Vendrell J, Ricart W. Circulating interleukin 6 levels, blood pressure, and insulin sensitivity in apparently healthy men and women.

Pickup JC, Mattock MB, Chusney GD, Burt D. NIDDM as a disease of the innate immune system: association of acute-phase reactants and interleukin-6 with metabolic syndrome X. Kern PA, Ranganathan S, Li C, Wood L, Ranganathan G. Adipose tissue tumor necrosis factor and interleukin-6 expression in human obesity and insulin resistance.

Am J Physiol Endocrinol Metab. CAS PubMed Google Scholar. Kirwan JP, Jing M. Modulation of insulin signaling in human skeletal muscle in response to exercise.

Exerc Sport Sci Rev. Emanuelli B, Peraldi P, Filloux C, Sawka-Verhelle D, Hilton D, Van Obberghen E. SOCS-3 is an insulin-induced negative regulator of insulin signaling. J Biol Chem. Krebs DL, Hilton DJ. SOCS: physiological suppressors of cytokine signaling.

J Cell Sci. Hwa V, Nadeau K, Wit JM, Rosenfeld RG. STAT5b deficiency: lessons from STAT5b gene mutations.

Best Pract Res Clin Endocrinol Metab. Tilg H, Moschen AR. Inflammatory mechanisms in the regulation of insulin resistance. Mol Med. CAS PubMed PubMed Central Google Scholar.

IL-1Ra and its delivery strategies: inserting the association in perspective. Pharm Res. Akash MSH, Rehman K, Li N, Gao JQ, Sun H, Chen S.

Sustained delivery of IL-1Ra from pluronic Fbased thermosensitive gel prolongs its therapeutic potentials. Akash MSH, Rehman K, Sun H, Chen S. Sustained delivery of IL-1Ra from PFgel reduces hyperglycemia in diabetic GK-rats. PLoS One. Jones SA, Horiuchi S, Topley N, Yamamoto N, Fuller GM.

The soluble interleukin 6 receptor: mechanisms of production and implications in disease. FASEB J. Ogata A, Morishima A, Hirano T, Hishitani Y, Hagihara K, Shima Y, Narazaki M, Tanaka T.

Improvement of HbA1c during treatment with humanised anti-interleukin 6 receptor antibody, tocilizumab. Ann Rheum Dis. 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. Swaroop JJ, Rajarajeswari D, Naidu JN. Association of TNF-alpha with insulin resistance in type 2 diabetes mellitus.

Indian J Med Res. Hotamisligil GS, Shargill NS, Spiegelman BM. Adipose expression of tumor necrosis factor-alpha: direct role in obesity-linked insulin resistance. Aguirre V, Uchida T, Yenush L, Davis R, White MF. The c-Jun NH 2 -terminal kinase promotes insulin resistance during association with insulin receptor substrate-1 and phosphorylation of Ser Chen G, Goeddel DV.

TNF-R1 signaling: a beautiful pathway. Sharma R, Anker SD. Cytokines, apoptosis and cachexia: the potential for TNF antagonism. Int J Cardiol. Trayhurn P. Endocrine and signalling role of adipose tissue: new perspectives on fat. Acta Physiol Scand. Kershaw EE, Flier JS. Adipose tissue as an endocrine organ.

Sartipy P, Loskutoff DJ. Monocyte chemoattractant protein 1 in obesity and insulin resistance. Proc Natl Acad Sci U S A. Yang Q, Graham TE, Mody N, Preitner F, Peroni OD, Zabolotny JM, Kotani K, Quadro L, Kahn BB.

Serum retinol binding protein 4 contributes to insulin resistance in obesity and type 2 diabetes. Argiles JM, Lopez-Soriano J, Almendro V, Busquets S, Lopez-Soriano FJ.

Cross-talk between skeletal muscle and adipose tissue: a link with obesity? Med Res Rev. Krebs M, Roden M. Molecular mechanisms of lipid-induced insulin resistance in muscle, liver and vasculature.

Diabetes Obes Metab. Sjoholm A, Nystrom T. Inflammation and the etiology of type 2 diabetes. Diabetes Metab Res Rev. Article PubMed CAS Google Scholar. Kahn BB, Flier JS. Obesity and insulin resistance. J Clin Invest. Zhang F, Chen Y, Heiman M, Dimarchi R. Leptin: structure, function and biology.

Vitam Horm. Lihn AS, Pedersen SB, Richelsen B. Adiponectin: action, regulation and association to insulin sensitivity. Obes Rev. Tschritter O, Fritsche A, Thamer C, Haap M, Shirkavand F, Rahe S, Staiger H, Maerker E, Haring H, Stumvoll M.

Plasma adiponectin concentrations predict insulin sensitivity of both glucose and lipid metabolism. Ota T. Chemokine systems link obesity to insulin resistance.

Diabetes Metab J. Article PubMed PubMed Central Google Scholar. Kanda H, Tateya S, Tamori Y, Kotani K, Hiasa K, Kitazawa R, Kitazawa S, Miyachi H, Maeda S, Egashira K, Kasuga M. MCP-1 contributes to macrophage infiltration into adipose tissue, insulin resistance, and hepatic steatosis in obesity.

Weisberg SP, Hunter D, Huber R, Lemieux J, Slaymaker S, Vaddi K, Charo I, Leibel RL, Ferrante Jr AW. CCR2 modulates inflammatory and metabolic effects of high-fat feeding. Kamei N, Tobe K, Suzuki R, Ohsugi M, Watanabe T, Kubota N, Ohtsuka-Kowatari N, Kumagai K, Sakamoto K, Kobayashi M, Yamauchi T, Ueki K, Oishi Y, Nishimura S, Manabe I, Hashimoto H, Ohnishi Y, Ogata H, Tokuyama K, Tsunoda M, Ide T, Murakami K, Nagai R, Kadowaki T.

Overexpression of monocyte chemoattractant protein-1 in adipose tissues causes macrophage recruitment and insulin resistance. Xu L, Kitade H, Ni Y, Ota T. Roles of Chem okines and Chemokine Receptors in Obesity-Associated Insulin Resistance and Nonalcoholic Fatty Liver Disease. Biomolecules ;5 3 Kitade H, Sawamoto K, Nagashimada M, Inoue H, Yamamoto Y, Sai Y, Takamura T, Yamamoto H, Miyamoto K, Ginsberg HN, Mukaida N, Kaneko S, Ota T.

Gerhardt CC, Romero IA, Cancello R, Camoin L, Strosberg AD. Chemokines control fat accumulation and leptin secretion by cultured human adipocytes. Mol Cell Endocrinol. Moran A, Steffen LM, Jacobs Jr DR, Steinberger J, Pankow JS, Hong CP, Tracy RP, Sinaiko AR.

Relation of C-reactive protein to insulin resistance and cardiovascular risk factors in youth. Diabetes Care. Yoneda M, Mawatari H, Fujita K, Iida H, Yonemitsu K, Kato S, Takahashi H, Kirikoshi H, Inamori M, Nozaki Y, Abe Y, Kubota K, Saito S, Iwasaki T, Terauchi Y, Togo S, Maeyama S, Nakajima A.

High-sensitivity C-reactive protein is an independent clinical feature of nonalcoholic steatohepatitis NASH and also of the severity of fibrosis in NASH. J Gastroenterol. Ridker PM. C-reactive protein and the prediction of cardiovascular events among those at intermediate risk: moving an inflammatory hypothesis toward consensus.

J Am Coll Cardiol. Nakanishi N, Shiraishi T, Wada M. Association between C-reactive protein and insulin resistance in a Japanese population: the Minoh Study. Intern Med. Ndumele CE, Pradhan AD, Ridker PM. Interrelationships between inflammation, C-reactive protein, and insulin resistance.

J Cardiometab Syndr. Pradhan AD, Manson JE, Rifai N, Buring JE, Ridker PM. C-reactive protein, interleukin 6, and risk of developing type 2 diabetes mellitus. Evans JL, Goldfine ID, Maddux BA, Grodsky GM.

Are oxidative stress-activated signaling pathways mediators of insulin resistance and beta-cell dysfunction? Hennig B, Meerarani P, Ramadass P, Watkins BA, Toborek M. Fatty acid-mediated activation of vascular endothelial cells.

Hirosumi J, Tuncman G, Chang L, Gorgun CZ, Uysal KT, Maeda K, Karin M, Hotamisligil GS. A central role for JNK in obesity and insulin resistance. Newsholme P, Haber EP, Hirabara SM, Rebelato EL, Procopio J, Morgan D, Oliveira-Emilio HC, Carpinelli AR, Curi R.

Diabetes associated cell stress and dysfunction: role of mitochondrial and non-mitochondrial ROS production and activity. J Physiol. Shulman GI. Cellular mechanisms of insulin resistance. Cooke JP. Asymmetrical dimethylarginine: the Uber marker? Stuhlinger MC, Abbasi F, Chu JW, Lamendola C, McLaughlin TL, Cooke JP, Reaven GM, Tsao PS.

Relationship between insulin resistance and an endogenous nitric oxide synthase inhibitor. Arcaro G, Cretti A, Balzano S, Lechi A, Muggeo M, Bonora E, Bonadonna RC. Insulin causes endothelial dysfunction in humans: sites and mechanisms. Ozcan U, Cao Q, Yilmaz E, Lee AH, Iwakoshi NN, Ozdelen E, Tuncman G, Gorgun C, Glimcher LH, Hotamisligil GS.

Endoplasmic reticulum stress links obesity, insulin action, and type 2 diabetes. Boden G. Salvado L, Palomer X, Barroso E, Vazquez-Carrera M. Targeting endoplasmic reticulum stress in insulin resistance. Barnes PJ, Karin M. Nuclear factor-kappaB: a pivotal transcription factor in chronic inflammatory diseases.

N Engl J Med. Chen LW, Egan L, Li ZW, Greten FR, Kagnoff MF, Karin M. The two faces of IKK and NF-kappaB inhibition: prevention of systemic inflammation but increased local injury following intestinal ischemia-reperfusion.

Nat Med. Li Q, Van Antwerp D, Mercurio F, Lee KF, Verma IM. Severe liver degeneration in mice lacking the IkappaB kinase 2 gene. Maeda S, Chang L, Li ZW, Luo JL, Leffert H, Karin M. IKKbeta is required for prevention of apoptosis mediated by cell-bound but not by circulating TNFalpha.

Grimble RF. Inflammatory status and insulin resistance. Curr Opin Clin Nutr Metab Care. Klover PJ, Zimmers TA, Koniaris LG, Mooney RA. Chronic exposure to interleukin-6 causes hepatic insulin resistance in mice.

Arkan MC, Hevener AL, Greten FR, Maeda S, Li ZW, Long JM, Wynshaw-Boris A, Poli G, Olefsky J, Karin M. IKK-beta links inflammation to obesity-induced insulin resistance. Yuan M, Konstantopoulos N, Lee J, Hansen L, Li ZW, Karin M, Shoelson SE. Reversal of obesity- and diet-induced insulin resistance with salicylates or targeted disruption of Ikkbeta.

Cai D, Yuan M, Frantz DF, Melendez PA, Hansen L, Lee J, Shoelson SE. Local and systemic insulin resistance resulting from hepatic activation of IKK-beta and NF-kappaB. Karin M, Yamamoto Y, Wang QM. The IKK NF-kappa B system: a treasure trove for drug development.

Nat Rev Drug Discov. Arena FP, Dugowson C, Saudek CD. Salicylate-induced hypoglycemia and ketoacidosis in a nondiabetic adult. Arch Intern Med. Olefsky JM, Glass CK.

Macrophages, inflammation, and insulin resistance. Toll-like receptor 2-deficient mice are protected from insulin resistance and β cell dysfunction induced by a high-fat diet.

Haversen, L. Induction of proinflammatory cytokines by long-chain saturated fatty acids in human macrophages. Atherosclerosis , — Lee, J. Saturated fatty acids, but not unsaturated fatty acids, induce the expression of cyclooxygenase-2 mediated through Toll-like receptor 4.

Saturated fatty acid activates but polyunsaturated fatty acid inhibits Toll-like receptor 2 dimerized with Toll-like receptor 6 or 1. Zhou, R. Thioredoxin-interacting protein links oxidative stress to inflammasome activation.

A study that describes the mechanism of glucose-induced IL-1β production in pancreatic islets. van de Veerdonk, F. Reactive oxygen species-independent activation of the IL-1β inflammasome in cells from patients with chronic granulomatous disease.

Meissner, F. Inflammasome activation in NADPH oxidase defective mononuclear phagocytes from patients with chronic granulomatous disease.

Blood , — Schroder, K. The NLRP3 inflammasome: a sensor for metabolic danger? Biologic basis for interleukin-1 in disease. Blood 87 , — Bruun, J. Monocyte chemoattractant protein-1 release is higher in visceral than subcutaneous human adipose tissue AT : implication of macrophages resident in the AT.

Harman-Boehm, I. Macrophage infiltration into omental versus subcutaneous fat across different populations: effect of regional adiposity and the comorbidities of obesity.

Kanda, H. MCP-1 contributes to macrophage infiltration into adipose tissue, insulin resistance, and hepatic steatosis in obesity. Sartipy, P. Monocyte chemoattractant protein 1 in obesity and insulin resistance.

Kamei, N. Overexpression of monocyte chemoattractant protein-1 in adipose tissues causes macrophage recruitment and insulin resistance. Jiao, P. Obesity-related upregulation of monocyte chemotactic factors in adipocytes: involvement of nuclear factor-κB and c-Jun NH2-terminal kinase pathways.

IL-1β-MyD88 signaling is central to islet chemokine secretion in response to metabolic stress: evidence from a spontaneous model of type 2 diabetes, the GK rat. Diabetologia 50 S Marselli, L. Evidence of inflammatory markers in β cells of type 2 diabetic subjects.

Diabetologia 50 , S—S Wolf, A. Adiponectin induces the anti-inflammatory cytokines IL and IL-1RA in human leukocytes. Greenstein, A. Local inflammation and hypoxia abolish the protective anticontractile properties of perivascular fat in obese patients.

Rutkowski, J. Mechanisms of obesity and related pathologies: the macro- and microcirculation of adipose tissue. FEBS J. Fleischman, A. Salsalate improves glycemia and inflammatory parameters in obese young adults. Diabetes Care 31 , — Sustained effects of interleukin-1 receptor antagonist treatment in type 2 diabetes.

Gonzalez-Gay, M. Anti-tumor necrosis factor-α blockade improves insulin resistance in patients with rheumatoid arthritis.

Insulin resistance in rheumatoid arthritis: the impact of the anti-TNF-α therapy. NY Acad. Huvers, F. Improved insulin sensitivity by anti-TNFα antibody treatment in patients with rheumatic diseases. Yuan, M. Reversal of obesity- and diet-induced insulin resistance with salicylates or targeted disruption of IKKβ.

This study initially identified a potential role for NF-κB in T2D and showed that salicylates improved blood glucose levels in rodent models.

Frantz, B. The effect of sodium salicylate and aspirin on NF-κB. Jurivich, D. Effect of sodium salicylate on the human heat shock response.

Hundal, R. Mechanism by which high-dose aspirin improves glucose metabolism in type 2 diabetes. Partial pancreatectomy in the rat and subsequent defect in glucose-induced insulin release.

Leahy, J. Chronic hyperglycemia is associated with impaired glucose influence on insulin secretion. A study in normal rats using chronic in vivo glucose infusions.

Yki-Jarvinen, H. Hyperglycemia decreases glucose uptake in type I diabetes. Diabetes 36 , — Rossetti, L. Correction of hyperglycemia with phlorizin normalizes tissue sensitivity to insulin in diabetic rats.

Reaven, G. Measurement of plasma glucose, free fatty acid, lactate, and insulin for 24 h in patients with NIDDM. Diabetes 37 , — Walker, K. Monounsaturated fatty acids prevent the deleterious effects of palmitate and high glucose on human pancreatic β-cell turnover and function.

Distinct effects of saturated and monounsaturated fatty acids on β-cell turnover and function. Diabetes 50 , 69—76 Unger, R. Lipotoxicity in the pathogenesis of obesity-dependent NIDDM.

Genetic and clinical implications. Diabetes 44 , — Are the β-cell signaling molecules malonyl-CoA and cystolic long-chain acyl-CoA implicated in multiple tissue defects of obesity and NIDDM?

Lipid homeostasis, lipotoxicity and the metabolic syndrome. Acta , — Poitout, V. Glucolipotoxicity: fuel excess and β-cell dysfunction. Evans, J. Oxidative stress and stress-activated signaling pathways: a unifying hypothesis of type 2 diabetes. Are oxidative stress-activated signaling pathways mediators of insulin resistance and β-cell dysfunction?

Diabetes 52 , 1—8 Araki, E. Endoplasmic reticulum stress and diabetes mellitus. Izumi, T. Dominant negative pathogenesis by mutant proinsulin in the Akita diabetic mouse. Endoplasmic reticulum stress and the inflammatory basis of metabolic disease.

Zraika, S. Toxic oligomers and islet β cell death: guilty by association or convicted by circumstantial evidence? American Diabetes Association. Diagnosis and classification of diabetes mellitus.

Diabetes Care 33 , S62—S69 Kibirige, M. Testing the accelerator hypothesis: the relationship between body mass and age at diagnosis of type 1 diabetes. Diabetes Care 26 , — Wilkin, T. The accelerator hypothesis: weight gain as the missing link between type I and type II diabetes.

Diabetologia 44 , — Decreased β-cell mass in diabetes: significance, mechanisms and therapeutic implications. Diabetologia 47 , — Hypponen, E. Obesity, increased linear growth, and risk of type 1 diabetes in children. Diabetes Care 23 , — Libman, I. Changing prevalence of overweight children and adolescents at onset of insulin-treated diabetes.

Fourlanos, S. Insulin resistance is a risk factor for progression to type 1 diabetes. Nathan, D. Medical management of hyperglycaemia in type 2 diabetes mellitus: a consensus algorithm for the initiation and adjustment of therapy.

Type 1, type 1. Use of salsalate to target inflammation in the treatment of insulin resistance and type 2 diabetes. Koska, J. The effect of salsalate on insulin action and glucose tolerance in obese non-diabetic patients: results of a randomised double-blind placebo-controlled study.

Diabetologia 52 , — Download references. The authors wish to thank their scientific collaborators who have contributed so much to these studies, in particular A. Goldfine, J. Lee, D. Mathis, K. Maedler, P.

Halban, T. Mandrup-Poulsen, J. Ehses and M. Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, CH, Basel, Switzerland.

Joslin Diabetes Center, Harvard Medical School, One Joslin Place, Boston, , Massachusetts, USA. You can also search for this author in PubMed Google Scholar.

Marc Y. Donath is listed as the inventor of a patent filed in for the use of an interleukin-1 receptor antagonist for the treatment of or prophylaxis against type 2 diabetes. He is a consultant for Novartis, XOMA, Eli Lilly and Company, Cytos, Merck and AstraZeneca.

Steven E. Shoelson holds patents on the use of salicylates in diabetes, prediabetes and cardiovascular disease. He has consulted for Catabasis, Amylin, AstraZeneca, Merck, Genentech, XOMA and Kowa. A pathological condition in which insulin becomes less effective at lowering blood glucose levels.

ER stress. A response by the ER that results in the disruption of protein folding and the accumulation of unfolded proteins in the ER. The toxic effects of elevated levels of free fatty acids.

These detrimental effects may be functional and reversible, or may lead to cell death. The toxic effects of hyperglycaemia. A disease resulting from an attack by the innate immune system on the body's own tissues.

By contrast, autoimmune diseases are caused by the pathological activation of adaptive immune responses. Autoimmune and autoinflammatory diseases have some characteristics in common, including shared effector mechanisms. A macrophage that is activated by Toll-like receptor ligands such as lipopolysaccharide and interferon-γ, and that expresses inducible nitric oxide synthase, which generates nitric oxide.

A macrophage that is stimulated by interleukin-4 IL-4 or IL and that expresses arginase 1, the mannose receptor CD and the IL-4 receptor α-chain. The Kit W—sh or sash mutation abolishes KIT expression in mast cells, and the mutant mice are deficient in mast cells. Inflammation of the pancreatic islets during the progression of diabetes.

Insulitis in type 1 diabetes is caused by autoimmunity and in type 2 diabetes by metabolic stressors such as hyperglycaemia and elevated levels of free fatty acids. A condition in which the flow of blood to a tissue or organs is less than normal, and which results in injury to that tissue or organ.

Severe weight loss, muscle wasting and debility caused by prolonged disease. It is thought to be mediated through neuroimmunoendocrine interactions.

A protein hormone that regulates energy intake and expenditure. It is one of the most important adipose-derived hormones and its production correlates with the mass of adipose tissue. A molecular complex of several proteins that, when activated, results in the production of active caspase 1, which cleaves pro-interleukin-1β pro-IL-1β and pro-IL to produce the active cytokines.

A prodrug form of salicylic acid that has fewer side effects than sodium salicylate. Salsalate is approved for use in humans as a source of salicylic acid. Reprints and permissions. Type 2 diabetes as an inflammatory disease. Nat Rev Immunol 11 , 98— Download citation. Published : 14 January Issue Date : February Anyone you share the following link with will be able to read this content:.

Sorry, a shareable link is not currently available for this article. Provided by the Springer Nature SharedIt content-sharing initiative. Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Skip to main content Thank you for visiting nature. nature nature reviews immunology review articles article. Subjects Immunopathogenesis Inflammation Therapeutics Type 2 diabetes. Key Points Type 2 diabetes is associated with obesity, ageing and inactivity.

Abstract Components of the immune system are altered in obesity and type 2 diabetes T2D , with the most apparent changes occurring in adipose tissue, the liver, pancreatic islets, the vasculature and circulating leukocytes. Access through your institution. Buy or subscribe. Change institution. Learn more.

Figure 1: Development of inflammation in type 2 diabetes. Figure 2: Interleukin-1β-induced inflammation in islets of patients with type 2 diabetes. References Shoelson, S. Article CAS PubMed PubMed Central Google Scholar Donath, M. Article CAS PubMed Google Scholar Bonner-Weir, S.

Article CAS PubMed Google Scholar Kahn, B. Article CAS PubMed Google Scholar Rhodes, C. Article CAS PubMed Google Scholar Robertson, R. Article CAS PubMed Google Scholar Weir, G. Article CAS PubMed Google Scholar Prentki, M. Article CAS PubMed PubMed Central Google Scholar Hull, R.

Article CAS PubMed Google Scholar Harding, H. Article CAS PubMed Google Scholar Hotamisligil, G. Article CAS Google Scholar Donath, M.

Article CAS PubMed Google Scholar Ehses, J. Article CAS PubMed Google Scholar Donath, M. Article CAS PubMed Google Scholar Masters, S. Article CAS Google Scholar Pickup, J. Article CAS PubMed Google Scholar Spranger, J. Article CAS PubMed Google Scholar Herder, C. Article PubMed Google Scholar Herder, C.

Article PubMed PubMed Central Google Scholar Pradhan, A. Article CAS PubMed Google Scholar Meier, C. Article CAS PubMed Google Scholar Carstensen, M. Article CAS PubMed PubMed Central Google Scholar Marculescu, R.

Article CAS PubMed Google Scholar Dinarello, C. Article CAS PubMed Google Scholar Larsen, C. Google Scholar Goldfine, A. Article PubMed PubMed Central Google Scholar Hotamisligil, G. Article CAS PubMed Google Scholar Schreyer, S. Article CAS PubMed PubMed Central Google Scholar Bernstein, L. Article CAS PubMed PubMed Central Google Scholar Dominguez, H.

Article CAS PubMed Google Scholar Lo, J. Article CAS PubMed Google Scholar Ofei, F. Article PubMed Google Scholar Paquot, N. CAS PubMed Google Scholar Rosenvinge, A.

Article CAS PubMed Google Scholar Kiortsis, D. Article CAS PubMed Google Scholar Stanley, T. Article CAS Google Scholar Yazdani-Biuki, B. Article PubMed Google Scholar Yazdani-Biuki, B. Article CAS PubMed Google Scholar Weisberg, S. Article CAS PubMed PubMed Central Google Scholar Xu, H.

Article CAS PubMed PubMed Central Google Scholar Lumeng, C. Article CAS PubMed Google Scholar Shaul, M. Article CAS PubMed PubMed Central Google Scholar Kosteli, A.

Article CAS PubMed PubMed Central Google Scholar Herrero, L. Article CAS PubMed Google Scholar Lumeng, C. Article CAS PubMed PubMed Central Google Scholar Liu, J.

Article CAS PubMed Google Scholar Wu, H. Article CAS PubMed Google Scholar Feuerer, M. Article CAS PubMed Google Scholar Ilan, Y. Article CAS PubMed PubMed Central Google Scholar Baecher-Allan, C. Article CAS PubMed Google Scholar Roncarolo, M. Article CAS Google Scholar Maedler, K. Article CAS PubMed PubMed Central Google Scholar Maedler, K.

Article CAS PubMed PubMed Central Google Scholar Ehses, J. Article CAS PubMed Google Scholar Carmeliet, P. Article CAS PubMed Google Scholar Hosogai, N. Article CAS PubMed Google Scholar Yin, J. Article CAS PubMed Google Scholar Pasarica, M. Article CAS PubMed PubMed Central Google Scholar Pasarica, M.

Article CAS PubMed PubMed Central Google Scholar Murdoch, C. Article CAS PubMed Google Scholar Burke, B.

Article CAS PubMed PubMed Central Google Scholar Cinti, S. Article CAS PubMed Google Scholar Strissel, K. Article CAS PubMed Google Scholar Arkan, M. Article CAS PubMed Google Scholar Cai, D.

Article CAS PubMed Google Scholar Solinas, G. Article CAS PubMed Google Scholar Shi, H. Article CAS PubMed PubMed Central Google Scholar Sabio, G. Article CAS PubMed PubMed Central Google Scholar Solinas, G.

Article CAS PubMed Google Scholar Tuncman, G. Article CAS PubMed PubMed Central Google Scholar Cai, D. Article CAS PubMed Google Scholar Zhang, X. Article CAS PubMed PubMed Central Google Scholar Eldor, R. Article CAS PubMed PubMed Central Google Scholar Mooney, R.

Article CAS PubMed Google Scholar Pedersen, B. Article CAS PubMed Google Scholar Fried, S. CAS PubMed Google Scholar Wunderlich, F. Article CAS PubMed Google Scholar Matthews, V. Article CAS PubMed Google Scholar Maedler, K. Article CAS PubMed Google Scholar Schumann, D. Article CAS PubMed PubMed Central Google Scholar Boni-Schnetzler, M.

Article PubMed PubMed Central CAS Google Scholar Boni-Schnetzler, M. Article CAS PubMed Google Scholar Haversen, L.

Article CAS PubMed Google Scholar Lee, J. Article CAS PubMed Google Scholar Zhou, R. Article CAS Google Scholar van de Veerdonk, F. Article CAS PubMed PubMed Central Google Scholar Meissner, F. Article CAS PubMed PubMed Central Google Scholar Schroder, K. Article CAS PubMed PubMed Central Google Scholar Bruun, J.

Article CAS PubMed Google Scholar Harman-Boehm, I. Article CAS PubMed Google Scholar Kanda, H. Article CAS PubMed PubMed Central Google Scholar Sartipy, P. Article CAS PubMed PubMed Central Google Scholar Kamei, N.

Article CAS PubMed Google Scholar Jiao, P. Google Scholar Marselli, L. Google Scholar Wolf, A. Article CAS PubMed Google Scholar Greenstein, A. Article CAS PubMed Google Scholar Rutkowski, J.

Detoxification Methods Explained Health. Ultimate Guide. Inflammation helps infalmmation your body, but chronic inflammation can cause serious damage. Chronic hyperglycemia and inflammation Schabelman, MD. Hyperglcemia can get an Ethical food practices response to an injury like a cut or a splinterto an infection from bacteria or a virus, or from other exposures that the body may see as a threat, such as stressdietary sugarand environmental toxins. Inflammation is usually divided into two types: acute and chronic. Inflammation is a term that gets tossed hyerglycemia a lot these days, Detoxification Methods Explained what does hypegglycemia actually mean? And how inflammatlon it Muscle development supplementation your blood sugar? Here's everything Chronjc need to know about inflammation when you have diabetes. Although she has a Masters degree in Marketing from New York University, Vanessa's first true love is science. Always inquisitive, she sought out the help of a Registered Dietitian in in order to understand the role food plays in one's overall health and build a better relationship with food.

Author: Malajind

1 thoughts on “Chronic hyperglycemia and inflammation

Leave a comment

Yours email will be published. Important fields a marked *

Design by ThemesDNA.com