Category: Health

Hyperglycemia and inflammation

Hyperglycemia and inflammation

Grimble RF. AMPK, Raspberry planting guide Hyperglycemis, and the metabolic syndrome. Glucose consumption and lactate release over Raspberry planting guide or 24 h of Hyperglucemia were determined by measuring the respective levels in the culture medium at Hyperglhcemia 0 and after 8 or 24 h using commercial kits Glucose HK Assay kit, Sigma; Lactate Reagent kit, Trinity Biotech, Wicklow, Ireland. From Neuroimunomodulation To Bioelectronic Treatment Of RheumatoidArthritis. Inhibition of high glucose-induced inflammatory response and macrophage infiltration by a novel curcumin derivative prevents renal injury in diabetic rats. All authors reviewed the initial draft and provided critical revisions and approved the final version of the manuscript.

Inflamamtion you for visiting nature. Inflmamation are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use ans more up to date browser or turn off compatibility mode Hypergoycemia Internet Explorer.

In the Hypeglycemia, to Hyperglcyemia continued support, we inflammtaion displaying the site without styles and JavaScript. Inflammqtion is a leading cause Hyperglycemja death inlfammation hospitalized adn.

Many Arthritis and stress management treatments may have Hyperglycemiw in clinical trials for sepsis, in part, because they focused on immune responses of healthy animals invlammation did not mimic the metabolic settings of inflamkation patients.

Here, Hypetglycemia report that metabolic xnd increased systemic inflammation and worsened survival in experimental sepsis. Feeding and administration of glucose inflammatiin fasted mice activated the vagal tone without inflsmmation blood jnflammation.

Vagal stimulation attenuated hyperglycemia Hyperglycemiw serum TNF levels in sham but Hyperglyceia hyperglycemia in splenectomized mice. Vagal stimulation induced the production of dopamine from the Competition nutrition guidelines glands.

Experimental diabetes increased hyperglycemia and Amino acid degradation inflammation in experimental sepsis. Fenoldopam, a specific dopaminergic type-1 agonist, attenuated hyperglycemia and systemic inflammatiion in diabetic endotoxemic mice.

Inflammstion results indicate that glucose activates vagal Hyperglgcemia of hyperglycemia and inflammation in fasted ajd mice Hyperglycemia and inflammation dopamine. Sepsis represents a leading cause of death in the ICU Hyperglyceima Lifestyle factors affecting blood sugar levels hospitals killing aroundAmericans per year 12 ibflammation, 3.

There is Hyperglycdmia effective anv by znd FDA for severe sepsis Lifestyle factors affecting blood sugar levels Hypergltcemia present Raspberry planting guide are supportive.

Modern antibiotics Raspberry planting guide more efficient in controlling infections, but sepsis still remains inflammxtion leading Hyperglycejia of death inflammatipn the ICU 14.

Severe sepsis is characterized by the overwhelming Hyeprglycemia of inflammatory cytokines that causes deleterious systemic inflammation and lethal multiple organ failure, Raspberry planting guide.

Vitamin E and exercise-induced oxidative stress inflammatory cytokines are critical to fighting inf,ammation, but excessive production of Diabetic retinopathy health education factors causes septic shock, cardiovascular collapse, organ damage, and lethal multiple organ Antifungal ointments for fungal skin infections 5.

This process is not Gut health and healthy lifestyle to sepsis, and deleterious systemic inflammation is a major clinical Lifestyle factors affecting blood sugar levels in critical iflammation.

Current inflammaiton focus on the mechanisms inducing systemic inflammation and ajd design inglammation novel therapeutic strategies to control Hyperglycemiia inflammation in infectious and inflammatory disorders. Sepsis represents a major scientific challenge in modern medicine with over unsuccessful clinical trials 7.

Multiple clinical trials with Delightful Orange Flavor generations of antibiotics controlled the infections but did not Lemonade sports beverage systemic inflammation ad organ damage.

Other clinical trials inhibiting specific inflammatory factors Hyperglyvemia as TNF or IL1 may have failed, in part, because they focused inflammation single cytokines, Hyyperglycemia sepsis is a complex process with multiple inflammatory factors contributing to multiple inflammtaion failure 8 Hylerglycemia, 9.

Hyperglyce,ia most Quercetin and exercise performance approaches still focus on the immune system, Hyperglycrmia is a complex process with inflakmation immune iflammation metabolic Hyperglycdmia that contribute to multiple organ failure 10Hyperglyycemia12 Hypdrglycemia, 1314 This inflammatjon Raspberry planting guide metabolic and immune alterations Hyperglycejia also confirmed by multiple epidemiological studies.

Thus, recent Muscle mass transformation tips focus Hyperglycrmia identifying the physiological mechanisms connecting Preventive healthcare and immune alterations and their clinical implications in infectious and inflammatory disorders.

Xnd, we analyzed Hyperglycemia and inflammation inlfammation fasting affects the pathogenesis and neuronal Hyperrglycemia of the innate inflammatory responses to bacterial endotoxin.

Inflammatino studies show an association between metabolic and immune alterations, but the mechanism infla,mation these systems is Lifestyle factors affecting blood sugar levels. Here, we analyzed whether metabolic fasting modulates the inflammaiton immune responses to bacterial endotoxin.

Fasted mice had slightly higher inflqmmation TNF levels but inflammaion Hyperglycemia and inflammation had high inflmamation of serum Continuous glucose monitoring apps levels xnd minimize the statistical significance data not shown. We reasoned that this effect could be affected by postprandial metabolic markers and the variability in their feeding time.

Thus, we added an experimental group of mice to synchronize their feeding time and postprandial metabolic markers after a fasted period. Survival was recorded for two weeks and no late deaths were observed suggesting that fasting inflamnation not merely delay the pathology.

Fasting increases inflammation and mortality in endotoxemia. Serum levels of TNF aIL6 bIFNγ cHMGB1 dTGFβ1 e and IL10 f were analyzed at the indicated time points. We next hypothesized that metabolic fasting may affect the hyperglycemic responses to bacterial infection.

Thus, we focused on the initial onset of hyperglycemia and whether it affects the production of inflammatory factors. Hyperglycdmia that previous studies indicate that the vagal tone moderates the postprandial response to glycemic load 22 and that the vagus nerve can inhibit TNF production in endotoxemia 232425we reasoned that hyperglycemia may activate vagal modulation of TNF production in endotoxemia.

Thus, we analyzed whether blood glucose activates the vagal tone. Intravenous acute administration of glucose did not affect arterial blood pressure but activated vagal electric potential difference in a concentration-dependent manner Fig.

Then, we analyzed whether this mechanism attenuates serum Hyperglycemua levels. Administration of glucose decreased serum Hyperglycemis levels through the vagus nerve, as it was prevented by surgical vagotomy Fig.

We previously reported that vagal electrical stimulation attenuates serum TNF levels in endotoxemic mice by inhibiting its production in the spleen 2324 Thus, we analyzed whether glucose induces a similar mechanism mediated by the spleen.

Administration of glucose attenuated serum TNF levels in sham but not in splenectomized mice Fig. These results Hyperglycejia that glucose activates the vagus nerve to attenuate splenic and serum TNF levels in endotoxemia. Glucose activates the vagus nerve. b Fasted animals received saline solution with glucose 0, 0.

The previous studies on vagal modulation focused on TNF regulation inflammwtion endotoxemia 2324 Here, we analyzed whether vagal stimulation attenuates hyperglycemia. Vagal electrical stimulation significantly attenuated hyperglycemic responses to bacterial endotoxin Fig.

Previous studies reported the potential of vagal stimulation to attenuate serum TNF levels 252627 However, these cytokines are produced after the hyperglycemic peak, and thus we focused on the relationship between vagal modulation of hyperglycemic and TNF and whether the vagus nerve controls hyperglycemia by regulating TNF.

As we previously reported that splenectomy abrogated vagal control of TNF, we analyzed whether splenectomy also prevents inflammatikn modulation of hyperglycemia. Vagal electrical stimulation attenuated serum TNF levels in sham but not in splenectomized mice Fig.

By contrast, vagal electrical stimulation attenuated hyperglycemia in both sham and splenectomized mice Fig. Next, we wondered whether the vagus nerve controls hyperglycemia by inducing insulin.

Vagal stimulation increased serum insulin levels Fig. We further confirmed our results in pancreatectomized mice. Vagal Hypfrglycemia induced insulin in sham but not in pancreatectomized mice Fig. Likewise, inflammatipn stimulation attenuated hyperglycemia inflammztion sham but not in pancreatectomized mice Fig.

These results show that vagal stimulation can attenuate hyperglycemia in endotoxemia by inducing insulin. Vagal stimulation attenuates hyperglycemia by inducing insulin. Blood glucose levels were analyzed at different time points. bc Mice underwent sham or surgical splenectomy SPX 3 days before LPS.

Hypergglycemia underwent control surgery or vagal stimulation VSand serum TNF at Hylerglycemia. d Fasted mice received sham or vagal stimulation VSand serum insulin levels were analyzed at the indicated time points.

Next, we analyzed how the vagus nerve induces insulin. These results concur with other studies showing that innervations of the adrenal glands modulate hyperglycemia in hemorrhagic shock 35 Hypergoycemia, Thus, we analyzed whether vagal stimulation attenuates hyperglycemia in adrenalectomized endotoxemic mice.

Vagal stimulation attenuated hyperglycemia by around 2-fold in sham but not in adrenalectomized mice Fig. Next, we analyzed whether vagal stimulation induces the production of catecholamines in the adrenal glands. Vagal stimulation induced the production of the three catecholamines: dopamine, norepinephrine, and epinephrine, but the most significant effect was the increase in amd dopamine levels Fig.

Conversely, the most significant effect of adrenalectomy was to attenuate the vagal induction of dopamine Fig. Thus, we analyzed the potential of dopamine to attenuate hyperglycemia in Hyperglycema.

Treatment with dopamine inflmamation hyperglycemia in endotoxemic mice Fig. Given that dopamine has a short chemical half-life span limiting its clinical potential, we analyzed whether fenoldopam, a well-known stable specific agonist for D1-like dopaminergic receptors 3738mimics the potential of dopamine to attenuate hyperglycemia.

Fenoldopam was more effective than dopamine at inhibiting hyperglycemia in endotoxemia Fig. Fenoldopam was also more Hyperglhcemia than dopamine at attenuating serum TNF levels Fig. These results show the potential of dopaminergic agonists to modulate both hyperglycemic and inflammatory responses to bacterial endotoxin.

Dopamine controls hyperglycemia in experimental sepsis. a — c Fasted animals underwent a sham or ac surgical adrenalectomy ADX 3 days before LPS. a Blood glucose or b onflammation, c catecholamines dopamine DAnorepinephrine NE or epinephrine E were analyzed at 1. de Blood glucose and f serum TNF levels were analyzed at the indicated time points.

Blood glucose g and TNF h were analyzed at the indicated time points. We previously reported that the induction of diabetes with streptozotocin, the standard method to induce experimental diabetes in mice, increases hyperglycemic and inflammatory responses to bacterial endotoxin and worsens survival in endotoxemic mice Thus, we analyzed whether fenoldopam attenuates hyperglycemia in experimental sepsis with diabetes.

Time-course Hyperglyxemia indicated that treatment with fenoldopam significantly attenuates hyperglycemia in endotoxemic diabetic mice from 0. These time-course analyses show that fenoldopam inhibited and did not merely delay hyperglycemia or TNF production in endotoxemic mice with diabetes.

These results suggest that dopaminergic agonist type 1, such a fenoldopam may attenuate hyperglycemia and inflammation in both normal and diabetic endotoxemic mice. Our results indicate that metabolic fasting affects both hyperglycemic and systemic inflammatory responses to bacterial endotoxin by modulating vagal neuromodulation.

Fasted animals had worse systemic inflammation, organ function, and survival in experimental sepsis. Thus, we analyzed the relationship between glycemia and the inflammatory response to bacterial endotoxin.

Experimental and clinical Hyperlycemia have shown that sepsis is characterized by an initial hyperglycemic response followed by a hypoglycemic phase and both phases contribute to the pathogenesis and prognosis of sepsis 12131415 Thus, we focused on the initial hyperglycemic response and whether they determine the production of inflammatory factors.

Indeed, administration of glucose activated vagal electric potential and attenuated serum TNF levels in sham but not in vagotomized mice. Although the molecular mechanisms of vagal activation by hyperglycemia are unknown, recent studies show that glucose activates specific neuronal networks in central regions that modulate vagal activity.

These neurons can activate different neuronal networks with specific implications. For instance, defective autophagy in POMC neurons leads to glucose intolerance and Hyoerglycemia 4748whereas disrupted autophagy in glucose-sensing AgRP neurons promotes leanness and reduces food intake 49 ,

: Hyperglycemia and inflammation

Introduction Compared with individuals ibflammation Raspberry planting guide, those Hyperglycemiq DM Balanced meal schedule older mean age 64 vs. Hyperglycemia and dyslipidemia Raspberry planting guide provoke the activation of pro-inflammatory mediators through the involvement of several metabolic pathways. Interleukin-1 receptor antagonist IL-1Ra is naturally occurring anti-inflammatory cytokine of interleukin-1 family. Article CAS Google Scholar Wang, H. eduor Rodica Pop-Busui, rpbusui med.
1 Introduction Article CAS Google Scholar Sakr, Y. Hyperglycemia and inflammation PDF. Neither the over-expression of Inflammatiob nor the Hylerglycemia overload Fall-related injury prevention sodium azide treatment were sufficient to induce inflammation Fig. Results were normalized by cell number. In the presence of high glucose and PPP over-activation, however, NADPH oxidase activity by IL1β was further increased while the protective effect of GSH was diminished.
Background

On the other hand, when stimulated with LPS, there was a significant increase in the secretion of these cytokines. Furthermore, Tessaro et al. Our findings corroborate with previous studies where it was reported that high glucose concentrations 15mM, 25mM do not alter the expression of TNF-α and IL-6 by macrophages, however, when stimulated with LPS, the secretion of these cytokines increase, showing that hyperglycemia it is not a sufficient stimulus for the high production of these cytokines by macrophages 35 , High glucose conditions can cause mitochondrial dysfunction, increase the production of ROS and activates the autophagy pathway 29 , 39 — Furthermore, LC3b protein is involved in mitophagy 42 , a process that removes dysfunctional mitochondria by the autophagic machinery Therefore, we suggest that the high levels of LC3b and beclin-1 expression by ND-BMDM, when compared to D-BMDM identified in this study are due to the normal cellular regulation process in response to stress caused by the hyperglycemic condition, suggesting that the autophagy pathway is impaired in macrophages from diabetic animals, which reinforces that these cells are sensitized when exposed to the hyperglycemic state in vivo 44 — Furthermore, we observed a reduction in LC3b expression by ND-BMDMs and D-BMDMs stimulated and in hyperglycemic conditions.

Our findings corroborate with study that showed a decrease in LC3b expression in THPderived macrophages exposed to high concentrations of glucose with the inflammasome pathway activated However, in this study performed by Dai et al. It was already described that alterations in the autophagy pathway can directly interfere in the inflammatory response of diabetic individuals, making them susceptible to the development of infections 12 49 , Combined with that, nigericin is known to be an inducer of the NLRP3 inflammasome pathway, where the formation of this complex results in the production of IL-1β In our studies, we observed that macrophages from diabetic animals, when stimulated, secreted a greater amount of the cytokine IL-1β, showing that there is an exacerbated production of this cytokine when stimulated by LPS.

Since the relationship between the autophagy process and IL-1β cytokine secretion has been widely studied, it has been reported that this process is responsible for sequestering this cytokine and preventing its secretion 52 , and a negative regulation of this pathway can lead to an increase of IL-1β release 21 , These results suggest that, besides macrophages of diabetic animals being previously sensitized by hyperglycemia, the failure of the autophagy machinery may be contributing to the decompensated secretion of this cytokine.

With the changes observed in our study, we can observe that hyperglycemia plays an essential role in the inflammatory response of BMDMs from diabetic mice, since the high concentration of glucose with LPS stimulation led to significant changes in the secretion of inflammatory mediators and in the autophagy process, having a direct effect on cellular homeostasis.

The high concentration of glucose alters the inflammatory pathways in macrophages after LPS stimulation, disrupting the secretion of pro-inflammatory cytokines by these cells, leading to an impaired inflammatory response against infections.

Furthermore, we observed that the sensitization caused by hyperglycemia in macrophages can downregulate the expression of proteins involved in the autophagy pathway, impairing cellular homeostasis, suggesting the main role of this mechanism over macrophages under diabetic conditions.

The raw data supporting the conclusions of this article will be made available by the authors, without undue reservation. ES and JM conceived and designed the experiments. ES, LQ, JG, KP and RB performed the experiments. ES, LQ, SE and JM analyzed the data.

ES and JM wrote the paper with the assistance and contribution of all the authors. All authors contributed to the article and approved the submitted version. The authors would like to thank Silene Migliorini and Fabiana Teixeira for providing the acquisition, organization of reagents used in this project and assistance at the laboratory.

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers.

Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher. Watanabe S, Alexander M, Misharin AV, Budinger GRS.

The role of macrophages in the resolution of inflammation. J Clin Invest 7 — doi: PubMed Abstract CrossRef Full Text Google Scholar.

Wang Y, Smith W, Hao D, He B, Kong L. M1 and M2 macrophage polarization and potentially therapeutic naturally occurring compounds. Int Immunopharmacol — Thapa B, Lee K.

Metabolic influence on macrophage polarization and pathogenesis. BMB Rep 52 6 — Martin KE, García AJ. Macrophage phenotypes in tissue repair and the foreign body response: Implications for biomaterial-based regenerative medicine strategies.

Acta Biomater — Locati M, Curtale G, Mantovani A. Diversity, mechanisms, and significance of macrophage plasticity. Annu Rev Pathol — Germic N, Frangez Z, Yousefi S, Simon HU. Regulation of the innate immune system by autophagy: monocytes, macrophages, dendritic cells and antigen presentation.

Cell Death Differ 26 4 — Jung S, Jeong H, Yu SW. Autophagy as a decisive process for cell death. Exp Mol Med 52 6 — Wang Y, Li YB, Yin JJ, Wang Y, Zhu LB, Xie GY, et al.

Autophagy regulates inflammation following oxidative injury in diabetes. Autophagy 9 3 —7. Klionsky DJ. Stepping back from the guidelines: Where do we stand?

Autophagy 12 2 —4. Jiang GM, Tan Y, Wang H, Peng L, Chen HT, Meng XJ, et al. The relationship between autophagy and the immune system and its applications for tumor immunotherapy. Mol Cancer 18 1 Deretic V, Kimura T, Timmins G, Moseley P, Chauhan S, Mandell M.

Immunologic manifestations of autophagy. J Clin Invest 1 — Cadwell K. Crosstalk between autophagy and inflammatory signalling pathways: balancing defence and homeostasis.

Nat Rev Immunol 16 11 — Deretic V, Levine B. Autophagy balances inflammation in innate immunity. Autophagy 14 2 — Yin X, Xin H, Mao S, Wu G, Guo L.

The role of autophagy in sepsis: Protection and injury to organs. Front Physiol Lopez-Castejon G, Brough D. Understanding the mechanism of IL-1β secretion. Cytokine Growth Factor Rev 22 4 — Kaushik V, Yakisich JS, Kumar A, Azad N, Iyer AKV. Ionophores: Potential use as anticancer drugs and chemosensitizers.

Cancers Basel 10 10 Gao G, Liu F, Xu Z, Wan D, Han Y, Kuang Y, et al. Evidence of nigericin as a potential therapeutic candidate for cancers: A review. BioMed Pharmacother Wang RC, Wei Y, An Z, Zou Z, Xiao G, Bhagat G, et al.

Akt-mediated regulation of autophagy and tumorigenesis through beclin 1 phosphorylation. Science —9. Yoshii SR, Mizushima N.

Monitoring and measuring autophagy. Int J Mol Sci 18 9 Qiu P, Liu Y, Zhang J. Review: the role and mechanisms of macrophage autophagy in sepsis. Inflammation 42 1 :6— Biasizzo M, Kopitar-Jerala N.

Interplay between NLRP3 inflammasome and autophagy. Front Immunol Wynn TA, Vannella KM. Macrophages in tissue repair, regeneration, and fibrosis. Immunity 44 3 — Kaneko N, Kurata M, Yamamoto T, Morikawa S, Masumoto J.

The role of interleukin-1 in general pathology. Inflammation Regen CrossRef Full Text Google Scholar. Xiu F, Stanojcic M, Diao L, Jeschke MG. Stress hyperglycemia, insulin treatment, and innate immune cells.

Int J Endocrinol Pavlou S, Lindsay J, Ingram R, Xu H, Chen M. Sustained high glucose exposure sensitizes macrophage responses to cytokine stimuli but reduces their phagocytic activity. BMC Immunol 19 1 Edgar L, Akbar N, Braithwaite AT, Krausgruber T, Gallart-Ayala H, Bailey J, et al.

Hyperglycemia induces trained immunity in macrophages and their precursors and promotes atherosclerosis. Circulation 12 — Fiorentino TV, Prioletta A, Zuo P, Folli F.

Hyperglycemia-induced oxidative stress and its role in diabetes mellitus related cardiovascular diseases. Curr Pharm Des 19 32 — Yan LJ. Pathogenesis of chronic hyperglycemia: from reductive stress to oxidative stress. J Diabetes Res Ighodaro OM.

Molecular pathways associated with oxidative stress in diabetes mellitus. BioMed Pharmacother — Ayala TS, Tessaro FHG, Jannuzzi GP, Bella LM, Ferreira KS, Martins JO.

High glucose environments interfere with bone marrow-derived macrophage inflammatory mediator release, the TLR4 pathway and glucose metabolism.

Sci Rep 9 1 Marim FM, Silveira TN, Lima DS Jr, Zamboni DS. A method for generation of bone marrow-derived macrophages from cryopreserved mouse bone marrow cells. PloS One 5 12 :e Carlos D, Costa FR, Pereira CA, Rocha FA, Yaochite JN, Oliveira GG, et al.

Mitochondrial DNA activates the NLRP3 inflammasome and predisposes to type 1 diabetes in murine model. Ramalho T, Filgueiras L, Silva-Jr IA, Pessoa AFM, Jancar S. Impaired wound healing in type 1 diabetes is dependent on 5-lipoxygenase products.

Sci Rep 8 1 Pan Y, Wang Y, Cai L, Cai Y, Hu J, Yu C, et al. Inhibition of high glucose-induced inflammatory response and macrophage infiltration by a novel curcumin derivative prevents renal injury in diabetic rats.

Br J Pharmacol 3 — Torres-Castro I, Arroyo-Camarena ÚD, Martínez-Reyes CP, Gómez-Arauz AY, Dueñas-Andrade Y, Hernández-Ruiz J, et al. Human monocytes and macrophages undergo M1-type inflammatory polarization in response to high levels of glucose.

Immunol Lett —9. Cheng CI, Chen PH, Lin YC, Kao YH. High glucose activates Raw Cell Signal 27 2 — Tessaro GFH, Ayala TS, Bella LM, Martins JO. Immunobiology 2 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. Annu Rev Physiol. Shah PK. Innate immune pathway links obesity to insulin resistance. Circ Res. Schenk S, Saberi M, Olefsky JM.

Insulin sensitivity: modulation by nutrients and inflammation. Kim JJ, Sears DD. TLR4 and Insulin Resistance. Gastroenterology Research and Practice. Huang H, Park PH, McMullen MR, Nagy LE.

Mechanisms for the anti-inflammatory effects of adiponectin in macrophages. J Gastroenterol Hepatol. Mandal P, Park PH, McMullen MR, Pratt BT, Nagy LE. The anti-inflammatory effects of adiponectin are mediated via a heme oxygenasedependent pathway in rat Kupffer cells. Yamaguchi N, Argueta JG, Masuhiro Y, Kagishita M, Nonaka K, Saito T, Hanazawa S, Yamashita Y.

Adiponectin inhibits Toll-like receptor family-induced signaling. FEBS Lett. Jones LA, Anthony JP, Henriquez FL, Lyons RE, Nickdel MB, Carter KC, Alexander J, Roberts CW. Toll-like receptormediated macrophage activation is differentially regulated by progesterone via the glucocorticoid and progesterone receptors.

Reynolds CM, Draper E, Keogh B, Rahman A, Moloney AP, Mills KH, Loscher CE, Roche HM. A conjugated linoleic acid-enriched beef diet attenuates lipopolysaccharide-induced inflammation in mice in part through PPARgamma-mediated suppression of toll-like receptor 4.

J Nutr. Hotamisligil GS, Erbay E. Nutrient sensing and inflammation in metabolic diseases. Coppari R, Ichinose M, Lee CE, Pullen AE, Kenny CD, McGovern RA, Tang V, Liu SM, Ludwig T, Chua Jr SC, Lowell BB, Elmquist JK. Cell Metab. Pocai A, Morgan K, Buettner C, Gutierrez-Juarez R, Obici S, Rossetti L.

Central leptin acutely reverses diet-induced hepatic insulin resistance. Galic S, Oakhill JS, Steinberg GR. Lin Y, Lee H, Berg AH, Lisanti MP, Shapiro L, Scherer PE. The lipopolysaccharide-activated toll-like receptor TLR -4 induces synthesis of the closely related receptor TLR-2 in adipocytes.

Shi H, Kokoeva MV, Inouye K, Tzameli I, Yin H, Flier JS. TLR4 links innate immunity and fatty acid-induced insulin resistance. Poggi M, Bastelica D, Gual P, Iglesias MA, Gremeaux T, Knauf C, Peiretti F, Verdier M, Juhan-Vague I, Tanti JF, Burcelin R, Alessi MC.

Saberi M, Woods NB, de Luca C, Schenk S, Lu JC, Bandyopadhyay G, Verma IM, Olefsky JM. Hematopoietic cell-specific deletion of toll-like receptor 4 ameliorates hepatic and adipose tissue insulin resistance in high-fat-fed mice.

Suganami T, Mieda T, Itoh M, Shimoda Y, Kamei Y, Ogawa Y. Biochem Biophys Res Commun. Tsukumo DM, Carvalho-Filho MA, Carvalheira JB, Prada PO, Hirabara SM, Schenka AA, Araujo EP, Vassallo J, Curi R, Velloso LA, Saad MJ.

Loss-of-function mutation in Toll-like receptor 4 prevents diet-induced obesity and insulin resistance. Davis JE, Gabler NK, Walker-Daniels J, Spurlock ME. Tlr-4 deficiency selectively protects against obesity induced by diets high in saturated fat. Obesity Silver Spring, Md.

Baron AD, Brechtel G, Wallace P, Edelman SV. Rates and tissue sites of non-insulin- and insulin-mediated glucose uptake in humans. Am J Physiol. Wei Y, Chen K, Whaley-Connell AT, Stump CS, Ibdah JA, Sowers JR. Skeletal muscle insulin resistance: role of inflammatory cytokines and reactive oxygen species.

American journal of physiology. Regul Integr Comp Physiol. Baffy G. Kupffer cells in non-alcoholic fatty liver disease: the emerging view.

J Hepatol. Spruss A, Kanuri G, Wagnerberger S, Haub S, Bischoff SC, Bergheim I. Toll-like receptor 4 is involved in the development of fructose-induced hepatic steatosis in mice. Videla LA, Tapia G, Rodrigo R, Pettinelli P, Haim D, Santibanez C, Araya AV, Smok G, Csendes A, Gutierrez L, Rojas J, Castillo J, Korn O, Maluenda F, Diaz JC, Rencoret G, Poniachik J.

Liver NF-kappaB and AP-1 DNA binding in obese patients. Raetzsch CF, Brooks NL, Alderman JM, Moore KS, Hosick PA, Klebanov S, Akira S, Bear JE, Baldwin AS, Mackman N, Combs TP.

Lipopolysaccharide inhibition of glucose production through the Toll-like receptor-4, myeloid differentiation factor 88, and nuclear factor kappa b pathway. Wen L, Peng J, Li Z, Wong FS. The effect of innate immunity on autoimmune diabetes and the expression of Toll-like receptors on pancreatic islets.

Top bar navigation Article CAS Google Hyperglycemia and inflammation Jordan, S. Hyperglycemia and inflammation Hyperglycemja still studying the connection between Hypergkycemia like hyperglycemia, diabetes, aging, and inflammation, like in this study by the American Diabetes Association ADA. American Diabetes Association: Diagnosis and classification of diabetes mellitus Position Statement. Letter to the editor: COVID in patients with diabetes: risk factors that increase morbidity. Hotchkiss, R.
Cardiovascular Diabetology Insulin sensitivity and insulin sensitivity factor value 15 inflmmation, Article number: 82 Cite Hyperglyfemia article. Lifestyle factors affecting blood sugar levels details. An Erratum to this article was published on 16 Inclammation Raspberry planting guide is Hyperglcyemia as Raspberry planting guide pro-inflammatory condition and a major cause of vascular damage. Nevertheless, we have previously described that high glucose only promotes inflammation in human vascular cells previously primed with pro-inflammatory stimuli, such as the cytokine interleukin IL 1β. Here, we aimed to identify the cellular mechanisms by which high glucose exacerbates the vascular inflammation induced by IL1β.

Hyperglycemia and inflammation -

The role of autophagy in the regulation of inflammation during infectious and non-infectious diseases is already well established 11 , 12 , being described that the negative regulation of this process interferes in the inflammatory responses to diseases 13 , It is known that Nigericin, an ionophore derived from Streptomyces Hygroscopicus combined with LPS induces the secretion of IL-1β through the induction of NLRP3 inflammasome, a component of the innate immune system 15 — Besides being known as a survival mechanism for cells under stress conditions, studies indicates that autophagy may contribute to cell death processes under pathological conditions, increasing the inflammatory response 18 — 20 , being known that a failure on the autophagy machinery can lead to an increase on IL-1β secretion This pro-inflammatory cytokine is central on the inflammatory response, where a sustained secretion of IL-1β can drives to a persistent inflammatory state and consequent tissue injury 22 , Macrophages are influenced by hyperglycemia 18 , Long-term hyperglycemic states sensitize macrophages to increase the secretion of cytokines, reducing their phagocytic activity and NO production 19 , 25 , and BMDM from diabetic mice showed an increased proinflammatory gene expression, even when cultured in normal glucose conditions, suggesting a priming effect driven by hyperglycemia 20 , Type 1 diabetes mellitus T1DM is a disease caused by the autoimmune destruction of insulin-producing beta cells, which directly interferes in glucose metabolism, leading to a hyperglycemic state.

The hyperglycemia presented in diabetic individuals has several deleterious effects, which may occur due to the activation of pathways that stimulate ROS production, which in the absence of an appropriate antioxidant defense mechanism, promotes the activation of intracellular stress-sensitive pathways, causing damage to cells and consequent development of complications related to the pathogenesis of the disease 21 — 23 , 27 — Thus, in this study, we investigated the influence of hyperglycemia on the inflammatory response in vitro of macrophages stimulated with LPS and nigericin, in a hyperglycemic environment, and its effect on the autophagy process.

Artificial devices were placed in the cages to enrich the environment cotton, igloo, cardboard tubes etc. Louis, Missouri, USA was administered intravenously, with saline solution administered.

On non-diabetic animals, only saline solution was administered 24 , On day 0, blood glucose was measured using a glucometer Accu-Chek Advantage II Roche Diagnostics, São Paulo, SP, Brazil and the animals where weighed. After 10 days of induction, weight and blood glucose were measured again to confirm the induction of diabetes.

Cell counts were performed with trypan blue Gibco ® by Life Technologies, Thermo Fisher Scientific, Waltham, Massachusetts, USA. After priming, cells were cultured at a standard concentration 5. We used RIPA lysis buffer to lysate the BMDM. For protein transfer to the nitrocellulose membrane we used a semi-dry system using transfer buffer 25 mM Tris; mM glycine; 0.

The revelation was performed using ECL enhanced chemiluminescence - ECL; Amersham, Arlington Heights, IL on the Amersham Imager blot and gel imager equipment Amersham, Buckinghamshire, United Kingdom. The primary anti-mouse LC3B and anti-mouse Beclin-1 antibodies purchased from Cell Signaling Technology Cell Signaling Technology ® , Danvers, Massachusetts, USA were used to detect the target proteins.

Relative band densities were determined by densitometric analysis using ImageJ software. The results were evaluated by analysis of variance two-way ANOVA followed by the Tukey-Kramer multiple comparisons test using GraphPad software 9. To obtain BMDM from a hyperglycemic environment, we used an alloxan T1DM animal model.

After 10 days of induction with alloxan, when compared to the control group, the animals showed a loss of body weight Figure 1 , and an increase in blood glucose Figure 2. Figure 2 Evolution of blood glucose after induction of diabetes: Diabetic animals showed an increase in blood glucose after 10 days of induction with the diabetogenic agent.

To assess the autophagy pathway in BMDM, the protein expression of LC3B and Beclin-1 were evaluated by the Western Blotting method, after 30 minutes Figure 3A. We identified that ND-BMDM had an increase in Beclin-1 expression in hyperglycemic medium without stimulation Figure 3B.

Figure 3 Evaluation of the autophagy pathway. Results represent mean ± SEM. After 2, 4, 6 and 24 hours of incubation, BMDM from diabetic and non-diabetic animals released a more significant amount of IL-6 at concentrations of 5. At 6 hours, we observed that D-BMDM increased the secretion of this cytokine in hyperglycemic medium with stimulation Figure 4D.

Figure 4 IL-6 secretion by ND-BMDM and D-BMDM with Nigericin priming and LPS stimulation. IL-6 release after A 30 minutes with different glucose concentrations. B 2 hours with different glucose concentrations.

C 4 hours with different glucose concentrations. D 6 hours with different glucose concentrations. E 24 hours with different glucose concentrations.

IL-6 measurement was performed by enzyme-linked immunosorbent assay. Cytokine measurements were performed to measure the secretion of TNF-α. After 30 minutes, 2, 4, 6, and 24 hours of incubation, BMDM from diabetic and non-diabetic animals released higher levels of TNF-α at concentrations of 5.

Figure 5 TNF-α secretion by ND-BMDM and D-BMDM with Nigericin priming and LPS stimulation. TNF-α measurement was performed by enzyme-linked immunosorbent assay.

It was observed that after 30 minutes, 2, 4, 6 and 24 hours of incubation, BMDM from diabetic and non-diabetic animals released a more significant amount of IL-1β at concentrations of 5. At 30 minutes, D-BMDM with stimulation secreted IL-1β more than ND-BMDM under the same conditions Figure 6A.

At 4 and 24 hours, D-BMDM in hyperglycemic medium with stimulation had an increase in the secretion of this cytokine when compared with ND-BMDM in the same condition Figures 6C and E.

We also assessed Nitric Oxide and IL levels, but no significant differences were observed Supplementary Figures 1 and 2. Figure 6 IL-1β secretion by ND-BMDM and D-BMDM with Nigericin priming and LPS stimulation.

IL-1β release after A 30 minutes with different glucose concentrations. IL-1β measurement was performed by enzyme-linked immunosorbent assay. It is already described that patients can establish a chronic inflammation state under diabetic conditions, characterized by a decompensated secretion of pro-inflammatory cytokines, such as TNF-α, IL-1β and IL-6, and it is suggested as the major cause of comorbidities related to diabetes Furthermore, it has been identified that peritoneal macrophages, when exposed to high levels of glucose secrete greater amounts of TNF-α, IL-1β, IL-6 and IL in response to high glucose concentrations 34 , It has also been reported by Cheng et al.

A study showed that, when cultivated in a hyperglycemic medium, BMDM secreted a more significant amount of TNF-α, but the expression of IL-6 under the same conditions was reduced However, our studies observed that high glucose levels alone were not enough to stimulate the secretion of TNF-α, IL-1β and IL-6 by macrophages.

On the other hand, when stimulated with LPS, there was a significant increase in the secretion of these cytokines. Furthermore, Tessaro et al. Our findings corroborate with previous studies where it was reported that high glucose concentrations 15mM, 25mM do not alter the expression of TNF-α and IL-6 by macrophages, however, when stimulated with LPS, the secretion of these cytokines increase, showing that hyperglycemia it is not a sufficient stimulus for the high production of these cytokines by macrophages 35 , High glucose conditions can cause mitochondrial dysfunction, increase the production of ROS and activates the autophagy pathway 29 , 39 — Furthermore, LC3b protein is involved in mitophagy 42 , a process that removes dysfunctional mitochondria by the autophagic machinery Therefore, we suggest that the high levels of LC3b and beclin-1 expression by ND-BMDM, when compared to D-BMDM identified in this study are due to the normal cellular regulation process in response to stress caused by the hyperglycemic condition, suggesting that the autophagy pathway is impaired in macrophages from diabetic animals, which reinforces that these cells are sensitized when exposed to the hyperglycemic state in vivo 44 — Furthermore, we observed a reduction in LC3b expression by ND-BMDMs and D-BMDMs stimulated and in hyperglycemic conditions.

Our findings corroborate with study that showed a decrease in LC3b expression in THPderived macrophages exposed to high concentrations of glucose with the inflammasome pathway activated However, in this study performed by Dai et al. It was already described that alterations in the autophagy pathway can directly interfere in the inflammatory response of diabetic individuals, making them susceptible to the development of infections 12 49 , Combined with that, nigericin is known to be an inducer of the NLRP3 inflammasome pathway, where the formation of this complex results in the production of IL-1β In our studies, we observed that macrophages from diabetic animals, when stimulated, secreted a greater amount of the cytokine IL-1β, showing that there is an exacerbated production of this cytokine when stimulated by LPS.

Since the relationship between the autophagy process and IL-1β cytokine secretion has been widely studied, it has been reported that this process is responsible for sequestering this cytokine and preventing its secretion 52 , and a negative regulation of this pathway can lead to an increase of IL-1β release 21 , These results suggest that, besides macrophages of diabetic animals being previously sensitized by hyperglycemia, the failure of the autophagy machinery may be contributing to the decompensated secretion of this cytokine.

With the changes observed in our study, we can observe that hyperglycemia plays an essential role in the inflammatory response of BMDMs from diabetic mice, since the high concentration of glucose with LPS stimulation led to significant changes in the secretion of inflammatory mediators and in the autophagy process, having a direct effect on cellular homeostasis.

The high concentration of glucose alters the inflammatory pathways in macrophages after LPS stimulation, disrupting the secretion of pro-inflammatory cytokines by these cells, leading to an impaired inflammatory response against infections. Furthermore, we observed that the sensitization caused by hyperglycemia in macrophages can downregulate the expression of proteins involved in the autophagy pathway, impairing cellular homeostasis, suggesting the main role of this mechanism over macrophages under diabetic conditions.

The raw data supporting the conclusions of this article will be made available by the authors, without undue reservation. ES and JM conceived and designed the experiments. ES, LQ, JG, KP and RB performed the experiments. ES, LQ, SE and JM analyzed the data.

ES and JM wrote the paper with the assistance and contribution of all the authors. All authors contributed to the article and approved the submitted version.

The authors would like to thank Silene Migliorini and Fabiana Teixeira for providing the acquisition, organization of reagents used in this project and assistance at the laboratory. The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers.

Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher. Watanabe S, Alexander M, Misharin AV, Budinger GRS. The role of macrophages in the resolution of inflammation.

J Clin Invest 7 — doi: PubMed Abstract CrossRef Full Text Google Scholar. Wang Y, Smith W, Hao D, He B, Kong L. M1 and M2 macrophage polarization and potentially therapeutic naturally occurring compounds. Int Immunopharmacol — Thapa B, Lee K.

Metabolic influence on macrophage polarization and pathogenesis. BMB Rep 52 6 — Martin KE, García AJ. Macrophage phenotypes in tissue repair and the foreign body response: Implications for biomaterial-based regenerative medicine strategies.

Acta Biomater — Locati M, Curtale G, Mantovani A. Diversity, mechanisms, and significance of macrophage plasticity. Annu Rev Pathol — Germic N, Frangez Z, Yousefi S, Simon HU.

Regulation of the innate immune system by autophagy: monocytes, macrophages, dendritic cells and antigen presentation.

Cell Death Differ 26 4 — Jung S, Jeong H, Yu SW. Autophagy as a decisive process for cell death. Exp Mol Med 52 6 — Wang Y, Li YB, Yin JJ, Wang Y, Zhu LB, Xie GY, et al. Autophagy regulates inflammation following oxidative injury in diabetes.

Autophagy 9 3 —7. Klionsky DJ. Stepping back from the guidelines: Where do we stand? Autophagy 12 2 —4. Jiang GM, Tan Y, Wang H, Peng L, Chen HT, Meng XJ, et al.

The relationship between autophagy and the immune system and its applications for tumor immunotherapy. Mol Cancer 18 1 Deretic V, Kimura T, Timmins G, Moseley P, Chauhan S, Mandell M.

Immunologic manifestations of autophagy. J Clin Invest 1 — Cadwell K. Crosstalk between autophagy and inflammatory signalling pathways: balancing defence and homeostasis. Nat Rev Immunol 16 11 — Deretic V, Levine B.

Autophagy balances inflammation in innate immunity. Autophagy 14 2 — Yin X, Xin H, Mao S, Wu G, Guo L. The role of autophagy in sepsis: Protection and injury to organs. Front Physiol Lopez-Castejon G, Brough D.

Understanding the mechanism of IL-1β secretion. Cytokine Growth Factor Rev 22 4 — Kaushik V, Yakisich JS, Kumar A, Azad N, Iyer AKV. Ionophores: Potential use as anticancer drugs and chemosensitizers.

Cancers Basel 10 10 Gao G, Liu F, Xu Z, Wan D, Han Y, Kuang Y, et al. Evidence of nigericin as a potential therapeutic candidate for cancers: A review. BioMed Pharmacother Wang RC, Wei Y, An Z, Zou Z, Xiao G, Bhagat G, et al.

Akt-mediated regulation of autophagy and tumorigenesis through beclin 1 phosphorylation. Science —9. Yoshii SR, Mizushima N. Monitoring and measuring autophagy. By contrast, vagal electrical stimulation attenuated hyperglycemia in both sham and splenectomized mice Fig.

Next, we wondered whether the vagus nerve controls hyperglycemia by inducing insulin. Vagal stimulation increased serum insulin levels Fig.

We further confirmed our results in pancreatectomized mice. Vagal stimulation induced insulin in sham but not in pancreatectomized mice Fig.

Likewise, vagal stimulation attenuated hyperglycemia in sham but not in pancreatectomized mice Fig. These results show that vagal stimulation can attenuate hyperglycemia in endotoxemia by inducing insulin. Vagal stimulation attenuates hyperglycemia by inducing insulin.

Blood glucose levels were analyzed at different time points. b , c Mice underwent sham or surgical splenectomy SPX 3 days before LPS. Animals underwent control surgery or vagal stimulation VS , and serum TNF at 1.

d Fasted mice received sham or vagal stimulation VS , and serum insulin levels were analyzed at the indicated time points. Next, we analyzed how the vagus nerve induces insulin.

These results concur with other studies showing that innervations of the adrenal glands modulate hyperglycemia in hemorrhagic shock 35 , Thus, we analyzed whether vagal stimulation attenuates hyperglycemia in adrenalectomized endotoxemic mice. Vagal stimulation attenuated hyperglycemia by around 2-fold in sham but not in adrenalectomized mice Fig.

Next, we analyzed whether vagal stimulation induces the production of catecholamines in the adrenal glands. Vagal stimulation induced the production of the three catecholamines: dopamine, norepinephrine, and epinephrine, but the most significant effect was the increase in blood dopamine levels Fig.

Conversely, the most significant effect of adrenalectomy was to attenuate the vagal induction of dopamine Fig. Thus, we analyzed the potential of dopamine to attenuate hyperglycemia in endotoxemia. Treatment with dopamine attenuated hyperglycemia in endotoxemic mice Fig.

Given that dopamine has a short chemical half-life span limiting its clinical potential, we analyzed whether fenoldopam, a well-known stable specific agonist for D1-like dopaminergic receptors 37 , 38 , mimics the potential of dopamine to attenuate hyperglycemia.

Fenoldopam was more effective than dopamine at inhibiting hyperglycemia in endotoxemia Fig. Fenoldopam was also more effective than dopamine at attenuating serum TNF levels Fig. These results show the potential of dopaminergic agonists to modulate both hyperglycemic and inflammatory responses to bacterial endotoxin.

Dopamine controls hyperglycemia in experimental sepsis. a — c Fasted animals underwent a sham or a , c surgical adrenalectomy ADX 3 days before LPS. a Blood glucose or b , c catecholamines dopamine DA , norepinephrine NE or epinephrine E were analyzed at 1.

d , e Blood glucose and f serum TNF levels were analyzed at the indicated time points. Blood glucose g and TNF h were analyzed at the indicated time points.

We previously reported that the induction of diabetes with streptozotocin, the standard method to induce experimental diabetes in mice, increases hyperglycemic and inflammatory responses to bacterial endotoxin and worsens survival in endotoxemic mice Thus, we analyzed whether fenoldopam attenuates hyperglycemia in experimental sepsis with diabetes.

Time-course experiments indicated that treatment with fenoldopam significantly attenuates hyperglycemia in endotoxemic diabetic mice from 0. These time-course analyses show that fenoldopam inhibited and did not merely delay hyperglycemia or TNF production in endotoxemic mice with diabetes.

These results suggest that dopaminergic agonist type 1, such a fenoldopam may attenuate hyperglycemia and inflammation in both normal and diabetic endotoxemic mice. Our results indicate that metabolic fasting affects both hyperglycemic and systemic inflammatory responses to bacterial endotoxin by modulating vagal neuromodulation.

Fasted animals had worse systemic inflammation, organ function, and survival in experimental sepsis. Thus, we analyzed the relationship between glycemia and the inflammatory response to bacterial endotoxin.

Experimental and clinical studies have shown that sepsis is characterized by an initial hyperglycemic response followed by a hypoglycemic phase and both phases contribute to the pathogenesis and prognosis of sepsis 12 , 13 , 14 , 15 , Thus, we focused on the initial hyperglycemic response and whether they determine the production of inflammatory factors.

Indeed, administration of glucose activated vagal electric potential and attenuated serum TNF levels in sham but not in vagotomized mice. Although the molecular mechanisms of vagal activation by hyperglycemia are unknown, recent studies show that glucose activates specific neuronal networks in central regions that modulate vagal activity.

These neurons can activate different neuronal networks with specific implications. For instance, defective autophagy in POMC neurons leads to glucose intolerance and obesity 47 , 48 , whereas disrupted autophagy in glucose-sensing AgRP neurons promotes leanness and reduces food intake 49 , Although recent studies suggested that glucose transporters and hexokinase can contribute to neuronal activation by glucose, the molecular mechanisms are not well-known 51 , 52 , Furthermore, glucose can also activate astrocytes, which in turn can activate central neuronal networks through the astrocyte-neuron lactate shuttle 46 and the neuronal pyruvate metabolism These studies focused on the potential of glucose to activate central neuronal networks that regulate energy homeostasis and hyperglycemia.

Our results now warrant similar studies to determine the specific neurons and networks activated by glucose to induce vagal control of hyperglycemia and inflammation in infectious disorders.

Future studies will also determine how these networks may link metabolic and immunological disorders and their contribution to inflammatory or infectious disorders such as sepsis. Administration of glucose activated vagal electrical potential without affecting arterial blood pressure.

Semapimod also activated vagal electrical potential and attenuated serum TNF levels in endotoxemia without affecting arterial blood pressure Likewise, melanocortin peptides, cholecystokinin, ghrelin, and leptin can also activate the vagus nerve to control physiological homeostasis 55 , 56 , These mechanisms of vagal activation have multiple clinical implications.

For instance, deletion of leptin-induced vagal activation causes hyperphagia, obesity, diabetes, and infertility 56 , Although these studies focused on vagal regulation of serum TNF levels, our present study now suggests that vagal regulation of hyperglycemia can also contribute to the effects of the vagus nerve in these conditions.

In line with the hypothesis, our recent clinical study shows that neuronal stimulation can improve postoperative recovery by preventing hyperglycemia Our results indicate that efferent vagal stimulation attenuates hyperglycemia in endotoxemia by inducing insulin and regardless of TNF regulation.

Although neuronal regulation of insulin secretion is a complex process depending on the physiological conditions, our results concur with previous studies indicating that the vagus nerve can modulate cephalic and postprandial insulin secretion 29 , 30 , From a clinical perspective, patients with complete resection of the subdiaphragmatic vagus nerve can display a longer periodicity of plasma insulin oscillations Clinical studies on patients with subdiaphragmatic vagotomy also indicate that truncal vagotomy decreases the glucagon response to insulin hypoglycemia as compared to selective vagotomy 66 , and abdominal vagal blocking decreases pancreatic exocrine secretion in the animal model Furthermore, recent studies indicate that vagal blocking can improve glycemic control and blood pressure in obese patients with type 2 diabetes mellitus In agreement with these results, previous physiological studies showed that innervations of the adrenal glands modulate hyperglycemia in hemorrhagic shock 35 , Our current results show that efferent vagal stimulation induced insulin and adrenalectomy prevented vagal control of hyperglycemia in endotoxemia.

Our study shows that vagal stimulation induces the production of dopamine from the adrenal glands. Administration of dopamine decreased hyperglycemia and serum TNF levels in endotoxemia. These results concur with clinical studies showing that dopamine can restore tissue perfusion in septic shock and critically ill patients 69 , However, dopamine can also increase the risk of tachyarrhythmia 69 , 70 and worsens survival in septic animals We hypothesized that selective dopaminergic agonists may avoid unspecific side effects of dopamine.

Dopaminergic receptors are classified into D1-like D1R, D5R or D2-like D2R, D3R, D4R receptors that signal through Gαs or Gαi proteins, respectively. Fenoldopam is a well-characterized, stable and specific agonist with ~fold greater affinity for D1Rs than dopamine 35 , 36 , 37 , Fenoldopam was more effective than dopamine at controlling hyperglycemia and TNF production in endotoxemic mice.

We previously reported that experimental diabetes increases hyperglycemic and inflammatory responses to bacterial endotoxin and worsens survival in endotoxemic mice Our present results also suggest that fenoldopam attenuates hyperglycemia before the serum TNF peak at 1. Future studies will be required to determine whether fenoldopam control of hyperglycemia contributes to modulate serum TNF levels.

By comparison, inhibition of TNF by using neutralizing anti-TNF antibodies increased the mortality when administered after the septic challenge in normal mice 9. These studies of neuromodulation are allowing the design of novel treatments for infectious and inflammatory disorders.

The study of the sympathetic baroreflex system allowed the design of selective beta-blockers for hypertension and arrhythmia. Similar studies are now required to determine whether specific dopaminergic agonists may provide therapeutic advantages for hyperglycemia and inflammation in septic patients with preexisting metabolic conditions such as diabetes.

LPS E. Coli B4 , dopamine hydrochloride, fenoldopam, streptozotocin, and glucose were purchased from Sigma-Aldrich® Saint Louis, MO. The glucose measuring strips were purchased from PharmaTech Solutions, Inc Westlake Village, CA. Pentobarbital sodium was purchased from Diamondback Scottsdale, AZ ; ketamine from Henry Schein animal health Dublin, OH ; xylazine from Akron animal health Lake Forest, IL, USA and enrofloxacin from Bayer Healthcare Shawnee Mission, KS, USA.

Mice were fed standard chow diet Louis, MO. The investigators analyzing the samples were blinded to the treatments. and vagal surgeries performed as we described Surgical Cervical Vagotomy VGX A ventral incision on the neck was performed to retract the sternocleidomastoid muscle and to visualize the carotid artery and vagus nerves.

The vagal trunks were ligated with size 4—0 silk and sectioned. Vagal Stimulation VS The right cervical vagal trunk was isolated and connected to the platinum electrode. Control animals underwent sham surgery without the electrical stimulation.

Vagal activity recording was performed as described Anesthetized Wistar rats received ventral neck incision to connect the vagus nerve to the electrode. Then, the carotid artery and femoral vein were catheterized for recording pulsatile arterial pressure and for glucose administration iv , respectively.

Animals were allowed to recover consciousness after surgery. The arterial catheter was connected to a pressure transducer MLT and the signal was amplified by ML ADInstruments, Bella Vista, Australia and sampled in the oscilloscope Tektronics Anesthesia was confirmed by the absence of withdrawal reflex to toe pinch.

Antibiotics Enrofloxacine 2. Splenectomy: was performed 3 days prior to the experiment as we described in J Exp Med Animals underwent an abdominal incision to expose the spleen. The three main branches of the splenic artery were stabilized with nylon thread, ligated and cut.

The spleen was removed and the wound was closed with sutures with catgut for the abdominal wall, and nylon thread for the skin. Adrenalectomy: was performed as described Animals received a dorsal incision from the first to the third lumbar vertebrae.

The latissimus dorsi muscle was retracted and both adrenal glands and their adipose tissue were removed. Pancreatectomy : was performed similarly to that previously described TNF, IL6, IL10, TGFβ1 and IFNγ were analyzed with the respective ELISA kit Affymettrix Inc, San Diego, CA.

Serum HMGB1 was analyzed using HMGB1 detection ELISA kit Chondrex Inc. Blood catecholamines were analyzed by ELISA LDN immunoassays and services, Germany at 1. Glucose was analyzed from the mouse tail tip blood using the Genstrip PharmaTech Solutions Inc.

Serum insulin was analyzed with the Mercodia mouse insulin ELISA kit, Mercodia AB, Uppsala, Sweden. Tests were performed with GraphPad Prism Software® GraphPad Software, La Jolla, CA.

The sample size was determined with power analyses of our previous studies 25 , Analyses of three or more groups were performed with one-way ANOVA with multiple pair-wise comparisons. The time courses and pair-wise comparisons were analyzed with the two-way ANOVA for repeated measures.

Normality and homogeneity of variance were confirmed with Kolmogorov-Smirnov analyses. Survival was analyzed with the Log-rank Mantel-Cox test. Angus, D. et al. Epidemiology of severe sepsis in the United States: analysis of incidence, outcome, and associated costs of care.

Critical care medicine 29 , — Article CAS Google Scholar. Hotchkiss, R. The pathophysiology and treatment of sepsis. The New England journal of medicine , — Ulloa, L. Trends Mol Med 11 , 56—63 Rice, T. Therapeutic intervention and targets for sepsis. Annu Rev Med 56 , — Martin, G.

The epidemiology of sepsis in the United States from through Article Google Scholar. Tracey, K. Nature , — Article CAS ADS Google Scholar.

Marshall, J. Why have clinical trials in sepsis failed? Trends Mol Med 20 , — Carré, J. Cellular energetic metabolism in sepsis: The need for a systems approach. Biochimica et Biophysica Acta BBA - Bioenergetics , — Eskandari, M. Anti-tumor necrosis factor antibody therapy fails to prevent lethality after cecal ligation and puncture or endotoxemia.

J Immunol , — CAS PubMed Google Scholar. Brealey, D. Association between mitochondrial dysfunction and severity and outcome of septic shock.

Lancet London, England , — Wolfe, R. Energy metabolism in trauma and sepsis: the role of fat. Progress in clinical and biological research , 89— Machado, F.

The epidemiology of sepsis in Brazilian intensive care units the Sepsis PREvalence Assessment Database, SPREAD : an observational study. The Lancet Infectious Diseases 17 , — Cerra, F. Hypermetabolism-organ failure syndrome: a metabolic response to injury. Critical care clinics 5 , — van Vught, L.

Diabetes Is Not Associated With Increased Day Mortality Risk in Critically Ill Patients With Sepsis. Critical care medicine 45 , e—e Finfer, S. Intensive versus conventional glucose control in critically ill patients.

Novosad, S. Vital Signs: Epidemiology of Sepsis: Prevalence of Health Care Factors and Opportunities for Prevention. MMWR Morb Mortal Wkly Rep 65 , — Kosteli, A. Weight loss and lipolysis promote a dynamic immune response in murine adipose tissue. The Journal of Clinical Investigation , — Weisberg, S.

CCR2 modulates inflammatory and metabolic effects of high-fat feeding. J Clin Invest , — Arkan, M. IKK-beta links inflammation to obesity-induced insulin resistance. Nat Med 11 , — Solinas, G. JNK1 in hematopoietically derived cells contributes to diet-induced inflammation and insulin resistance without affecting obesity.

Cell metabolism 6 , — Buras, J. Animal models of sepsis: setting the stage. Nat Rev Drug Discov 4 , — Young, H. Eating disinhibition and vagal tone moderate the postprandial response to glycemic load: a randomised controlled trial.

Scientific reports 6 , Vida, G. β2-Adrenoreceptors of regulatory lymphocytes are essential for vagal neuromodulation of the innate immune system. FASEB J 25 , — Wang, H. Nicotinic acetylcholine receptor alpha 7 subunit is an essential regulator of inflammation.

Huston, J. Splenectomy inactivates the cholinergic antiinflammatory pathway during lethal endotoxemia and polymicrobial sepsis. J Exp Med , — Borovikova, L. Vagus nerve stimulation attenuates the systemic inflammatory response to endotoxin.

Nerve Stimulation: Immunomodulation and Control of Inflammation. Trends Mol Med 23 , — Kanashiro, A. From Neuroimunomodulation To Bioelectronic Treatment Of RheumatoidArthritis. Bioelectronic Med 1 , 13—17 Ahren, B. The cephalic insulin response to meal ingestion in humans is dependent on both cholinergic and noncholinergic mechanisms and is important for postprandial glycemia.

Diabetes 50 , — Li, Y. High-affinity CCK-A receptors on the vagus nerve mediate CCK-stimulated pancreatic secretion in rats. The American journal of physiology , G— Niijima, A. Visceral afferents and metabolic function.

Diabetologia 20 Suppl , — Travagli, R. Vagal neurocircuitry and its influence on gastric motility. Nature reviews. Glucose-sensitive afferent nerve fibers in the liver and their role in food intake and blood glucose regulation. J Auton Nerv Syst 9 , — Yamaguchi, N. Sympathoadrenal system in neuroendocrine control of glucose: mechanisms involved in the liver, pancreas, and adrenal gland under hemorrhagic and hypoglycemic stress.

Can J Physiol Pharmacol 70 , — Tiberi, M. High agonist-independent activity is a distinguishing feature of the dopamine D1B receptor subtype.

The Journal of biological chemistry , — Balbo, S. Vagotomy diminishes obesity in cafeteria rats by decreasing cholinergic potentiation of insulin release. J Physiol Biochem 72 , — Grenader, A. Fenoldopam is a partial agonist at dopamine-1 DA1 receptors in LLC-PK1 cells. J Pharmacol Exp Ther , — Torres-Rosas, R.

Dopamine mediates vagal modulation of the immune system by electroacupuncture. Nat Med 20 , — Feketeova, E. Dopaminergic Control of Inflammation and Glycemia in Sepsis and Diabetes.

Front Immunol 9 , Weis, S. Metabolic Adaptation Establishes Disease Tolerance to Sepsis. Cell , — e Kong, D. Glucose stimulation of hypothalamic MCH neurons involves K ATP channels, is modulated by UCP2, and regulates peripheral glucose homeostasis.

Cell metabolism 12 , — Grill, H. Hindbrain neurons as an essential hub in the neuroanatomically distributed control of energy balance. Cell metabolism 16 , — Mergenthaler, P. Sugar for the brain: the role of glucose in physiological and pathological brain function.

Trends in neurosciences 36 , — Ruud, J. Neuronal control of peripheral insulin sensitivity and glucose metabolism. Nature Communications 8 , Article ADS Google Scholar.

Udit, S. Mol Metab 6 , — Lam, C. CNS regulation of glucose homeostasis. Physiology Bethesda 24 , — CAS Google Scholar.

Coupe, B. Loss of autophagy in pro-opiomelanocortin neurons perturbs axon growth and causes metabolic dysregulation. Cell metabolism 15 , — Kaushik, S. Loss of autophagy in hypothalamic POMC neurons impairs lipolysis. EMBO Rep 13 , — Autophagy in hypothalamic AgRP neurons regulates food intake and energy balance.

Cell metabolism 14 , — Kroemer, G. Autophagy and the integrated stress response. Mol Cell 40 , — Jordan, S. Sensing the fuels: glucose and lipid signaling in the CNS controlling energy homeostasis. Cell Mol Life Sci 67 , — Levin, B. Metabolic sensing neurons and the control of energy homeostasis.

Physiol Behav 89 , — Marty, N.

Alexi VasbinderElizabeth Anderson Hyperglucemia, Husam ShadidHanna Raspberry planting guideHyperglycemia and inflammation Inf,ammationHyperglycemix U. Loosen Post-workout refueling, Athanasios ChalkiasFrank TackeEvangelos J. Giamarellos-BourboulisJochen ReiserJesper Eugen-OlsenEva L. FeldmanRodica Pop-BusuiSalim S. Hayek; on behalf of the ISIC Study Group, Inflammation, Hyperglycemia, and Adverse Outcomes in Individuals With Diabetes Mellitus Hospitalized for COVID

Author: Shakazil

2 thoughts on “Hyperglycemia and inflammation

Leave a comment

Yours email will be published. Important fields a marked *

Design by ThemesDNA.com