301
|
Inouye KE, Shi H, Howard JK, Daly CH, Lord GM, Rollins BJ, Flier JS. Absence of CC chemokine ligand 2 does not limit obesity-associated infiltration of macrophages into adipose tissue. Diabetes 2007; 56:2242-50. [PMID: 17473219 DOI: 10.2337/db07-0425] [Citation(s) in RCA: 180] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Macrophage recruitment to adipose tissue in obesity contributes to enhanced adipose tissue inflammatory activity and thus may underlie obesity-associated metabolic dysfunction. Obese adipose tissue exhibits increases in CC chemokine ligand 2 (CCL2, or monocyte chemoattractant protein-1), an important macrophage-recruiting factor. We therefore hypothesized that elevated CCL2 may contribute to obesity-associated adipose tissue macrophage recruitment. Male 6-week-old CCL2(-/-) and wild-type mice (n = 11-14 per group) were fed standard and high-fat diets until 34 weeks of age. At 12-16 and 25-29 weeks of age, blood was collected for plasma glucose and hormone measurements, and glucose tolerance and insulin tolerance tests were performed. Adipose tissue was collected at 34 weeks for analysis of macrophage infiltration. Surprisingly, CCL2(-/-) mice on high-fat diet showed no reductions in adipose tissue macrophages. CCL2(-/-) mice on standard and high-fat diet were also glucose intolerant and had mildly increased plasma glucose and decreased serum adiponectin levels compared with wild-type mice. On high-fat diet, CCL2(-/-) mice also gained slightly more weight and were hyperinsulinemic compared with wild-type mice. Because macrophage levels were unchanged in CCL2(-/-) mice, the phenotype appears to be caused by lack of CCL2 itself. The fact that metabolic function was altered in CCL2(-/-) mice, despite no changes in adipose tissue macrophage levels, suggests that CCL2 has effects on metabolism that are independent of its macrophage-recruiting capabilities. Importantly, we conclude that CCL2 is not critical for adipose tissue macrophage recruitment. The dominant factor for recruiting macrophages in adipose tissue during obesity therefore remains to be identified.
Collapse
Affiliation(s)
- Karen E Inouye
- Department of Medicine, Division of Endocrinology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| | | | | | | | | | | | | |
Collapse
|
302
|
Kose H, Bando Y, Izumi K, Yamada T, Matsumoto K. Epistasis between hyperglycemic QTLs revealed in a double congenic of the OLETF rat. Mamm Genome 2007; 18:609-15. [PMID: 17710484 DOI: 10.1007/s00335-007-9031-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2007] [Accepted: 04/25/2007] [Indexed: 01/14/2023]
Abstract
Glucose homeostasis is believed to be regulated by multiple genetic components, in addition to numerous external factors. It is therefore crucial to dissect and understand what roles each causative gene plays in maintaining proper glucose metabolism. In OLETF (Otsuka Long-Evans Tokushima Fatty) rat, a model of polygenic type 2 diabetes, at least 14 quantitative trait loci (QTLs) influencing plasma glucose levels were identified. In congenic strains some of the OLETF allelic variants were shown to increase glucose levels. In this study the focus was on two of the hyperglycemic loci, Nidd1/of and Nidd2/of. Congenic rats possessing OLETF genome fragment at either locus showed similar levels of mild hyperglycemia. A newly established double congenic rat showed a further aggravation of hyperglycemia. The Nidd1/of locus was also shown to function in the reduction of plasma leptin levels and fat weights, while the Nidd2/of locus led to increased plasma insulin and fat weights. Interestingly, both plasma leptin and fat weights reverted to the control levels in the double congenic rat. These results indicate that there is an epistatic interaction between the two loci. However, it is unlikely that the abnormal level of enhanced glucose homeostasis is mediated, at least not directly, by leptin or fat mass.
Collapse
Affiliation(s)
- Hiroyuki Kose
- Division for Animal Research Resources, Institute of Health Biosciences, The University of Tokushima Graduate School, Kuramoto-cho, Tokushima, 770-8503, Japan
| | | | | | | | | |
Collapse
|
303
|
Tsukumo DML, Carvalho-Filho MA, Carvalheira JBC, Prada PO, Hirabara SM, Schenka AA, Araújo EP, Vassallo J, Curi R, Velloso LA, Saad MJA. Loss-of-function mutation in Toll-like receptor 4 prevents diet-induced obesity and insulin resistance. Diabetes 2007; 56:1986-98. [PMID: 17519423 DOI: 10.2337/db06-1595] [Citation(s) in RCA: 615] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Obesity is associated with insulin resistance and a state of abnormal inflammatory response. The Toll-like receptor (TLR)4 has an important role in inflammation and immunity, and its expression has been reported in most tissues of the body, including the insulin-sensitive ones. Because it is activated by lipopolysaccharide and saturated fatty acids, which are inducers of insulin resistance, TLR4 may be a candidate for participation in the cross-talk between inflammatory and metabolic signals. Here, we show that C3H/HeJ mice, which have a loss-of-function mutation in TLR4, are protected against the development of diet-induced obesity. In addition, these mice demonstrate decreased adiposity, increased oxygen consumption, a decreased respiratory exchange ratio, improved insulin sensitivity, and enhanced insulin-signaling capacity in adipose tissue, muscle, and liver compared with control mice during high-fat feeding. Moreover, in these tissues, control mice fed a high-fat diet show an increase in IkappaB kinase complex and c-Jun NH(2)-terminal kinase activity, which is prevented in C3H/HeJ mice. In isolated muscles from C3H/HeJ mice, protection from saturated fatty acid-induced insulin resistance is observed. Thus, TLR4 appears to be an important mediator of obesity and insulin resistance and a potential target for the therapy of these highly prevalent medical conditions.
Collapse
Affiliation(s)
- Daniela M L Tsukumo
- Department of Internal Medicine, State University of Campinas, Campinas, São Paulo, Brazil
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
304
|
Abstract
Recently, the high responsiveness of omental adipocytes to positive lipolytic stimuli has been clearly demonstrated in women. We conclude that adipose tissue fatty acid release, storage capacity, and secreted cytokines may all be involved in the etiology of the metabolic syndrome. The anatomical location of visceral adipocytes close to the liver, combined with possible depot-specific alterations in various adipocyte or adipose tissue features likely play critical roles in this process. This highly complex etiology is concordant with the heterogeneous clinical manifestations of the metabolic syndrome, and suggests possible interindividual variability in the extent to which each pathophysiological mechanism is involved.
Collapse
Affiliation(s)
- André Tchernof
- Molecular Endocrinology and Oncology Research Center, Department of Nutrition, Laval University Medical Research Center and Laval University, Quebec City, Province of Quebec, Canada.
| |
Collapse
|
305
|
Eriksson JW. Metabolic stress in insulin's target cells leads to ROS accumulation - A hypothetical common pathway causing insulin resistance. FEBS Lett 2007; 581:3734-42. [PMID: 17628546 DOI: 10.1016/j.febslet.2007.06.044] [Citation(s) in RCA: 114] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2007] [Revised: 06/16/2007] [Accepted: 06/18/2007] [Indexed: 01/04/2023]
Abstract
The metabolic syndrome is a cluster of cardiovascular risk factors, and visceral adiposity is a central component that is also strongly associated with insulin resistance. Both visceral obesity and insulin resistance are important risk factors for the development of type 2 diabetes. It is likely that adipose tissue, particularly in the intra-abdominal depot, is part of a complex interplay involving several tissues and that dysregulated hormonal, metabolic and neural signalling within and between organs can trigger development of metabolic disease. One attractive hypothesis is that many factors leading to insulin resistance are mediated via the generation of abnormal amounts of reactive oxygen species (ROS). There is much evidence supporting that detrimental effects of glucose, fatty acids, hormones and cytokines leading to insulin resistance can be exerted via such a common pathway. This review paper mainly focuses on metabolic and other 'stress' factors that affect insulin's target cells, in particular adipocytes, and it will highlight oxidative stress as a potential unifying mechanism by which these stress factors promote insulin resistance and the development and progression of type 2 diabetes.
Collapse
Affiliation(s)
- Jan W Eriksson
- The Lundberg Laboratory for Diabetes Research, Institute of Medicine, Sahlgrenska University Hospital, SE 41345 Gothenburg, Sweden.
| |
Collapse
|
306
|
Rouleau P, Ung RV, Lapointe NP, Guertin PA. Hormonal and immunological changes in mice after spinal cord injury. J Neurotrauma 2007; 24:367-78. [PMID: 17376000 DOI: 10.1089/neu.2006.0117] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Spinal cord injury (SCI) is associated with immune deficiencies and life-threatening infections. However, the specific mechanisms underlying this pathological condition remain unclear. In recent years, increasing evidence has suggested that anabolic hormones may be involved in immunological complications. Here, we monitored candidate hormone concentrations and immune cell counts, in CD1 mice, for 4 weeks after low-thoracic transection of the spinal cord (Tx). Serum dihydroepiandrosterone (DHEA), insulin, and parathyroid hormone (PTH) levels decreased throughout the time period studied compared with control, non-Tx mice. In turn, testosterone and growth hormone (GH) levels were only transiently changed, with a decrease of testosterone during the first 2 weeks and an increase of GH at 1 week post-Tx. A complete blood count revealed either unchanged or moderately decreased erythrocyte, platelet, hemoglobin and hematocrit levels. Total leukocyte, lymphocyte, and eosinophil counts also decreased, whereas neutrophils and monocytes did not change significantly. In the bone marrow, lymphocyte numbers decreased and neutrophils increased, whereas monocytes, eosinophils, and megakariocytes did not change significantly. These results revealed significant changes occurring rapidly (<1-2 weeks) after Tx in both hormonal and immunological systems, providing compelling evidence of a role for anabolic hormones in SCI-related immune deficiencies.
Collapse
Affiliation(s)
- Pascal Rouleau
- Neuroscience Unit, Laval University Medical Center (CHUQ-CHUL), Laval University, Quebec City, Quebec, Canada
| | | | | | | |
Collapse
|
307
|
Yi P, Lu FE, Chen G, Xu LJ, Wang KF. Molecular mechanism of insulin resistance induced by free fatty acids in 3T3-L1 adipocytes through targeting nuclear-kB p65. Shijie Huaren Xiaohua Zazhi 2007; 15:1706-1712. [DOI: 10.11569/wcjd.v15.i15.1706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effect of free fatty acids (FFAs) on nuclear factor-kB p65 (NF-kBp65) expression and translocation in 3T3-L1 adipocytes and its possible molecular mechanism.
METHODS: 3T3-L1 adipocytes were treated for 6 to 24 hours with palmic acid (0.3, 0.5, 1.0 mmol/L) to induce insulin resistance. Glucose oxidase method was employed to measure the glucose consumption in the medium and 2-deoxy--[3H]-D-glucose method was used for the determination of glucose uptake. Western blot was used to determine the protein expression of total NF-kBp65 and nuclear NF-kBp65. The distribution of NF-kBp65 was investigated by confocal laser scanning microscopy (CLSM).
RESULTS: After the treatment with 0.3, 0.5 and 1.0 mmol/L of palmic acid for 6 to 24 hours, the glucose consumption (3.03 ± 0.34, 2.71 ± 0.36, 2.64 ± 0.25 mmol/L) and the insulin-stimulated glucose transport of 3T3-L1 adipose cells (64%, 33%, 32%) were decreased in a dose- and time-dependent manner. Both the expression of nuclear NF-kBp65 and nuclear translocation of NF-kBp65 were increased in a dose- and time-dependent manner. But the total NF-kBp65 protein expression had no marked changes during this study.
CONCLUSION: FFAs can induce insulin resistance, the molecular mechanism of which might be associated with the activation and translocation of NF-kBp65.
Collapse
|
308
|
Abstract
Weight gain and obesity are major risk factors for conditions and diseases ranging from insulin resistance and type 2 diabetes mellitus to atherosclerosis and the sequelae of nonalcoholic fatty liver disease. A chronic, subacute state of inflammation often accompanies the accumulation of excess lipid in adipose tissue and liver (hepatic steatosis), evidenced by changes in both inflammatory cells and biochemical markers of inflammation. These changes can be seen in the involved tissues and systemically, in terms of elevated circulating levels of inflammatory markers. The link between obesity and inflammation has therefore raised the important question of whether obesity-induced inflammation plays a pathogenic role in the development and progression of these disorders. We review the rapidly expanding body of animal and clinical data that support potential roles for inflammation in the pathogenesis of insulin resistance and type 2 diabetes mellitus.
Collapse
|
309
|
Kempf K, Rose B, Herder C, Kleophas U, Martin S, Kolb H. Inflammation in metabolic syndrome and type 2 diabetes: Impact of dietary glucose. Ann N Y Acad Sci 2007; 1084:30-48. [PMID: 17151291 DOI: 10.1196/annals.1372.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Chronic overnutrition combined with a lack of exercise is the main cause for the rapidly increasing prevalence of overweight and obesity. It seems accepted that adipositis (macrophage infiltration and inflammation of adipose tissue in obesity) and systemic low grade inflammation affect the pathogenesis of the metabolic syndrome or type 2 diabetes mellitus (T2DM). Therefore, modern weight reduction programs additionally focus on strategies to attenuate the inflammation state. Exercise is one major factor, which contributes to the reduction of both the incidence of T2DM and inflammation, and the immunomodulatory effects of exercise are supported by similarly beneficial effects of dietary changes. In this context, glucose is the most extensively studied nutrient and current investigations focus on postprandial glucose-induced inflammation, one possible reason why hyperglycemia is detrimental. Indeed, glucose may modulate the mRNA expression and serum concentrations of immune parameters but these alterations rapidly normalize in normoglycemic subjects. In case of an impaired metabolic state, however, postprandial hyperglycemia increases magnitude and duration of systemic inflammatory responses, which probably promotes the development of T2DM and of cardiovascular disease.
Collapse
Affiliation(s)
- Kerstin Kempf
- German Diabetes Clinic, German Diabetes Center, Leibniz Institute at Heinrich-Heine-University Düsseldorf, Auf'm Hennekamp 65, 40225 Düsseldorf, Germany.
| | | | | | | | | | | |
Collapse
|
310
|
Shaw P, Clarke AR. Murine models of intestinal cancer: recent advances. DNA Repair (Amst) 2007; 6:1403-12. [PMID: 17376749 DOI: 10.1016/j.dnarep.2007.02.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2007] [Accepted: 02/14/2007] [Indexed: 10/23/2022]
Abstract
Since the advent of strategies capable of manipulating the germline of mice, there has been a rapid expansion in the number of murine models of intestinal cancer. These have largely been developed with the specific aim of elucidating the molecular mechanisms underlying tumour initiation and progression. In attempting this goal, these models have become increasingly sophisticated, allowing ever more precise recapitulation of the genetic events that underlie human disease. Such technological advances include both temporal and spatial control over mutant allele expression. This review highlights some of notable recent advances using these approaches, with particular focus upon the role of a number of key signalling pathways, DNA repair mechanisms and inflammation.
Collapse
Affiliation(s)
- Paul Shaw
- Cardiff School of Biosciences, Cardiff University, Cardiff, UK
| | | |
Collapse
|
311
|
Abstract
Insulin resistance is a major causative factor for type 2 diabetes and is associated with increased risk of cardiovascular disease. Despite intense investigation for a number of years, molecular mechanisms underlying insulin resistance remain to be determined. Recently, chronic inflammation has been highlighted as a culprit for obesity-induced insulin resistance. Nonetheless, upstream regulators and downstream effectors of chronic inflammation in insulin resistance remain unclarified. Inducible nitric oxide synthase (iNOS), a mediator of inflammation, has emerged as an important player in insulin resistance. Obesity is associated with increased iNOS expression in insulin-sensitive tissues in rodents and humans. Inhibition of iNOS ameliorates obesity-induced insulin resistance. However, molecular mechanisms by which iNOS mediates insulin resistance remain largely unknown. Protein S-nitrosylation, a covalent attachment of NO moiety to thiol sulfhydryls, has emerged as a major mediator of a broad array of NO actions. S-nitrosylation is elevated in patients with type 2 diabetes, and increased S-nitrosylation of insulin signaling molecules, including insulin receptor, insulin receptor substrate-1, and Akt/PKB, has been shown in skeletal muscle of obese, diabetic mice. Akt/PKB is reversibly inactivated by S-nitrosylation. Based on these findings, S-nitrosylation has recently been proposed to play an important role in the pathogenesis of insulin resistance.
Collapse
Affiliation(s)
- Masao Kaneki
- Department of Anesthesia & Critical Care, Massachusetts General Hospital, Shriners Hospital for Children, Harvard Medical School, Charlestown, Massachusetts 02129, USA.
| | | | | | | |
Collapse
|
312
|
Flowers JB, Oler AT, Nadler ST, Choi Y, Schueler KL, Yandell BS, Kendziorski CM, Attie AD. Abdominal obesity in BTBR male mice is associated with peripheral but not hepatic insulin resistance. Am J Physiol Endocrinol Metab 2007; 292:E936-45. [PMID: 17132824 DOI: 10.1152/ajpendo.00370.2006] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Insulin resistance is a common feature of obesity. BTBR mice have more fat mass than most other inbred mouse strains. On a chow diet, BTBR mice have elevated insulin levels relative to the C57BL/6J (B6) strain. Male F1 progeny of a B6 x BTBR cross are insulin resistant. Previously, we reported insulin resistance in isolated muscle and in isolated adipocytes in this strain. Whereas the muscle insulin resistance was observed only in male F1 mice, adipocyte insulin resistance was also present in male BTBR mice. We examined in vivo mechanisms of insulin resistance with the hyperinsulinemic euglycemic clamp technique. At 10 wk of age, BTBR and F1 mice had a >30% reduction in whole body glucose disposal primarily due to insulin resistance in heart, soleus muscle, and adipose tissue. The increased adipose tissue mass and decreased muscle mass in BTBR and F1 mice were negatively and positively correlated with whole body glucose disposal, respectively. Genes involved in focal adhesion, actin cytoskeleton, and inflammation were more highly expressed in BTBR and F1 than in B6 adipose tissue. The BTBR and F1 mice have higher levels of testosterone, which may be related to the pathological changes in adipose tissue that lead to systemic insulin resistance. Despite profound peripheral insulin resistance, BTBR and F1 mice retained hepatic insulin sensitivity. These studies reveal a genetic difference in body composition that correlates with large differences in peripheral insulin sensitivity.
Collapse
Affiliation(s)
- Jessica B Flowers
- Department of Nutritional Sciences, University of Wisconsin-Madison, 433 Babcock Dr., Madison, WI 53706, USA
| | | | | | | | | | | | | | | |
Collapse
|
313
|
Wright WS, Longo KA, Dolinsky VW, Gerin I, Kang S, Bennett CN, Chiang SH, Prestwich TC, Gress C, Burant CF, Susulic VS, MacDougald OA. Wnt10b inhibits obesity in ob/ob and agouti mice. Diabetes 2007; 56:295-303. [PMID: 17259372 DOI: 10.2337/db06-1339] [Citation(s) in RCA: 138] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The Wnt family of secreted signaling molecules has profound effects on diverse developmental processes, including the fate of mesenchymal progenitors. While activation of Wnt signaling blocks adipogenesis, inhibition of endogenous Wnt/beta-catenin signaling by Wnt10b promotes spontaneous preadipocyte differentiation. Transgenic mice with expression of Wnt10b from the FABP4 promoter (FABP4-Wnt10b) have less adipose tissue when maintained on a normal chow diet and are resistant to diet-induced obesity. Here we demonstrate that FABP4-Wnt10b mice largely avert weight gain and metabolic abnormalities associated with genetic obesity. FABP4-Wnt10b mice do not gain significant body weight on the ob/ob background, and at 8 weeks of age, they have an approximately 70% reduction in visceral and subcutaneous adipose tissues compared with ob/ob mice. Similarly, on the lethal yellow agouti (A(y)) background, FABP4-Wnt10b mice have 50-70% less adipose tissue weight and circulating leptin at 5 months of age. Wnt10b-Ay mice are more glucose tolerant and insulin sensitive than A(y) controls, perhaps due to reduced expression and circulation of resistin. Reduced expression of inflammatory cytokines may also contribute to improved glucose homeostasis.
Collapse
Affiliation(s)
- Wendy S Wright
- Department of Molecular and Integrative Physiology, University of Michigan, 1301 E. Catherine Drive, Ann Arbor, MI 48109-0622, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
314
|
Chow FY, Nikolic-Paterson DJ, Ma FY, Ozols E, Rollins BJ, Tesch GH. Monocyte chemoattractant protein-1-induced tissue inflammation is critical for the development of renal injury but not type 2 diabetes in obese db/db mice. Diabetologia 2007; 50:471-80. [PMID: 17160673 DOI: 10.1007/s00125-006-0497-8] [Citation(s) in RCA: 190] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2006] [Accepted: 09/05/2006] [Indexed: 01/12/2023]
Abstract
AIMS/HYPOTHESIS Tissue macrophage accumulation is thought to induce insulin resistance during obesity and stimulate the progression of diabetic nephropathy. Monocyte chemoattractant protein-1 (MCP-1) is a potent stimulator of macrophage recruitment. It is increased in adipose tissue during obesity and in diabetic kidneys, suggesting that inflammation of these tissues may be MCP-1-dependent. Based on these findings, the aim of this study was to examine whether a deficiency in MCP-1 would alter the development of type 2 diabetes and its renal complications. MATERIALS AND METHODS The role of MCP-1 in the progression of type 2 diabetes and its associated renal injury was assessed in obese db/db mice that were deficient in the gene encoding MCP-1 (Ccl2). RESULTS The incidence and development of type 2 diabetes were similar in Ccl2(+/+) and Ccl2(-/-) db/db mice between 8 and 32 weeks of age. Body mass, hyperglycaemia, hyperinsulinaemia, glucose and insulin tolerance, plasma triacylglycerol and serum NEFA were not different between these strains. Pathological changes in epididymal adipose tissue, including increases in macrophage accumulation and Tnfa mRNA and reductions in Adipoq mRNA, were unaffected by the absence of MCP-1. In contrast, kidney macrophage accumulation and the progression of diabetic renal injury (albuminuria, histopathology, renal fibrosis) were substantially reduced in Ccl2(-/-) compared with Ccl2(+/+) db/db mice with equivalent diabetes. CONCLUSIONS/INTERPRETATION Our study demonstrates that MCP-1 promotes type 2 diabetic renal injury but does not influence the development of obesity, insulin resistance or type 2 diabetes in db/db mice. MCP-1 plays a critical role in inflammation of the kidney, but not adipose tissue, during the progression of type 2 diabetes.
Collapse
Affiliation(s)
- F Y Chow
- Department of Nephrology, Monash Medical Centre, Clayton, VIC, 3168, Australia
| | | | | | | | | | | |
Collapse
|
315
|
Kahn SE, Hull RL, Utzschneider KM. Mechanisms linking obesity to insulin resistance and type 2 diabetes. Nature 2007; 444:840-6. [PMID: 17167471 DOI: 10.1038/nature05482] [Citation(s) in RCA: 3582] [Impact Index Per Article: 199.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Obesity is associated with an increased risk of developing insulin resistance and type 2 diabetes. In obese individuals, adipose tissue releases increased amounts of non-esterified fatty acids, glycerol, hormones, pro-inflammatory cytokines and other factors that are involved in the development of insulin resistance. When insulin resistance is accompanied by dysfunction of pancreatic islet beta-cells - the cells that release insulin - failure to control blood glucose levels results. Abnormalities in beta-cell function are therefore critical in defining the risk and development of type 2 diabetes. This knowledge is fostering exploration of the molecular and genetic basis of the disease and new approaches to its treatment and prevention.
Collapse
Affiliation(s)
- Steven E Kahn
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, VA Puget Sound Health Care System and University of Washington, 1660 South Columbian Way, Seattle, Washington 98108, USA.
| | | | | |
Collapse
|
316
|
Juan CC, Chang CL, Chuang TY, Huang SW, Kwok CF, Ho LT. Insulin sensitivity and resistin expression in nitric oxide-deficient rats. Diabetologia 2006; 49:3017-26. [PMID: 17063326 DOI: 10.1007/s00125-006-0403-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2006] [Accepted: 07/03/2006] [Indexed: 12/30/2022]
Abstract
AIMS/HYPOTHESIS The aim of this study was to investigate changes in insulin sensitivity and expression of the gene encoding resistin (Retn) in adipocytes from long-term nitric oxide (NO)-deficient rats. METHODS Male Sprague-Dawley rats received [Formula: see text]-nitro-L: -arginine methyl ester (L-NAME 0.5 mg/ml) in their drinking water for 4 weeks, while control rats received plain drinking water. During the experimental period, changes in plasma glucose, insulin and C-peptide levels were measured. After administration of L-NAME for 4 weeks, insulin sensitivity was evaluated in vivo and in vitro. An insulin binding assay was also performed to determine the number and binding affinity of insulin receptors in adipocytes. Adipocyte Retn mRNA levels were examined using northern blotting. RESULTS Successful induction of NO deficiency was demonstrated by an increase in systemic blood pressure. No difference in plasma glucose levels was found between the two groups. Compared with the control rats, plasma insulin and C-peptide levels were significantly decreased in the NO-deficient rats, and insulin sensitivity was significantly increased. Insulin-stimulated glucose uptake and insulin binding capacity, but not binding affinity, were significantly increased in adipocytes isolated from NO-deficient rats. In addition, adipocyte Retn mRNA levels, but not plasma resistin levels, were significantly decreased in NO-deficient rats, and the Retn mRNA levels were negatively correlated with insulin sensitivity. CONCLUSIONS/INTERPRETATION Insulin sensitivity was increased in NO-deficient rats and this was associated with insulin binding capacity and downregulated Retn expression. These findings suggest that NO plays a regulatory role in metabolism. Dysregulation of NO production may result in the development of metabolic disorders.
Collapse
Affiliation(s)
- C C Juan
- Department of Physiology, School of Medicine, National Yang-Ming University, Taipei, Taiwan.
| | | | | | | | | | | |
Collapse
|
317
|
Park J, Choe SS, Choi AH, Kim KH, Yoon MJ, Suganami T, Ogawa Y, Kim JB. Increase in glucose-6-phosphate dehydrogenase in adipocytes stimulates oxidative stress and inflammatory signals. Diabetes 2006; 55:2939-49. [PMID: 17065329 DOI: 10.2337/db05-1570] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
In adipocytes, oxidative stress and chronic inflammation are closely associated with metabolic disorders, including insulin resistance, obesity, cardiovascular disease, and type 2 diabetes. However, the molecular mechanisms underlying these metabolic disorders have not been thoroughly elucidated. In this report, we demonstrate that overexpression of glucose-6-phosphate dehydrogenase (G6PD) in adipocytes stimulates oxidative stress and inflammatory responses, thus affecting the neighboring macrophages. Adipogenic G6PD overexpression promotes the expression of pro-oxidative enzymes, including inducible nitric oxide synthase and NADPH oxidase, and the activation of nuclear factor-kappaB (NF-kappaB) signaling, which eventually leads to the dysregulation of adipocytokines and inflammatory signals. Furthermore, secretory factors from G6PD-overexpressing adipocytes stimulate macrophages to express more proinflammatory cytokines and to be recruited to the adipocytes; this would cause chronic inflammatory conditions in the adipose tissue of obesity. These effects of G6PD overexpression in adipocytes were abolished by pretreatment with NF-kappaB inhibitors or antioxidant drugs. Thus, we propose that a high level of G6PD in adipocytes may mediate the onset of metabolic disorders in obesity by increasing the oxidative stress and inflammatory signals.
Collapse
Affiliation(s)
- Jiyoung Park
- Department of Biological Sciences, Seoul National University, San 56-1, Sillim-Dong, Kwanak-Gu, Seoul, Korea
| | | | | | | | | | | | | | | |
Collapse
|
318
|
Jang EH, Park CS, Lee SK, Pie JE, Kang JH. Excessive nitric oxide attenuates leptin-mediated signal transducer and activator of transcription 3 activation. Life Sci 2006; 80:609-17. [PMID: 17097687 DOI: 10.1016/j.lfs.2006.10.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2006] [Revised: 09/15/2006] [Accepted: 10/12/2006] [Indexed: 11/24/2022]
Abstract
The mechanisms of leptin resistance observed in most cases of human obesity are poorly understood. Therefore, we evaluated the effects of nitric oxide (NO) on the leptin-induced activation of Janus kinase/signal transducer and activator of transcription 3 (JAK/STAT3) pathways and on the leptin receptor (LEPR) expression using SH-SY5Y cells. Here, we show that the NO donor spermine/NONOate inhibited leptin-induced activation of STAT3 in vitro. The inhibition of leptin-mediated STAT3 phosphorylation caused by excessive NO was partially prevented by a sulfhydryl reducing agent, ascorbic acid. Cellular experiments show that reduced expression of long form leptin receptor (LEPR-b) and STAT3 protein instability induced by NO may be mechanisms of the NO-mediated inhibition of leptin-STAT3 signaling. We also present data showing that the hypothalamic NO content of high-fat (HF)-diet-induced obese mice was higher than that of control mice; this is likely caused by decreased caveolin-1 expression and increased nNOS expression induced by HF diet over 19 weeks. Concurrently with the overproduction of NO, the decrease of hypothalamic LEPR-b in obese mice also supports these in vitro data. Combined results suggest that excess of NO can induce the attenuation of leptin-mediated STAT3 activation through reduced expression of LEPR-b mRNA and instability of STAT3 protein at least in part. Furthermore, our in vivo data indicate that long-term HF diet induces hypothalamic overproduction of NO, which may be related with leptin insensitivity. However, further study is required to warrant direct in vivo evidence of a causal relationship between endogenous excess of hypothalamic NO and central leptin resistance.
Collapse
Affiliation(s)
- Eun-Hee Jang
- Department of Pharmacology and Medicinal Toxicology Research Center, Inha Research Institute for Medical Sciences, Center for Advanced Medical Education, Inha University, College of Medicine by BK-21 Project, South Korea
| | | | | | | | | |
Collapse
|
319
|
Beard KM, Lu H, Ho K, Fantus IG. Bradykinin augments insulin-stimulated glucose transport in rat adipocytes via endothelial nitric oxide synthase-mediated inhibition of Jun NH2-terminal kinase. Diabetes 2006; 55:2678-87. [PMID: 17003331 DOI: 10.2337/db05-1538] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
An increase in bradykinin has been suggested to contribute to the enhanced insulin sensitivity observed in the presence of ACE inhibitors. To investigate a potential direct, nonvascular effect on an insulin target tissue, the effect of bradykinin on glucose uptake and insulin signaling was studied in primary rat adipocytes. Whereas basal glucose uptake was not altered, bradykinin augmented insulin-stimulated glucose uptake twofold, which was blocked by HOE-140, a bradykinin B2 receptor antagonist. The bradykinin effect on glucose uptake was nitric oxide (NO) dependent, mimicked by NO donors and absent in adipocytes from endothelial NO synthase-/- mice. Investigation of insulin signaling revealed that bradykinin enhanced insulin receptor substrate-1 (IRS-1) Tyr phosphorylation, Akt/protein kinase B phosphorylation, and GLUT4 translocation. In contrast, insulin-stimulated extracellular signal-regulated kinase1/2 and Jun NH2-terminal kinase (JNK) activation were decreased in the presence of bradykinin, accompanied by decreased IRS-1 Ser307 phosphorylation. Furthermore, bradykinin did not enhance insulin action in the presence of the JNK inhibitor, SP-600125, or in adipocytes from JNK1-/- mice. These data indicate that bradykinin enhances insulin sensitivity in adipocytes via an NO-dependent pathway that acts by modulating the feedback inhibition of insulin signaling at the level of IRS-1.
Collapse
Affiliation(s)
- Kristin M Beard
- Department of Medicine and Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, ON, Canada M5G 1X6
| | | | | | | |
Collapse
|
320
|
Valerio A, Cardile A, Cozzi V, Bracale R, Tedesco L, Pisconti A, Palomba L, Cantoni O, Clementi E, Moncada S, Carruba MO, Nisoli E. TNF-alpha downregulates eNOS expression and mitochondrial biogenesis in fat and muscle of obese rodents. J Clin Invest 2006; 116:2791-8. [PMID: 16981010 PMCID: PMC1564431 DOI: 10.1172/jci28570] [Citation(s) in RCA: 241] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2006] [Accepted: 07/25/2006] [Indexed: 12/16/2022] Open
Abstract
Obesity is associated with chronic low-grade inflammation. Thus, at metabolically relevant sites, including adipose tissue and muscle, there is abnormal production of proinflammatory cytokines such as TNF-alpha. Here we demonstrate that eNOS expression was reduced, with a concomitant reduction of mitochondrial biogenesis and function, in white and brown adipose tissue and in the soleus muscle of 3 different animal models of obesity. The genetic deletion of TNF receptor 1 in obese mice restored eNOS expression and mitochondrial biogenesis in fat and muscle; this was associated with less body weight gain than in obese wild-type controls. Furthermore, TNF-alpha downregulated eNOS expression and mitochondrial biogenesis in cultured white and brown adipocytes and muscle satellite cells of mice. The NO donors DETA-NO and SNAP prevented the reduction of mitochondrial biogenesis observed with TNF-alpha. Our findings demonstrate that TNF-alpha impairs mitochondrial biogenesis and function in different tissues of obese rodents by downregulating eNOS expression and suggest a novel pathophysiological process that sustains obesity.
Collapse
Affiliation(s)
- Alessandra Valerio
- Integrated Laboratories Network, Center for Study and Research on Obesity, Department of Pharmacology, School of Medicine, University of Milan, Milan, Italy.
Department of Preclinical Sciences, University of Milan, Milan, Italy.
CEINGE Biotecnologie Avanzate, Naples, Italy.
Istituto Auxologico Italiano, Milan, Italy.
Stem Cell Research Institute, San Raffaele Scientific Institute, Milan, Italy.
Istituto di Farmacologia e Farmacognosia, University of Urbino “Carlo Bo,” Urbino, Italy.
Eugenio Medea Scientific Institute, Lecco, Italy.
Wolfson Institute for Biomedical Research, University College London, London, United Kingdom
| | - Annalisa Cardile
- Integrated Laboratories Network, Center for Study and Research on Obesity, Department of Pharmacology, School of Medicine, University of Milan, Milan, Italy.
Department of Preclinical Sciences, University of Milan, Milan, Italy.
CEINGE Biotecnologie Avanzate, Naples, Italy.
Istituto Auxologico Italiano, Milan, Italy.
Stem Cell Research Institute, San Raffaele Scientific Institute, Milan, Italy.
Istituto di Farmacologia e Farmacognosia, University of Urbino “Carlo Bo,” Urbino, Italy.
Eugenio Medea Scientific Institute, Lecco, Italy.
Wolfson Institute for Biomedical Research, University College London, London, United Kingdom
| | - Valeria Cozzi
- Integrated Laboratories Network, Center for Study and Research on Obesity, Department of Pharmacology, School of Medicine, University of Milan, Milan, Italy.
Department of Preclinical Sciences, University of Milan, Milan, Italy.
CEINGE Biotecnologie Avanzate, Naples, Italy.
Istituto Auxologico Italiano, Milan, Italy.
Stem Cell Research Institute, San Raffaele Scientific Institute, Milan, Italy.
Istituto di Farmacologia e Farmacognosia, University of Urbino “Carlo Bo,” Urbino, Italy.
Eugenio Medea Scientific Institute, Lecco, Italy.
Wolfson Institute for Biomedical Research, University College London, London, United Kingdom
| | - Renata Bracale
- Integrated Laboratories Network, Center for Study and Research on Obesity, Department of Pharmacology, School of Medicine, University of Milan, Milan, Italy.
Department of Preclinical Sciences, University of Milan, Milan, Italy.
CEINGE Biotecnologie Avanzate, Naples, Italy.
Istituto Auxologico Italiano, Milan, Italy.
Stem Cell Research Institute, San Raffaele Scientific Institute, Milan, Italy.
Istituto di Farmacologia e Farmacognosia, University of Urbino “Carlo Bo,” Urbino, Italy.
Eugenio Medea Scientific Institute, Lecco, Italy.
Wolfson Institute for Biomedical Research, University College London, London, United Kingdom
| | - Laura Tedesco
- Integrated Laboratories Network, Center for Study and Research on Obesity, Department of Pharmacology, School of Medicine, University of Milan, Milan, Italy.
Department of Preclinical Sciences, University of Milan, Milan, Italy.
CEINGE Biotecnologie Avanzate, Naples, Italy.
Istituto Auxologico Italiano, Milan, Italy.
Stem Cell Research Institute, San Raffaele Scientific Institute, Milan, Italy.
Istituto di Farmacologia e Farmacognosia, University of Urbino “Carlo Bo,” Urbino, Italy.
Eugenio Medea Scientific Institute, Lecco, Italy.
Wolfson Institute for Biomedical Research, University College London, London, United Kingdom
| | - Addolorata Pisconti
- Integrated Laboratories Network, Center for Study and Research on Obesity, Department of Pharmacology, School of Medicine, University of Milan, Milan, Italy.
Department of Preclinical Sciences, University of Milan, Milan, Italy.
CEINGE Biotecnologie Avanzate, Naples, Italy.
Istituto Auxologico Italiano, Milan, Italy.
Stem Cell Research Institute, San Raffaele Scientific Institute, Milan, Italy.
Istituto di Farmacologia e Farmacognosia, University of Urbino “Carlo Bo,” Urbino, Italy.
Eugenio Medea Scientific Institute, Lecco, Italy.
Wolfson Institute for Biomedical Research, University College London, London, United Kingdom
| | - Letizia Palomba
- Integrated Laboratories Network, Center for Study and Research on Obesity, Department of Pharmacology, School of Medicine, University of Milan, Milan, Italy.
Department of Preclinical Sciences, University of Milan, Milan, Italy.
CEINGE Biotecnologie Avanzate, Naples, Italy.
Istituto Auxologico Italiano, Milan, Italy.
Stem Cell Research Institute, San Raffaele Scientific Institute, Milan, Italy.
Istituto di Farmacologia e Farmacognosia, University of Urbino “Carlo Bo,” Urbino, Italy.
Eugenio Medea Scientific Institute, Lecco, Italy.
Wolfson Institute for Biomedical Research, University College London, London, United Kingdom
| | - Orazio Cantoni
- Integrated Laboratories Network, Center for Study and Research on Obesity, Department of Pharmacology, School of Medicine, University of Milan, Milan, Italy.
Department of Preclinical Sciences, University of Milan, Milan, Italy.
CEINGE Biotecnologie Avanzate, Naples, Italy.
Istituto Auxologico Italiano, Milan, Italy.
Stem Cell Research Institute, San Raffaele Scientific Institute, Milan, Italy.
Istituto di Farmacologia e Farmacognosia, University of Urbino “Carlo Bo,” Urbino, Italy.
Eugenio Medea Scientific Institute, Lecco, Italy.
Wolfson Institute for Biomedical Research, University College London, London, United Kingdom
| | - Emilio Clementi
- Integrated Laboratories Network, Center for Study and Research on Obesity, Department of Pharmacology, School of Medicine, University of Milan, Milan, Italy.
Department of Preclinical Sciences, University of Milan, Milan, Italy.
CEINGE Biotecnologie Avanzate, Naples, Italy.
Istituto Auxologico Italiano, Milan, Italy.
Stem Cell Research Institute, San Raffaele Scientific Institute, Milan, Italy.
Istituto di Farmacologia e Farmacognosia, University of Urbino “Carlo Bo,” Urbino, Italy.
Eugenio Medea Scientific Institute, Lecco, Italy.
Wolfson Institute for Biomedical Research, University College London, London, United Kingdom
| | - Salvador Moncada
- Integrated Laboratories Network, Center for Study and Research on Obesity, Department of Pharmacology, School of Medicine, University of Milan, Milan, Italy.
Department of Preclinical Sciences, University of Milan, Milan, Italy.
CEINGE Biotecnologie Avanzate, Naples, Italy.
Istituto Auxologico Italiano, Milan, Italy.
Stem Cell Research Institute, San Raffaele Scientific Institute, Milan, Italy.
Istituto di Farmacologia e Farmacognosia, University of Urbino “Carlo Bo,” Urbino, Italy.
Eugenio Medea Scientific Institute, Lecco, Italy.
Wolfson Institute for Biomedical Research, University College London, London, United Kingdom
| | - Michele O. Carruba
- Integrated Laboratories Network, Center for Study and Research on Obesity, Department of Pharmacology, School of Medicine, University of Milan, Milan, Italy.
Department of Preclinical Sciences, University of Milan, Milan, Italy.
CEINGE Biotecnologie Avanzate, Naples, Italy.
Istituto Auxologico Italiano, Milan, Italy.
Stem Cell Research Institute, San Raffaele Scientific Institute, Milan, Italy.
Istituto di Farmacologia e Farmacognosia, University of Urbino “Carlo Bo,” Urbino, Italy.
Eugenio Medea Scientific Institute, Lecco, Italy.
Wolfson Institute for Biomedical Research, University College London, London, United Kingdom
| | - Enzo Nisoli
- Integrated Laboratories Network, Center for Study and Research on Obesity, Department of Pharmacology, School of Medicine, University of Milan, Milan, Italy.
Department of Preclinical Sciences, University of Milan, Milan, Italy.
CEINGE Biotecnologie Avanzate, Naples, Italy.
Istituto Auxologico Italiano, Milan, Italy.
Stem Cell Research Institute, San Raffaele Scientific Institute, Milan, Italy.
Istituto di Farmacologia e Farmacognosia, University of Urbino “Carlo Bo,” Urbino, Italy.
Eugenio Medea Scientific Institute, Lecco, Italy.
Wolfson Institute for Biomedical Research, University College London, London, United Kingdom
| |
Collapse
|
321
|
Carvalho-Filho MA, Ueno M, Carvalheira JBC, Velloso LA, Saad MJA. Targeted disruption of iNOS prevents LPS-induced S-nitrosation of IRbeta/IRS-1 and Akt and insulin resistance in muscle of mice. Am J Physiol Endocrinol Metab 2006; 291:E476-82. [PMID: 16638822 DOI: 10.1152/ajpendo.00422.2005] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
We have previously demonstrated that the insulin resistance associated with inducible nitric oxide synthase (iNOS) induction in two different models of obesity, diet-induced obesity and the ob/ob mice, is mediated by S-nitrosation of proteins involved in insulin signal transduction: insulin receptor beta-subunit (IRbeta), insulin receptor substrate 1(IRS-1), and Akt. S-nitrosation of IRbeta and Akt impairs their kinase activities, and S-nitrosation of IRS-1 reduces its tissue expression. In this study, we observed that LPS-induced insulin resistance in the muscle of wild-type mice, as demonstrated by reduced insulin-induced tyrosine phosphorylation of IRbeta and IRS-1, reduced IRS-1 expression and reduced insulin-induced serine phosphorylation of Akt. This resistance occurred in parallel with enhanced iNOS expression, which was accompanied by S-nitrosation of IRbeta/IRS-1 and Akt. In the muscle of iNOS(-/-) mice, we did not observe enhanced iNOS expression or any S-nitrosation of IRbeta/IRS-1 and Akt after LPS treatment. Moreover, insulin resistance was not present. The preservation of insulin-induced tyrosine phosphorylation of IRbeta and IRS-1, of IRS-1 protein expression, and of insulin-induced serine phosphorylation of Akt observed in LPS-treated iNOS(-/-) mice strongly suggests that the insulin resistance induced by LPS is iNOS mediated, probably through S-nitrosation of proteins of early steps of insulin signaling.
Collapse
Affiliation(s)
- Marco A Carvalho-Filho
- Departamento de Clínica Médica, Universidade Estadual de Campinas, UNICAMP, 13081 970, Campinas SP, Brazil
| | | | | | | | | |
Collapse
|
322
|
Abstract
Signal transduction via PI 3-kinases plays an important role in regulating the cellular processes of cell growth, survival, proliferation, and motility. The stimulated generation of reactive oxygen species is a necessary component of the signal transduction mechanisms by which many growth factors and cytokines activate this signaling pathway and elicit their cellular responses. Evidence now supports the oxidative inactivation of both tyrosine phosphatases acting upstream of PI 3-kinase, and of the lipid phosphatase PTEN as components of the normal stimulated regulation of PI 3-kinase signaling. However, the effects of chronic oxidative stress appear rather different, particularly a proposed role for nitrosylation of Akt and other targets leading to inhibition of PI 3-kinase signaling during diabetic insulin resistance in muscle. Recently, evidence has also begun to emerge, indicating that physiological redox signaling may display the same tight spatial and temporal specificity as seen with many other signal transduction systems in terms of targeting individual proteins for modification, and of enzymatic reversal mechanisms. This review will focus upon the details of these and other roles for reactive oxygen and nitrogen species in the regulation of PI 3-kinase signaling, both during acute stimulation and chronic oxidative stress, and the evidence for their significance.
Collapse
Affiliation(s)
- Nick R Leslie
- Division of Molecular Physiology, School of Life Sciences, University of Dundee, Wellcome Trust Biocentre, Dundee, United Kingdom.
| |
Collapse
|
323
|
Jobgen WS, Fried SK, Fu WJ, Meininger CJ, Wu G. Regulatory role for the arginine–nitric oxide pathway in metabolism of energy substrates. J Nutr Biochem 2006; 17:571-88. [PMID: 16524713 DOI: 10.1016/j.jnutbio.2005.12.001] [Citation(s) in RCA: 460] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2005] [Revised: 11/30/2005] [Accepted: 12/02/2005] [Indexed: 12/13/2022]
Abstract
Nitric oxide (NO) is synthesized from L-arginine by NO synthase in virtually all cell types. Emerging evidence shows that NO regulates the metabolism of glucose, fatty acids and amino acids in mammals. As an oxidant, pathological levels of NO inhibit nearly all enzyme-catalyzed reactions through protein oxidation. However, as a signaling molecule, physiological levels of NO stimulate glucose uptake as well as glucose and fatty acid oxidation in skeletal muscle, heart, liver and adipose tissue; inhibit the synthesis of glucose, glycogen, and fat in target tissues (e.g., liver and adipose); and enhance lipolysis in adipocytes. Thus, an inhibition of NO synthesis causes hyperlipidemia and fat accretion in rats, whereas dietary arginine supplementation reduces fat mass in diabetic fatty rats. The putative underlying mechanisms may involve multiple cyclic guanosine-3',5'-monophosphate-dependent pathways. First, NO stimulates the phosphorylation of adenosine-3',5'-monophosphate-activated protein kinase, resulting in (1) a decreased level of malonyl-CoA via inhibition of acetyl-CoA carboxylase and activation of malonyl-CoA decarboxylase and (2) a decreased expression of genes related to lipogenesis and gluconeogenesis (glycerol-3-phosphate acyltransferase, sterol regulatory element binding protein-1c and phosphoenolpyruvate carboxykinase). Second, NO increases the phosphorylation of hormone-sensitive lipase and perilipins, leading to the translocation of the lipase to the neutral lipid droplets and, hence, the stimulation of lipolysis. Third, NO activates expression of peroxisome proliferator-activated receptor-gamma coactivator-1alpha, thereby enhancing mitochondrial biogenesis and oxidative phosphorylation. Fourth, NO increases blood flow to insulin-sensitive tissues, promoting substrate uptake and product removal via the circulation. Modulation of the arginine-NO pathway through dietary supplementation with L-arginine or L-citrulline may aid in the prevention and treatment of the metabolic syndrome in obese humans and companion animals, and in reducing unfavorable fat mass in animals of agricultural importance.
Collapse
|
324
|
Unno Y, Akuta T, Sakamoto YI, Horiuchi S, Akaike T. Nitric oxide-induced downregulation of leptin production by 3T3-L1 adipocytes. Nitric Oxide 2006; 15:125-32. [PMID: 16442319 DOI: 10.1016/j.niox.2005.12.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2005] [Revised: 11/21/2005] [Accepted: 12/10/2005] [Indexed: 02/06/2023]
Abstract
Leptin secreted mainly by adipocytes plays an important role in insulin sensitivity in metabolic syndrome. Inducible nitric oxide synthase (iNOS) in 3T3-L1 adipocytes is induced by lipopolysaccharide (LPS) and several proinflammatory cytokines such as tumor necrosis factor-alpha and interferon-gamma (IFN-gamma). Because the role of iNOS-derived nitric oxide (NO) in adipocyte function has not been fully clarified, the question that we addressed in the present study was whether iNOS-derived NO is involved in regulation of leptin secretion by adipocytes. Incubation of 3T3-L1 adipocytes for 12h with a mixture of IFN-gamma and LPS caused not only a 55% reduction in leptin secretion and a 52% reduction in leptin mRNA, but also significant induction of iNOS at both protein and mRNA levels. Inhibition of leptin secretion that had been induced by the IFN-gamma-LPS mixture was completely nullified by NOS inhibitors such as Nomega-monomethyl-L-arginine and aminoguanidine. Treatment of adipocytes with NO donors such as an NONOate and S-nitrosoglutathione produced an effect on leptin secretion similar to that of the IFN-gamma-LPS mixture. It is likely therefore that NO mediates downregulation of leptin caused by the IFN-gamma-LPS mixture in 3T3-L1 adipocytes, which suggests an important role for NO in adipocyte functions.
Collapse
Affiliation(s)
- Yuka Unno
- Department of Microbiology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | | | | | | | | |
Collapse
|
325
|
Schwartz EA, Reaven PD. Molecular and signaling mechanisms of atherosclerosis in insulin resistance. Endocrinol Metab Clin North Am 2006; 35:525-49, viii. [PMID: 16959584 DOI: 10.1016/j.ecl.2006.06.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Although the prevalence of cardiovascular complications is increased in insulin-resistant individuals, the underlying causes of this link have been elusive. Recent work suggests that several intracellular signal transduction pathways are inappropriately activated by hyperinsulinemia, hyperglycemia, increased free fatty acids, dyslipidemia, various inflammatory cytokines and adipokines--factors that are increased in insulin resistance. Once activated, substantial cross talk occurs between these pathways, especially a self-reinforcing cascade of vascular inflammation and cell dysfunction, greatly increasing the risk and severity of atherosclerosis in the insulin-resistant individual. We review several key cell-signalling pathways, describe how they are activated in they insulin-resistant state and the damage they induce, and discusses possible therapeutic approaches to limit vascular damage.
Collapse
Affiliation(s)
- Eric A Schwartz
- Division of Research, Carl T. Hayden VA Medical Center, 650 East Indian School Road, Phoenix, AZ 85012, USA
| | | |
Collapse
|
326
|
Woo CH, Massett MP, Shishido T, Itoh S, Ding B, McClain C, Che W, Vulapalli SR, Yan C, Abe JI. ERK5 activation inhibits inflammatory responses via peroxisome proliferator-activated receptor delta (PPARdelta) stimulation. J Biol Chem 2006; 281:32164-74. [PMID: 16943204 DOI: 10.1074/jbc.m602369200] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPAR) decrease the production of cytokine and inducible nitric-oxide synthase (iNOS) expression, which are associated with aging-related inflammation and insulin resistance. Recently, the involvement of the induction of heme oxygenase-1 (HO-1) in regulating inflammation has been suggested, but the exact mechanisms for reducing inflammation by HO-1 remains unclear. We found that overexpression of HO-1 and [Ru(CO)(3)Cl(2)](2), a carbon monoxide (CO)-releasing compound, increased not only ERK5 kinase activity, but also its transcriptional activity measured by luciferase assay with the transfection of the Gal4-ERK5 reporter gene. This transcriptional activity is required for coactivation of PPARdelta by ERK5 in C2C12 cells. [Ru(CO)(3)Cl(2)](2) activated PPARdelta transcriptional activity via the MEK5/ERK5 signaling pathway. The inhibition of NF-kappaB activity by ERK5 activation was reversed by a dominant negative form of PPARdelta suggesting that ERK5/PPARdelta activation is required for the anti-inflammatory effects of CO and HO-1. Based on these data, we propose a new mechanism by which CO and HO-1 mediate anti-inflammatory effects via activating ERK5/PPARdelta, and ERK5 mediates CO and HO-1-induced PPARdelta activation via its interaction with PPARdelta.
Collapse
Affiliation(s)
- Chang-Hoon Woo
- Cardiovascular Research Institute, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
327
|
Mélançon S, Bachelard H, Badeau M, Bourgoin F, Pitre M, Larivière R, Nadeau A. Effects of high-sucrose feeding on insulin resistance and hemodynamic responses to insulin in spontaneously hypertensive rats. Am J Physiol Heart Circ Physiol 2006; 290:H2571-81. [PMID: 16443676 DOI: 10.1152/ajpheart.01002.2005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
This study was designed to investigate the effects of a sucrose diet on vascular and metabolic actions of insulin in spontaneously hypertensive rats (SHR). Male SHR were randomized to receive a sucrose or regular chow diet for 4 wk. Age-matched, chow-fed Wistar-Kyoto (WKY) rats were used as normotensive control. In a first series of experiments, the three groups of rats had pulsed Doppler flow probes and intravascular catheters implanted to determine blood pressure, heart rate, and blood flows. Insulin sensitivity was assessed during a euglycemic hyperinsulinemic clamp performed in conscious rats. In a second series of experiments, new groups of rats were used to examine glucose transport activity in isolated muscles and to determine endothelial nitric oxide synthase (eNOS) protein expression in muscles and endothelin content in vascular tissues. Sucrose feeding was shown to markedly enhance the pressor response to insulin and its hindquarter vasoconstrictor effect when compared with chow-fed SHR. A reduction in eNOS protein content in muscle, but no change in vascular endothelin-1 protein, was noted in sucrose-fed SHR when compared with WKY rats, but these changes were not different from those noted in chow-fed SHR. Similar reductions in insulin-stimulated glucose transport were observed in soleus muscles from both groups of SHR when compared with WKY rats. In extensor digitorum longus muscles, a significant reduction in insulin-stimulated glucose transport was only seen in sucrose-fed rats when compared with the other two groups. Environmental factors, that is, high intake of simple sugars, could possibly potentiate the genetic predisposition in SHR to endothelial dysfunction and insulin resistance.
Collapse
Affiliation(s)
- Sébastien Mélançon
- Department of Medicine and Lipid Research Unit, Centre de Recherche du CHUL, Centre Hospitalier Universitaire de Québec, 2705 Blvd. Laurier, Ste-Foy, Québec, Canada, G1V 4G2
| | | | | | | | | | | | | |
Collapse
|
328
|
Linscheid P, Seboek D, Zulewski H, Scherberich A, Blau N, Keller U, Müller B. Cytokine-induced metabolic effects in human adipocytes are independent of endogenous nitric oxide. Am J Physiol Endocrinol Metab 2006; 290:E1068-77. [PMID: 16380391 DOI: 10.1152/ajpendo.00374.2005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Nitric oxide (NO) has been recognized as a potential mediator of inflammation-induced metabolic alterations, including insulin resistance. However, expression mechanisms and potential roles of endothelial and inducible NO synthases (eNOS and iNOS, respectively) in human adipocytes are poorly understood. In the present study, we aimed to analyze several aspects of NO-related gene expression and metabolite synthesis in basal and inflammation-activated human adipocyte models. eNOS mRNA was highly expressed in omental and to a lesser extent in human subcutaneous adipose tissue biopsies, but not in purified adipocytes, in mesenchymal stem cell (MSC)- and in preadipocyte-derived adipocytes, respectively. Trace amounts of iNOS mRNA were detected in adipose tissue samples of donors with abdominal infection, as opposed to noninfected subjects. Interferon-gamma, in combination with interleukin-1beta or lipopolysaccharide, evoked a transient (4 h < time < 24 h) iNOS mRNA expression in human MSC and preadipocyte-derived adipocytes, respectively. This induction was preceded by cytokine-specific mRNAs. In addition, it was accompanied by an activation of the tetrahydrobiopterin synthesis pathway and by inhibition of peroxisome proliferator-activated receptor-gamma2. In contrast to murine 3T3-L1-derived adipocytes, iNOS protein and NO oxidation products remained undetectable in iNOS mRNA-positive human adipocytes. Accordingly, coadministration of NOS inhibitors (i.e., Nomega-nitro-L-arginine methyl ester, Nomega-monomethyl-L-arginine, and 1400W) had no effects on insulin-mediated glucose uptake and lipolysis. We conclude that, in human adipocytes, endogenous NO is not involved in metabolic regulation during either basal or cytokine-activated conditions.
Collapse
Affiliation(s)
- Philippe Linscheid
- Dept. of Research, Univ. Hospital Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland.
| | | | | | | | | | | | | |
Collapse
|
329
|
Luque RM, Kineman RD. Impact of obesity on the growth hormone axis: evidence for a direct inhibitory effect of hyperinsulinemia on pituitary function. Endocrinology 2006; 147:2754-63. [PMID: 16513828 DOI: 10.1210/en.2005-1549] [Citation(s) in RCA: 114] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
There is a negative relationship between obesity and GH. However, it is not known how metabolic changes, associated with obesity, lead to a reduction in GH output. This study examined the GH axis of two mouse models of obesity, the leptin-deficient (ob/ob) mouse and the diet-induced obese (DIO; high-fat fed) mouse. Both models displayed hyperglycemia and hyperinsulinemia with reduced expression of GH as well as reduced expression of pituitary receptors important for GH synthesis and release [GHRH receptor (DIO only) and the ghrelin receptor (ob/ob and DIO)]. These pituitary changes were not accompanied by changes in hypothalamic expression of GHRH or somatostatin; suggesting that alterations in pituitary function may be precipitated in part by direct effects of systemic signals. Of the metabolic and hormonal parameters examined (insulin, glucose, corticosterone, free fatty acids, ghrelin, and IGF-I), only insulin/glucose showed a significant, and negative, correlation with pituitary expression. Pituitaries of DIO mice remained responsive to the acute in vivo actions of insulin, as assessed by phosphorylation of Akt, despite systemic (skeletal muscle and fat) insulin resistance. In addition, treating primary pituitary cell cultures from lean mice with insulin reduced GH release as well as GH, GHRH receptor, and ghrelin receptor mRNA levels compared with vehicle-treated controls, where the magnitude of suppression of pituitary mRNA levels was similar to that observed in the DIO mouse. These results coupled with the fact that the pituitary expresses the insulin receptor at levels comparable to tissues classically considered insulin sensitive, indicates high circulating insulin levels can directly contribute to the suppression of GH synthesis and release in the obese state.
Collapse
Affiliation(s)
- Raul M Luque
- Section of Endocrinology and Metabolism, Department of Medicine, University of Illinois at Chicago, Illinois, USA
| | | |
Collapse
|
330
|
Dubois MJ, Bergeron S, Kim HJ, Dombrowski L, Perreault M, Fournès B, Faure R, Olivier M, Beauchemin N, Shulman GI, Siminovitch KA, Kim JK, Marette A. The SHP-1 protein tyrosine phosphatase negatively modulates glucose homeostasis. Nat Med 2006; 12:549-56. [PMID: 16617349 DOI: 10.1038/nm1397] [Citation(s) in RCA: 112] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2005] [Accepted: 03/17/2006] [Indexed: 12/17/2022]
Abstract
The protein tyrosine phosphatase SHP-1 is a well-known inhibitor of activation-promoting signaling cascades in hematopoietic cells but its potential role in insulin target tissues is unknown. Here we show that Ptpn6(me-v/me-v) (also known as viable motheaten) mice bearing a functionally deficient SHP-1 protein are markedly glucose tolerant and insulin sensitive as compared to wild-type littermates, as a result of enhanced insulin receptor signaling to IRS-PI3K-Akt in liver and muscle. Downregulation of SHP-1 activity in liver of normal mice by adenoviral expression of a catalytically inert mutant of SHP-1, or after small hairpin RNA-mediated SHP-1 silencing, further confirmed this phenotype. Tyrosine phosphorylation of CEACAM1, a modulator of hepatic insulin clearance, and clearance of serum [125I]-insulin were markedly increased in SHP-1-deficient mice or SHP-1-deficient hepatic cells in vitro. These findings show a novel role for SHP-1 in the regulation of glucose homeostasis through modulation of insulin signaling in liver and muscle as well as hepatic insulin clearance.
Collapse
Affiliation(s)
- Marie-Julie Dubois
- Department of Anatomy-Physiology and Lipid Research Unit, Laval University Hospital Research Center, 2705 Laurier Boulevard, Québec, Québec G1V 4G2, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
331
|
Chen H. Cellular inflammatory responses: novel insights for obesity and insulin resistance. Pharmacol Res 2006; 53:469-77. [PMID: 16632376 DOI: 10.1016/j.phrs.2006.03.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2005] [Revised: 12/02/2005] [Accepted: 03/13/2006] [Indexed: 01/04/2023]
Abstract
Type 2 diabetes is rapidly becoming a worldwide epidemic. Obesity and sedentary lifestyle are the main environmental causes for the development of insulin resistance and type 2 diabetes. In the past decade, it has been increasingly recognized that obesity and insulin resistance are associated with chronic, low-grade systemic inflammation. This review will cover the recent advances in this field and provide a working model explaining how cellular inflammatory responses arise to cope with obesity-induced metabolic stresses and how these inflammatory responses underlie insulin resistance.
Collapse
Affiliation(s)
- Hong Chen
- Novartis Institutes for BioMedical Research Inc., 100 Technology Square, Cambridge, MA 02139, USA.
| |
Collapse
|
332
|
Greenberg AS, Obin MS. Obesity and the role of adipose tissue in inflammation and metabolism. Am J Clin Nutr 2006; 83:461S-465S. [PMID: 16470013 DOI: 10.1093/ajcn/83.2.461s] [Citation(s) in RCA: 895] [Impact Index Per Article: 47.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Recent discoveries, notably of the hormones leptin and adiponectin, have revised the notion that adipocytes are simply a storage depot for body energy. Instead, adipocytes are also endocrine organs, with multiple metabolic roles in regulating whole-body physiology. Small adipocytes in lean individuals promote metabolic homeostasis; the enlarged adipocytes of obese individuals recruit macrophages and promote inflammation and the release of a range of factors that predispose toward insulin resistance. Exercise activates the AMP-activated protein kinase (AMPK) in muscle and other tissues, a pathway that increases fat oxidation and glucose transport. Importantly, the adipocyte hormones leptin and adiponectin also activate AMPK; remarkably, the same pathway is activated by certain antidiabetic agents such as thiazolidinediones. Increasingly, our understanding of the adipocyte as an endocrine organ is leading to new insights into obesity and health.
Collapse
Affiliation(s)
- Andrew S Greenberg
- Obesity and Metabolism Laboratory, Jean Mayer US Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston 02111-1524,USA.
| | | |
Collapse
|
333
|
Meigs JB, O'donnell CJ, Tofler GH, Benjamin EJ, Fox CS, Lipinska I, Nathan DM, Sullivan LM, D'Agostino RB, Wilson PWF. Hemostatic markers of endothelial dysfunction and risk of incident type 2 diabetes: the Framingham Offspring Study. Diabetes 2006; 55:530-7. [PMID: 16443791 DOI: 10.2337/diabetes.55.02.06.db05-1041] [Citation(s) in RCA: 135] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Endothelial dysfunction may precede development of type 2 diabetes. We tested the hypothesis that elevated levels of hemostatic markers of endothelial dysfunction, plasminogen activator inhibitor-1 (PAI-1) antigen, and von Willebrand factor (vWF) antigen predicted incident diabetes independent of other diabetes risk factors. We followed 2,924 Framingham Offspring subjects (54% women, mean age 54 years) without diabetes at baseline (defined by treatment, fasting plasma glucose > or =7 or 2-h postchallenge glucose > or =11.1 mmol/l) over 7 years for new cases of diabetes (treatment or fasting plasma glucose > or =7.0 mmol/l). We used a series of regression models to estimate relative risks for diabetes per interquartile range (IQR) increase in PAI-1 (IQR 16.8 ng/ml) and vWF (IQR 66.8% of control) conditioned on baseline characteristics. Over follow-up, there were 153 new cases of diabetes. Age- and sex-adjusted relative risks of diabetes were 1.55 per IQR for PAI-1 (95% CI 1.41-1.70) and 1.49 for vWF (1.21-1.85). These effects remained after further adjustment for diabetes risk factors (including physical activity; HDL cholesterol, triglyceride, and blood pressure levels; smoking; parental history of diabetes; use of alcohol, nonsteroidal anti-inflammatory drugs, exogenous estrogen, or hypertension therapy; and impaired glucose tolerance), waist circumference, homeostasis model assessment of insulin resistance, and inflammation (assessed by levels of C-reactive protein): the adjusted relative risks were 1.18 per IQR for PAI-1 (1.01-1.37) and 1.39 for vWF (1.09-1.77). We conclude that in this community-based sample, plasma markers of endothelial dysfunction increased risk of incident diabetes independent of other diabetes risk factors including obesity, insulin resistance, and inflammation.
Collapse
Affiliation(s)
- James B Meigs
- General Medicine Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, 02114, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
334
|
Hotamisligil GS. Role of endoplasmic reticulum stress and c-Jun NH2-terminal kinase pathways in inflammation and origin of obesity and diabetes. Diabetes 2005; 54 Suppl 2:S73-8. [PMID: 16306344 DOI: 10.2337/diabetes.54.suppl_2.s73] [Citation(s) in RCA: 273] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Metabolic and immune systems are the most fundamental requirements for survival, and many metabolic and immune response pathways or nutrient- and pathogen-sensing systems have been evolutionarily highly conserved. Consequently, metabolic and immune pathways are also highly integrated and interdependent. In the past decade, it became apparent that this interface plays a critical role in the pathogenesis of chronic metabolic diseases, particularly obesity and type 2 diabetes. Importantly, the inflammatory component in obesity and diabetes is now firmly established with the discovery of causal links between inflammatory mediators, such as tumor necrosis factor (TNF)-alpha and insulin receptor signaling and the elucidation of the underlying molecular mechanisms, such as c-Jun NH2-terminal kinase (JNK)- and inhibitor of nuclear factor-kappaB kinase-mediated transcriptional and posttranslational modifications that inhibit insulin action. More recently, obesity-induced endoplasmic reticulum stress has been demonstrated to underlie the initiation of obesity-induced JNK activation, inflammatory responses, and generation of peripheral insulin resistance. This article will review the link between stress, inflammation, and metabolic disease, particularly type 2 diabetes, and discuss the mechanistic and therapeutic opportunities that emerge from this platform by focusing on JNK and endoplasmic reticulum stress responses.
Collapse
Affiliation(s)
- Gökhan S Hotamisligil
- Department of Genetics and Complex Diseases, Harvard School of Public Health, Boston, Massachusetts, USA.
| |
Collapse
|
335
|
Wiernsperger NF. Is non-insulin dependent glucose uptake a therapeutic alternative? Part 1: physiology, mechanisms and role of non insulin-dependent glucose uptake in type 2 diabetes. DIABETES & METABOLISM 2005; 31:415-26. [PMID: 16357785 DOI: 10.1016/s1262-3636(07)70212-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Several decades of research for treating type 2 diabetes have yielded new drugs but the actual experience with the available oral antidiabetic compounds clearly shows that therapeutic needs are not matched. This highlights the urgent need for exploring other pathways. All cell types have the capacity to take up glucose independently of insulin, whereby basal but also hyperglycaemia-promoted glucose supply is ensured. Although poorly explored, insulin-independent glucose uptake might nevertheless represent a therapeutic target, as an alternative to the clear limits of actual drug treatments. This review not only critically examines some major pathways not requiring insulin (although they may be influenced by the hormone) but importantly, this analysis extends to the clinical applicability of these potential therapeutic principles by also considering their predictable tolerability for long-term therapy. In particular vascular safety (the ultimate problem linked with diabetes) will be envisaged because of the ubiquitous distribution of glucose transporters and some linked mechanisms. Several mechanisms can be identified which do not require insulin for their functioning. The first part of this review deals with the description, the regulation and the limits of some mechanisms representing potential pharmacological targets capable of having a highly significant impact on glucose uptake. These selected topics are: a) unmasking and/or activation of glucose transporters in cell plasma membranes, b) insulin mimetics acting at postreceptor level, c) activation of AMPK, d) increasing nitric oxide and e) increasing glucose-6P and glycogen stores.
Collapse
Affiliation(s)
- N F Wiernsperger
- INSERM UMR 585, Bâtiment Louis Pasteur, INSA Lyon, Cedex, France.
| |
Collapse
|
336
|
Schmidt MI, Saad MJ, Duncan BB. Subclinical inflammation and obesity, diabetes and related disorders. ACTA ACUST UNITED AC 2005. [DOI: 10.1016/j.ddmec.2005.08.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
337
|
Okon EB, Chung AWY, Rauniyar P, Padilla E, Tejerina T, McManus BM, Luo H, van Breemen C. Compromised arterial function in human type 2 diabetic patients. Diabetes 2005; 54:2415-23. [PMID: 16046309 DOI: 10.2337/diabetes.54.8.2415] [Citation(s) in RCA: 108] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Diabetes is associated with a perturbation of signaling pathways in vascular tissue, which causes vasomotor dysfunction such as hypertension and accelerated atherosclerosis. In the present study, the mechanisms of vasomotor dysfunction, Akt (Thr308 and Ser473) phosphorylation and expression of endothelial NO (nitric oxide) synthase, and inducible NO synthase were investigated in human diabetic internal mammary arteries. The phospho-Akt (Thr308) level in arteries from diabetic patients was reduced to about one-half of the level in nondiabetic patients, suggesting impaired insulin signaling in human diabetic vascular tissue. Augmented vasoconstriction was observed in diabetic arteries, due in part to deficiency of basal and stimulated NO production. This correlated with decreased endothelial NO synthase expression and activity in diabetic vessels. The sensitivity of diabetic vessels to the NO donor, sodium nitroprusside, was reduced as well, suggesting that NO breakdown and/or decreased sensitivity of smooth muscle to NO are also responsible for abnormal vasoconstriction. In addition, the abnormal vasoconstriction in diabetic vessels was not completely abolished in the presence of Nomega-nitro-L-arginine methyl ester, revealing that NO-independent mechanisms also contribute to vasomotor dysfunction in diabetes. In conclusion, diabetes downregulates the Akt-signaling pathway and compromises human arterial function through a decrease in NO availability as well as through NO-independent mechanisms.
Collapse
Affiliation(s)
- Elena B Okon
- James Hogg iCAPTURE Center, St. Paul's Hospital, Room 166, 1081 Burrard St., Vancouver, BC, Canada V6Z 1Y6.
| | | | | | | | | | | | | | | |
Collapse
|
338
|
Abstract
Over the last decade, an abundance of evidence has emerged demonstrating a close link between metabolism and immunity. It is now clear that obesity is associated with a state of chronic low-level inflammation. In this article, we discuss the molecular and cellular underpinnings of obesity-induced inflammation and the signaling pathways at the intersection of metabolism and inflammation that contribute to diabetes. We also consider mechanisms through which the inflammatory response may be initiated and discuss the reasons for the inflammatory response in obesity. We put forth for consideration some hypotheses regarding important unanswered questions in the field and suggest a model for the integration of inflammatory and metabolic pathways in metabolic disease.
Collapse
Affiliation(s)
- Kathryn E Wellen
- Department of Genetics & Complex Diseases, Harvard School of Public Health, Boston, Massachusetts 02115, USA
| | | |
Collapse
|
339
|
Abstract
Over the last decade, an abundance of evidence has emerged demonstrating a close link between metabolism and immunity. It is now clear that obesity is associated with a state of chronic low-level inflammation. In this article, we discuss the molecular and cellular underpinnings of obesity-induced inflammation and the signaling pathways at the intersection of metabolism and inflammation that contribute to diabetes. We also consider mechanisms through which the inflammatory response may be initiated and discuss the reasons for the inflammatory response in obesity. We put forth for consideration some hypotheses regarding important unanswered questions in the field and suggest a model for the integration of inflammatory and metabolic pathways in metabolic disease.
Collapse
Affiliation(s)
- Kathryn E Wellen
- Department of Genetics & Complex Diseases, Harvard School of Public Health, Boston, Massachusetts 02115, USA
| | | |
Collapse
|
340
|
Abstract
Over the last decade, an abundance of evidence has emerged demonstrating a close link between metabolism and immunity. It is now clear that obesity is associated with a state of chronic low-level inflammation. In this article, we discuss the molecular and cellular underpinnings of obesity-induced inflammation and the signaling pathways at the intersection of metabolism and inflammation that contribute to diabetes. We also consider mechanisms through which the inflammatory response may be initiated and discuss the reasons for the inflammatory response in obesity. We put forth for consideration some hypotheses regarding important unanswered questions in the field and suggest a model for the integration of inflammatory and metabolic pathways in metabolic disease.
Collapse
Affiliation(s)
- Kathryn E Wellen
- Department of Genetics & Complex Diseases, Harvard School of Public Health, Boston, Massachusetts 02115, USA
| | | |
Collapse
|
341
|
Abstract
Reactive oxygen and nitrogen species (ROS and RNS) recently emerged as critical signaling molecules in cardiovascular research. Several studies over the past decade have shown that physiological effects of vasoactive factors are mediated by these reactive species and, conversely, that altered redox mechanisms are implicated in the occurrence of metabolic and cardiovascular diseases. Oxidant stress occurs when ROS and/or RNS production exceeds the cell natural antioxidant systems, and pathological events ensue. Cardiovascular risk factors are associated with an imbalance of the redox equilibrium toward oxidative stress, leading to endothelial activation and proinflammatory processes implicated in atherogenesis and metabolic disorders. Recent studies indicate that insulin and insulin-sensitizing drugs activate antiinflammatory pathways that may limit oxidant stress in insulin target tissues. The main goal of this brief review is to discuss recent progress in the field of cellular redox signaling as it pertains to insulin modulation of vascular endothelial function in cardiovascular diseases.
Collapse
Affiliation(s)
- Raymond Christon
- Lipides Membranaires et Fonctions Cardiovasculaires, UMR INRA-Université Paris XI, Faculté de Pharmacie, Châtenay-Malabry
| | | | | |
Collapse
|
342
|
Affiliation(s)
- Ashok K Srivastava
- Laboratory of Cell Signaling, Research Centre, Centre hospitalier de l'Université de Montréal (CHUM)-Hôtel-Dieu and Department of Medicine, Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
343
|
Abstract
Subclinical, low-grade systemic inflammation has been observed in patients with type 2 diabetes and in those at increased risk of the disease. This may be more than an epiphenomenon. Alleles of genes encoding immune/inflammatory mediators are associated with the disease, and the two major environmental factors the contribute to the risk of type 2 diabetes-diet and physical activity-have a direct impact on levels of systemic immune mediators. In animal models, targeting of immune genes enhanced or suppressed the development of obesity or diabetes. Obesity is associated with the infiltration and proinflammatory activity of macrophages in adipose tissue, and immune mediators may be important regulators of insulin resistance, mitochondrial function, ectopic lipid storage and beta cell dysfunction or death. Intervention studies targeting these pathways would help to determine the contribution of an activated innate immune system to the development of type 2 diabetes.
Collapse
Affiliation(s)
- H Kolb
- German Diabetes Center, Leibniz-Institute at the University of Düsseldorf, Düsseldorf, Germany.
| | | |
Collapse
|
344
|
Fujimoto M, Shimizu N, Kunii K, Martyn JAJ, Ueki K, Kaneki M. A role for iNOS in fasting hyperglycemia and impaired insulin signaling in the liver of obese diabetic mice. Diabetes 2005; 54:1340-8. [PMID: 15855318 DOI: 10.2337/diabetes.54.5.1340] [Citation(s) in RCA: 143] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Chronic inflammation has been postulated to play an important role in the pathogenesis of insulin resistance. Inducible nitric oxide synthase (iNOS) has been implicated in many human diseases associated with inflammation. iNOS deficiency was shown to prevent high-fat diet-induced insulin resistance in skeletal muscle but not in the liver. A role for iNOS in fasting hyperglycemia and hepatic insulin resistance, however, remains to be investigated in obesity-related diabetes. To address this issue, we examined the effects of a specific inhibitor for iNOS, L-NIL, in obese diabetic (ob/ob) mice. iNOS expression was increased in the liver of ob/ob mice compared with wild-type mice. Treatment with iNOS inhibitor reversed fasting hyperglycemia with concomitant amelioration of hyperinsulinemia and improved insulin sensitivity in ob/ob mice. iNOS inhibitor also increased the protein expression of insulin receptor substrate (IRS)-1 and -2 1.5- and 2-fold, respectively, and enhanced IRS-1- and IRS-2-mediated insulin signaling in the liver of ob/ob mice. Exposure to NO donor and ectopically expressed iNOS decreased the protein expression of IRS-1 and -2 in cultured hepatocytes. These results suggest that iNOS plays a role in fasting hyperglycemia and contributes to hepatic insulin resistance in ob/ob mice.
Collapse
Affiliation(s)
- Masaki Fujimoto
- Department of Anesthesia and Critical Care, Massachusetts General Hospital, Harvard Medical School, 149 13th St., Rm. 6604, Charlestown, MA 02129, USA
| | | | | | | | | | | |
Collapse
|
345
|
Hamel C, Millette E, Lamontagne D. Role of nitric oxide and protein kinase C in the tachyphylaxis to vasopressin in rat aortic rings. Life Sci 2005; 77:1069-81. [PMID: 15978263 DOI: 10.1016/j.lfs.2004.12.039] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2004] [Accepted: 12/10/2004] [Indexed: 11/20/2022]
Abstract
The contribution of endothelium-derived mediators and protein kinase C in the tachyphylaxis to arginine vasopressin (AVP) was assessed in the rat aorta. Endothelium-intact (E+) and denuded rings (E-) obtained from the rat thoracic aorta were exposed to three administrations of a supramaximal concentration of AVP (100 nM), lasting 20 min and 45 min apart. N-Omega-nitro-L-arginine (NNLA), a non-selective inhibitor of all isoforms of NO synthase, and AMT, a selective inhibitor for the inducible (iNOS) and neuronal (nNOS) isoforms, diminished the tachyphylaxis to AVP significantly in both E+ and in E- rings. No iNOS could be detected by Western blots in freshly isolated rings or in rings exposed to AVP, despite a strong signal in rings isolated from LPS-treated rats, while nNOS could be constitutively detected. Inhibition of prostaglandins or epoxyeicosatrienoic acids (EETs) synthesis by diclofenac or clotrimazole, respectively, had no effect on tachyphylaxis while combination of these agents diminished tachyphylaxis in E+ only. Combination of NNLA, diclofenac and clotrimazole blocked completely the tachyphylaxis. Inhibition of PKC by either chelerythrine or bisindolylmaleimide I-HCl (BisI) led to a significant diminution of AVP tachyphylaxis only in E-. Activation of PKC with phorbol-12-myristate-13-acetate (PMA) simulated tachyphylaxis to AVP in E- only, effect blocked by the NO donor, SNP. In conclusion, NO produced from constitutive nNOS present in vascular smooth muscle cells participates in tachyphylaxis to AVP. PKC is involved in this tachyphylaxis only in E- rings, the presence of NO probably diminishing the effects of this kinase.
Collapse
Affiliation(s)
- Christine Hamel
- Faculté de Pharmacie, Université de Montréal, C.P. 6128, Succursale centre-ville, Montréal, QC, Canada H3C 3J7
| | | | | |
Collapse
|
346
|
Carvalho-Filho MA, Ueno M, Hirabara SM, Seabra AB, Carvalheira JBC, de Oliveira MG, Velloso LA, Curi R, Saad MJA. S-nitrosation of the insulin receptor, insulin receptor substrate 1, and protein kinase B/Akt: a novel mechanism of insulin resistance. Diabetes 2005; 54:959-67. [PMID: 15793233 DOI: 10.2337/diabetes.54.4.959] [Citation(s) in RCA: 207] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Evidence demonstrates that exogenous nitric oxide (NO) and the NO produced by inducible nitric oxide synthase (iNOS) can induce insulin resistance in muscle. Here, we investigated whether this insulin resistance could be mediated by S-nitrosation of proteins involved in early steps of the insulin signal transduction pathway. Exogenous NO donated by S-nitrosoglutathione (GSNO) induced in vitro and in vivo S-nitrosation of the insulin receptor beta subunit (IRbeta) and protein kinase B/Akt (Akt) and reduced their kinase activity in muscle. Insulin receptor substrate (IRS)-1 was also rapidly S-nitrosated, and its expression was reduced after chronic GSNO treatment. In two distinct models of insulin resistance associated with enhanced iNOS expression-diet-induced obesity and the ob/ob diabetic mice-we observed enhanced S-nitrosation of IRbeta/IRS-1 and Akt in muscle. Reversal of S-nitrosation of these proteins by reducing iNOS expression yielded an improvement in insulin action in both animal models. Thus, S-nitrosation of proteins involved in insulin signal transduction is a novel molecular mechanism of iNOS-induced insulin resistance.
Collapse
Affiliation(s)
- Marco A Carvalho-Filho
- Department of Internal Medicine, State University of Campinas, UNICAMP, Campinas, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
347
|
Noronha BT, Li JM, Wheatcroft SB, Shah AM, Kearney MT. Inducible nitric oxide synthase has divergent effects on vascular and metabolic function in obesity. Diabetes 2005; 54:1082-9. [PMID: 15793247 DOI: 10.2337/diabetes.54.4.1082] [Citation(s) in RCA: 114] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Previous studies have suggested an involvement of inducible nitric oxide synthase (iNOS) in obesity, but the relation, if any, between this and mechanisms underlying endothelial dysfunction in obesity is unknown. We studied mice fed an obesogenic high-fat or standard diet for up to 8 weeks. Obesity was associated with elevated blood pressure; resistance to the glucoregulatory actions of insulin; resistance to the vascular actions of insulin, assessed as the reduction in phenylephrine constrictor response of aortic rings after insulin preincubation (lean -21.7 +/- 11.5 vs. obese 18.2 +/- 15.5%; P < 0.05); and evidence of reactive oxygen species (ROS)-dependent vasodilatation in response to acetylcholine in aortic rings (change in maximal relaxation to acetylcholine after exposure to catalase: lean -2.1 +/- 6.0 vs. obese -15.0 +/- 3.8%; P = 0.04). Obese mice had increased expression of iNOS in aorta, with evidence of increased vascular NO production, assessed as the increase in maximal constriction to phenylephrine after iNOS inhibition with 1400W (lean -3.5 +/- 9.1 vs. obese 42.1 +/- 11.2%; P < 0.001). To further address the role of iNOS in obesity-induced vascular and metabolic dysfunction, we studied the effect of a high-fat diet in iNOS knockout mice (iNOS KO). Obese iNOS KO mice were protected against the development of resistance to insulin's glucoregulatory and vascular effects (insulin-dependent reduction in maximal phenylephrine response: obese wild-type 11.2 +/- 15.0 vs. obese iNOS KO -20.0 +/- 7.7%; P = 0.02). However, obese iNOS KO mice remained hypertensive (124.0 +/- 0.7 vs. 114.9 +/- 0.5 mmHg; P < 0.01) and had evidence of increased vascular ROS production. Although these data support iNOS as a target to protect against the adverse effects of obesity on glucoregulation and vascular insulin resistance, iNOS inhibition does not prevent the development of raised blood pressure or oxidative stress.
Collapse
|
348
|
Abstract
Advances in adipose tissue biology over the past 10 years have led to an improved understanding of the mechanisms linking obesity with the metabolic syndrome and other complications. Obesity is characterized by a chronic, systemic low-grade state of inflammation. Biomarkers of inflammation, such as the leukocyte count, tumor necrosis factor-alpha (TNF-alpha), interleukin 6 (IL-6), and C-reactive protein, are increased in obesity, associated with insulin resistance, and predict the development of type 2 diabetes and cardiovascular disease. It is now clear that the adipocyte is an active participant in the generation of the inflammatory state in obesity. Adipocytes secrete a variety of cytokines, including IL-6 and TNF-alpha, that promote inflammation. Moreover, recent studies suggest that obesity is associated with an increase in adipose tissue macrophages, which also participate in the inflammatory process through the elaboration of cytokines. An improved understanding of the role of adipose tissue in the activation of inflammatory pathways may suggest novel treatment and prevention strategies aimed at reducing obesity-associated morbidities and mortality.
Collapse
Affiliation(s)
- Yong-Ho Lee
- Diabetes and Metabolism Translational Medicine Unit, Division of Endocrinology and Metabolism, Department of Medicine, University of Vermont College of Medicine, FAHC/UHC--Arnold 3412, One South Prospect Street, Burlington, VT 05401, USA.
| | | |
Collapse
|
349
|
Sugita H, Fujimoto M, Yasukawa T, Shimizu N, Sugita M, Yasuhara S, Martyn JAJ, Kaneki M. Inducible nitric-oxide synthase and NO donor induce insulin receptor substrate-1 degradation in skeletal muscle cells. J Biol Chem 2005; 280:14203-11. [PMID: 15805118 DOI: 10.1074/jbc.m411226200] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Chronic inflammation plays an important role in insulin resistance. Inducible nitric-oxide synthase (iNOS), a mediator of inflammation, has been implicated in many human diseases including insulin resistance. However, the molecular mechanisms by which iNOS mediates insulin resistance remain largely unknown. Here we demonstrate that exposure to NO donor or iNOS transfection reduced insulin receptor substrate (IRS)-1 protein expression without altering the mRNA level in cultured skeletal muscle cells. NO donor increased IRS-1 ubiquitination, and proteasome inhibitors blocked NO donor-induced reduction in IRS-1 expression in cultured skeletal muscle cells. The effect of NO donor on IRS-1 expression was cGMP-independent and accentuated by concomitant oxidative stress, suggesting an involvement of nitrosative stress. Inhibitors for phosphatidylinositol-3 kinase, mammalian target of rapamycin, and c-Jun amino-terminal kinase failed to block NO donor-induced IRS-1 reduction, whereas these inhibitors prevented insulin-stimulated IRS-1 decrease. Moreover iNOS expression was increased in skeletal muscle of diabetic (ob/ob) mice compared with lean wild-type mice. iNOS gene disruption or treatment with iNOS inhibitor ameliorated depressed IRS-1 expression in skeletal muscle of diabetic (ob/ob) mice. These findings indicate that iNOS reduces IRS-1 expression in skeletal muscle via proteasome-mediated degradation and thereby may contribute to obesity-related insulin resistance.
Collapse
Affiliation(s)
- Hiroki Sugita
- Department of Anesthesia and Critical Care, Massachusetts General Hospital, Shriners Hospital for Children, Harvard Medical School, Boston, Massachusetts 02129, USA
| | | | | | | | | | | | | | | |
Collapse
|
350
|
Yasukawa T, Tokunaga E, Ota H, Sugita H, Martyn JAJ, Kaneki M. S-nitrosylation-dependent inactivation of Akt/protein kinase B in insulin resistance. J Biol Chem 2005; 280:7511-8. [PMID: 15632167 DOI: 10.1074/jbc.m411871200] [Citation(s) in RCA: 202] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Inducible nitric-oxide synthase (iNOS) has been implicated in many human diseases including insulin resistance. However, how iNOS causes or exacerbates insulin resistance remains largely unknown. Protein S-nitrosylation is now recognized as a prototype of a redox-dependent, cGMP-independent signaling component that mediates a variety of actions of nitric oxide (NO). Here we describe the mechanism of inactivation of Akt/protein kinase B (PKB) in NO donor-treated cells and diabetic (db/db) mice. NO donors induced S-nitrosylation and inactivation of Akt/PKB in vitro and in intact cells. The inhibitory effects of NO donor were independent of phosphatidylinositol 3-kinase and cGMP. In contrast, the concomitant presence of oxidative stress accelerated S-nitrosylation and inactivation of Akt/PKB. In vitro denitrosylation with reducing agent reactivated recombinant and cellular Akt/PKB from NO donor-treated cells. Mutated Akt1/PKBalpha (C224S), in which cysteine 224 was substituted by serine, was resistant to NO donor-induced S-nitrosylation and inactivation, indicating that cysteine 224 is a major S-nitrosylation acceptor site. In addition, S-nitrosylation of Akt/PKB was increased in skeletal muscle of diabetic (db/db) mice compared with wild-type mice. These data suggest that S-nitrosylation-mediated inactivation may contribute to the pathogenesis of iNOS- and/or oxidative stress-involved insulin resistance.
Collapse
Affiliation(s)
- Takashi Yasukawa
- Department of Anesthesia and Critical Care, Massachusetts General Hospital, Shriners Hospital for Children, Harvard Medical School, Boston, Massachusetts 02114, USA
| | | | | | | | | | | |
Collapse
|