51
|
Virtue A, Johnson C, Lopez-Pastraña J, Shao Y, Fu H, Li X, Li YF, Yin Y, Mai J, Rizzo V, Tordoff M, Bagi Z, Shan H, Jiang X, Wang H, Yang XF. MicroRNA-155 Deficiency Leads to Decreased Atherosclerosis, Increased White Adipose Tissue Obesity, and Non-alcoholic Fatty Liver Disease: A NOVEL MOUSE MODEL OF OBESITY PARADOX. J Biol Chem 2016; 292:1267-1287. [PMID: 27856635 DOI: 10.1074/jbc.m116.739839] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 10/27/2016] [Indexed: 12/17/2022] Open
Abstract
Obesity paradox (OP) describes a widely observed clinical finding of improved cardiovascular fitness and survival in some overweight or obese patients. The molecular mechanisms underlying OP remain enigmatic partly due to a lack of animal models mirroring OP in patients. Using apolipoprotein E knock-out (apoE-/-) mice on a high fat (HF) diet as an atherosclerotic obesity model, we demonstrated 1) microRNA-155 (miRNA-155, miR-155) is significantly up-regulated in the aortas of apoE-/- mice, and miR-155 deficiency in apoE-/- mice inhibits atherosclerosis; 2) apoE-/-/miR-155-/- (double knock-out (DKO)) mice show HF diet-induced obesity, adipocyte hypertrophy, and present with non-alcoholic fatty liver disease; 3) DKO mice demonstrate HF diet-induced elevations of plasma leptin, resistin, fed-state and fasting insulin and increased expression of adipogenic transcription factors but lack glucose intolerance and insulin resistance. Our results are the first to present an OP model using DKO mice with features of decreased atherosclerosis, increased obesity, and non-alcoholic fatty liver disease. Our findings suggest the mechanistic role of reduced miR-155 expression in OP and present a new OP working model based on a single miRNA deficiency in diet-induced obese atherogenic mice. Furthermore, our results serve as a breakthrough in understanding the potential mechanism underlying OP and provide a new biomarker and novel therapeutic target for OP-related metabolic diseases.
Collapse
Affiliation(s)
- Anthony Virtue
- From the Centers for Metabolic Disease Research, Cardiovascular Research and Thrombosis Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania 19140
| | - Candice Johnson
- From the Centers for Metabolic Disease Research, Cardiovascular Research and Thrombosis Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania 19140
| | - Jahaira Lopez-Pastraña
- From the Centers for Metabolic Disease Research, Cardiovascular Research and Thrombosis Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania 19140
| | - Ying Shao
- From the Centers for Metabolic Disease Research, Cardiovascular Research and Thrombosis Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania 19140
| | - Hangfei Fu
- From the Centers for Metabolic Disease Research, Cardiovascular Research and Thrombosis Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania 19140
| | - Xinyuan Li
- From the Centers for Metabolic Disease Research, Cardiovascular Research and Thrombosis Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania 19140
| | - Ya-Feng Li
- From the Centers for Metabolic Disease Research, Cardiovascular Research and Thrombosis Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania 19140
| | - Ying Yin
- From the Centers for Metabolic Disease Research, Cardiovascular Research and Thrombosis Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania 19140
| | - Jietang Mai
- From the Centers for Metabolic Disease Research, Cardiovascular Research and Thrombosis Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania 19140
| | - Victor Rizzo
- From the Centers for Metabolic Disease Research, Cardiovascular Research and Thrombosis Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania 19140
| | - Michael Tordoff
- the Monell Chemical Senses Center, Philadelphia, Pennsylvania 19104, and
| | - Zsolt Bagi
- the Vascular Biology Center, Augusta University, Augusta, Georgia 30912
| | - Huimin Shan
- From the Centers for Metabolic Disease Research, Cardiovascular Research and Thrombosis Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania 19140
| | - Xiaohua Jiang
- From the Centers for Metabolic Disease Research, Cardiovascular Research and Thrombosis Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania 19140
| | - Hong Wang
- From the Centers for Metabolic Disease Research, Cardiovascular Research and Thrombosis Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania 19140
| | - Xiao-Feng Yang
- From the Centers for Metabolic Disease Research, Cardiovascular Research and Thrombosis Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania 19140,
| |
Collapse
|
52
|
McMurray F, Patten DA, Harper ME. Reactive Oxygen Species and Oxidative Stress in Obesity-Recent Findings and Empirical Approaches. Obesity (Silver Spring) 2016; 24:2301-2310. [PMID: 27804267 DOI: 10.1002/oby.21654] [Citation(s) in RCA: 173] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 07/26/2016] [Accepted: 08/01/2016] [Indexed: 12/12/2022]
Abstract
OBJECTIVE High levels of reactive oxygen species (ROS) are intricately linked to obesity and associated pathologies, notably insulin resistance and type 2 diabetes. However, ROS are also thought to be important in intracellular signaling, which may paradoxically be required for insulin sensitivity. Many theories have been developed to explain this apparent paradox, which have broadened our understanding of these important small molecules. While many sites for intracellular ROS production have been described, mitochondrial generated ROS remain a major contributor in most cell types. Mitochondrial ROS generation is controlled by a number of factors described in this review. Moreover, these studies have established both a demand for novel sensitive approaches to measure ROS, as well as a need to standardize and review their suitability for different applications. METHODS To properly assess levels of ROS and mitochondrial ROS in the development of obesity and its complications, a growing number of tools have been developed. This paper reviews many of the common methods for the investigation of ROS in mitochondria, cell, animal, and human models. RESULTS Available approaches can be generally divided into those that measure ROS-induced damage (e.g., DNA, lipid, and protein damage); those that measure antioxidant levels and redox ratios; and those that use novel biosensors and probes for a more direct measure of different forms of ROS (e.g., 2',7'-di-chlorofluorescein (DCF), dihydroethidium (DHE) and its mitochondrial targeted form (MitoSOX), Amplex Red, roGFP, HyPer, mt-cpYFP, ratiometric H2 O2 probes, and their derivatives). Moreover, this review provides caveats and strengths for the use of these techniques in different models. CONCLUSIONS Advances in these techniques will undoubtedly advance the understanding of ROS in obesity and may help resolve unanswered questions in the field.
Collapse
Affiliation(s)
- Fiona McMurray
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - David A Patten
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Mary-Ellen Harper
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada.
| |
Collapse
|
53
|
Jager J, Aparicio-Vergara M, Aouadi M. Liver innate immune cells and insulin resistance: the multiple facets of Kupffer cells. J Intern Med 2016; 280:209-20. [PMID: 26864622 DOI: 10.1111/joim.12483] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Obesity, which affects 600 million adults worldwide, is a major risk factor for type 2 diabetes (T2D) and insulin resistance. Current therapies for these metabolic disorders include weight management by lifestyle intervention or bariatric surgery and pharmacological treatment with the aim of regulating blood glucose. Probably because of their short-term effectiveness, these therapies have not been able to stop the rapidly rising prevalence of T2D over the past decades, highlighting an urgent need to develop new therapeutic strategies. The role of immune cells, such as macrophages, in insulin resistance has been extensively studied. Major advances have been made to elucidate the role of adipose tissue macrophages in these pathogeneses. Recently, anti-inflammatory drugs have been suggested as an alternative treatment for T2D, and clinical trials of these agents are currently ongoing. In addition, results of previous clinical trials using antibodies against inflammatory cytokines, which showed modest effects, are now being rigorously re-evaluated. However, it is still unclear how liver macrophages [termed Kupffer cells (KCs)], which constitute the major source of macrophages in the body, contribute to the development of insulin resistance. In this review, we will discuss the present understanding of the role of liver immune cells in the development of insulin resistance. We will particularly focus on KCs, which could represent an attractive target for the treatment of metabolic diseases.
Collapse
Affiliation(s)
- J Jager
- Department of Medicine, KI/AZ Integrated CardioMetabolic Center, Karolinska Institutet at Karolinska University Hospital Huddinge, C2-84, S-141 86, Stockholm, Sweden
| | - M Aparicio-Vergara
- Department of Medicine, KI/AZ Integrated CardioMetabolic Center, Karolinska Institutet at Karolinska University Hospital Huddinge, C2-84, S-141 86, Stockholm, Sweden
| | - M Aouadi
- Department of Medicine, KI/AZ Integrated CardioMetabolic Center, Karolinska Institutet at Karolinska University Hospital Huddinge, C2-84, S-141 86, Stockholm, Sweden
| |
Collapse
|
54
|
Kumari M, Wang X, Lantier L, Lyubetskaya A, Eguchi J, Kang S, Tenen D, Roh HC, Kong X, Kazak L, Ahmad R, Rosen ED. IRF3 promotes adipose inflammation and insulin resistance and represses browning. J Clin Invest 2016; 126:2839-54. [PMID: 27400129 PMCID: PMC4966307 DOI: 10.1172/jci86080] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 05/12/2016] [Indexed: 02/06/2023] Open
Abstract
The chronic inflammatory state that accompanies obesity is a major contributor to insulin resistance and other dysfunctional adaptations in adipose tissue. Cellular and secreted factors promote the inflammatory milieu of obesity, but the transcriptional pathways that drive these processes are not well described. Although the canonical inflammatory transcription factor NF-κB is considered to be the major driver of adipocyte inflammation, members of the interferon regulatory factor (IRF) family may also play a role in this process. Here, we determined that IRF3 expression is upregulated in the adipocytes of obese mice and humans. Signaling through TLR3 and TLR4, which lie upstream of IRF3, induced insulin resistance in murine adipocytes, while IRF3 knockdown prevented insulin resistance. Furthermore, improved insulin sensitivity in IRF3-deficient mice was associated with reductions in intra-adipose and systemic inflammation in the high fat-fed state, enhanced browning of subcutaneous fat, and increased adipose expression of GLUT4. Taken together, the data indicate that IRF3 is a major transcriptional regulator of adipose inflammation and is involved in maintaining systemic glucose and energy homeostasis.
Collapse
|
55
|
Park SH, Liu Z, Sui Y, Helsley RN, Zhu B, Powell DK, Kern PA, Zhou C. IKKβ Is Essential for Adipocyte Survival and Adaptive Adipose Remodeling in Obesity. Diabetes 2016; 65:1616-29. [PMID: 26993069 PMCID: PMC4878418 DOI: 10.2337/db15-1156] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 03/09/2016] [Indexed: 02/06/2023]
Abstract
IκB kinase β (IKKβ), a central coordinator of inflammatory responses through activation of nuclear factor-κB (NF-κB), has been implicated as a critical molecular link between inflammation and metabolic disorders; however, the role of adipocyte IKKβ in obesity and related metabolic disorders remains elusive. Here we report an essential role of IKKβ in the regulation of adipose remodeling and adipocyte survival in diet-induced obesity. Targeted deletion of IKKβ in adipocytes does not affect body weight, food intake, and energy expenditure but results in an exaggerated diabetic phenotype when challenged with a high-fat diet (HFD). IKKβ-deficient mice have multiple histopathologies in visceral adipose tissue, including increased adipocyte death, amplified macrophage infiltration, and defective adaptive adipose remodeling. Deficiency of IKKβ also leads to increased adipose lipolysis, elevated plasma free fatty acid (FFA) levels, and impaired insulin signaling. Mechanistic studies demonstrated that IKKβ is a key adipocyte survival factor and that IKKβ protects murine and human adipocytes from HFD- or FFA-elicited cell death through NF-κB-dependent upregulation of antiapoptotic proteins and NF-κB-independent inactivation of proapoptotic BAD protein. Our findings establish IKKβ as critical for adipocyte survival and adaptive adipose remodeling in obesity.
Collapse
Affiliation(s)
- Se-Hyung Park
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY
| | - Zun Liu
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY
| | - Yipeng Sui
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY
| | - Robert N Helsley
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY
| | - Beibei Zhu
- Department of Medicine, University of Kentucky, Lexington, KY
| | - David K Powell
- Department of Biomedical Engineering, University of Kentucky, Lexington, KY
| | - Philip A Kern
- Department of Medicine, University of Kentucky, Lexington, KY
| | - Changcheng Zhou
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
| |
Collapse
|
56
|
Stefano GB, Challenger S, Kream RM. Hyperglycemia-associated alterations in cellular signaling and dysregulated mitochondrial bioenergetics in human metabolic disorders. Eur J Nutr 2016; 55:2339-2345. [PMID: 27084094 PMCID: PMC5122622 DOI: 10.1007/s00394-016-1212-2] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 03/31/2016] [Indexed: 12/16/2022]
Abstract
Purpose The severity of untreated or refractory diabetes mellitus has been functionally linked to elevated concentrations of free plasma glucose, clinically defined as hyperglycemia. Operationally, the pathophysiological presentations of prolonged hyperglycemia may be categorized within insulin-dependent and insulin-independent, type 1 and type 2 diabetic phenotypes, respectively. Accordingly, major areas of empirical biomedical research have focused on the elucidation of underlying mechanisms driving key cellular signaling systems that are significantly altered in patients presenting with diabetes-associated chronic hyperglycemia. Methods Presently, we provide a translationally oriented review of key studies evaluating the aberrant effects of hyperglycemia on two major signaling pathways linked to debilitating cellular and systemic effects via targeted disruption of mitochondrial bioenergetics: (1) advanced glycation end-products (AGEs)/and their cognate receptor for advanced glycation end-products (RAGEs), and (2) the hexosamine biosynthetic pathway (HBP). Results In preclinical models, cultured vascular endothelial cells exposed to hyperglycemic glucose concentrations were observed to produce enhanced levels of reactive oxygen species (ROS) functionally linked to increased formation of AGEs and expression of their cognate RAGEs. Importantly, inhibitors of AGEs formation, mitochondrial complex II, or un-couplers of oxidative phosphorylation, were observed to significantly reduce the effects of hyperglycemia on ROS production and cellular damage, thereby establishing a critical linkage to multiple levels of mitochondrial functioning. Hyperglycemia-mediated enhancement of mitochondrial ROS/superoxide production in vascular endothelial cells has been functionally linked to the shunting of glucose into the HBP with resultant long-term activation of pro-inflammatory signaling processes. Additionally, exposure of cultured cells to hyperglycemic conditions resulted in enhanced HBP-mediated inhibition of protein subunits of mitochondrial respiratory complexes I, III, and IV, intimately associated with normative cellular bioenergetics and ATP production. Conclusions Convergent lines of evidence link chronic hyperglycemic conditions to aberrant expression of AGEs/RAGEs and HBP signaling pathways in relation to the pathophysiological formation of ROS and pro-inflammatory processes on the functional dysregulation of mitochondrial bioenergetics.
Collapse
Affiliation(s)
- George B Stefano
- MitoGenetics LLC, 3 Bioscience Park Drive, Suite 307, Farmingdale, NY, 11735, USA.
| | - Sean Challenger
- MitoGenetics LLC, 3 Bioscience Park Drive, Suite 307, Farmingdale, NY, 11735, USA
| | - Richard M Kream
- MitoGenetics LLC, 3 Bioscience Park Drive, Suite 307, Farmingdale, NY, 11735, USA
| |
Collapse
|
57
|
Karkeni E, Astier J, Tourniaire F, El Abed M, Romier B, Gouranton E, Wan L, Borel P, Salles J, Walrand S, Ye J, Landrier JF. Obesity-associated Inflammation Induces microRNA-155 Expression in Adipocytes and Adipose Tissue: Outcome on Adipocyte Function. J Clin Endocrinol Metab 2016; 101:1615-26. [PMID: 26829440 PMCID: PMC4880153 DOI: 10.1210/jc.2015-3410] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT Obesity alters adipose tissue's metabolic and endocrine functions and causes a chronic local and systemic low-grade inflammatory state to develop, generating obesity-associated complications. In the last decade, many entities contributing to and regulating this inflammatory state have been identified, among which are microRNAs. OBJECTIVE This study aimed to identify microRNA regulated in inflamed adipocytes and adipose tissue, and its effect on adipocyte biology. DESIGN AND RESULTS We screened the expression profile of TNFα-treated adipocytes (a major pro-inflammatory protein expressed in obese adipose tissue), and identified miR-155 as the most responsive microRNA. The involvement of TNFα on the basal miR-155 expression was confirmed in the adipose tissue of Tnfa−/− mice where miR-155 was significantly reduced. Also, mice overexpressing p65 or invalidated for p65 in adipose tissue respectively increased and decreased miR-155 expression, in line with the involvement of the nuclear factor κB (NF-κB) pathway in miR-155 induction. miR-155 expression was higher in obese subjects' adipose tissue than in that of normal-weight subjects, and correlated with TNFα expression and body mass index. Gain and loss of function of miR-155 showed its effect on adipocyte function, probably via its ability to target PPARγ mRNA 3′UTR. Interestingly, miR-155 overexpression also resulted in an increased inflammatory state in adipocytes. CONCLUSION Altogether, these data are evidence of a proinflammatory loop mediated by NF-κB and miR-155 that could participate in the amplification of inflammatory status in adipocytes.
Collapse
Affiliation(s)
- Esma Karkeni
- Institut National de Recherche Agronomique (INRA) (E.K., J.A., F.T., M.E.A., B.R., E.G., P.B., J.-F.L.), UMR 1260, F-13385, Marseille, France; Institut National de la Santé et de la Recherche Médicale (INSERM), Unité Mixte de Recherche (UMR) 1062 (E.K., J.A., F.T., M.E.A., B.R., E.G., P.B., J.F.L.), Nutrition, Obésité et Risque Thrombotique F-13385, Marseille, France; Faculté de Médecine (E.K., J.A., F.T., M.E.A., B.R., E.G., P.B., J.-F.L.), Aix-Marseille Université, F-13385, Marseille, France; Pennington Biomedical Research Center (L.W., J.Y.), Louisiana State University System, Baton Rouge, LA 70808; Laboratory of Transplant Immunology, Regeneration Medicine Research Center, Sichuan University, Sichuan Province, 610041 China; and UMR INRA 1019 Unité de Nutrition Humaine (J.S., S.W.), Centre de Recherches INRA de Clermont-Ferrand/Theix, 63122 St-Genès-Champanelle, France
| | - Julien Astier
- Institut National de Recherche Agronomique (INRA) (E.K., J.A., F.T., M.E.A., B.R., E.G., P.B., J.-F.L.), UMR 1260, F-13385, Marseille, France; Institut National de la Santé et de la Recherche Médicale (INSERM), Unité Mixte de Recherche (UMR) 1062 (E.K., J.A., F.T., M.E.A., B.R., E.G., P.B., J.F.L.), Nutrition, Obésité et Risque Thrombotique F-13385, Marseille, France; Faculté de Médecine (E.K., J.A., F.T., M.E.A., B.R., E.G., P.B., J.-F.L.), Aix-Marseille Université, F-13385, Marseille, France; Pennington Biomedical Research Center (L.W., J.Y.), Louisiana State University System, Baton Rouge, LA 70808; Laboratory of Transplant Immunology, Regeneration Medicine Research Center, Sichuan University, Sichuan Province, 610041 China; and UMR INRA 1019 Unité de Nutrition Humaine (J.S., S.W.), Centre de Recherches INRA de Clermont-Ferrand/Theix, 63122 St-Genès-Champanelle, France
| | - Franck Tourniaire
- Institut National de Recherche Agronomique (INRA) (E.K., J.A., F.T., M.E.A., B.R., E.G., P.B., J.-F.L.), UMR 1260, F-13385, Marseille, France; Institut National de la Santé et de la Recherche Médicale (INSERM), Unité Mixte de Recherche (UMR) 1062 (E.K., J.A., F.T., M.E.A., B.R., E.G., P.B., J.F.L.), Nutrition, Obésité et Risque Thrombotique F-13385, Marseille, France; Faculté de Médecine (E.K., J.A., F.T., M.E.A., B.R., E.G., P.B., J.-F.L.), Aix-Marseille Université, F-13385, Marseille, France; Pennington Biomedical Research Center (L.W., J.Y.), Louisiana State University System, Baton Rouge, LA 70808; Laboratory of Transplant Immunology, Regeneration Medicine Research Center, Sichuan University, Sichuan Province, 610041 China; and UMR INRA 1019 Unité de Nutrition Humaine (J.S., S.W.), Centre de Recherches INRA de Clermont-Ferrand/Theix, 63122 St-Genès-Champanelle, France
| | - Mouna El Abed
- Institut National de Recherche Agronomique (INRA) (E.K., J.A., F.T., M.E.A., B.R., E.G., P.B., J.-F.L.), UMR 1260, F-13385, Marseille, France; Institut National de la Santé et de la Recherche Médicale (INSERM), Unité Mixte de Recherche (UMR) 1062 (E.K., J.A., F.T., M.E.A., B.R., E.G., P.B., J.F.L.), Nutrition, Obésité et Risque Thrombotique F-13385, Marseille, France; Faculté de Médecine (E.K., J.A., F.T., M.E.A., B.R., E.G., P.B., J.-F.L.), Aix-Marseille Université, F-13385, Marseille, France; Pennington Biomedical Research Center (L.W., J.Y.), Louisiana State University System, Baton Rouge, LA 70808; Laboratory of Transplant Immunology, Regeneration Medicine Research Center, Sichuan University, Sichuan Province, 610041 China; and UMR INRA 1019 Unité de Nutrition Humaine (J.S., S.W.), Centre de Recherches INRA de Clermont-Ferrand/Theix, 63122 St-Genès-Champanelle, France
| | - Béatrice Romier
- Institut National de Recherche Agronomique (INRA) (E.K., J.A., F.T., M.E.A., B.R., E.G., P.B., J.-F.L.), UMR 1260, F-13385, Marseille, France; Institut National de la Santé et de la Recherche Médicale (INSERM), Unité Mixte de Recherche (UMR) 1062 (E.K., J.A., F.T., M.E.A., B.R., E.G., P.B., J.F.L.), Nutrition, Obésité et Risque Thrombotique F-13385, Marseille, France; Faculté de Médecine (E.K., J.A., F.T., M.E.A., B.R., E.G., P.B., J.-F.L.), Aix-Marseille Université, F-13385, Marseille, France; Pennington Biomedical Research Center (L.W., J.Y.), Louisiana State University System, Baton Rouge, LA 70808; Laboratory of Transplant Immunology, Regeneration Medicine Research Center, Sichuan University, Sichuan Province, 610041 China; and UMR INRA 1019 Unité de Nutrition Humaine (J.S., S.W.), Centre de Recherches INRA de Clermont-Ferrand/Theix, 63122 St-Genès-Champanelle, France
| | - Erwan Gouranton
- Institut National de Recherche Agronomique (INRA) (E.K., J.A., F.T., M.E.A., B.R., E.G., P.B., J.-F.L.), UMR 1260, F-13385, Marseille, France; Institut National de la Santé et de la Recherche Médicale (INSERM), Unité Mixte de Recherche (UMR) 1062 (E.K., J.A., F.T., M.E.A., B.R., E.G., P.B., J.F.L.), Nutrition, Obésité et Risque Thrombotique F-13385, Marseille, France; Faculté de Médecine (E.K., J.A., F.T., M.E.A., B.R., E.G., P.B., J.-F.L.), Aix-Marseille Université, F-13385, Marseille, France; Pennington Biomedical Research Center (L.W., J.Y.), Louisiana State University System, Baton Rouge, LA 70808; Laboratory of Transplant Immunology, Regeneration Medicine Research Center, Sichuan University, Sichuan Province, 610041 China; and UMR INRA 1019 Unité de Nutrition Humaine (J.S., S.W.), Centre de Recherches INRA de Clermont-Ferrand/Theix, 63122 St-Genès-Champanelle, France
| | - Lin Wan
- Institut National de Recherche Agronomique (INRA) (E.K., J.A., F.T., M.E.A., B.R., E.G., P.B., J.-F.L.), UMR 1260, F-13385, Marseille, France; Institut National de la Santé et de la Recherche Médicale (INSERM), Unité Mixte de Recherche (UMR) 1062 (E.K., J.A., F.T., M.E.A., B.R., E.G., P.B., J.F.L.), Nutrition, Obésité et Risque Thrombotique F-13385, Marseille, France; Faculté de Médecine (E.K., J.A., F.T., M.E.A., B.R., E.G., P.B., J.-F.L.), Aix-Marseille Université, F-13385, Marseille, France; Pennington Biomedical Research Center (L.W., J.Y.), Louisiana State University System, Baton Rouge, LA 70808; Laboratory of Transplant Immunology, Regeneration Medicine Research Center, Sichuan University, Sichuan Province, 610041 China; and UMR INRA 1019 Unité de Nutrition Humaine (J.S., S.W.), Centre de Recherches INRA de Clermont-Ferrand/Theix, 63122 St-Genès-Champanelle, France
| | - Patrick Borel
- Institut National de Recherche Agronomique (INRA) (E.K., J.A., F.T., M.E.A., B.R., E.G., P.B., J.-F.L.), UMR 1260, F-13385, Marseille, France; Institut National de la Santé et de la Recherche Médicale (INSERM), Unité Mixte de Recherche (UMR) 1062 (E.K., J.A., F.T., M.E.A., B.R., E.G., P.B., J.F.L.), Nutrition, Obésité et Risque Thrombotique F-13385, Marseille, France; Faculté de Médecine (E.K., J.A., F.T., M.E.A., B.R., E.G., P.B., J.-F.L.), Aix-Marseille Université, F-13385, Marseille, France; Pennington Biomedical Research Center (L.W., J.Y.), Louisiana State University System, Baton Rouge, LA 70808; Laboratory of Transplant Immunology, Regeneration Medicine Research Center, Sichuan University, Sichuan Province, 610041 China; and UMR INRA 1019 Unité de Nutrition Humaine (J.S., S.W.), Centre de Recherches INRA de Clermont-Ferrand/Theix, 63122 St-Genès-Champanelle, France
| | - Jérôme Salles
- Institut National de Recherche Agronomique (INRA) (E.K., J.A., F.T., M.E.A., B.R., E.G., P.B., J.-F.L.), UMR 1260, F-13385, Marseille, France; Institut National de la Santé et de la Recherche Médicale (INSERM), Unité Mixte de Recherche (UMR) 1062 (E.K., J.A., F.T., M.E.A., B.R., E.G., P.B., J.F.L.), Nutrition, Obésité et Risque Thrombotique F-13385, Marseille, France; Faculté de Médecine (E.K., J.A., F.T., M.E.A., B.R., E.G., P.B., J.-F.L.), Aix-Marseille Université, F-13385, Marseille, France; Pennington Biomedical Research Center (L.W., J.Y.), Louisiana State University System, Baton Rouge, LA 70808; Laboratory of Transplant Immunology, Regeneration Medicine Research Center, Sichuan University, Sichuan Province, 610041 China; and UMR INRA 1019 Unité de Nutrition Humaine (J.S., S.W.), Centre de Recherches INRA de Clermont-Ferrand/Theix, 63122 St-Genès-Champanelle, France
| | - Stéphane Walrand
- Institut National de Recherche Agronomique (INRA) (E.K., J.A., F.T., M.E.A., B.R., E.G., P.B., J.-F.L.), UMR 1260, F-13385, Marseille, France; Institut National de la Santé et de la Recherche Médicale (INSERM), Unité Mixte de Recherche (UMR) 1062 (E.K., J.A., F.T., M.E.A., B.R., E.G., P.B., J.F.L.), Nutrition, Obésité et Risque Thrombotique F-13385, Marseille, France; Faculté de Médecine (E.K., J.A., F.T., M.E.A., B.R., E.G., P.B., J.-F.L.), Aix-Marseille Université, F-13385, Marseille, France; Pennington Biomedical Research Center (L.W., J.Y.), Louisiana State University System, Baton Rouge, LA 70808; Laboratory of Transplant Immunology, Regeneration Medicine Research Center, Sichuan University, Sichuan Province, 610041 China; and UMR INRA 1019 Unité de Nutrition Humaine (J.S., S.W.), Centre de Recherches INRA de Clermont-Ferrand/Theix, 63122 St-Genès-Champanelle, France
| | - Jianping Ye
- Institut National de Recherche Agronomique (INRA) (E.K., J.A., F.T., M.E.A., B.R., E.G., P.B., J.-F.L.), UMR 1260, F-13385, Marseille, France; Institut National de la Santé et de la Recherche Médicale (INSERM), Unité Mixte de Recherche (UMR) 1062 (E.K., J.A., F.T., M.E.A., B.R., E.G., P.B., J.F.L.), Nutrition, Obésité et Risque Thrombotique F-13385, Marseille, France; Faculté de Médecine (E.K., J.A., F.T., M.E.A., B.R., E.G., P.B., J.-F.L.), Aix-Marseille Université, F-13385, Marseille, France; Pennington Biomedical Research Center (L.W., J.Y.), Louisiana State University System, Baton Rouge, LA 70808; Laboratory of Transplant Immunology, Regeneration Medicine Research Center, Sichuan University, Sichuan Province, 610041 China; and UMR INRA 1019 Unité de Nutrition Humaine (J.S., S.W.), Centre de Recherches INRA de Clermont-Ferrand/Theix, 63122 St-Genès-Champanelle, France
| | - Jean-François Landrier
- Institut National de Recherche Agronomique (INRA) (E.K., J.A., F.T., M.E.A., B.R., E.G., P.B., J.-F.L.), UMR 1260, F-13385, Marseille, France; Institut National de la Santé et de la Recherche Médicale (INSERM), Unité Mixte de Recherche (UMR) 1062 (E.K., J.A., F.T., M.E.A., B.R., E.G., P.B., J.F.L.), Nutrition, Obésité et Risque Thrombotique F-13385, Marseille, France; Faculté de Médecine (E.K., J.A., F.T., M.E.A., B.R., E.G., P.B., J.-F.L.), Aix-Marseille Université, F-13385, Marseille, France; Pennington Biomedical Research Center (L.W., J.Y.), Louisiana State University System, Baton Rouge, LA 70808; Laboratory of Transplant Immunology, Regeneration Medicine Research Center, Sichuan University, Sichuan Province, 610041 China; and UMR INRA 1019 Unité de Nutrition Humaine (J.S., S.W.), Centre de Recherches INRA de Clermont-Ferrand/Theix, 63122 St-Genès-Champanelle, France
| |
Collapse
|
58
|
Geng S, Zhu W, Xie C, Li X, Wu J, Liang Z, Xie W, Zhu J, Huang C, Zhu M, Wu R, Zhong C. Medium-chain triglyceride ameliorates insulin resistance and inflammation in high fat diet-induced obese mice. Eur J Nutr 2016; 55:931-940. [PMID: 25911003 DOI: 10.1007/s00394-015-0907-0] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 04/15/2015] [Indexed: 02/06/2023]
Abstract
PURPOSE The aim of the present study was to investigate the in vivo effects of dietary medium-chain triglyceride (MCT) on inflammation and insulin resistance as well as the underlying potential molecular mechanisms in high fat diet-induced obese mice. METHODS Male C57BL/6J mice (n = 24) were fed one of the following three diets for a period of 12 weeks: (1) a modified AIN-76 diet with 5 % corn oil (normal diet); (2) a high-fat control diet (17 % w/w lard and 3 % w/w corn oil, HFC); (3) an isocaloric high-fat diet supplemented with MCT (17 % w/w MCT and 3 % w/w corn oil, HF-MCT). Glucose metabolism was evaluated by fasting blood glucose levels and intraperitoneal glucose tolerance test. Insulin sensitivity was evaluated by fasting serum insulin levels and the index of homeostasis model assessment-insulin resistance. The levels of serum interleukin-6 (IL-6), interleukin-10 (IL-10), and tumor necrosis factor-α were measured by ELISA, and hepatic activation of nuclear factor κB (NF-κB) and mitogen-activated protein kinase (MAPK) pathways was determined using western blot analysis. RESULTS Compared to HFC diet, consumption of HF-MCT did not induce body weight gain and white adipose tissue accumulation in mice. HFC-induced increases in serum fasting glucose and insulin levels as well as glucose intolerance were prevented by HF-MCT diet. Meanwhile, HF-MCT resulted in significantly lower serum IL-6 level and higher IL-10 level, and lower expression levels of inducible nitric oxide synthase and cyclooxygenase-2 protein in liver tissues when compared to HFC. In addition, HF-MCT attenuated HFC-triggered hepatic activation of NF-κB and p38 MAPK. CONCLUSIONS Our study demonstrated that MCT was efficacious in suppressing body fat accumulation, insulin resistance, inflammatory response, and NF-κB and p38 MAPK activation in high fat diet-fed mice. These data suggest that MCT may exert beneficial effects against high fat diet-induced insulin resistance and inflammation.
Collapse
Affiliation(s)
- Shanshan Geng
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Weiwei Zhu
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Chunfeng Xie
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Xiaoting Li
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Jieshu Wu
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Zhaofeng Liang
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Wei Xie
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Jianyun Zhu
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Cong Huang
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Mingming Zhu
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Rui Wu
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Caiyun Zhong
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
59
|
Zhang Y, Zhao Z, Ke B, Wan L, Wang H, Ye J. Induction of Posttranslational Modifications of Mitochondrial Proteins by ATP Contributes to Negative Regulation of Mitochondrial Function. PLoS One 2016; 11:e0150454. [PMID: 26930489 PMCID: PMC4773252 DOI: 10.1371/journal.pone.0150454] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 02/15/2016] [Indexed: 02/05/2023] Open
Abstract
It is generally accepted that ATP regulates mitochondrial function through the AMPK signaling pathway. However, the AMPK-independent pathway remains largely unknown. In this study, we investigated ATP surplus in the negative regulation of mitochondrial function with a focus on pyruvate dehydrogenase (PDH) phosphorylation and protein acetylation. PDH phosphorylation was induced by a high fat diet in the liver of obese mice, which was associated with ATP elevation. In 1c1c7 hepatoma cells, the phosphorylation was induced by palmitate treatment through induction of ATP production. The phosphorylation was associated with a reduction in mitochondria oxygen consumption after 4 h treatment. The palmitate effect was blocked by etomoxir, which inhibited ATP production through suppression of fatty acid β-oxidation. The PDH phosphorylation was induced by incubation of mitochondrial lysate with ATP in vitro without altering the expression of PDH kinase 2 (PDK2) and 4 (PDK4). In addition, acetylation of multiple mitochondrial proteins was induced by ATP in the same conditions. Acetyl-CoA exhibited a similar activity to ATP in induction of the phosphorylation and acetylation. These data suggest that ATP elevation may inhibit mitochondrial function through induction of the phosphorylation and acetylation of mitochondrial proteins. The results suggest an AMPK-independent mechanism for ATP regulation of mitochondrial function.
Collapse
Affiliation(s)
- Yong Zhang
- Antioxidant and Gene Regulation Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, United States of America
| | - Zhiyun Zhao
- Antioxidant and Gene Regulation Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, United States of America
| | - Bilun Ke
- Antioxidant and Gene Regulation Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, United States of America
| | - Lin Wan
- Antioxidant and Gene Regulation Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, United States of America
- Laboratory of Transplantation Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Hui Wang
- Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine in Henan Province, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, China
| | - Jianping Ye
- Antioxidant and Gene Regulation Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, United States of America
- * E-mail:
| |
Collapse
|
60
|
García-Martín R, Alexaki VI, Qin N, Rubín de Celis MF, Economopoulou M, Ziogas A, Gercken B, Kotlabova K, Phieler J, Ehrhart-Bornstein M, Bornstein SR, Eisenhofer G, Breier G, Blüher M, Hampe J, El-Armouche A, Chatzigeorgiou A, Chung KJ, Chavakis T. Adipocyte-Specific Hypoxia-Inducible Factor 2α Deficiency Exacerbates Obesity-Induced Brown Adipose Tissue Dysfunction and Metabolic Dysregulation. Mol Cell Biol 2016; 36:376-393. [PMID: 26572826 PMCID: PMC4719429 DOI: 10.1128/mcb.00430-15] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 05/21/2015] [Accepted: 11/05/2015] [Indexed: 12/04/2022] Open
Abstract
Angiogenesis is a central regulator for white (WAT) and brown (BAT) adipose tissue adaptation in the course of obesity. Here we show that deletion of hypoxia-inducible factor 2α (HIF2α) in adipocytes (by using Fabp4-Cre transgenic mice) but not in myeloid or endothelial cells negatively impacted WAT angiogenesis and promoted WAT inflammation, WAT dysfunction, hepatosteatosis, and systemic insulin resistance in obesity. Importantly, adipocyte HIF2α regulated vascular endothelial growth factor (VEGF) expression and angiogenesis of obese BAT as well as its thermogenic function. Consistently, obese adipocyte-specific HIF2α-deficient mice displayed BAT dysregulation, associated with reduced levels of uncoupling protein 1 (UCP1) and a dysfunctional thermogenic response to cold exposure. VEGF administration reversed WAT and BAT inflammation and BAT dysfunction in adipocyte HIF2α-deficient mice. Together, our findings show that adipocyte HIF2α is protective against maladaptation to obesity and metabolic dysregulation by promoting angiogenesis in both WAT and BAT and by counteracting obesity-mediated BAT dysfunction.
Collapse
Affiliation(s)
- Rubén García-Martín
- Department of Clinical Pathobiochemistry, Medical Faculty, Technische Universität Dresden, Dresden, Germany
| | - Vasileia I Alexaki
- Department of Clinical Pathobiochemistry, Medical Faculty, Technische Universität Dresden, Dresden, Germany
| | - Nan Qin
- Institute of Clinical Chemistry and Laboratory Medicine, Medical Faculty, Technische Universität Dresden, Dresden, Germany
| | - María F Rubín de Celis
- Department of Medicine III, Medical Faculty, Technische Universität Dresden, Dresden, Germany
| | - Matina Economopoulou
- Department of Ophthalmology, Medical Faculty, Technische Universität Dresden, Dresden, Germany
| | - Athanasios Ziogas
- Department of Clinical Pathobiochemistry, Medical Faculty, Technische Universität Dresden, Dresden, Germany
| | - Bettina Gercken
- Department of Clinical Pathobiochemistry, Medical Faculty, Technische Universität Dresden, Dresden, Germany
| | - Klara Kotlabova
- Department of Clinical Pathobiochemistry, Medical Faculty, Technische Universität Dresden, Dresden, Germany
| | - Julia Phieler
- Department of Clinical Pathobiochemistry, Medical Faculty, Technische Universität Dresden, Dresden, Germany
| | - Monika Ehrhart-Bornstein
- Department of Medicine III, Medical Faculty, Technische Universität Dresden, Dresden, Germany Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Stefan R Bornstein
- Department of Medicine III, Medical Faculty, Technische Universität Dresden, Dresden, Germany
| | - Graeme Eisenhofer
- Institute of Clinical Chemistry and Laboratory Medicine, Medical Faculty, Technische Universität Dresden, Dresden, Germany
| | - Georg Breier
- Department of Psychiatry, Medical Faculty, Technische Universität Dresden, Dresden, Germany Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Matthias Blüher
- Department of Endocrinology and Nephrology, University of Leipzig, Leipzig, Germany
| | - Jochen Hampe
- Department of Medicine I, Medical Faculty, Technische Universität Dresden, Dresden, Germany
| | - Ali El-Armouche
- Department of Pharmacology and Toxicology, Medical Faculty, Technische Universität Dresden, Dresden, Germany
| | - Antonios Chatzigeorgiou
- Department of Clinical Pathobiochemistry, Medical Faculty, Technische Universität Dresden, Dresden, Germany Institute of Clinical Chemistry and Laboratory Medicine, Medical Faculty, Technische Universität Dresden, Dresden, Germany Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine, TU Dresden, Dresden, and German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Kyoung-Jin Chung
- Department of Clinical Pathobiochemistry, Medical Faculty, Technische Universität Dresden, Dresden, Germany
| | - Triantafyllos Chavakis
- Department of Clinical Pathobiochemistry, Medical Faculty, Technische Universität Dresden, Dresden, Germany Institute of Clinical Chemistry and Laboratory Medicine, Medical Faculty, Technische Universität Dresden, Dresden, Germany Center for Regenerative Therapies Dresden, Dresden, Germany Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine, TU Dresden, Dresden, and German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| |
Collapse
|
61
|
Xu X, Lu L, Dong Q, Li X, Zhang N, Xin Y, Xuan S. Research advances in the relationship between nonalcoholic fatty liver disease and atherosclerosis. Lipids Health Dis 2015; 14:158. [PMID: 26631018 PMCID: PMC4668687 DOI: 10.1186/s12944-015-0141-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 10/22/2015] [Indexed: 02/08/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a metabolic stress-induced liver disease that is closely related not only to genetic susceptibility but also to insulin resistance and highly linked with metabolic syndrome. In recent years, the prevalence of NAFLD has increased rapidly, paralleling the epidemic of type 2 diabetes mellitus and obesity leading to cardiovascular disease. It has been demonstrated that NAFLD is highly associated with atherosclerosis. With recently gained knowledge, it appears that NAFLD may induce insulin resistance, dyslipidemia, oxidative stress, inflammation, and fluctuation of adipokines associated with atherosclerosis. In this review, we aimed to summarize recent discoveries related to both NAFLD and atherosclerosis, and to identify possible mechanisms linking them.
Collapse
Affiliation(s)
- Xin Xu
- Department of Gastroenterology, Qingdao Municipal Hospital, Dalian Medical University, Qingdao, China.,Digestive Disease Key Laboratory of Qingdao, Qingdao, China
| | - Linlin Lu
- Digestive Disease Key Laboratory of Qingdao, Qingdao, China.,Central Laboratories, Qingdao Municipal Hospital, Qingdao, China
| | - Quanyong Dong
- Department of Gastroenterology, Qingdao Municipal Hospital, Dalian Medical University, Qingdao, China.,Digestive Disease Key Laboratory of Qingdao, Qingdao, China
| | - Xiaolin Li
- Department of Gastroenterology, Qingdao Municipal Hospital, Dalian Medical University, Qingdao, China.,Digestive Disease Key Laboratory of Qingdao, Qingdao, China
| | - Nannan Zhang
- Department of Gastroenterology, Qingdao Municipal Hospital, Dalian Medical University, Qingdao, China.,Digestive Disease Key Laboratory of Qingdao, Qingdao, China
| | - Yongning Xin
- Department of Gastroenterology, Qingdao Municipal Hospital, Dalian Medical University, Qingdao, China. .,Digestive Disease Key Laboratory of Qingdao, Qingdao, China. .,Central Laboratories, Qingdao Municipal Hospital, Qingdao, China.
| | - Shiying Xuan
- Department of Gastroenterology, Qingdao Municipal Hospital, Dalian Medical University, Qingdao, China. .,Digestive Disease Key Laboratory of Qingdao, Qingdao, China. .,Central Laboratories, Qingdao Municipal Hospital, Qingdao, China.
| |
Collapse
|
62
|
Ke B, Zhao Z, Ye X, Gao Z, Manganiello V, Wu B, Ye J. Inactivation of NF-κB p65 (RelA) in Liver Improves Insulin Sensitivity and Inhibits cAMP/PKA Pathway. Diabetes 2015; 64:3355-62. [PMID: 26038580 PMCID: PMC4587638 DOI: 10.2337/db15-0242] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2015] [Accepted: 05/20/2015] [Indexed: 01/07/2023]
Abstract
The transcription factor nuclear factor-κB (NF-κB) mediates inflammation and stress signals in cells. To test NF-κB in the control of hepatic insulin sensitivity, we inactivated NF-κB in the livers of C57BL/6 mice through deletion of the p65 gene, which was achieved by crossing floxed-p65 and Alb-cre mice to generate L-p65-knockout (KO) mice. KO mice did not exhibit any alterations in growth, reproduction, and body weight while on a chow diet. However, the mice on a high-fat diet (HFD) exhibited an improvement in systemic insulin sensitivity. Hepatic insulin sensitivity was enhanced as indicated by increased pyruvate tolerance, Akt phosphorylation, and decreased gene expression in hepatic gluconeogenesis. In the liver, a decrease in intracellular cAMP was observed with decreased CREB phosphorylation. Cyclic nucleotide phosphodiesterase-3B (PDE3B), a cAMP-degrading enzyme, was increased in mRNA and protein as a result of the absence of NF-κB activity. NF-κB was found to inhibit PDE3B transcription through three DNA-binding sites in the gene promoter in response to tumor necrosis factor-α. Body composition, food intake, energy expenditure, and systemic and hepatic inflammation were not significantly altered in KO mice on HFD. These data suggest that NF-κB inhibits hepatic insulin sensitivity by upregulating cAMP through suppression of PDE3B gene transcription.
Collapse
Affiliation(s)
- Bilun Ke
- Department of Gastroenterology, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China Antioxidant and Gene Regulation Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA
| | - Zhiyun Zhao
- Antioxidant and Gene Regulation Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA
| | - Xin Ye
- Antioxidant and Gene Regulation Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA
| | - Zhanguo Gao
- Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine in Henan Province, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| | - Vincent Manganiello
- Pulmonary Cardiovascular Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Bin Wu
- Department of Gastroenterology, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jianping Ye
- Antioxidant and Gene Regulation Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA
| |
Collapse
|
63
|
Minegishi Y, Haramizu S, Misawa K, Shimotoyodome A, Hase T, Murase T. Deletion of nuclear factor-κB p50 upregulates fatty acid utilization and contributes to an anti-obesity and high-endurance phenotype in mice. Am J Physiol Endocrinol Metab 2015; 309:E523-33. [PMID: 26173458 DOI: 10.1152/ajpendo.00071.2015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 07/12/2015] [Indexed: 02/06/2023]
Abstract
The transcription factor nuclear factor-κB (NF-κB) plays an important role in regulating physiological processes such as immunity and inflammation. In addition to this primary role, NF-κB interacts physically with peroxisome proliferator-activated receptors regulating lipid metabolism-related gene expression and inhibits their transcriptional activity. Therefore, inhibition of NF-κB may promote fatty acid utilization, which could ameliorate obesity and improve endurance capacity. To test this hypothesis, we attempted to elucidate the energy metabolic status of mice lacking the p50 subunit of NF-κB (p50 KO mice) from the tissue to whole body level. p50 KO mice showed a significantly lower respiratory quotient throughout the day than did wild-type (WT) mice; this decrease was associated with increased fatty acid oxidation activity in liver and gastrocnemius muscle of p50 KO mice. p50 KO mice that were fed a high-fat diet were also resistant to fat accumulation and adipose tissue inflammation. Furthermore, p50 KO mice showed a significantly longer maximum running time compared with WT mice, with a lower respiratory exchange ratio during exercise as well as higher residual muscle glycogen content and lower blood lactate levels after exercise. These results suggest that p50 deletion facilitates fatty acid catabolism, leading to an anti-obesity and high-endurance phenotype of mice and supporting the idea that NF-κB is an important regulator of energy metabolism.
Collapse
Affiliation(s)
| | - Satoshi Haramizu
- Biological Science Laboratories, Kao Corporation, Tochigi, Japan
| | - Koichi Misawa
- Biological Science Laboratories, Kao Corporation, Tochigi, Japan
| | | | - Tadashi Hase
- Biological Science Laboratories, Kao Corporation, Tochigi, Japan
| | - Takatoshi Murase
- Biological Science Laboratories, Kao Corporation, Tochigi, Japan
| |
Collapse
|
64
|
Zhou J, Hao Z, Irwin N, Berthoud HR, Ye J. Gastric inhibitory polypeptide (GIP) is selectively decreased in the roux-limb of dietary obese mice after RYGB surgery. PLoS One 2015; 10:e0134728. [PMID: 26266950 PMCID: PMC4534413 DOI: 10.1371/journal.pone.0134728] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 07/13/2015] [Indexed: 01/27/2023] Open
Abstract
Gastric inhibitory polypeptide (GIP, glucose-dependent insulinotropic polypeptide) is expressed by intestinal K cells to regulate glucose-induced insulin secretion. The impact of Roux-en Y bypass (RYGB) surgery on blood GIP is highly contraversial. This study was conducted to address the mechanism of controversy. GIP mRNA was examined in the intestine, and serum GIP was determined using Luminex and ELISA in diet-induced obese (DIO) mice. The assays were conducted in RYGB mice in fasting and fed conditions. Food preference, weight loss and insulin sensitivity were monitored in RYGB mice. In DIO mice, GIP mRNA was increased by 80% in all sections of the small intestine over the lean control. The increase was observed in both fasting and fed conditions. After RYGB surgery, the food-induced GIP expression was selectively reduced in the Roux-limb, but not in the biliopancreatic and common limbs of intestine in fed condition. Lack of stimulation by glucose or cholesterol contributed to the reduction. Jejunal mucosa of Roux-limb exhibited hypertrophy, but villous surface was decreased by the undigested food. Serum GIP (total) was significantly higher in the fasting condition, but not in the fed condition due to attenuated GIP response to food intake in RYGB mice. The GIP alteration was associated with chow diet preference, sustained weight loss and insulin sensitization in RYGB mice. RYGB increased serum GIP in the fasting, but not in the fed conditions. The loss of food-induced GIP response in Roux-limb of intestine likely contributes to the attenuated serum GIP response to feeding.
Collapse
Affiliation(s)
- Jiaqiang Zhou
- Department of Endocrinology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Antioxidant and Gene Regulation Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, United States of America
| | - Zheng Hao
- Antioxidant and Gene Regulation Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, United States of America
| | - Nigel Irwin
- School of Biomedical Sciences, University of Ulster, Coleraine, United Kingdom
| | - Hans-Rudolf Berthoud
- Neurobiology of Nutrition Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, United States of America
| | - Jianping Ye
- Antioxidant and Gene Regulation Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, United States of America
| |
Collapse
|
65
|
Wang X, He G, Peng Y, Zhong W, Wang Y, Zhang B. Sodium butyrate alleviates adipocyte inflammation by inhibiting NLRP3 pathway. Sci Rep 2015; 5:12676. [PMID: 26234821 PMCID: PMC4522654 DOI: 10.1038/srep12676] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 07/06/2015] [Indexed: 02/06/2023] Open
Abstract
Insulin resistance (IR) is a common feature of Type II diabetes, metabolic disorders, hypertension and other vascular diseases. Recent studies showed that obesity-induced inflammation may be critical for IR. To investigate the anti-inflammatory effect of sodium butyrate (NaB) on obesity-induced inflammation, the db/db mice were intraperitoneally injected with NaB for 6 weeks. Glucose control was evaluated by glucose tolerance test (GTT) and insulin tolerance test (ITT). Adipose tissue was harvested for gene expression analysis. 3T3-L1 adipocytes were treated with Tnf-α to mimic the inflammatory state and gene expression was detected by realtime PCR and Western blotting. Our results showed that NaB treatment improved glucose control in db/db mice as determined by GTT and ITT tests. Gene expression analysis showed that NaB inhibited cytokines and immunological markers including CD68, Interferon-γ and Mcp in adipose tissues in db/db mice. Moreover, NaB inhibited cytokine releasing in 3T3-L1 adipocytes treated with TNF-α. Further analysis of inflammation pathway showed that NLRP3 was activated in db/db mice, which was efficiently inhibited by NaB treatment. Our data suggest that inhibition of obesity-induced inflammation alleviates IR, and NaB might be a potential anti-inflammatory agent for obesity.
Collapse
Affiliation(s)
- Xukai Wang
- Department of Cardiovascular Internal Medicine, Institute of Field Surgery, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Gang He
- Department of Medical Genetics, College of Basic Medicine, Third Military Medical University, Chongqing, China
| | - Yan Peng
- Department of Cardiovascular Internal Medicine, Institute of Field Surgery, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Weitian Zhong
- Department of Cardiovascular Internal Medicine, Institute of Field Surgery, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Yan Wang
- Department of Medical Genetics, College of Basic Medicine, Third Military Medical University, Chongqing, China
| | - Bo Zhang
- Department of Medical Genetics, College of Basic Medicine, Third Military Medical University, Chongqing, China
| |
Collapse
|
66
|
Bradford BJ, Yuan K, Farney JK, Mamedova LK, Carpenter AJ. Invited review: Inflammation during the transition to lactation: New adventures with an old flame. J Dairy Sci 2015. [PMID: 26210279 DOI: 10.3168/jds.2015-9683] [Citation(s) in RCA: 297] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
For dairy cattle, the first several weeks of lactation represent the highest-risk period in their lives after their own neonatal period. Although more than 50% of cows during this period are estimated to suffer from at least one subclinical disorder, the complicated admixture of normal adaptations to lactation, infectious challenges, and metabolic disorders has made it difficult to determine which physiological processes are adaptive and which are pathological during this time. Subacute inflammation, a condition that has been well documented in obesity, has been a subject of great interest among dairy cattle physiologists in the past decade. Many studies have now clearly shown that essentially all cows experience some degree of systemic inflammation in the several days after parturition. The magnitude and likely persistence of the inflammatory state varies widely among cows, and several studies have linked the degree of postpartum inflammation to increased disease risk and decreased whole-lactation milk production. In addition to these associations, enhancing postpartum inflammation with repeated subacute administration of cytokines has impaired productivity and markers of health, whereas targeted use of nonsteroidal anti-inflammatory drugs during this window of time has enhanced whole-lactation productivity in several studies. Despite these findings, many questions remain about postpartum inflammation, including which organs are key initiators of this state and what signaling molecules are responsible for systemic and tissue-specific inflammatory states. Continued in vivo work should help clarify the degree to which mild postpartum inflammation is adaptive and whether the targeted use of anti-inflammatory drugs or nutrients can improve the health and productivity of dairy cows.
Collapse
Affiliation(s)
- B J Bradford
- Department of Animal Sciences and Industry, Kansas State University, Manhattan 66506.
| | - K Yuan
- Department of Animal Sciences and Industry, Kansas State University, Manhattan 66506
| | - J K Farney
- Department of Animal Sciences and Industry, Kansas State University, Manhattan 66506
| | - L K Mamedova
- Department of Animal Sciences and Industry, Kansas State University, Manhattan 66506
| | - A J Carpenter
- Department of Animal Sciences and Industry, Kansas State University, Manhattan 66506
| |
Collapse
|
67
|
Basse AL, Dixen K, Yadav R, Tygesen MP, Qvortrup K, Kristiansen K, Quistorff B, Gupta R, Wang J, Hansen JB. Global gene expression profiling of brown to white adipose tissue transformation in sheep reveals novel transcriptional components linked to adipose remodeling. BMC Genomics 2015; 16:215. [PMID: 25887780 PMCID: PMC4407871 DOI: 10.1186/s12864-015-1405-8] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 02/26/2015] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Large mammals are capable of thermoregulation shortly after birth due to the presence of brown adipose tissue (BAT). The majority of BAT disappears after birth and is replaced by white adipose tissue (WAT). RESULTS We analyzed the postnatal transformation of adipose in sheep with a time course study of the perirenal adipose depot. We observed changes in tissue morphology, gene expression and metabolism within the first two weeks of postnatal life consistent with the expected transition from BAT to WAT. The transformation was characterized by massively decreased mitochondrial abundance and down-regulation of gene expression related to mitochondrial function and oxidative phosphorylation. Global gene expression profiling demonstrated that the time points grouped into three phases: a brown adipose phase, a transition phase and a white adipose phase. Between the brown adipose and the transition phase 170 genes were differentially expressed, and 717 genes were differentially expressed between the transition and the white adipose phase. Thirty-eight genes were shared among the two sets of differentially expressed genes. We identified a number of regulated transcription factors, including NR1H3, MYC, KLF4, ESR1, RELA and BCL6, which were linked to the overall changes in gene expression during the adipose tissue remodeling. Finally, the perirenal adipose tissue expressed both brown and brite/beige adipocyte marker genes at birth, the expression of which changed substantially over time. CONCLUSIONS Using global gene expression profiling of the postnatal BAT to WAT transformation in sheep, we provide novel insight into adipose tissue plasticity in a large mammal, including identification of novel transcriptional components linked to adipose tissue remodeling. Moreover, our data set provides a useful resource for further studies in adipose tissue plasticity.
Collapse
Affiliation(s)
- Astrid L Basse
- Department of Biomedical Sciences, University of Copenhagen, DK-2200, Copenhagen, Denmark.
- Department of Systems Biology, Center for Biological Sequence Analysis, Technical University of Denmark, DK-2800, Kongens Lyngby, Denmark.
| | - Karen Dixen
- Department of Biomedical Sciences, University of Copenhagen, DK-2200, Copenhagen, Denmark.
- Department of Systems Biology, Center for Biological Sequence Analysis, Technical University of Denmark, DK-2800, Kongens Lyngby, Denmark.
| | - Rachita Yadav
- Department of Biology, University of Copenhagen, DK-2100, Copenhagen, Denmark.
- Department of Systems Biology, Center for Biological Sequence Analysis, Technical University of Denmark, DK-2800, Kongens Lyngby, Denmark.
| | - Malin P Tygesen
- Department of Veterinary Clinical and Animal Sciences, University of Copenhagen, DK-1870, Frederiksberg, Denmark.
| | - Klaus Qvortrup
- Department of Biomedical Sciences, University of Copenhagen, DK-2200, Copenhagen, Denmark.
| | - Karsten Kristiansen
- Department of Systems Biology, Center for Biological Sequence Analysis, Technical University of Denmark, DK-2800, Kongens Lyngby, Denmark.
| | - Bjørn Quistorff
- Department of Biomedical Sciences, University of Copenhagen, DK-2200, Copenhagen, Denmark.
| | - Ramneek Gupta
- Department of Biology, University of Copenhagen, DK-2100, Copenhagen, Denmark.
| | - Jun Wang
- Department of Systems Biology, Center for Biological Sequence Analysis, Technical University of Denmark, DK-2800, Kongens Lyngby, Denmark.
- BGI-Shenzhen, Shenzhen, 518083, China.
- Princess Al Jawhara Center of Excellence in the Research of Hereditary Disorders, King Abdulaziz University, Jeddah, 21589, Saudi Arabia.
- Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau, 999078, China.
| | - Jacob B Hansen
- Department of Systems Biology, Center for Biological Sequence Analysis, Technical University of Denmark, DK-2800, Kongens Lyngby, Denmark.
| |
Collapse
|
68
|
Gao Z, Zhang J, Henagan TM, Lee JH, Ye X, Wang H, Ye J. P65 inactivation in adipocytes and macrophages attenuates adipose inflammatory response in lean but not in obese mice. Am J Physiol Endocrinol Metab 2015; 308:E496-505. [PMID: 25564477 PMCID: PMC4360014 DOI: 10.1152/ajpendo.00532.2014] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
NF-κB induces transcriptional expression of proinflammatory genes and antiapoptotic genes. The two activities of NF-κB remain to be characterized in the mechanism of chronic inflammation in obesity. To address this issue, we inactivated NF-κB in adipose tissue by knocking out p65 (RelA) in mice (F-p65-KO) and examined the inflammation in lean and obese conditions. In the lean condition, KO mice exhibited a reduced inflammation in adipose tissue with a decrease in macrophage infiltration, M1 polarization, and proinflammatory cytokine expression. In the obese condition, KO mice had elevated inflammation with more macrophage infiltration, M1 polarization, and cytokine expression. In the mechanism of enhanced inflammation, adipocytes and macrophages exhibited an increase in cellular apoptosis, which was observed with more formation of crown-like structures (CLS) in fat tissue of KO mice. Body weight, glucose metabolism, and insulin sensitivity were not significantly altered in KO mice under the lean and obese conditions. A modest but significant reduction in body fat mass was observed in KO mice on HFD with an elevation in energy expenditure. The data suggest that in the control of adipose inflammation, NF-κB exhibits different activities in the lean vs. obese condition. NF-κB is required for expression of proinflammatory genes in the lean but not in the obese condition. NF-κB is required for inhibition of apoptosis in the obese condition, in which proinflammation is enhanced by NF-κB inactivation.
Collapse
Affiliation(s)
- Zhanguo Gao
- School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan Province, China; Antioxidant and Gene Regulation Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana
| | - Jin Zhang
- Antioxidant and Gene Regulation Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana
| | - Tara M Henagan
- Antioxidant and Gene Regulation Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana; Department of Nutrition Science, Purdue University, West Lafayette, Indiana; and
| | - Jong Han Lee
- Antioxidant and Gene Regulation Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana
| | - Xin Ye
- Antioxidant and Gene Regulation Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana
| | - Hui Wang
- School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan Province, China; Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine in Henan Province, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| | - Jianping Ye
- Antioxidant and Gene Regulation Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana;
| |
Collapse
|
69
|
Tai N, Wong FS, Wen L. The role of gut microbiota in the development of type 1, type 2 diabetes mellitus and obesity. Rev Endocr Metab Disord 2015; 16:55-65. [PMID: 25619480 PMCID: PMC4348024 DOI: 10.1007/s11154-015-9309-0] [Citation(s) in RCA: 186] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Diabetes is a group of metabolic disorders characterized by persistent hyperglycemia and has become a major public health concern. Autoimmune type 1 diabetes (T1D) and insulin resistant type 2 diabetes (T2D) are the two main types. A combination of genetic and environmental factors contributes to the development of these diseases. Gut microbiota have emerged recently as an essential player in the development of T1D, T2D and obesity. Altered gut microbiota have been strongly linked to disease in both rodent models and humans. Both classic 16S rRNA sequencing and shot-gun metagenomic pyrosequencing analysis have been successfully applied to explore the gut microbiota composition and functionality. This review focuses on the association between gut microbiota and diabetes and discusses the potential mechanisms by which gut microbiota regulate disease development in T1D, T2D and obesity.
Collapse
Affiliation(s)
- Ningwen Tai
- Section of Endocrinology, Department of Internal Medicine, Yale School of Medicine, New Haven, USA
| | | | | |
Collapse
|
70
|
Abstract
Inflammation regulates energy metabolism in both physiological and pathological conditions. Pro-inflammatory cytokines involves in energy regulation in several conditions, such as obesity, aging (calorie restriction), sports (exercise), and cancer (cachexia). Here, we introduce a view of integrative physiology to understand pro-inflammatory cytokines in the control of energy expenditure. In obesity, chronic inflammation is derived from energy surplus that induces adipose tissue expansion and adipose tissue hypoxia. In addition to the detrimental effect on insulin sensitivity, pro-inflammatory cytokines also stimulate energy expenditure and facilitate adipose tissue remodeling. In caloric restriction (CR), inflammatory status is decreased by low energy intake that results in less energy supply to immune cells to favor energy saving under caloric restriction. During physical exercise, inflammatory status is elevated due to muscle production of pro-inflammatory cytokines, which promote fatty acid mobilization from adipose tissue to meet the muscle energy demand. In cancer cachexia, chronic inflammation is elevated by the immune response in the fight against cancer. The energy expenditure from chronic inflammation contributes to weight loss. Immune tolerant cancer cells gains more nutrients during the inflammation. In these conditions, inflammation coordinates energy distribution and energy demand between tissues. If the body lacks response to the pro-inflammatory cytokines (Inflammation Resistance), the energy metabolism will be impaired leading to an increased risk for obesity. In contrast, super-induction of the inflammation activity leads to weight loss and malnutrition in cancer cachexia. In summary, inflammation is a critical component in the maintenance of energy balance in the body. Literature is reviewed in above fields to support this view.
Collapse
Affiliation(s)
- Hui Wang
- Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine in Henan Province, Xinxiang Medical University, Xinxiang 453003, P. R. China
| | - Jianping Ye
- Pennington Biomedical Research Center, Louisiana State University System
- Correspondence:
| |
Collapse
|
71
|
Low level of trans-10, cis-12 conjugated linoleic acid decreases adiposity and increases browning independent of inflammatory signaling in overweight Sv129 mice. J Nutr Biochem 2015; 26:616-25. [PMID: 25801353 DOI: 10.1016/j.jnutbio.2014.12.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 11/06/2014] [Accepted: 12/19/2014] [Indexed: 01/04/2023]
Abstract
The objective of this study was to determine the extent to which a low level of trans-10, cis-12 (10,12) conjugated linoleic acid (CLA) decreases adiposity and increases browning in overweight mice, its dependence on inflammatory signaling and potential synergistic effects of daily exercise. Young, Sv129 male mice were fed a high-fat diet for 5 weeks to make them fat and glucose intolerant and then switch them to a low-fat diet with or without 0.1% 10,12 CLA, sodium salicylate or exercise for another 7 weeks. 10,12 CLA decreased white adipose tissue (WAT) and brown adipose tissue mass, and increased the messenger RNA and protein levels, and activities of enzymes associated with thermogenesis or fatty acid oxidation in WAT. Mice fed 10,12 CLA had lower body temperatures compared to controls during cold exposure, which coincided with decreased adiposity. Although sodium salicylate decreased 10,12 CLA-mediated increases in markers of inflammation in WAT, it did not affect other outcomes. Exercise had no further effect on the outcomes measured. Collectively, these data indicate that 10,12 CLA-mediated reduction of adiposity is independent of inflammatory signaling, and possibly due to up-regulation of fatty acid oxidation and heat production in order to regulate body temperature. Although this low level of 10,12 CLA reduced adiposity in overweight mice, hepatomegaly and inflammation are major health concerns.
Collapse
|
72
|
Carpenter A, Pencharz P, Mouzaki M. Accurate estimation of energy requirements of young patients. J Pediatr Gastroenterol Nutr 2015; 60:4-10. [PMID: 25238120 DOI: 10.1097/mpg.0000000000000572] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
The provision of optimal nutritional care is based on accurate estimations of patients' resting energy expenditure. The latter can be calculated with the use of predictive equations or measured with indirect calorimetry (IC). Owing to their ease of use, mathematical equations have largely replaced IC in clinical practice. This article examines the limitations and predictive inaccuracy of commonly used equations in pediatrics, which may contribute to the provision of poor nutritional care and directly affect patient outcomes. In addition, the role of IC is discussed and the physiology of nutrient metabolism, in terms of energy expenditure, is reviewed.
Collapse
Affiliation(s)
- Andrea Carpenter
- Division of Gastroenterology, Hepatology and Nutrition, Hospital for Sick Children, University of Toronto, Toronto, Canada
| | | | | |
Collapse
|
73
|
Kang S, Tsai LT, Zhou Y, Evertts A, Xu S, Griffin MJ, Issner R, Whitton HJ, Garcia BA, Epstein CB, Mikkelsen TS, Rosen ED. Identification of nuclear hormone receptor pathways causing insulin resistance by transcriptional and epigenomic analysis. Nat Cell Biol 2015; 17:44-56. [PMID: 25503565 PMCID: PMC4281178 DOI: 10.1038/ncb3080] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 11/06/2014] [Indexed: 02/06/2023]
Abstract
Insulin resistance is a cardinal feature of Type 2 diabetes (T2D) and a frequent complication of multiple clinical conditions, including obesity, ageing and steroid use, among others. How such a panoply of insults can result in a common phenotype is incompletely understood. Furthermore, very little is known about the transcriptional and epigenetic basis of this disorder, despite evidence that such pathways are likely to play a fundamental role. Here, we compare cell autonomous models of insulin resistance induced by the cytokine tumour necrosis factor-α or by the steroid dexamethasone to construct detailed transcriptional and epigenomic maps associated with cellular insulin resistance. These data predict that the glucocorticoid receptor and vitamin D receptor are common mediators of insulin resistance, which we validate using gain- and loss-of-function studies. These studies define a common transcriptional and epigenomic signature in cellular insulin resistance enabling the identification of pathogenic mechanisms.
Collapse
Affiliation(s)
- Sona Kang
- Division of Endocrinology, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, USA
| | - Linus T Tsai
- Division of Endocrinology, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, USA
| | - Yiming Zhou
- Division of Endocrinology, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, USA
| | - Adam Evertts
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA
| | - Su Xu
- Division of Endocrinology, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, USA
| | - Michael J Griffin
- Division of Endocrinology, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, USA
| | - Robbyn Issner
- Broad Institute, Cambridge, Massachusetts 02142, USA
| | | | - Benjamin A Garcia
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | | | - Evan D Rosen
- 1] Division of Endocrinology, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, USA [2] Broad Institute, Cambridge, Massachusetts 02142, USA [3] Harvard Medical School, Boston, Massachusetts 02215, USA
| |
Collapse
|
74
|
Sun LN, Yang ZY, Lv SS, Liu XC, Guan GJ, Liu G. Curcumin prevents diabetic nephropathy against inflammatory response via reversing caveolin-1 Tyr14 phosphorylation influenced TLR4 activation. Int Immunopharmacol 2014; 23:236-46. [DOI: 10.1016/j.intimp.2014.08.023] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Revised: 08/21/2014] [Accepted: 08/25/2014] [Indexed: 11/29/2022]
|
75
|
Leptin deficient ob/ob mice and diet-induced obese mice responded differently to Roux-en-Y bypass surgery. Int J Obes (Lond) 2014; 39:798-805. [PMID: 25349056 DOI: 10.1038/ijo.2014.189] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 10/08/2014] [Accepted: 10/18/2014] [Indexed: 12/25/2022]
Abstract
OBJECTIVE Weight regain contributes to the therapeutic failure in 15-20% of type 2 diabetic patients after Roux-en-Y gastric bypass surgery (RYGB), and the mechanism remains largely unknown. This study was conducted to explore the mechanism of weight regain. RESEARCH DESIGN Wild-type (WT) diet-induced obese (DIO) mice were used to mimic human obesity, and ob/ob mice were used for leptin deficiency-induced obesity. Two groups of mice were compared in weight regain for 10 months after RYGB. Weight loss, food intake, fecal energy loss and energy expenditure were monitored in the study of weight regain. Fasting insulin, insulin tolerance and homeostatic model assessment-insulin resistance were tested for insulin sensitivity under the weight regain. Weight loss from RYGB and calorie restriction was compared for the impact in insulin sensitivity. RESULTS In WT mice, RYGB induced a sustained weight loss and insulin sensitization over the sham operation in this 10-month study. However, RYGB failed to generate the same effects in leptin-deficient ob/ob mice, which suffered a weight regain over the pre-surgery level. In ob/ob mice, body weight was reduced by RYGB transiently in the first week, recovered in the second week and increased over the baseline thereafter. Weight loss was induced by RYGB relative to that of sham mice, but the loss was not sufficient to keep body weight below the pre-surgery levels. In addition, insulin sensitivity was not improved by the weight loss. The response to RYGB was improved in ob/ob mice by 2 weeks of leptin treatment. Weight loss from calorie restriction had an equivalent effect on insulin sensitization compared with that of RYGB. CONCLUSION Those data demonstrate that ob/ob mice and DIO mice responded differently to RYGB surgery, suggesting that leptin may be one of the factors required for RYGB to prevent weight regain and diabetes recurrence.
Collapse
|
76
|
Nuclear factor-κB is a common upstream signal for growth differentiation factor-5 expression in brown adipocytes exposed to pro-inflammatory cytokines and palmitate. Biochem Biophys Res Commun 2014; 452:974-9. [DOI: 10.1016/j.bbrc.2014.09.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 09/07/2014] [Indexed: 02/08/2023]
|
77
|
Bou M, Todorčević M, Rodríguez J, Capilla E, Gutiérrez J, Navarro I. Interplay of adiponectin, TNFα and insulin on gene expression, glucose uptake and PPARγ, AKT and TOR pathways in rainbow trout cultured adipocytes. Gen Comp Endocrinol 2014; 205:218-25. [PMID: 24846393 DOI: 10.1016/j.ygcen.2014.05.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 04/30/2014] [Accepted: 05/04/2014] [Indexed: 12/15/2022]
Abstract
Adipose tissue is being increasingly recognized as an important endocrine organ that produces and releases a variety of factors. In the present study we have evaluated in primary cultures of rainbow trout adipocytes, obtained from visceral adipose tissue, the interplay of the adiponectin system, TNFα and insulin at a transcriptional level and, their effects on the adipogenic transcription factor PPARγ, as well as on the activation of main insulin signaling pathways. Likewise, the implication of these adipokines in the regulation of glucose uptake in the adipocyte and their interactions with insulin or IGF-I were also evaluated. Similarly to the mammalian model, insulin enhanced adiponectin gene expression, while it exerted a negative modulation on adiponectin receptors. TNFα increased the mRNA levels of adiponectin receptor 1, but neither adiponectin nor TNFα modulated each other expression. Therefore, the reciprocal suppressive effect of both adipokines previously reported in mammals was not present in this model. Furthermore, the anti-adipogenic effect of TNFα was revealed by the down-regulation of PPARγ at a protein level, meanwhile adiponectin increased PPARγ expression in insulin-stimulated adipocytes, supporting its insulin-sensitizing role. Both adipokines stimulated glucose uptake without modifying AKT or TOR phosphorylation; however, glucose uptake in insulin-treated adipocytes was enhanced by TNFα but not by adiponectin. All in all, these results contribute to gain knowledge on the role of adipokines in rainbow trout adipose tissue and, to better understand the mechanisms that regulate glucose metabolism in this species.
Collapse
Affiliation(s)
- Marta Bou
- Department of Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona 08028, Spain
| | | | - Júlia Rodríguez
- Department of Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona 08028, Spain
| | - Encarnación Capilla
- Department of Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona 08028, Spain
| | - Joaquim Gutiérrez
- Department of Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona 08028, Spain
| | - Isabel Navarro
- Department of Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona 08028, Spain.
| |
Collapse
|
78
|
Blank T, Prinz M. NF-κB signaling regulates myelination in the CNS. Front Mol Neurosci 2014; 7:47. [PMID: 24904273 PMCID: PMC4033361 DOI: 10.3389/fnmol.2014.00047] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 05/05/2014] [Indexed: 01/12/2023] Open
Abstract
Besides myelination of neuronal axons by oligodendrocytes to facilitate propagation of action potentials, oligodendrocytes also support axon survival and function. A key transcription factor involved in these processes is nuclear factor-κB (NF-κB), a hetero or homodimer of the Rel family of proteins, including p65, c-Rel, RelB, p50, and p52. Under unstimulated, NF-κB remains inactive in the cytoplasm through interaction with NF-κB inhibitors (IκBs). Upon activation of NF-κB the cytoplasmic IκBs gets degradated, allowing the translocation of NF-κB into the nucleus where the dimer binds to the κB consensus DNA sequence and regulates gene transcription. In this review we describe how oligodendrocytes are, directly or indirectly via neighboring cells, regulated by NF-κB signaling with consequences for innate and adaptive immunity and for regulation of cell apoptosis and survival.
Collapse
Affiliation(s)
- Thomas Blank
- Institute of Neuropathology, University of Freiburg Freiburg, Germany
| | - Marco Prinz
- Institute of Neuropathology, University of Freiburg Freiburg, Germany ; BIOSS Centre for Biological Signalling Studies, University of Freiburg Freiburg, Germany
| |
Collapse
|
79
|
Wei X, Ke B, Zhao Z, Ye X, Gao Z, Ye J. Regulation of insulin degrading enzyme activity by obesity-associated factors and pioglitazone in liver of diet-induced obese mice. PLoS One 2014; 9:e95399. [PMID: 24740421 PMCID: PMC3989328 DOI: 10.1371/journal.pone.0095399] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 03/26/2014] [Indexed: 12/23/2022] Open
Abstract
Insulin degrading enzyme (IDE) is a potential drug target in the treatment of type 2 diabetes (T2D). IDE controls circulating insulin through a degradation-dependent clearance mechanism in multiple tissues. However, there is not sufficient information about IDE regulation in obesity. In this study, we test obesity-associated factors and pioglitazone in the regulation of IDE in diet-induced obese (DIO) C57BL/6 mice. The enzyme activity and protein level of IDE were increased in the liver of DIO mice. Pioglitazone (10 mg/kg/day) administration for 2 months significantly enhanced the enzyme activity (75%), protein (180%) and mRNA (100%) of IDE in DIO mice. The pioglitazone-induced changes were coupled with 50% reduction in fasting insulin and 20% reduction in fasting blood glucose. The mechanism of IDE regulation in liver was investigated in the mouse hepatoma cell line (Hepa 1c1c7 cells), in which pioglitazone (5 µM) increased IDE protein and mRNA in a time-dependent manner in an 8 h study. Free fatty acid (palmitate 300 µM) induced IDE protein, but reduced the mRNA. Glucagon induced, and TNF-α decreased IDE protein. Insulin did not exhibit any activity in the same condition. In summary, pioglitazone, FFA and glucagon directly increased, but TNF-α decreased the IDE activity in hepatocytes. The results suggest that IDE activity is regulated in liver by multiple factors in obesity and pioglitazone may induce IDE activity in the control of T2D.
Collapse
Affiliation(s)
- Xiuqing Wei
- Department of Digestive Disease, Third Affiliated Hospital, Sun Yet-Sen University, Guangzhou, China
- Antioxidant and Gene Regulation Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, United States of America
| | - Bilun Ke
- Department of Digestive Disease, Third Affiliated Hospital, Sun Yet-Sen University, Guangzhou, China
- Antioxidant and Gene Regulation Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, United States of America
| | - Zhiyun Zhao
- Antioxidant and Gene Regulation Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, United States of America
| | - Xin Ye
- Antioxidant and Gene Regulation Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, United States of America
| | - Zhanguo Gao
- Antioxidant and Gene Regulation Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, United States of America
| | - Jianping Ye
- Antioxidant and Gene Regulation Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, United States of America
- * E-mail:
| |
Collapse
|
80
|
Yin J, Lee JH, Zhang J, Gao Z, Polotsky VY, Ye J. Regulation of hepatocyte growth factor expression by NF-κB and PPARγ in adipose tissue. Am J Physiol Endocrinol Metab 2014; 306:E929-36. [PMID: 24569592 PMCID: PMC3989740 DOI: 10.1152/ajpendo.00687.2013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Hepatocyte growth factor (HGF) is expressed as an angiogenic factor in adipose tissue. However, the molecular mechanism of Hgf expression remains largely unknown in the tissue. We addressed the issue by studying Hgf expression in adipocytes and macrophages. Hgf was expressed more in the stromal-vascular fraction than the adipocyte fraction. The expression was fivefold more in macrophages than the stromal-vascular faction and was reduced by 50% after macrophage deletion in adipose tissue. The expression was reduced by differentiation in adipocytes and by tumor necrosis factor-α or lipopolysaccharide treatment in macrophages. The expression was suppressed by nuclear factor (NF)-κB in C57BL/6 mice with NF-κB p65 overexpression under the aP2 gene promoter (aP2-p65 mice) but enhanced by inactivation of NF-κB p65 in mouse embryonic fibroblasts. The Hgf gene promoter was suppressed by p65 overexpression, which blocked peroxisome proliferator-activated receptor-γ (PPARγ) interaction with RNA polymerase II. The p65 activity was abolished by knockdown of histone deacetylase 3. Hgf expression was upregulated by hypoxia in vitro and in vivo. Compared with vascular endothelial growth factor (Vegf), which was predominately expressed in mature adipocytes, Hgf was mainly expressed in nonadipocytes, suggesting that Hgf and Vegf may have different cell sources in adipose tissue. In mechanism, Hgf expression is inhibited by NF-κB through suppression of PPARγ function in the Hgf gene promoter. Both Hgf and Vegf are induced by hypoxia. The study provides a molecular mechanism for the difference of inflammation and hypoxia in the regulation of angiogenic factors.
Collapse
Affiliation(s)
- Jun Yin
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana
| | | | | | | | | | | |
Collapse
|
81
|
Makki K, Taront S, Molendi-Coste O, Bouchaert E, Neve B, Eury E, Lobbens S, Labalette M, Duez H, Staels B, Dombrowicz D, Froguel P, Wolowczuk I. Beneficial metabolic effects of rapamycin are associated with enhanced regulatory cells in diet-induced obese mice. PLoS One 2014; 9:e92684. [PMID: 24710396 PMCID: PMC3977858 DOI: 10.1371/journal.pone.0092684] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Accepted: 02/25/2014] [Indexed: 12/20/2022] Open
Abstract
The “mechanistic target of rapamycin” (mTOR) is a central controller of growth, proliferation and/or motility of various cell-types ranging from adipocytes to immune cells, thereby linking metabolism and immunity. mTOR signaling is overactivated in obesity, promoting inflammation and insulin resistance. Therefore, great interest exists in the development of mTOR inhibitors as therapeutic drugs for obesity or diabetes. However, despite a plethora of studies characterizing the metabolic consequences of mTOR inhibition in rodent models, its impact on immune changes associated with the obese condition has never been questioned so far. To address this, we used a mouse model of high-fat diet (HFD)-fed mice with and without pharmacologic mTOR inhibition by rapamycin. Rapamycin was weekly administrated to HFD-fed C57BL/6 mice for 22 weeks. Metabolic effects were determined by glucose and insulin tolerance tests and by indirect calorimetry measures of energy expenditure. Inflammatory response and immune cell populations were characterized in blood, adipose tissue and liver. In parallel, the activities of both mTOR complexes (e. g. mTORC1 and mTORC2) were determined in adipose tissue, muscle and liver. We show that rapamycin-treated mice are leaner, have enhanced energy expenditure and are protected against insulin resistance. These beneficial metabolic effects of rapamycin were associated to significant changes of the inflammatory profiles of both adipose tissue and liver. Importantly, immune cells with regulatory functions such as regulatory T-cells (Tregs) and myeloid-derived suppressor cells (MDSCs) were increased in adipose tissue. These rapamycin-triggered metabolic and immune effects resulted from mTORC1 inhibition whilst mTORC2 activity was intact. Taken together, our results reinforce the notion that controlling immune regulatory cells in metabolic tissues is crucial to maintain a proper metabolic status and, more generally, comfort the need to search for novel pharmacological inhibitors of the mTOR signaling pathway to prevent and/or treat metabolic diseases.
Collapse
Affiliation(s)
- Kassem Makki
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR)8199, Lille Pasteur Institute, Lille, France
- Lille 2 University, Lille, France
- European Genomic Institute for Diabetes (EGID), Lille, France
| | - Solenne Taront
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR)8199, Lille Pasteur Institute, Lille, France
- Lille 2 University, Lille, France
- European Genomic Institute for Diabetes (EGID), Lille, France
| | - Olivier Molendi-Coste
- Lille 2 University, Lille, France
- European Genomic Institute for Diabetes (EGID), Lille, France
- Institut National de la Santé et de la Recherche Médicale (Inserm), UMR1011, Lille Pasteur Institute, Lille, France
| | - Emmanuel Bouchaert
- Lille 2 University, Lille, France
- European Genomic Institute for Diabetes (EGID), Lille, France
- Institut National de la Santé et de la Recherche Médicale (Inserm), UMR1011, Lille Pasteur Institute, Lille, France
| | - Bernadette Neve
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR)8199, Lille Pasteur Institute, Lille, France
- Lille 2 University, Lille, France
- European Genomic Institute for Diabetes (EGID), Lille, France
| | - Elodie Eury
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR)8199, Lille Pasteur Institute, Lille, France
- Lille 2 University, Lille, France
- European Genomic Institute for Diabetes (EGID), Lille, France
| | - Stéphane Lobbens
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR)8199, Lille Pasteur Institute, Lille, France
- Lille 2 University, Lille, France
- European Genomic Institute for Diabetes (EGID), Lille, France
| | - Myriam Labalette
- Lille 2 University, Lille, France
- Immunology Institute, Centre Hospitalier Régional Universitaire (CHRU) Lille and Equipe d'Accueil (EA)2686, Lille 2 University, Lille, France
| | - Hélène Duez
- Lille 2 University, Lille, France
- European Genomic Institute for Diabetes (EGID), Lille, France
- Institut National de la Santé et de la Recherche Médicale (Inserm), UMR1011, Lille Pasteur Institute, Lille, France
| | - Bart Staels
- Lille 2 University, Lille, France
- European Genomic Institute for Diabetes (EGID), Lille, France
- Institut National de la Santé et de la Recherche Médicale (Inserm), UMR1011, Lille Pasteur Institute, Lille, France
| | - David Dombrowicz
- Lille 2 University, Lille, France
- European Genomic Institute for Diabetes (EGID), Lille, France
- Institut National de la Santé et de la Recherche Médicale (Inserm), UMR1011, Lille Pasteur Institute, Lille, France
| | - Philippe Froguel
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR)8199, Lille Pasteur Institute, Lille, France
- Lille 2 University, Lille, France
- European Genomic Institute for Diabetes (EGID), Lille, France
- Department of Genomics of Common Disease, School of Public Health, Imperial College London, London, United Kingdom
- * E-mail: (PF); (IW)
| | - Isabelle Wolowczuk
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR)8199, Lille Pasteur Institute, Lille, France
- Lille 2 University, Lille, France
- European Genomic Institute for Diabetes (EGID), Lille, France
- * E-mail: (PF); (IW)
| |
Collapse
|
82
|
Hasan AU, Ohmori K, Hashimoto T, Kamitori K, Yamaguchi F, Ishihara Y, Ishihara N, Noma T, Tokuda M, Kohno M. Valsartan ameliorates the constitutive adipokine expression pattern in mature adipocytes: a role for inverse agonism of the angiotensin II type 1 receptor in obesity. Hypertens Res 2014; 37:621-8. [PMID: 24599011 DOI: 10.1038/hr.2014.51] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Revised: 10/21/2013] [Accepted: 11/17/2013] [Indexed: 01/23/2023]
Abstract
Angiotensin (Ang) II receptor blockers (ARBs) alleviate obesity-related insulin resistance, which suggests an important role for the Ang II type 1 receptor (AT1R) in the regulation of adipocytokines. Therefore, we treated mature 3T3-L1 adipocytes with 50 μmol l(-1) of valsartan, a selective AT1R blocker without direct agonism to peroxisome proliferator-activated receptor (PPAR)-γ. In the absence of effective concentrations of Ang II, unstimulated mature adipocytes expressed and secreted high levels of interleukin (IL)-6. This constitutive proinflammatory activity was attenuated by the suppression of extracellular signal-regulated kinase phosphorylation by valsartan but was unaffected by the Ang II type 2 receptor blocker PD123319. COS7 cells co-transfected with AT1R and IL-6, which expressed NF-κB but lacked PPAR-γ, showed no constitutive but substantial ligand-dependent IL-6 reporter activity, which was counteracted by valsartan. Valsartan preserved cytosolic IκB-α and subsequently reduced nuclear NF-κB1 protein expression in mature adipocytes. Interestingly, valsartan did not increase PPAR-γ messenger RNA expression per se but enhanced the transcriptional activity of PPAR-γ in mature adipocytes; this enhancement was accompanied by upregulation of the PPAR coactivator (PGC)-1α. Moreover, T0090907, a PPAR-γ inhibitor, increased IL-6 expression, and this increase was attenuated by valsartan. Indeed, addition of valsartan without direct PPAR-γ agonism increased adiponectin production in mature adipocytes. Together, the findings indicate that valsartan blocks the constitutive AT1R activity involving the NF-κB pathway that limits PPAR-γ activity in mature adipocytes. Thus, inverse agonism of AT1R attenuates the spontaneous proinflammatory response and enhances the constitutive insulin-sensitizing activities of mature adipocytes, which may underlie the beneficial metabolic impacts of ARBs.
Collapse
Affiliation(s)
- Arif U Hasan
- Department of Cardiorenal and Cerebrovascular Medicine, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Koji Ohmori
- Department of Cardiorenal and Cerebrovascular Medicine, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Takeshi Hashimoto
- Department of Cardiovascular Physiology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Kazuyo Kamitori
- Department of Cell Physiology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Fuminori Yamaguchi
- Department of Cell Physiology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Yasuhiro Ishihara
- Department of Cardiorenal and Cerebrovascular Medicine, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Naoko Ishihara
- Department of Cardiorenal and Cerebrovascular Medicine, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Takahisa Noma
- Department of Cardiorenal and Cerebrovascular Medicine, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Masaaki Tokuda
- Department of Cell Physiology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Masakazu Kohno
- Department of Cardiorenal and Cerebrovascular Medicine, Faculty of Medicine, Kagawa University, Kagawa, Japan
| |
Collapse
|
83
|
Abstract
Obesity is now recognised as a low grade, chronic inflammatory disease that is linked to a myriad of disorders including cancer, cardiovascular disease and type 2 diabetes (T2D). With respect to T2D, work in the last decade has revealed that cells of the immune system are recruited to white adipose tissue beds (WAT), where they can secrete cytokines to modulate metabolism within WAT. As many of these cytokines are known to impair insulin action, blocking the recruitment of immune cells has been purported to have therapeutic utility for the treatment of obesity-induced T2D. As inflammation is critical for host defence, and energy consuming in nature, the blockade of inflammatory processes may, however, result in unwanted complications. In this review, we outline the immunological changes that occur within the WAT with respect to systemic glucose homeostasis. In particular, we focus on the role of major immune cell types in regulating nutrient homeostasis and potential initiating stimuli for WAT inflammation.
Collapse
Affiliation(s)
- H L Kammoun
- Cellular and Molecular Metabolism Laboratory, BakerIDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | | | | |
Collapse
|
84
|
Li Y, Wong K, Giles A, Jiang J, Lee JW, Adams AC, Kharitonenkov A, Yang Q, Gao B, Guarente L, Zang M. Hepatic SIRT1 attenuates hepatic steatosis and controls energy balance in mice by inducing fibroblast growth factor 21. Gastroenterology 2014; 146:539-49.e7. [PMID: 24184811 PMCID: PMC4228483 DOI: 10.1053/j.gastro.2013.10.059] [Citation(s) in RCA: 232] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Revised: 09/22/2013] [Accepted: 10/24/2013] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS The hepatocyte-derived hormone fibroblast growth factor 21 (FGF21) is a hormone-like regulator of metabolism. The nicotinamide adenine dinucleotide-dependent deacetylase SIRT1 regulates fatty acid metabolism through multiple nutrient sensors. Hepatic overexpression of SIRT1 reduces steatosis and glucose intolerance in obese mice. We investigated mechanisms by which SIRT1 controls hepatic steatosis in mice. METHODS Liver-specific SIRT1 knockout (SIRT1 LKO) mice and their wild-type littermates (controls) were divided into groups that were placed on a normal chow diet, fasted for 24 hours, or fasted for 24 hours and then fed for 6 hours. Liver tissues were collected and analyzed by histologic examination, gene expression profiling, and real-time polymerase chain reaction assays. Human HepG2 cells were incubated with pharmacologic activators of SIRT1 (resveratrol or SRT1720) and mitochondrion oxidation consumption rate and immunoblot analyses were performed. FGF21 was overexpressed in SIRT1 LKO mice using an adenoviral vector. Energy expenditure was assessed by indirect calorimetry. RESULTS Prolonged fasting induced lipid deposition in livers of control mice, but severe hepatic steatosis in SIRT1 LKO mice. Gene expression analysis showed that fasting up-regulated FGF21 in livers of control mice but not in SIRT1 LKO mice. Decreased hepatic and circulating levels of FGF21 in fasted SIRT1 LKO mice were associated with reduced hepatic expression of genes involved in fatty acid oxidation and ketogenesis, and increased expression of genes that control lipogenesis, compared with fasted control mice. Resveratrol or SRT1720 each increased the transcriptional activity of the FGF21 promoter (-2070/+117) and levels of FGF21 messenger RNA and protein in HepG2 cells. Surprisingly, SIRT1 LKO mice developed late-onset obesity with impaired whole-body energy expenditure. Hepatic overexpression of FGF21 in SIRT1 LKO mice increased the expression of genes that regulate fatty acid oxidation, decreased fasting-induced steatosis, reduced obesity, increased energy expenditure, and promoted browning of white adipose tissue. CONCLUSIONS SIRT1-mediated activation of FGF21 prevents liver steatosis caused by fasting. This hepatocyte-derived endocrine signaling appears to regulate expression of genes that control a brown fat-like program in white adipose tissue, energy expenditure, and adiposity. Strategies to activate SIRT1 or FGF21 could be used to treat fatty liver disease and obesity.
Collapse
Affiliation(s)
- Yu Li
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts.
| | - Kimberly Wong
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Amber Giles
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Jianwei Jiang
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Jong Woo Lee
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Andrew C Adams
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana
| | | | - Qin Yang
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Bin Gao
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Leonard Guarente
- Department of Biology, Paul F. Glenn Laboratory, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Mengwei Zang
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts.
| |
Collapse
|
85
|
Zhao R, Tang D, Yi S, Li W, Wu C, Lu Y, Hou X, Song J, Lin P, Chen L, Sun L. Elevated peripheral frequencies of Th22 cells: a novel potent participant in obesity and type 2 diabetes. PLoS One 2014; 9:e85770. [PMID: 24465695 PMCID: PMC3894984 DOI: 10.1371/journal.pone.0085770] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Accepted: 12/06/2013] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVE Chronic low-grade inflammation has long been recognized as the central link between obesity and type 2 diabetes (T2D). The novel subset of T helper (Th) cells, Th22, plays an emerging role in chronic inflammation. We investigated the potential association between Th22 and the pathogenesis of obesity and T2D. METHODS Ninety T2D inpatients (T2D group), 30 healthy participants with BMI ranged from 19 to 23.9 kg/m2 (CTL group) and 30 metabolically healthy obese controls with BMI ≥ 30 kg/m2 (MHO group) were employed in our study. Peripheral frequencies of Th22 and Th1 and Th17 cells were determined by flow cytometry based on their specific cytokine patterns. Cytokine levels in fresh plasma were quantified by ELISA. RESULTS Compared to that in CTL group (1.18±0.06%, n = 28), peripheral frequency of Th22 cells was significantly increased in MHO group (1.88±0.10%, n = 30) and in T2D group (2.247±0.10%, n = 89). There was a consistent notable increase in plasma interleukin (IL)-22 of T2D patients [47.56 (30.55-76.89) pg/mL] as compared with that of MHO group [36.65 (29.52-55.70) pg/ml; *P<0.0001] and CTLs [36.33 (31.93-40.62) pg/mL; *P<0.0001]. Furthermore, other than Th1/Th17, previously frequently described participants in obesity and T2D, there was a strong correlation between Th22 frequency and the homeostasis model of assessment for insulin resistance index (r = 0.6771, *P<0.0001) and HOMA for β-cell function (r = -0.7264, *P<0.0001). CONCLUSIONS There were increased Th22 frequencies and IL-22 levels in obesity and T2D. Elevated Th22 and IL-22 also aided in the differentiation of MHO from T2D patients. The notable correlation implied that Th22 might play a more determinant role in both insulin resistance and β-cell impairment.
Collapse
Affiliation(s)
- Ruxing Zhao
- Department of Endocrinology, Qilu Hospital, Shandong University, Jinan, China
- Institute of Endocrinology and Metabolism, Shandong University, Jinan, China
- Research Center for Cell Therapy, Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan, China
| | - Dongqi Tang
- Research Center for Cell Therapy, Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan, China
| | - Shounan Yi
- Centre for Transplant and Renal Research, Westmead Millennium Institute, The University of Sydney at Westmead Hospital, Westmead, New South Wales, Australia
| | - Wenjuan Li
- Department of Endocrinology, Qilu Hospital, Shandong University, Jinan, China
| | - Chuanlong Wu
- Department of Endocrinology, Qilu Hospital, Shandong University, Jinan, China
| | - Yiran Lu
- Department of Endocrinology, Qilu Hospital, Shandong University, Jinan, China
| | - Xinguo Hou
- Department of Endocrinology, Qilu Hospital, Shandong University, Jinan, China
| | - Jun Song
- Department of Endocrinology, Qilu Hospital, Shandong University, Jinan, China
| | - Peng Lin
- Department of Endocrinology, Qilu Hospital, Shandong University, Jinan, China
| | - Li Chen
- Department of Endocrinology, Qilu Hospital, Shandong University, Jinan, China
- Institute of Endocrinology and Metabolism, Shandong University, Jinan, China
| | - Lei Sun
- Department of Endocrinology, Qilu Hospital, Shandong University, Jinan, China
- Institute of Endocrinology and Metabolism, Shandong University, Jinan, China
| |
Collapse
|
86
|
Zhou Y, Xu F, Deng H, Bi Y, Sun W, Zhao Y, Chen Z, Weng J. PEDF expression is inhibited by insulin treatment in adipose tissue via suppressing 11β-HSD1. PLoS One 2013; 8:e84016. [PMID: 24367624 PMCID: PMC3867502 DOI: 10.1371/journal.pone.0084016] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2013] [Accepted: 11/11/2013] [Indexed: 11/23/2022] Open
Abstract
Early intensive insulin therapy improves insulin sensitivity in type 2 diabetic patients; while the underlying mechanism remains largely unknown. Pigment epithelium-derived factor (PEDF), an anti-angiogenic factor, is believed to be involved in the pathogenesis of insulin resistance. Here, we hypothesize that PEDF might be down regulated by insulin and then lead to the improved insulin resistance in type 2 diabetic patients during insulin therapy. We addressed this issue by investigating insulin regulation of PEDF expression in diabetic conditions. The results showed that serum PEDF was reduced by 15% in newly diagnosed type 2 diabetic patients after insulin therapy. In adipose tissue of diabetic Sprague-Dawley rats, PEDF expression was associated with TNF-α elevation and it could be decreased both in serum and in adipose tissue by insulin treatment. In adipocytes, PEDF was induced by TNF-α through activation of NF-κB. The response was inhibited by knockdown and enhanced by over expression of NF-κB p65. However, PEDF expression was indirectly, not directly, induced by NF-κB which promoted 11β-hydroxysteroid dehydrogenase 1 (11β-HSD1) expression in adipocytes. 11β-HSD1 is likely to stimulate PEDF expression through production of active form of glucocorticoids as dexamethasone induced PEDF expression in adipose tissue. Insulin inhibited PEDF by down-regulating 11β-HSD1 expression. The results suggest that PEDF activity is induced by inflammation and decreased by insulin through targeting 11β-HSD1/glucocorticoid pathway in adipose tissue of diabetic patients.
Collapse
Affiliation(s)
- Yinli Zhou
- Department of Endocrinology and Metabolism, the Third Affiliated Hospital of Sun Yat-Sen University; Key Laboratory of Diabetology of Guangdong Province, Guangzhou, China
| | - Fen Xu
- Department of Endocrinology and Metabolism, the Third Affiliated Hospital of Sun Yat-Sen University; Key Laboratory of Diabetology of Guangdong Province, Guangzhou, China
| | - Hongrong Deng
- Department of Endocrinology and Metabolism, the Third Affiliated Hospital of Sun Yat-Sen University; Key Laboratory of Diabetology of Guangdong Province, Guangzhou, China
| | - Yan Bi
- Department of Endocrinology and Metabolism, the Third Affiliated Hospital of Sun Yat-Sen University; Key Laboratory of Diabetology of Guangdong Province, Guangzhou, China
| | - Weiping Sun
- Department of Endocrinology and Metabolism, the Third Affiliated Hospital of Sun Yat-Sen University; Key Laboratory of Diabetology of Guangdong Province, Guangzhou, China
| | - Yi Zhao
- Department of Endocrinology and Metabolism, the Third Affiliated Hospital of Sun Yat-Sen University; Key Laboratory of Diabetology of Guangdong Province, Guangzhou, China
| | - Zonglan Chen
- Department of Endocrinology and Metabolism, the Third Affiliated Hospital of Sun Yat-Sen University; Key Laboratory of Diabetology of Guangdong Province, Guangzhou, China
| | - Jianping Weng
- Department of Endocrinology and Metabolism, the Third Affiliated Hospital of Sun Yat-Sen University; Key Laboratory of Diabetology of Guangdong Province, Guangzhou, China
- * E-mail:
| |
Collapse
|
87
|
Ye X, Jiang X, Guo W, Clark K, Gao Z. Overexpression of NF-κB p65 in macrophages ameliorates atherosclerosis in apoE-knockout mice. Am J Physiol Endocrinol Metab 2013; 305:E1375-83. [PMID: 24105415 PMCID: PMC3882374 DOI: 10.1152/ajpendo.00307.2013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The transcription factor NF-κB p65 is a key regulator in the regulation of an inflammatory response and in the pathology of atherosclerosis. However, there is no direct evidence for the role of NF-κB in macrophages in the development of atherosclerosis. We investigated whether macrophage overexpression of p65 in apoE-knockout mice could improve atherosclerosis. Transgenic (Tg) mice overexpressing p65 in macrophages were generated by crossing fatty acid-binding protein 4 (aP2) promoter-controlled p65 mice with apoE-knockout (KO) mice. Tg mice exhibited functional activation of NF-κB signaling in macrophages and fat tissues. We observed that the atherosclerotic lesion was 40% less in the Tg mice compared with the apoE-KO controls fed a standard atherogenic diet for 16 wk (n = 12). The Tg mice were leaner from reduced fat mass by increased energy expenditure. Moreover, the overexpression of p65 in macrophages suppressed foam cell formation. Our results show that there is 1) an increased fatty acid oxidation in macrophages, 2) a reduced scavenger receptor CD36 expression and lipid accumulation in microphages, 3) reduced-inflammation cytokines in serum, and 4) enhanced energy expenditure in Tg mice. Our data suggest that activation of NF-κB in macrophages has atheroprotective effects in mice by enhancing lipid metabolism and energy expenditure.
Collapse
Affiliation(s)
- Xin Ye
- Pennington Biomedical Research Center, Louisiana State University Systems, Baton Rouge, Louisiana; and
| | | | | | | | | |
Collapse
|
88
|
Wang Y, Wang H, Hegde V, Dubuisson O, Gao Z, Dhurandhar NV, Ye J. Interplay of pro- and anti-inflammatory cytokines to determine lipid accretion in adipocytes. Int J Obes (Lond) 2013; 37:1490-8. [PMID: 23381555 PMCID: PMC3657600 DOI: 10.1038/ijo.2013.9] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2012] [Revised: 12/20/2012] [Accepted: 01/04/2013] [Indexed: 01/04/2023]
Abstract
OBJECTIVE Obesity is associated with an increase in various pro-inflammatory and anti-inflammatory cytokines, but the interplay of these cytokines is incompletely understood. We conducted experiments to test a broader hypothesis that a dynamic interplay of pro-inflammatory and anti-inflammatory cytokines controls lipid storage in adipocytes. DESIGN Three experiments were designed to test the overall hypothesis that proinflammatory cytokine (for example, tumor necrosis factor-α (TNF-α) inhibits anti-inflammatory cytokine (for example, adiponectin) activity in an attempt to limit excess lipid accumulation in adipocytes. RESULTS Experiment one showed that in pro-inflammatory animal models (ap2-P65, ob/ob and high-fat diet-induced obese mice), the increase in TNF-α expression was associated with a decrease in adiponectin expression. Experiment two showed that in 3T3-L1 adipocytes, TNF-α significantly reduced lipid accumulation and glucose uptake induced by adiponectin, and increased lipolysis. Experiment three showed that in 3T3-L1 adipocytes, TNF-α reduced mRNA and protein expression of adiponectin. Adiponectin gene transcription and mRNA stability were both reduced by TNF-α. The expression of peroxisome proliferator-activated receptor gamma, an activator of adiponectin gene promoter, was reduced by TNF-α. The inhibitory activity of TNF-α was blocked by the chemical inhibitors of NF-κB and super suppressor IκBα. CONCLUSIONS TNF-α opposes the action of adiponectin in the regulation of lipid metabolism, and inhibits adiponectin expression at transcriptional and post-transcriptional levels. The results suggest that pro-inflammatory cytokine inhibit anti-inflammatory cytokine in adipocytes to reduce lipid storage. This suggests a potential role of anti-inflammatory cytokines in the control of adipose tissue expansion.
Collapse
Affiliation(s)
- Yanning Wang
- Antioxidant and Gene Regulation Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808
| | - Hui Wang
- Research Center for Immunology, Xinxiang Medical University, Xinxiang 453003, Henan Province, P.R. China
| | - Vijay Hegde
- Infection and Obesity Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808
| | - Olga Dubuisson
- Infection and Obesity Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808
| | - Zhanguo Gao
- Antioxidant and Gene Regulation Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808
| | - Nikhil V. Dhurandhar
- Infection and Obesity Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808
| | - Jianping Ye
- Antioxidant and Gene Regulation Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808
| |
Collapse
|
89
|
|
90
|
Shen Q, Chitchumroonchokchai C, Thomas JL, Gushchina LV, Disilvestro D, Failla ML, Ziouzenkova O. Adipocyte reporter assays: application for identification of anti-inflammatory and antioxidant properties of mangosteen xanthones. Mol Nutr Food Res 2013; 58:239-47. [PMID: 24039005 DOI: 10.1002/mnfr.201300181] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 06/14/2013] [Accepted: 06/26/2013] [Indexed: 12/22/2022]
Abstract
SCOPE Three fluorescence biosensors were developed based on a 3T3-L1 preadipocyte line that stably expressed Nfkb-RE/GFP, Fabp4-P/CFP, and Nrf2-P/YFP fluorescent reporters. We hypothesized that nutraceuticals' inflammatory, adipogenic, and antioxidant status will be identified based on the change in fluorescence in reporter adipocytes. We validated these assays with activators of NFκB, FABP4-regulating peroxisome proliferator activated receptor gamma, NFR2 and, thereafter, tested known and unknown properties of mangostines (MGs), the xanthone metabolites in mangosteen fruit. METHODS AND RESULTS We validated inflammatory and adipogenic properties of α-MG using an Nfkb-RE/GFP biosensor assay. Next, we identified unique properties of γ-MG, a minor MG xanthone. γ-MG suppressed adipogenesis and expression of adiponectin, but inhibited the Nfkb-RE/GFP reporter and secretion of inflammatory monocyte chemotactic protein 1 as compared to the control adipocytes. We found that the inhibition of adipogenesis and Nfkb-mediated inflammation depends on a dose-dependent reduction of Nrf2 promoter activity by α-MG. The Nrf2 inhibition resulted in the reduced Pparg expression. α-MG did not directly influence Pparg activity in Fabp4-P/CFP adipocytes. CONCLUSION α-MG-mediated antioxidant response via Nrf2 is a mechanism preventing adipogenesis and inflammation in adipocytes. Combined application of high-throughput biosensors could provide an effective platform for the identification of nutraceuticals and the mechanism of their actions in adipocytes and, potentially, in obese patients.
Collapse
Affiliation(s)
- Qiwen Shen
- Department of Human Sciences, The Ohio State University, Columbus, OH, USA
| | | | | | | | | | | | | |
Collapse
|
91
|
Thaler JP, Guyenet SJ, Dorfman MD, Wisse BE, Schwartz MW. Hypothalamic inflammation: marker or mechanism of obesity pathogenesis? Diabetes 2013; 62:2629-34. [PMID: 23881189 PMCID: PMC3717869 DOI: 10.2337/db12-1605] [Citation(s) in RCA: 148] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Accepted: 04/03/2013] [Indexed: 12/22/2022]
Affiliation(s)
- Joshua P. Thaler
- Diabetes and Obesity Center of Excellence and Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, Washington
| | - Stephan J. Guyenet
- Diabetes and Obesity Center of Excellence and Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, Washington
| | - Mauricio D. Dorfman
- Diabetes and Obesity Center of Excellence and Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, Washington
| | - Brent E. Wisse
- Diabetes and Obesity Center of Excellence and Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, Washington
| | - Michael W. Schwartz
- Diabetes and Obesity Center of Excellence and Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, Washington
| |
Collapse
|
92
|
Li Y, Nie Y, Helou Y, Ding G, Feng B, Xu G, Salomon A, Xu H. Identification of sucrose non-fermenting-related kinase (SNRK) as a suppressor of adipocyte inflammation. Diabetes 2013; 62:2396-409. [PMID: 23520131 PMCID: PMC3712026 DOI: 10.2337/db12-1081] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Accepted: 03/13/2013] [Indexed: 01/10/2023]
Abstract
In this study, the role of sucrose non-fermenting-related kinase (SNRK) in white adipocyte biology was investigated. SNRK is abundantly expressed in adipose tissue, and the expression level is decreased in obese mice. SNRK expression is repressed by inflammatory signals but increased by insulin sensitizer in cultured adipocytes. In vivo, adipose tissue SNRK expression can be decreased by lipid injection but enhanced by macrophage ablation. Knocking down SNRK in cultured adipocytes activates both JNK and IKKβ pathways as well as promotes lipolysis. Insulin-stimulated Akt phosphorylation and glucose uptake are impaired in SNRK knockdown adipocytes. Phosphoproteomic analysis with SNRK knockdown adipocytes revealed significantly decreased phosphorylation of 49 proteins by 25% or more, which are involved in various aspects of adipocyte function with a clear indication of attenuated mTORC1 signaling. Phosphorylation of 43 proteins is significantly increased by onefold or higher, among which several proteins are known to be involved in inflammatory pathways. The inflammatory responses in SNRK knockdown adipocytes can be partially attributable to defective mTORC1 signaling, since rapamycin treatment activates IKKβ and induces lipolysis in adipocytes. In summary, SNRK may act as a suppressor of adipocyte inflammation and its presence is necessary for maintaining normal adipocyte function.
Collapse
Affiliation(s)
- Yujie Li
- Hallett Center for Diabetes and Endocrinology, Rhode Island Hospital, Warren Alpert
- Department of Geriatric Endocrinology, Jiangsu Province Hospital, Nanjing Medical University, Nanjing, China
| | - Yaohui Nie
- Hallett Center for Diabetes and Endocrinology, Rhode Island Hospital, Warren Alpert
- Department of Medicine and Therapeutics, The Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Ynes Helou
- Department of Molecular Pharmacology and Physiology, Brown University, Providence, Rhode Island
| | - Guoxian Ding
- Department of Geriatric Endocrinology, Jiangsu Province Hospital, Nanjing Medical University, Nanjing, China
| | - Bin Feng
- Hallett Center for Diabetes and Endocrinology, Rhode Island Hospital, Warren Alpert
| | - Gang Xu
- Department of Medicine and Therapeutics, The Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Arthur Salomon
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island
- Department of Chemistry, Brown University, Providence, Rhode Island
| | - Haiyan Xu
- Hallett Center for Diabetes and Endocrinology, Rhode Island Hospital, Warren Alpert
- Pathobiology Program, Brown University, Providence, Rhode Island
| |
Collapse
|
93
|
Tourniaire F, Romier-Crouzet B, Lee JH, Marcotorchino J, Gouranton E, Salles J, Malezet C, Astier J, Darmon P, Blouin E, Walrand S, Ye J, Landrier JF. Chemokine Expression in Inflamed Adipose Tissue Is Mainly Mediated by NF-κB. PLoS One 2013; 8:e66515. [PMID: 23824685 PMCID: PMC3688928 DOI: 10.1371/journal.pone.0066515] [Citation(s) in RCA: 102] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Accepted: 05/10/2013] [Indexed: 01/08/2023] Open
Abstract
Immune cell infiltration of expanding adipose tissue during obesity and its role in insulin resistance has been described and involves chemokines. However, studies so far have focused on a single chemokine or its receptor (especially CCL2 and CCL5) whereas redundant functions of chemokines have been described. The objective of this work was to explore the expression of chemokines in inflamed adipose tissue in obesity. Human and mouse adipocytes were analyzed for expression of chemokines in response to inflammatory signal (TNF-α) using microarrays and gene set enrichment analysis. Gene expression was verified by qRT-PCR. Chemokine protein was determined in culture medium with ELISA. Chemokine expression was investigated in human subcutaneous adipose tissue biopsies and mechanism of chemokine expression was investigated using chemical inhibitors and cellular and animal transgenic models. Chemokine encoding genes were the most responsive genes in TNF-α treated human and mouse adipocytes. mRNA and protein of 34 chemokine genes were induced in a dose-dependent manner in the culture system. Furthermore, expression of those chemokines was elevated in human obese adipose tissue. Finally, chemokine expression was reduced by NF-κB inactivation and elevated by NF-κB activation. Our data indicate that besides CCL2 and CCL5, numerous other chemokines such as CCL19 are expressed by adipocytes under obesity-associated chronic inflammation. Their expression is regulated predominantly by NF-κB. Those chemokines could be involved in the initiation of infiltration of leukocytes into obese adipose tissue.
Collapse
Affiliation(s)
- Franck Tourniaire
- INRA, UMR 1260, Marseille, France
- INSERM, UMR 1062, « Nutrition, Obésité et Risque Thrombotique », Marseille, France
- Université d’Aix-Marseille, Faculté de Médecine, Marseille, France
| | - Beatrice Romier-Crouzet
- INRA, UMR 1260, Marseille, France
- INSERM, UMR 1062, « Nutrition, Obésité et Risque Thrombotique », Marseille, France
- Université d’Aix-Marseille, Faculté de Médecine, Marseille, France
| | - Jong Han Lee
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, United States of America
| | - Julie Marcotorchino
- INRA, UMR 1260, Marseille, France
- INSERM, UMR 1062, « Nutrition, Obésité et Risque Thrombotique », Marseille, France
- Université d’Aix-Marseille, Faculté de Médecine, Marseille, France
| | - Erwan Gouranton
- INRA, UMR 1260, Marseille, France
- INSERM, UMR 1062, « Nutrition, Obésité et Risque Thrombotique », Marseille, France
- Université d’Aix-Marseille, Faculté de Médecine, Marseille, France
| | - Jerome Salles
- UMR INRA 1019 Unité de nutrition humaine, Centre de Recherches INRA de Clermont-Ferrand/Theix, St Genès Champanelle, France
| | - Christiane Malezet
- INRA, UMR 1260, Marseille, France
- INSERM, UMR 1062, « Nutrition, Obésité et Risque Thrombotique », Marseille, France
- Université d’Aix-Marseille, Faculté de Médecine, Marseille, France
| | - Julien Astier
- INRA, UMR 1260, Marseille, France
- INSERM, UMR 1062, « Nutrition, Obésité et Risque Thrombotique », Marseille, France
- Université d’Aix-Marseille, Faculté de Médecine, Marseille, France
| | - Patrice Darmon
- INRA, UMR 1260, Marseille, France
- INSERM, UMR 1062, « Nutrition, Obésité et Risque Thrombotique », Marseille, France
- Université d’Aix-Marseille, Faculté de Médecine, Marseille, France
| | | | - Stephane Walrand
- UMR INRA 1019 Unité de nutrition humaine, Centre de Recherches INRA de Clermont-Ferrand/Theix, St Genès Champanelle, France
| | - Jianping Ye
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, United States of America
| | - Jean-Francois Landrier
- INRA, UMR 1260, Marseille, France
- INSERM, UMR 1062, « Nutrition, Obésité et Risque Thrombotique », Marseille, France
- Université d’Aix-Marseille, Faculté de Médecine, Marseille, France
- * E-mail:
| |
Collapse
|
94
|
Cildir G, Akıncılar SC, Tergaonkar V. Chronic adipose tissue inflammation: all immune cells on the stage. Trends Mol Med 2013; 19:487-500. [PMID: 23746697 DOI: 10.1016/j.molmed.2013.05.001] [Citation(s) in RCA: 216] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Revised: 05/03/2013] [Accepted: 05/06/2013] [Indexed: 12/12/2022]
Abstract
Inflammation is indispensable for host homeostasis against invading pathogens and efficient wound healing upon tissue malfunction and has to be tightly controlled by various mechanisms to limit excess responses harmful to host tissues. A myriad of disease conditions ranging from type 2 diabetes (T2D) to neurodegenerative and cardiovascular disorders are now shown to progress due to persistent, unresolved inflammation in metabolic tissues such as adipose, liver, pancreas, muscle, and brain. However, their underlying mechanisms are incompletely understood. The actions of innate and adaptive immune cells in these ailments are increasingly appreciated so much so that a new research area called 'immunometabolism' has emerged. In this review, we will highlight the fundamental roles of various immune cells in adipose tissue during the initiation and progression of obesity-induced inflammation and discuss potential anti-inflammatory therapies from different mechanistic points of view.
Collapse
Affiliation(s)
- Gökhan Cildir
- Laboratory of NF-κB Signaling, Institute of Molecular and Cell Biology (IMCB), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | | | | |
Collapse
|
95
|
Bjursell M, Wedin M, Admyre T, Hermansson M, Böttcher G, Göransson M, Lindén D, Bamberg K, Oscarsson J, Bohlooly-Y M. Ageing Fxr deficient mice develop increased energy expenditure, improved glucose control and liver damage resembling NASH. PLoS One 2013; 8:e64721. [PMID: 23700488 PMCID: PMC3659114 DOI: 10.1371/journal.pone.0064721] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 04/17/2013] [Indexed: 12/17/2022] Open
Abstract
Nuclear receptor subfamily 1, group H, member 4 (Nr1h4, FXR) is a bile acid activated nuclear receptor mainly expressed in the liver, intestine, kidney and adrenal glands. Upon activation, the primary function is to suppress cholesterol 7 alpha-hydroxylase (Cyp7a1), the rate-limiting enzyme in the classic or neutral bile acid synthesis pathway. In the present study, a novel Fxr deficient mouse line was created and studied with respect to metabolism and liver function in ageing mice fed chow diet. The Fxr deficient mice were similar to wild type mice in terms of body weight, body composition, energy intake and expenditure as well as behaviours at a young age. However, from 15 weeks of age and onwards, the Fxr deficient mice had almost no body weight increase up to 39 weeks of age mainly because of lower body fat mass. The lower body weight gain was associated with increased energy expenditure that was not compensated by increased food intake. Fasting levels of glucose and insulin were lower and glucose tolerance was improved in old and lean Fxr deficient mice. However, the Fxr deficient mice displayed significantly increased liver weight, steatosis, hepatocyte ballooning degeneration and lobular inflammation together with elevated plasma levels of ALT, bilirubin and bile acids, findings compatible with non-alcoholic steatohepatitis (NASH) and cholestasis. In conclusion, ageing Fxr deficient mice display late onset leanness associated with elevated energy expenditure and improved glucose control but develop severe NASH-like liver pathology.
Collapse
|
96
|
Lee JH, Gao Z, Ye J. Regulation of 11β-HSD1 expression during adipose tissue expansion by hypoxia through different activities of NF-κB and HIF-1α. Am J Physiol Endocrinol Metab 2013; 304:E1035-41. [PMID: 23512810 PMCID: PMC3651619 DOI: 10.1152/ajpendo.00029.2013] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
11β-Hydroxysteroid dehydrogenase type 1 (11β-HSD1) is involved in the pathogenesis of type 2 diabetes by generating active glucocorticoids (cortisol and corticosterone) that are strong inhibitors of angiogenesis. However, the mechanism of 11β-HSD1 gene expression and its relationship to adipose angiogenesis are largely unknown. To address this issue, we examined 11β-HSD1 expression in visceral and subcutaneous adipose tissue (AT) of diet-induced obese (DIO) mice during weight gain and investigated the gene regulation by hypoxia in vitro. 11β-HSD1 mRNA was reduced in the adipose tissues during weight gain in DIO mice, and the reduction was associated with an elevated expression of angiogenic factors. In vitro, 11β-HSD1 expression was induced in mRNA and protein by hypoxia. Of the two transcription factors activated by hypoxia, the nuclear factor-κB (NF-κB) enhanced but the hypoxia inducible factor-1α (HIF-1α) reduced 11β-HSD1 expression. 11β-HSD1 expression was elevated by NF-κB in epididymal fat of aP2-p65 mice. The hypoxia-induced 11β-HSD1 expression was attenuated by NF-κB inactivation in p65-deficient cells but enhanced by HIF-1 inactivation in HIF-1α-null cells. These data suggest that 11β-HSD1 expression is upregulated by NF-κB and downregulated by HIF-1α. During AT expansion in DIO mice, the reduction of 11β-HSD1 expression may reflect a dominant HIF-1α activity in the adipose tissue. This study suggests that NF-κB may mediate the inflammatory cytokine signal to upregulate 11β-HSD1 expression.
Collapse
MESH Headings
- 11-beta-Hydroxysteroid Dehydrogenase Type 1/biosynthesis
- 11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics
- 11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism
- 3T3-L1 Cells
- Animals
- Blotting, Western
- Cell Hypoxia/physiology
- Diabetes Mellitus, Type 2/enzymology
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/metabolism
- Gene Expression Regulation, Enzymologic
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Intra-Abdominal Fat/enzymology
- Intra-Abdominal Fat/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- NF-kappa B/metabolism
- Obesity/enzymology
- Obesity/genetics
- Obesity/metabolism
- RNA, Messenger/chemistry
- RNA, Messenger/genetics
- Real-Time Polymerase Chain Reaction
- Subcutaneous Fat/enzymology
- Subcutaneous Fat/metabolism
- Weight Gain/physiology
Collapse
Affiliation(s)
- Jong Han Lee
- Pennington Biomedical Research Center, Louisiana State University System, 6400 Perkins Road, Baton Rouge, LA 70808, USA
| | | | | |
Collapse
|
97
|
Kim JE, Lee MH, Nam DH, Song HK, Kang YS, Lee JE, Kim HW, Cha JJ, Hyun YY, Han SY, Han KH, Han JY, Cha DR. Celastrol, an NF-κB inhibitor, improves insulin resistance and attenuates renal injury in db/db mice. PLoS One 2013; 8:e62068. [PMID: 23637966 PMCID: PMC3637455 DOI: 10.1371/journal.pone.0062068] [Citation(s) in RCA: 126] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Accepted: 03/18/2013] [Indexed: 12/12/2022] Open
Abstract
The NF-κB pathway plays an important role in chronic inflammatory and autoimmune diseases. Recently, NF-κB has also been suggested as an important mechanism linking obesity, inflammation, and metabolic disorders. However, there is no current evidence regarding the mechanism of action of NF-κB inhibition in insulin resistance and diabetic nephropathy in type 2 diabetic animal models. We investigated the effects of the NF-κB inhibitor celastrol in db/db mice. The treatment with celastrol for 2 months significantly lowered fasting plasma glucose (FPG), HbA1C and homeostasis model assessment index (HOMA-IR) levels. Celastrol also exhibited significant decreases in body weight, kidney/body weight and adiposity. Celastrol reduced insulin resistance and lipid abnormalities and led to higher plasma adiponectin levels. Celastrol treatment also significantly mitigated lipid accumulation and oxidative stress in organs including the kidney, liver and adipose tissue. The treated group also exhibited significantly lower creatinine levels and urinary albumin excretion was markedly reduced. Celastrol treatment significantly lowered mesangial expansion and suppressed type IV collagen, PAI-1 and TGFβ1 expressions in renal tissues. Celastrol also improved abnormal lipid metabolism, oxidative stress and proinflammatory cytokine activity in the kidney. In cultured podocytes, celastrol treatment abolished saturated fatty acid-induced proinflammatory cytokine synthesis. Taken together, celastrol treatment not only improved insulin resistance, glycemic control and oxidative stress, but also improved renal functional and structural changes through both metabolic and anti-inflammatory effects in the kidney. These results suggest that targeted therapy for NF-κB may be a useful new therapeutic approach for the management of type II diabetes and diabetic nephropathy.
Collapse
Affiliation(s)
- Jung Eun Kim
- Department of Internal Medicine, Division of Nephrology, Korea University, Ansan City, Kyungki-Do, Korea
| | - Mi Hwa Lee
- Department of Internal Medicine, Division of Nephrology, Korea University, Ansan City, Kyungki-Do, Korea
| | - Deok Hwa Nam
- Department of Internal Medicine, Division of Nephrology, Korea University, Ansan City, Kyungki-Do, Korea
| | - Hye Kyoung Song
- Department of Internal Medicine, Division of Nephrology, Korea University, Ansan City, Kyungki-Do, Korea
| | - Young Sun Kang
- Department of Internal Medicine, Division of Nephrology, Korea University, Ansan City, Kyungki-Do, Korea
| | - Ji Eun Lee
- Department of Internal Medicine, Division of Nephrology, Wonkwang University, Gunpo City, Kyungki-Do, Korea
| | - Hyun Wook Kim
- Department of Internal Medicine, Division of Nephrology, Wonkwang University, Gunpo City, Kyungki-Do, Korea
| | - Jin Joo Cha
- Department of Internal Medicine, Division of Nephrology, Korea University, Ansan City, Kyungki-Do, Korea
| | - Young Youl Hyun
- Department of Internal Medicine, Division of Nephrology, Sungkyunkwan University, Seoul, Korea
| | - Sang Youb Han
- Department of Internal Medicine, Division of Nephrology, Inje University, Goyang City, Kyungki-Do, Korea
| | - Kum Hyun Han
- Department of Internal Medicine, Division of Nephrology, Inje University, Goyang City, Kyungki-Do, Korea
| | - Jee Young Han
- Department of Pathology, Inha University, Incheon City, Kyungki-Do, Korea
| | - Dae Ryong Cha
- Department of Internal Medicine, Division of Nephrology, Korea University, Ansan City, Kyungki-Do, Korea
| |
Collapse
|
98
|
Walters RG, Coin LJM, Ruokonen A, de Smith AJ, El-Sayed Moustafa JS, Jacquemont S, Elliott P, Esko T, Hartikainen AL, Laitinen J, Männik K, Martinet D, Meyre D, Nauck M, Schurmann C, Sladek R, Thorleifsson G, Thorsteinsdóttir U, Valsesia A, Waeber G, Zufferey F, Balkau B, Pattou F, Metspalu A, Völzke H, Vollenweider P, Stefansson K, Järvelin MR, Beckmann JS, Froguel P, Blakemore AIF. Rare genomic structural variants in complex disease: lessons from the replication of associations with obesity. PLoS One 2013; 8:e58048. [PMID: 23554873 PMCID: PMC3595275 DOI: 10.1371/journal.pone.0058048] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Accepted: 01/30/2013] [Indexed: 01/19/2023] Open
Abstract
The limited ability of common variants to account for the genetic contribution to complex disease has prompted searches for rare variants of large effect, to partly explain the 'missing heritability'. Analyses of genome-wide genotyping data have identified genomic structural variants (GSVs) as a source of such rare causal variants. Recent studies have reported multiple GSV loci associated with risk of obesity. We attempted to replicate these associations by similar analysis of two familial-obesity case-control cohorts and a population cohort, and detected GSVs at 11 out of 18 loci, at frequencies similar to those previously reported. Based on their reported frequencies and effect sizes (OR≥25), we had sufficient statistical power to detect the large majority (80%) of genuine associations at these loci. However, only one obesity association was replicated. Deletion of a 220 kb region on chromosome 16p11.2 has a carrier population frequency of 2×10(-4) (95% confidence interval [9.6×10(-5)-3.1×10(-4)]); accounts overall for 0.5% [0.19%-0.82%] of severe childhood obesity cases (P = 3.8×10(-10); odds ratio = 25.0 [9.9-60.6]); and results in a mean body mass index (BMI) increase of 5.8 kg.m(-2) [1.8-10.3] in adults from the general population. We also attempted replication using BMI as a quantitative trait in our population cohort; associations with BMI at or near nominal significance were detected at two further loci near KIF2B and within FOXP2, but these did not survive correction for multiple testing. These findings emphasise several issues of importance when conducting rare GSV association, including the need for careful cohort selection and replication strategy, accurate GSV identification, and appropriate correction for multiple testing and/or control of false discovery rate. Moreover, they highlight the potential difficulty in replicating rare CNV associations across different populations. Nevertheless, we show that such studies are potentially valuable for the identification of variants making an appreciable contribution to complex disease.
Collapse
Affiliation(s)
- Robin G. Walters
- Department of Genomics of Common Disease, Imperial College London, London, United Kingdom
- Clinical Trial Service Unit and Epidemiological Studies Unit, University of Oxford, Oxford, United Kingdom
| | - Lachlan J. M. Coin
- Department of Genomics of Common Disease, Imperial College London, London, United Kingdom
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - Aimo Ruokonen
- Institute of Diagnostics, Clinical Chemistry, University of Oulu, Oulu, Finland
- Oulu University Hospital, Oulu, Finland
| | - Adam J. de Smith
- Department of Genomics of Common Disease, Imperial College London, London, United Kingdom
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California, United States of America
| | | | - Sebastien Jacquemont
- Service of Medical Genetics, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Paul Elliott
- Department of Epidemiology and Biostatistics, Imperial College London, London, United Kingdom
- MRC Health Protection Agency (HPA) Centre for Environment and Health, Imperial College London, London, United Kingdom
| | - Tõnu Esko
- Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Anna-Liisa Hartikainen
- Institute of Clinical Sciences/Obstetrics and Gynecology, University of Oulu, Oulu, Finland
| | | | - Katrin Männik
- Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
- The Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Danielle Martinet
- Service of Medical Genetics, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - David Meyre
- CNRS 8199-Institute of Biology, Pasteur Institute, Lille, France
- Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, Ontario, Canada
| | - Matthias Nauck
- Institute of Clinical Chemistry and Laboratory Medicine, Ernst-Moritz-Arndt-University, Greifswald, Germany
| | - Claudia Schurmann
- Interfaculty Institute for Genetics and Functional Genomics, Ernst-Moritz-Arndt-University, Greifswald, Germany
| | - Rob Sladek
- McGill University and Genome Quebec Innovation Centre, Montreal, Canada
- Department of Medicine and Human Genetics, McGill University, Montreal, Canada
| | | | - Unnur Thorsteinsdóttir
- deCODE Genetics, Reykjavík, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Armand Valsesia
- Department of Medical Genetics, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Gerard Waeber
- Department of Internal Medicine, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Flore Zufferey
- Service of Medical Genetics, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Beverley Balkau
- INSERM, CESP Centre for Research in Epidemiology and Population Health, U1018, Villejuif, France
- University Paris Sud 11, UMRS 1018, Villejuif, France
| | - François Pattou
- INSERM U859, Lille, France
- Université Lille Nord de France, Centre Hospitalier Universitaire Lille, Lille, France
| | - Andres Metspalu
- Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Henry Völzke
- Institute for Community Medicine, Ernst-Moritz-Arndt-University, Greifswald, Germany
| | - Peter Vollenweider
- Department of Internal Medicine, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Kári Stefansson
- deCODE Genetics, Reykjavík, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Marjo-Riitta Järvelin
- Department of Epidemiology and Biostatistics, Imperial College London, London, United Kingdom
- MRC Health Protection Agency (HPA) Centre for Environment and Health, Imperial College London, London, United Kingdom
- Institute of Health Sciences, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
- Department of Lifecourse and Services, National Institute for Health and Welfare, Oulu, Finland
| | - Jacques S. Beckmann
- Service of Medical Genetics, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
- Department of Medical Genetics, University of Lausanne, Lausanne, Switzerland
| | - Philippe Froguel
- Department of Genomics of Common Disease, Imperial College London, London, United Kingdom
- CNRS 8199-Institute of Biology, Pasteur Institute, Lille, France
| | - Alexandra I. F. Blakemore
- Department of Genomics of Common Disease, Imperial College London, London, United Kingdom
- Section of Investigative Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
99
|
Abstract
Obesity increases the risk for type 2 diabetes through induction of insulin resistance. Treatment of type 2 diabetes has been limited by little translational knowledge of insulin resistance although there have been several well-documented hypotheses for insulin resistance. In those hypotheses, inflammation, mitochondrial dysfunction, hyperinsulinemia and lipotoxicity have been the major concepts and have received a lot of attention. Oxidative stress, endoplasmic reticulum (ER) stress, genetic background, aging, fatty liver, hypoxia and lipodystrophy are active subjects in the study of these concepts. However, none of those concepts or views has led to an effective therapy for type 2 diabetes. The reason is that there has been no consensus for a unifying mechanism of insulin resistance. In this review article, literature is critically analyzed and reinterpreted for a new energy-based concept of insulin resistance, in which insulin resistance is a result of energy surplus in cells. The energy surplus signal is mediated by ATP and sensed by adenosine monophosphate-activated protein kinase (AMPK) signaling pathway. Decreasing ATP level by suppression of production or stimulation of utilization is a promising approach in the treatment of insulin resistance. In support, many of existing insulin sensitizing medicines inhibit ATP production in mitochondria. The effective therapies such as weight loss, exercise, and caloric restriction all reduce ATP in insulin sensitive cells. This new concept provides a unifying cellular and molecular mechanism of insulin resistance in obesity, which may apply to insulin resistance in aging and lipodystrophy.
Collapse
|
100
|
Abstract
Chronic inflammation is a characteristic of obesity and is associated with accompanying insulin resistance, a hallmark of type 2 diabetes mellitus (T2DM). Although proinflammatory cytokines are known for their detrimental effects on adipose tissue function and insulin sensitivity, their beneficial effects in the regulation of metabolism have not drawn sufficient attention. In obesity, inflammation is initiated by a local hypoxia to augment angiogenesis and improve adipose tissue blood supply. A growing body of evidence suggests that macrophages and proinflammatory cytokines are essential for adipose remodeling and adipocyte differentiation. Phenotypes of multiple lines of transgenic mice consistently suggest that proinflammatory cytokines increase energy expenditure and act to prevent obesity. Removal of proinflammatory cytokines by gene knockout decreases energy expenditure and induces adult-onset obesity. In contrast, elevation of proinflammatory cytokines augments energy expenditure and decreases the risk for obesity. Anti-inflammatory therapies have been tested in more than a dozen clinical trials to improve insulin sensitivity and glucose homeostasis in patients with T2DM, and the results are not encouraging. One possible explanation is that anti-inflammatory therapies also attenuate the beneficial effects of inflammation in stimulating energy expenditure, which may have limited the efficacy of the treatment by promoting energy accumulation. Thus, the positive effects of proinflammatory events should be considered in evaluating the impact of inflammation in obesity and type 2 diabetes.
Collapse
Affiliation(s)
- Jianping Ye
- Antioxidant and Gene Regulation Laboratory, Pennington Biomedical Research Center, Louisiana State Univ. System, Baton Rouge, LA 70808, USA.
| | | |
Collapse
|