901
|
Halberg N, Schraw TD, Wang ZV, Kim JY, Yi J, Hamilton MP, Luby-Phelps K, Scherer PE. Systemic fate of the adipocyte-derived factor adiponectin. Diabetes 2009; 58:1961-70. [PMID: 19581422 PMCID: PMC2731534 DOI: 10.2337/db08-1750] [Citation(s) in RCA: 160] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE The adipocyte-derived secretory protein adiponectin has been widely studied and shown to have potent insulin-sensitizing, antiapoptotic, and anti-inflammatory properties. While its biosynthesis is well understood, its fate, once in circulation, is less well established. RESEARCH DESIGN AND METHODS Here, we examine the half-life of adiponectin in circulation by tracking fluorescently labeled recombinant adiponectin in the circulation, following it to its final destination in the hepatocyte. RESULTS Despite its abundant presence in plasma, adiponectin is cleared rapidly with a half-life of approximately 75 min. A more bioactive version carrying a mutation at cysteine 39 is cleared within minutes. Even though steady-state levels of adiponectin differ between male and female mice, we failed to detect any differences in clearance rates, suggesting that differences in plasma are mostly due to differential production rates. In a metabolically challenged state (high-fat diet exposure or in an ob/ob background), adiponectin levels are reduced in plasma and clearance is significantly prolonged, reflecting a dramatic drop in adiponectin production levels. CONCLUSIONS Combined, these results show a surprisingly rapid turnover of adiponectin with multiple fat pads contributing to the plasma levels of adiponectin and clearance mediated primarily by the liver. It is surprising that despite high-level production and rapid clearance, plasma levels of adiponectin remain remarkably constant.
Collapse
Affiliation(s)
- Nils Halberg
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Biomedical Sciences, Faculty of Health Science, University of Copenhagen, Copenhagen, Denmark
| | - Todd D. Schraw
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Zhao V. Wang
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Ja-Young Kim
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - James Yi
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Mark P. Hamilton
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Kate Luby-Phelps
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Philipp E. Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas
- Corresponding author: Philipp E. Scherer,
| |
Collapse
|
902
|
Bloomgarden ZT. The 6th Annual World Congress on the insulin resistance syndrome. Diabetes Care 2009; 32:e104-11. [PMID: 19717816 PMCID: PMC2744119 DOI: 10.2337/dc09-zb09] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
903
|
Kienesberger PC, Lee D, Pulinilkunnil T, Brenner DS, Cai L, Magnes C, Koefeler HC, Streith IE, Rechberger GN, Haemmerle G, Flier JS, Zechner R, Kim YB, Kershaw EE. Adipose triglyceride lipase deficiency causes tissue-specific changes in insulin signaling. J Biol Chem 2009; 284:30218-29. [PMID: 19723629 DOI: 10.1074/jbc.m109.047787] [Citation(s) in RCA: 98] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Triacylglycerol accumulation in insulin target tissues is associated with insulin resistance. Paradoxically, mice with global targeted deletion of adipose triglyceride lipase (ATGL), the rate-limiting enzyme in triacylglycerol hydrolysis, display improved glucose tolerance and insulin sensitivity despite triacylglycerol accumulation in multiple tissues. To determine the molecular mechanisms for this phenotype, ATGL-deficient (ATGL(-/-)) and wild-type mice were injected with saline or insulin (10 units/kg, intraperitoneally), and then phosphorylation and activities of key insulin-signaling proteins were determined in insulin target tissues (liver, adipose tissue, and muscle). Insulin signaling and/or glucose transport was also evaluated in isolated adipocytes and skeletal muscle ex vivo. In ATGL(-/-) mice, insulin-stimulated phosphatidylinositol 3-kinase and Akt activities as well as phosphorylation of critical residues of IRS1 (Tyr(P)-612) and Akt (Ser(P)-473) were increased in skeletal muscle in vivo. Insulin-stimulated phosphatidylinositol 3-kinase activity and total insulin receptor and insulin receptor substrate 1, but not other parameters, were also increased in white adipose tissue in vivo. In contrast, in vivo measures of insulin signaling were decreased in brown adipose tissue and liver. Interestingly, the enhanced components of insulin signaling identified in skeletal muscle and white adipose tissue in vivo and their expected downstream effects on glucose transport were not present ex vivo. ATGL deficiency altered intramyocellular lipids as well as serum factors known to influence insulin sensitivity. Thus, skeletal muscle, rather than other tissues, primarily contributes to enhanced insulin sensitivity in ATGL(-/-) mice in vivo despite triacylglycerol accumulation, and both local and systemic factors contribute to tissue-specific effects of global ATGL deficiency on insulin action.
Collapse
Affiliation(s)
- Petra C Kienesberger
- From the Institute of Molecular Biosciences, University of Graz, A-8010 Graz, Austria
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
904
|
Wang C, Xin X, Xiang R, Ramos FJ, Liu M, Lee HJ, Chen H, Mao X, Kikani CK, Liu F, Dong LQ. Yin-Yang regulation of adiponectin signaling by APPL isoforms in muscle cells. J Biol Chem 2009; 284:31608-15. [PMID: 19661063 DOI: 10.1074/jbc.m109.010355] [Citation(s) in RCA: 113] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
APPL1 is a newly identified adiponectin receptor-binding protein that positively mediates adiponectin signaling in cells. Here we report that APPL2, an isoform of APPL1 that forms a dimer with APPL1, can interacts with both AdipoR1 and AdipoR2 and acts as a negative regulator of adiponectin signaling in muscle cells. Overexpression of APPL2 inhibits the interaction between APPL1 and AdipoR1, leading to down-regulation of adiponectin signaling in C2C12 myotubes. In contrast, suppressing APPL2 expression by RNAi significantly enhances adiponectin-stimulated glucose uptake and fatty acid oxidation. In addition to targeting directly to and competing with APPL1 in binding with the adiponectin receptors, APPL2 also suppresses adiponectin and insulin signaling by sequestrating APPL1 from these two pathways. In addition to adiponectin, metformin also induces APPL1-APPL2 dissociation. Taken together, our results reveal that APPL isoforms function as an integrated Yin-Yang regulator of adiponectin signaling and mediate the cross-talk between adiponectin and insulin signaling pathways in muscle cells.
Collapse
Affiliation(s)
- Changhua Wang
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229-3900, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
905
|
Winer S, Chan Y, Paltser G, Truong D, Tsui H, Bahrami J, Dorfman R, Wang Y, Zielenski J, Mastronardi F, Maezawa Y, Drucker DJ, Engleman E, Winer D, Dosch HM. Normalization of obesity-associated insulin resistance through immunotherapy. Nat Med 2009; 15:921-9. [PMID: 19633657 PMCID: PMC3063199 DOI: 10.1038/nm.2001] [Citation(s) in RCA: 1094] [Impact Index Per Article: 68.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2009] [Accepted: 06/09/2009] [Indexed: 12/13/2022]
Abstract
Obesity and its associated metabolic syndromes represent a growing global challenge, yet mechanistic understanding of this pathology and current therapeutics are unsatisfactory. We discovered that CD4(+) T lymphocytes, resident in visceral adipose tissue (VAT), control insulin resistance in mice with diet-induced obesity (DIO). Analyses of human tissue suggest that a similar process may also occur in humans. DIO VAT-associated T cells show severely biased T cell receptor V(alpha) repertoires, suggesting antigen-specific expansion. CD4(+) T lymphocyte control of glucose homeostasis is compromised in DIO progression, when VAT accumulates pathogenic interferon-gamma (IFN-gamma)-secreting T helper type 1 (T(H)1) cells, overwhelming static numbers of T(H)2 (CD4(+)GATA-binding protein-3 (GATA-3)(+)) and regulatory forkhead box P3 (Foxp3)(+) T cells. CD4(+) (but not CD8(+)) T cell transfer into lymphocyte-free Rag1-null DIO mice reversed weight gain and insulin resistance, predominantly through T(H)2 cells. In obese WT and ob/ob (leptin-deficient) mice, brief treatment with CD3-specific antibody or its F(ab')(2) fragment, reduces the predominance of T(H)1 cells over Foxp3(+) cells, reversing insulin resistance for months, despite continuation of a high-fat diet. Our data suggest that the progression of obesity-associated metabolic abnormalities is under the pathophysiological control of CD4(+) T cells. The eventual failure of this control, with expanding adiposity and pathogenic VAT T cells, can successfully be reversed by immunotherapy.
Collapse
Affiliation(s)
- Shawn Winer
- Department of Pediatrics & Immunology, Research Institute, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
906
|
Shi H, Seeley RJ, Clegg DJ. Sexual differences in the control of energy homeostasis. Front Neuroendocrinol 2009; 30:396-404. [PMID: 19341761 PMCID: PMC4517605 DOI: 10.1016/j.yfrne.2009.03.004] [Citation(s) in RCA: 181] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2008] [Revised: 03/09/2009] [Accepted: 03/17/2009] [Indexed: 12/20/2022]
Abstract
The prevalence of obesity has reached epidemic proportion with enormous costs in both human lives and healthcare dollars spent. Obesity-related metabolic disorders are much lower in premenopausal women than men; however, there is a dramatic increase following menopause in women. The health risks associated with obesity vary depending on the location of adipose tissue. Adipose tissue distributed in the abdominal visceral carry a much greater risk for metabolic disorders than does adipose tissue distributed subcutaneously. There are distinct sex-dependent differences in the regional fat distribution, women carry more fat subcutaneously whereas men carry more fat viscerally. Males and females differ with respect to their regulation of energy homeostasis. Peripheral adiposity hormones such as leptin and insulin as well as sex hormones directly influence energy balance. Sexual dimorphisms in energy balance, body fat distribution, and the role sex hormones have in mediating these differences are the focus of this review.
Collapse
Affiliation(s)
- Haifei Shi
- Obesity Research Center, University of Cincinnati, Cincinnati, OH, USA
| | | | | |
Collapse
|
907
|
|
908
|
Perspectives on adipose tissue, chagas disease and implications for the metabolic syndrome. Interdiscip Perspect Infect Dis 2009; 2009:824324. [PMID: 19644556 PMCID: PMC2715900 DOI: 10.1155/2009/824324] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2009] [Accepted: 05/27/2009] [Indexed: 01/07/2023] Open
Abstract
The contribution of adipose tissue an
autocrine and endocrine organ in the
pathogenesis of infectious disease and metabolic
syndrome is gaining attention. Adipose tissue
and adipocytes
are one of the major targets of T. cruzi infection. Parasites are detected 300 days postinfection in adipose tissue. Infection of adipose tissue and cultured adipocytes triggered local
expression of inflammatory mediators resulting in the upregulation of cytokine and chemokine
levels. Adipose tissue obtained from infected mice display an increased infiltration of
inflammatory cells. Adiponectin, an adipocyte specific protein, which exerts antiinflammatory
effects, is reduced during the acute phase of infection. The antiinflammatory regulator
peroxisome proliferator activated receptor-γ (PPAR-γ) is downregulated in infected cultured
adipocytes and adipose tissue. T. cruzi infection is associated with an upregulation of signaling
pathways such as MAPKs, Notch and cyclin D, and reduced caveolin-1 expression.
Adiponectin null mice have a cardiomyopathy and thus we speculate that the T. cruzi-induced
reduction in adiponectin contributes to the T. cruzi-induced cardiomyopathy. While T. cruzi infection causes hypoglycemia which correlates with mortality, hyperglycemia is associated
with increased parasitemia and mortality. The T. cruzi-induced increase in macrophages in
adipose tissue taken together with the reduction in adiponectin and the associated
cardiomyopathy is reminiscent of the metabolic syndrome.
Collapse
|
909
|
Abstract
PURPOSE OF REVIEW To introduce the healthy obese phenotype, characterized by favorable cardiometabolic risk factors despite excess adipose tissue. The epidemiology of the healthy obese phenotype is presented, including associated risk of cardiovascular disease (CVD), and potential biologic mechanisms which may give rise to the phenotype are discussed. RECENT FINDINGS Although it appears that approximately 30% of obese individuals maintain healthy cardiometabolic profiles, little published data exist examining the healthy obese phenotype. The healthy obese do not appear to be at increased risk of incident CVD events compared with at-risk obese, and the location of adipose tissue and the metabolic characteristics of the fat in a given location, including the presence of ectopic fat and associated adipocytokine response, may give rise to the phenotype. Recent data also suggest that weight loss among healthy obese may adversely impact their favorable cardiometabolic profile. SUMMARY A high prevalence of the healthy obese phenotype has been reported, and these individuals appear to be at no increased risk of CVD. Further research is needed into the mechanisms that allow these individuals to maintain low risk of CVD despite excess adiposity and appropriate weight loss recommendations for this group.
Collapse
Affiliation(s)
- Rachel P Wildman
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York 10461, USA.
| |
Collapse
|
910
|
Hypoxia-inducible factor 1alpha induces fibrosis and insulin resistance in white adipose tissue. Mol Cell Biol 2009; 29:4467-83. [PMID: 19546236 DOI: 10.1128/mcb.00192-09] [Citation(s) in RCA: 666] [Impact Index Per Article: 41.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Adipose tissue can undergo rapid expansion during times of excess caloric intake. Like a rapidly expanding tumor mass, obese adipose tissue becomes hypoxic due to the inability of the vasculature to keep pace with tissue growth. Consequently, during the early stages of obesity, hypoxic conditions cause an increase in the level of hypoxia-inducible factor 1alpha (HIF1alpha) expression. Using a transgenic model of overexpression of a constitutively active form of HIF1alpha, we determined that HIF1alpha fails to induce the expected proangiogenic response. In contrast, we observed that HIF1alpha initiates adipose tissue fibrosis, with an associated increase in local inflammation. "Trichrome- and picrosirius red-positive streaks," enriched in fibrillar collagens, are a hallmark of adipose tissue suffering from the early stages of hypoxia-induced fibrosis. Lysyl oxidase (LOX) is a transcriptional target of HIF1alpha and acts by cross-linking collagen I and III to form the fibrillar collagen fibers. Inhibition of LOX activity by beta-aminoproprionitrile treatment results in a significant improvement in several metabolic parameters and further reduces local adipose tissue inflammation. Collectively, our observations are consistent with a model in which adipose tissue hypoxia serves as an early upstream initiator for adipose tissue dysfunction by inducing a local state of fibrosis.
Collapse
|
911
|
Affiliation(s)
- Jorge Plutzky
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
912
|
Richey JM, Woolcott OO, Stefanovski D, Harrison LN, Zheng D, Lottati M, Hsu IR, Kim SP, Kabir M, Catalano KJ, Chiu JD, Ionut V, Kolka C, Mooradian V, Bergman RN. Rimonabant prevents additional accumulation of visceral and subcutaneous fat during high-fat feeding in dogs. Am J Physiol Endocrinol Metab 2009; 296:E1311-8. [PMID: 19366874 PMCID: PMC3833919 DOI: 10.1152/ajpendo.90972.2008] [Citation(s) in RCA: 24] [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: 01/01/2023]
Abstract
We investigated whether rimonabant, a type 1 cannabinoid receptor antagonist, reduces visceral adipose tissue (VAT) and subcutaneous adipose tissue (SAT) in dogs maintained on a hypercaloric high-fat diet (HHFD). To determine whether energy expenditure contributed to body weight changes, we also calculated resting metabolic rate. Twenty male dogs received either rimonabant (1.25 mg.kg(-1).day(-1), orally; n = 11) or placebo (n = 9) for 16 wk, concomitant with a HHFD. VAT, SAT, and nonfat tissue were measured by magnetic resonance imaging. Resting metabolic rate was assessed by indirect calorimetry. By week 16 of treatment, rimonabant dogs lost 2.5% of their body weight (P = 0.029), whereas in placebo dogs body weight increased by 6.2% (P < 0.001). Rimonabant reduced food intake (P = 0.027), concomitant with a reduction of SAT by 19.5% (P < 0.001). In contrast with the VAT increase with placebo (P < 0.01), VAT did not change with rimonabant. Nonfat tissue remained unchanged in both groups. Body weight loss was not associated with either resting metabolic rate (r(2) = 0.24; P = 0.154) or food intake (r(2) = 0.24; P = 0.166). In conclusion, rimonabant reduced body weight together with a reduction in abdominal fat, mainly because of SAT loss. Body weight changes were not associated with either resting metabolic rate or food intake. The findings provide evidence of a peripheral effect of rimonabant to reduce adiposity and body weight, possibly through a direct effect on adipose tissue.
Collapse
Affiliation(s)
- Joyce M Richey
- Dept. of Physiology, Keck School of Medicine, Univ. of Southern California, 1333 San Pablo St., MMR 626, Los Angeles, CA 90033, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
913
|
Lee DE, Kehlenbrink S, Lee H, Hawkins M, Yudkin JS. Getting the message across: mechanisms of physiological cross talk by adipose tissue. Am J Physiol Endocrinol Metab 2009; 296:E1210-29. [PMID: 19258492 DOI: 10.1152/ajpendo.00015.2009] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Obesity is associated with resistance of skeletal muscle to insulin-mediated glucose uptake, as well as resistance of different organs and tissues to other metabolic and vascular actions of insulin. In addition, the body is exquisitely sensitive to nutrient imbalance, with energy excess or a high-fat diet rapidly increasing insulin resistance, even before noticeable changes occur in fat mass. There is a growing acceptance of the fact that, as well as acting as a storage site for surplus energy, adipose tissue is an important source of signals relevant to, inter alia, energy homeostasis, fertility, and bone turnover. It has also been widely recognized that obesity is a state of low-grade inflammation, with adipose tissue generating substantial quantities of proinflammatory molecules. At a cellular level, the understanding of the signaling pathways responsible for such alterations has been intensively investigated. What is less clear, however, is how alterations of physiology, and of signaling, within one cell or one tissue are communicated to other parts of the body. The concepts of cell signals being disseminated systemically through a circulating "endocrine" signal have been complemented by the view that local signaling may similarly occur through autocrine or paracrine mechanisms. Yet, while much elegant work has focused on the alterations in signaling that are found in obesity or energy excess, there has been less attention paid to ways in which such signals may propagate to remote organs. This review of the integrative physiology of obesity critically appraises the data and outlines a series of hypotheses as to how interorgan cross talk takes place. The hypotheses presented include the "fatty acid hypothesis,", the "portal hypothesis,", the "endocrine hypothesis,", the "inflammatory hypothesis,", the "overflow hypothesis,", a novel "vasocrine hypothesis," and a "neural hypothesis," and the strengths and weaknesses of each hypothesis are discussed.
Collapse
Affiliation(s)
- Do-Eun Lee
- Department of Internal Medicine, Division of Endocrinology, Winthrop University Hospital, London, United Kingdom
| | | | | | | | | |
Collapse
|
914
|
Landskroner-Eiger S, Qian B, Muise ES, Nawrocki AR, Berger JP, Fine EJ, Koba W, Deng Y, Pollard JW, Scherer PE. Proangiogenic contribution of adiponectin toward mammary tumor growth in vivo. Clin Cancer Res 2009; 15:3265-76. [PMID: 19447867 PMCID: PMC3237387 DOI: 10.1158/1078-0432.ccr-08-2649] [Citation(s) in RCA: 119] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
PURPOSE Adipocytes represent one of the most abundant constituents of the mammary gland. They are essential for mammary tumor growth and survival. Metabolically, one of the more important fat-derived factors ("adipokines") is adiponectin (APN). Serum concentrations of APN negatively correlate with body mass index and insulin resistance. To explore the association of APN with breast cancer and tumor angiogenesis, we took an in vivo approach aiming to study its role in the mouse mammary tumor virus (MMTV)-polyoma middle T antigen (PyMT) mammary tumor model. EXPERIMENTAL DESIGN We compared the rates of tumor growth in MMTV-PyMT mice in wild-type and APN-null backgrounds. RESULTS Histology and micro-positron emission tomography imaging show that the rate of tumor growth is significantly reduced in the absence of APN at early stages. PyMT/APN knockout mice exhibit a reduction in their angiogenic profile resulting in nutrient deprivation of the tumors and tumor-associated cell death. Surprisingly, in more advanced malignant stages of the disease, tumor growth develops more aggressively in mice lacking APN, giving rise to a larger tumor burden, an increase in the mobilization of circulating endothelial progenitor cells, and a gene expression fingerprint indicative of more aggressive tumor cells. CONCLUSIONS These observations highlight a novel important contribution of APN in mammary tumor development and angiogenesis, indicating that APN has potent angio-mimetic properties in tumor vascularization. However, in tumors deprived of APN, this antiangiogenic stress results in an adaptive response that fuels tumor growth through mobilization of circulating endothelial progenitor cells and the development of mechanisms enabling massive cell proliferation despite a chronically hypoxic microenvironment.
Collapse
MESH Headings
- Adiponectin/blood
- Adiponectin/genetics
- Adiponectin/metabolism
- Animals
- Antigens, Viral, Tumor/genetics
- Apoptosis
- Blotting, Western
- Female
- Flow Cytometry
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic/drug effects
- Glucose/metabolism
- Glucose/pharmacokinetics
- Male
- Mammary Neoplasms, Experimental/blood supply
- Mammary Neoplasms, Experimental/genetics
- Mammary Neoplasms, Experimental/pathology
- Mammary Tumor Virus, Mouse/genetics
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
- PPAR gamma/agonists
- PPAR gamma/metabolism
- Polyomavirus/genetics
- Positron-Emission Tomography
- Reverse Transcriptase Polymerase Chain Reaction
- Thiazolidinediones/pharmacology
- Time Factors
- Tumor Burden/drug effects
- Vascular Endothelial Growth Factor A/genetics
Collapse
Affiliation(s)
- Shira Landskroner-Eiger
- Department of Cell Biology, Center of Reproductive Biology and Womens’ Health, Albert Einstein Cancer Center, Bronx, New York
| | - Binzhi Qian
- Department of Developmental and Molecular Biology, Center of Reproductive Biology and Womens’ Health, Albert Einstein Cancer Center, Bronx, New York
| | - Eric S. Muise
- Department of Molecular Profiling, Merck Research Laboratories, Rahway, New Jersey
| | - Andrea R. Nawrocki
- Department of Cell Biology, Center of Reproductive Biology and Womens’ Health, Albert Einstein Cancer Center, Bronx, New York
| | - Joel P. Berger
- Department of Metabolic Disorders, Merck Research Laboratories, Rahway, New Jersey
| | - Eugene J. Fine
- Department of Nuclear Medicine, M. Donald Blaufox Laboratory for Molecular Imaging, Albert Einstein College of Medicine, Bronx, New York
| | - Wade Koba
- Department of Nuclear Medicine, M. Donald Blaufox Laboratory for Molecular Imaging, Albert Einstein College of Medicine, Bronx, New York
| | - Yingfeng Deng
- Touchstone Diabetes Center, Departments of Internal Medicine, Cell Biology and Simmons Cancer, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jeffrey W. Pollard
- Department of Developmental and Molecular Biology, Center of Reproductive Biology and Womens’ Health, Albert Einstein Cancer Center, Bronx, New York
| | - Philipp E. Scherer
- Touchstone Diabetes Center, Departments of Internal Medicine, Cell Biology and Simmons Cancer, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
915
|
The immunopathogenesis of alcoholic and nonalcoholic steatohepatitis: two triggers for one disease? Semin Immunopathol 2009; 31:359-69. [DOI: 10.1007/s00281-009-0152-9] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2009] [Accepted: 04/28/2009] [Indexed: 02/08/2023]
|
916
|
Hegarty BD, Turner N, Cooney GJ, Kraegen EW. Insulin resistance and fuel homeostasis: the role of AMP-activated protein kinase. Acta Physiol (Oxf) 2009; 196:129-45. [PMID: 19245658 DOI: 10.1111/j.1748-1716.2009.01968.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The worldwide prevalence of type 2 diabetes (T2D) and related disorders of the metabolic syndrome (MS) has reached epidemic proportions. Insulin resistance (IR) is a major perturbation that characterizes these disorders. Extra-adipose accumulation of lipid, particularly within the liver and skeletal muscle, is closely linked with the development of IR. The AMP-activated protein kinase (AMPK) pathway plays an important role in the regulation of both lipid and glucose metabolism. Through its effects to increase fatty acid oxidation and inhibit lipogenesis, AMPK activity in the liver and skeletal muscle could be expected to ameliorate lipid accumulation and associated IR in these tissues. In addition, AMPK promotes glucose uptake into skeletal muscle and suppresses glucose output from the liver via insulin-independent mechanisms. These characteristics make AMPK a highly attractive target for the development of strategies to curb the prevalence and costs of T2D. Recent insights into the regulation of AMPK and mechanisms by which it modulates fuel metabolism in liver and skeletal muscle are discussed here. In addition, we consider the arguments for and against the hypothesis that dysfunctional AMPK contributes to IR. Finally we review studies which assess AMPK as an appropriate target for the prevention and treatment of T2D and MS.
Collapse
Affiliation(s)
- B D Hegarty
- Diabetes and Obesity Research Program, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, NSW 2010, Australia.
| | | | | | | |
Collapse
|
917
|
Yoshida T, Nagasaki H, Asato Y, Ohta T. The ratio of high-molecular weight adiponectin and total adiponectin differs in preterm and term infants. Pediatr Res 2009; 65:580-3. [PMID: 19127209 DOI: 10.1203/pdr.0b013e3181995103] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Adiponectin consists of three subspecies (high-, middle- and low-molecular weight adiponectin). Among these, high-molecular weight adiponectin (H-adn) is suggested to be an active form of this protein. To assess the relationship between H-adn and postnatal growth in preterm infants (PIs), serum H-adn and total adiponectin (T-adn) were measured in 46 PIs at birth and at corrected term, and 26 term infants (TI) at birth. T-adn and H-adn concentrations, and the ratio of H-adn to T-adn (H/T-adn) were significantly greater in TI and PI at corrected term than in PI at birth (p < 0.001). T-adn and H-adn concentrations in PI at corrected term were similar to those in TI, but H/T-adn in PI at corrected term was less than that in TI (p < 0.02). Stepwise multiple regression analysis revealed that the factors contributing to H/T-adn and serum concentrations of T- and H-adn in PI at corrected term were different from those in TI. These data suggest that quality of early postnatal growth in PIs is different from that in normally developed TI. Postnatal growth accompanying adipose tissue similar to TI may be important for PI to prevent future development of cardiovascular disease.
Collapse
Affiliation(s)
- Tomohide Yoshida
- Department of Pediatrics, University of the Ryukyus, Nishihara, Okinawa 903-0125, Japan
| | | | | | | |
Collapse
|
918
|
Cao J, Inoue K, Li X, Drummond G, Abraham NG. Physiological significance of heme oxygenase in hypertension. Int J Biochem Cell Biol 2009; 41:1025-33. [PMID: 19027871 PMCID: PMC2745554 DOI: 10.1016/j.biocel.2008.10.025] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2008] [Revised: 10/21/2008] [Accepted: 10/27/2008] [Indexed: 02/06/2023]
Abstract
The last decade has witnessed an explosion in the elucidation of the role that the heme oxygenase system plays in human physiology. This system encompasses not only the heme degradative pathway, including heme oxygenase and biliverdin reductase, but also the products of heme degradation, carbon monoxide, iron, and biliverdin/bilirubin. Their role in diabetes, inflammation, heart disease, hypertension, transplantation, and pulmonary disease are areas of burgeoning research. The research has focused not only on heme itself but also on its metabolic products as well as endogenous compounds involved in a vast number of genetic and metabolic processes that are affected when heme metabolism is perturbed. It should be noted, however, that although the use of carbon monoxide and biliverdin/bilirubin as therapeutic agents has been successful, these agents can be toxic at high levels in tissue, e.g., kernicterus. Care must be used to ensure that when these compounds are used as therapeutic agents their deleterious effects are minimized or avoided. On balance, however, the strategies to target heme oxygenase-1 as described in this review offer promising therapeutic approaches to clinicians for the effective management of hypertension and renal function. The approaches detailed may prove to be seminal in the development of a new therapeutic strategy to treat hypertension.
Collapse
Affiliation(s)
- Jian Cao
- New York Medical College, Department of Pharmacology, Valhalla, NY 10595, United States
| | - Kazuyoshi Inoue
- Department of Geriatric Cardiology, Chinese PLA General Hospital, Beijing 100853, China
| | - Xiaoying Li
- Department of Geriatric Cardiology, Chinese PLA General Hospital, Beijing 100853, China
| | - George Drummond
- New York Medical College, Department of Pharmacology, Valhalla, NY 10595, United States
| | - Nader G. Abraham
- New York Medical College, Department of Pharmacology, Valhalla, NY 10595, United States
| |
Collapse
|
919
|
Nunn AVW, Bell JD, Guy GW. Lifestyle-induced metabolic inflexibility and accelerated ageing syndrome: insulin resistance, friend or foe? Nutr Metab (Lond) 2009; 6:16. [PMID: 19371409 PMCID: PMC2678135 DOI: 10.1186/1743-7075-6-16] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2008] [Accepted: 04/16/2009] [Indexed: 12/16/2022] Open
Abstract
The metabolic syndrome may have its origins in thriftiness, insulin resistance and one of the most ancient of all signalling systems, redox. Thriftiness results from an evolutionarily-driven propensity to minimise energy expenditure. This has to be balanced with the need to resist the oxidative stress from cellular signalling and pathogen resistance, giving rise to something we call 'redox-thriftiness'. This is based on the notion that mitochondria may be able to both amplify membrane-derived redox growth signals as well as negatively regulate them, resulting in an increased ATP/ROS ratio. We suggest that 'redox-thriftiness' leads to insulin resistance, which has the effect of both protecting the individual cell from excessive growth/inflammatory stress, while ensuring energy is channelled to the brain, the immune system, and for storage. We also suggest that fine tuning of redox-thriftiness is achieved by hormetic (mild stress) signals that stimulate mitochondrial biogenesis and resistance to oxidative stress, which improves metabolic flexibility. However, in a non-hormetic environment with excessive calories, the protective nature of this system may lead to escalating insulin resistance and rising oxidative stress due to metabolic inflexibility and mitochondrial overload. Thus, the mitochondrially-associated resistance to oxidative stress (and metabolic flexibility) may determine insulin resistance. Genetically and environmentally determined mitochondrial function may define a 'tipping point' where protective insulin resistance tips over to inflammatory insulin resistance. Many hormetic factors may induce mild mitochondrial stress and biogenesis, including exercise, fasting, temperature extremes, unsaturated fats, polyphenols, alcohol, and even metformin and statins. Without hormesis, a proposed redox-thriftiness tipping point might lead to a feed forward insulin resistance cycle in the presence of excess calories. We therefore suggest that as oxidative stress determines functional longevity, a rather more descriptive term for the metabolic syndrome is the 'lifestyle-induced metabolic inflexibility and accelerated ageing syndrome'. Ultimately, thriftiness is good for us as long as we have hormetic stimuli; unfortunately, mankind is attempting to remove all hormetic (stressful) stimuli from his environment.
Collapse
Affiliation(s)
- Alistair VW Nunn
- Metabolic and Molecular Imaging Group, MRC Clinical Sciences Centre, Hammersmith Hospital, Imperial College London, Du Cane Road, London, W12 OHS, UK
| | - Jimmy D Bell
- Metabolic and Molecular Imaging Group, MRC Clinical Sciences Centre, Hammersmith Hospital, Imperial College London, Du Cane Road, London, W12 OHS, UK
| | | |
Collapse
|
920
|
|
921
|
Shetty S, Kusminski CM, Scherer PE. Adiponectin in health and disease: evaluation of adiponectin-targeted drug development strategies. Trends Pharmacol Sci 2009; 30:234-9. [PMID: 19359049 DOI: 10.1016/j.tips.2009.02.004] [Citation(s) in RCA: 186] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2008] [Revised: 02/16/2009] [Accepted: 02/18/2009] [Indexed: 12/15/2022]
Abstract
Adiponectin is a secretory protein predominantly expressed by adipocytes and released at a high rate into circulation. The ease with which the levels of adiponectin can be measured owing to its high abundance, small diurnal variation and high stability in plasma have made it a popular target for measurements in many clinical studies. It has emerged as a valuable biomarker for insulin sensitivity, cardiovascular risk and inflammation. However, adiponectin levels have been measured in many additional disease states. Preclinical studies not only have implicated adiponectin as an outstanding biomarker but also have demonstrated direct cardio-protective and insulin-sensitizing properties to be associated with the protein. Adiponectin might, therefore, be a viable protein therapeutic that could be supplied in a recombinant form in the context of type 2 diabetes and cardiovascular disease. However, the high abundance, complex tertiary and quaternary structure and rapid turnover might make chronic administration of the protein rather challenging.
Collapse
Affiliation(s)
- Shoba Shetty
- Touchstone Diabetes Center, Departments of Internal Medicine and Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA
| | | | | |
Collapse
|
922
|
Chakravarthy MV, Zhu Y, Yin L, Coleman T, Pappan KL, Marshall CA, McDaniel ML, Semenkovich CF. Inactivation of hypothalamic FAS protects mice from diet-induced obesity and inflammation. J Lipid Res 2009; 50:630-40. [PMID: 19029118 PMCID: PMC2656656 DOI: 10.1194/jlr.m800379-jlr200] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2008] [Revised: 10/24/2008] [Indexed: 01/22/2023] Open
Abstract
Obesity promotes insulin resistance and chronic inflammation. Disrupting any of several distinct steps in lipid synthesis decreases adiposity, but it is unclear if this approach coordinately corrects the environment that propagates metabolic disease. We tested the hypothesis that inactivation of FAS in the hypothalamus prevents diet-induced obesity and systemic inflammation. Ten weeks of high-fat feeding to mice with inactivation of FAS (FASKO) limited to the hypothalamus and pancreatic beta cells protected them from diet-induced obesity. Though high-fat fed FASKO mice had no beta-cell phenotype, they were hypophagic and hypermetabolic, and they had increased insulin sensitivity at the liver but not the periphery as demonstrated by hyperinsulinemic-euglycemic clamps, and biochemically by increased phosphorylated Akt, glycogen synthase kinase-3beta, and FOXO1 compared with wild-type mice. High-fat fed FASKO mice had decreased excretion of urinary isoprostanes, suggesting less oxidative stress and blunted tumor necrosis factor alpha (TNFalpha) and interleukin-6 (IL-6) responses to endotoxin, suggesting less systemic inflammation. Pair-feeding studies demonstrated that these beneficial effects were dependent on central FAS disruption and not merely a consequence of decreased adiposity. Thus, inducing central FAS deficiency may be a valuable integrative strategy for treating several components of the metabolic syndrome, in part by correcting hepatic insulin resistance and suppressing inflammation.
Collapse
Affiliation(s)
- Manu V. Chakravarthy
- Department of Medicine, Division of Endocrinology, Metabolism & Lipid Research, Washington University School of Medicine in St. Louis, MO 63110
| | - Yimin Zhu
- Department of Medicine, Division of Endocrinology, Metabolism & Lipid Research, Washington University School of Medicine in St. Louis, MO 63110
| | - Li Yin
- Department of Medicine, Division of Endocrinology, Metabolism & Lipid Research, Washington University School of Medicine in St. Louis, MO 63110
| | - Trey Coleman
- Department of Medicine, Division of Endocrinology, Metabolism & Lipid Research, Washington University School of Medicine in St. Louis, MO 63110
| | - Kirk L. Pappan
- Department of Pathology & Immunology, Washington University School of Medicine in St. Louis, MO 63110
| | - Connie A. Marshall
- Department of Pathology & Immunology, Washington University School of Medicine in St. Louis, MO 63110
| | - Michael L. McDaniel
- Department of Pathology & Immunology, Washington University School of Medicine in St. Louis, MO 63110
| | - Clay F. Semenkovich
- Department of Medicine, Division of Endocrinology, Metabolism & Lipid Research, Washington University School of Medicine in St. Louis, MO 63110
- Department of Cell Biology & Physiology, Washington University School of Medicine in St. Louis, MO 63110
| |
Collapse
|
923
|
Holland WL, Scherer PE. PAQRs: a counteracting force to ceramides? Mol Pharmacol 2009; 75:740-3. [PMID: 19158359 PMCID: PMC2684919 DOI: 10.1124/mol.109.054817] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2009] [Accepted: 01/21/2009] [Indexed: 12/25/2022] Open
Abstract
In recent years, sphingolipids have garnered increasing attention for their roles in modulating intracellular signaling events. Circulating factors associated with obesity promote excess accumulation of ceramide or glucosylceramide derivatives, which impair insulin action in peripheral tissues. In this issue, Villa et al. (p. 866) provide evidence that, in yeast, the progestin and adipoQ receptor superfamily of receptors mediate their effects via a novel ceramidase activity, generating sphingoid base as a second messenger.
Collapse
Affiliation(s)
- William L Holland
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390-8549, USA
| | | |
Collapse
|
924
|
Liu HY, Yehuda-Shnaidman E, Hong T, Han J, Pi J, Liu Z, Cao W. Prolonged exposure to insulin suppresses mitochondrial production in primary hepatocytes. J Biol Chem 2009; 284:14087-95. [PMID: 19336408 DOI: 10.1074/jbc.m807992200] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Insulin is the central regulator of metabolism and is necessary for storing energy as fat efficiently. Mitochondria are primary sites of energy consumption of most cells. Increased plasma insulin level and mitochondrial dysfunction are features of insulin resistance. The exact role of insulin in regulation of mitochondrial production and function remains unestablished. In this study, we observed that mitochondrial production in liver and skeletal muscle gastrocnemius was increased in mice with insulin deficiency (streptozotocin-induced type 1 diabetes). In contrast, prolonged exposure (24 h) of isolated hepatocytes to insulin decreased mitochondrial mass, mitochondrial DNA (mtDNA), intracellular ATP content, and cellular O(2) consumption. Transcript levels of genes associated with mitochondrial production and beta oxidation were decreased, whereas those of lipogenic genes were increased by the prolonged exposure to insulin. Insulin-induced changes in mtDNA, mitochondrial mass, intracellular ATP content, and transcripts of mitochondrion-associated genes were prevented by blockade of Akt activation with the phosphatidylinositol 3-kinase inhibitor LY294002. Conversely, levels of mtDNA, intracellular ATP content, and expression of mitochondrion-associated genes were decreased by overexpression of the constitutively active Akt. Finally, insulin suppression of mtDNA, ATP production, and expression of mitochondrion-related genes was largely prevented by inhibition of cyclic nucleotide phosphodiesterase with isobutylmethylxanthine. Together, our results show prolonged exposure of isolated hepatocytes to insulin suppresses mitochondrial production and function through the classical Akt-dependent insulin signaling pathway.
Collapse
Affiliation(s)
- Hui-Yu Liu
- Translational Biology, The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina 27709, USA
| | | | | | | | | | | | | |
Collapse
|
925
|
van Eijk M, Aten J, Bijl N, Ottenhoff R, van Roomen CPAA, Dubbelhuis PF, Seeman I, Ghauharali-van der Vlugt K, Overkleeft HS, Arbeeny C, Groen AK, Aerts JMFG. Reducing glycosphingolipid content in adipose tissue of obese mice restores insulin sensitivity, adipogenesis and reduces inflammation. PLoS One 2009; 4:e4723. [PMID: 19305508 PMCID: PMC2654925 DOI: 10.1371/journal.pone.0004723] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2008] [Accepted: 01/14/2009] [Indexed: 01/09/2023] Open
Abstract
Adipose tissue is a critical mediator in obesity-induced insulin resistance. Previously we have demonstrated that pharmacological lowering of glycosphingolipids and subsequently GM3 by using the iminosugar AMP-DNM, strikingly improves glycemic control. Here we studied the effects of AMP-DNM on adipose tissue function and inflammation in detail to provide an explanation for the observed improved glucose homeostasis. Leptin-deficient obese (LepOb) mice were fed AMP-DNM and its effects on insulin signalling, adipogenesis and inflammation were monitored in fat tissue. We show that reduction of glycosphingolipid biosynthesis in adipose tissue of LepOb mice restores insulin signalling in isolated ex vivo insulin-stimulated adipocytes. We observed improved adipogenesis as the number of larger adipocytes was reduced and expression of genes like peroxisome proliferator-activated receptor (PPAR) γ, insulin responsive glucose transporter (GLUT)-4 and adipsin increased. In addition, we found that adiponectin gene expression and protein were increased by AMP-DNM. As a consequence of this improved function of fat tissue we observed less inflammation, which was characterized by reduced numbers of adipose tissue macrophages (crown-like structures) and reduced levels of the macrophage chemo attractants monocyte-chemoattractant protein-1 (Mcp-1/Ccl2) and osteopontin (OPN). In conclusion, pharmacological lowering of glycosphingolipids by inhibition of glucosylceramide biosynthesis improves adipocyte function and as a consequence reduces inflammation in adipose tissue of obese animals.
Collapse
Affiliation(s)
- Marco van Eijk
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
926
|
Abstract
PURPOSE OF REVIEW Earlier studies investigating the risk of developing coronary heart disease in relation to body fat distribution showed inconsistent results, and any sex-related difference in disease risk has not been adequately examined. This review aims to assess current findings on the prospective association between body fat distribution measures and coronary heart disease in men and women. RECENT FINDINGS Current epidemiologic evidence suggests that waist circumference and waist-hip ratio, as indicators of abdominal adiposity, are positively related to coronary heart disease in men and women independently of body mass index and conventional coronary heart disease risk factors. But the magnitude and shape of the associations for these abdominal adiposity indices varied with adjustments for mediating and confounding factors. Interestingly, hip waist circumference was inversely associated with coronary heart disease after adjusting for waist circumference. Because waist and hips are positively correlated but have separate and opposite associations with coronary disease, using waist circumference alone may provide underestimated risk estimate if hip girth is not accounted for in the calculation of this risk. SUMMARY For adipose tissue distribution assessment to be clinically useful, the ideal adiposity phenotype should provide a single risk estimate that captures the separate 'effects' of abdominal and peripheral adiposity. Although far from perfect, waist-hip ratio may capture separate effects of central and peripheral adiposity. This simple and inexpensive measure could be used to help improve coronary heart disease risk assessment.
Collapse
|
927
|
Nicolai A, Li M, Kim DH, Peterson SJ, Vanella L, Positano V, Gastaldelli A, Rezzani R, Rodella LF, Drummond G, Kusmic C, L’Abbate A, Kappas A, Abraham NG. Heme oxygenase-1 induction remodels adipose tissue and improves insulin sensitivity in obesity-induced diabetic rats. Hypertension 2009; 53:508-15. [PMID: 19171794 PMCID: PMC2745551 DOI: 10.1161/hypertensionaha.108.124701] [Citation(s) in RCA: 149] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2008] [Accepted: 12/06/2008] [Indexed: 12/16/2022]
Abstract
Obesity-associated inflammation causes insulin resistance. Obese adipose tissue displays hypertrophied adipocytes and increased expression of the cannabinoid-1 receptor. Cobalt protoporphyrin (CoPP) increases heme oxygenase-1 (HO-1) activity, increasing adiponectin and reducing inflammatory cytokines. We hypothesize that CoPP administration to Zucker diabetic fat (ZDF) rats would improve insulin sensitivity and remodel adipose tissue. Twelve-week-old Zucker lean and ZDF rats were divided into 4 groups: Zucker lean, Zucker lean-CoPP, ZDF, and ZDF-CoPP. Control groups received vehicle and treatment groups received CoPP (2 mg/kg body weight) once weekly for 6 weeks. Serum insulin levels and glucose response to insulin injection were measured. At 18 weeks of age, rats were euthanized, and aorta, kidney, and subcutaneous and visceral adipose tissues were harvested. HO-1 expression was measured by Western blot analysis and HO-1 activity by serum carbon monoxide content. Adipocyte size and cannabinoid-1 expression were measured. Adipose tissue volumes were determined using MRI. CoPP significantly increased HO-1 activity, phosphorylated AKT and phosphorylated AMP kinase, and serum adiponectin in ZDF rats. HO-1 induction improved hyperinsulinemia and insulin sensitivity in ZDF rats. Subcutaneous and visceral adipose tissue volumes were significantly decreased in ZDF rats. Adipocyte size and cannabinoid-1 expression were both significantly reduced in ZDF-CoPP rats in subcutaneous and visceral adipose tissues. This study demonstrates that HO-1 induction improves insulin sensitivity, downregulates the peripheral endocannabinoid system, reduces adipose tissue volume, and causes adipose tissue remodeling in a model of obesity-induced insulin resistance. These findings suggest HO-1 as a potential therapeutic target for obesity and its associated health risks.
Collapse
Affiliation(s)
| | - Ming Li
- Departments of Pharmacology New York Medical College, Valhalla
| | - Dong Hyun Kim
- Departments of Pharmacology New York Medical College, Valhalla
| | - Stephen J. Peterson
- Departments of Pharmacology New York Medical College, Valhalla
- Departments of Medicine New York Medical College, Valhalla
| | - Luca Vanella
- Departments of Pharmacology New York Medical College, Valhalla
| | - Vincenzo Positano
- Scuola Superiore Sant’Anna and Consiglio Nazionale delle Ricerche Institute of Clinical Physiology Pisa, Italy
| | - Amalia Gastaldelli
- Scuola Superiore Sant’Anna and Consiglio Nazionale delle Ricerche Institute of Clinical Physiology Pisa, Italy
| | | | | | - George Drummond
- Departments of Pharmacology New York Medical College, Valhalla
| | - Claudia Kusmic
- Scuola Superiore Sant’Anna and Consiglio Nazionale delle Ricerche Institute of Clinical Physiology Pisa, Italy
| | - Antonio L’Abbate
- Scuola Superiore Sant’Anna and Consiglio Nazionale delle Ricerche Institute of Clinical Physiology Pisa, Italy
| | | | - Nader G. Abraham
- Departments of Pharmacology New York Medical College, Valhalla
- Departments of Medicine New York Medical College, Valhalla
| |
Collapse
|
928
|
Egger G, Dixon J. Should obesity be the main game? Or do we need an environmental makeover to combat the inflammatory and chronic disease epidemics? Obes Rev 2009; 10:237-49. [PMID: 19055538 DOI: 10.1111/j.1467-789x.2008.00542.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
There is a link between obesity and chronic disease. However, the causal relationship is complicated. Some forms of obesity are associated with low-level systemic inflammation, which is linked to disease. But lifestyle behaviours that may not necessarily cause obesity (poor diet, inadequate sleep, smoking, etc.) can independently cause inflammation and consequent disease. It is proposed here that it is the environment driving modern lifestyles, which is the true cause of much chronic disease, rather than obesity per se, and that obesity may be a marker of environmental derangement, rather than the primary cause of the problem. Attempts to clinically manage obesity alone on a large scale are therefore unlikely to be successful at the population level without significant lifestyle or environmental change. Environmental factors influencing obesity and health have now also been implicated in ecological perturbations such as climate change, through the shift to positive energy balance in humans caused by the exponential use of fossil fuels in such areas as transport, and consequent rises in carbon emissions into the atmosphere. It is proposed therefore that a more policy-based approach to dealing with obesity, which attacks the common causes of both biological and ecological 'dis-ease', could have positive effects on both chronic disease and environmental problems. A plea is thus made for a greater health input into discussions on environmental regulation for chronic disease control, as well as climate change.
Collapse
Affiliation(s)
- G Egger
- Health and Applied Sciences, Southern Cross University, Australia.
| | | |
Collapse
|
929
|
Abstract
UNLABELLED Nonalcoholic fatty liver disease is a burgeoning problem. We have previously shown that Hispanics were at greater risk for nonalcoholic fatty liver disease than were African-Americans despite a similar prevalence of risk factors between these groups. We have performed the largest, population-based study to date (n = 2170) utilizing proton magnetic resonance (MR) spectroscopy, dual-energy x-ray absorptiometry, and multislice abdominal MR imaging to determine the contribution of body fat distribution to the differing prevalence of hepatic steatosis in the three major U.S. ethnic groups (African-American, Hispanic, Caucasian). Despite controlling for age and total adiposity, African-Americans had less intraperitoneal (IP) fat and more lower extremity fat than their Hispanic and Caucasian counterparts. The differences in hepatic triglyceride content (HTGC) between these groups remained after controlling for total, abdominal subcutaneous, and lower extremity adiposity; however, controlling for IP fat nearly abolished the differences in HTGC, indicating a close association between IP and liver fat regardless of ethnicity. Despite the lower levels of IP and liver fat in African-Americans, their prevalence of insulin resistance was similar to Hispanics, who had the highest levels of IP and liver fat. Furthermore, insulin levels and homeostasis model assessment values were highest and serum triglyceride levels were lowest among African-Americans after controlling for IP fat. CONCLUSION IP fat is linked to HTGC, irrespective of ethnicity. The differing prevalence of hepatic steatosis between these groups was associated with similar differences in visceral adiposity. The metabolic response to obesity and insulin resistance differs in African-Americans when compared to either Hispanics or Caucasians: African-Americans appear to be more resistant to both the accretion of triglyceride in the abdominal visceral compartment (adipose tissue and liver) and hypertriglyceridemia associated with insulin resistance.
Collapse
Affiliation(s)
- Richard Guerrero
- Department of Internal Medicine, The University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
- Advanced Imaging Research Center, The University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| | - Gloria L. Vega
- Donald W. Reynolds Cardiovascular Clinical Research Center, The University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
- Center for Human Nutrition, The University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| | - Scott M. Grundy
- Donald W. Reynolds Cardiovascular Clinical Research Center, The University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
- Center for Human Nutrition, The University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
- Veterans Affairs Medical Center, Dallas, Texas, USA
| | - Jeffrey D. Browning
- Department of Internal Medicine, The University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
- Advanced Imaging Research Center, The University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| |
Collapse
|
930
|
Abstract
Obesity leads to insulin resistance because the larger adipocytes in obese persons secrete proinflammatory cytokines that cause chronic inflammation in adipose tissue. This, in turn, leads to the alteration of adipokine secretion, which can induce insulin resistance. However, the development of insulin resistance without obesity is still obscure. We aimed to use an animal inflammation model with cotton pellet granuloma (CPG) in adipose tissue to characterize insulin resistance formation. We found that CPG in epididymal white adipose tissue (WAT), rather than in interscapular brown adipose tissue, impaired insulin sensitivity, and glucose utilization, and that it decreased levels of phosphoinsulin receptor and phospho-Akt in both muscle and liver tissue, but that it did not modify the body weight or food intake in mice. Macrophage infiltration in adipose tissue, leukocyte counts, monocyte chemoattractant protein-1, and interleukin-6 were elevated in CPG-treated mice. However, we found a marked decrease of plasma adiponectin only in the WAT group, which might have been because of the lower level of peroxisome proliferator-activated receptor-gamma in WAT. These results show that granuloma formation in WAT by implantation of a cotton pellet may induce insulin resistance under nonobese condition through circulating inflammatory mediators, especially the low level of adiponectin.
Collapse
|
931
|
Qi L, Saberi M, Zmuda E, Wang Y, Altarejos J, Zhang X, Dentin R, Hedrick S, Bandyopadhyay G, Hai T, Olefsky J, Montminy M. Adipocyte CREB promotes insulin resistance in obesity. Cell Metab 2009; 9:277-86. [PMID: 19254572 PMCID: PMC2730923 DOI: 10.1016/j.cmet.2009.01.006] [Citation(s) in RCA: 150] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2008] [Revised: 10/20/2008] [Accepted: 01/14/2009] [Indexed: 10/21/2022]
Abstract
Increases in adiposity trigger metabolic and inflammatory changes that interfere with insulin action in peripheral tissues, culminating in beta cell failure and overt diabetes. We found that the cAMP Response Element Binding protein (CREB) is activated in adipose cells under obese conditions, where it promotes insulin resistance by triggering expression of the transcriptional repressor ATF3 and thereby downregulating expression of the adipokine hormone adiponectin as well as the insulin-sensitive glucose transporter 4 (GLUT4). Transgenic mice expressing a dominant-negative CREB transgene in adipocytes displayed increased whole-body insulin sensitivity in the contexts of diet-induced and genetic obesity, and they were protected from the development of hepatic steatosis and adipose tissue inflammation. These results indicate that adipocyte CREB provides an early signal in the progression to type 2 diabetes.
Collapse
Affiliation(s)
- Ling Qi
- Peptide Biology Laboratories, Salk Institute for Biological Studies
| | - Maziyar Saberi
- Division of Endocrinology and Metabolism, UCSD, La Jolla, CA 92037
| | - Erik Zmuda
- Dept. of Molecular and Cellular Biochemistry, Ohio State University, Columbus, OH 43210
| | - Yiguo Wang
- Peptide Biology Laboratories, Salk Institute for Biological Studies
| | - Judith Altarejos
- Peptide Biology Laboratories, Salk Institute for Biological Studies
| | | | - Renaud Dentin
- Peptide Biology Laboratories, Salk Institute for Biological Studies
| | - Susie Hedrick
- Peptide Biology Laboratories, Salk Institute for Biological Studies
| | | | - Tsonwin Hai
- Dept. of Molecular and Cellular Biochemistry, Ohio State University, Columbus, OH 43210
| | - Jerry Olefsky
- Division of Endocrinology and Metabolism, UCSD, La Jolla, CA 92037
| | - Marc Montminy
- Peptide Biology Laboratories, Salk Institute for Biological Studies
| |
Collapse
|
932
|
Cellular and molecular effects of n-3 polyunsaturated fatty acids on adipose tissue biology and metabolism. Clin Sci (Lond) 2009; 116:1-16. [PMID: 19037880 DOI: 10.1042/cs20070456] [Citation(s) in RCA: 211] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Adipose tissue and its secreted products, adipokines, have a major role in the development of obesity-associated metabolic derangements including Type 2 diabetes. Conversely, obesity and its metabolic sequelae may be counteracted by modulating metabolism and secretory functions of adipose tissue. LC-PUFAs (long-chain polyunsaturated fatty acids) of the n-3 series, namely DHA (docosahexaenoic acid; C(22:6n-3)) and EPA (eicosapentaenoic acid; C(20:5n-3)), exert numerous beneficial effects, such as improvements in lipid metabolism and prevention of obesity and diabetes, which partially result from the metabolic action of n-3 LC-PUFAs in adipose tissue. Recent studies highlight the importance of mitochondria in adipose tissue for the maintenance of systemic insulin sensitivity. For instance, both n-3 LC-PUFAs and the antidiabetic drugs TZDs (thiazolidinediones) induce mitochondrial biogenesis and beta-oxidation. The activation of this 'metabolic switch' in adipocytes leads to a decrease in adiposity. Both n-3 LC-PUFAs and TZDs ameliorate a low-grade inflammation of adipose tissue associated with obesity and induce changes in the pattern of secreted adipokines, resulting in improved systemic insulin sensitivity. In contrast with TZDs, which act as agonists of PPARgamma (peroxisome-proliferator-activated receptor-gamma) and promote differentiation of adipocytes and adipose tissue growth, n-3 LC-PUFAs affect fat cells by different mechanisms, including the transcription factors PPARalpha and PPARdelta. Some of the effects of n-3 LC-PUFAs on adipose tissue depend on their active metabolites, especially eicosanoids. Thus treatments affecting adipose tissue by multiple mechanisms, such as combining n-3 LC-PUFAs with either caloric restriction or antidiabetic/anti-obesity drugs, should be explored.
Collapse
|
933
|
Lucas S, Verwaerde C, Wolowczuk I. Is the Adipose Tissue the Key Road to Inflammation? ACTA ACUST UNITED AC 2009. [DOI: 10.4137/iii.s2145] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Stéphanie Lucas
- Laboratoire de NeuroImmunoEndocrinologie Institut Pasteur de Lille, BP447 and IFR 142 1, rue A. Calmette Lille, F-59019, France
| | - Claudie Verwaerde
- Laboratoire de NeuroImmunoEndocrinologie Institut Pasteur de Lille, BP447 and IFR 142 1, rue A. Calmette Lille, F-59019, France
| | - Isabelle Wolowczuk
- Laboratoire de NeuroImmunoEndocrinologie Institut Pasteur de Lille, BP447 and IFR 142 1, rue A. Calmette Lille, F-59019, France
| |
Collapse
|
934
|
Peterson SJ, Kim DH, Li M, Positano V, Vanella L, Rodella LF, Piccolomini F, Puri N, Gastaldelli A, Kusmic C, L'Abbate A, Abraham NG. The L-4F mimetic peptide prevents insulin resistance through increased levels of HO-1, pAMPK, and pAKT in obese mice. J Lipid Res 2009; 50:1293-304. [PMID: 19224872 PMCID: PMC2694329 DOI: 10.1194/jlr.m800610-jlr200] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
We examined mechanisms by which L-4F reduces obesity and diabetes in obese (ob) diabetic mice. We hypothesized that L-4F reduces adiposity via increased pAMPK, pAKT, HO-1, and increased insulin receptor phosphorylation in ob mice. Obese and lean mice were divided into five groups: lean, lean-L-4F-treated, ob, ob-L-4F-treated, and ob-L-4F-LY294002. Food intake, insulin, glucose adipocyte stem cells, pAMPK, pAKT, CB1, and insulin receptor phosphorylation were determined. Subcutaneous (SAT) and visceral adipose tissue (VAT) were determined by MRI and hepatic lipid content by magnetic resonance spectroscopy. SAT and VAT volumes decreased in ob-L-4F-treated animals compared with control. L-4F treatment decreased hepatic lipid content and increased the numbers of small adipocytes (P < 0.05) and phosphorylation of insulin receptors. L-4F decreased CB1 in SAT and VAT and increased pAKT and pAMPK in endothelium. L-4F-mediated improvement in endothelium was prevented by LY294002. Inhibition of pAKT and pAMPK by LY294002 was associated with an increase in glucose levels. Upregulation of HO-1 by L-4F produced adipose remodeling and increased the number of small differentiated adipocytes. The anti-obesity effects of L-4F are manifested by a decrease in visceral fat content with reciprocal increases in adiponectin, pAMPK, pAKT, and phosphorylation of insulin receptors with improved insulin sensitivity.
Collapse
Affiliation(s)
- Stephen J Peterson
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
935
|
Yue L, Mazzone T. Peroxisome proliferator-activated receptor {gamma} stimulation of adipocyte ApoE gene transcription mediated by the liver receptor X pathway. J Biol Chem 2009; 284:10453-61. [PMID: 19218241 DOI: 10.1074/jbc.m808482200] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Peroxisome proliferator-activated receptor (PPARgamma) agonists increase insulin sensitivity in humans and are useful for treating human diabetes. Treatment with these agonists leads to increased apoE expression and triglyceride accumulation in adipocytes. The importance of apoE for adipocyte triglyceride accumulation is demonstrated by observations that triglyceride accumulation is impaired in apoE knockout adipocytes treated with PPARgamma agonists. The current studies investigate the molecular mechanism for PPARgamma stimulation of the adipocyte apoE gene and demonstrate that the liver receptor X (LXR) response element within an apoE gene downstream enhancer is required for the apoE response to PPARgamma agonists. The response of the apoE gene to treatment with PPARgamma agonists was delayed beyond 12 h suggesting the involvement of an intermediary pathway. The combined addition of PPARgamma and LXR agonists did not increase apoE response beyond that observed with addition of either alone. Deletion or mutation of the LXR response element completely eliminated the adipocyte apoE gene response to a PPARgamma agonist. Chromatin immunoprecipitation analyses performed using isolated adipocytes, or adipose tissue from mice treated with PPARgamma agonists, showed increased LXR binding to the apoE gene after PPARgamma agonist treatment. Knockdown of LXR expression completely eliminated the increase in apoE message, protein, and triglyceride in response to PPARgamma stimulation. The LXR response element has been previously shown to mediate sterol responsiveness of the apoE gene, and apoE expression plays an important role in adipocyte triglyceride balance. The current observations suggest that the PPARgamma-LXR-apoE regulatory cascade could be an important molecular link for cross-talk between adipocyte triglyceride and cholesterol homeostasis.
Collapse
Affiliation(s)
- Lili Yue
- Departments of Medicine, Pharmacology, and Human Nutrition, University of Illinois, Chicago, Illinois 60612
| | | |
Collapse
|
936
|
McAuley P, Pittsley J, Myers J, Abella J, Froelicher VF. Fitness and Fatness as Mortality Predictors in Healthy Older Men: The Veterans Exercise Testing Study. J Gerontol A Biol Sci Med Sci 2009; 64:695-9. [DOI: 10.1093/gerona/gln039] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
937
|
Minokoshi Y, Shiuchi T, Lee S, Suzuki A, Okamoto S. Role of hypothalamic AMP-kinase in food intake regulation. Nutrition 2009; 24:786-90. [PMID: 18725075 DOI: 10.1016/j.nut.2008.06.002] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2008] [Accepted: 06/03/2008] [Indexed: 12/14/2022]
Abstract
Adenosine monophosphate-activated protein kinase (AMPK) functions as a cellular fuel gauge that regulates metabolic pathways in nutrient metabolism. Recent studies have strongly implicated that AMPK in the hypothalamus regulates energy metabolism by integrating inputs from multiple hormones, peptides, neurotransmitters, and nutrients. Leptin is an adipocyte hormone that regulates food intake and energy expenditure in peripheral tissues. Leptin inhibits AMPK activity in the arcuate and paraventricular hypothalamus, and its inhibition is necessary for the anorexic effect of leptin. Alteration of hypothalamic AMPK activity is sufficient to change food intake and body weight. Furthermore, fasting/refeeding, glucose, and melanocortin receptor alter AMPK activity in the hypothalamus. Adiponectin has also been shown to increase food intake by activating AMPK in the arcuate hypothalamus. Recent data have shown that acetyl-coenzyme A carboxylase/malonyl-coenzyme A/carnitine palmitoyltransferase-1/fatty acid oxidation and mammalian target of rapamycin signalings are putative downstream pathways for food intake regulation in response to hypothalamic AMPK. Thus, these results suggest that food intake and nutrient metabolism are coordinately regulated by the common signaling pathway of AMPK in the hypothalamus.
Collapse
Affiliation(s)
- Yasuhiko Minokoshi
- Division of Endocrinology and Metabolism, National Institute for Physiological Sciences, Aichi, Japan.
| | | | | | | | | |
Collapse
|
938
|
Schupp M, Lefterova MI, Janke J, Leitner K, Cristancho AG, Mullican SE, Qatanani M, Szwergold N, Steger DJ, Curtin JC, Kim RJ, Suh MJ, Albert MR, Engeli S, Gudas LJ, Lazar MA. Retinol saturase promotes adipogenesis and is downregulated in obesity. Proc Natl Acad Sci U S A 2009; 106:1105-10. [PMID: 19139408 PMCID: PMC2633572 DOI: 10.1073/pnas.0812065106] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2008] [Indexed: 12/19/2022] Open
Abstract
Adipocyte differentiation is controlled by many transcription factors, but few known downstream targets of these factors are necessary for adipogenesis. Here we report that retinol saturase (RetSat), which is an enzyme implicated in the generation of dihydroretinoid metabolites, is induced during adipogenesis and is directly regulated by the transcription factor peroxisome proliferator activated receptor gamma (PPARgamma). Ablation of RetSat dramatically inhibited adipogenesis but, surprisingly, this block was not overcome by the putative product of RetSat enzymatic activity. On the other hand, ectopic RetSat with an intact, but not a mutated, FAD/NAD dinucleotide-binding motif increased endogenous PPARgamma transcriptional activity and promoted adipogenesis. Indeed, RetSat was not required for adipogenesis when cells were provided with exogenous PPARgamma ligands. In adipose tissue, RetSat is expressed in adipocytes but is unexpectedly downregulated in obesity, most likely owing to infiltration of macrophages that we demonstrate to repress RetSat expression. Thiazolidinedione treatment reversed low RetSat expression in adipose tissue of obese mice. Thus, RetSat plays an important role in the biology of adipocytes, where it favors normal differentiation, yet is reduced in the obese state. RetSat is thus a novel target for therapeutic intervention in metabolic disease.
Collapse
Affiliation(s)
- Michael Schupp
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine and Department of Genetics, and The Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Martina I. Lefterova
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine and Department of Genetics, and The Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Jürgen Janke
- Franz-Volhard Clinical Research Center, Medical Faculty of the Charité and HELIOS Klinikum, 13125 Berlin, Germany
| | - Kirstin Leitner
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine and Department of Genetics, and The Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Ana G. Cristancho
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine and Department of Genetics, and The Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Shannon E. Mullican
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine and Department of Genetics, and The Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Mohammed Qatanani
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine and Department of Genetics, and The Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Nava Szwergold
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine and Department of Genetics, and The Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - David J. Steger
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine and Department of Genetics, and The Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Joshua C. Curtin
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine and Department of Genetics, and The Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Roy J. Kim
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine and Department of Genetics, and The Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
- Division of Endocrinology, Children's Hospital of Philadelphia, Philadelphia, PA 19104
| | - Moo-jin Suh
- Department of Pharmacology, Weill Cornell Medical College, New York, NY 10021; and
| | - Martin R. Albert
- Department of Pharmacology, Weill Cornell Medical College, New York, NY 10021; and
| | - Stefan Engeli
- Institute of Clinical Pharmacology, Medical School of Hannover, 30625 Hannover, Germany
| | - Lorraine J. Gudas
- Department of Pharmacology, Weill Cornell Medical College, New York, NY 10021; and
| | - Mitchell A. Lazar
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine and Department of Genetics, and The Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| |
Collapse
|
939
|
Christodoulides C, Lagathu C, Sethi JK, Vidal-Puig A. Adipogenesis and WNT signalling. Trends Endocrinol Metab 2009; 20:16-24. [PMID: 19008118 PMCID: PMC4304002 DOI: 10.1016/j.tem.2008.09.002] [Citation(s) in RCA: 449] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2008] [Revised: 09/22/2008] [Accepted: 09/22/2008] [Indexed: 12/21/2022]
Abstract
An inability of adipose tissue to expand consequent to exhausted capacity to recruit new adipocytes might underlie the association between obesity and insulin resistance. Adipocytes arise from mesenchymal precursors whose commitment and differentiation along the adipocytic lineage is tightly regulated. These regulatory factors mediate cross-talk between adipose cells, ensuring that adipocyte growth and differentiation are coupled to energy storage demands. The WNT family of autocrine and paracrine growth factors regulates adult tissue maintenance and remodelling and, consequently, is well suited to mediate adipose cell communication. Indeed, several recent reports, summarized in this review, implicate WNT signalling in regulating adipogenesis. Manipulating the WNT pathway to alter adipose cellular makeup, therefore, constitutes an attractive drug-development target to combat obesity-associated metabolic complications.
Collapse
Affiliation(s)
- Constantinos Christodoulides
- Institute of Metabolic Science, MRC Centre for Obesity and Associated Diseases, Biochemistry, University of Cambridge, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK.
| | | | | | | |
Collapse
|
940
|
Abstract
Adiponectin, an adipokine secreted by the white adipose tissue, plays an important role in regulating glucose and lipid metabolism and controlling energy homeostasis in insulin-sensitive tissues. A decrease in the circulating level of adiponectin has been linked to insulin resistance, type 2 diabetes, atherosclerosis, and metabolic syndrome. Adiponectin exerts its effects through two membrane receptors, AdipoR1 and AdipoR2. APPL1 is the first identified protein that interacts directly with adiponectin receptors. APPL1 is an adaptor protein with multiple functional domains, the Bin1/amphiphysin/rvs167, pleckstrin homology, and phosphotyrosine binding domains. The PTB domain of APPL1 interacts directly with the intracellular region of adiponectin receptors. Through this interaction, APPL1 mediates adiponectin signaling and its effects on metabolism. APPL1 also functions in insulin-signaling pathway and is an important mediator of adiponectin-dependent insulin sensitization in skeletal muscle. Adiponectin signaling through APPL1 is necessary to exert its anti-inflammatory and cytoprotective effects on endothelial cells. APPL1 also acts as a mediator of other signaling pathways by interacting directly with membrane receptors or signaling proteins, thereby playing critical roles in cell proliferation, apoptosis, cell survival, endosomal trafficking, and chromatin remodeling. This review focuses mainly on our current understanding of adiponectin signaling in various tissues, the role of APPL1 in mediating adiponectin signaling, and also its role in the cross-talk between adiponectin/insulin-signaling pathways.
Collapse
Affiliation(s)
- Sathyaseelan S Deepa
- Dept. of Cellular & Structural Biology, Univ. of Texas Health Science Ctr., 7703 Floyd Curl Dr., San Antonio, TX 78229, USA
| | | |
Collapse
|
941
|
Kennedy A, Martinez K, Chuang CC, LaPoint K, McIntosh M. Saturated fatty acid-mediated inflammation and insulin resistance in adipose tissue: mechanisms of action and implications. J Nutr 2009; 139:1-4. [PMID: 19056664 DOI: 10.3945/jn.108.098269] [Citation(s) in RCA: 348] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
This review highlights the inflammatory and insulin-antagonizing effects of saturated fatty acids (SFA), which contribute to the development of metabolic syndrome. Mechanisms responsible for these unhealthy effects of SFA include: 1) accumulation of diacylglycerol and ceramide; 2) activation of nuclear factor-kappaB, protein kinase C-, and mitogen-activated protein kinases, and subsequent induction of inflammatory genes in white adipose tissue, immune cells, and myotubes; 3) decreased PPARgamma coactivator-1 alpha/beta activation and adiponectin production, which decreases the oxidation of glucose and fatty acids (FA); and 4) recruitment of immune cells like macrophages, neutrophils, and bone marrow-derived dendritic cells to WAT and muscle. Several studies have demonstrated potential health benefits of substituting SFA with unsaturated FA, particularly oleic acid and (n-3) FA. Thus, reducing consumption of foods rich in SFA and increasing consumption of whole grains, fruits, vegetables, lean meats and poultry, fish, low-fat dairy products, and oils containing oleic acid or (n-3) FA is likely to reduce the incidence of metabolic disease.
Collapse
Affiliation(s)
- Arion Kennedy
- Department of Nutrition, University of North Carolina at Greensboro, Greensboro, NC 27402, USA
| | | | | | | | | |
Collapse
|
942
|
Abstract
Adiponectin is one of the most effective adipokines in the context of correcting obesity-induced insulin resistance. However, adiponectin-deficient animal models show a relatively modest phenotype unless metabolically challenged. This suggests that potent compensatory mechanisms are in place. In this issue of the Biochemical Journal, Wong et al. characterize new members of the CTRPs [C1q-TNFalpha (tumour necrosis factor alpha)-related proteins]. They establish that some CTRPs are produced primarily in the stromal vascular fraction of adipose tissue, and that expression of CRTP1, in particular (like adiponectin), is induced by PPARgamma (peroxisome-proliferator-activated receptor gamma) agonists. Moreover, injection of recombinant CTRP1 displays glucose-lowering effects. These observations suggest that CTRP1 may have partially overlapping functions and, along with other paralogues, may effectively compensate for the chronic loss of adiponectin function.
Collapse
|
943
|
Scaffold-based discovery of indeglitazar, a PPAR pan-active anti-diabetic agent. Proc Natl Acad Sci U S A 2008; 106:262-7. [PMID: 19116277 DOI: 10.1073/pnas.0811325106] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
In a search for more effective anti-diabetic treatment, we used a process coupling low-affinity biochemical screening with high-throughput co-crystallography in the design of a series of compounds that selectively modulate the activities of all three peroxisome proliferator-activated receptors (PPARs), PPARalpha, PPARgamma, and PPARdelta. Transcriptional transactivation assays were used to select compounds from this chemical series with a bias toward partial agonism toward PPARgamma, to circumvent the clinically observed side effects of full PPARgamma agonists. Co-crystallographic characterization of the lead molecule, indeglitazar, in complex with each of the 3 PPARs revealed the structural basis for its PPAR pan-activity and its partial agonistic response toward PPARgamma. Compared with full PPARgamma-agonists, indeglitazar is less potent in promoting adipocyte differentiation and only partially effective in stimulating adiponectin gene expression. Evaluation of the compound in vivo confirmed the reduced adiponectin response in animal models of obesity and diabetes while revealing strong beneficial effects on glucose, triglycerides, cholesterol, body weight, and other metabolic parameters. Indeglitazar has now progressed to Phase II clinical evaluations for Type 2 diabetes mellitus (T2DM).
Collapse
|
944
|
Abstract
Adipocytes are embedded in a unique extracellular matrix whose main function is to provide mechanical support, in addition to participating in a variety of signaling events. During adipose tissue expansion, the extracellular matrix requires remodeling to accommodate adipocyte growth. Here, we demonstrate a general upregulation of several extracellular matrix components in adipose tissue in the diabetic state, therefore implicating "adipose tissue fibrosis" as a hallmark of metabolically challenged adipocytes. Collagen VI is a highly enriched extracellular matrix component of adipose tissue. The absence of collagen VI results in the uninhibited expansion of individual adipocytes and is paradoxically associated with substantial improvements in whole-body energy homeostasis, both with high-fat diet exposure and in the ob/ob background. Collectively, our data suggest that weakening the extracellular scaffold of adipocytes enables their stress-free expansion during states of positive energy balance, which is consequently associated with an improved inflammatory profile. Therefore, the disproportionate accumulation of extracellular matrix components in adipose tissue may not be merely an epiphenomenon of metabolically challenging conditions but may also directly contribute to a failure to expand adipose tissue mass during states of excess caloric intake.
Collapse
|
945
|
Poussin C, Hall D, Minehira K, Galzin AM, Tarussio D, Thorens B. Different transcriptional control of metabolism and extracellular matrix in visceral and subcutaneous fat of obese and rimonabant treated mice. PLoS One 2008; 3:e3385. [PMID: 19030233 PMCID: PMC2586343 DOI: 10.1371/journal.pone.0003385] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2008] [Accepted: 09/16/2008] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND The visceral (VAT) and subcutaneous (SCAT) adipose tissues play different roles in physiology and obesity. The molecular mechanisms underlying their expansion in obesity and following body weight reduction are poorly defined. METHODOLOGY C57Bl/6 mice fed a high fat diet (HFD) for 6 months developed low, medium, or high body weight as compared to normal chow fed mice. Mice from each groups were then treated with the cannabinoid receptor 1 antagonist rimonabant or vehicle for 24 days to normalize their body weight. Transcriptomic data for visceral and subcutaneous adipose tissues from each group of mice were obtained and analyzed to identify: i) genes regulated by HFD irrespective of body weight, ii) genes whose expression correlated with body weight, iii) the biological processes activated in each tissue using gene set enrichment analysis (GSEA), iv) the transcriptional programs affected by rimonabant. PRINCIPAL FINDINGS In VAT, "metabolic" genes encoding enzymes for lipid and steroid biosynthesis and glucose catabolism were down-regulated irrespective of body weight whereas "structure" genes controlling cell architecture and tissue remodeling had expression levels correlated with body weight. In SCAT, the identified "metabolic" and "structure" genes were mostly different from those identified in VAT and were regulated irrespective of body weight. GSEA indicated active adipogenesis in both tissues but a more prominent involvement of tissue stroma in VAT than in SCAT. Rimonabant treatment normalized most gene expression but further reduced oxidative phosphorylation gene expression in SCAT but not in VAT. CONCLUSION VAT and SCAT show strikingly different gene expression programs in response to high fat diet and rimonabant treatment. Our results may lead to identification of therapeutic targets acting on specific fat depots to control obesity.
Collapse
Affiliation(s)
- Carine Poussin
- Center for Integrative Genomics and Department of Physiology, University of Lausanne, Lausanna, Switzerland
| | - Diana Hall
- Center for Integrative Genomics and Department of Physiology, University of Lausanne, Lausanna, Switzerland
| | - Kaori Minehira
- Center for Integrative Genomics and Department of Physiology, University of Lausanne, Lausanna, Switzerland
| | | | - David Tarussio
- Center for Integrative Genomics and Department of Physiology, University of Lausanne, Lausanna, Switzerland
| | - Bernard Thorens
- Center for Integrative Genomics and Department of Physiology, University of Lausanne, Lausanna, Switzerland
- * E-mail:
| |
Collapse
|
946
|
Singh SP, Niemczyk M, Zimniak L, Zimniak P. Fat accumulation in Caenorhabditis elegans triggered by the electrophilic lipid peroxidation product 4-hydroxynonenal (4-HNE). Aging (Albany NY) 2008; 1:68-80. [PMID: 20157589 PMCID: PMC2815766 DOI: 10.18632/aging.100005] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2008] [Accepted: 12/12/2008] [Indexed: 04/29/2023]
Abstract
Deposition and mobilization of fat in an organism are tightly controlled by multiple levels of endocrine and neuroendocrine regulation. Because these hormonal mechanisms ultimately act by affecting biochemical reactions of fat synthesis or utilization, obesity could be also modulated by altering directly the underlying lipid biochemistry. We have previously shown that genetically modified mice with an elevated level of the lipid peroxidation product 4-HNE become obese. We now demonstrate that the process is phylogenetically conserved and thus likely to be universal. In the nematode C. elegans, disruption of either conjugation or oxidation of 4-HNE leads to fat accumulation, whereas augmentation of 4-HNE conjugation results in a lean phenotype. Moreover, direct treatment of C. elegans with synthetic 4-HNE causes increased lipid storage, directly demonstrating a causative role of 4-HNE. The postulated mechanism, which involves modulation of acetyl-CoA carboxylase activity, could contribute to the triggering and maintenance of the obese phenotype on a purely metabolic level.
Collapse
Affiliation(s)
- Sharda P. Singh
- Department of Pharmacology and
Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Maciej Niemczyk
- Department of Pharmacology and
Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Ludwika Zimniak
- Department of Pharmacology and
Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Piotr Zimniak
- Department of Pharmacology and
Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Central Arkansas Veterans
Healthcare System, Little Rock, AR 72205, USA
| |
Collapse
|
947
|
Kumar KG, Trevaskis JL, Lam DD, Sutton GM, Koza RA, Chouljenko VN, Kousoulas KG, Rogers PM, Kesterson RA, Thearle M, Ferrante AW, Mynatt RL, Burris TP, Dong JZ, Halem HA, Culler MD, Heisler LK, Stephens JM, Butler AA. Identification of adropin as a secreted factor linking dietary macronutrient intake with energy homeostasis and lipid metabolism. Cell Metab 2008; 8:468-81. [PMID: 19041763 PMCID: PMC2746325 DOI: 10.1016/j.cmet.2008.10.011] [Citation(s) in RCA: 344] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2007] [Revised: 04/23/2008] [Accepted: 10/29/2008] [Indexed: 12/15/2022]
Abstract
Obesity and nutrient homeostasis are linked by mechanisms that are not fully elucidated. Here we describe a secreted protein, adropin, encoded by a gene, Energy Homeostasis Associated (Enho), expressed in liver and brain. Liver Enho expression is regulated by nutrition: lean C57BL/6J mice fed high-fat diet (HFD) exhibited a rapid increase, while fasting reduced expression compared to controls. However, liver Enho expression declines with diet-induced obesity (DIO) associated with 3 months of HFD or with genetically induced obesity, suggesting an association with metabolic disorders in the obese state. In DIO mice, transgenic overexpression or systemic adropin treatment attenuated hepatosteatosis and insulin resistance independently of effects on adiposity or food intake. Adropin regulated expression of hepatic lipogenic genes and adipose tissue peroxisome proliferator-activated receptor gamma, a major regulator of lipogenesis. Adropin may therefore be a factor governing glucose and lipid homeostasis, which protects against hepatosteatosis and hyperinsulinemia associated with obesity.
Collapse
MESH Headings
- Adipose Tissue, Brown/metabolism
- Adipose Tissue, White/metabolism
- Amino Acid Sequence
- Animals
- Base Sequence
- Benzoates/chemistry
- Benzoates/metabolism
- Benzylamines/chemistry
- Benzylamines/metabolism
- Blood Proteins/genetics
- Blood Proteins/metabolism
- Blood Proteins/physiology
- Cells, Cultured
- DNA-Binding Proteins/agonists
- DNA-Binding Proteins/metabolism
- Energy Metabolism
- Fasting
- Fatty Liver/metabolism
- Female
- Humans
- Intercellular Signaling Peptides and Proteins
- Leptin/metabolism
- Lipid Metabolism
- Liver X Receptors
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Molecular Sequence Data
- Obesity/genetics
- Obesity/metabolism
- Orphan Nuclear Receptors
- Peptides
- Proteins/genetics
- Proteins/metabolism
- Proteins/physiology
- RNA Interference
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Receptors, Cytoplasmic and Nuclear/agonists
- Receptors, Cytoplasmic and Nuclear/metabolism
Collapse
Affiliation(s)
- K Ganesh Kumar
- Neuropeptides Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
948
|
Tilg H, Moschen AR. Insulin resistance, inflammation, and non-alcoholic fatty liver disease. Trends Endocrinol Metab 2008; 19:371-9. [PMID: 18929493 DOI: 10.1016/j.tem.2008.08.005] [Citation(s) in RCA: 368] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2008] [Revised: 08/27/2008] [Accepted: 08/28/2008] [Indexed: 02/07/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD), the major cause of abnormal liver function in the western world, is often associated with obesity and diabetes. In obese individuals, fat accumulation in the abdominal region affects both lipid and glucose metabolism, and a liver loaded with fat is insulin resistant. Insulin resistance (IR) is often associated with chronic low-grade inflammation, and numerous mediators released from immune cells and adipocytes contribute to development of IR. Recent results showing an important role for these mediators in NAFLD are providing us with a better understanding of this highly prevalent disease with implications for novel therapy development. This review highlights new aspects in development of liver steatosis and the relevance of various cytokines and adipocytokines in NAFLD.
Collapse
Affiliation(s)
- Herbert Tilg
- Department of Medicine, Christian Doppler Research Laboratory for Gut Inflammation, Medical University Innsbruck, Austria.
| | | |
Collapse
|
949
|
Perlemuter G, Naveau S, Belle-Croix F, Buffet C, Agostini H, Laromiguière M, Cassard-Doulcier AM, Oppert JM. Independent and opposite associations of trunk fat and leg fat with liver enzyme levels. Liver Int 2008; 28:1381-8. [PMID: 18482272 DOI: 10.1111/j.1478-3231.2008.01764.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND In contrast to trunk fat mass (TFM), which is associated with cardiovascular risk markers, leg fat mass (LFM) displays independent protective effects against atherosclerosis. Little is known about the respective influence of central and peripheral adiposity on liver enzyme levels. AIMS To assess the respective influence of TFM and LFM on alanine aminotransferase (ALT), aspartate aminotransferase (AST) and gamma-glutamyltransferase (GGT) levels, and to test whether LFM might protect against an increase of liver enzyme levels. METHODS Cross-sectional study on 1442 patients (women: 1155; men: 287) referred for overweight/obesity over 3 years. Body composition was analysed by dual-energy X-ray absorptiometry. The relationships among liver enzymes, age, weight, height, body mass index (BMI), biological indices and body composition were studied. RESULTS The mean BMI was 39.7 +/- 7.9 kg/m(2) in women and 38.2 +/- 6.6 kg/m(2) in men. In women, after adjustement for confounding factors, ALT, AST and GGT were negatively and independently correlated with LFM and positively with TFM. Similar independent associations were observed for ALT and AST in men. The strongest associations were found for ALT in both women and men. CONCLUSIONS As observed for cardiovascular risk factors, LFM and TFM are inversely and independently correlated with liver enzyme levels in obese patients. LFM may confer independent protective effects against obesity-associated liver damage.
Collapse
Affiliation(s)
- Gabriel Perlemuter
- AP-HP, Hôpital Antoine Béclère, Service d'hépato-gastroentérologie, Clamart, France.
| | | | | | | | | | | | | | | |
Collapse
|
950
|
Ledoux S, Queguiner I, Msika S, Calderari S, Rufat P, Gasc JM, Corvol P, Larger E. Angiogenesis associated with visceral and subcutaneous adipose tissue in severe human obesity. Diabetes 2008; 57:3247-57. [PMID: 18835936 PMCID: PMC2584130 DOI: 10.2337/db07-1812] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2007] [Accepted: 09/16/2008] [Indexed: 01/01/2023]
Abstract
OBJECTIVE The expansion of adipose tissue is linked to the development of its vasculature. However, the regulation of adipose tissue angiogenesis in humans has not been extensively studied. Our aim was to compare the angiogenesis associated with subcutaneous adipose tissue (SAT) and visceral adipose tissue (VAT) from the same obese patients in an in vivo model. RESEARCH DESIGN AND METHODS Adipose tissue samples from visceral (VAT) and subcutaneous (SAT) sites, obtained from 36 obese patients (mean BMI 46.5 kg/m(2)) during bariatric surgery, were layered on chick chorioallantoïc membrane (CAM). RESULTS Both SAT and VAT expressed angiogenic factors without significant difference for vascular endothelial growth factor (VEGF) expression. Adipose tissue layered on CAM stimulated angiogenesis. Angiogenic stimulation was macroscopically detectable, with engulfment of the samples, in 39% and was evidenced by angiography in 59% of the samples. A connection between CAM and adipose tissue vessels was evidenced by immunohistochemistry, with recruitment of both avian and human endothelial cells. The angiogenic potency of adipose tissue was not related to its localization (with an angiogenic stimulation in 60% of SAT samples and 61% of VAT samples) or to adipocyte size or inflammatory infiltrate assessed in adipose samples before the graft on CAM. Stimulation of angiogenesis by adipose tissue was nearly abolished by bevacizumab, which specifically targets human VEGF. CONCLUSIONS We have established a model to study the regulation of angiogenesis by human adipose tissue. This model highlighted the role of VEGF in angiogenesis in both SAT and VAT.
Collapse
Affiliation(s)
- Séverine Ledoux
- Institut National de la Santé et de la Recherche Médicale U833, Chaire de Médecine Expérimentale, Collège de France, Paris, France.
| | | | | | | | | | | | | | | |
Collapse
|