101
|
Kang C, Yijun L, Jingtao D, Changyu P, Wenhua Y, Baoan W, Fangling M, Xianling W, Guoqing Y, Yiming M, Juming L. Effects of telmisartan on lipid metabolisms and proinflammatory factors secretion of differentiated 3T3-L1 adipocytes. J Renin Angiotensin Aldosterone Syst 2014; 16:1061-8. [PMID: 24591527 DOI: 10.1177/1470320313518252] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Affiliation(s)
- Chen Kang
- Department of Endocrinology, PLA General Hospital, Beijing, China
| | - Li Yijun
- Department of Endocrinology, PLA General Hospital, Beijing, China
| | - Dou Jingtao
- Department of Endocrinology, PLA General Hospital, Beijing, China
| | - Pan Changyu
- Department of Endocrinology, PLA General Hospital, Beijing, China
| | - Yan Wenhua
- Department of Endocrinology, PLA General Hospital, Beijing, China
| | - Wang Baoan
- Department of Endocrinology, PLA General Hospital, Beijing, China
| | - Ma Fangling
- Department of Endocrinology, PLA General Hospital, Beijing, China
| | - Wang Xianling
- Department of Endocrinology, PLA General Hospital, Beijing, China
| | - Yang Guoqing
- Department of Endocrinology, PLA General Hospital, Beijing, China
| | - Mu Yiming
- Department of Endocrinology, PLA General Hospital, Beijing, China
| | - Lu Juming
- Department of Endocrinology, PLA General Hospital, Beijing, China
| |
Collapse
|
102
|
Sanchez-Gurmaches J, Guertin DA. Adipocyte lineages: tracing back the origins of fat. BIOCHIMICA ET BIOPHYSICA ACTA 2014; 1842:340-51. [PMID: 23747579 PMCID: PMC3805734 DOI: 10.1016/j.bbadis.2013.05.027] [Citation(s) in RCA: 178] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Revised: 05/22/2013] [Accepted: 05/24/2013] [Indexed: 12/25/2022]
Abstract
The obesity epidemic has intensified efforts to understand the mechanisms controlling adipose tissue development. Adipose tissue is generally classified as white adipose tissue (WAT), the major energy storing tissue, or brown adipose tissue (BAT), which mediates non-shivering thermogenesis. It is hypothesized that brite adipocytes (brown in white) may represent a third adipocyte class. The recent realization that brown fat exist in adult humans suggests increasing brown fat energy expenditure could be a therapeutic strategy to combat obesity. To understand adipose tissue development, several groups are tracing the origins of mature adipocytes back to their adult precursor and embryonic ancestors. From these studies emerged a model that brown adipocytes originate from a precursor shared with skeletal muscle that expresses Myf5-Cre, while all white adipocytes originate from a Myf5-negative precursors. While this provided a rational explanation to why BAT is more metabolically favorable than WAT, recent work indicates the situation is more complex because subsets of white adipocytes also arise from Myf5-Cre expressing precursors. Lineage tracing studies further suggest that the vasculature may provide a niche supporting both brown and white adipocyte progenitors; however, the identity of the adipocyte progenitor cell is under debate. Differences in origin between adipocytes could explain metabolic heterogeneity between depots and/or influence body fat patterning particularly in lipodystrophy disorders. Here, we discuss recent insights into adipose tissue origins highlighting lineage-tracing studies in mice, how variations in metabolism or signaling between lineages could affect body fat distribution, and the questions that remain unresolved. This article is part of a Special Issue entitled: Modulation of Adipose Tissue in Health and Disease.
Collapse
Affiliation(s)
- Joan Sanchez-Gurmaches
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - David A Guertin
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
103
|
Masuda T, Fu Y, Eguchi A, Czogalla J, Rose MA, Kuczkowski A, Gerasimova M, Feldstein AE, Scadeng M, Vallon V. Dipeptidyl peptidase IV inhibitor lowers PPARγ agonist-induced body weight gain by affecting food intake, fat mass, and beige/brown fat but not fluid retention. Am J Physiol Endocrinol Metab 2014; 306:E388-98. [PMID: 24347054 PMCID: PMC3923087 DOI: 10.1152/ajpendo.00124.2013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Peroxisome proliferator-activated receptor-γ (PPARγ) agonists like pioglitazone (PGZ) are effective antidiabetic drugs, but they induce fluid retention and body weight (BW) gain. Dipeptidyl peptidase IV (DPP IV) inhibitors are antidiabetic drugs that enhance renal Na(+) and fluid excretion. Therefore, we examined whether the DPP IV inhibitor alogliptin (ALG) ameliorates PGZ-induced BW gain. Male Sv129 mice were treated with vehicle (repelleted diet), PGZ (220 mg/kg diet), ALG (300 mg/kg diet), or a combination of PGZ and ALG (PGZ + ALG) for 14 days. PGZ + ALG prevented the increase in BW observed with PGZ but did not attenuate the increase in body fluid content determined by bioimpedance spectroscopy (BIS). BIS revealed that ALG alone had no effect on fat mass (FM) but enhanced the FM-lowering effect of PGZ; MRI analysis confirmed the latter and showed reductions in visceral and inguinal subcutaneous (sc) white adipose tissue (WAT). ALG but not PGZ decreased food intake and plasma free fatty acid concentrations. Conversely, PGZ but not ALG increased mRNA expression of thermogenesis mediator uncoupling protein 1 in epididymal WAT. Adding ALG to PGZ treatment increased the abundance of multilocular cell islets in sc WAT, and PGZ + ALG increased the expression of brown-fat-like "beige" cell marker TMEM26 in sc WAT and interscapular brown adipose tissue and increased rectal temperature vs. vehicle. In summary, DPP IV inhibition did not attenuate PPARγ agonist-induced fluid retention but prevented BW gain by reducing FM. This involved ALG inhibition of food intake and was associated with food intake-independent synergistic effects of PPARγ agonism and DPP-IV inhibition on beige/brown fat cells and thermogenesis.
Collapse
|
104
|
Abstract
The obesogen hypothesis postulates the role of environmental chemical pollutants that disrupt homeostatic controls and adaptive mechanisms to promote adipose-dependent weight gain leading to obesity and metabolic syndrome complications. One of the most direct molecular mechanisms for coupling environmental chemical exposures to perturbed physiology invokes pollutants mimicking endogenous endocrine hormones or bioactive dietary signaling metabolites that serve as nuclear receptor ligands. The organotin pollutant tributyltin can exert toxicity through multiple mechanisms but most recently has been shown to bind, activate, and mediate RXR-PPARγ transcriptional regulation central to lipid metabolism and adipocyte biology. Data in support of long-term obesogenic effects on whole body adipose tissue are also reported. Organotins represent an important model test system for evaluating the impact and epidemiological significance of chemical insults as contributing factors for obesity and human metabolic health.
Collapse
Affiliation(s)
- Felix Grün
- The Center for Complex Biological Systems, University of California Irvine, Irvine, California, USA.
| |
Collapse
|
105
|
Nomaguchi K, Tanaka M, Misawa E, Yamada M, Toida T, Iwatsuki K, Goto T, Kawada T. Aloe vera phytosterols act as ligands for PPAR and improve the expression levels of PPAR target genes in the livers of mice with diet-induced obesity. Obes Res Clin Pract 2013; 5:e190–e201. [PMID: 24331101 DOI: 10.1016/j.orcp.2011.01.002] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2010] [Revised: 01/14/2011] [Accepted: 01/18/2011] [Indexed: 12/14/2022]
Abstract
SUMMARY Lophenol (Lo) and cycloartanol (Cy), minor phytosterols of Aloe vera gel, were previously identified as anti-diabetic compounds, and these compounds also reduced body fat in a type 2 diabetic model animal. In this study, we investigated the effects of Lo and Cy on peroxisome proliferator activated receptors (PPAR) using a luciferase reporter assay. DNA microarray and real-time quantitative RT-PCR (qPCR) analyses were also performed in a diet-induced obesity (DIO) mouse model. The Aloe phytosterols activated PPAR in a dose-dependent manner. The expression levels of many PPAR target genes were changed in the Aloe phytosterol group compared with those in the control high-fat diet (HFD) group. In particular, the expression levels of Fatp1, Acox1, Cpt1, and Hmgcs2 were significantly increased in the Aloe phytosterol group compared with those in the control HFD group; however, the expression level of ApoCIII was significantly decreased in the Aloe phytosterol group. We confirmed that Aloe phytosterols activate PPAR transcription in vitro. In addition, quantitative gene expression analysis in DIO mice suggested that Aloe phytosterols improve fatty acid metabolism in the liver.:
Collapse
Affiliation(s)
- Kouji Nomaguchi
- Functional Food Research Department, Food Science & Technology Institute, Morinaga Milk Industry Co., Ltd., Zama, Japan.
| | - Miyuki Tanaka
- Functional Food Research Department, Food Science & Technology Institute, Morinaga Milk Industry Co., Ltd., Zama, Japan
| | - Eriko Misawa
- Functional Food Research Department, Food Science & Technology Institute, Morinaga Milk Industry Co., Ltd., Zama, Japan
| | - Muneo Yamada
- Functional Food Research Department, Food Science & Technology Institute, Morinaga Milk Industry Co., Ltd., Zama, Japan
| | - Tomohiro Toida
- Functional Food Research Department, Food Science & Technology Institute, Morinaga Milk Industry Co., Ltd., Zama, Japan
| | - Keiji Iwatsuki
- Functional Food Research Department, Food Science & Technology Institute, Morinaga Milk Industry Co., Ltd., Zama, Japan
| | - Tsuyoshi Goto
- Laboratory of Molecular Function of Food, Division of Food Sciences and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Teruo Kawada
- Laboratory of Molecular Function of Food, Division of Food Sciences and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| |
Collapse
|
106
|
Nemanich S, Rani S, Shoghi K. In vivo multi-tissue efficacy of peroxisome proliferator-activated receptor-γ therapy on glucose and fatty acid metabolism in obese type 2 diabetic rats. Obesity (Silver Spring) 2013; 21:2522-9. [PMID: 23512563 PMCID: PMC3695080 DOI: 10.1002/oby.20378] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Accepted: 01/07/2013] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To identify the disturbances in glucose and lipid metabolism observed in type 2 diabetes mellitus, we examined the interaction and contribution of multiple tissues (liver, heart, muscle, and brown adipose tissue) and monitored the effects of the Peroxisome Proliferator-Activated Receptor-γ (PPARγ) agonist rosiglitazone (RGZ) on metabolism in these tissues. DESIGN AND METHODS Rates of [(18) F]fluorodeoxyglucose ([(18) F]FDG) and [(11) C]Palmitate uptake and utilization in the Zucker diabetic fatty (ZDF) rat were quantified using noninvasive positron emission tomography imaging and quantitative modeling in comparison to lean Zucker rats. Furthermore, we studied two separate groups of RGZ-treated and untreated ZDF rats. RESULTS Glucose uptake is impaired in ZDF brown fat, muscle, and heart tissues compared to leans, while RGZ treatment increased glucose uptake compared to untreated ZDF rats. Fatty acid (FA) uptake decreased, but FA flux increased in brown fat and skeletal muscle of ZDF rats. RGZ treatment increased uptake of FA in brown fat but decreased uptake and utilization in liver, muscle, and heart. CONCLUSION Our data indicate tissue-specific mechanisms for glucose and FA disposal as well as differential action of insulin-sensitizing drugs to normalize substrate handling and highlight the role that preclinical imaging may play in screening drugs for obesity and diabetes.
Collapse
Affiliation(s)
- Samuel Nemanich
- Department of Radiology, Washington University in St. Louis, Saint Louis, MO
| | - Sudheer Rani
- Department of Radiology, Washington University in St. Louis, Saint Louis, MO
| | - Kooresh Shoghi
- Department of Radiology, Washington University in St. Louis, Saint Louis, MO
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO
- Division of Biology and Biomedical Sciences, Washington University in St. Louis, Saint Louis, MO
| |
Collapse
|
107
|
Abstract
Adipose tissue is formed at stereotypic times and locations in a diverse array of organisms. Once formed, the tissue is dynamic, responding to homeostatic and external cues and capable of a 15-fold expansion. The formation and maintenance of adipose tissue is essential to many biological processes and when perturbed leads to significant diseases. Despite this basic and clinical significance, understanding of the developmental biology of adipose tissue has languished. In this Review, we highlight recent efforts to unveil adipose developmental cues, adipose stem cell biology and the regulators of adipose tissue homeostasis and dynamism.
Collapse
Affiliation(s)
- Daniel C Berry
- Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | | | | | | |
Collapse
|
108
|
Bak EJ, Kim J, Jang S, Woo GH, Yoon HG, Yoo YJ, Cha JH. Gallic acid improves glucose tolerance and triglyceride concentration in diet-induced obesity mice. Scandinavian Journal of Clinical and Laboratory Investigation 2013; 73:607-14. [DOI: 10.3109/00365513.2013.831470] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
109
|
Cignarelli A, Giorgino F, Vettor R. Pharmacologic agents for type 2 diabetes therapy and regulation of adipogenesis. Arch Physiol Biochem 2013; 119:139-50. [PMID: 23724947 DOI: 10.3109/13813455.2013.796996] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The close link between type 2 diabetes and excess body weight highlights the need to consider the effects on weight of different treatments used for correction of hyperglycaemia. Indeed, specific currently available diabetes therapies can cause weight gain, including insulin and its analogues, sulphonylureas, and thiazolidinediones, while others, such as metformin and the GLP-1 receptor agonists, can promote weight loss. Excess body weight in patients with diabetes is largely due to expansion of adipose tissue, and these drugs could interfere with the mechanisms underlying the expansion and differentiation of adipocyte precursors. Almost all anti-diabetes drugs could also potentially affect adipocyte metabolism directly, by modulating lipogenesis, lipolysis, and fat oxidation. This review will examine the available evidence for specific effects of various anti-diabetes drugs on adipose tissue development and function with the ultimate goal of increasing our understanding of how pharmacological agents can modulate energy balance and body fat.
Collapse
Affiliation(s)
- A Cignarelli
- Department of Emergency and Organ Transplantation, Section of Internal Medicine, Endocrinology, Andrology, and Metabolic Diseases, University of Bari "Aldo Moro" , Bari , Italy and
| | | | | |
Collapse
|
110
|
Yang MH, Vasquez Y, Ali Z, Khan IA, Khan SI. Constituents from Terminalia species increase PPARα and PPARγ levels and stimulate glucose uptake without enhancing adipocyte differentiation. JOURNAL OF ETHNOPHARMACOLOGY 2013; 149:490-498. [PMID: 23850833 DOI: 10.1016/j.jep.2013.07.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Revised: 06/19/2013] [Accepted: 07/04/2013] [Indexed: 06/02/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The fruits of Terminalia bellerica Roxb. (Combretaceae) and T. chebula Retz. (Combretaceae) are important components of triphala, a popular Ayurvedic formulation, for treating diabetes in Indian traditional medicine. AIM OF THE STUDY The aim of this study was to evaluate the effects of the constituents of T. bellerica and T. chebula fruit extracts on PPARα and PPARγ signaling/expression, cellular glucose uptake and adipogenesis. MATERIALS AND METHODS PPARα and PPARγ signaling and expression (luciferase assay and western blot) and the insulin-stimulated uptake of 2-NBDG were determined in HepG2 cells. The effects on adipogenesis were determined in 3T3-L1 cells by Oil red O staining and measurement of lipid content by AdipoRed reagent. RESULTS Out of the 20 compounds, two ellagitannins, chebulagic acid (1) and corilagin (2), and three gallotannins, 2,3,6-tri-O-galloyl-β-D-glucose (3), 1,2,3,6-tetra-O-galloyl-β-D-glucose (4), and 1,2,3,4,6-penta-O-galloyl-β-D-glucose (5), showed the enhancement of PPARα and/or PPARγ signaling. Two of the gallotannins (4 and 5) also increased PPARα and PPARγ protein expression, while all three (3-5) enhanced insulin-stimulated glucose uptake into HepG2 cells. Compound 1,2,3,6-tetra-O-galloyl-β-D-glucose (4) was the most potent in increasing cellular glucose uptake (9.92-fold increase at 50 μM). In the test for adipogenesis, 3-5 did not enhance the differentiation of 3T3-L1 preadipocytes but inhibited the adipogenic effect of rosiglitazone. CONCLUSION Three gallotannins (3-5) from Terminalia fruits acting as enhancers of both PPARα and PPARγ signaling increased insulin-stimulated glucose uptake without inducing the adipogenesis, with 1,2,3,6-tetra-O-galloyl-β-D-glucose (4) being the most effective in stimulating glucose uptake and 1,2,3,4,6-penta-O-galloyl-β-D-glucose (5) being most effective in increasing PPAR protein expression.
Collapse
Affiliation(s)
- Min Hye Yang
- National Center for Natural Products Research, Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, MS 38677, USA
| | | | | | | | | |
Collapse
|
111
|
Valsamakis G, Lois K, Kumar S, Mastorakos G. Metabolic and other effects of pioglitazone as an add-on therapy to metformin in the treatment of polycystic ovary syndrome (PCOS). Hormones (Athens) 2013; 12:363-78. [PMID: 24121378 DOI: 10.1007/bf03401302] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Insulin resistance is a key pathogenic defect of the clustered metabolic disturbances seen in polycystic ovary syndrome (PCOS). Metformin is an insulin sensitizer acting in the liver and the peripheral tissues that ameliorates the metabolic and reproductive defects in PCOS. In addition, pioglitazone is an insulin sensitizer used in diabetes mellitus type 2 (T2DM), improving insulin resistance (IR) in adipose tissue and muscles. In T2DM, these drugs are also used as a combined treatment due to their "add-on effect" on insulin resistance. Although the beneficial role of troglitazone (a member of the thiazolidinediones (TZDs) family) in PCOS has been shown in the past, currently only pioglitazone is available in the market. A few small randomized controlled trials have directly compared the effectiveness of pioglitazone in women with PCOS, while there are a limited number of small studies that support the beneficial metabolic add-on effect of pioglitazone on metformin-treated PCOS women as compared to metformin or pioglitazone monotherapy. These findings suggest a potentially promising role for combined pioglitazone/metformin treatment in the management of PCOS in metformin-resistant patients. In view of recent concerns regarding pioglitazone usage and its associated health risk, we aim to compare the pros and cons of each drug regarding their metabolic and other hormonal effects in women with PCOS and to explore the possible beneficial effect of combined therapy in certain cases, taking into consideration the teratogenic effect of pioglitazone. Finally, we discuss the need for a randomized controlled trial that will evaluate the metabolic and other hormonal effects of combined metformin/pioglitazone treatment in PCOS with selective treatment targets.
Collapse
Affiliation(s)
- Georgios Valsamakis
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire, Warwick Medical School, Coventry, UK, Endocrine Unit, Aretaieion University Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | | | | | | |
Collapse
|
112
|
Age-Related Differences in Response to High-Fat Feeding on Adipose Tissue and Metabolic Profile in ZDSD Rats. ISRN OBESITY 2013; 2013:584547. [PMID: 24555150 PMCID: PMC3901986 DOI: 10.1155/2013/584547] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Accepted: 04/29/2013] [Indexed: 11/17/2022]
Abstract
The recruitment of new fat cells through adipogenesis may prevent the development of obesity-related comorbidities. However, adipogenic capacity is markedly reduced in mature adults. This study examined how initiation of high-fat feeding at different phases of adulthood modified adipose tissue (AT) morphology and obesity phenotype in obese and diabetic Zucker Diabetic Sprague Dawley (ZDSD) rats. For this, rodents were provided high-fat diet (HFD) beginning at 63, 84, or 112 d after parturition until termination (n = 6). At termination, ZDSD rats fed HFD beginning at 63 d after parturition (early adulthood) exhibited greater body fat and lower lean mass without significant changes to energy intake or body weight. Moreover, early high fat feeding increased adipocyte size and number, whereas these effects were absent at 84 or 112 d after parturition. At 126 d after parturition, there were no detectable transcript differences in PPARγ or C/EBPα. However, rodents provided HFD in early adolescence exhibited lower expression of canonical Wnt signaling intermediates. Corresponding with these changes was a marked reduction in AT-specific inflammation, as well as overall improvement in systemic glucose, lipid, and inflammatory homeostasis. Taken together, these data indicate that dietary regulation of adipocyte recruitment in adolescence may represent a major determinant of obesity phenotype.
Collapse
|
113
|
Searcy JL, Phelps JT, Pancani T, Kadish I, Popovic J, Anderson KL, Beckett TL, Murphy MP, Chen KC, Blalock EM, Landfield PW, Porter NM, Thibault O. Long-term pioglitazone treatment improves learning and attenuates pathological markers in a mouse model of Alzheimer's disease. J Alzheimers Dis 2013; 30:943-61. [PMID: 22495349 DOI: 10.3233/jad-2012-111661] [Citation(s) in RCA: 116] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Thiazolidinediones (TZDs) are agonists at peroxisome proliferator-activated gamma-type (PPAR-γ) receptors and are used clinically for the treatment of type 2 diabetes where they have been shown to reestablish insulin sensitivity, improve lipid profiles, and reduce inflammation. Recent work also suggests that TZDs may be beneficial in Alzheimer's disease (AD), ameliorating cognitive decline early in the disease process. However, there have been only a few studies identifying mechanisms through which cognitive benefits may be exerted. Starting at 10 months of age, the triple transgenic mouse model of AD (3xTg-AD) with accelerated amyloid-β (Aβ) deposition and tau pathology was treated with the TZD pioglitazone (PIO-Actos) at 18 mg/Kg body weight/day. After four months, PIO-treated animals showed multiple beneficial effects, including improved learning on the active avoidance task, reduced serum cholesterol, decreased hippocampal amyloid-β and tau deposits, and enhanced short- and long-term plasticity. Electrophysiological membrane properties and post-treatment blood glucose levels were unchanged by PIO. Gene microarray analyses of hippocampal tissue identified predicted transcriptional responses following TZD treatment as well as potentially novel targets of TZDs, including facilitation of estrogenic processes and decreases in glutamatergic and lipid metabolic/cholesterol dependent processes. Taken together, these results confirm prior animal studies showing that TZDs can ameliorate cognitive deficits associated with AD-related pathology, but also extend these findings by pointing to novel molecular targets in the brain.
Collapse
Affiliation(s)
- James L Searcy
- Department of Molecular and Biomedical Pharmacology, University of Kentucky Medical Center, Lexington, KY 40536-0084, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
114
|
Brännmark C, Nyman E, Fagerholm S, Bergenholm L, Ekstrand EM, Cedersund G, Strålfors P. Insulin signaling in type 2 diabetes: experimental and modeling analyses reveal mechanisms of insulin resistance in human adipocytes. J Biol Chem 2013; 288:9867-9880. [PMID: 23400783 DOI: 10.1074/jbc.m112.432062] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Type 2 diabetes originates in an expanding adipose tissue that for unknown reasons becomes insulin resistant. Insulin resistance reflects impairments in insulin signaling, but mechanisms involved are unclear because current research is fragmented. We report a systems level mechanistic understanding of insulin resistance, using systems wide and internally consistent data from human adipocytes. Based on quantitative steady-state and dynamic time course data on signaling intermediaries, normally and in diabetes, we developed a dynamic mathematical model of insulin signaling. The model structure and parameters are identical in the normal and diabetic states of the model, except for three parameters that change in diabetes: (i) reduced concentration of insulin receptor, (ii) reduced concentration of insulin-regulated glucose transporter GLUT4, and (iii) changed feedback from mammalian target of rapamycin in complex with raptor (mTORC1). Modeling reveals that at the core of insulin resistance in human adipocytes is attenuation of a positive feedback from mTORC1 to the insulin receptor substrate-1, which explains reduced sensitivity and signal strength throughout the signaling network. Model simulations with inhibition of mTORC1 are comparable with experimental data on inhibition of mTORC1 using rapamycin in human adipocytes. We demonstrate the potential of the model for identification of drug targets, e.g. increasing the feedback restores insulin signaling, both at the cellular level and, using a multilevel model, at the whole body level. Our findings suggest that insulin resistance in an expanded adipose tissue results from cell growth restriction to prevent cell necrosis.
Collapse
Affiliation(s)
- Cecilia Brännmark
- Department of Clinical and Experimental Medicine, Linköping University, SE58185 Linköping, Sweden
| | - Elin Nyman
- Department of Clinical and Experimental Medicine, Linköping University, SE58185 Linköping, Sweden
| | - Siri Fagerholm
- Department of Clinical and Experimental Medicine, Linköping University, SE58185 Linköping, Sweden
| | - Linnéa Bergenholm
- Department of Clinical and Experimental Medicine, Linköping University, SE58185 Linköping, Sweden
| | - Eva-Maria Ekstrand
- Department of Clinical and Experimental Medicine, Linköping University, SE58185 Linköping, Sweden
| | - Gunnar Cedersund
- Department of Clinical and Experimental Medicine, Linköping University, SE58185 Linköping, Sweden; Department of Biomedical Engineering, Linköping University, SE58185 Linköping, Sweden
| | - Peter Strålfors
- Department of Clinical and Experimental Medicine, Linköping University, SE58185 Linköping, Sweden.
| |
Collapse
|
115
|
Bénardeau A, Verry P, Atzpodien EA, Funk JM, Meyer M, Mizrahi J, Winter M, Wright MB, Uhles S, Sebokova E. Effects of the dual PPAR-α/γ agonist aleglitazar on glycaemic control and organ protection in the Zucker diabetic fatty rat. Diabetes Obes Metab 2013; 15:164-74. [PMID: 22958363 DOI: 10.1111/dom.12006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Revised: 06/12/2012] [Accepted: 09/02/2012] [Indexed: 12/27/2022]
Abstract
AIMS To evaluate the effects of aleglitazar, a dual peroxisome proliferator-activated receptor-α/γ agonist, on the development of diabetes-related organ dysfunction, in relation to glycaemic and lipid changes, in Zucker diabetic fatty (ZDF) rats. METHODS Six-week-old, male ZDF rats received aleglitazar 0.3 mg/kg/day or vehicle as food admix for 13 weeks (n = 10 per group). Age-matched male Zucker lean rats served as non-diabetic controls. Plasma and renal markers were measured at several time points. Histopathology and quantitative immunohistochemistry were performed at 13 weeks. RESULTS Glycated haemoglobin (5.4 vs. 9.2%) and blood glucose (8.3 ± 0.3 vs. 26.1 ± 1.0 mmol/l) were significantly reduced at 12 weeks with aleglitazar versus vehicle-treated ZDF rats (both p < 0.01), while aleglitazar preserved near-normal plasma insulin levels. Aleglitazar prevented the development of hypertriglyceridaemia (1.4 ± 0.1 vs. 8.5 ± 0.9 mmol/l) and reduced plasma non-esterified fatty acids (0.09 ± 0.02 vs. 0.26 ± 0.04 mmol/l) relative to vehicle-treated animals (both p < 0.01). Urinary glucose and protein concentrations were significantly reduced at 13 weeks with aleglitazar versus vehicle-treated rats (both p < 0.01). Consistent with its effect on glycaemic control, aleglitazar protected β-cell morphology, as evidenced by preservation of islet integrity, and reduction of β-cell apoptosis and islet fibrosis. Aleglitazar prevented renal glomerular hypertrophy, podocyte degeneration, glomerulosclerosis, tubulo-interstitial lesions and development of cataracts. CONCLUSIONS Aleglitazar strongly improved glycaemic and lipid parameters while protecting key tissues, including the pancreas, kidneys and eyes, against diabetes-associated structural and functional changes in the ZDF rat.
Collapse
Affiliation(s)
- A Bénardeau
- pRED, Pharma Research & Early Development, DTA Cardiovascular & Metabolism, F. Hoffmann-La Roche AG, Basel, Switzerland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
116
|
Arunachalam S, Tirupathi Pichiah PB, Achiraman S. Doxorubicin treatment inhibits PPARγ and may induce lipotoxicity by mimicking a type 2 diabetes-like condition in rodent models. FEBS Lett 2012; 587:105-10. [PMID: 23219922 DOI: 10.1016/j.febslet.2012.11.019] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Revised: 10/16/2012] [Accepted: 11/16/2012] [Indexed: 01/04/2023]
Abstract
Doxorubicin-treated animals show elevated serum triglyceride and blood glucose levels. Adipocytes play an important role in buffering blood glucose and lipids. A raise in serum lipid level triggers adipogenesis in order to increase the lipid absorption capacity of adipose tissue. Doxorubicin inhibits adipogenesis through the down-regulation of PPARγ, a crucial component of the lipid metabolic pathway which controls the expression of glucose and fatty acid transporters. Doxorubicin-mediated down-regulation of PPARγ inhibits blood glucose and lipid clearance thereby causing hyperglycemia and hyperlipidemia resulting in lipotoxicity, glucotoxicity, inflammation and insulin resistance. Therefore we hypothesize that doxorubicin treatment could mimic a type 2 diabetic condition.
Collapse
Affiliation(s)
- Sankarganesh Arunachalam
- Department of Environmental Biotechnology, Bharathidasan University, Tiruchirappalli 620 024, Tamil Nadu, India
| | | | | |
Collapse
|
117
|
Bidar AW, Ploj K, Lelliott C, Nelander K, Winzell MS, Böttcher G, Oscarsson J, Storlien L, Hockings PD. In vivo imaging of lipid storage and regression in diet-induced obesity during nutrition manipulation. Am J Physiol Endocrinol Metab 2012; 303:E1287-95. [PMID: 23032688 DOI: 10.1152/ajpendo.00274.2012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Changes in adipose tissue distribution and ectopic fat storage in, liver and skeletal muscle tissue impact whole body insulin sensitivity in both humans and experimental animals. Numerous mouse models of obesity, insulin resistance, and diabetes exist; however, current methods to assess mouse phenotypes commonly involve direct harvesting of the tissues of interest, precluding the possibility of repeated measurements in the same animal. In this study, we demonstrate that whole body 3-D imaging of body fat composition can be used to analyze distribution as well as redistribution of fat after intervention by repeated assessment of intrahepatocellular lipids (IHCL), intra-abdominal, subcutaneous, and total adipose tissue (IAT, SAT, and TAT) and brown adipose tissue (BAT). C57BL/6J mice fed a cafeteria diet for 16 wk were compared with mice fed standard chow for 16 wk and mice switched from café diet to standard chow after 12 wk. MRI determinations were made at 9 and 15 wk, and autopsy was performed at 16 wk. There was a strong correlation between MRI-calculated weights in vivo at 15 wk and measured weights at 16 wk ex vivo for IAT (r = 0.99), BAT (r = 0.93), and IHCL (r = 0.97). IHCL and plasma insulin increased steeply relative to body weight at body weights above 45 g. This study demonstrates that the use of 3-D imaging to assess body fat composition may allow substantial reductions in animal usage. The dietary interventions indicated that a marked metabolic deterioration occurred when the mice had gained a certain fat mass.
Collapse
Affiliation(s)
- Abdel Wahad Bidar
- AstraZeneca R&D, PHB Imaging, Pepparedsleden 1, SE-43183 Mölndal, Sweden.
| | | | | | | | | | | | | | | | | |
Collapse
|
118
|
Alonso-Castro AJ, Zapata-Bustos R, Gómez-Espinoza G, Salazar-Olivo LA. Isoorientin reverts TNF-α-induced insulin resistance in adipocytes activating the insulin signaling pathway. Endocrinology 2012; 153:5222-30. [PMID: 22948221 DOI: 10.1210/en.2012-1290] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Isoorientin (ISO) is a plant C-glycosylflavonoid with purported antidiabetic effects but unexplored mechanisms of action. To gain insight into its antidiabetic mechanisms, we assayed nontoxic ISO concentrations on the 2-(N-(7-nitrobenz-2-oxa-1, 3-diazol-4-yl) amino)-2-deoxy-d-glucose (2-NBDG) uptake by murine 3T3-F442A and human sc adipocytes. In insulin-sensitive adipocytes, ISO stimulated the 2-NBDG uptake by 210% (murine) and 67% (human), compared with insulin treatment. Notably, ISO also induced 2-NBDG uptake in murine (139%) and human (60%) adipocytes made resistant to insulin by treatment with TNF-α, compared with the incorporation induced in these cells by rosiglitazone. ISO induction of glucose uptake in adipocytes was abolished by inhibitors of the insulin signaling pathway. These inhibitors also blocked the proper phosphorylation of insulin signaling pathway components induced by ISO in both insulin-sensitive and insulin-resistant adipocytes. Additionally, ISO stimulated the transcription of genes encoding components of insulin signaling pathway in murine insulin-sensitive and insulin-resistant adipocytes. In summary, we show here that ISO exerts its antidiabetic effects by activating the insulin signaling pathway in adipocytes, reverts the insulin resistance caused in these cells by TNF-α by stimulating the proper phosphorylation of proteins in this signaling pathway, and induces the expression of genes encoding these proteins.
Collapse
Affiliation(s)
- Angel Josabad Alonso-Castro
- Instituto Potosino de Investigación Científica y Tecnológica, Molecular Biology Division, San Luis Potosí, México
| | | | | | | |
Collapse
|
119
|
Skrzypski M, Kaczmarek P, Le TT, Wojciechowicz T, Pruszyńska-Oszmalek E, Szczepankiewicz D, Sassek M, Arafat A, Wiedenmann B, Nowak KW, Strowski MZ. Effects of orexin A on proliferation, survival, apoptosis and differentiation of 3T3-L1 preadipocytes into mature adipocytes. FEBS Lett 2012; 586:4157-64. [PMID: 23123090 DOI: 10.1016/j.febslet.2012.10.013] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2012] [Revised: 09/11/2012] [Accepted: 10/07/2012] [Indexed: 12/29/2022]
Abstract
Metabolic activities of orexin A (OXA) in mature adipocytes are mediated via PI3K/PKB and PPARγ. However, the effects of OXA on preadipocytes are largely unknown. We report here that OXA stimulates the proliferation and viability of 3T3-L1 preadipocytes and protects them from apoptosis via ERK1/2, but not through PKB. OXA reduces proapoptotic activity of caspase-3 via ERK1/2. Inhibition of ERK1/2 prevents the differentiation of preadipocytes into adipocytes. Unlike insulin, neither short-term nor prolonged exposure of 3T3-L1 preadipocytes to OXA induces preadipocyte differentiation to adipocytes, despite increased ERK1/2 phosphorylation. Unlike insulin, OXA fails to activate PKB, which explains its inability to induce the differentiation of preadipocytes.
Collapse
Affiliation(s)
- M Skrzypski
- Department of Hepatology and Gastroenterology & Interdisciplinary Centre of Metabolism: Endocrinology, Diabetes and Metabolism, Charité-University Medicine Berlin, 13353 Berlin, Germany; Department of Animal Physiology and Biochemistry, Poznań University of Life Sciences, 60-637 Poznań, Poland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
120
|
Liao Z, Wu Z, Wu M. Cirsium japonicum flavones enhance adipocyte differentiation and glucose uptake in 3T3-L1 cells. Biol Pharm Bull 2012; 35:855-60. [PMID: 22687475 DOI: 10.1248/bpb.35.855] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cirsium japonicum flavones have been demonstrated to possess anti-diabetic effects in diabetic rats, but the functional mechanism remains unknown. The nuclear receptor peroxisome proliferator-activated receptor γ (PPARγ) plays an important role in glucose and lipid homeostasis. In this study, we report the effects of Cirsium japonicum flavones (pectolinarin and 5,7-dihydroxy-6,4-dimethoxy flavone) on PPARγ activation, adipocyte differentiation, and glucose uptake in 3T3-L1 cells. Reporter gene assays and Oil Red O staining showed that Cirsium japonicum flavones induced PPARγ activation and enhanced adipocyte differentiation of 3T3-L1 cells in a dose-dependent manner. In addition, Cirsium japonicum flavones increased the expression of PPARγ target genes, such as adiponectin and glucose transporter 4 (GLUT4), and enhanced the translocation of intracellular GLUT4 to the plasma membrane. In mature 3T3-L1 adipocytes, Cirsium japonicum flavones significantly enhanced the basal and insulin-stimulated glucose uptake. The flavones-induced effects in 3T3-L1 cells were abolished by the PPARγ antagonist, GW9662, and by the phosphatidylinositol 3-kinase (PI3K) inhibitor, wortmannin. This study suggests that Cirsium japonicum flavones promote adipocyte differentiation and glucose uptake by inducing PPARγ activation and then modulating the insulin signaling pathway in some way, which could benefit diabetes patients.
Collapse
Affiliation(s)
- Zhiyong Liao
- College of Life and Environmental Science, Wenzhou University, China.
| | | | | |
Collapse
|
121
|
Effect of pioglitazone on the fructose-induced abdominal adipose tissue dysfunction. PPAR Res 2012; 2012:259093. [PMID: 23091482 PMCID: PMC3469242 DOI: 10.1155/2012/259093] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Accepted: 08/23/2012] [Indexed: 12/31/2022] Open
Abstract
Aim. To test the potential role of PPARγ in the endocrine abdominal tissue dysfunction induced by feeding normal rats with a fructose rich diet (FRD) during three weeks. Methodology. Adult normal male rats received a standard commercial diet (CD) or FRD, (10% in drinking water) without or with pioglitazone (PIO) (i.p. 0.25 mg/Kg BW/day; CD-PIO and FRD-PIO). Thereafter, we measured circulating metabolic, endocrine, and oxidative stress (OS) markers, abdominal adipose tissue (AAT) mass, leptin (LEP) and plasminogen activator inhibitor-1 (PAI-1) tissue content/expression, and leptin release by isolated adipocytes incubated with different concentrations of insulin. Results. Plasma glucose, insulin, triglyceride, TBARS, LEP, and PAI-1 levels were higher in FRD rats; PIO coadministration fully prevented all these increments. AAT adipocytes from FRD rats were larger, secreted a higher amount of LEP, and displayed decreased sensitivity to insulin stimulation; these effects were significantly ameliorated by PIO. Whereas AAT LEP and PAI-1 (mRNA) concentrations increased significantly in FRD rats, those of insulin-receptor-substrate- (IRS-) 1 and IRS-2 were reduced. PIO coadministration prevented FRD effects on LEP, PAI-1, and IRS-2 (fully) and IRS-1 (partially) mRNAs in AAT. Conclusion. PPARγ would play a relevant role in the development of the FRD-induced metabolic-endocrine dysfunction.
Collapse
|
122
|
Abstract
Adipocyte progenitors are thought to play a fundamental role in white adipose tissue (WAT) plasticity, which enables dynamic modulation of WAT metabolic and cellular characteristics in response to various stimuli. In general, two main strategies have been used to identify adipocyte progenitor cells: fluorescence-activated cell sorting (FACS)-based prospective analysis and lineage tracing. Although FACS-isolation is highly useful in defining multipotential stem cell populations for in vitro analysis and transplantation, lineage tracing is essential to identify endogenous progenitors that do, in fact, differentiate into adipocytes in vivo. Our recent lineage tracing studies have shown that cells expressing the surface marker platelet-derived growth factor receptor α (PDGFRα) give rise to white and brown adipocytes in adult WAT, depending on inductive cues. PDGFRα+ cells are a subpopulation of those expressing CD34 and Sca1, and have unique morphology whereby long dendritic processes contact numerous cell types in the microenvironment. The significant contribution of PDGFRα+ cells to browning and hyperplastic expansion of WAT leads us to propose that PDGFRα+ cells are remodeling stem cells in adult WAT. Application of advanced imaging technology and genetic tools to this progenitor population will allow greater understanding of cellular plasticity in adipose tissue.
Collapse
|
123
|
Maternal obesity, lipotoxicity and cardiovascular diseases in offspring. J Mol Cell Cardiol 2012; 55:111-6. [PMID: 22982026 DOI: 10.1016/j.yjmcc.2012.08.023] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2012] [Revised: 08/28/2012] [Accepted: 08/28/2012] [Indexed: 02/02/2023]
Abstract
Maternal obesity has risen dramatically over the past 20 years, by nearly 42% in African-Americans and 29% in Caucasians. Maternal obesity is afflicted with many maternal obstetric complications in the offspring including high blood pressure, obesity, gestational diabetes and increased perinatal morbidity. Maternal nutritional environment plays a rather important role in the programming of the health set-points in the offspring such as glucose and insulin metabolism, energy balance and predisposition to metabolic disorders. In particular, maternal obesity is associated with elevated prevalence of cardiovascular diseases in the offspring. Evidence from human and experimental studies including rodents and nonhuman primates has indicated that maternal obesity or overnutrition programs offspring for an increased risk of adult obesity. Maternal obesity or fat diet exposure predisposes the onset and development of obesity, insulin resistance, cardiac hypertrophy and myocardial contractile anomalies in the offspring. A number of mechanisms including elevated hormones (leptin, insulin), nutrients (fatty acids, triglycerides and glucose) and inflammatory cytokines have been postulated to play a key role in maternal obesity-induced postnatal cardiovascular sequelae. In addition, lipotoxicity (accumulation of lipid metabolites) resulting from maternal obesity is capable of activating a number of stress signaling cascades including pro-inflammatory cytokines and oxidative stress to exacerbate maternal obesity-induced cardiovascular complications later on in adult life. This mini-review summarizes the recent knowledge with regard to the role of lipotoxicity in maternal obesity-induced change in cardiovascular function in the offspring. This article is part of a Special Issue entitled "Focus on Cardiac Metabolism".
Collapse
|
124
|
Sanchez-Gurmaches J, Hung CM, Sparks CA, Tang Y, Li H, Guertin DA. PTEN loss in the Myf5 lineage redistributes body fat and reveals subsets of white adipocytes that arise from Myf5 precursors. Cell Metab 2012; 16:348-62. [PMID: 22940198 PMCID: PMC3488151 DOI: 10.1016/j.cmet.2012.08.003] [Citation(s) in RCA: 265] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Revised: 03/16/2012] [Accepted: 08/09/2012] [Indexed: 02/07/2023]
Abstract
The developmental origin of adipose tissue and what controls its distribution is poorly understood. By lineage tracing and gene expression analysis in mice, we provide evidence that mesenchymal precursors expressing Myf5--which are thought to give rise only to brown adipocytes and skeletal muscle--also give rise to a subset of white adipocytes. Furthermore, individual brown and white fats contain a mixture of adipocyte progenitor cells derived from Myf5(+) and Myf5(neg) lineages, the number of which varies with depot location. Subsets of white adipocytes originating from both Myf5(+) and Myf5(neg) precursors respond to β(3)-adrenoreceptor stimulation, suggesting "brite" adipocytes may also have multiple origins. We additionally find that deleting PTEN with myf5-cre causes lipomatosis and partial lipodystrophy by selectively expanding the Myf5(+) adipocyte lineages. Thus, the spectrum of adipocytes arising from Myf5(+) precursors is broader than previously thought, and differences in PI3K activity between adipocyte lineages alter body fat distribution.
Collapse
Affiliation(s)
- Joan Sanchez-Gurmaches
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | | | | | | | | | | |
Collapse
|
125
|
Kakazu E, Kondo Y, Ninomiya M, Kimura O, Nagasaki F, Ueno Y, Shimosegawa T. The influence of pioglitazone on the plasma amino acid profile in patients with nonalcoholic steatohepatitis (NASH). Hepatol Int 2012. [DOI: 10.1007/s12072-012-9395-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
126
|
Lodhi IJ, Yin L, Jensen-Urstad APL, Funai K, Coleman T, Baird JH, El Ramahi MK, Razani B, Song H, Fu-Hsu F, Turk J, Semenkovich CF. Inhibiting adipose tissue lipogenesis reprograms thermogenesis and PPARγ activation to decrease diet-induced obesity. Cell Metab 2012; 16:189-201. [PMID: 22863804 PMCID: PMC3467338 DOI: 10.1016/j.cmet.2012.06.013] [Citation(s) in RCA: 195] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2011] [Revised: 03/07/2012] [Accepted: 06/22/2012] [Indexed: 10/28/2022]
Abstract
De novo lipogenesis in adipocytes, especially with high fat feeding, is poorly understood. We demonstrate that an adipocyte lipogenic pathway encompassing fatty acid synthase (FAS) and PexRAP (peroxisomal reductase activating PPARγ) modulates endogenous PPARγ activation and adiposity. Mice lacking FAS in adult adipose tissue manifested increased energy expenditure, increased brown fat-like adipocytes in subcutaneous adipose tissue, and resistance to diet-induced obesity. FAS knockdown in embryonic fibroblasts decreased PPARγ transcriptional activity and adipogenesis. FAS-dependent alkyl ether phosphatidylcholine species were associated with PPARγ and treatment of 3T3-L1 cells with one such ether lipid increased PPARγ transcriptional activity. PexRAP, a protein required for alkyl ether lipid synthesis, was associated with peroxisomes and induced during adipogenesis. PexRAP knockdown in cells decreased PPARγ transcriptional activity and adipogenesis. PexRAP knockdown in mice decreased expression of PPARγ-dependent genes and reduced diet-induced adiposity. These findings suggest that inhibiting PexRAP or related lipogenic enzymes could treat obesity and diabetes.
Collapse
Affiliation(s)
- Irfan J Lodhi
- Division of Endocrinology, Metabolism & Lipid Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
127
|
Miesel A, Müller-Fielitz H, Jöhren O, Vogt FM, Raasch W. Double blockade of angiotensin II (AT(1) )-receptors and ACE does not improve weight gain and glucose homeostasis better than single-drug treatments in obese rats. Br J Pharmacol 2012; 165:2721-35. [PMID: 22014027 DOI: 10.1111/j.1476-5381.2011.01726.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND AND PURPOSE Combination therapies are becoming increasingly important for the treatment of high blood pressure. Little is known about whether double blockade of angiotensin II (AT(1) ) receptors and angiotensin-converting enzyme (ACE) exert synergistic metabolic effects. EXPERIMENTAL APPROACH Spontaneously hypertensive rats were allowed to choose between palatable chocolate bars and standard chow and were simultaneously treated with the AT(1) blocker telmisartan (8 mg·kg(bw) (-1) ·day(-1) ), the ACE inhibitor ramipril (4 mg·kg(bw) (-1) ·day(-1) ) or a combination of the two (8 + 4 mg·kg(bw) (-1) ·day(-1) ) for 12 weeks. KEY RESULTS Although food-dependent energy intake was increased by telmisartan and telmisartan + ramipril compared with ramipril or controls, body weight gain, abundance of fat and plasma leptin levels were decreased. Increased insulin levels in response to an oral glucose tolerance test were comparably attenuated by telmisartan and telmisartan + ramipril, but not by ramipril. During an insulin tolerance test, glucose utilization was equally as effectively improved by telmisartan and telmisartan + ramipril. In response to a stress test, ACTH, corticosterone and glucose increased in controls. These stress reactions were attenuated by telmisartan and telmisartan + ramipril. CONCLUSIONS AND IMPLICATIONS The combination of telmisartan + ramipril was no more efficacious in regulating body weight and glucose homeostasis than telmisartan alone. However, telmisartan was more effective than ramipril in improving metabolic parameters and in reducing body weight. The association between the decrease in stress responses and the diminished glucose levels after stress supports our hypothesis that the ability of telmisartan, as an AT(1) receptor blocker, to alleviate stress reactions may contribute to its hypoglycaemic actions.
Collapse
Affiliation(s)
- Anja Miesel
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | | | | | | | | |
Collapse
|
128
|
Lange C, Brunswig-Spickenheier B, Eissing L, Scheja L. Platelet lysate suppresses the expression of lipocalin-type prostaglandin D2 synthase that positively controls adipogenic differentiation of human mesenchymal stromal cells. Exp Cell Res 2012; 318:2284-96. [PMID: 22698646 DOI: 10.1016/j.yexcr.2012.06.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2011] [Revised: 05/31/2012] [Accepted: 06/04/2012] [Indexed: 12/19/2022]
Abstract
Mesenchymal stromal cells (MSCs) have been shown to display a considerable therapeutic potential in cellular therapies. However, harmful adipogenic maldifferentiation of transplanted MSCs may seriously threaten the success of this therapeutic approach. We have previously demonstrated that using platelet lysate (PL) instead of widely used fetal calf serum (FCS) diminished lipid accumulation in adipogenically stimulated human MSCs and identified, among others, lipocalin-type prostaglandin D2 synthase (L-PGDS) as a gene suppressed in PL-supplemented MSCs. Here, we investigated the role of PL and putatively pro-adipogenic L-PGDS in human MSC adipogenesis. Next to strongly reduced levels of L-PGDS we show that PL-supplemented MSCs display markedly decreased expression of adipogenic master regulators and differentiation markers, both before and after induction of adipocyte differentiation. The low adipogenic differentiation capability of PL-supplemented MSCs could be partially restored by exogenous addition of L-PGDS protein. Conversely, siRNA-mediated downregulation of L-PGDS in FCS-supplemented MSCs profoundly reduced adipocyte differentiation. In contrast, inhibiting endogenous prostaglandin synthesis by aspirin did not reduce differentiation, suggesting that a mechanism such as lipid shuttling but not the prostaglandin D2 synthase activity of L-PGDS is critical for adipogenesis. Our data demonstrate that L-PGDS is a novel pro-adipogenic factor in human MSCs which might be of relevance in adipocyte metabolism and disease. L-PGDS gene expression is a potential quality marker for human MSCs, as it might predict unwanted adipogenic differentiation after MSC transplantation.
Collapse
Affiliation(s)
- Claudia Lange
- University Medical Center Hamburg-Eppendorf, Clinic for Stem Cell Transplantation and Research Dept. Cell and Gene Therapy, Martinistr. 52, 20246 Hamburg, Germany.
| | | | | | | |
Collapse
|
129
|
Abstract
Adipose cells are unique in the dynamism of their sizes, a requisite for their main function of storing and releasing lipid. Lipid metabolism is crucial for energy homeostasis. However, the regulation of lipid storage capacity in conditions of energy excess and scarcity is still not clear. It is not technically feasible to monitor every process affecting storage capacity such as recruitment, growth/shrinkage and death of individual adipose cells in real time for a sufficiently long period. However, recent computational approaches have allowed an examination of the detailed dynamics of adipose cells using statistical information in the form of precise measurements of adipose cell-size probability distributions. One interesting finding is that the growth/shrinkage of adipose cells (> 50 μm diameter) under positive/negative energy balance is proportional to the surface area of cells, limiting efficient lipid absorption/release from larger adipose cells. In addition to the physical characteristics of adipose cells, quantitative modeling integrates dynamics of adipose cells, providing the mechanism of cell turnover under normal and drug-treated conditions. Thus, further use of mathematical modeling applied to experimental measurements of adipose cell-size probability distributions in conjunction with physiological measurements of metabolic state may help unravel the intricate network of interactions underlying metabolic syndromes in obesity.
Collapse
|
130
|
Effects of the roughage/concentrate ratio on the expression of angiogenic growth factors in adipose tissue of fattening Wagyu steers. Meat Sci 2012; 90:807-13. [DOI: 10.1016/j.meatsci.2011.11.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Revised: 07/20/2011] [Accepted: 11/02/2011] [Indexed: 01/07/2023]
|
131
|
Müller-Fielitz H, Landolt J, Heidbreder M, Werth S, Vogt FM, Jöhren O, Raasch W. Improved insulin sensitivity after long-term treatment with AT1 blockers is not associated with PPARγ target gene regulation. Endocrinology 2012; 153:1103-15. [PMID: 22253423 DOI: 10.1210/en.2011-0183] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
In both cell culture experiments and in vivo studies, a number of angiotensin II type 1 (AT(1)) receptor antagonists activated the peroxisome proliferator-activated receptor-γ (PPARγ). This mechanism has been discussed to be, at least in part, responsible for the improvement in glucose metabolism observed in animal studies and clinical trials. To investigate whether the PPARγ-dependent mechanism may represent a valid target for chronic therapy, spontaneously hypertensive rats (SHR) were fed either with a cafeteria diet (CD) or standard chow. CD-fed SHR were simultaneously treated with either telmisartan (TEL; 8 mg/kg(body weight)· d) or candesartan (CAND; 10 mg/kg(body weight)· d) for 3 months because TEL, but not CAND, has been demonstrated to be a strong activator of PPARγ. After 3 months, chow- and CD-fed controls were hypertensive, whereas TEL and CAND treatment resulted in normalized blood pressures in SHR. Body weight and the amount of abdominal fat (determined by magnetic resonance imaging) were higher in CD- than in chow-fed SHR. After TEL or CAND, body weight, abdominal fat quantity, and adipocyte size returned to normal. In glucose tolerance tests, the glucose responses were comparable in the TEL- and CAND-treated SHR and obese controls, whereas the insulin response was almost halved by AT(1) blockade. Expression of PPARγ target genes aP2, FAT CD36, FASn, and PEPCK remained unaltered at the protein level in visceral fat after TEL and CAND compared with the CD-fed controls. Because the expression of examined PPARγ target genes was not affected, we concluded that improved insulin sensitivity after long-term treatment with AT(1) blockers was not related to a PPARγ-dependent mechanism.
Collapse
Affiliation(s)
- Helge Müller-Fielitz
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany
| | | | | | | | | | | | | |
Collapse
|
132
|
Kim MK, Yang S, Lee KH, Um JH, Liu M, Kang H, Park SJ, Chung JH. Promyelocytic leukemia inhibits adipogenesis, and loss of promyelocytic leukemia results in fat accumulation in mice. Am J Physiol Endocrinol Metab 2011; 301:E1130-42. [PMID: 21846906 PMCID: PMC3233778 DOI: 10.1152/ajpendo.00092.2011] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The function of the tumor suppressor promyelocytic leukemia (PML) protein is disrupted in promyelocytic leukemia. PML has been reported to function as a negative regulator of mTOR (mammalian target of rapamycin) and nuclear Akt under some conditions. mTOR and Akt pathways regulate a diverse array of pathways, including those that control insulin signaling, energy metabolism, growth, cellular survival, and lifespan. Although the PML-mTOR/Akt link suggests that PML may have metabolic functions in the whole organism, very little is known about the metabolic functions of PML. Here we report that PML(-/-) mice did not show any significant metabolic defects. There was no impairment in the mTOR/Akt or AMPK signaling in white adipose tissue, liver, or muscle. However, despite having normal food intake and activity levels, PML(-/-) mice gained body weight faster and had more fat mass, particularly subcutaneous fat mass, in the diet-induced obesity model. Using in vitro adipogenesis models, we discovered that PML is a suppressor of adipogenesis. PML expression decreased during adipogenesis and was undetectable in fully differentiated adipocytes. Loss of PML increased expression of the adipogenic transcription factors CCAAT/enhancer binding protein-α and peroxisome proliferator-activated receptor-γ. We found that the Sirt1-NCor-SMRT corepressor complex, which represses pparg transcription, does not bind to the pparg promoter efficiently upon PML depletion. On the basis of these findings, we propose that PML is a negative regulator of the adipogenic transcription factors and that, in times of energy excess, PML may limit fat accumulation by suppressing the differentiation of preadipocytes into adipocytes.
Collapse
Affiliation(s)
- Myung K Kim
- Laboratory of Obesity and Aging Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, USA.
| | | | | | | | | | | | | | | |
Collapse
|
133
|
Samaan MC. The macrophage at the intersection of immunity and metabolism in obesity. Diabetol Metab Syndr 2011; 3:29. [PMID: 22035457 PMCID: PMC3223491 DOI: 10.1186/1758-5996-3-29] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2011] [Accepted: 10/28/2011] [Indexed: 12/23/2022] Open
Abstract
Obesity is a worldwide pandemic representing one of the major challenges that societies face around the globe. Identifying the mechanisms involved in its development and propagation will help the development of preventative and therapeutic strategies that may help control its rising rates.Obesity is associated with chronic low-grade inflammation, and this is believed to be one of the major contributors to the development of insulin resistance, which is an early event in obesity and leads to type 2 diabetes when the pancreas fails to keep up with increased demand for insulin. In this review, we discuss the role of macrophages in mediation of inflammation in obesity in metabolic organs including adipose tissue, skeletal muscle and liver. The presence of immune cells at the interface with metabolic organs modulates both metabolic function and inflammatory responses in these organs, and may provide a potential therapeutic target to modulate metabolic function in obesity.
Collapse
Affiliation(s)
- M Constantine Samaan
- Division of Pediatric Endocrinology, Department of Pediatrics, McMaster Children's Hospital, McMaster University, 1200 Main Street West, Hamilton, Ontario, Canada.
| |
Collapse
|
134
|
Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ) regulates metabolic homeostasis and is a molecular target for anti-diabetic drugs. We report here the identification of a steroid receptor ligand, RU-486, as an unexpected PPARγ agonist, thereby uncovering a novel signaling route for this steroid drug. Similar to rosiglitazone, RU-486 modulates the expression of key PPARγ target genes and promotes adipocyte differentiation, but with a lower adipogenic activity. Structural and functional studies of receptor-ligand interactions reveal the molecular basis for a unique binding mode for RU-486 in the PPARγ ligand-binding pocket with distinctive properties and epitopes, providing the molecular mechanisms for the discrimination of RU-486 from thiazolidinediones (TZDs) drugs. Our findings together indicate that steroid compounds may represent an alternative approach for designing non-TZD PPARγ ligands in the treatment of insulin resistance.
Collapse
|
135
|
Kusunoki M, Tsutsumi K, Sato D, Nakamura A, Habu S, Mori Y, Morishita M, Yonemoto T, Miyata T, Nakaya Y, Nakamura T. Pioglitazone-induced body weight gain is prevented by combined administration with the lipoprotein lipase activator NO-1886. Eur J Pharmacol 2011; 668:486-91. [DOI: 10.1016/j.ejphar.2011.07.030] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2011] [Accepted: 07/27/2011] [Indexed: 12/31/2022]
|
136
|
Inulin-type fructans with prebiotic properties counteract GPR43 overexpression and PPARγ-related adipogenesis in the white adipose tissue of high-fat diet-fed mice. J Nutr Biochem 2011; 22:712-22. [DOI: 10.1016/j.jnutbio.2010.05.009] [Citation(s) in RCA: 202] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2010] [Revised: 05/04/2010] [Accepted: 05/29/2010] [Indexed: 11/21/2022]
|
137
|
Hexarelin Signaling to PPARgamma in Metabolic Diseases. PPAR Res 2011; 2008:364784. [PMID: 18288286 PMCID: PMC2233980 DOI: 10.1155/2008/364784] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2007] [Accepted: 11/27/2007] [Indexed: 12/23/2022] Open
Abstract
Investigating the metabolic functions of the nuclear receptor peroxisome proliferator-activated receptor γ (PPARγ) has been extremely rewarding over the past years.
Uncovering the biologic roles of PPARγ and its mechanism of action has greatly advanced our understanding of the transcriptional control of lipid and glucose metabolism, and compounds such as thiazolidinediones which directly regulate PPARγ have proven to exhibit potent insulin-sensitizer effects in the treatment of diabetes. We review here recent advances on the emerging role of growth hormone releasing peptides in regulating PPARγ through interaction with scavenger receptor CD36 and ghrelin GHS-R1a receptor. With the impact that these peptides exert on the metabolic pathways involved in lipid metabolism and energy homeostasis, it is hoped that the development of novel approaches in the regulation of PPAR functions will bring additional therapeutic possibilities to face problems related to metabolic diseases.
Collapse
|
138
|
Tang W, Zeve D, Seo J, Jo AY, Graff JM. Thiazolidinediones regulate adipose lineage dynamics. Cell Metab 2011; 14:116-22. [PMID: 21723509 PMCID: PMC3163675 DOI: 10.1016/j.cmet.2011.05.012] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2010] [Revised: 03/03/2011] [Accepted: 05/31/2011] [Indexed: 12/13/2022]
Abstract
White adipose tissue regulates metabolism; the importance of this control is highlighted by the ongoing pandemic of obesity and associated complications such as diabetes, atherosclerosis, and cancer. White adipose tissue maintenance is a dynamic process, yet very little is known about how pharmacologic stimuli affect such plasticity. Combining in vivo lineage marking and BrdU labeling strategies, we found that rosiglitazone, a member of the thiazolidinedione class of glucose-lowering medicines, markedly increases the evolution of adipose progenitors into adipocytes. Notably, chronic rosiglitazone administration disrupts the adipogenic and self-renewal capacities of the stem cell compartment and alters its molecular characteristics. These data unravel unknown aspects of adipose dynamics and provide a basis to manipulate the adipose lineage for therapeutic ends.
Collapse
Affiliation(s)
- Wei Tang
- Department of Developmental Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX 75390-9133, USA
| | | | | | | | | |
Collapse
|
139
|
Pioglitazone promotes preadipocyte proliferation by downregulating p16(Ink4a). Biochem Biophys Res Commun 2011; 411:375-80. [PMID: 21741366 DOI: 10.1016/j.bbrc.2011.06.152] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Accepted: 06/24/2011] [Indexed: 12/11/2022]
Abstract
Pioglitazone, a synthetic ligand of peroxisome proliferator-activated receptor (PPAR)γ, causes preadipocyte proliferation through a mechanism which still remains elusive. Here, to address the mechanism, we investigated the effects of PPARγ and pioglitazone on the kinetics of cyclin-dependent kinase inhibitors, especially with p16(Ink4a) (p16) centered, by employing 3T3-L1 preadipocytes. Pioglitazone promoted preadipocyte proliferation by increasing S and G(2)/M cell-cycle entry, which was accompanied by decreased p16 mRNA expression. PPARγ overexpression along with the luciferase reporter assay confirmed that PPARγ was crucial for the downregulation of p16 mRNA transcription, and that the action was augmented by pioglitazone. Thus, pioglitazone exerted cell-cycle dependent promoting effect on preadipocyte proliferation, of which mechanisms include p16-downregulation through PPARγ.
Collapse
|
140
|
Yoon M, Kim MY. The anti-angiogenic herbal composition Ob-X from Morus alba, Melissa officinalis, and Artemisia capillaris regulates obesity in genetically obese ob/ob mice. PHARMACEUTICAL BIOLOGY 2011; 49:614-619. [PMID: 21554004 DOI: 10.3109/13880209.2010.539617] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
CONTEXT The growth and development of adipose tissue leading to obesity is suggested to depend on angiogenesis. Our previous study showed that Melissa officinalis L. (Labiatae), Morus alba L. (Moraceae), and Artemisia capillaris Thunb. (Compositae) are involved in the regulation of angiogenesis. We hypothesized that Ob-X, a mixture of three herbs, M. alba, M. officinalis, and A. capillaris, can regulate obesity. OBJECTIVE To investigate the inhibitory effect of Ob-X on obesity in genetically obese ob/ob mice. MATERIALS AND METHODS The effect of Ob-X on angiogenesis was measured using a mouse Matrigel plug assay. The effects of Ob-X on obesity were investigated in ob/ob mice. RESULTS Ob-X inhibited angiogenesis in a dose-dependent manner, as evidenced by decreased blood vessel density in a mouse matrigel plug assay. Administration of Ob-X to ob/ob mice for 5 weeks produced a significant reduction in body weight gain by 27% compared with control (12.1 ± 3.01 vs. 16.6 ± 2.24 g, respectively). Ob-X also significantly decreased visceral adipose tissue mass by 15% (0.87 ± 0.12 vs. 1.02 ± 0.15 g, respectively). The size of adipocytes in visceral adipose tissue was reduced by 46% in Ob-X-treated mice. Ob-X treatment inhibited hepatic lipid accumulation and significantly decreased circulating glucose levels compared with controls (197 ± 56.5 vs. 365 ± 115 mg/dL, respectively). DISCUSSION AND CONCLUSION These results suggest that Ob-X, which has an anti-angiogenic activity, reduces body weight gain and visceral adipose tissue mass in genetically obese mice, providing evidence that obesity can be prevented by angiogenesis inhibitors.
Collapse
Affiliation(s)
- Michung Yoon
- Department of Life Sciences, Mokwon University, Daejeon, Korea.
| | | |
Collapse
|
141
|
Hata T, Mera Y, Tadaki H, Kuroki Y, Kawai T, Ohta T, Kakutani M. JTT-130, a novel intestine-specific inhibitor of microsomal triglyceride transfer protein, suppresses high fat diet-induced obesity and glucose intolerance in Sprague-Dawley rats. Diabetes Obes Metab 2011; 13:446-54. [PMID: 21255216 DOI: 10.1111/j.1463-1326.2011.01368.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
AIM Microsomal triglyceride transfer protein (MTP) takes part in the mobilization and secretion of triglyceride-rich lipoproteins from enterocytes and hepatocytes. We investigated the effects of JTT-130, a novel intestine-specific MTP inhibitor, on high fat diet-induced obesity and glucose intolerance. METHODS Male Sprague-Dawley rats were fed a 3.1% fat diet or a 35% fat diet with or without JTT-130 as a food admixture (0.029%). Food intake, body weight, abdominal fat, hepatic triglyceride, faecal free fatty acids and plasma levels of glucagon-like peptide-1 (GLP-1) and peptide YY (PYY) were assessed. Plasma levels of glucose and insulin were measured during intraperitoneal glucose tolerance tests. In addition, indirect calorimetry was performed on rats fed with a 35% fat diet. RESULTS JTT-130 treatment decreased body weights, abdominal fat and hepatic triglyceride with suppression of food intake and elevation of faecal free fatty acids and plasma GLP-1 and PYY levels in rats fed with the 35% fat diet, whereas no significant effects on these parameters except for increased faecal free fatty acids were observed in rats fed with the 3.1% fat diet. JTT-130 treatment decreased plasma levels of glucose and insulin during intraperitoneal glucose tolerance tests on rats fed with the 35% fat diet, but not on rats fed with the 3.1% fat diet. JTT-130-treated rats showed increased O(2) consumption and CO(2) production on a 35% fat diet. CONCLUSIONS JTT-130 suppresses high fat diet-induced obesity and glucose intolerance with suppression of food intake and fat absorption and could be useful for prevention and treatment of obesity and obesity-related insulin resistance.
Collapse
Affiliation(s)
- T Hata
- Japan Tobacco, Central Pharmaceutical Research Institute, Biological/Pharmacological Research Laboratories, Osaka.
| | | | | | | | | | | | | |
Collapse
|
142
|
Hwang HH, Moon PG, Lee JE, Kim JG, Lee W, Ryu SH, Baek MC. Identification of the target proteins of rosiglitazone in 3T3-L1 adipocytes through proteomic analysis of cytosolic and secreted proteins. Mol Cells 2011; 31:239-46. [PMID: 21347706 PMCID: PMC3932691 DOI: 10.1007/s10059-011-0026-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2010] [Revised: 11/27/2010] [Accepted: 12/06/2010] [Indexed: 12/13/2022] Open
Abstract
Rosiglitazone, one of the thiazolidinedione (TZD), is an oral antidiabetic drug that activates a gamma isoform of peroxisome proliferator-activated receptor (PPARγ). To identify target proteins induced by rosiglitazone in adipocytes, we first performed simultaneous in-depth proteomic profiling of cytosolic proteins and secreted proteins (secretome) from 3T3-L1 adipocytes using a label-free quantification method with nano-UPLC MS/MS. In total, we identified 646 proteins from 3T3-L1 adipocytes, of which 172 and 162 proteins were upregulated and downregulated >1.5-fold, respectively, in rosiglitazone-treated cells, as compared to controls. Some differentially expressed proteins in particular, including fatty acid translocase (FAT)/CD36, fatty acid binding protein, lipoprotein lipase, acetyl CoA acyltransferase, carnitine O-palmitoyltransferase 2, sterol carrier protein, adiponectin, and phosphoenolpyruvate carboxykinase could explain the current action mechanism of TZDs. Furthermore, this study is the first to report on two potential target proteins of rosiglitazone, such as adenomatosis polyposis coli 2 (APC2), and eukaryotic translation initiation factor 5A-1 (eIF5A) related to apoptosis and cell division. Our data clearly suggest that in-depth proteomic approaches using cytosolic and secreted proteins are important and necessary for identification of drug targets at the protein level.
Collapse
Affiliation(s)
- Hyun-Ho Hwang
- Department of Molecular Medicine, Kyungpook National University, Daegu 700-422, Korea
- Cell and Matrix Biology Research Institute, Kyungpook National University, Daegu 700-422, Korea
| | - Pyong-Gon Moon
- Department of Molecular Medicine, Kyungpook National University, Daegu 700-422, Korea
- Cell and Matrix Biology Research Institute, Kyungpook National University, Daegu 700-422, Korea
| | - Jeong-Eun Lee
- Department of Molecular Medicine, Kyungpook National University, Daegu 700-422, Korea
- Cell and Matrix Biology Research Institute, Kyungpook National University, Daegu 700-422, Korea
| | - Jung-Guk Kim
- Department of Internal Medicine, Kyungpook National University Hospital, Daegu, 700-721, Korea
| | - Wan Lee
- College of Medicine, Dongguk University, Gyeongju 780-714, Korea
| | - Sung-Ho Ryu
- Pohang University of Science and Technology, Pohang 790-784, Korea
| | - Moon-Chang Baek
- Department of Molecular Medicine, Kyungpook National University, Daegu 700-422, Korea
- Cell and Matrix Biology Research Institute, Kyungpook National University, Daegu 700-422, Korea
| |
Collapse
|
143
|
Weight loss and hypophagia after high-dose AT1-blockade is only observed after high dosing and depends on regular leptin signalling but not blood pressure. Naunyn Schmiedebergs Arch Pharmacol 2011; 383:373-84. [PMID: 21287150 DOI: 10.1007/s00210-011-0602-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2010] [Accepted: 01/12/2011] [Indexed: 01/11/2023]
Abstract
AT(1)-blockade has been shown to induce weight loss in animals or patients. The aim of this study was to investigate whether weight reduction after AT(1)-blockade is dependent on dose, blood pressure reduction and leptin signalling. Spontaneously hypertensive rats (SHR) and lean and obese Zucker rats were treated for 4 weeks with candesartan (0, 2, 6 or 16 mg/kg/day). Body weight, food intake and hypothalamic mRNA levels of (an)orexigenic peptides were determined. Obese Zucker rats served as a model of primary leptin resistance. In SHR, body mass index and food intake were decreased selectively by 16 mg/kg/day candesartan but not after using normal (2 mg/kg/day) or supranormal (6 mg/kg/day) doses. Correlation analysis between blood pressure and body weight indicated no relationship of hypotensive potency on weight loss. The hypothalamic mRNA levels of the orexigenic peptide MCH (melanin-concentrating hormone) were diminished in parallel. Consistent to the results in SHRs, 16 mg/kg/day candesartan revealed a decrease of body weight, food intake and hypothalamic MCH mRNA levels in lean Zucker rats. In obese Zucker rats, none of these parameters were reduced by candesartan. Loss of body weight and hypophagia are not general features of AT(1)-blockers, since neither was seen after normal or moderately supranormal doses, but they were, after the highest doses. These actions of AT(1)-blockers occur independently of their ability to lower blood pressure. They do depend on an intact leptin signalling, since they were absent in obese Zucker rats that feature a genetic mutation of the leptin receptor.
Collapse
|
144
|
Choi SS, Cha BY, Iida K, Lee YS, Yonezawa T, Teruya T, Nagai K, Woo JT. Artepillin C, as a PPARγ ligand, enhances adipocyte differentiation and glucose uptake in 3T3-L1 cells. Biochem Pharmacol 2011; 81:925-33. [PMID: 21219874 DOI: 10.1016/j.bcp.2011.01.002] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2010] [Revised: 12/27/2010] [Accepted: 01/04/2011] [Indexed: 01/01/2023]
Abstract
The nuclear receptor peroxisome proliferator-activated receptor (PPAR) γ plays an important role in adipocyte differentiation. Its ligands, including thiazolidinediones, improve insulin sensitivity in type 2 diabetes. We investigated the effects of artepillin C, an ingredient of Baccharis dracunculifolia, on adipogenesis and glucose uptake using 3T3-L1 cells. In PPARγ ligand-binding assays, artepillin C exhibited binding affinity toward PPARγ. Artepillin C dose-dependently enhanced adipocyte differentiation of 3T3-L1 cells. As a result of the artepillin C-induced adipocyte differentiation, the gene expression of PPARγ and its target genes, such as aP2, adiponectin and glucose transporter (GLUT) 4, was increased. These increases were abolished by cotreatment with GW9662, a PPARγ antagonist. In mature 3T3-L1 adipocytes, artepillin C significantly enhanced the basal and insulin-stimulated glucose uptake. These effects were decreased by cotreatment with a PI3K inhibitor. Although artepillin C had no effects on the insulin signaling cascade, artepillin C enhanced the expression and plasma membrane translocation of GLUT1 and GLUT4 in mature adipocytes. In conclusion, these findings suggest that artepillin C promotes adipocyte differentiation and glucose uptake in part by direct binding to PPARγ, which could be the basis of the pharmacological benefits of green propolis intake in reducing the risk of type 2 diabetes.
Collapse
Affiliation(s)
- Sun-Sil Choi
- Research Institute for Biological Functions, Chubu University, 1200 Matsumoto, Kasugai, Aichi, Japan
| | | | | | | | | | | | | | | |
Collapse
|
145
|
Bjørndal B, Burri L, Staalesen V, Skorve J, Berge RK. Different adipose depots: their role in the development of metabolic syndrome and mitochondrial response to hypolipidemic agents. J Obes 2011; 2011:490650. [PMID: 21403826 PMCID: PMC3042633 DOI: 10.1155/2011/490650] [Citation(s) in RCA: 236] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2010] [Accepted: 12/27/2010] [Indexed: 12/16/2022] Open
Abstract
Adipose tissue metabolism is closely linked to insulin resistance, and differential fat distributions are associated with disorders like hypertension, diabetes, and cardiovascular disease. Adipose tissues vary in their impact on metabolic risk due to diverse gene expression profiles, leading to differences in lipolysis and in the production and release of adipokines and cytokines, thereby affecting the function of other tissues. In this paper, the roles of the various adipose tissues in obesity are summarized, with particular focus on mitochondrial function. In addition, we discuss how a functionally mitochondrial-targeted compound, the modified fatty acid tetradecylthioacetic acid (TTA), can influence mitochondrial function and decrease the size of specific fat depots.
Collapse
Affiliation(s)
- Bodil Bjørndal
- Institute of Medicine, University of Bergen, N 5021 Bergen, Norway
- *Bodil Bjørndal:
| | - Lena Burri
- Institute of Medicine, University of Bergen, N 5021 Bergen, Norway
| | - Vidar Staalesen
- Institute of Medicine, University of Bergen, N 5021 Bergen, Norway
| | - Jon Skorve
- Institute of Medicine, University of Bergen, N 5021 Bergen, Norway
| | - Rolf K. Berge
- Institute of Medicine, University of Bergen, N 5021 Bergen, Norway
- Department of Heart Disease, Haukeland University Hospital, N 5021 Bergen, Norway
| |
Collapse
|
146
|
Rosell M, Jones MC, Parker MG. Role of nuclear receptor corepressor RIP140 in metabolic syndrome. Biochim Biophys Acta Mol Basis Dis 2010; 1812:919-28. [PMID: 21193034 PMCID: PMC3117993 DOI: 10.1016/j.bbadis.2010.12.016] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Revised: 12/15/2010] [Accepted: 12/17/2010] [Indexed: 01/04/2023]
Abstract
Obesity and its associated complications, which can lead to the development of metabolic syndrome, are a worldwide major public health concern especially in developed countries where they have a very high prevalence. RIP140 is a nuclear coregulator with a pivotal role in controlling lipid and glucose metabolism. Genetically manipulated mice devoid of RIP140 are lean with increased oxygen consumption and are resistant to high-fat diet-induced obesity and hepatic steatosis with improved insulin sensitivity. Moreover, white adipocytes with targeted disruption of RIP140 express genes characteristic of brown fat including CIDEA and UCP1 while skeletal muscles show a shift in fibre type composition enriched in more oxidative fibres. Thus, RIP140 is a potential therapeutic target in metabolic disorders. In this article we will review the role of RIP140 in tissues relevant to the appearance and progression of the metabolic syndrome and discuss how the manipulation of RIP140 levels or activity might represent a therapeutic approach to combat obesity and associated metabolic disorders. This article is part of a Special Issue entitled: Translating nuclear receptors from health to disease.
Collapse
Affiliation(s)
- Meritxell Rosell
- Institute of Reproductive and Developmental Biology, Imperial College London, Faculty of Medicine, Hammersmith Campus 158 Du Cane Road, W12 0NN, UK.
| | | | | |
Collapse
|
147
|
Coordinate Transcriptomic and Metabolomic Effects of the Insulin Sensitizer Rosiglitazone on Fundamental Metabolic Pathways in Liver, Soleus Muscle, and Adipose Tissue in Diabetic db/db Mice. PPAR Res 2010; 2010. [PMID: 20953342 PMCID: PMC2953354 DOI: 10.1155/2010/679184] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2010] [Revised: 07/16/2010] [Accepted: 08/11/2010] [Indexed: 11/17/2022] Open
Abstract
Rosiglitazone (RSG), developed for the treatment of type 2 diabetes mellitus, is known to have potent effects on carbohydrate and lipid metabolism leading to the improvement of insulin sensitivity in target tissues. To further assess the capacity of RSG to normalize gene expression in insulin-sensitive tissues, we compared groups of 18-day-treated db/db mice with increasing oral doses of RSG (10, 30, and 100 mg/kg/d) with untreated non-diabetic littermates (db/+). For this aim, transcriptional changes were measured in liver, inguinal adipose tissue (IAT) and soleus muscle using microarrays and real-time PCR. In parallel, targeted metabolomic assessment of lipids (triglycerides (TGs) and free fatty acids (FFAs)) in plasma and tissues was performed by UPLC-MS methods. Multivariate analyses revealed a relationship between the differential gene expressions in liver and liver trioleate content and between blood glucose levels and a combination of differentially expressed genes measured in liver, IAT, and muscle. In summary, we have integrated gene expression and targeted metabolomic data to present a comprehensive overview of RSG-induced changes in a diabetes mouse model and improved the molecular understanding of how RSG ameliorates diabetes through its effect on the major insulin-sensitive tissues.
Collapse
|
148
|
Duan SZ, Usher MG, Foley EL, Milstone DS, Brosius FC, Mortensen RM. Sex dimorphic actions of rosiglitazone in generalised peroxisome proliferator-activated receptor-gamma (PPAR-gamma)-deficient mice. Diabetologia 2010; 53:1493-505. [PMID: 20401461 PMCID: PMC2896206 DOI: 10.1007/s00125-010-1748-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2009] [Accepted: 03/11/2010] [Indexed: 01/19/2023]
Abstract
AIMS/HYPOTHESIS The aim of this study was to determine the dependency on peroxisome proliferator-activated receptor-gamma (PPAR-gamma) of insulin sensitisation and glucose homeostasis by thiazolidinediones using a global Ppar-gamma (also known as Pparg)-knockout mouse model. METHODS Global Mox2-Cre-Ppar-gamma-knockout (MORE-PGKO) mice were treated with rosiglitazone and analysed for insulin sensitivity and glucose metabolism. Metabolic and hormonal variables were determined. Adipose and other tissues were measured and analysed for gene expression. RESULTS Rosiglitazone induced regrowth of fat in female but not male MORE-PGKO mice, and only in specific depots. Insulin sensitivity increased but, surprisingly, was not associated with the typical changes in adipokines, plasma NEFA or tissue triacylglycerol. However, increases in alternatively activated macrophage markers, which have been previously associated with metabolic improvement, were observed in the regrown fat. Rosiglitazone improved glucose homeostasis but not insulin sensitivity in male MORE-PGKO mice, with further increase of insulin associated with an apparent expansion of pancreatic islets. CONCLUSIONS/INTERPRETATION Stimulating fat growth by rosiglitazone is sufficient to improve insulin sensitivity in female mice with 95% PPAR-gamma deficiency. This increase in insulin sensitivity is not likely to be due to changes typically seen in adipokines or lipids but may involve changes in macrophage polarisation that occur independent of PPAR-gamma. In contrast, rosiglitazone improves glucose homeostasis in male mice with similar PPAR-gamma deficiency by increasing insulin production independent of changes in adiposity. Further, the insulin-sensitising effect of rosiglitazone is dependent on PPAR-gamma in this male lipodystrophic model.
Collapse
Affiliation(s)
- S. Z. Duan
- Department of Internal Medicine, Nephrology Division, University of Michigan Medical School, Ann Arbor, MI, USA
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - M. G. Usher
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, 7641 Med. Sci. II, 1150 W. Med. Ctr. Dr., Ann Arbor, Michigan 48109-5622, Ann Arbor, MI, USA
| | - E. L. Foley
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, 7641 Med. Sci. II, 1150 W. Med. Ctr. Dr., Ann Arbor, Michigan 48109-5622, Ann Arbor, MI, USA
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI, USA
| | - D. S. Milstone
- Vascular Research Division, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - F. C. Brosius
- Department of Internal Medicine, Nephrology Division, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, 7641 Med. Sci. II, 1150 W. Med. Ctr. Dr., Ann Arbor, Michigan 48109-5622, Ann Arbor, MI, USA
| | - R. M. Mortensen
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, 7641 Med. Sci. II, 1150 W. Med. Ctr. Dr., Ann Arbor, Michigan 48109-5622, Ann Arbor, MI, USA
- Department of Internal Medicine, Metabolism Endocrinology and Diabetes Division, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
149
|
Abstract
PURPOSE OF REVIEW Recent studies demonstrate that adipose tissue undergoes a continuous process of remodeling that is pathologically accelerated in the obese state. Contrary to earlier dogma, adipocytes die and are replaced by newly differentiated ones. This review will summarize recent advances of our knowledge of the mechanisms that regulate adipose tissue remodeling and highlight the influences of obesity, depot, and sex, as well as the relevance of rodent models to humans. RECENT FINDINGS A substantial literature now points to the importance of dynamic changes in adipocyte and immune cell turnover, angiogenesis, and extracellular matrix remodeling in regulating the expandability and functional integrity of this tissue. In obesity, the macrophages are recruited, surrounding dead adipocytes and polarized toward an inflammatory phenotype. The number of dead adipocytes is closely associated with the pathophysiological consequences of obesity, including insulin resistance and hepatic steatosis. Further, there are substantial depot, sex and species differences in the extent of remodeling. SUMMARY Adipose tissue undergoes a continuous remodeling process that normally maintains tissue health, but may spin out of control and lead to adipocyte death in association with the recruitment and activation of macrophages, and systemic insulin resistance.
Collapse
Affiliation(s)
| | | | - Susan K. Fried
- Correspondence to Susan K. Fried PhD, Division of Endocrinology, Diabetes, and Nutrition, Department of Medicine, Boston University, School of Medicine, 650 Albany St, EBRC-810, Boston, MA 02118, Tel: 617-638-7110; Fax: 617-638-7124;
| |
Collapse
|
150
|
Inskip J, Plunet W, Ramer L, Ramsey JB, Yung A, Kozlowski P, Ramer M, Krassioukov A. Cardiometabolic risk factors in experimental spinal cord injury. J Neurotrauma 2010; 27:275-85. [PMID: 19772460 DOI: 10.1089/neu.2009.1064] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Cardiometabolic risk factors are sorely underreported after spinal cord injury (SCI), despite the high prevalence of metabolic disorders and cardiovascular mortality in this population. Body-composition analysis and serum-lipid profiling are two assessments that are beginning to be more widely used to document metabolic changes after clinical SCI. Individuals with SCI have been reported to carry increased visceral fat and to exhibit altered serum-lipid levels. However, little is known about the development of these cardiometabolic risk factors in animal models. Using a combination of magnetic resonance imaging (MRI) and adipose tissue dissection, we show that visceral and subcutaneous adipose tissue were both increased at 1 month, but not at 1 week, after complete T3 SCI in rats. Additionally, at 1 month post injury, T3 SCI rats exhibited nonfasting serum hypertriglyceridemia, a result obtained using both standard clinical methods and a home cholesterol monitoring device (CardioChek). Interestingly, at 1 month post injury, rats with complete T10 SCI did not show an increase in either visceral adiposity or serum triglyceride levels. The fact that complete high-thoracic SCI disrupts lipid metabolism and perturbs fat storage in the subacute period, while low-thoracic SCI does not, suggests that differences in descending sympathetic control of adipose tissue might play a role in these changes. These results provide the first evidence of cardiometabolic risk factors in experimental animals with SCI, and are a starting point for investigations of the etiology of obesity and metabolic dysfunctions that often accompany SCI.
Collapse
Affiliation(s)
- Jessica Inskip
- International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | | | | | | | | | | |
Collapse
|