101
|
Valdearcos M, Xu AW, Koliwad SK. Hypothalamic inflammation in the control of metabolic function. Annu Rev Physiol 2015; 77:131-60. [PMID: 25668019 DOI: 10.1146/annurev-physiol-021014-071656] [Citation(s) in RCA: 146] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Diet-induced obesity leads to devastating and common chronic diseases, fueling ongoing interest in determining new mechanisms underlying both obesity and its consequences. It is now well known that chronic overnutrition produces a unique form of inflammation in peripheral insulin target tissues, and efforts to limit this inflammation have met with some success in preserving insulin sensitivity in obese individuals. Recently, the activation of inflammatory pathways by dietary excess has also been observed among cells located in the mediobasal hypothalamus, a brain area that exerts central control over peripheral glucose, fat, and energy metabolism. Here we review progress in the field of diet-induced hypothalamic inflammation, drawing key distinctions between metabolic inflammation in the hypothalamus and that occurring in peripheral tissues. We focus on specific stimuli of the inflammatory response, the roles of individual hypothalamic cell types, and the links between hypothalamic inflammation and metabolic function under normal and pathophysiological circumstances. Finally, we explore the concept of controlling hypothalamic inflammation to mitigate metabolic disease.
Collapse
|
102
|
Santoro A, Mattace Raso G, Meli R. Drug targeting of leptin resistance. Life Sci 2015; 140:64-74. [PMID: 26071010 DOI: 10.1016/j.lfs.2015.05.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 05/06/2015] [Accepted: 05/13/2015] [Indexed: 12/21/2022]
|
103
|
McCormick SM, Heller NM. Regulation of Macrophage, Dendritic Cell, and Microglial Phenotype and Function by the SOCS Proteins. Front Immunol 2015; 6:549. [PMID: 26579124 PMCID: PMC4621458 DOI: 10.3389/fimmu.2015.00549] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 10/13/2015] [Indexed: 12/11/2022] Open
Abstract
Macrophages are innate immune cells of dynamic phenotype that rapidly respond to external stimuli in the microenvironment by altering their phenotype to respond to and to direct the immune response. The ability to dynamically change phenotype must be carefully regulated to prevent uncontrolled inflammatory responses and subsequently to promote resolution of inflammation. The suppressor of cytokine signaling (SOCS) proteins play a key role in regulating macrophage phenotype. In this review, we summarize research to date from mouse and human studies on the role of the SOCS proteins in determining the phenotype and function of macrophages. We will also touch on the influence of the SOCS on dendritic cell (DC) and microglial phenotype and function. The molecular mechanisms of SOCS function in macrophages and DCs are discussed, along with how dysregulation of SOCS expression or function can lead to alterations in macrophage/DC/microglial phenotype and function and to disease. Regulation of SOCS expression by microRNA is discussed. Novel therapies and unanswered questions with regard to SOCS regulation of monocyte-macrophage phenotype and function are highlighted.
Collapse
Affiliation(s)
- Sarah M McCormick
- Anesthesiology and Critical Care Medicine, The Johns Hopkins University , Baltimore, MD , USA
| | - Nicola M Heller
- Anesthesiology and Critical Care Medicine, The Johns Hopkins University , Baltimore, MD , USA ; Anesthesiology and Critical Care Medicine, The Johns Hopkins University , Baltimore, MD , USA
| |
Collapse
|
104
|
Yin Y, Liu W, Dai Y. SOCS3 and its role in associated diseases. Hum Immunol 2015; 76:775-80. [DOI: 10.1016/j.humimm.2015.09.037] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 08/03/2015] [Accepted: 09/26/2015] [Indexed: 11/27/2022]
|
105
|
Abstract
The adipokine leptin realizes signal transduction via four different leptin receptor (OB-R) isoforms. The amount of functionally active OB-R, however, is affected by constitutive shedding of the extracellular domain. The product of the cleavage process, the so-called soluble leptin receptor (sOB-R), is the main binding protein for leptin in human blood and modulates its bioavailability. Concentrations of sOB-R are differentially regulated in metabolic disorders, such as type 1 diabetes mellitus or obesity, and can, therefore, enhance or reduce leptin sensitivity. Lipotoxicity and apoptosis increase OB-R cleavage via ADAM10-dependent mechanisms. In contrast, although increased sOB-R concentrations seem to directly inhibit leptin effects, reduced amounts of sOB-R may reflect decreased membrane expression of OB-R. These findings, in part, explain alterations of leptin sensitivity that are associated with changes in serum sOB-R concentrations seen in metabolic disorders.
Collapse
Affiliation(s)
- Michael Schaab
- Institute of Laboratory Medicine Clinical Chemistry and Molecular Diagnostics, Leipzig, Germany.
| | - Juergen Kratzsch
- Institute of Laboratory Medicine Clinical Chemistry and Molecular Diagnostics, Leipzig, Germany
| |
Collapse
|
106
|
Moravcová S, Červená K, Pačesová D, Bendová Z. Identification of STAT3 and STAT5 proteins in the rat suprachiasmatic nucleus and the Day/Night difference in astrocytic STAT3 phosphorylation in response to lipopolysaccharide. J Neurosci Res 2015; 94:99-108. [PMID: 26420542 DOI: 10.1002/jnr.23673] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 09/01/2015] [Accepted: 09/08/2015] [Indexed: 12/12/2022]
Abstract
Signal transducers and activators of transcription (STAT) proteins regulate many aspects of cellular physiology from growth and differentiations to immune responses. Using immunohistochemistry, we show the daily rhythm of STAT3 protein in the rat suprachiasmatic nucleus (SCN), with low but significant amplitude peaking in the morning. We also reveal the strong expression of STAT5A in astrocytes of the SCN and the STAT5B signal in nonastrocytic cells. Administration of lipopolysaccharide (LPS) acutely induced phosphorylation of STAT3 on Tyr705 during both the day and the night and induced phosphorylation on Ser727 but only after the daytime application. The LPS-induced phospho-STAT3 (Tyr705) remained elevated for 24 hr after the daytime application but declined within 8 hr when LPS was applied at night.
Collapse
Affiliation(s)
- Simona Moravcová
- Department of Physiology, Faculty of Science, Charles University in Prague, Prague, Czech Republic
| | - Kateřina Červená
- Department of Physiology, Faculty of Science, Charles University in Prague, Prague, Czech Republic
| | - Dominika Pačesová
- Department of Physiology, Faculty of Science, Charles University in Prague, Prague, Czech Republic
| | - Zdeňka Bendová
- Department of Physiology, Faculty of Science, Charles University in Prague, Prague, Czech Republic
| |
Collapse
|
107
|
Yang L, McKnight GS. Hypothalamic PKA regulates leptin sensitivity and adiposity. Nat Commun 2015; 6:8237. [PMID: 26381935 PMCID: PMC4576457 DOI: 10.1038/ncomms9237] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 07/30/2015] [Indexed: 12/17/2022] Open
Abstract
Mice lacking the RIIβ regulatory subunit of cyclic AMP-dependent protein kinase A (PKA) display reduced adiposity and resistance to diet-induced obesity. Here we show that RIIβ knockout (KO) mice have enhanced sensitivity to leptin's effects on both feeding and energy metabolism. After administration of a low dose of leptin, the duration of hypothalamic JAK/STAT3 signalling is increased, resulting in enhanced POMC mRNA induction. Consistent with the extended JAK/STAT3 activation, we find that the negative feedback regulator of leptin receptor signalling, Socs3, is inhibited in the hypothalamus of RIIβ KO mice. During fasting, RIIβ–PKA is activated and this correlates with an increase in CREB phosphorylation. The increase in CREB phosphorylation is absent in the fasted RIIβ KO hypothalamus. Selective inhibition of PKA activity in AgRP neurons partially recapitulates the leanness and resistance to diet-induced obesity of RIIβ KO mice. Our findings suggest that RIIβ–PKA modulates the duration of leptin receptor signalling and therefore the magnitude of the catabolic response to leptin. Mice lacking RIIβ, a regulatory subunit of protein kinase A, are lean and resistant to diet-induced obesity. Here, the authors show that RIIβ regulates leptin sensitivity, acting as a physiological brake on leptin responsiveness and the duration of leptin signalling in the hypothalamus.
Collapse
Affiliation(s)
- Linghai Yang
- Department of Pharmacology, University of Washington School of Medicine, 1959 North East Pacific Street, Box 357280, Seattle, Washington 98195, USA
| | - G Stanley McKnight
- Department of Pharmacology, University of Washington School of Medicine, 1959 North East Pacific Street, Box 357280, Seattle, Washington 98195, USA
| |
Collapse
|
108
|
Ericson E, Wennberg Huldt C, Strömstedt M, Brodin P. A novel role of the checkpoint kinase ATR in leptin signaling. Mol Cell Endocrinol 2015; 412:257-64. [PMID: 25980679 DOI: 10.1016/j.mce.2015.04.034] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Revised: 03/30/2015] [Accepted: 04/27/2015] [Indexed: 11/29/2022]
Abstract
In a world with increasing incidences of obesity, it becomes critical to understand the detailed regulation of appetite. To identify novel regulators of the signaling mediated by one of the key hormones of energy homeostasis, leptin, we screened a set of compounds for their effect on the downstream Signal Transducer and Activator of Transcription 3 (STAT3) signaling. Interestingly, cells exposed to inhibitors of the Ataxia Telangiectasia and RAD3-related protein ATR increased their leptin dependent STAT3 activity. This was due to failure of the cells to induce the negative feedback mediator Suppressor of Cytokine Signaling 3 (SOCS3), suggesting that ATR has a previously unknown role in the negative feedback regulation of leptin signaling. This is an important finding not only because it sheds light on additional genes involved in leptin signaling, but also because it brings forward a new potential therapeutic intervention point for increasing leptin signaling in obese individuals.
Collapse
Affiliation(s)
- Elke Ericson
- Bioscience, Cardiovascular and Metabolic Diseases, AstraZeneca R&D, Pepparedsleden 1, 431 83 Mölndal, Sweden.
| | - Charlotte Wennberg Huldt
- Bioscience, Cardiovascular and Metabolic Diseases, AstraZeneca R&D, Pepparedsleden 1, 431 83 Mölndal, Sweden
| | - Maria Strömstedt
- Bioscience, Cardiovascular and Metabolic Diseases, AstraZeneca R&D, Pepparedsleden 1, 431 83 Mölndal, Sweden
| | - Peter Brodin
- Bioscience, Cardiovascular and Metabolic Diseases, AstraZeneca R&D, Pepparedsleden 1, 431 83 Mölndal, Sweden
| |
Collapse
|
109
|
Sahu M, Anamthathmakula P, Sahu A. Phosphodiesterase-3B-cAMP pathway of leptin signalling in the hypothalamus is impaired during the development of diet-induced obesity in FVB/N mice. J Neuroendocrinol 2015; 27:293-302. [PMID: 25702569 DOI: 10.1111/jne.12266] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 01/16/2015] [Accepted: 02/12/2015] [Indexed: 01/05/2023]
Abstract
The phosphodiesterase-3B (PDE3B)-cAMP pathway plays an important role in transducing the action of leptin in the hypothalamus. Obesity is usually associated with hyperleptinaemia and resistance to anorectic and body weight-reducing effects of leptin. To determine whether the hypothalamic PDE3B-cAMP pathway of leptin signalling is impaired during the development of diet-induced obesity (DIO), we fed male FVB/N mice a high-fat diet (HFD: 58% kcal as fat) or low-fat diet (LFD: 6% kcal as fat) for 4 weeks. HFD fed mice developed DIO in association with hyperphagia, hyperleptinaemia and hyperinsulinaemia. Leptin (i.p.) significantly increased hypothalamic PDE3B activity and phosphorylated (p)-Akt levels in LFD-fed but not in HFD-fed mice. However, basal p-Akt levels in hypothalamus were increased in DIO mice. Additionally, amongst six-microdissected brain nuclei examined, leptin selectively decreased cAMP levels in the arcuate nucleus (ARC) of LFD-fed mice but failed to do so in HFD-fed mice. We next tested whether both the PBE3B and Akt pathways of leptin signalling remained impaired in DIO mice on the HFD for 12 weeks (long-term). DIO mice were hyperinsulinaemic and hyperleptinaemic in association with impaired glucose and insulin tolerance. Although, in LFD-fed mice, leptin significantly increased PDE3B activity and p-Akt levels in the hypothalamus, it failed to do so in HFD-fed mice. Also, basal p-Akt levels in the hypothalamus were increased in DIO mice and leptin had no further effect. Similarly, immunocytochemistry showed that leptin increased the number of p-Akt-positive cells in the ARC of LFD-fed but not in HFD-fed mice, and there was an increased basal number of p-Akt positive cells in the ARC of DIO mice. These results suggest that the PDE3B-cAMP- and Akt-pathways of leptin signalling in the hypothalamus are impaired during the development of DIO. Thus, a defect in the regulation by leptin of the hypothalamic PDE3B-cAMP pathway and Akt signalling may be one of the mechanisms of central leptin resistance and the development of DIO.
Collapse
Affiliation(s)
- M Sahu
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | | |
Collapse
|
110
|
Martínez de Morentin PB, Lage R, González-García I, Ruíz-Pino F, Martins L, Fernández-Mallo D, Gallego R, Fernø J, Señarís R, Saha AK, Tovar S, Diéguez C, Nogueiras R, Tena-Sempere M, López M. Pregnancy induces resistance to the anorectic effect of hypothalamic malonyl-CoA and the thermogenic effect of hypothalamic AMPK inhibition in female rats. Endocrinology 2015; 156:947-60. [PMID: 25535827 PMCID: PMC4330316 DOI: 10.1210/en.2014-1611] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Accepted: 12/19/2014] [Indexed: 12/20/2022]
Abstract
During gestation, hyperphagia is necessary to cope with the metabolic demands of embryonic development. There were three main aims of this study: Firstly, to investigate the effect of pregnancy on hypothalamic fatty acid metabolism, a key pathway for the regulation of energy balance; secondly, to study whether pregnancy induces resistance to the anorectic effect of fatty acid synthase (FAS) inhibition and accumulation of malonyl-coenzyme A (CoA) in the hypothalamus; and, thirdly, to study whether changes in hypothalamic AMPK signaling are associated with brown adipose tissue (BAT) thermogenesis during pregnancy. Our data suggest that in pregnant rats, the hypothalamic fatty acid pathway shows an overall state that should lead to anorexia and elevated BAT thermogenesis: decreased activities of AMP-activated protein kinase (AMPK), FAS, and carnitine palmitoyltransferase 1, coupled with increased acetyl-CoA carboxylase function with subsequent elevation of malonyl-CoA levels. This profile seems dependent of estradiol levels but not prolactin or progesterone. Despite the apparent anorexic and thermogenic signaling in the hypothalamus, pregnant rats remain hyperphagic and display reduced temperature and BAT function. Actually, pregnant rats develop resistance to the anorectic effects of central FAS inhibition, which is associated with a reduction of proopiomelanocortin (POMC) expression and its transcription factors phospho-signal transducer and activator of transcription 3, and phospho-forkhead box O1. This evidence demonstrates that pregnancy induces a state of resistance to the anorectic and thermogenic actions of hypothalamic cellular signals of energy surplus, which, in parallel to the already known refractoriness to leptin effects, likely contributes to gestational hyperphagia and adiposity.
Collapse
Affiliation(s)
- Pablo B Martínez de Morentin
- Department of Physiology, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS) (P.B.M.d.M., R.L., I.G.-G., L.M., D.F.M., R.S., S.T., C.D., R.N., M.L.), University of Santiago de Compostela (USC)-Instituto de Investigación Sanitaria (IDIS), Santiago de Compostela 15782, Spain; Centro de Investigación Biomédica en Red (CIBER) Fisiopatología de la Obesidad y Nutrición (CIBERobn) (P.B.M.d.M., R.L., I.G.-G., F.R.-P., L.M., D.F.M., S.T., C.D., R.N., M.T.-S., M.L.), Santiago de Compostela 15706, Spain; Department of Cell Biology, Physiology and Immunology (F.R.-P., M.T.-S.), University of Córdoba, Córdoba 14004, Spain; Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC)/Hospital Universitario Reina Sofía (F.R.-P., M.T.-S.), Córdoba 14004, Spain; Department of Morphological Sciences (R.G.), School of Medicine, University of Santiago de Compostela, Santiago de Compostela 15782, Spain; Department of Clinical Science (J.F.), K. G. Jebsen Center for Diabetes Research, University of Bergen, Bergen, N-5021, Norway; and Diabetes Research Unit, EBRC-827 (A.K.S.), Boston Medical Center, Boston, Massachusetts 02118
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
111
|
FTO is necessary for the induction of leptin resistance by high-fat feeding. Mol Metab 2015; 4:287-98. [PMID: 25830092 PMCID: PMC4354923 DOI: 10.1016/j.molmet.2015.01.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 01/25/2015] [Accepted: 01/28/2015] [Indexed: 11/23/2022] Open
Abstract
OBJECTIVE Loss of function FTO mutations significantly impact body composition in humans and mice, with Fto-deficient mice reported to resist the development of obesity in response to a high-fat diet (HFD). We aimed to further explore the interactions between FTO and HFD and determine if FTO can influence the adverse metabolic consequence of HFD. METHODS We studied mice deficient in FTO in two well validated models of leptin resistance (HFD feeding and central palmitate injection) to determine how Fto genotype may influence the action of leptin. Using transcriptomic analysis of hypothalamic tissue to identify relevant pathways affected by the loss of Fto, we combined data from co-immunoprecipitation, yeast 2-hybrid and luciferase reporter assays to identify mechanisms through which FTO can influence the development of leptin resistant states. RESULTS Mice deficient in Fto significantly increased their fat mass in response to HFD. Fto (+/-) and Fto (-/-) mice remained sensitive to the anorexigenic effects of leptin, both after exposure to a HFD or after acute central application of palmitate. Genes encoding components of the NFкB signalling pathway were down-regulated in the hypothalami of Fto-deficient mice following a HFD. When this pathway was reactivated in Fto-deficient mice with a single low central dose of TNFα, the mice became less sensitive to the effect of leptin. We identified a transcriptional coactivator of NFкB, TRIP4, as a binding partner of FTO and a molecule that is required for TRIP4 dependent transactivation of NFкB. CONCLUSIONS Our study demonstrates that, independent of body weight, Fto influences the metabolic outcomes of a HFD through alteration of hypothalamic NFкB signalling. This supports the notion that pharmacological modulation of FTO activity might have the potential for therapeutic benefit in improving leptin sensitivity, in a manner that is influenced by the nutritional environment.
Collapse
Key Words
- FTO, FaT mass and Obesity related
- Fto
- GWAS, Genome-wide association studies
- HFD, high-fat diet
- High-fat diet
- Hypothalamus
- ICV, intracerebroventricular injection
- Irx3, Iroquois Homeobox 3
- Leptin resistance
- MEF, Mouse embryonic fibroblasts
- NFкB
- Ob-R, leptin receptor
- PTPs, protein-tyrosine phosphatase
- SNPs, single nucleotide polymorphisms
- SOCS3
- SOCS3, suppressor of cytokine signalling
- TRIP4
- Tlr4, Toll-like receptor 4
- WAT, white adipose tissue
- Y2H, Yeast two-hybrid
Collapse
|
112
|
Calorie restriction-mediated restoration of hypothalamic signal transducer and activator of transcription 3 (STAT3) phosphorylation is not effective for lowering the body weight set point in IRS-2 knockout obese mice. Diabetol Int 2015. [DOI: 10.1007/s13340-015-0205-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
113
|
Identification of eQTLs for hepatic Xbp1s and Socs3 gene expression in mice fed a high-fat, high-caloric diet. G3-GENES GENOMES GENETICS 2015; 5:487-96. [PMID: 25617409 PMCID: PMC4390565 DOI: 10.1534/g3.115.016626] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a highly prevalent form of human hepatic disease and feeding mice a high-fat, high-caloric (HFHC) diet is a standard model of NAFLD. To better understand the genetic basis of NAFLD, we conducted an expression quantitative trait locus (eQTL) analysis of mice fed a HFHC diet. Two-hundred sixty-five (A/J × C57BL/6J) F2 male mice were fed a HFHC diet for 8 wk. eQTL analysis was utilized to identify genomic regions that regulate hepatic gene expression of Xbp1s and Socs3. We identified two overlapping loci for Xbp1s and Socs3 on Chr 1 (164.0–185.4 Mb and 174.4–190.5 Mb, respectively) and Chr 11 (41.1–73.1 Mb and 44.0–68.6 Mb, respectively), and an additional locus for Socs3 on Chr 12 (109.9–117.4 Mb). C57BL/6J-Chr 11A/J/ NaJ mice fed a HFHC diet manifested the A/J phenotype of increased Xbp1s and Socs3 gene expression (P < 0.05), whereas C57BL/6J-Chr 1A/J/ NaJ mice retained the C57BL/6J phenotype. In addition, we replicated the eQTLs on Chr 1 and Chr 12 (LOD scores ≥3.5) using mice from the BXD murine reference panel challenged with CCl4 to induce chronic liver injury and fibrosis. We have identified overlapping eQTLs for Xbp1 and Socs3 on Chr 1 and Chr 11, and consomic mice confirmed that replacing the C57BL/6J Chr 11 with the A/J Chr 11 resulted in an A/J phenotype for Xbp1 and Socs3 gene expression. Identification of the genes for these eQTLs will lead to a better understanding of the genetic factors responsible for NAFLD and potentially other hepatic diseases.
Collapse
|
114
|
Münzberg H, Morrison CD. Structure, production and signaling of leptin. Metabolism 2015; 64:13-23. [PMID: 25305050 PMCID: PMC4267896 DOI: 10.1016/j.metabol.2014.09.010] [Citation(s) in RCA: 282] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Revised: 09/17/2014] [Accepted: 09/20/2014] [Indexed: 01/07/2023]
Abstract
The cloning of leptin in 1994 was an important milestone in obesity research. In those days obesity was stigmatized as a condition caused by lack of character and self-control. Mutations in either leptin or its receptor were the first single gene mutations found to cause morbid obesity, and it is now appreciated that obesity is caused by a dysregulation of central neuronal circuits. From the first discovery of the leptin deficient obese mouse (ob/ob), to the cloning of leptin (ob aka lep) and leptin receptor (db aka lepr) genes, much has been learned about leptin and its action in the central nervous system. The initial high hopes that leptin would cure obesity were quickly dampened by the discovery that most obese humans have increased leptin levels and develop leptin resistance. Nevertheless, leptin target sites in the brain represent an excellent blueprint for distinct neuronal circuits that control energy homeostasis. A better understanding of the regulation and interconnection of these circuits will further guide and improve the development of safe and effective interventions to treat obesity. This review will highlight our current knowledge about the hormone leptin, its signaling pathways and its central actions to mediate distinct physiological functions.
Collapse
Affiliation(s)
- Heike Münzberg
- Pennington Biomedical Research Center, LSU System, Baton Rouge, LA.
| | | |
Collapse
|
115
|
Zampieri TT, Ramos-Lobo AM, Furigo IC, Pedroso JAB, Buonfiglio DC, Donato J. SOCS3 deficiency in leptin receptor-expressing cells mitigates the development of pregnancy-induced metabolic changes. Mol Metab 2014; 4:237-45. [PMID: 25737950 PMCID: PMC4338315 DOI: 10.1016/j.molmet.2014.12.005] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 12/08/2014] [Accepted: 12/12/2014] [Indexed: 12/25/2022] Open
Abstract
Objective During pregnancy, women normally increase their food intake and body fat mass, and exhibit insulin resistance. However, an increasing number of women are developing metabolic imbalances during pregnancy, including excessive gestational weight gain and gestational diabetes mellitus. Despite the negative health impacts of pregnancy-induced metabolic imbalances, their molecular causes remain unclear. Therefore, the present study investigated the molecular mechanisms responsible for orchestrating the metabolic changes observed during pregnancy. Methods Initially, we investigated the hypothalamic expression of key genes that could influence the energy balance and glucose homeostasis during pregnancy. Based on these results, we generated a conditional knockout mouse that lacks the suppressor of cytokine signaling-3 (SOCS3) only in leptin receptor-expressing cells and studied these animals during pregnancy. Results Among several genes involved in leptin resistance, only SOCS3 was increased in the hypothalamus of pregnant mice. Remarkably, SOCS3 deletion from leptin receptor-expressing cells prevented pregnancy-induced hyperphagia, body fat accumulation as well as leptin and insulin resistance without affecting the ability of the females to carry their gestation to term. Additionally, we found that SOCS3 conditional deletion protected females against long-term postpartum fat retention and streptozotocin-induced gestational diabetes. Conclusions Our study identified the increased hypothalamic expression of SOCS3 as a key mechanism responsible for triggering pregnancy-induced leptin resistance and metabolic adaptations. These findings not only help to explain a common phenomenon of the mammalian physiology, but it may also aid in the development of approaches to prevent and treat gestational metabolic imbalances.
Collapse
Key Words
- ARH, arcuate nucleus of the hypothalamus
- DIO, diet-induced obesity
- DMH, dorsomedial nucleus of the hypothalamus
- EGWG, excessive gestational weight gain
- GDM, gestational diabetes mellitus
- GH-V, placental growth hormone
- GTT, glucose tolerance test
- Gestational diabetes
- Hypothalamus
- IR, insulin receptor
- ITT, insulin tolerance test
- LepR, leptin receptor
- Leptin
- Leptin resistance
- Obesity
- PKC, protein kinase C
- RP, retroperitoneal
- SOCS3, suppressor of cytokine signaling-3
- STZ, streptozotocin
- Suppressor of cytokine signaling
- VMH, ventromedial nucleus of the hypothalamus
- pSTAT3, phosphorylation of the signal transducer and activator of transcription 3
- pSTAT3-ir, pSTAT3-immunoreactive
Collapse
Affiliation(s)
- Thais T Zampieri
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 1524, São Paulo, SP, 05508-000, Brazil
| | - Angela M Ramos-Lobo
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 1524, São Paulo, SP, 05508-000, Brazil
| | - Isadora C Furigo
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 1524, São Paulo, SP, 05508-000, Brazil
| | - João A B Pedroso
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 1524, São Paulo, SP, 05508-000, Brazil
| | - Daniella C Buonfiglio
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 1524, São Paulo, SP, 05508-000, Brazil
| | - Jose Donato
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 1524, São Paulo, SP, 05508-000, Brazil
| |
Collapse
|
116
|
Tang Y, Purkayastha S, Cai D. Hypothalamic microinflammation: a common basis of metabolic syndrome and aging. Trends Neurosci 2014; 38:36-44. [PMID: 25458920 DOI: 10.1016/j.tins.2014.10.002] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Revised: 10/13/2014] [Accepted: 10/14/2014] [Indexed: 12/19/2022]
Abstract
Chronic microinflammation is a hallmark of many aging-related neurodegenerative diseases as well as metabolic syndrome-driven diseases. Recent research indicates that chronic caloric excess can lead to hypothalamic microinflammation, which in turn participates in the development and progression of metabolic syndrome disorders such as obesity, glucose intolerance, and hypertension. Additionally, it was recently shown that increasing age after young adulthood can cause hypothalamic microinflammation independently of nutritional status, mediating a central mechanism of systemic aging. Taken together, these findings suggest that the hypothalamus has a fundamental role, via hypothalamic microinflammation, in translating overnutrition and aging into complex outcomes. Here we summarize recent work and suggest a conceptual model in which hypothalamic microinflammation is a common mediator of metabolic syndrome and aging.
Collapse
Affiliation(s)
- Yizhe Tang
- Department of Molecular Pharmacology, Diabetes Research Center, Institute of Aging, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Sudarshana Purkayastha
- Department of Molecular Pharmacology, Diabetes Research Center, Institute of Aging, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Dongsheng Cai
- Department of Molecular Pharmacology, Diabetes Research Center, Institute of Aging, Albert Einstein College of Medicine, New York, NY 10461, USA.
| |
Collapse
|
117
|
Tsaousidou E, Paeger L, Belgardt BF, Pal M, Wunderlich CM, Brönneke H, Collienne U, Hampel B, Wunderlich FT, Schmidt-Supprian M, Kloppenburg P, Brüning JC. Distinct Roles for JNK and IKK Activation in Agouti-Related Peptide Neurons in the Development of Obesity and Insulin Resistance. Cell Rep 2014; 9:1495-506. [PMID: 25456138 DOI: 10.1016/j.celrep.2014.10.045] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 07/10/2014] [Accepted: 10/16/2014] [Indexed: 01/01/2023] Open
Abstract
Activation of c-Jun N-terminal kinase 1 (JNK1)- and inhibitor of nuclear factor kappa-B kinase 2 (IKK2)-dependent signaling plays a crucial role in the development of obesity-associated insulin and leptin resistance not only in peripheral tissues but also in the CNS. Here, we demonstrate that constitutive JNK activation in agouti-related peptide (AgRP)-expressing neurons of the hypothalamus is sufficient to induce weight gain and adiposity in mice as a consequence of hyperphagia. JNK activation increases spontaneous action potential firing of AgRP cells and causes both neuronal and systemic leptin resistance. Similarly, activation of IKK2 signaling in AgRP neurons also increases firing of these cells but fails to cause obesity and leptin resistance. In contrast to JNK activation, IKK2 activation blunts insulin signaling in AgRP neurons and impairs systemic glucose homeostasis. Collectively, these experiments reveal both overlapping and nonredundant effects of JNK- and IKK-dependent signaling in AgRP neurons, which cooperate in the manifestation of the metabolic syndrome.
Collapse
Affiliation(s)
- Eva Tsaousidou
- Department of Mouse Genetics and Metabolism, Institute for Genetics, Cologne 50674, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), Cologne 50931, Germany; Center for Molecular Medicine (CMMC), University of Cologne, Cologne 50931, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Cologne 50937, Germany; Max Planck Institute for Metabolism Research, Cologne 50931, Germany
| | - Lars Paeger
- Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), Cologne 50931, Germany; Institute for Zoology, University of Cologne, Cologne 50674, Germany
| | - Bengt F Belgardt
- Department of Mouse Genetics and Metabolism, Institute for Genetics, Cologne 50674, Germany
| | - Martin Pal
- Baker IDI Heart and Diabetes Institute, Cellular and Molecular Metabolism Laboratory, Melbourne, VIC 3004, Australia
| | - Claudia M Wunderlich
- Department of Mouse Genetics and Metabolism, Institute for Genetics, Cologne 50674, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), Cologne 50931, Germany; Center for Molecular Medicine (CMMC), University of Cologne, Cologne 50931, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Cologne 50937, Germany; Max Planck Institute for Metabolism Research, Cologne 50931, Germany
| | - Hella Brönneke
- Mouse Phenotyping Core Facility, Cologne Excellence Cluster on Cellular Stress Responses, Cologne 50931, Germany
| | - Ursel Collienne
- Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), Cologne 50931, Germany; Institute for Zoology, University of Cologne, Cologne 50674, Germany
| | - Brigitte Hampel
- Department of Mouse Genetics and Metabolism, Institute for Genetics, Cologne 50674, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), Cologne 50931, Germany; Center for Molecular Medicine (CMMC), University of Cologne, Cologne 50931, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Cologne 50937, Germany; Max Planck Institute for Metabolism Research, Cologne 50931, Germany
| | - F Thomas Wunderlich
- Department of Mouse Genetics and Metabolism, Institute for Genetics, Cologne 50674, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), Cologne 50931, Germany; Center for Molecular Medicine (CMMC), University of Cologne, Cologne 50931, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Cologne 50937, Germany; Max Planck Institute for Metabolism Research, Cologne 50931, Germany
| | - Marc Schmidt-Supprian
- Department of Hematology and Oncology, Klinikum Rechts der Isar, Technical University of Munich, Munich 81675, Germany
| | - Peter Kloppenburg
- Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), Cologne 50931, Germany; Institute for Zoology, University of Cologne, Cologne 50674, Germany
| | - Jens C Brüning
- Department of Mouse Genetics and Metabolism, Institute for Genetics, Cologne 50674, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), Cologne 50931, Germany; Center for Molecular Medicine (CMMC), University of Cologne, Cologne 50931, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Cologne 50937, Germany; Max Planck Institute for Metabolism Research, Cologne 50931, Germany.
| |
Collapse
|
118
|
Contreras C, González-García I, Martínez-Sánchez N, Seoane-Collazo P, Jacas J, Morgan DA, Serra D, Gallego R, Gonzalez F, Casals N, Nogueiras R, Rahmouni K, Diéguez C, López M. Central ceramide-induced hypothalamic lipotoxicity and ER stress regulate energy balance. Cell Rep 2014; 9:366-377. [PMID: 25284795 PMCID: PMC5157160 DOI: 10.1016/j.celrep.2014.08.057] [Citation(s) in RCA: 187] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Revised: 08/07/2014] [Accepted: 08/23/2014] [Indexed: 12/30/2022] Open
Abstract
Hypothalamic endoplasmic reticulum (ER) stress is a key mechanism leading to obesity. Here, we demonstrate that ceramides induce lipotoxicity and hypothalamic ER stress, leading to sympathetic inhibition, reduced brown adipose tissue (BAT) thermogenesis, and weight gain. Genetic overexpression of the chaperone GRP78/BiP (glucose-regulated protein 78 kDa/binding immunoglobulin protein) in the ventromedial nucleus of the hypothalamus (VMH) abolishes ceramide action by reducing hypothalamic ER stress and increasing BAT thermogenesis, which leads to weight loss and improved glucose homeostasis. The pathophysiological relevance of this mechanism is demonstrated in obese Zucker rats, which show increased hypothalamic ceramide levels and ER stress. Overexpression of GRP78 in the VMH of these animals reduced body weight by increasing BAT thermogenesis as well as decreasing leptin and insulin resistance and hepatic steatosis. Overall, these data identify a triangulated signaling network involving central ceramides, hypothalamic lipotoxicity/ER stress, and BAT thermogenesis as a pathophysiological mechanism of obesity.
Collapse
Affiliation(s)
- Cristina Contreras
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782 Santiago de Compostela, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706 Santiago de Compostela, Spain
| | - Ismael González-García
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782 Santiago de Compostela, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706 Santiago de Compostela, Spain
| | - Noelia Martínez-Sánchez
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782 Santiago de Compostela, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706 Santiago de Compostela, Spain
| | - Patricia Seoane-Collazo
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782 Santiago de Compostela, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706 Santiago de Compostela, Spain
| | - Jordi Jacas
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706 Santiago de Compostela, Spain; Basic Sciences Department, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, Sant Cugat del Vallés, 08195 Barcelona, Spain
| | - Donald A Morgan
- Department of Pharmacology, University of Iowa, Iowa City, IA 52242, USA
| | - Dolors Serra
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706 Santiago de Compostela, Spain; Department of Biochemistry and Molecular Biology, School of Pharmacy, Institut de Biomedicina (IBUB), Universitat de Barcelona, 08028 Barcelona, Spain
| | - Rosalía Gallego
- Department of Morphological Sciences, School of Medicine, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Francisco Gonzalez
- Department of Surgery, CIMUS, University of Santiago de Compostela-Instituto de Invesstiagacion Sanitaria, 15782 Santiago de Compostela, Spain; Service of Ophthalmology, Complejo Hospitalario Universitario de Santiago de Compostela, 15706 Santiago de Compostela, Spain
| | - Núria Casals
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706 Santiago de Compostela, Spain; Basic Sciences Department, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, Sant Cugat del Vallés, 08195 Barcelona, Spain
| | - Rubén Nogueiras
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782 Santiago de Compostela, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706 Santiago de Compostela, Spain
| | - Kamal Rahmouni
- Department of Pharmacology, University of Iowa, Iowa City, IA 52242, USA; Department of Internal Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Carlos Diéguez
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782 Santiago de Compostela, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706 Santiago de Compostela, Spain
| | - Miguel López
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782 Santiago de Compostela, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706 Santiago de Compostela, Spain.
| |
Collapse
|
119
|
Heldsinger A, Grabauskas G, Wu X, Zhou S, Lu Y, Song I, Owyang C. Ghrelin induces leptin resistance by activation of suppressor of cytokine signaling 3 expression in male rats: implications in satiety regulation. Endocrinology 2014; 155:3956-69. [PMID: 25060362 PMCID: PMC4164930 DOI: 10.1210/en.2013-2095] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The anorexigenic adipocyte-derived hormone leptin and the orexigenic hormone ghrelin act in opposition to regulate feeding behavior via the vagal afferent pathways. The mechanisms by which ghrelin exerts its inhibitory effects on leptin are unknown. We hypothesized that ghrelin activates the exchange protein activated by cAMP (Epac), inducing increased SOCS3 expression, which negatively affects leptin signal transduction and neuronal firing in nodose ganglia (NG) neurons. We showed that 91 ± 3% of leptin receptor (LRb) -bearing neurons contained ghrelin receptors (GHS-R1a) and that ghrelin significantly inhibited leptin-stimulated STAT3 phosphorylation in rat NG neurons. Studies of the signaling cascades used by ghrelin showed that ghrelin caused a significant increase in Epac and suppressor of cytokine signaling 3 (SOCS3) expression in cultured rat NG neurons. Transient transfection of cultured NG neurons to silence SOCS3 and Epac genes reversed the inhibitory effects of ghrelin on leptin-stimulated STAT3 phosphorylation. Patch-clamp studies and recordings of single neuronal discharges of vagal primary afferent neurons showed that ghrelin markedly inhibited leptin-stimulated neuronal firing, an action abolished by silencing SOCS3 expression in NG. Plasma ghrelin levels increased significantly during fasting. This was accompanied by enhanced SOCS3 expression in the NG and prevented by treatment with a ghrelin antagonist. Feeding studies showed that silencing SOCS3 expression in the NG reduced food intake evoked by endogenous leptin. We conclude that ghrelin exerts its inhibitory effects on leptin-stimulated neuronal firing by increasing SOCS3 expression. The SOCS3 signaling pathway plays a pivotal role in ghrelin's inhibitory effect on STAT3 phosphorylation, neuronal firing, and feeding behavior.
Collapse
Affiliation(s)
- Andrea Heldsinger
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48019
| | | | | | | | | | | | | |
Collapse
|
120
|
Xbp1s in Pomc neurons connects ER stress with energy balance and glucose homeostasis. Cell Metab 2014; 20:471-82. [PMID: 25017942 PMCID: PMC4186248 DOI: 10.1016/j.cmet.2014.06.002] [Citation(s) in RCA: 212] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Revised: 05/20/2014] [Accepted: 05/29/2014] [Indexed: 01/14/2023]
Abstract
The molecular mechanisms underlying neuronal leptin and insulin resistance in obesity and diabetes remain unclear. Here we show that induction of the unfolded protein response transcription factor spliced X-box binding protein 1 (Xbp1s) in pro-opiomelanocortin (Pomc) neurons alone is sufficient to protect against diet-induced obesity as well as improve leptin and insulin sensitivity, even in the presence of strong activators of ER stress. We also demonstrate that constitutive expression of Xbp1s in Pomc neurons contributes to improved hepatic insulin sensitivity and suppression of endogenous glucose production. Notably, elevated Xbp1s levels in Pomc neurons also resulted in activation of the Xbp1s axis in the liver via a cell-nonautonomous mechanism. Together our results identify critical molecular mechanisms linking ER stress in arcuate Pomc neurons to acute leptin and insulin resistance as well as liver metabolism in diet-induced obesity and diabetes.
Collapse
|
121
|
Ladyman SR, Grattan DR. JAK-STAT and feeding. JAKSTAT 2014; 2:e23675. [PMID: 24058809 PMCID: PMC3710322 DOI: 10.4161/jkst.23675] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Revised: 01/17/2013] [Accepted: 01/18/2013] [Indexed: 12/26/2022] Open
Abstract
The regulation of energy balance requires a complex system to homeostatically maintain the adult body at a precise set point. The central nervous system, particularly the hypothalamus, plays a key role in integrating a variety of signals that can relay information about the body's energy stores. As part of this system, numerous cytokines and hormones contribute to the regulation of food intake and energy homeostasis. Cytokines, and some hormones, are known to act through JAK-STAT intracellular signaling pathways. The hormone leptin, which plays a vital role in appetite regulation, signals through the JAK-STAT pathway, and it is through this involvement that the JAK-STAT pathway has become an established component in the mechanisms regulating food intake within the body. Emerging research, however, is now showing that this involvement of JAK-STAT is not limited to its activation by leptin. Furthermore, while the JAK-STAT pathway may simply act to transmit the anorectic signal of circulating factors, this intracellular signaling pathway may also become impaired when normal regulation of energy balance is disrupted. Thus, altered JAK-STAT signaling may contribute to the breakdown of the normal homeostatic mechanisms maintaining body weight in obesity.
Collapse
Affiliation(s)
- Sharon R Ladyman
- Centre for Neuroendocrinology and Department of Anatomy; School of Medical Sciences; University of Otago; Dunedin, New Zealand
| | | |
Collapse
|
122
|
Wunderlich CM, Hövelmeyer N, Wunderlich FT. Mechanisms of chronic JAK-STAT3-SOCS3 signaling in obesity. JAKSTAT 2014; 2:e23878. [PMID: 24058813 PMCID: PMC3710326 DOI: 10.4161/jkst.23878] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Revised: 02/03/2013] [Accepted: 02/04/2013] [Indexed: 02/01/2023] Open
Abstract
Janus kinase (JAK)-signal transducers and activators of transcription (STAT) signaling pathways are critical for the maintenance of homeostatic and developmental processes; however, deregulation and chronic activation of JAK-STAT3 results in numerous diseases. Among others, obesity is currently being intensively studied. In obesity, chronic JAK-STAT3 is activated by the CNS by increased circulating leptin levels leading to the development of leptin resistance, whereas in the peripheral organs chronic IL-6-induced JAK-STAT3 impairs insulin action. We report the consequences of chronic JAK-STAT3 induced signaling as present under obese conditions in the main metabolic organs.
Collapse
Affiliation(s)
- Claudia M Wunderlich
- Max Planck Institute for Neurological Research; Institute for Genetics; University of Cologne and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC); Cologne, Germany
| | | | | |
Collapse
|
123
|
Abstract
The suppressors of cytokine signaling (SOCS) family of proteins are cytokine-inducible inhibitors of Janus kinase (JAK)-signal transducer and activator of the transcription (STAT) signaling pathways. Among the family, SOCS1 and SOCS3 potently suppress cytokine actions by inhibiting JAK kinase activities. The generation of mice lacking individual SOCS genes has been instrumental in defining the role of individual SOCS proteins in specific cytokine pathways in vivo; SOCS1 is an essential negative regulator of interferon-γ (IFNγ) and SOCS3 is an essential negative regulator of leukemia inhibitory factor (LIF). JAK-STAT3 activating cytokines have exhibited cardioprotective roles in the heart. The cardiac-specific deletion of SOCS3 enhances the activation of cardioprotective signaling pathways, inhibits myocardial apoptosis and fibrosis and results in the inhibition of left ventricular remodeling after myocardial infarction (MI). We propose that myocardial SOCS3 is a key determinant of left ventricular remodeling after MI, and SOCS3 may serve as a novel therapeutic target to prevent left ventricular remodeling after MI. In this review, we discuss the signaling pathways mediated by JAK-STAT and SOCS proteins and their roles in the development of myocardial injury under stress (e.g., pressure overload, viral infection and ischemia).
Collapse
Affiliation(s)
- Hideo Yasukawa
- Division of Cardiovascular Medicine; Department of Internal Medicine; Kurume University School of Medicine; Kurume, Japan ; Cardiovascular Research Institute; Kurume University School of Medicine; Kurume, Japan
| | | | | | | |
Collapse
|
124
|
Pedroso JAB, Buonfiglio DC, Cardinali LI, Furigo IC, Ramos-Lobo AM, Tirapegui J, Elias CF, Donato J. Inactivation of SOCS3 in leptin receptor-expressing cells protects mice from diet-induced insulin resistance but does not prevent obesity. Mol Metab 2014; 3:608-18. [PMID: 25161884 PMCID: PMC4142399 DOI: 10.1016/j.molmet.2014.06.001] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Revised: 05/29/2014] [Accepted: 06/03/2014] [Indexed: 02/05/2023] Open
Abstract
Therapies that improve leptin sensitivity have potential as an alternative treatment approach against obesity and related comorbidities. We investigated the effects of Socs3 gene ablation in different mouse models to understand the role of SOCS3 in the regulation of leptin sensitivity, diet-induced obesity (DIO) and glucose homeostasis. Neuronal deletion of SOCS3 partially prevented DIO and improved glucose homeostasis. Inactivation of SOCS3 only in LepR-expressing cells protected against leptin resistance induced by HFD, but did not prevent DIO. However, inactivation of SOCS3 in LepR-expressing cells protected mice from diet-induced insulin resistance by increasing hypothalamic expression of Katp channel subunits and c-Fos expression in POMC neurons. In summary, the regulation of leptin signaling by SOCS3 orchestrates diet-induced changes on glycemic control. These findings help to understand the molecular mechanisms linking obesity and type 2 diabetes, and highlight the potential of SOCS3 inhibitors as a promising therapeutic approach for the treatment of diabetes.
Collapse
Key Words
- AP, area postrema
- ARH, arcuate nucleus of the hypothalamus
- DIO, diet-induced obesity
- DMV, dorsal motor nucleus of the vagus
- GTT, glucose tolerance test
- HFD, high-fat diet
- High-fat diet
- Hypothalamus
- ITT, insulin tolerance test
- KO, knockout
- LepR, leptin receptor
- Leptin resistance
- NTS, nucleus of the solitary tract
- PI3K, phosphatidylinositol 3-kinase
- PKC, protein kinase C
- POMC
- POMC, proopiomelanocortin
- PTPs, protein-tyrosine phosphatases
- SOCS3, suppressor of cytokine signaling-3
- Suppressor of cytokine signaling-3
- T2DM, type 2 diabetes mellitus
- Type 2 diabetes mellitus
- VMH, ventromedial nucleus of the hypothalamus
Collapse
Affiliation(s)
- João A B Pedroso
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Daniella C Buonfiglio
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Lais I Cardinali
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Isadora C Furigo
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Angela M Ramos-Lobo
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Julio Tirapegui
- Department of Food Science and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Carol F Elias
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA ; Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA ; Department of Internal Medicine, Division of Hypothalamic Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jose Donato
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
125
|
Nakamura K, Fuster JJ, Walsh K. Adipokines: a link between obesity and cardiovascular disease. J Cardiol 2014; 63:250-9. [PMID: 24355497 PMCID: PMC3989503 DOI: 10.1016/j.jjcc.2013.11.006] [Citation(s) in RCA: 365] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 11/11/2013] [Indexed: 12/12/2022]
Abstract
Obesity is a risk factor for various cardiovascular diseases including hypertension, atherosclerosis, and myocardial infarction. Recent studies aimed at understanding the microenvironment of adipose tissue and its impact on systemic metabolism have shed light on the pathogenesis of obesity-linked cardiovascular diseases. Adipose tissue functions as an endocrine organ by secreting multiple immune-modulatory proteins known as adipokines. Obesity leads to increased expression of pro-inflammatory adipokines and diminished expression of anti-inflammatory adipokines, resulting in the development of a chronic, low-grade inflammatory state. This adipokine imbalance is thought to be a key event in promoting both systemic metabolic dysfunction and cardiovascular disease. This review will focus on the adipose tissue microenvironment and the role of adipokines in modulating systemic inflammatory responses that contribute to cardiovascular disease.
Collapse
Affiliation(s)
- Kazuto Nakamura
- Molecular Cardiology/Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - José J Fuster
- Molecular Cardiology/Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Kenneth Walsh
- Molecular Cardiology/Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA.
| |
Collapse
|
126
|
Liu Z, Gan L, Yang X, Zhang Z, Sun C. Hydrodynamic tail vein injection of SOCS3 eukaryotic expression vector in vivo promoted liver lipid metabolism and hepatocyte apoptosis in mouse. Biochem Cell Biol 2014; 92:119-25. [DOI: 10.1139/bcb-2013-0117] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Suppressor of cytokine signaling 3 (SOCS3), a signal transduction cytokine, is involved in lipid metabolism as well as in cell proliferation, differentiation, apoptosis, and so on. To explore the effects of SOCS3 on apoptosis and lipid metabolism in liver, we used a simple effective method named hydrodynamic tail vein injection to overexpress SOCS3. Then orbital blood was obtained for the assessment of blood lipid after injection. Lipid metabolism related genes were detected by Western blot after the determination of serum lipids. Meanwhile, liver cell apoptosis was observed by Hoechst and TUNEL staining and the expression of apoptosis related proteins Bax, Bcl-2, and Caspase3 were detected as well as the JAK2/STAT3 signaling pathway. In addition, we also demonstrated the effect of SOCS3 in prime hepatocyte by overexpression or interference of SOCS3 along with SD1008, which is a specific inhibitor of the JAK2/STAT3 signaling pathway. Taken together, all the results indicated that SOCS3 promoted lipid synthesis in mice liver and promoted hepatocyte apoptosis by inhibiting the activation of the JAK2/STAT3 signaling pathway, however the detailed regulation mechanism had not yet been fully understood and needs further study.
Collapse
Affiliation(s)
- Zhenjiang Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100. China
| | - Lu Gan
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100. China
| | - Xiaobo Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100. China
| | - Zhenzhen Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100. China
| | - Chao Sun
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100. China
| |
Collapse
|
127
|
Carow B, Rottenberg ME. SOCS3, a Major Regulator of Infection and Inflammation. Front Immunol 2014; 5:58. [PMID: 24600449 PMCID: PMC3928676 DOI: 10.3389/fimmu.2014.00058] [Citation(s) in RCA: 393] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 01/31/2014] [Indexed: 12/18/2022] Open
Abstract
In this review, we describe the role of suppressor of cytokine signaling-3 (SOCS3) in modulating the outcome of infections and autoimmune diseases as well as the underlying mechanisms. SOCS3 regulates cytokine or hormone signaling usually preventing, but in some cases aggravating, a variety of diseases. A main role of SOCS3 results from its binding to both the JAK kinase and the cytokine receptor, which results in the inhibition of STAT3 activation. Available data also indicate that SOCS3 can regulate signaling via other STATs than STAT3 and also controls cellular pathways unrelated to STAT activation. SOCS3 might either act directly by hampering JAK activation or by mediating the ubiquitination and subsequent proteasome degradation of the cytokine/growth factor/hormone receptor. Inflammation and infection stimulate SOCS3 expression in different myeloid and lymphoid cell populations as well as in diverse non-hematopoietic cells. The accumulated data suggest a relevant program coordinated by SOCS3 in different cell populations, devoted to the control of immune homeostasis in physiological and pathological conditions such as infection and autoimmunity.
Collapse
Affiliation(s)
- Berit Carow
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet , Stockholm , Sweden
| | - Martin E Rottenberg
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet , Stockholm , Sweden
| |
Collapse
|
128
|
Rasmussen BA, Breen DM, Duca FA, Côté CD, Zadeh-Tahmasebi M, Filippi BM, Lam TKT. Jejunal leptin-PI3K signaling lowers glucose production. Cell Metab 2014; 19:155-61. [PMID: 24361011 DOI: 10.1016/j.cmet.2013.11.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 10/08/2013] [Accepted: 11/11/2013] [Indexed: 10/25/2022]
Abstract
The fat-derived hormone leptin binds to its hypothalamic receptors to regulate glucose homeostasis. Leptin is also synthesized in the stomach and subsequently binds to its receptors expressed in the intestine, although the functional relevance of such activation remains largely unknown. We report here that intrajejunal leptin administration activates jejunal leptin receptors and signals through a phosphatidylinositol 3-kinase (PI3K)-dependent and signal transducer and activator of transcription 3 (STAT3)-independent signaling pathway to lower glucose production in healthy rodents. Jejunal leptin action is sufficient to lower glucose production in uncontrolled diabetic and high-fat-fed rodents and contributes to the early antidiabetic effect of duodenal-jejunal bypass surgery. These data unveil a glucoregulatory site of leptin action and suggest that enhancing leptin-PI3K signaling in the jejunum lowers plasma glucose concentrations in diabetes.
Collapse
Affiliation(s)
- Brittany A Rasmussen
- Toronto General Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Danna M Breen
- Toronto General Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Frank A Duca
- Toronto General Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Clémence D Côté
- Toronto General Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Melika Zadeh-Tahmasebi
- Toronto General Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Beatrice M Filippi
- Toronto General Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Tony K T Lam
- Toronto General Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; Banting and Best Diabetes Centre, University of Toronto, Toronto, ON M5G 2C4, Canada.
| |
Collapse
|
129
|
Do HJ, Jin T, Chung JH, Hwang JW, Shin MJ. Voglibose administration regulates body weight and energy intake in high fat-induced obese mice. Biochem Biophys Res Commun 2014; 443:1110-7. [PMID: 24388987 DOI: 10.1016/j.bbrc.2013.12.120] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 12/22/2013] [Indexed: 02/06/2023]
Abstract
We tested whether long-term administration of voglibose (VO) prevents diet induced obesity in addition to hypoglycemic effects in high fat fed mice and further investigated the underlying mechanisms by which voglibose exerts its weight lowering effect. Male C57BL/6 mice were fed ad libitum for 12 weeks with the control diet (CTL), high-fat diet (HFD) or the HFD with VO supplementations. Blood lipid profile, plasma leptin levels and hepatic triglyceride content, as well as expressions of genes involved in appetite and mitochondrial function were examined. The results showed that VO significantly reduced body weight, fat mass and energy intakes in high fat fed mice. VO showed improved metabolic profiles including blood glucose, triglyceride and free fatty acid. Elevated levels of plasma leptin in HFD were significantly reduced with the VO, furthermore, VO modulated the hypothalamic expressions of leptin receptors and appetite related genes. VO showed the upregulated expressions of PGC-1 in the liver and epididymal adipose tissue. In conclusion, VO may exert antiobesity properties through reductions in energy intake and improvement in mitochondrial function, indicating that VO has potential therapeutic use in patients with obesity, type 2 diabetes, and related complications.
Collapse
Affiliation(s)
- Hyun Ju Do
- Department of Food and Nutrition, Korea University, Seoul 136-703, Republic of Korea
| | - Taeon Jin
- Department of Applied Bioscience, CHA University, Gyeonggi-do 463-836, Republic of Korea
| | - Ji Hyung Chung
- Department of Applied Bioscience, CHA University, Gyeonggi-do 463-836, Republic of Korea.
| | - Ji Won Hwang
- Department of Food and Nutrition, Korea University, Seoul 136-703, Republic of Korea; Department of Public Health Sciences, Graduate School, Korea University, Seoul 136-703, Republic of Korea
| | - Min-Jeong Shin
- Department of Food and Nutrition, Korea University, Seoul 136-703, Republic of Korea; Department of Public Health Sciences, Graduate School, Korea University, Seoul 136-703, Republic of Korea; Korea University Guro Hospital, Korea University, Seoul 152-703, Republic of Korea.
| |
Collapse
|
130
|
Martin Agnoux A, Alexandre-Gouabau MC, Le Dréan G, Antignac JP, Parnet P. Relative contribution of foetal and post-natal nutritional periods on feeding regulation in adult rats. Acta Physiol (Oxf) 2014; 210:188-201. [PMID: 24010762 DOI: 10.1111/apha.12163] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Revised: 06/17/2013] [Accepted: 09/02/2013] [Indexed: 01/21/2023]
Abstract
AIM The aim of this study was to assess the contribution of both foetal and/or post-natal nutritional periods on feeding regulation in adult rats. METHODS Body weight gain, adipose tissue development, food preferences and feeding pattern under regular chow or Western diets were characterized on four experimental groups of rats: pups born from protein-restricted dams (R) and weaned by control (RC) or R dams (RR) and pups born from control dams weaned by C (CC) or R dams (CR). RESULTS Rats born with intrauterine growth restriction (IUGR) and fed a Western diet at adulthood appeared predisposed to body weight gain and more fat accretion, whereas CR rats, despite their preference for high-fat diet and their hyperphagia for Western diet, did not show significant increase in fat tissue. Daytime food intakes, as well as their speed of ingestion, were found modified in RC and RR. Alterations in the hypothalamic appetite regulatory mechanisms were investigated through neuropeptide expression analysis. IUGR rats showed altered expression of key elements of leptin and NPY signalling, while CR rats exhibited lesser expression of enterostatin, MC4r and HT-1Br mRNA. CONCLUSION Altogether, these results indicate that peri-natal nutrition has different lasting effects on feeding pattern and hypothalamic appetite regulation, depending on the time window insult.
Collapse
Affiliation(s)
- A. Martin Agnoux
- INRA, UMR1280, Physiologie des Adaptations Nutritionnelles; Nantes France
- Université de Nantes; UMR 1280, Physiologie des Adaptations Nutritionnelles; Nantes France
- IMAD, Institut des Maladies de l'Appareil Digestif, CRNH (Centre de Recherche en Nutrition Humaine); Nantes France
| | - M. -C. Alexandre-Gouabau
- INRA, UMR1280, Physiologie des Adaptations Nutritionnelles; Nantes France
- Université de Nantes; UMR 1280, Physiologie des Adaptations Nutritionnelles; Nantes France
- IMAD, Institut des Maladies de l'Appareil Digestif, CRNH (Centre de Recherche en Nutrition Humaine); Nantes France
| | - G. Le Dréan
- INRA, UMR1280, Physiologie des Adaptations Nutritionnelles; Nantes France
- Université de Nantes; UMR 1280, Physiologie des Adaptations Nutritionnelles; Nantes France
- IMAD, Institut des Maladies de l'Appareil Digestif, CRNH (Centre de Recherche en Nutrition Humaine); Nantes France
| | - J. -P. Antignac
- LUNAM université; Oniris, Laboratoire d'Etude des Résidus et Contaminants dans les Aliments (LABERCA); USC INRA 1329; Nantes France
| | - P. Parnet
- INRA, UMR1280, Physiologie des Adaptations Nutritionnelles; Nantes France
- Université de Nantes; UMR 1280, Physiologie des Adaptations Nutritionnelles; Nantes France
- IMAD, Institut des Maladies de l'Appareil Digestif, CRNH (Centre de Recherche en Nutrition Humaine); Nantes France
| |
Collapse
|
131
|
Roberts CK, Hevener AL, Barnard RJ. Metabolic syndrome and insulin resistance: underlying causes and modification by exercise training. Compr Physiol 2013; 3:1-58. [PMID: 23720280 DOI: 10.1002/cphy.c110062] [Citation(s) in RCA: 304] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Metabolic syndrome (MS) is a collection of cardiometabolic risk factors that includes obesity, insulin resistance, hypertension, and dyslipidemia. Although there has been significant debate regarding the criteria and concept of the syndrome, this clustering of risk factors is unequivocally linked to an increased risk of developing type 2 diabetes and cardiovascular disease. Regardless of the true definition, based on current population estimates, nearly 100 million have MS. It is often characterized by insulin resistance, which some have suggested is a major underpinning link between physical inactivity and MS. The purpose of this review is to: (i) provide an overview of the history, causes and clinical aspects of MS, (ii) review the molecular mechanisms of insulin action and the causes of insulin resistance, and (iii) discuss the epidemiological and intervention data on the effects of exercise on MS and insulin sensitivity.
Collapse
Affiliation(s)
- Christian K Roberts
- Exercise and Metabolic Disease Research Laboratory, Translational Sciences Section, School of Nursing, University of California at Los Angeles, Los Angeles, California, USA.
| | | | | |
Collapse
|
132
|
Purkayastha S, Cai D. Neuroinflammatory basis of metabolic syndrome. Mol Metab 2013; 2:356-63. [PMID: 24327952 DOI: 10.1016/j.molmet.2013.09.005] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2013] [Revised: 09/23/2013] [Accepted: 09/25/2013] [Indexed: 01/07/2023] Open
Abstract
Inflammatory reaction is a fundamental defense mechanism against threat towards normal integrity and physiology. On the other hand, chronic diseases such as obesity, type 2 diabetes, hypertension and atherosclerosis, have been causally linked to chronic, low-grade inflammation in various metabolic tissues. Recent cross-disciplinary research has led to identification of hypothalamic inflammatory changes that are triggered by overnutrition, orchestrated by hypothalamic immune system, and sustained through metabolic syndrome-associated pathophysiology. While continuing research is actively trying to underpin the identity and mechanisms of these inflammatory stimuli and actions involved in metabolic syndrome disorders and related diseases, proinflammatory IκB kinase-β (IKKβ), the downstream nuclear transcription factor NF-κB and some related molecules in the hypothalamus were discovered to be pathogenically significant. This article is to summarize recent progresses in the field of neuroendocrine research addressing the central integrative role of neuroinflammation in metabolic syndrome components ranging from obesity, glucose intolerance to cardiovascular dysfunctions.
Collapse
Affiliation(s)
- Sudarshana Purkayastha
- Department of Molecular Pharmacology, Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | |
Collapse
|
133
|
Schéle E, Grahnemo L, Anesten F, Hallén A, Bäckhed F, Jansson JO. The gut microbiota reduces leptin sensitivity and the expression of the obesity-suppressing neuropeptides proglucagon (Gcg) and brain-derived neurotrophic factor (Bdnf) in the central nervous system. Endocrinology 2013; 154:3643-51. [PMID: 23892476 DOI: 10.1210/en.2012-2151] [Citation(s) in RCA: 160] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The gut microbiota contributes to fat mass and the susceptibility to obesity. However, the underlying mechanisms are not completely understood. To investigate whether the gut microbiota affects hypothalamic and brainstem body fat-regulating circuits, we compared gene expression of food intake-regulating neuropeptides between germ-free and conventionally raised (CONV-R) mice. We found that CONV-R mice had decreased expression of the antiobesity neuropeptide glucagon-like peptide-1 (GLP-1) precursor proglucagon (Gcg) in the brainstem. Moreover, in both the hypothalamus and the brainstem, CONV-R mice had decreased expression of the antiobesity neuropeptide brain-derived neurotrophic factor (Bdnf). CONV-R mice had reduced expression of the pro-obesity peptides neuropeptide-Y (Npy) and agouti-related protein (Agrp), and increased expression of the antiobesity peptides proopiomelanocortin (Pomc) and cocaine- and amphetamine-regulated transcript (Cart) in the hypothalamus. The latter changes in neuropeptide expression could be secondary to elevated fat mass in CONV-R mice. Leptin treatment caused less weight reduction and less suppression of orexigenic Npy and Agrp expression in CONV-R mice compared with germ-free mice. The hypothalamic expression of leptin resistance-associated suppressor of cytokine signaling 3 (Socs-3) was increased in CONV-R mice. In conclusion, the gut microbiota reduces the expression of 2 genes coding for body fat-suppressing neuropeptides, Gcg and Bdnf, an alteration that may contribute to fat mass induction by the gut microbiota. Moreover, the presence of body fat-inducing gut microbiota is associated with hypothalamic signs of Socs-3-mediated leptin resistance, which may be linked to failed compensatory body fat reduction.
Collapse
Affiliation(s)
- Erik Schéle
- Sahlgrenska Academy at the University of Gothenburg, Institute of Neuroscience and Physiology/Endocrinology Medicinaregatan 11, Goteborg-41390, Sweden.
| | | | | | | | | | | |
Collapse
|
134
|
Huang H, Lee SH, Ye C, Lima IS, Oh BC, Lowell BB, Zabolotny JM, Kim YB. ROCK1 in AgRP neurons regulates energy expenditure and locomotor activity in male mice. Endocrinology 2013; 154:3660-70. [PMID: 23885017 PMCID: PMC3776869 DOI: 10.1210/en.2013-1343] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Normal leptin signaling is essential for the maintenance of body weight homeostasis. Proopiomelanocortin- and agouti-related peptide (AgRP)-producing neurons play critical roles in regulating energy metabolism. Our recent work demonstrates that deletion of Rho-kinase 1 (ROCK1) in the AgRP neurons of mice increased body weight and adiposity. Here, we report that selective loss of ROCK1 in AgRP neurons caused a significant decrease in energy expenditure and locomotor activity of mice. These effects were independent of any change in food intake. Furthermore, AgRP neuron-specific ROCK1-deficient mice displayed central leptin resistance, as evidenced by impaired Signal Transducer and Activator of Transcription 3 activation in response to leptin administration. Leptin's ability to hyperpolarize and decrease firing rate of AgRP neurons was also abolished in the absence of ROCK1. Moreover, diet-induced and genetic forms of obesity resulted in reduced ROCK1 activity in murine arcuate nucleus. Of note, high-fat diet also impaired leptin-stimulated ROCK1 activity in arcuate nucleus, suggesting that a defect in hypothalamic ROCK1 activity may contribute to the pathogenesis of central leptin resistance in obesity. Together, these data demonstrate that ROCK1 activation in hypothalamic AgRP neurons is required for the homeostatic regulation of energy expenditure and adiposity. These results further support previous work identifying ROCK1 as a key regulator of energy balance and suggest that targeting ROCK1 in the hypothalamus may lead to development of antiobesity therapeutics.
Collapse
Affiliation(s)
- Hu Huang
- PhD, Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, Massachusetts 02215.
| | | | | | | | | | | | | | | |
Collapse
|
135
|
Improved leptin sensitivity as a potential candidate responsible for the spontaneous food restriction of the Lou/C rat. PLoS One 2013; 8:e73452. [PMID: 24039946 PMCID: PMC3765307 DOI: 10.1371/journal.pone.0073452] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Accepted: 07/23/2013] [Indexed: 12/18/2022] Open
Abstract
The Lou/C rat, an inbred strain of Wistar origin, was described as a model of resistance to age- and diet-induced obesity. Although such a resistance involves many metabolic parameters described in our previous studies, Lou/C rats also exhibit a spontaneous food restriction due to decreased food consumption during the nocturnal period. We then attempted to delineate the leptin sensitivity and mechanisms implicated in this strain, using different protocols of acute central and peripheral leptin administration. A first analysis of the meal patterns revealed that Lou/C rats eat smaller meals, without any change in meal number compared to age-matched Wistar animals. Although the expression of the recognized leptin transporters (leptin receptors and megalin) measured in the choroid plexus was normal in Lou/C rats, the decreased triglyceridemia observed in these animals is compatible with an increased leptin transport across the blood brain barrier. Improved hypothalamic leptin signaling in Lou/C rats was also suggested by the higher pSTAT3/STAT3 (signal transducer and activator of transcription 3) ratio observed following acute peripheral leptin administration, as well as by the lower hypothalamic mRNA expression of the suppressor of cytokine signaling 3 (SOCS3), known to downregulate leptin signaling. To conclude, spontaneous hypophagia of Lou/C rats appears to be related to improved leptin sensitivity. The main mechanism underlying such a phenomenon consists in improved leptin signaling through the Ob-Rb leptin receptor isoform, which seems to consequently lead to overexpression of brain-derived neurotrophic factor (BDNF) and thyrotropin-releasing hormone (TRH).
Collapse
|
136
|
Misra M. Obesity pharmacotherapy: current perspectives and future directions. Curr Cardiol Rev 2013; 9:33-54. [PMID: 23092275 PMCID: PMC3584306 DOI: 10.2174/157340313805076322] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Revised: 07/31/2012] [Accepted: 08/27/2012] [Indexed: 02/06/2023] Open
Abstract
The rising tide of obesity and its related disorders is one of the most pressing health concerns worldwide, yet existing medicines to combat the problem are disappointingly limited in number and effectiveness. Recent advances in mechanistic insights into the neuroendocrine regulation of body weight have revealed an expanding list of molecular targets for novel, rationally designed antiobesity pharmaceutical agents. Antiobesity drugs act via any of four mechanisms: 1) decreasing energy intake, 2) increasing energy expenditure or modulating lipid metabolism, 3) modulating fat stores or adipocyte differentiation, and 4) mimicking caloric restriction. Various novel drug candidates and targets directed against obesity are currently being explored. A few of them are also in the later phases of clinical trials. This review discusses the development of novel antiobesity drugs based on current understanding of energy homeostasis
Collapse
Affiliation(s)
- Monika Misra
- Department of Pharmacology, Jawaharlal Nehru Medical College, Aligarh Muslim University, Aligarh, Uttar Pradesh, 202002, India.
| |
Collapse
|
137
|
Vitzel KF, Bikopoulos G, Hung S, Curi R, Ceddia RB. Loss of the anorexic response to systemic 5-aminoimidazole-4-carboxamide-1-β-D-ribofuranoside administration despite reducing hypothalamic AMP-activated protein kinase phosphorylation in insulin-deficient rats. PLoS One 2013; 8:e71944. [PMID: 23967267 PMCID: PMC3743807 DOI: 10.1371/journal.pone.0071944] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 07/09/2013] [Indexed: 02/07/2023] Open
Abstract
This study tested whether chronic systemic administration of 5-aminoimidazole-4-carboxamide-1-β-D-ribofuranoside (AICAR) could attenuate hyperphagia, reduce lean and fat mass losses, and improve whole-body energy homeostasis in insulin-deficient rats. Male Wistar rats were first rendered diabetic through streptozotocin (STZ) administration and then intraperitoneally injected with AICAR for 7 consecutive days. Food and water intake, ambulatory activity, and energy expenditure were assessed at the end of the AICAR-treatment period. Blood was collected for circulating leptin measurement and the hypothalami were extracted for the determination of suppressor of cytokine signaling 3 (SOCS3) content, as well as the content and phosphorylation of AMP-kinase (AMPK), acetyl-CoA carboxylase (ACC), and the signal transducer and activator of transcription 3 (STAT3). Rats were thoroughly dissected for adiposity and lean body mass (LBM) determinations. In non-diabetic rats, despite reducing adiposity, AICAR increased (∼1.7-fold) circulating leptin and reduced hypothalamic SOCS3 content and food intake by 67% and 25%, respectively. The anorexic effect of AICAR was lost in diabetic rats, even though hypothalamic AMPK and ACC phosphorylation markedly decreased in these animals. Importantly, hypothalamic SOCS3 and STAT3 levels remained elevated and reduced, respectively, after treatment of insulin-deficient rats with AICAR. Diabetic rats were lethargic and displayed marked losses of fat and LBM. AICAR treatment increased ambulatory activity and whole-body energy expenditure while also attenuating diabetes-induced fat and LBM losses. In conclusion, AICAR did not reverse hyperphagia, but it promoted anti-catabolic effects on skeletal muscle and fat, enhanced spontaneous physical activity, and improved the ability of rats to cope with the diabetes-induced dysfunctional alterations in glucose metabolism and whole-body energy homeostasis.
Collapse
Affiliation(s)
- Kaio F. Vitzel
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, SP, Brazil
| | - George Bikopoulos
- School of Kinesiology and Health Science, York University, Toronto, ON, Canada
| | - Steven Hung
- School of Kinesiology and Health Science, York University, Toronto, ON, Canada
| | - Rui Curi
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, SP, Brazil
| | - Rolando B. Ceddia
- School of Kinesiology and Health Science, York University, Toronto, ON, Canada
- * E-mail:
| |
Collapse
|
138
|
White CA, Nicola NA. SOCS3: An essential physiological inhibitor of signaling by interleukin-6 and G-CSF family cytokines. JAKSTAT 2013; 2:e25045. [PMID: 24416642 PMCID: PMC3876435 DOI: 10.4161/jkst.25045] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Accepted: 05/15/2013] [Indexed: 12/21/2022] Open
Abstract
SOCS3 is an inducible negative feedback inhibitor of cytokine signaling. Conditional deletion of SOCS3 in mice using the Cre-lox system has now been applied to a range of cell types in the steady-state and under inflammatory, pathogenic, or tumorigenic stress, with the resulting phenotypes demonstrating the effects of SOCS3 in physiological and disease contexts. Together with recent structural and biochemical studies on the mechanisms of SOCS3 binding to cytokine receptors and associated kinases, we now have a better understanding of the non-redundant roles of SOCS3 in the inhibition of cytokine signaling via the receptors gp130, G-CSFR, leptinR, and IL-12Rβ. This review discusses the known functional activities of SOCS3 in fertility and development, inflammation, innate and adaptive immunity, and malignancy as determined by genetic studies in mice.
Collapse
Affiliation(s)
- Christine A White
- Walter and Eliza Hall Institute of Medical Research; Parkville, VIC Australia ; Department of Medical Biology; University of Melbourne; Parkville, VIC Australia
| | - Nicos A Nicola
- Walter and Eliza Hall Institute of Medical Research; Parkville, VIC Australia ; Department of Medical Biology; University of Melbourne; Parkville, VIC Australia
| |
Collapse
|
139
|
Messenger SA, Moreau JM, Ciriello J. Effect of chronic intermittent hypoxia on leptin and leptin receptor protein expression in the carotid body. Brain Res 2013; 1513:51-60. [DOI: 10.1016/j.brainres.2013.03.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Revised: 03/18/2013] [Accepted: 03/20/2013] [Indexed: 01/20/2023]
|
140
|
Lu Y, Qian L, Zhang Q, Chen B, Gui L, Huang D, Chen G, Chen L. Palmitate induces apoptosis in mouse aortic endothelial cells and endothelial dysfunction in mice fed high-calorie and high-cholesterol diets. Life Sci 2013; 92:1165-73. [PMID: 23680379 DOI: 10.1016/j.lfs.2013.05.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2012] [Revised: 03/30/2013] [Accepted: 05/01/2013] [Indexed: 12/13/2022]
Abstract
AIMS Obesity is associated with hypertriglyceridemia and elevated circulating free fatty acids (FFA), resulting in endothelial dysfunction. Endoplasmic reticulum (ER) stress has been implicated in many of these processes. To determine if ER stress participates in palmitate-induced apoptosis, we investigated the effects of diet-induced obesity and palmitate on mouse aortic endothelial cells (MAEC) in vivo and in vitro. MAIN METHODS Male C57BL/6 mice were fed standard chow diets (SCD) or high-calorie and high-cholesterol diets (HCD) for 3 months. Insulin resistance was detected, and the serum, including proinflammatory indices and markers of endothelial function, was also analyzed. The ultrastructure and apoptosis of the endothelial cells in the thoracic aorta were observed. The primary MAEC were separated and treated with palmitate at different concentrations or different times respectively to observe any changes in cellular proliferation, intracellular reactive oxygen species (ROS) levels and apoptosis. Finally, the ER stress markers C/EBP homologous protein (CHOP) and glucose-regulated protein 78 (GRP78) were analyzed. KEY FINDINGS HCD-fed obese mice became inflammation-activated and insulin-resistant. Swollen mitochondria, expanded ER and apoptosis in the endothelial cells of the thoracic aorta were observed in HCD-fed mice. Palmitate inhibited cell proliferation, increased production of ROS and induced apoptosis in MAEC. CHOP was overexpressed and shifted into the nucleus (mainly), while the expression of GRP78 was upregulated in the palmitate-treated MAEC. SIGNIFICANCE Our results indicate that diet-induced obesity results in endothelial dysfunction in vivo, and that oxidative and ER stress may be involved in apoptosis induced by the palmitate in vitro.
Collapse
MESH Headings
- Animals
- Aorta, Thoracic/drug effects
- Aorta, Thoracic/metabolism
- Aorta, Thoracic/pathology
- Apoptosis/drug effects
- Apoptosis/physiology
- Cells, Cultured
- Cholesterol, Dietary/adverse effects
- Cholesterol, Dietary/metabolism
- Endoplasmic Reticulum Chaperone BiP
- Endothelial Cells/drug effects
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/pathology
- Endothelium, Vascular/physiopathology
- Energy Intake/physiology
- Male
- Mice
- Mice, Inbred C57BL
- Obesity/metabolism
- Obesity/pathology
- Palmitic Acid/toxicity
- Random Allocation
Collapse
Affiliation(s)
- Yunxia Lu
- Department of Biochemistry and Molecular Biology, Anhui Medical University, Hefei, Anhui 230032, PR China.
| | | | | | | | | | | | | | | |
Collapse
|
141
|
Abstract
Leptin is secreted into the bloodstream by adipocytes and is required for the maintenance of energy homeostasis and body weight. Leptin deficiency or genetic defects in the components of the leptin signaling pathways cause obesity. Leptin controls energy balance and body weight mainly through leptin receptor b (LEPRb)-expressing neurons in the brain, particularly in the hypothalamus. These LEPRb-expressing neurons function as the first-order neurons that project to the second-order neurons located within and outside the hypothalamus, forming a neural network that controls the energy homeostasis and body weight. Multiple factors, including inflammation and endoplasmic reticulum (ER) stress, contribute to leptin resistance. Leptin resistance is the key risk factor for obesity. This review is focused on recent advance about leptin action, leptin signaling, and leptin resistance.
Collapse
|
142
|
Nazarians-Armavil A, Menchella JA, Belsham DD. Cellular insulin resistance disrupts leptin-mediated control of neuronal signaling and transcription. Mol Endocrinol 2013; 27:990-1003. [PMID: 23579487 DOI: 10.1210/me.2012-1338] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Central resistance to the actions of insulin and leptin is associated with the onset of obesity and type 2 diabetes mellitus, whereas leptin and insulin signaling is essential for both glucose and energy homeostasis. Although it is known that leptin resistance can lead to attenuated insulin signaling, whether insulin resistance can lead to or exacerbate leptin resistance is unknown. To investigate the molecular events underlying crosstalk between these signaling pathways, immortalized hypothalamic neuronal models, rHypoE-19 and mHypoA-2/10, were used. Prolonged insulin exposure was used to induce cellular insulin resistance, and thereafter leptin-mediated regulation of signal transduction and gene expression was assessed. Leptin directly repressed agouti-related peptide mRNA levels but induced urocortin-2, insulin receptor substrate (IRS)-1, IRS2, and IR transcription, through leptin-mediated phosphatidylinositol 3-kinase/Akt activation. Neuronal insulin resistance, as assessed by attenuated Akt phosphorylation, blocked leptin-mediated signal transduction and agouti-related peptide, urocortin-2, IRS1, IRS2, and insulin receptor synthesis. Insulin resistance caused a substantial decrease in insulin receptor protein levels, forkhead box protein 1 phosphorylation, and an increase in suppressor of cytokine signaling 3 protein levels. Cellular insulin resistance may cause or exacerbate neuronal leptin resistance and, by extension, obesity. It is essential to unravel the effects of neuronal insulin resistance given that both peripheral, as well as the less widely studied central insulin resistance, may contribute to the development of metabolic, reproductive, and cardiovascular disorders. This study provides improved understanding of the complex cellular crosstalk between insulin-leptin signal transduction that is disrupted during neuronal insulin resistance.
Collapse
Affiliation(s)
- Anaies Nazarians-Armavil
- Department of Physiology, University of Toronto, Medical Sciences Building 3247A, 1 Kings College Circle, Toronto, Ontario, Canada M5S 1A8
| | | | | |
Collapse
|
143
|
Stolarczyk E, Vong C, Perucha E, Jackson I, Cawthorne M, Wargent E, Powell N, Canavan J, Lord G, Howard J. Improved insulin sensitivity despite increased visceral adiposity in mice deficient for the immune cell transcription factor T-bet. Cell Metab 2013; 17:520-33. [PMID: 23562076 PMCID: PMC3685808 DOI: 10.1016/j.cmet.2013.02.019] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Revised: 12/18/2012] [Accepted: 02/27/2013] [Indexed: 12/19/2022]
Abstract
Low-grade inflammation in fat is associated with insulin resistance, although the mechanisms are unclear. We report that mice deficient in the immune cell transcription factor T-bet have lower energy expenditure and increased visceral fat compared with wild-type mice, yet paradoxically are more insulin sensitive. This striking phenotype, present in young T-bet(-/-) mice, persisted with high-fat diet and increasing host age and was associated with altered immune cell numbers and cytokine secretion specifically in visceral adipose tissue. However, the favorable metabolic phenotype observed in T-bet-deficient hosts was lost in T-bet(-/-) mice also lacking adaptive immunity (T-bet(-/-)xRag2(-/-)), demonstrating that T-bet expression in the adaptive rather than the innate immune system impacts host glucose homeostasis. Indeed, adoptive transfer of T-bet-deficient, but not wild-type, CD4(+) T cells to Rag2(-/-) mice improved insulin sensitivity. Our results reveal a role for T-bet in metabolic physiology and obesity-associated insulin resistance.
Collapse
Affiliation(s)
- Emilie Stolarczyk
- Division of Diabetes and Nutritional Sciences, King’s College London, London SE1 9RT, UK
| | - Chi Teng Vong
- Division of Diabetes and Nutritional Sciences, King’s College London, London SE1 9RT, UK
| | - Esperanza Perucha
- Department of Experimental Immunobiology, King’s College London, London SE1 9RT, UK
| | - Ian Jackson
- Department of Experimental Immunobiology, King’s College London, London SE1 9RT, UK
| | | | | | - Nick Powell
- Department of Experimental Immunobiology, King’s College London, London SE1 9RT, UK
- Centre for Immunology and Infectious Disease, Blizard Institute of Cell and Molecular Science, Bart’s and The London School of Medicine and Dentistry, London E1 2AT, UK
| | - James B. Canavan
- Department of Experimental Immunobiology, King’s College London, London SE1 9RT, UK
- Centre for Immunology and Infectious Disease, Blizard Institute of Cell and Molecular Science, Bart’s and The London School of Medicine and Dentistry, London E1 2AT, UK
| | - Graham M. Lord
- Department of Experimental Immunobiology, King’s College London, London SE1 9RT, UK
| | - Jane K. Howard
- Division of Diabetes and Nutritional Sciences, King’s College London, London SE1 9RT, UK
- Corresponding author
| |
Collapse
|
144
|
Abstract
Obesity is characterized by a chronic and low-grade inflammation in tissues including the hypothalamus. Hypothalamic inflammation is considered an early and determining factor for the onset of obesity, a factor that occurs even before body weight gain. Within the hypothalamus, microglia and astrocytes produce cytokines that drive inflammatory responses. Astrocytes are directly affected by nutrient excess and might play a unique role in promoting hypothalamic inflammatory responses in obesity. This article reviews evidence supporting the role of hypothalamic astrocytes in obesity, and suggests a new approach for neuroendocrine research designed to reveal pathogenesis and develop novel treatment strategies against obesity.
Collapse
Affiliation(s)
- Cristina García-Cáceres
- Institute for Diabetes and Obesity, Helmholtz Centre Munich, 85748 Garching, Munich, Germany
| | | | | |
Collapse
|
145
|
Implications of leptin in neuroendocrine regulation of male reproduction. Reprod Biol 2013; 13:1-14. [DOI: 10.1016/j.repbio.2012.12.001] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Revised: 12/20/2012] [Accepted: 12/21/2012] [Indexed: 01/14/2023]
|
146
|
Inactivation of Socs3 in the hypothalamus enhances the hindbrain response to endogenous satiety signals via oxytocin signaling. J Neurosci 2013. [PMID: 23197703 DOI: 10.1523/jneurosci.1669-12.2012] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Leptin is an adipocyte-derived hormone that controls energy balance by acting primarily in the CNS, but its action is lost in common forms of obesity due to central leptin resistance. One potential mechanism for such leptin resistance is an increased hypothalamic expression of Suppressor of cytokine signaling 3 (Socs3), a feedback inhibitor of the Jak-Stat pathway that prevents Stat3 activation. Ample studies have confirmed the important role of Socs3 in leptin resistance and obesity. However, the degree to which Socs3 participates in the regulation of energy homeostasis in nonobese conditions remains largely undetermined. In this study, using adult mice maintained under standard diet, we demonstrate that Socs3 deficiency in the mediobasal hypothalamus (MBH) reduces food intake, protects against body weight gain, and limits adiposity, suggesting that Socs3 is necessary for normal body weight maintenance. Mechanistically, MBH Socs3-deficient mice display increased hindbrain sensitivity to endogenous, meal-related satiety signals, mediated by oxytocin signaling. Thus, oxytocin signaling likely mediates the effect of hypothalamic leptin on satiety circuits of the caudal brainstem. This provides an anatomical substrate for the effect of leptin on meal size, and more generally, a mechanism for how the brain controls short-term food intake as a function of the energetic stores available in the organism to maintain energy homeostasis. Any dysfunction in this pathway could potentially lead to overeating and obesity.
Collapse
|
147
|
Modulation of AgRP-neuronal function by SOCS3 as an initiating event in diet-induced hypothalamic leptin resistance. Proc Natl Acad Sci U S A 2013; 110:E697-706. [PMID: 23386726 DOI: 10.1073/pnas.1218284110] [Citation(s) in RCA: 110] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Chronic consumption of a fat-rich diet leads to attenuation of leptin signaling in hypothalamic neurons, a hallmark feature of cellular leptin resistance. To date, little is known about the temporal and spatial dysregulation of neuronal function under conditions of nutrient excess. We show that agouti-related protein (AgRP)-expressing neurons precede proopiomelanocortin neurons in developing diet-induced cellular leptin resistance. High-fat diet-induced up-regulation of suppressor of cytokine signaling-3 (SOCS3) occurs in AgRP neurons before proopiomelanocortin and other hypothalamic neurons. SOCS3 expression in AgRP neurons increases after 2 d of high-fat feeding, but reduces after switching to a low-fat diet for 1 d. Consistently, transgenic overexpression of SOCS3 in AgRP neurons produces metabolic phenotypes resembling those observed after short-term high-fat feeding. We further show that AgRP neurons are the predominant cell type situated outside the blood-brain barrier in the mediobasal hypothalamus. AgRP neurons are more responsive to low levels of circulating leptin, but they are also more prone to development of leptin resistance in response to a small increase in blood leptin concentrations. Collectively, these results suggest that AgRP neurons are able to sense slight changes in plasma metabolic signals, allowing them to serve as first-line responders to fluctuation of energy intake. Furthermore, modulation of SOCS3 expression in AgRP neurons may play a dynamic and physiological role in metabolic fine tuning in response to short-term changes of nutritional status.
Collapse
|
148
|
Jung CH, Kim MS. Molecular mechanisms of central leptin resistance in obesity. Arch Pharm Res 2013; 36:201-7. [DOI: 10.1007/s12272-013-0020-y] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2012] [Accepted: 12/24/2012] [Indexed: 02/06/2023]
|
149
|
Apolzan JW, Harris RBS. Rapid onset and reversal of peripheral and central leptin resistance in rats offered chow, sucrose solution, and lard. Appetite 2013; 60:65-73. [PMID: 23022555 PMCID: PMC3996830 DOI: 10.1016/j.appet.2012.09.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Revised: 09/03/2012] [Accepted: 09/17/2012] [Indexed: 12/23/2022]
Abstract
We previously reported that rats offered choice diet (chow, 30% sucrose solution, lard) increase body fat by 130% within 3 weeks. We tested the effects of choice diet on the development of leptin resistance in rats. Intraperitoneal injection of 2mg/kg leptin inhibited 14 h food intake and weight gain of all rats after 2 days and 4 days of diet. On day 8, choice rats were leptin insensitive and by day 16 they were resistant. Chow rats remained leptin responsive. A second study showed that on day 16 choice, but not chow rats, were centrally leptin resistant (1.5 μg leptin, 3rd ventricle). In both studies, rats were switched back to chow only after approximately 3 weeks on choice diet and were leptin responsive after 4 days. A third study showed that carcass fat was reduced by 30% 4 days after switching back to chow. A final experiment showed that leptin responsive chow rats, but not leptin resistant choice rats, increased energy expenditure by 12% during the 2.6h after a central leptin injection. Thus, choice diet rapidly induces leptin resistance, but leptin responsiveness is quickly restored when choice is replaced with chow. This rapid onset and reversal of leptin resistance may be associated with changes in either substrate metabolism or adiposity.
Collapse
Affiliation(s)
- John W Apolzan
- Department of Physiology, Georgia Health Sciences University, 1120 15th Street, Augusta, GA 30912, USA; Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA(1).
| | - Ruth B S Harris
- Department of Physiology, Georgia Health Sciences University, 1120 15th Street, Augusta, GA 30912, USA
| |
Collapse
|
150
|
Sarvas JL, Khaper N, Lees SJ. The IL-6 Paradox: Context Dependent Interplay of SOCS3 and AMPK. ACTA ACUST UNITED AC 2013; Suppl 13. [PMID: 24244888 DOI: 10.4172/2155-6156.s13-003] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Insulin resistance is the principle step towards the progression of type 2 diabetes, and has been linked to increased circulating levels of cytokines, leading to chronic low-grade inflammation. Specifically, in chronic disease states increased IL-6 is thought to play a critical role in the regulation of insulin resistance in the peripheral tissues, and has been used as a marker of insulin resistance. There is also an endogenous up-regulation of IL-6 in response to exercise, which has been linked to improved insulin sensitivity. This leads to the question "how can elevated IL-6 lead to the development of insulin resistance, and yet also lead to increased insulin sensitivity?" Resolving the dual role of IL-6 in regulating insulin resistance/sensitivity is critical to the development of potential therapeutic interventions. This review summarizes the literature on the seemingly paradoxical role of elevated IL-6 on insulin signalling, including the activation of AMPK and the involvement of leptin and SOCS3.
Collapse
|