51
|
Muthuramu I, Amin R, Postnov A, Mishra M, Jacobs F, Gheysens O, Van Veldhoven PP, De Geest B. Coconut Oil Aggravates Pressure Overload-Induced Cardiomyopathy without Inducing Obesity, Systemic Insulin Resistance, or Cardiac Steatosis. Int J Mol Sci 2017; 18:ijms18071565. [PMID: 28718833 PMCID: PMC5536053 DOI: 10.3390/ijms18071565] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 07/05/2017] [Accepted: 07/15/2017] [Indexed: 12/16/2022] Open
Abstract
Studies evaluating the effects of high-saturated fat diets on cardiac function are most often confounded by diet-induced obesity and by systemic insulin resistance. We evaluated whether coconut oil, containing C12:0 and C14:0 as main fatty acids, aggravates pressure overload-induced cardiomyopathy induced by transverse aortic constriction (TAC) in C57BL/6 mice. Mortality rate after TAC was higher (p < 0.05) in 0.2% cholesterol 10% coconut oil diet-fed mice than in standard chow-fed mice (hazard ratio 2.32, 95% confidence interval 1.16 to 4.64) during eight weeks of follow-up. The effects of coconut oil on cardiac remodeling occurred in the absence of weight gain and of systemic insulin resistance. Wet lung weight was 1.76-fold (p < 0.01) higher in coconut oil mice than in standard chow mice. Myocardial capillary density (p < 0.001) was decreased, interstitial fibrosis was 1.88-fold (p < 0.001) higher, and systolic and diastolic function was worse in coconut oil mice than in standard chow mice. Myocardial glucose uptake was 1.86-fold (p < 0.001) higher in coconut oil mice and was accompanied by higher myocardial pyruvate dehydrogenase levels and higher acetyl-CoA carboxylase levels. The coconut oil diet increased oxidative stress. Myocardial triglycerides and free fatty acids were lower (p < 0.05) in coconut oil mice. In conclusion, coconut oil aggravates pressure overload-induced cardiomyopathy.
Collapse
Affiliation(s)
- Ilayaraja Muthuramu
- Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, Catholic University of Leuven, Leuven 3000, Belgium.
| | - Ruhul Amin
- Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, Catholic University of Leuven, Leuven 3000, Belgium.
| | - Andrey Postnov
- Nuclear Medicine & Molecular Imaging, Department of Imaging & Pathology, Catholic University of Leuven, Leuven 3000, Belgium.
| | - Mudit Mishra
- Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, Catholic University of Leuven, Leuven 3000, Belgium.
| | - Frank Jacobs
- Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, Catholic University of Leuven, Leuven 3000, Belgium.
| | - Olivier Gheysens
- Nuclear Medicine & Molecular Imaging, Department of Imaging & Pathology, Catholic University of Leuven, Leuven 3000, Belgium.
| | - Paul P Van Veldhoven
- Laboratory of Lipid Biochemistry and Protein Interactions, Department of Cellular and Molecular Medicine, Catholic University of Leuven, Leuven 3000, Belgium.
| | - Bart De Geest
- Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, Catholic University of Leuven, Leuven 3000, Belgium.
| |
Collapse
|
52
|
Yan L, Sundaram S, Nielsen FH. Voluntary running of defined distances reduces body adiposity and its associated inflammation in C57BL/6 mice fed a high-fat diet. Appl Physiol Nutr Metab 2017; 42:1179-1184. [PMID: 28715640 DOI: 10.1139/apnm-2017-0285] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
This study investigated the effect of voluntary running of defined distances on body adiposity in male C57BL/6 mice fed a high-fat diet. Mice were assigned to 6 groups and fed a standard AIN93G diet (sedentary) or a modified high-fat AIN93G diet (sedentary; unrestricted running; or 75%, 50%, or 25% of unrestricted running) for 12 weeks. The average running distance was 8.3, 6.3, 4.2, and 2.1 km/day for the unrestricted, 75%, 50%, and 25% of unrestricted runners, respectively. Body adiposity was 46% higher in sedentary mice when fed the high-fat diet instead of the standard diet. Running decreased adiposity in mice fed the high-fat diet in a dose-dependent manner but with no significant difference between sedentary mice and those running 2.1 km/day. In sedentary mice, the high-fat instead of the standard diet increased insulin resistance, hepatic triacylglycerides, and adipose and plasma concentrations of leptin and monocyte chemotactic protein-1 (MCP-1). Running reduced these variables in a dose-dependent manner. Adipose adiponectin was lowest in sedentary mice fed the high-fat diet; running raised adiponectin in both adipose tissue and plasma. Running 8.3 and 6.3 km/day had the greatest, but similar, effects on the aforementioned variables. Running 2.1 km/day did not affect these variables except, when compared with sedentariness, it significantly decreased MCP-1. The findings showed that running 6.3 kg/day was optimal for reducing adiposity and associated inflammation that was increased in mice by feeding a high-fat diet. The findings suggest that voluntary running of defined distances may counteract the obesogenic effects of a high-fat diet.
Collapse
Affiliation(s)
- Lin Yan
- U.S. Department of Agriculture, Agricultural Research Service, Grand Forks Human Nutrition Research Center, 2420 2nd Avenue North, Grand Forks, ND 58202, USA.,U.S. Department of Agriculture, Agricultural Research Service, Grand Forks Human Nutrition Research Center, 2420 2nd Avenue North, Grand Forks, ND 58202, USA
| | - Sneha Sundaram
- U.S. Department of Agriculture, Agricultural Research Service, Grand Forks Human Nutrition Research Center, 2420 2nd Avenue North, Grand Forks, ND 58202, USA.,U.S. Department of Agriculture, Agricultural Research Service, Grand Forks Human Nutrition Research Center, 2420 2nd Avenue North, Grand Forks, ND 58202, USA
| | - Forrest H Nielsen
- U.S. Department of Agriculture, Agricultural Research Service, Grand Forks Human Nutrition Research Center, 2420 2nd Avenue North, Grand Forks, ND 58202, USA.,U.S. Department of Agriculture, Agricultural Research Service, Grand Forks Human Nutrition Research Center, 2420 2nd Avenue North, Grand Forks, ND 58202, USA
| |
Collapse
|
53
|
Costa R, Rodrigues I, Guardão L, Rocha-Rodrigues S, Silva C, Magalhães J, Ferreira-de-Almeida M, Negrão R, Soares R. Xanthohumol and 8-prenylnaringenin ameliorate diabetic-related metabolic dysfunctions in mice. J Nutr Biochem 2017; 45:39-47. [DOI: 10.1016/j.jnutbio.2017.03.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 01/31/2017] [Accepted: 03/16/2017] [Indexed: 01/12/2023]
|
54
|
An Increased Dietary Supply of Medium-Chain Fatty Acids during Early Weaning in Rodents Prevents Excessive Fat Accumulation in Adulthood. Nutrients 2017. [PMID: 28632178 PMCID: PMC5490610 DOI: 10.3390/nu9060631] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Medium-chain fatty acids (MCFA) are a directly and readily absorbed source of energy. Exposure early-in-life to increased MCFA levels might affect development and impact (lipid) metabolism later in life. We tested whether an increased MCFA intake early-in-life positively affects adult body composition and metabolic status when challenged by a western-style diet (WSD). Male offspring of C57Bl/6j mice and Wistar rats were fed a control diet (CTRL; 10 w% fat, 14% MCFA) or a medium-chain triglycerides (MCT) diet with 20% MCFA until postnatal (PN) day 42, whereupon animals were fed a WSD (10 w% fat) until PN day 98. Body composition was monitored by Dual Energy X-ray Absorptiometry (DEXA). In rats, glucose homeostasis was assessed by glucose tolerance test (GTT) and insulin tolerance test (ITT); in mice, the HOmeostasis Model Assessment of Insulin Resistance (HOMA-IR) was calculated. At autopsy on PN day 98, plasma lipid profiles, glucose, insulin, and adipokines were measured; organs and fat pads were collected and the adipocyte size distribution was analysed. Milk analysis in mice showed that the maternal MCT diet was not translated into milk, and pups were thus only exposed to high MCT levels from early weaning onward: PN day 16 until 42. Mice exposed to MCT showed 28% less fat accumulation vs. CTRL during WSD. The average adipocyte cell size, fasting plasma triglycerides (TG), and leptin levels were reduced in MCT mice. In rats, no effects were found on the adult body composition, but the adipocyte cell size distribution shifted towards smaller adipocytes. Particularly mice showed positive effects on glucose homeostasis and insulin sensitivity. Increased MCFA intake early-in-life protected against the detrimental effects of an obesogenic diet in adulthood.
Collapse
|
55
|
Murphy AM, Thomas A, Crinion SJ, Kent BD, Tambuwala MM, Fabre A, Pepin JL, Roche HM, Arnaud C, Ryan S. Intermittent hypoxia in obstructive sleep apnoea mediates insulin resistance through adipose tissue inflammation. Eur Respir J 2017; 49:49/4/1601731. [PMID: 28424360 DOI: 10.1183/13993003.01731-2016] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 12/20/2016] [Indexed: 11/05/2022]
Abstract
Obstructive sleep apnoea (OSA) is increasingly associated with insulin resistance. The underlying pathophysiology remains unclear but intermittent hypoxia (IH)-mediated inflammation and subsequent dysfunction of the adipose tissue has been hypothesised to play a key role.We tested this hypothesis employing a comprehensive translational approach using a murine IH model of lean and diet-induced obese mice, an innovative IH system for cell cultures and a tightly controlled patient cohort.IH led to the development of insulin resistance in mice, corrected for the degree of obesity, and reduced insulin-mediated glucose uptake in 3T3-L1 adipocytes, associated with inhibition of the insulin-signalling pathway and downregulation of insulin-receptor substrate-1 mRNA. Providing mechanistic insight, IH induced a pro-inflammatory phenotype of visceral adipose tissue in mice with pro-inflammatory M1 macrophage polarisation correlating with the severity of insulin resistance. Complimentary in vitro analysis demonstrated that IH led to M1 polarisation of THP1-derived macrophages. In subjects without comorbidities (n=186), OSA was independently associated with insulin resistance. Furthermore, we found an independent correlation of OSA severity with the M1 macrophage inflammatory marker sCD163.This study provides evidence that IH induces a pro-inflammatory phenotype of the adipose tissue, which may be a crucial link between OSA and the development of insulin resistance.
Collapse
Affiliation(s)
- Aoife M Murphy
- Nutrigenomics Research Group, School of Public Health, Physiotherapy and Sports Science, Conway Institute, University College Dublin, Dublin, Ireland
| | - Amandine Thomas
- Université Grenoble Alpes, HP2, Inserm, U1042, CHU de Grenoble, Laboratoire EFCR, Pôle Thorax et Vaisseaux Grenoble, Grenoble, France
| | - Sophie J Crinion
- Pulmonary and Sleep Disorders Unit, St Vincent's University Hospital, Dublin, Ireland
| | - Brian D Kent
- Pulmonary and Sleep Disorders Unit, Guy's and St Thomas' Hospital, London, UK
| | - Murtaza M Tambuwala
- School of Pharmacy and Pharmaceutical Science, Ulster University, Coleraine, UK
| | - Aurelie Fabre
- Dept of Pathology, St Vincent's University Hospital, Dublin, Ireland.,Research Pathology Core Technology, Conway Institute, University College Dublin, Dublin, Ireland
| | - Jean-Louis Pepin
- Université Grenoble Alpes, HP2, Inserm, U1042, CHU de Grenoble, Laboratoire EFCR, Pôle Thorax et Vaisseaux Grenoble, Grenoble, France
| | - Helen M Roche
- Nutrigenomics Research Group, School of Public Health, Physiotherapy and Sports Science, Conway Institute, University College Dublin, Dublin, Ireland
| | - Claire Arnaud
- Université Grenoble Alpes, HP2, Inserm, U1042, CHU de Grenoble, Laboratoire EFCR, Pôle Thorax et Vaisseaux Grenoble, Grenoble, France
| | - Silke Ryan
- Pulmonary and Sleep Disorders Unit, St Vincent's University Hospital, Dublin, Ireland .,School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland
| |
Collapse
|
56
|
Petersen PS, Lei X, Wolf RM, Rodriguez S, Tan SY, Little HC, Schweitzer MA, Magnuson TH, Steele KE, Wong GW. CTRP7 deletion attenuates obesity-linked glucose intolerance, adipose tissue inflammation, and hepatic stress. Am J Physiol Endocrinol Metab 2017; 312:E309-E325. [PMID: 28223291 PMCID: PMC5406989 DOI: 10.1152/ajpendo.00344.2016] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 01/10/2017] [Accepted: 02/06/2017] [Indexed: 12/22/2022]
Abstract
Chronic low-grade inflammation and cellular stress are important contributors to obesity-linked metabolic dysfunction. Here, we uncover an immune-metabolic role for C1q/TNF-related protein 7 (CTRP7), a secretory protein of the C1q family with previously unknown function. In obese humans, circulating CTRP7 levels were markedly elevated and positively correlated with body mass index, glucose, insulin, insulin resistance index, hemoglobin A1c, and triglyceride levels. Expression of CTRP7 in liver was also significantly upregulated in obese humans and positively correlated with gluconeogenic genes. In mice, Ctrp7 expression was differentially modulated in various tissues by fasting and refeeding and by diet-induced obesity. A genetic loss-of-function mouse model was used to determine the requirement of CTRP7 for metabolic homeostasis. When fed a control low-fat diet, male or female mice lacking CTRP7 were indistinguishable from wild-type littermates. In obese male mice consuming a high-fat diet, however, CTRP7 deficiency attenuated insulin resistance and enhanced glucose tolerance, effects that were independent of body weight, metabolic rate, and physical activity level. Improved glucose metabolism in CTRP7-deficient mice was associated with reduced adipose tissue inflammation, as well as decreased liver fibrosis and cellular oxidative and endoplasmic reticulum stress. These results provide a link between elevated CTRP7 levels and impaired glucose metabolism, frequently associated with obesity. Inhibiting CTRP7 action may confer beneficial metabolic outcomes in the setting of obesity and diabetes.
Collapse
Affiliation(s)
- Pia S Petersen
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Xia Lei
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Risa M Wolf
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland; and
| | - Susana Rodriguez
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Stefanie Y Tan
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Hannah C Little
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Michael A Schweitzer
- Department of Surgery, Johns Hopkins Center for Bariatric Surgery, Johns Hopkins Bayview Medical Center, Baltimore, Maryland
| | - Thomas H Magnuson
- Department of Surgery, Johns Hopkins Center for Bariatric Surgery, Johns Hopkins Bayview Medical Center, Baltimore, Maryland
| | - Kimberley E Steele
- Department of Surgery, Johns Hopkins Center for Bariatric Surgery, Johns Hopkins Bayview Medical Center, Baltimore, Maryland
| | - G William Wong
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland;
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
57
|
Relationship between the indexes of insulin resistance and metabolic status in dairy cows during early lactation. ACTA VET-BEOGRAD 2017. [DOI: 10.1515/acve-2017-0006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Abstract
Insulin resistance is a phenomenon which accompanies the ongoing metabolic adaptation in cows during early lactation. The aim of our study was to determine the linear correlations of HOMA (Homeostatic Model Assessment), QUICKI (Quantitative Insulin Sensitivity Check Index) and RQUICKI (Revised Quantitative Insulin Sensitivity Check Index) indexes of insulin resistance with the metabolic status of cows (concentration of hormones, metabolites and body condition score). The experiment included 40 Holstein-Frisian cows in the first week after calving. Indexes of insulin resistance valued: 18.68±5.43 (HOMA), 0.39±0.06 (QUICKI) and 0.45±0.06 (RQUICKI). Linear correlations were examined by testing the coefficient of correlation (r), determination (r2,%) and regression parameter beta (b) in linear equation. A negative correlation was found between HOMA and IGF-I (insulin growth factor I) (r=−0.51, r2=25.0, b=−1.1257, p<0.01). HOMA showed a positive correlation with BHB (betahidroxybutyrate) (r=0.48, r2=23.2, b=0.0234, p<0.01). A positive correlation was found between QUICKI and IGF-I (r=0.30, r2=10.0 b=46.7900, p<0.05) and cholesterol (r=0.44, r2=18.3, b=1.9021, p<0.01). In contrast, QUICKI and BHB (r=0.51, r2=27.1, b=−1.7241, p<0.01), just like QUICKI and BCS (r=0.46, r2=20.9, b=−2.424, p<0.01), showed a negative correlation. RQUICKI showed positive correlations with IGF-I (r=0.48, r2=22.8, b=28.1230, p<0.01), T4 (r=0.47, r2=22.1, b=87.142, p<0.01) and triglycerides (r=0.36, r2=13, b=0.0407, p<0.05) but negative correlations with cortisol (r=−0.36, r2=13.0, b=−9.0332, p<0.05), STH (somatotropic hormone) (r=−0.42, r2=17.3, b=−5.4976, p<0.01), BHB (r=−0.62, r2=38.3, b=−1.1872, p<0.01), total bilirubin (r=−0.58, r2=33.7, b=−7.131, p<0.01) and BCS (body condition score) (r=−0.6, r2=36.4, b=−1.8347, p<0.01). In conclusion, indexes of insulin resistance may be used to evaluate the metabolic status of cows in early lactation. RQUICKI might be the most appropriate predictor of metabolic status due to its linear relationship with most of the parameters included in homeorhetic process.
Collapse
|
58
|
Sangüesa G, Baena M, Hutter N, Montañés JC, Sánchez RM, Roglans N, Laguna JC, Alegret M. The Addition of Liquid Fructose to a Western-Type Diet in LDL-R -/- Mice Induces Liver Inflammation and Fibrogenesis Markers without Disrupting Insulin Receptor Signalling after an Insulin Challenge. Nutrients 2017; 9:nu9030278. [PMID: 28294959 PMCID: PMC5372941 DOI: 10.3390/nu9030278] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 03/03/2017] [Accepted: 03/09/2017] [Indexed: 01/17/2023] Open
Abstract
A high consumption of fat and simple sugars, especially fructose, has been related to the development of insulin resistance, but the mechanisms involved in the effects of these nutrients are not fully understood. This study investigates the effects of a Western-type diet and liquid fructose supplementation, alone and combined, on insulin signalling and inflammation in low-density lipoprotein (LDL) receptor-deficient mice (LDL-R−/−). LDL-R−/− mice were fed chow or Western diet ±15% fructose solution for 12 weeks. Plasma glucose and insulin, and the expression of genes related to inflammation in the liver and visceral white adipose tissue (vWAT), were analysed. V-akt murine thymoma viral oncogene homolog-2 (Akt) activation was measured in the liver of the mice after a single injection of saline or insulin. None of the dietary interventions caused inflammation in vWAT, whereas the Western diet induced hepatic inflammation, which was further enhanced by liquid fructose, leading also to a significant increase in fibrogenesis markers. However, there was no change in plasma glucose or insulin, or insulin-induced Akt phosphorylation. In conclusion, hepatic inflammation and fibrogenesis markers induced by a Western diet supplemented with liquid fructose in LDL-R−/− mice are not associated with a significant impairment of hepatic insulin signalling.
Collapse
Affiliation(s)
- Gemma Sangüesa
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, School of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain.
- Institute of Biomedicine, University of Barcelona, 08028 Barcelona, Spain.
| | - Miguel Baena
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, School of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain.
- Institute of Biomedicine, University of Barcelona, 08028 Barcelona, Spain.
| | - Natalia Hutter
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, School of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain.
| | - José Carlos Montañés
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, School of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain.
| | - Rosa María Sánchez
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, School of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain.
- Institute of Biomedicine, University of Barcelona, 08028 Barcelona, Spain.
- CIBER Fisiología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Spain.
| | - Núria Roglans
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, School of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain.
- Institute of Biomedicine, University of Barcelona, 08028 Barcelona, Spain.
- CIBER Fisiología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Spain.
| | - Juan Carlos Laguna
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, School of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain.
- Institute of Biomedicine, University of Barcelona, 08028 Barcelona, Spain.
- CIBER Fisiología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Spain.
| | - Marta Alegret
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, School of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain.
- Institute of Biomedicine, University of Barcelona, 08028 Barcelona, Spain.
- CIBER Fisiología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Spain.
| |
Collapse
|
59
|
Morettini M, Faelli E, Perasso L, Fioretti S, Burattini L, Ruggeri P, Di Nardo F. IVGTT-based simple assessment of glucose tolerance in the Zucker fatty rat: Validation against minimal models. PLoS One 2017; 12:e0173200. [PMID: 28264067 PMCID: PMC5338807 DOI: 10.1371/journal.pone.0173200] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 02/16/2017] [Indexed: 12/29/2022] Open
Abstract
For the assessment of glucose tolerance from IVGTT data in Zucker rat, minimal model methodology is reliable but time- and money-consuming. This study aimed to validate for the first time in Zucker rat, simple surrogate indexes of insulin sensitivity and secretion against the glucose-minimal-model insulin sensitivity index (SI) and against first- (Φ1) and second-phase (Φ2) β-cell responsiveness indexes provided by C-peptide minimal model. Validation of the surrogate insulin sensitivity index (ISI) and of two sets of coupled insulin-based indexes for insulin secretion, differing from the cut-off point between phases (FPIR3-SPIR3, t = 3 min and FPIR5-SPIR5, t = 5 min), was carried out in a population of ten Zucker fatty rats (ZFR) and ten Zucker lean rats (ZLR). Considering the whole rat population (ZLR+ZFR), ISI showed a significant strong correlation with SI (Spearman's correlation coefficient, r = 0.88; P<0.001). Both FPIR3 and FPIR5 showed a significant (P<0.001) strong correlation with Φ1 (r = 0.76 and r = 0.75, respectively). Both SPIR3 and SPIR5 showed a significant (P<0.001) strong correlation with Φ2 (r = 0.85 and r = 0.83, respectively). ISI is able to detect (P<0.001) the well-recognized reduction in insulin sensitivity in ZFRs, compared to ZLRs. The insulin-based indexes of insulin secretion are able to detect in ZFRs (P<0.001) the compensatory increase of first- and second-phase secretion, associated to the insulin-resistant state. The ability of the surrogate indexes in describing glucose tolerance in the ZFRs was confirmed by the Disposition Index analysis. The model-based validation performed in the present study supports the utilization of low-cost, insulin-based indexes for the assessment of glucose tolerance in Zucker rat, reliable animal model of human metabolic syndrome.
Collapse
Affiliation(s)
- Micaela Morettini
- Department of Information Engineering, Università Politecnica delle Marche, Ancona, Italy
| | - Emanuela Faelli
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Luisa Perasso
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Sandro Fioretti
- Department of Information Engineering, Università Politecnica delle Marche, Ancona, Italy
| | - Laura Burattini
- Department of Information Engineering, Università Politecnica delle Marche, Ancona, Italy
| | - Piero Ruggeri
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Francesco Di Nardo
- Department of Information Engineering, Università Politecnica delle Marche, Ancona, Italy
| |
Collapse
|
60
|
Bindels LB, Segura Munoz RR, Gomes-Neto JC, Mutemberezi V, Martínez I, Salazar N, Cody EA, Quintero-Villegas MI, Kittana H, de Los Reyes-Gavilán CG, Schmaltz RJ, Muccioli GG, Walter J, Ramer-Tait AE. Resistant starch can improve insulin sensitivity independently of the gut microbiota. MICROBIOME 2017; 5:12. [PMID: 28166818 PMCID: PMC5294823 DOI: 10.1186/s40168-017-0230-5] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 01/09/2017] [Indexed: 05/13/2023]
Abstract
BACKGROUND Obesity-related diseases, including type 2 diabetes and cardiovascular disease, have reached epidemic proportions in industrialized nations, and dietary interventions for their prevention are therefore important. Resistant starches (RS) improve insulin sensitivity in clinical trials, but the mechanisms underlying this health benefit remain poorly understood. Because RS fermentation by the gut microbiota results in the formation of physiologically active metabolites, we chose to specifically determine the role of the gut microbiota in mediating the metabolic benefits of RS. To achieve this goal, we determined the effects of RS when added to a Western diet on host metabolism in mice with and without a microbiota. RESULTS RS feeding of conventionalized mice improved insulin sensitivity and redressed some of the Western diet-induced changes in microbiome composition. However, parallel experiments in germ-free littermates revealed that RS-mediated improvements in insulin levels also occurred in the absence of a microbiota. RS reduced gene expression of adipose tissue macrophage markers and altered cecal concentrations of several bile acids in both germ-free and conventionalized mice; these effects were strongly correlated with the metabolic benefits, providing a potential microbiota-independent mechanism to explain the physiological effects of RS. CONCLUSIONS This study demonstrated that some metabolic benefits exerted by dietary RS, especially improvements in insulin levels, occur independently of the microbiota and could involve alterations in the bile acid cycle and adipose tissue immune modulation. This work also sets a precedent for future mechanistic studies aimed at establishing the causative role of the gut microbiota in mediating the benefits of bioactive compounds and functional foods.
Collapse
Affiliation(s)
- Laure B Bindels
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Rafael R Segura Munoz
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - João Carlos Gomes-Neto
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Valentin Mutemberezi
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Inés Martínez
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Nuria Salazar
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias-Consejo Superior de Investigaciones Científicas (IPLA-CSIC), Asturias, Spain
| | - Elizabeth A Cody
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, USA
| | | | - Hatem Kittana
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Clara G de Los Reyes-Gavilán
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias-Consejo Superior de Investigaciones Científicas (IPLA-CSIC), Asturias, Spain
| | - Robert J Schmaltz
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Giulio G Muccioli
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Jens Walter
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Amanda E Ramer-Tait
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, USA.
| |
Collapse
|
61
|
Tucci S, Floegel U, Beermann F, Behringer S, Spiekerkoetter U. Triheptanoin: long-term effects in the very long-chain acyl-CoA dehydrogenase-deficient mouse. J Lipid Res 2016; 58:196-207. [PMID: 27884962 DOI: 10.1194/jlr.m072033] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 11/07/2016] [Indexed: 12/31/2022] Open
Abstract
A rather new approach in the treatment of long-chain fatty acid oxidation disorders is represented by triheptanoin, a triglyceride with three medium-odd-chain heptanoic acids (C7), due to its anaplerotic potential. We here investigate the effects of a 1-year triheptanoin-based diet on the clinical phenotype of very long-chain-acyl-CoA-dehydrogenase-deficient (VLCAD-/-) mice. The cardiac function was assessed in VLCAD-/- mice by in vivo MRI. Metabolic adaptations were identified by the expression of genes regulating energy metabolism and anaplerotic processes using real-time PCR, and the results were correlated with the measurement of the glycolytic enzymes pyruvate dehydrogenase and pyruvate kinase. Finally, the intrahepatic lipid accumulation and oxidative stress in response to the long-term triheptanoin diet were assessed. Triheptanoin was not able to prevent the development of systolic dysfunction in VLCAD-/- mice despite an upregulation of cardiac glucose oxidation. Strikingly, the anaplerotic effects of triheptanoin were restricted to the liver. Despite this, the hepatic lipic content was increased upon triheptanoin supplementation. Our data demonstrate that the concept of anaplerosis does not apply to all tissues equally.
Collapse
Affiliation(s)
- Sara Tucci
- Department of General Pediatrics, Center for Pediatrics and Adolescent Medicine, University Hospital Freiburg, Freiburg, Germany
| | - Ulrich Floegel
- Department of Molecular Cardiology, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Frauke Beermann
- Department of General Pediatrics, Center for Pediatrics and Adolescent Medicine, University Hospital Freiburg, Freiburg, Germany
| | - Sidney Behringer
- Department of General Pediatrics, Center for Pediatrics and Adolescent Medicine, University Hospital Freiburg, Freiburg, Germany
| | - Ute Spiekerkoetter
- Department of General Pediatrics, Center for Pediatrics and Adolescent Medicine, University Hospital Freiburg, Freiburg, Germany
| |
Collapse
|
62
|
Rodriguez S, Lei X, Petersen PS, Tan SY, Little HC, Wong GW. Loss of CTRP1 disrupts glucose and lipid homeostasis. Am J Physiol Endocrinol Metab 2016; 311:E678-E697. [PMID: 27555298 PMCID: PMC5241556 DOI: 10.1152/ajpendo.00087.2016] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 08/18/2016] [Indexed: 12/22/2022]
Abstract
C1q/TNF-related protein 1 (CTRP1) is a conserved plasma protein of the C1q family with notable metabolic and cardiovascular functions. We have previously shown that CTRP1 infusion lowers blood glucose and that transgenic mice with elevated circulating CTRP1 are protected from diet-induced obesity and insulin resistance. Here, we used a genetic loss-of-function mouse model to address the requirement of CTRP1 for metabolic homeostasis. Despite similar body weight, food intake, and energy expenditure, Ctrp1 knockout (KO) mice fed a low-fat diet developed insulin resistance and hepatic steatosis. Impaired glucose metabolism in Ctrp1 KO mice was associated with increased hepatic gluconeogenic gene expression and decreased skeletal muscle glucose transporter glucose transporter 4 levels and AMP-activated protein kinase activation. Loss of CTRP1 enhanced the clearance of orally administered lipids but did not affect intestinal lipid absorption, hepatic VLDL-triglyceride export, or lipoprotein lipase activity. In contrast to triglycerides, hepatic cholesterol levels were reduced in Ctrp1 KO mice, paralleling the reduced expression of cholesterol synthesis genes. Contrary to expectations, when challenged with a high-fat diet to induce obesity, Ctrp1 KO mice had increased physical activity and reduced body weight, adiposity, and expression of lipid synthesis and fibrotic genes in adipose tissue; these phenotypes were linked to elevated FGF-21 levels. Due in part to increased hepatic AMP-activated protein kinase activation and reduced expression of lipid synthesis genes, Ctrp1 KO mice fed a high-fat diet also had reduced liver and serum triglyceride and cholesterol levels. Taken together, these results provide genetic evidence to establish the significance of CTRP1 to systemic energy metabolism in different metabolic and dietary contexts.
Collapse
Affiliation(s)
- Susana Rodriguez
- Department of Physiology and Center for Metabolism and Obesity Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Xia Lei
- Department of Physiology and Center for Metabolism and Obesity Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Pia S Petersen
- Department of Physiology and Center for Metabolism and Obesity Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Stefanie Y Tan
- Department of Physiology and Center for Metabolism and Obesity Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Hannah C Little
- Department of Physiology and Center for Metabolism and Obesity Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - G William Wong
- Department of Physiology and Center for Metabolism and Obesity Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
63
|
Baudrand R, Gupta N, Garza AE, Vaidya A, Leopold JA, Hopkins PN, Jeunemaitre X, Ferri C, Romero JR, Williams J, Loscalzo J, Adler GK, Williams GH, Pojoga LH. Caveolin 1 Modulates Aldosterone-Mediated Pathways of Glucose and Lipid Homeostasis. J Am Heart Assoc 2016; 5:JAHA.116.003845. [PMID: 27680666 PMCID: PMC5121487 DOI: 10.1161/jaha.116.003845] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background Overactivation of the aldosterone and mineralocorticoid receptor (MR) pathway is associated with hyperglycemia and dyslipidemia. Caveolin 1 (cav‐1) is involved in glucose/lipid homeostasis and may modulate MR signaling. We investigated the interplay between cav‐1 and aldosterone signaling in modulating insulin resistance and dyslipidemia in cav‐1–null mice and humans with a prevalent variant in the CAV1 gene. Methods and Results In mouse studies, cav‐1 knockout mice exhibited higher levels of homeostatic model assessment of insulin resistance, cholesterol, and resistin and lower ratios of high‐ to low‐density lipoprotein (all P<0.001 versus wild type). Moreover, cav‐1 knockout mice displayed hypertriglyceridemia and higher mRNA levels for resistin, retinol binding protein 4, NADPH oxidase 4, and aldose reductase in liver and/or fat tissues. MR blockade with eplerenone significantly decreased glycemia (P<0.01), total cholesterol (P<0.05), resistin (P<0.05), and described enzymes, with no effect on insulin or triglycerides. In the human study, we analyzed the CAV1 gene polymorphism rs926198 in 556 white participants; 58% were minor allele carriers and displayed higher odds of insulin resistance (odds ratio 2.26 [95% CI 1.40–3.64]) and low high‐density lipoprotein (odds ratio 1.54 [95% CI 1.01–3.37]). Aldosterone levels correlated with higher homeostatic model assessment of insulin resistance and resistin and lower high‐density lipoprotein only in minor allele carriers. CAV1 gene expression quantitative trait loci data revealed lower cav‐1 expression in adipose tissues by the rs926198 minor allele. Conclusions Our findings in mice and humans suggested that decreased cav‐1 expression may activate the effect of aldosterone/MR signaling on several pathways of glycemia, dyslipidemia, and resistin. In contrast, hyperinsulinemia and hypertriglyceridemia are likely mediated by MR‐independent mechanisms. Future human studies will elucidate the clinical relevance of MR blockade in patients with genotype‐mediated cav‐1 deficiency.
Collapse
Affiliation(s)
- Rene Baudrand
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA Department of Endocrinology, School of Medicine, Pontificia Universidad Catolica De Chile, Santiago, Chile
| | - Nidhi Gupta
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Amanda E Garza
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Anand Vaidya
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Jane A Leopold
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Paul N Hopkins
- Cardiovascular Genetics, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT
| | - Xavier Jeunemaitre
- Centre d'Investigation Clinique Inserm/AP, Departement de Genetique, Hȏpital European Georges Pompidou, Paris, France
| | - Claudio Ferri
- Department MeSVA, San Salvatore Hospital, University of L'Aquila, Italy
| | - Jose R Romero
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Jonathan Williams
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Joseph Loscalzo
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Gail K Adler
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Gordon H Williams
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Luminita H Pojoga
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
64
|
Hernández-Aguilera A, Fernández-Arroyo S, Cuyàs E, Luciano-Mateo F, Cabre N, Camps J, Lopez-Miranda J, Menendez JA, Joven J. Epigenetics and nutrition-related epidemics of metabolic diseases: Current perspectives and challenges. Food Chem Toxicol 2016; 96:191-204. [PMID: 27503834 DOI: 10.1016/j.fct.2016.08.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 08/03/2016] [Accepted: 08/04/2016] [Indexed: 02/07/2023]
Abstract
We live in a world fascinated by the relationship between disease and nutritional disequilibrium. The subtle and slow effects of chronic nutrient toxicity are a major public health concern. Since food is potentially important for the development of "metabolic memory", there is a need for more information on the type of nutrients causing adverse or toxic effects. We now know that metabolic alterations produced by excessive intake of some nutrients, drugs and chemicals directly impact epigenetic regulation. We envision that understanding how metabolic pathways are coordinated by environmental and genetic factors will provide novel insights for the treatment of metabolic diseases. New methods will enable the assembly and analysis of large sets of complex molecular and clinical data for understanding how inflammation and mitochondria affect bioenergetics, epigenetics and health. Collectively, the observations we highlight indicate that energy utilization and disease are intimately connected by epigenetics. The challenge is to incorporate metabolo-epigenetic data in better interpretations of disease, to expedite therapeutic targeting of key pathways linking nutritional toxicity and metabolism. An additional concern is that changes in the parental phenotype are detectable in the methylome of subsequent offspring. The effect might create a menace to future generations and preconceptional considerations.
Collapse
Affiliation(s)
- Anna Hernández-Aguilera
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain
| | - Salvador Fernández-Arroyo
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain
| | - Elisabet Cuyàs
- Molecular Oncology Group, Girona Biomedical Research Institute (IDIBGI), Girona, Spain; ProCURE (Program Against Cancer Therapeutic Resistance), Metabolism and Cancer Group, Catalan Institute of Oncology, Girona, Spain
| | - Fedra Luciano-Mateo
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain
| | - Noemi Cabre
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain
| | - Jordi Camps
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain
| | - Jose Lopez-Miranda
- Lipid and Atherosclerosis Unit, IMIBIC, Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain; CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Javier A Menendez
- Molecular Oncology Group, Girona Biomedical Research Institute (IDIBGI), Girona, Spain; ProCURE (Program Against Cancer Therapeutic Resistance), Metabolism and Cancer Group, Catalan Institute of Oncology, Girona, Spain.
| | - Jorge Joven
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain; The Campus of International Excellence Southern Catalonia, Tarragona, Spain.
| |
Collapse
|
65
|
Diet-induced obesity regulates adipose-resident stromal cell quantity and extracellular matrix gene expression. Stem Cell Res 2016; 17:181-90. [DOI: 10.1016/j.scr.2016.07.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 06/30/2016] [Accepted: 07/03/2016] [Indexed: 01/10/2023] Open
|
66
|
Lei X, Rodriguez S, Petersen PS, Seldin MM, Bowman CE, Wolfgang MJ, Wong GW. Loss of CTRP5 improves insulin action and hepatic steatosis. Am J Physiol Endocrinol Metab 2016; 310:E1036-52. [PMID: 27143553 PMCID: PMC4935138 DOI: 10.1152/ajpendo.00010.2016] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 04/26/2016] [Indexed: 01/01/2023]
Abstract
The gene that encodes C1q/TNF-related protein 5 (CTRP5), a secreted protein of the C1q family, is mutated in individuals with late-onset retinal degeneration. CTRP5 is widely expressed outside the eye and also circulates in plasma. Its physiological role in peripheral tissues, however, has yet to be elucidated. Here, we show that Ctrp5 expression is modulated by fasting and refeeding, and by different diets, in mice. Adipose expression of CTRP5 was markedly upregulated in obese and diabetic humans and in genetic and dietary models of obesity in rodents. Furthermore, human CTRP5 expression in the subcutaneous fat depot positively correlated with BMI. A genetic loss-of-function mouse model was used to address the metabolic function of CTRP5 in vivo. On a standard chow diet, CTRP5-deficient mice had reduced fasting insulin but were otherwise comparable with wild-type littermate controls in body weight and adiposity. However, when fed a high-fat diet, CTRP5-deficient animals had attenuated hepatic steatosis and improved insulin action. Loss of CTRP5 also improved the capacity of chow-fed aged mice to respond to subsequent high-fat feeding, as evidenced by decreased insulin resistance. In cultured adipocytes and myotubes, recombinant CTRP5 treatment attenuated insulin-stimulated Akt phosphorylation. Our results provide the first genetic and physiological evidence for CTRP5 as a negative regulator of glucose metabolism and insulin sensitivity. Inhibition of CTRP5 action may result in the alleviation of insulin resistance associated with obesity and diabetes.
Collapse
Affiliation(s)
- Xia Lei
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Susana Rodriguez
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Pia S Petersen
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Marcus M Seldin
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Caitlyn E Bowman
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland; and Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Michael J Wolfgang
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland; and Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - G William Wong
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
67
|
Fuhrmeister J, Zota A, Sijmonsma TP, Seibert O, Cıngır Ş, Schmidt K, Vallon N, de Guia RM, Niopek K, Berriel Diaz M, Maida A, Blüher M, Okun JG, Herzig S, Rose AJ. Fasting-induced liver GADD45β restrains hepatic fatty acid uptake and improves metabolic health. EMBO Mol Med 2016; 8:654-69. [PMID: 27137487 PMCID: PMC4888855 DOI: 10.15252/emmm.201505801] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Recent studies have demonstrated that repeated short‐term nutrient withdrawal (i.e. fasting) has pleiotropic actions to promote organismal health and longevity. Despite this, the molecular physiological mechanisms by which fasting is protective against metabolic disease are largely unknown. Here, we show that, metabolic control, particularly systemic and liver lipid metabolism, is aberrantly regulated in the fasted state in mouse models of metabolic dysfunction. Liver transcript assays between lean/healthy and obese/diabetic mice in fasted and fed states uncovered “growth arrest and DNA damage‐inducible” GADD45β as a dysregulated gene transcript during fasting in several models of metabolic dysfunction including ageing, obesity/pre‐diabetes and type 2 diabetes, in both mice and humans. Using whole‐body knockout mice as well as liver/hepatocyte‐specific gain‐ and loss‐of‐function strategies, we revealed a role for liver GADD45β in the coordination of liver fatty acid uptake, through cytoplasmic retention of FABP1, ultimately impacting obesity‐driven hyperglycaemia. In summary, fasting stress‐induced GADD45β represents a liver‐specific molecular event promoting adaptive metabolic function.
Collapse
Affiliation(s)
- Jessica Fuhrmeister
- Joint Research Division Molecular Metabolic Control, German Cancer Research Center, Center for Molecular Biology, Heidelberg University and Heidelberg University Hospital, Heidelberg, Germany
| | - Annika Zota
- Joint Research Division Molecular Metabolic Control, German Cancer Research Center, Center for Molecular Biology, Heidelberg University and Heidelberg University Hospital, Heidelberg, Germany Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, Neuherberg, Germany Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine I, Heidelberg University Hospital, Neuherberg, Germany
| | - Tjeerd P Sijmonsma
- Joint Research Division Molecular Metabolic Control, German Cancer Research Center, Center for Molecular Biology, Heidelberg University and Heidelberg University Hospital, Heidelberg, Germany
| | - Oksana Seibert
- Joint Research Division Molecular Metabolic Control, German Cancer Research Center, Center for Molecular Biology, Heidelberg University and Heidelberg University Hospital, Heidelberg, Germany
| | - Şahika Cıngır
- Joint Research Division Molecular Metabolic Control, German Cancer Research Center, Center for Molecular Biology, Heidelberg University and Heidelberg University Hospital, Heidelberg, Germany
| | - Kathrin Schmidt
- Division of Inherited Metabolic Diseases, University Children's Hospital, Heidelberg, Germany
| | - Nicola Vallon
- Joint Research Division Molecular Metabolic Control, German Cancer Research Center, Center for Molecular Biology, Heidelberg University and Heidelberg University Hospital, Heidelberg, Germany
| | - Roldan M de Guia
- Joint Research Division Molecular Metabolic Control, German Cancer Research Center, Center for Molecular Biology, Heidelberg University and Heidelberg University Hospital, Heidelberg, Germany
| | - Katharina Niopek
- Joint Research Division Molecular Metabolic Control, German Cancer Research Center, Center for Molecular Biology, Heidelberg University and Heidelberg University Hospital, Heidelberg, Germany Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, Neuherberg, Germany Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine I, Heidelberg University Hospital, Neuherberg, Germany
| | - Mauricio Berriel Diaz
- Joint Research Division Molecular Metabolic Control, German Cancer Research Center, Center for Molecular Biology, Heidelberg University and Heidelberg University Hospital, Heidelberg, Germany Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, Neuherberg, Germany Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine I, Heidelberg University Hospital, Neuherberg, Germany
| | - Adriano Maida
- Joint Research Division Molecular Metabolic Control, German Cancer Research Center, Center for Molecular Biology, Heidelberg University and Heidelberg University Hospital, Heidelberg, Germany Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, Neuherberg, Germany Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine I, Heidelberg University Hospital, Neuherberg, Germany
| | - Matthias Blüher
- Department of Medicine, University of Leipzig, Leipzig, Germany
| | - Jürgen G Okun
- Division of Inherited Metabolic Diseases, University Children's Hospital, Heidelberg, Germany
| | - Stephan Herzig
- Joint Research Division Molecular Metabolic Control, German Cancer Research Center, Center for Molecular Biology, Heidelberg University and Heidelberg University Hospital, Heidelberg, Germany Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, Neuherberg, Germany Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine I, Heidelberg University Hospital, Neuherberg, Germany
| | - Adam J Rose
- Joint Research Division Molecular Metabolic Control, German Cancer Research Center, Center for Molecular Biology, Heidelberg University and Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
68
|
Antunes LC, Elkfury JL, Jornada MN, Foletto KC, Bertoluci MC. Validation of HOMA-IR in a model of insulin-resistance induced by a high-fat diet in Wistar rats. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2016; 60:138-42. [PMID: 27191048 DOI: 10.1590/2359-3997000000169] [Citation(s) in RCA: 130] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 03/07/2016] [Indexed: 11/22/2022]
Abstract
Objective The present study aimed to validate homeostasis model assessment of insulin resistance (HOMA-IR) in relation to the insulin tolerance test (ITT) in a model of insulin-resistance in Wistar rats induced by a 19-week high-fat diet. Materials and methods A total of 30 male Wistar rats weighing 200-300 g were allocated into a high-fat diet group (HFD) (55% fat-enriched chow, ad lib, n = 15) and a standard-diet group (CD) standard chow, ad lib, n = 15), for 19 weeks. ITT was determined at baseline and in the 19th week. HOMA-IR was determined between the 18-19th week in three different days and the mean was considered for analysis. Area under the curve (AUC-ITT) of the blood glucose excursion along 120 minutes after intra-peritoneal insulin injection was determined and correlated with the corresponding fasting values for HOMA-IR. Results AUC-ITT and HOMA-IR were significantly greater after 19th week in HFD compared to CD (p < 0.001 for both). AUC-OGTT was also higher in HFD rats (p = 0.003). HOMA-IR was strongly correlated (Pearson's) with AUC-ITT r = 0.637; p < 0.0001. ROC curves of HOMA-IR and AUC-ITT showed similar sensitivity and specificity. Conclusion HOMA-IR is a valid measure to determine insulin-resistance in Wistar rats. Arch Endocrinol Metab. 2016;60(2):138-42.
Collapse
Affiliation(s)
- Luciana C Antunes
- Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brasil
| | | | - Manoela N Jornada
- Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brasil
| | - Kelly C Foletto
- Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brasil
| | - Marcello C Bertoluci
- Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brasil
| |
Collapse
|
69
|
Hypoxia-inducible factor prolyl hydroxylase 1 (PHD1) deficiency promotes hepatic steatosis and liver-specific insulin resistance in mice. Sci Rep 2016; 6:24618. [PMID: 27094951 PMCID: PMC4837354 DOI: 10.1038/srep24618] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 03/31/2016] [Indexed: 01/03/2023] Open
Abstract
Obesity is associated with local tissue hypoxia and elevated hypoxia-inducible factor 1 alpha (HIF-1α) in metabolic tissues. Prolyl hydroxylases (PHDs) play an important role in regulating HIF-α isoform stability. In the present study, we investigated the consequence of whole-body PHD1 gene (Egln2) inactivation on metabolic homeostasis in mice. At baseline, PHD1-/- mice exhibited higher white adipose tissue (WAT) mass, despite lower body weight, and impaired insulin sensitivity and glucose tolerance when compared to age-matched wild-type (WT) mice. When fed a synthetic low-fat diet, PHD1-/- mice also exhibit a higher body weight gain and WAT mass along with glucose intolerance and systemic insulin resistance compared to WT mice. PHD1 deficiency led to increase in glycolytic gene expression, lipogenic proteins ACC and FAS, hepatic steatosis and liver-specific insulin resistance. Furthermore, gene markers of inflammation were also increased in the liver, but not in WAT or skeletal muscle, of PHD1-/- mice. As expected, high-fat diet (HFD) promoted obesity, hepatic steatosis, tissue-specific inflammation and systemic insulin resistance in WT mice but these diet-induced metabolic alterations were not exacerbated in PHD1-/- mice. In conclusion, PHD1 deficiency promotes hepatic steatosis and liver-specific insulin resistance but does not worsen the deleterious effects of HFD on metabolic homeostasis.
Collapse
|
70
|
Malmgren S, Ahrén B. Evidence for time dependent variation of glucagon secretion in mice. Peptides 2016; 76:102-7. [PMID: 26774585 DOI: 10.1016/j.peptides.2016.01.008] [Citation(s) in RCA: 2] [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: 10/18/2015] [Revised: 01/05/2016] [Accepted: 01/09/2016] [Indexed: 12/15/2022]
Abstract
Glucose metabolism is subjected to diurnal variation, which might be mediated by alterations in the transcription pattern of clock genes and regulated by hormonal factors, as has been demonstrated for insulin. However, whether also glucagon is involved in the diurnal variation of glucose homeostasis is not known. We therefore examined glucagon secretion after meal ingestion (meal tolerance test) and during hypoglycemia (hyperinsulinemic hypoglycemic clamp at 2.5mmol/L glucose) and in vitro from isolated islets at ZT3 versus ZT15 in normal C57BL/6J mice and, furthermore, glucose levels and the insulin response to meal ingestion were also examined at these time points in glucagon receptor knockout mice (GCGR-/-) and their wildtype (wt) littermates. We found in normal mice that whereas the glucagon response to meal ingestion was not different between ZT3 and ZT15, the glucagon response to hypoglycemia was lower at ZT3 than at ZT15 and glucagon secretion from isolated islets was higher at ZT3 than at ZT15. GCGR-/- mice displayed lower basal glucose, a lower insulin response to meal and a higher insulin sensitivity than wt mice at ZT3 but not at ZT15. We conclude that there is a time dependent variation in glucagon secretion in normal mice, which is dependent both on intraislet and extraislet regulatory mechanisms and that the phenotype characteristics of a lower glucose and reduced insulin response to meal in GCGR-/- mice are evident only during the light phase. These findings suggest that glucagon signaling is a plausible contributor to the diurnal variation in glucose homeostasis which may explain that the phenotype of the GCGR-/- mice is dependent on the time of the day when it is examined.
Collapse
Affiliation(s)
- Siri Malmgren
- Department of Clinical Sciences in Lund, Section of Medicine, Lund University, Lund, Sweden
| | - Bo Ahrén
- Department of Clinical Sciences in Lund, Section of Medicine, Lund University, Lund, Sweden.
| |
Collapse
|
71
|
Picklo MJ, Newman JW. Antioxidant supplementation and obesity have independent effects on hepatic oxylipin profiles in insulin-resistant, obesity-prone rats. Free Radic Biol Med 2015; 89:182-91. [PMID: 26398714 DOI: 10.1016/j.freeradbiomed.2015.07.152] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 07/01/2015] [Accepted: 07/21/2015] [Indexed: 01/25/2023]
Abstract
Obesity-induced changes in lipid metabolism are mechanistically associated with the development of insulin resistance and prediabetes. Recent studies have focused on the extent to which obesity-induced insulin resistance is mediated through oxylipins, derived from enzymatic and nonenzymatic lipid peroxidation. Vitamin E and vitamin C are widely used antioxidant supplements, but conflicting data exist as to whether supplementation with vitamins E and C reduces insulin resistance. The purpose of this work is (1) to test the hypothesis that supplementation with vitamin E and vitamin C prevents the development of insulin resistance and (2) to determine the extent to which antioxidant supplementation modifies obesity-induced changes in hepatic oxylipins. Using obesity-prone Sprague-Dawley rats fed a high-fat, hypercaloric diet, we found that vitamin E and C supplementation did not block the development of insulin resistance, despite increased plasma levels of these antioxidants and decreased hepatic F2-isoprostane (F2-IsoP) concentrations. The obese phenotype was associated with increased hepatic concentrations of cytochrome P450 (CYP450)-dependent linoleic acid and α-linolenic acid-derived epoxides. Antioxidant supplementation, but not obesity, decreased levels of the lipoxygenase (LOX)-dependent, arachidonic acid-derived products lipoxin A4 (LXA4), 8,15-dihydroxtetraenoate (8,15-DiHETE), and 5,15-DiHETE. Our data demonstrate that antioxidant supplementation and obesity impact hepatic LOX- and CYP450-dependent oxylipin metabolism.
Collapse
Affiliation(s)
- Matthew J Picklo
- USDA-ARS Grand Forks Human Nutrition Research Center 2420 2(nd) Avenue North Grand Forks, ND 58203-9034, USA; Department of Chemistry, University of North Dakota, Grand Forks, ND 58201, USA.
| | - John W Newman
- USDA-ARS Western Human Nutrition Research Center, Davis, CA 95616, USA; West Coast Metabolomics Center, University of California Davis, Davis, CA 95616, USA; Department of Nutrition, University of California Davis, Davis, CA 95616, USA
| |
Collapse
|
72
|
Zhou J, Xu G, Bai Z, Li K, Yan J, Li F, Ma S, Xu H, Huang K. Selenite exacerbates hepatic insulin resistance in mouse model of type 2 diabetes through oxidative stress-mediated JNK pathway. Toxicol Appl Pharmacol 2015; 289:409-18. [PMID: 26522834 DOI: 10.1016/j.taap.2015.10.019] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Revised: 10/25/2015] [Accepted: 10/27/2015] [Indexed: 12/20/2022]
Abstract
Recent evidence suggests a potential pro-diabetic effect of selenite treatment in type 2 diabetics; however, the underlying mechanisms remain elusive. Here we investigated the effects and the underlying mechanisms of selenite treatment in a nongenetic mouse model of type 2 diabetes. High-fat diet (HFD)/streptozotocin (STZ)-induced diabetic mice were orally gavaged with selenite at 0.5 or 2.0mg/kg body weight/day or vehicle for 4 weeks. High-dose selenite treatment significantly elevated fasting plasma insulin levels and insulin resistance index, in parallel with impaired glucose tolerance, insulin tolerance and pyruvate tolerance. High-dose selenite treatment also attenuated hepatic IRS1/Akt/FoxO1 signaling and pyruvate kinase gene expressions, but elevated the gene expressions of phosphoenolpyruvate carboxyl kinase (PEPCK), glucose 6-phosphatase (G6Pase), peroxisomal proliferator-activated receptor-γ coactivator 1α (PGC-1α) and selenoprotein P (SelP) in the liver. Furthermore, high-dose selenite treatment caused significant increases in MDA contents, protein carbonyl contents, and a decrease in GSH/GSSG ratio in the liver, concurrent with enhanced ASK1/MKK4/JNK signaling. Taken together, these findings suggest that high-dose selenite treatment exacerbates hepatic insulin resistance in mouse model of type 2 diabetes, at least in part through oxidative stress-mediated JNK pathway, providing new mechanistic insights into the pro-diabetic effect of selenite in type 2 diabetes.
Collapse
Affiliation(s)
- Jun Zhou
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, PR China.
| | - Gang Xu
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, PR China
| | - Zhaoshuai Bai
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, PR China
| | - Kaicheng Li
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, PR China
| | - Junyan Yan
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, PR China
| | - Fen Li
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, PR China
| | - Shuai Ma
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, PR China
| | - Huibi Xu
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, PR China
| | - Kaixun Huang
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, PR China.
| |
Collapse
|
73
|
Reduced linoleic acid intake in early postnatal life improves metabolic outcomes in adult rodents following a Western-style diet challenge. Nutr Res 2015; 35:800-11. [PMID: 26239950 DOI: 10.1016/j.nutres.2015.06.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 06/04/2015] [Accepted: 06/26/2015] [Indexed: 10/23/2022]
Abstract
The global increase in dietary n-6 polyunsaturated fatty acid (PUFA) intake has been suggested to contribute to the rise in obesity incidence. We hypothesized that reduced n-6 PUFA intake during early postnatal life improves adult body composition and metabolic phenotype upon a Western diet challenge. Male offspring of C57Bl/6j mice and Wistar rats were subjected to a control diet (CTRL; 3.16 En% linoleic acid [LA]) or a low n-6 PUFA diet (low LA; 1.36 En% LA) from postnatal days (PNs) 2 to 42. Subsequently, all animals were switched to a Western-style diet (2.54 En% LA) until PN98. We monitored body composition by dual-energy x-ray absorptiometry and glucose homeostasis by an intravenous glucose and insulin tolerance test in rats and by the homeostasis model assessment of insulin resistance (HOMA-IR) in mice. At PN98, plasma lipids, glucose, insulin, and adipokines were measured and adipocyte number and size were analyzed. In mice, the postnatal low-LA diet decreased fat accumulation during the adult Western-style diet challenge (-27% compared with CTRL, P < .001). Simultaneously, it reduced fasting triglyceride levels and lowered fasting resistin and leptin levels. In rats, the low-LA diet did not affect adult body composition, but decreased the number of retroperitoneal adipocytes and increased the number of large adipocytes. In conclusion, lowering dietary n-6 PUFA intake in early life protected against detrimental effects of an obesogenic diet in adulthood on metabolic homeostasis and fat mass accumulation.
Collapse
|
74
|
Tiano JP, Tate CR, Yang BS, DiMarchi R, Mauvais-Jarvis F. Effect of targeted estrogen delivery using glucagon-like peptide-1 on insulin secretion, insulin sensitivity and glucose homeostasis. Sci Rep 2015; 5:10211. [PMID: 25970118 PMCID: PMC4429560 DOI: 10.1038/srep10211] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 04/07/2015] [Indexed: 12/21/2022] Open
Abstract
The female estrogen 17β-estradiol (E2) enhances pancreatic β-cell function via estrogen receptors (ERs). However, the risk of hormone dependent cancer precludes the use of general estrogen therapy as a chronic treatment for diabetes. To target E2 to β-cells without the undesirable effects of general estrogen therapy, we created fusion peptides combining active or inactive glucagon-like peptide-1 (GLP-1) and E2 in a single molecule (aGLP1-E2 and iGLP1-E2 respectively). By combining the activities of GLP-1 and E2, we envisioned synergistic insulinotropic activities of these molecules on β-cells. In cultured human islets and in C57BL/6 mice, both aGLP1 and aGLP1-E2 enhanced glucose-stimulated insulin secretion (GSIS) compared to vehicle and iGLP1-E2 without superior efficacy of aGLP1-E2 compared to GLP-1 alone. However, aGLP1-E2 decreased fasting and fed blood glucose to a greater extent than aGLP1 and iGLP1-E2 alone. Further, aGLP1-E2 exhibited improved insulin sensitivity compared to aGLP1 and iGLP1-E2 alone (HOMA-IR and insulin tolerance test). In conclusion, targeted estrogen delivery to non-diabetic islets in the presence of GLP-1 does not enhance GSIS. However, combining GLP-1 to estrogen delivers additional efficacy relative to GLP-1 alone on insulin sensitivity and glucose homeostasis in non-diabetic mice.
Collapse
Affiliation(s)
- Joseph P Tiano
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University School of Medicine, Chicago, IL 60611
| | - Chandra R Tate
- Division of Endocrinology &Metabolism, Department of Medicine, Tulane University Health Sciences Center, School of Medicine, New Orleans, LA 70112
| | - Bin S Yang
- Department of Chemistry, Indiana University, Bloomington, IN, 47405
| | - Richard DiMarchi
- Department of Chemistry, Indiana University, Bloomington, IN, 47405
| | - Franck Mauvais-Jarvis
- 1] Division of Endocrinology &Metabolism, Department of Medicine, Tulane University Health Sciences Center, School of Medicine, New Orleans, LA 70112 [2] Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University School of Medicine, Chicago, IL 60611
| |
Collapse
|
75
|
Metformin suppresses diethylnitrosamine-induced liver tumorigenesis in obese and diabetic C57BL/KsJ-+Leprdb/+Leprdb mice. PLoS One 2015; 10:e0124081. [PMID: 25879666 PMCID: PMC4399835 DOI: 10.1371/journal.pone.0124081] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 03/03/2015] [Indexed: 12/22/2022] Open
Abstract
Obesity and related metabolic disorders, such as diabetes mellitus, raise the risk of liver carcinogenesis. Metformin, which is widely used in the treatment of diabetes, ameliorates insulin sensitivity. Metformin is also thought to have antineoplastic activities and to reduce cancer risk. The present study examined the preventive effect of metformin on the development of diethylnitrosamine (DEN)-induced liver tumorigenesis in C57BL/KsJ-+Leprdb/+Leprdb (db/db) obese and diabetic mice. The mice were given a single injection of DEN at 2 weeks of age and subsequently received drinking water containing metformin for 20 weeks. Metformin administration significantly reduced the multiplicity of hepatic premalignant lesions and inhibited liver cell neoplasms. Metformin also markedly decreased serum levels of insulin and reduced insulin resistance, and inhibited phosphorylation of Akt, mammalian target of rapamycin (mTOR), and p70S6 in the liver. Furthermore, serum levels of leptin were decreased, while those of adiponectin were increased by metformin. These findings suggest that metformin prevents liver tumorigenesis by ameliorating insulin sensitivity, inhibiting the activation of Akt/mTOR/p70S6 signaling, and improving adipokine imbalance. Therefore, metformin may be a potent candidate for chemoprevention of liver tumorigenesis in patients with obesity or diabetes.
Collapse
|
76
|
Tucci S, Flögel U, Spiekerkoetter U. Sexual dimorphism of lipid metabolism in very long-chain acyl-CoA dehydrogenase deficient (VLCAD-/-) mice in response to medium-chain triglycerides (MCT). Biochim Biophys Acta Mol Basis Dis 2015; 1852:1442-50. [PMID: 25887160 DOI: 10.1016/j.bbadis.2015.04.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 03/17/2015] [Accepted: 04/07/2015] [Indexed: 02/08/2023]
Abstract
Medium-chain triglycerides (MCT) are widely applied in the treatment of long-chain fatty acid oxidation disorders. Previously it was shown that long-term MCT supplementation strongly affects lipid metabolism in mice. We here investigate sex-specific effects in mice with very-long-chain-acyl-CoA dehydrogenase (VLCAD) deficiency in response to a long-term MCT modified diet. We quantified blood lipids, acylcarnitines, glucose, insulin and free fatty acids, as well as tissue triglycerides in the liver and skeletal muscle under a control and an MCT diet over 1 year. In addition, visceral and hepatic fat content and muscular intramyocellular lipids (IMCL) were assessed by in vivo(1)H magnetic resonance spectroscopy (MRS) techniques. The long-term application of an MCT diet induced a marked alteration of glucose homeostasis. However, only VLCAD-/- female mice developed a severe metabolic syndrome characterized by marked insulin resistance, dyslipidemia, severe hepatic and visceral steatosis, whereas VLCAD-/- males seemed to be protected and only presented with milder insulin resistance. Moreover, the highly saturated MCT diet is associated with a decreased hepatic stearoyl-CoA desaturase 1 (SCD1) activity in females aggravating the harmful effects of a saturated MCT diet. Long-term MCT supplementation deeply affects lipid metabolism in a sexual dimorphic manner resulting in a severe metabolic syndrome only in female mice. These findings are striking since the first signs of insulin resistance already occur in female VLCAD-/- mice during their reproductive period. How these metabolic adaptations are finally regulated needs to be determined. More important, the relevance of these findings for humans under these dietary modifications needs to be investigated.
Collapse
Affiliation(s)
- Sara Tucci
- Department of General Pediatrics, Center for Pediatrics and Adolescent Medicine, University Hospital Freiburg, 79106 Freiburg, Germany.
| | - Ulrich Flögel
- Department of Molecular Cardiology, Heinrich-Heine-University Duesseldorf, 40225 Düsseldorf, Germany
| | - Ute Spiekerkoetter
- Department of General Pediatrics, Center for Pediatrics and Adolescent Medicine, University Hospital Freiburg, 79106 Freiburg, Germany
| |
Collapse
|
77
|
Wu Y, Williams EG, Dubuis S, Mottis A, Jovaisaite V, Houten SM, Argmann CA, Faridi P, Wolski W, Kutalik Z, Zamboni N, Auwerx J, Aebersold R. Multilayered genetic and omics dissection of mitochondrial activity in a mouse reference population. Cell 2014; 158:1415-1430. [PMID: 25215496 DOI: 10.1016/j.cell.2014.07.039] [Citation(s) in RCA: 181] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Revised: 06/02/2014] [Accepted: 07/28/2014] [Indexed: 02/06/2023]
Abstract
The manner by which genotype and environment affect complex phenotypes is one of the fundamental questions in biology. In this study, we quantified the transcriptome--a subset of the metabolome--and, using targeted proteomics, quantified a subset of the liver proteome from 40 strains of the BXD mouse genetic reference population on two diverse diets. We discovered dozens of transcript, protein, and metabolite QTLs, several of which linked to metabolic phenotypes. Most prominently, Dhtkd1 was identified as a primary regulator of 2-aminoadipate, explaining variance in fasted glucose and diabetes status in both mice and humans. These integrated molecular profiles also allowed further characterization of complex pathways, particularly the mitochondrial unfolded protein response (UPR(mt)). UPR(mt) shows strikingly variant responses at the transcript and protein level that are remarkably conserved among C. elegans, mice, and humans. Overall, these examples demonstrate the value of an integrated multilayered omics approach to characterize complex metabolic phenotypes.
Collapse
Affiliation(s)
- Yibo Wu
- Department of Biology, Institute of Molecular Systems Biology, Eidgenössische Technische Hochschule Zürich (ETHZ), Zurich 8093, Switzerland
| | - Evan G Williams
- Laboratory of Integrative and Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Sébastien Dubuis
- Department of Biology, Institute of Molecular Systems Biology, Eidgenössische Technische Hochschule Zürich (ETHZ), Zurich 8093, Switzerland
| | - Adrienne Mottis
- Laboratory of Integrative and Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Virginija Jovaisaite
- Laboratory of Integrative and Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Sander M Houten
- Department of Clinical Chemistry, Laboratory Genetic Metabolic Diseases and Department of Pediatrics, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
| | - Carmen A Argmann
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
| | - Pouya Faridi
- Department of Biology, Institute of Molecular Systems Biology, Eidgenössische Technische Hochschule Zürich (ETHZ), Zurich 8093, Switzerland; Department of Traditional Pharmacy, School of Pharmacy and Pharmaceutical Sciences, Shiraz University of Medical Sciences, Shiraz 71349-14693, Iran
| | - Witold Wolski
- Department of Biology, Institute of Molecular Systems Biology, Eidgenössische Technische Hochschule Zürich (ETHZ), Zurich 8093, Switzerland
| | - Zoltán Kutalik
- Institute of Social and Preventive Medicine (IUMSP), Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne 1010, Switzerland; Swiss Institute of Bioinformatics, Lausanne 1015, Switzerland
| | - Nicola Zamboni
- Department of Biology, Institute of Molecular Systems Biology, Eidgenössische Technische Hochschule Zürich (ETHZ), Zurich 8093, Switzerland
| | - Johan Auwerx
- Laboratory of Integrative and Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland.
| | - Ruedi Aebersold
- Department of Biology, Institute of Molecular Systems Biology, Eidgenössische Technische Hochschule Zürich (ETHZ), Zurich 8093, Switzerland; Faculty of Science, University of Zurich, Zurich 8057, Switzerland.
| |
Collapse
|
78
|
Jelenik T, Séquaris G, Kaul K, Ouwens DM, Phielix E, Kotzka J, Knebel B, Weiß J, Reinbeck AL, Janke L, Nowotny P, Partke HJ, Zhang D, Shulman GI, Szendroedi J, Roden M. Tissue-specific differences in the development of insulin resistance in a mouse model for type 1 diabetes. Diabetes 2014; 63:3856-67. [PMID: 24917575 DOI: 10.2337/db13-1794] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Although insulin resistance is known to underlie type 2 diabetes, its role in the development of type 1 diabetes has been gaining increasing interest. In a model of type 1 diabetes, the nonobese diabetic (NOD) mouse, we found that insulin resistance driven by lipid- and glucose-independent mechanisms is already present in the liver of prediabetic mice. Hepatic insulin resistance is associated with a transient rise in mitochondrial respiration followed by increased production of lipid peroxides and c-Jun N-terminal kinase activity. At the onset of diabetes, increased adipose tissue lipolysis promotes myocellular diacylglycerol accumulation. This is paralleled by increased myocellular protein kinase C θ activity and serum fetuin A levels. Muscle mitochondrial oxidative capacity is unchanged at the onset but decreases at later stages of diabetes. In conclusion, hepatic and muscle insulin resistance manifest at different stages and involve distinct cellular mechanisms during the development of diabetes in the NOD mouse.
Collapse
Affiliation(s)
- Tomas Jelenik
- Institute for Clinical Diabetology, German Diabetes Center, Düsseldorf, Germany German Center for Diabetes Research, Partner Düsseldorf, Germany
| | - Gilles Séquaris
- Institute for Clinical Diabetology, German Diabetes Center, Düsseldorf, Germany German Center for Diabetes Research, Partner Düsseldorf, Germany
| | - Kirti Kaul
- Institute for Clinical Diabetology, German Diabetes Center, Düsseldorf, Germany German Center for Diabetes Research, Partner Düsseldorf, Germany
| | - D Margriet Ouwens
- German Center for Diabetes Research, Partner Düsseldorf, Germany Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Düsseldorf, Germany Department of Endocrinology, Ghent University Hospital, Ghent, Belgium
| | - Esther Phielix
- Institute for Clinical Diabetology, German Diabetes Center, Düsseldorf, Germany German Center for Diabetes Research, Partner Düsseldorf, Germany
| | - Jörg Kotzka
- German Center for Diabetes Research, Partner Düsseldorf, Germany Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Düsseldorf, Germany
| | - Birgit Knebel
- German Center for Diabetes Research, Partner Düsseldorf, Germany Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Düsseldorf, Germany
| | - Jürgen Weiß
- German Center for Diabetes Research, Partner Düsseldorf, Germany Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Düsseldorf, Germany
| | - Anna Lena Reinbeck
- Institute for Clinical Diabetology, German Diabetes Center, Düsseldorf, Germany German Center for Diabetes Research, Partner Düsseldorf, Germany
| | - Linda Janke
- Institute for Clinical Diabetology, German Diabetes Center, Düsseldorf, Germany German Center for Diabetes Research, Partner Düsseldorf, Germany
| | - Peter Nowotny
- Institute for Clinical Diabetology, German Diabetes Center, Düsseldorf, Germany German Center for Diabetes Research, Partner Düsseldorf, Germany
| | - Hans-Joachim Partke
- Institute for Clinical Diabetology, German Diabetes Center, Düsseldorf, Germany German Center for Diabetes Research, Partner Düsseldorf, Germany Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Düsseldorf, Germany
| | - Dongyan Zhang
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT
| | - Gerald I Shulman
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT
| | - Julia Szendroedi
- Institute for Clinical Diabetology, German Diabetes Center, Düsseldorf, Germany German Center for Diabetes Research, Partner Düsseldorf, Germany Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Düsseldorf, Germany Department of Endocrinology and Diabetology, Heinrich-Heine University, Düsseldorf, Germany
| | - Michael Roden
- Institute for Clinical Diabetology, German Diabetes Center, Düsseldorf, Germany German Center for Diabetes Research, Partner Düsseldorf, Germany Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Düsseldorf, Germany Department of Endocrinology and Diabetology, Heinrich-Heine University, Düsseldorf, Germany
| |
Collapse
|
79
|
Long-term corticosterone exposure decreases insulin sensitivity and induces depressive-like behaviour in the C57BL/6NCrl mouse. PLoS One 2014; 9:e106960. [PMID: 25310187 PMCID: PMC4195581 DOI: 10.1371/journal.pone.0106960] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 08/04/2014] [Indexed: 12/21/2022] Open
Abstract
Chronic stress or long-term administration of glucocorticoids disrupts the hypothalamus-pituitary-adrenal system leading to continuous high levels of glucocorticoids and insulin resistance (IR). This pre-diabetic state can eventually develop into type 2 diabetes mellitus and has been associated with a higher risk to develop depressive disorders. The mechanisms underlying the link between chronic stress, IR and depression remains unclear. The present study aimed to establish a stress-depression model in mice to further study the effects of stress-induced changes upon insulin sensitivity and behavioural consequences. A pilot study was conducted to establish the optimal administration route and a pragmatic measurement of IR. Subsequently, 6-month-old C57BL/6NCrl mice were exposed to long-term oral corticosterone treatment via the drinking water. To evaluate insulin sensitivity changes, blood glucose and plasma insulin levels were measured at different time-points throughout treatment and mice were behaviourally assessed in the elevated zero maze (EZM), forced swimming test (FST) and open field test to reveal behavioural changes. Long-term corticosterone treatment increased body weight and decreased insulin sensitivity. The latter was revealed by a higher IR index and increased insulin in the plasma, whereas blood glucose levels remained unchanged. Corticosterone treatment induced longer immobility times in the FST, reflecting depressive-like behaviour. No effects were observed upon anxiety as measured in the EZM. The effect of the higher body weight of the CORT treated animals at time of testing did not influence behaviour in the EZM or FST, as no differences were found in general locomotor activity. Long-term corticosterone treatment via the drinking water reduces insulin sensitivity and induces depressive-like behaviour in the C57BL/6 mouse. This mouse model could thus be used to further explore the underlying mechanisms of chronic stress-induced T2DM and its association with increased prevalence of major depressive disorder on the short-term and other behavioural adaptations on the longer term.
Collapse
|
80
|
Bowe JE, Franklin ZJ, Hauge-Evans AC, King AJ, Persaud SJ, Jones PM. Metabolic phenotyping guidelines: assessing glucose homeostasis in rodent models. J Endocrinol 2014; 222:G13-25. [PMID: 25056117 DOI: 10.1530/joe-14-0182] [Citation(s) in RCA: 171] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The pathophysiology of diabetes as a disease is characterised by an inability to maintain normal glucose homeostasis. In type 1 diabetes, this is due to autoimmune destruction of the pancreatic β-cells and subsequent lack of insulin production, and in type 2 diabetes it is due to a combination of both insulin resistance and an inability of the β-cells to compensate adequately with increased insulin release. Animal models, in particular genetically modified mice, are increasingly being used to elucidate the mechanisms underlying both type 1 and type 2 diabetes, and as such the ability to study glucose homeostasis in vivo has become an essential tool. Several techniques exist for measuring different aspects of glucose tolerance and each of these methods has distinct advantages and disadvantages. Thus the appropriate methodology may vary from study to study depending on the desired end-points, the animal model, and other practical considerations. This review outlines the most commonly used techniques for assessing glucose tolerance in rodents and details the factors that should be taken into account in their use. Representative scenarios illustrating some of the practical considerations of designing in vivo experiments for the measurement of glucose homeostasis are also discussed.
Collapse
Affiliation(s)
- James E Bowe
- Diabetes Research GroupDivision of Diabetes and Nutritional Sciences, King's College London, Guy's Campus, London SE1 1UL, UK
| | - Zara J Franklin
- Diabetes Research GroupDivision of Diabetes and Nutritional Sciences, King's College London, Guy's Campus, London SE1 1UL, UK
| | - Astrid C Hauge-Evans
- Diabetes Research GroupDivision of Diabetes and Nutritional Sciences, King's College London, Guy's Campus, London SE1 1UL, UK
| | - Aileen J King
- Diabetes Research GroupDivision of Diabetes and Nutritional Sciences, King's College London, Guy's Campus, London SE1 1UL, UK
| | - Shanta J Persaud
- Diabetes Research GroupDivision of Diabetes and Nutritional Sciences, King's College London, Guy's Campus, London SE1 1UL, UK
| | - Peter M Jones
- Diabetes Research GroupDivision of Diabetes and Nutritional Sciences, King's College London, Guy's Campus, London SE1 1UL, UK
| |
Collapse
|
81
|
Andreassen KV, Feigh M, Hjuler ST, Gydesen S, Henriksen JE, Beck-Nielsen H, Christiansen C, Karsdal MA, Henriksen K. A novel oral dual amylin and calcitonin receptor agonist (KBP-042) exerts antiobesity and antidiabetic effects in rats. Am J Physiol Endocrinol Metab 2014; 307:E24-33. [PMID: 24801386 DOI: 10.1152/ajpendo.00121.2014] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The present study investigated a novel oral dual amylin and calcitonin receptor agonist (DACRA), KBP-042, in head-to-head comparison with salmon calcitonin (sCT) with regard to in vitro receptor pharmacology, ex vivo pancreatic islet studies, and in vivo proof of concept studies in diet-induced obese (DIO) and Zucker diabetic fatty (ZDF) rats. In vitro, KBP-042 demonstrated superior binding affinity and activation of amylin and calcitonin receptors, and ex vivo, KBP-042 exerted inhibitory action on stimulated insulin and glucagon release from isolated islets. In vivo, KBP-042 induced a superior and pronounced reduction in food intake in conjunction with a sustained pair-fed corrected weight loss in DIO rats. Concomitantly, KBP-042 improved glucose homeostasis and reduced hyperinsulinemia and hyperleptinemia in conjunction with enhanced insulin sensitivity. In ZDF rats, KBP-042 induced a superior attenuation of diabetic hyperglycemia and alleviated impaired glucose and insulin tolerance. Concomitantly, KBP-042 preserved insulinotropic and induced glucagonostatic action, ultimately preserving pancreatic insulin and glucagon content. In conclusion, oral KBP-042 is a novel DACRA, which exerts antiobesity and antidiabetic efficacy by dual modulation of insulin sensitivity and directly decelerating stress on the pancreatic α- and β-cells. These results could provide the basis for oral KBP-042 as a novel therapeutic agent in type 2 diabetes.
Collapse
Affiliation(s)
| | - Michael Feigh
- Nordic Bioscience, Herlev, Denmark; and Diabetes Research Centre, Department of Endocrinology, University of Southern Denmark, Odense, Denmark
| | | | | | - Jan Erik Henriksen
- Diabetes Research Centre, Department of Endocrinology, University of Southern Denmark, Odense, Denmark
| | - Henning Beck-Nielsen
- Diabetes Research Centre, Department of Endocrinology, University of Southern Denmark, Odense, Denmark
| | | | | | | |
Collapse
|
82
|
Affiliation(s)
- Jerry Radziuk
- Department of Medicine, University of Ottawa, Ottawa Hospital, Ottawa, Ontario, Canada
| |
Collapse
|
83
|
Dib LH, Ortega MT, Fleming SD, Chapes SK, Melgarejo T. Bone marrow leptin signaling mediates obesity-associated adipose tissue inflammation in male mice. Endocrinology 2014; 155:40-6. [PMID: 24169547 PMCID: PMC3868799 DOI: 10.1210/en.2013-1607] [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] [Indexed: 12/18/2022]
Abstract
Obesity is characterized by an increased recruitment of proinflammatory macrophages to the adipose tissue (AT), leading to systemic inflammation and metabolic disease. The pathogenesis of this AT inflammation, however, remains to be elucidated. The circulating adipokine leptin is increased in obesity and is involved in immune cell function and activation. In the present study, we investigated the role of leptin in the induction of obesity-associated inflammation. We generated radiation chimeric C57BL/6J mice reconstituted with either leptin receptor-deficient (db/db) or wild-type (WT) bone marrow and challenged them with a high-fat diet (HFD) for 16 weeks. Mice reconstituted with db/db bone marrow (WT/db), had significantly lower body weight and adiposity compared with mice with WT bone marrow (WT/WT). Gonadal AT in WT/db mice displayed a 2-fold lower expression of the inflammatory genes Tnfa, Il6, and Ccl2. In addition, gonadal fat of WT/db mice contained significantly fewer crown-like structures compared with WT/WT mice, and most of their AT macrophages expressed macrophage galactose-type C type lectin 1 (MGL1) and were C-C chemokine receptor type 2 (CCR2)-negative, indicative of an anti-inflammatory phenotype. Moreover, WT/db mice exhibited greater insulin sensitivity compared with WT/WT mice. These data show that disrupted leptin signaling in bone marrow-derived cells attenuates the proinflammatory conditions that mediate many of the metabolic complications that characterize obesity. Our findings establish a novel mechanism involved in the regulation of obesity-associated systemic inflammation and support the hypothesis that leptin is a proinflammatory cytokine.
Collapse
|
84
|
Baudrand R, Lian CG, Lian BQ, Ricchiuti V, Yao TM, Li J, Williams GH, Adler GK. Long-term dietary sodium restriction increases adiponectin expression and ameliorates the proinflammatory adipokine profile in obesity. Nutr Metab Cardiovasc Dis 2014; 24:34-41. [PMID: 24418377 PMCID: PMC4405158 DOI: 10.1016/j.numecd.2013.07.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Revised: 07/08/2013] [Accepted: 07/25/2013] [Indexed: 01/08/2023]
Abstract
BACKGROUND/AIM Obesity is associated with changes in adiponectin and pro-inflammatory adipokines. Sodium intake can affect adipokine secretion suggesting a role in cardiovascular dysfunction. We tested if long-term dietary sodium restriction modifies the expression of adiponectin and ameliorates the pro-inflammatory profile of obese, diabetic mice. METHODS/RESULTS Db/db mice were randomized to high sodium (HS 1.6% Na+, n = 6) or low sodium (LS 0.03% Na+, n = 8) diet for 16 weeks and compared with lean, db/+ mice on HS diet (n = 8). Insulin levels were 50% lower in the db/db mice on LS diet when compared with HS db/db (p < 0.05). LS diet increased cardiac adiponectin mRNA levels in db/db mice by 5-fold when compared with db/db mice on HS diet and by 2-fold when compared with HS lean mice (both p < 0.01). LS diet increased adiponectin in adipose tissue compared with db/db mice on HS diet, achieving levels similar to those of lean mice. MCP-1, IL-6 and TNF-α expression were reduced more than 50% in adipose tissue of db/db mice on LS diet when compared with HS db/db mice (all p < 0.05), to levels observed in the HS lean mice. Further, LS db/db mice had significantly reduced circulating MCP-1 and IL-6 levels when compared with HS db/db mice (both p < 0.01). CONCLUSION In obese-diabetic mice, long-term LS diet increases adiponectin in heart and adipose tissue and reduces pro-inflammatory factors in adipose tissue and plasma. These additive mechanisms may contribute to the potential cardioprotective benefits of LS diet in obesity-related metabolic disorders.
Collapse
Affiliation(s)
- R Baudrand
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 221 Longwood Avenue, Boston, MA 02115, USA; Department of Endocrinology, School of Medicine, Pontificia Universidad Catolica de Chile, Santiago 8330074, Chile
| | - C G Lian
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 221 Longwood Avenue, Boston, MA 02115, USA; Division of Dermatopathology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - B Q Lian
- Department of Medicine, University of Massachusetts, Worcester, MA 01605, USA
| | - V Ricchiuti
- Department of Pathology and Medicine, University of Cincinnati, Cincinnati, OH 45219, USA
| | - T M Yao
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 221 Longwood Avenue, Boston, MA 02115, USA
| | - J Li
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 221 Longwood Avenue, Boston, MA 02115, USA
| | - G H Williams
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 221 Longwood Avenue, Boston, MA 02115, USA
| | - G K Adler
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 221 Longwood Avenue, Boston, MA 02115, USA.
| |
Collapse
|
85
|
Mokadem M, Zechner JF, Margolskee RF, Drucker DJ, Aguirre V. Effects of Roux-en-Y gastric bypass on energy and glucose homeostasis are preserved in two mouse models of functional glucagon-like peptide-1 deficiency. Mol Metab 2013; 3:191-201. [PMID: 24634822 DOI: 10.1016/j.molmet.2013.11.010] [Citation(s) in RCA: 139] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 11/21/2013] [Accepted: 11/25/2013] [Indexed: 12/13/2022] Open
Abstract
Glucagon-like peptide-1 (GLP-1) secretion is greatly enhanced after Roux-en-Y gastric bypass (RYGB). While intact GLP-1exerts its metabolic effects via the classical GLP-1 receptor (GLP-1R), proteolytic processing of circulating GLP-1 yields metabolites such as GLP-1(9-36)amide/GLP-1(28-36)amide, that exert similar effects independent of the classical GLP-1R. We investigated the hypothesis that GLP-1, acting via these metabolites or through its known receptor, is required for the beneficial effects of RYGB using two models of functional GLP-1 deficiency - α-gustducin-deficient (α-Gust (-/-)) mice, which exhibit attenuated nutrient-stimulated GLP-1 secretion, and GLP-1R-deficient mice. We show that the effect of RYGB to enhance glucose-stimulated GLP-1 secretion was greatly attenuated in α-Gust (-/-) mice. In both genetic models, RYGB reduced body weight and improved glucose homeostasis to levels observed in lean control mice. Therefore, GLP-1, acting through its classical GLP-1R or its bioactive metabolites, does not seem to be involved in the effects of RYGB on body weight and glucose homeostasis.
Collapse
Affiliation(s)
- Mohamad Mokadem
- Center for Hypothalamic Research, Division of Digestive and Liver Diseases, Department of Internal Medicine, UT Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Juliet F Zechner
- Center for Hypothalamic Research, Division of Digestive and Liver Diseases, Department of Internal Medicine, UT Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Robert F Margolskee
- Monell Chemical Senses Center, 3500 Market Street, Philadelphia, PA 19104, USA
| | - Daniel J Drucker
- Department of Medicine, Mt. Sinai Hospital, Samuel Lunenfeld Research Institute, University of Toronto, 600 University Avenue, Toronto, Ontario, Canada M5G 1X5
| | - Vincent Aguirre
- Center for Hypothalamic Research, Division of Digestive and Liver Diseases, Department of Internal Medicine, UT Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| |
Collapse
|
86
|
Lu Y, Xi W, Ding X, Fan S, Zhang Y, Jiang D, Li Y, Huang C, Zhou Z. Citrange fruit extracts alleviate obesity-associated metabolic disorder in high-fat diet-induced obese C57BL/6 mouse. Int J Mol Sci 2013; 14:23736-50. [PMID: 24317433 PMCID: PMC3876074 DOI: 10.3390/ijms141223736] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2013] [Revised: 11/13/2013] [Accepted: 11/18/2013] [Indexed: 02/06/2023] Open
Abstract
Obesity is becoming one of the global epidemics of the 21st century. In this study, the effects of citrange (Citrus sinensis × Poncirus trifoliata) fruit extracts in high-fat (HF) diet-induced obesity mice were studied. Female C57BL/6 mice were fed respectively a chow diet (control), an HF diet, HF diet supplemented with 1% w/w citrange peel extract (CPE) or 1% w/w citrange flesh and seed extract (CFSE) for 8 weeks. Our results showed that both CPE and CFSE regulated the glucose metabolic disorders of obese mice. In CPE and CFSE-treated groups, the body weight gain, blood glucose, serum total cholesterol (TC) and low density lipoprotein cholesterol (LDL-c) levels were significantly (p < 0.05) reduced relative to those in the HF group. To explore the mechanisms of action of CPE and CFSE on the metabolism of glucose and lipid, related genes’ expressions in liver were assayed. In liver tissue, the expression level of peroxisome proliferator-activated receptor γ (PPARγ) and its target genes were down-regulated by CPE and CFSE supplementation as revealed by qPCR tests. In addition, both CPE and CFSE decreased the expression level of liver X receptor (LXR) α and β, which are involved in lipid and glucose metabolism. Taken together, these results suggest that CPE and CFSE administration could ameliorate obesity and related metabolic disorders in HF diet-induced obesity mice probably through the inhibition of PPARγ and LXRs gene expressions.
Collapse
Affiliation(s)
- Yan Lu
- College of Horticulture and Landscape Architecture, Southwest University, Chongqing 400716, China; E-Mails: (Y.L.); (W.X.); (X.D.)
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; E-Mails: (S.F.); (Y.Z.); (Y.L.)
- Key Laboratory of Horticulture Science for Southern Mountainous Regions, Ministry of Education, Chongqing 400715, China
| | - Wanpeng Xi
- College of Horticulture and Landscape Architecture, Southwest University, Chongqing 400716, China; E-Mails: (Y.L.); (W.X.); (X.D.)
| | - Xiaobo Ding
- College of Horticulture and Landscape Architecture, Southwest University, Chongqing 400716, China; E-Mails: (Y.L.); (W.X.); (X.D.)
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; E-Mails: (S.F.); (Y.Z.); (Y.L.)
- Key Laboratory of Horticulture Science for Southern Mountainous Regions, Ministry of Education, Chongqing 400715, China
| | - Shengjie Fan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; E-Mails: (S.F.); (Y.Z.); (Y.L.)
| | - Yu Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; E-Mails: (S.F.); (Y.Z.); (Y.L.)
| | - Dong Jiang
- Citrus Research Institute, Chinese Academy Agricultural Science, Chongqing 400712, China; E-Mail:
| | - Yiming Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; E-Mails: (S.F.); (Y.Z.); (Y.L.)
| | - Cheng Huang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; E-Mails: (S.F.); (Y.Z.); (Y.L.)
- Authors to whom correspondence should be addressed; E-Mails: (Z.Z.); (C.H.); Tel.: +86-23-6825-0229 (Z.Z.); +86-21-5132-2182 (C.H.); Fax: +86-23-6825-1274 (Z.Z.); +86-21-5132-2193 (C.H.)
| | - Zhiqin Zhou
- College of Horticulture and Landscape Architecture, Southwest University, Chongqing 400716, China; E-Mails: (Y.L.); (W.X.); (X.D.)
- Key Laboratory of Horticulture Science for Southern Mountainous Regions, Ministry of Education, Chongqing 400715, China
- Authors to whom correspondence should be addressed; E-Mails: (Z.Z.); (C.H.); Tel.: +86-23-6825-0229 (Z.Z.); +86-21-5132-2182 (C.H.); Fax: +86-23-6825-1274 (Z.Z.); +86-21-5132-2193 (C.H.)
| |
Collapse
|
87
|
Jang HJ, Ridgeway SD, Kim JA. Effects of the green tea polyphenol epigallocatechin-3-gallate on high-fat diet-induced insulin resistance and endothelial dysfunction. Am J Physiol Endocrinol Metab 2013; 305:E1444-51. [PMID: 24148349 PMCID: PMC3882381 DOI: 10.1152/ajpendo.00434.2013] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Insulin resistance, a hallmark of metabolic disorders, is a risk factor for diabetes and cardiovascular disease. Impairment of insulin responsiveness in vascular endothelium contributes to insulin resistance. The reciprocal relationship between insulin resistance and endothelial dysfunction augments the pathophysiology of metabolism and cardiovascular functions. The most abundant green tea polyphenol, epigallocatechin-3-gallate (EGCG), has been shown to have vasodilator action in vessels by activation of endothelial nitric oxide synthase (eNOS). However, it is not known whether EGCG has a beneficial effect in high-fat diet (HFD)-induced endothelial dysfunction. Male C57BL/6J mice were fed either a normal chow diet (NCD) or HFD with or without EGCG supplement (50 mg·kg(-1)·day(-1)) for 10 wk. Mice fed a HFD with EGCG supplement gained less body weight and showed improved insulin sensitivity. In vehicle-treated HFD mice, endothelial function was impaired in response to insulin but not to acetylcholine, whereas the EGCG-treated HFD group showed improved insulin-stimulated vasodilation. Interestingly, EGCG intake reduced macrophage infiltration into aortic tissues in HFD mice. Treatment with EGCG restored the insulin-stimulated phosphorylation of eNOS, insulin receptor substrate-1 (IRS-1), and protein kinase B (Akt), which was inhibited by palmitate (200 μM, 5 h) in primary bovine aortic endothelial cells. From these results, we conclude that supplementation of EGCG improves glucose tolerance, insulin sensitivity, and endothelial function. The results suggest that EGCG may have beneficial health effects in glucose metabolism and endothelial function through modulating HFD-induced inflammatory response.
Collapse
Affiliation(s)
- Hyun-Ju Jang
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, Alabama
| | | | | |
Collapse
|
88
|
|
89
|
Wang X, Hansen BC, Shi D, Fang Y, Du F, Wang B, Chen YM, Gregoire FM, Wang YXJ. Quantification of β-cell insulin secretory function using a graded glucose infusion with C-peptide deconvolution in dysmetabolic, and diabetic cynomolgus monkeys. Diabetol Metab Syndr 2013; 5:40. [PMID: 23886319 PMCID: PMC3728033 DOI: 10.1186/1758-5996-5-40] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Accepted: 07/16/2013] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Quantitation of β-cell function is critical in better understanding of the dynamic interactions of insulin secretion, clearance and action at different phases in the progression of diabetes. The present study aimed to quantify β-cell secretory function independently of insulin sensitivity in the context of differential metabolic clearance rates of insulin (MCRI) in nonhuman primates (NHPs). METHODS Insulin secretion rate (ISR) was derived from deconvolution of serial C-peptide concentrations measured during a 5 stage graded glucose infusion (GGI) in 12 nondiabetic (N), 8 prediabetic or dysmetabolic (DYS) and 4 overtly diabetic (DM) cynomolgus monkeys. The characterization of the monkeys was based on the fasting glucose and insulin concentrations, glucose clearance rate measured by intravenous glucose tolerance test, and insulin resistance indices measured in separate experiments. The molar ratio of C-peptide/insulin (C/I) was used as a surrogate index of hepatic MCRI. RESULTS Compared to the N monkeys, the DYS with normal glycemia and hyperinsulinemia had significantly higher basal and GGI-induced elevation of insulin and C-peptide concentrations and lower C/I, however, each unit of glucose-stimulated ISR increment was not significantly different from that in the N monkeys. In contrast, the DM monkeys with β-cell failure and hyperglycemia had a depressed GGI-stimulated ISR response and elevated C/I. CONCLUSIONS The present data demonstrated that in addition to β-cell hypersecretion of insulin, reduced hepatic MCRI may also contribute to the development of hyperinsulinemia in the DYS monkeys. On the other hand, hyperinsulinemia may cause the saturation of hepatic insulin extraction capacity, which in turn reduced MCRI in the DYS monkeys. The differential contribution of ISR and MCRI in causing hyperinsulinemia provides a new insight into the trajectory of β-cell dysfunction in the development of diabetes. The present study was the first to use the GGI and C-peptide deconvolution method to quantify the β-cell function in NHPs.
Collapse
Affiliation(s)
- Xiaoli Wang
- Cardiovascular and Metabolic Diseases Research, Crown Bioscience Inc., 6 Beijing West Road, Science & Technology Park, Taicang Economic Development Area, Taicang, Jiangsu Province 215400, P.R. China
- International Institute of Biomedical Research (IIBR), a Crown Bioscience Company at David H. Murdock Research Institute (DHMRI), Kannapolis, NC, USA
| | - Barbara C Hansen
- International Institute of Biomedical Research (IIBR), a Crown Bioscience Company at David H. Murdock Research Institute (DHMRI), Kannapolis, NC, USA
- Departments of Internal Medicine and Pediatrics, and Obesity, Diabetes and Aging Research Center, University of South Florida, Tampa, FL, USA
| | - Da Shi
- Cardiovascular and Metabolic Diseases Research, Crown Bioscience Inc., 6 Beijing West Road, Science & Technology Park, Taicang Economic Development Area, Taicang, Jiangsu Province 215400, P.R. China
| | - Yupeng Fang
- Cardiovascular and Metabolic Diseases Research, Crown Bioscience Inc., 6 Beijing West Road, Science & Technology Park, Taicang Economic Development Area, Taicang, Jiangsu Province 215400, P.R. China
- International Institute of Biomedical Research (IIBR), a Crown Bioscience Company at David H. Murdock Research Institute (DHMRI), Kannapolis, NC, USA
| | - Fenglai Du
- Cardiovascular and Metabolic Diseases Research, Crown Bioscience Inc., 6 Beijing West Road, Science & Technology Park, Taicang Economic Development Area, Taicang, Jiangsu Province 215400, P.R. China
- International Institute of Biomedical Research (IIBR), a Crown Bioscience Company at David H. Murdock Research Institute (DHMRI), Kannapolis, NC, USA
| | - Bingdi Wang
- Cardiovascular and Metabolic Diseases Research, Crown Bioscience Inc., 6 Beijing West Road, Science & Technology Park, Taicang Economic Development Area, Taicang, Jiangsu Province 215400, P.R. China
- International Institute of Biomedical Research (IIBR), a Crown Bioscience Company at David H. Murdock Research Institute (DHMRI), Kannapolis, NC, USA
| | - Yaxiong Michael Chen
- Cardiovascular and Metabolic Diseases Research, Crown Bioscience Inc., 6 Beijing West Road, Science & Technology Park, Taicang Economic Development Area, Taicang, Jiangsu Province 215400, P.R. China
| | - Francine M Gregoire
- Cardiovascular and Metabolic Diseases Research, Crown Bioscience Inc., 6 Beijing West Road, Science & Technology Park, Taicang Economic Development Area, Taicang, Jiangsu Province 215400, P.R. China
- International Institute of Biomedical Research (IIBR), a Crown Bioscience Company at David H. Murdock Research Institute (DHMRI), Kannapolis, NC, USA
| | - Yi-Xin Jim Wang
- Cardiovascular and Metabolic Diseases Research, Crown Bioscience Inc., 6 Beijing West Road, Science & Technology Park, Taicang Economic Development Area, Taicang, Jiangsu Province 215400, P.R. China
- International Institute of Biomedical Research (IIBR), a Crown Bioscience Company at David H. Murdock Research Institute (DHMRI), Kannapolis, NC, USA
| |
Collapse
|
90
|
Jang HJ, Kim HS, Hwang DH, Quon MJ, Kim JA. Toll-like receptor 2 mediates high-fat diet-induced impairment of vasodilator actions of insulin. Am J Physiol Endocrinol Metab 2013; 304:E1077-88. [PMID: 23531618 PMCID: PMC3651621 DOI: 10.1152/ajpendo.00578.2012] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Obesity is characterized by a chronic proinflammatory state that leads to endothelial dysfunction. Saturated fatty acids (SFA) stimulate Toll-like receptors (TLR) that promote metabolic insulin resistance. However, it is not known whether TLR2 mediates impairment of vascular actions of insulin in response to high-fat diet (HFD) to cause endothelial dysfunction. siRNA knockdown of TLR2 in primary endothelial cells opposed palmitate-stimulated expression of proinflammatory cytokines and splicing of X box protein 1 (XBP-1). Inhibition of unfolding protein response (UPR) reduced SFA-stimulated expression of TNFα. Thus, SFA stimulates UPR and proinflammatory response through activation of TLR2 in endothelial cells. Knockdown of TLR2 also opposed impairment of insulin-stimulated phosphorylation of eNOS and subsequent production of NO. Importantly, insulin-stimulated vasorelaxation of mesenteric arteries from TLR2 knockout mice was preserved even on HFD (in contrast with results from arteries examined in wild-type mice on HFD). We conclude that TLR2 in vascular endothelium mediates HFD-stimulated proinflammatory responses and UPR that accompany impairment of vasodilator actions of insulin, leading to endothelial dysfunction. These results are relevant to understanding the pathophysiology of the cardiovascular complications of diabetes and obesity.
Collapse
Affiliation(s)
- Hyun-Ju Jang
- Dept. of Medicine, Div. of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | | | | |
Collapse
|
91
|
Clemmensen C, Smajilovic S, Madsen AN, Klein AB, Holst B, Bräuner-Osborne H. Increased susceptibility to diet-induced obesity in GPRC6A receptor knockout mice. J Endocrinol 2013; 217:151-60. [PMID: 23428581 DOI: 10.1530/joe-12-0550] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The recently identified G protein-coupled receptor GPRC6A is activated by dietary amino acids and expressed in multiple tissues. Although the receptor is hypothesised to exert biological impact on metabolic and endocrine-related parameters, the role of the receptor in obesity and metabolic complications is still elusive. In the present study, we investigated the impact of GPRC6A deficiency in a murine model of diet-induced obesity (DIO). Male Gprc6a knockout (KO) mice and WT littermates were subjected to a high-fat diet (HFD) for 25 weeks and exposed to comprehensive metabolic phenotyping. A significant increase in body weight, corresponding to a selective increase in body fat, was observed in Gprc6a KO mice exposed to an HFD relative to WT controls. The obese phenotype was linked to subtle perturbations in energy homoeostasis as GPRC6A deficiency resulted in chronic hyperphagia and decreased locomotor activity. Moreover, diet-induced obese Gprc6a KO mice had increased circulating insulin and leptin levels relative to WT animals, thereby demonstrating that endocrine abnormalities associate with the reported disturbances in energy balance. The phenotype was further accompanied by disruptions in glucose metabolism showing that Gprc6a KO mice on an HFD display increased susceptibility to develop metabolic-related disorders. Altogether, these data suggest that the amino acid sensing receptor GPRC6A plays an important role in resistance to DIO and metabolic complications. Future studies will illuminate the underlying molecular mechanisms mediating the herein reported findings and potentially facilitate the development of novel therapeutic compounds targeting the GPRC6A receptor.
Collapse
Affiliation(s)
- Christoffer Clemmensen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | | | | | | | | | | |
Collapse
|
92
|
Zechner JF, Mirshahi UL, Satapati S, Berglund ED, Rossi J, Scott MM, Still CD, Gerhard GS, Burgess SC, Mirshahi T, Aguirre V. Weight-independent effects of roux-en-Y gastric bypass on glucose homeostasis via melanocortin-4 receptors in mice and humans. Gastroenterology 2013; 144:580-590.e7. [PMID: 23159449 PMCID: PMC3835150 DOI: 10.1053/j.gastro.2012.11.022] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Revised: 11/08/2012] [Accepted: 11/11/2012] [Indexed: 01/06/2023]
Abstract
BACKGROUND & AIMS Roux-en-Y gastric bypass (RYGB) improves glucose homeostasis independently of changes in body weight by unknown mechanisms. Melanocortin-4 receptors (MC4R) have weight-independent effects on glucose homeostasis, via autonomic neurons, and also might contribute to weight loss after RYGB. We investigated whether MC4Rs mediate effects of RYGB, such as its weight-independent effects on glucose homeostasis, in mice and humans. METHODS We studied C57BL/6 mice with diet-induced obesity, MC4R-deficient mice, and mice that re-express MC4R specifically in autonomic neurons after RYGB or sham surgeries. We also sequenced the MC4R locus in patients undergoing RYGB to investigate diabetes resolution in carriers of rare MC4R variants. RESULTS MC4Rs in autonomic brainstem neurons (including the parasympathetic dorsal motor vagus) mediated improved glucose homeostasis independent of changes in body weight. In contrast, MC4Rs in cholinergic preganglionic motor neurons (sympathetic and parasympathetic) mediated RYGB-induced increased energy expenditure and weight loss. Increased energy expenditure after RYGB is the predominant mechanism of weight loss and confers resistance to weight gain from a high-fat diet, the effects of which are MC4R-dependent. MC4R-dependent effects of RYGB still occurred in mice with Mc4r haplosufficiency, and early stage diabetes resolved at a similar rate in patients with rare variants of MC4R and noncarriers. However, carriers of MC4R (I251L), a rare variant associated with increased weight loss after RYGB and increased basal activity in vitro, were more likely to have early and weight-independent resolution of diabetes than noncarriers, indicating a role for MC4Rs in the effects of RYGB. CONCLUSIONS MC4Rs in autonomic neurons mediate beneficial effects of RYGB, including weight-independent improved glucose homeostasis, in mice and humans.
Collapse
Affiliation(s)
- Juliet F. Zechner
- UT Southwestern Medical Center, Dallas, TX 75390, USA
- Center for Hypothalamic Research, Department of Medicine
| | - Uyenlinh L. Mirshahi
- Weis Center for Research and Geisinger Obesity Research Institute, Geisinger Clinic, Danville, PA 17822, USA
| | - Santhosh Satapati
- UT Southwestern Medical Center, Dallas, TX 75390, USA
- Advanced Imaging Center, Department of Pharmacology
| | - Eric D. Berglund
- UT Southwestern Medical Center, Dallas, TX 75390, USA
- Center for Hypothalamic Research, Department of Medicine
| | - Jari Rossi
- Institute of Biomedicine, Anatomy, University of Helsinki, Helsinki, Finland
| | - Michael M. Scott
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22903, USA
| | - Christopher D. Still
- Weis Center for Research and Geisinger Obesity Research Institute, Geisinger Clinic, Danville, PA 17822, USA
| | - Glenn S. Gerhard
- Weis Center for Research and Geisinger Obesity Research Institute, Geisinger Clinic, Danville, PA 17822, USA
| | - Shawn C. Burgess
- UT Southwestern Medical Center, Dallas, TX 75390, USA
- Advanced Imaging Center, Department of Pharmacology
| | - Tooraj Mirshahi
- Weis Center for Research and Geisinger Obesity Research Institute, Geisinger Clinic, Danville, PA 17822, USA
| | - Vincent Aguirre
- UT Southwestern Medical Center, Dallas, TX 75390, USA
- Center for Hypothalamic Research, Department of Medicine
| |
Collapse
|
93
|
Gargani S, Thévenet J, Yuan JE, Lefebvre B, Delalleau N, Gmyr V, Hubert T, Duhamel A, Pattou F, Kerr-Conte J. Adaptive changes of human islets to an obesogenic environment in the mouse. Diabetologia 2013; 56. [PMID: 23192693 PMCID: PMC3536990 DOI: 10.1007/s00125-012-2775-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
AIMS/HYPOTHESIS In this study, we used an immunodeficient mouse model to explore, in vivo, the longitudinal adaptation of human islets to an obesogenic environment. METHODS Non-diabetic Rag2 (-/-) mice (n = 61) were transplanted with human islets (400 islet equivalents [IEQ]) from six pancreases: four non-diabetic and two with overt metabolic dysfunction (older, high HbA(lc) or history of diabetes). Animals were fed for 12 weeks with a control or high-fat diet (HFD), and followed for weight, serum triacylglycerol, fasting blood glucose and human C-peptide. After the mice were killed, human grafts and the endogenous pancreas were analysed for endocrine volume, distribution of beta and alpha cells, and proliferation. RESULTS Transplanted mice on an HFD gained significantly more weight (p < 0.001) and had higher fasting glycaemia (2-12 weeks; p = 0.0002) and consistently higher fasting human C-peptide levels (2-12 weeks; p = 0.04) compared with those on the control diet. Histology demonstrated doubling of human islet graft volume at 12 weeks in animals on the HFD and increased beta cell volume (p < 0.001), but no change in alpha cell volume. Human islet function (hyperbolic product HOMA2%BS) at 12 weeks was four times lower in HFD animals (p < 0.001 vs controls) because of insufficient beta cell adaptation to decreased (70%) sensitivity (HOMA%S). Human islets obtained from donors with metabolic dysfunction failed to adapt to the HFD. CONCLUSIONS/INTERPRETATION This longitudinal study provides direct evidence that human islets adapt both endocrine and beta cell mass, function and gene expression to obesity in vivo. The present model will facilitate the identification of mechanisms by which human islets adapt to obesity in vivo and the cell type(s) responsible, and factors predisposing human beta cells to decompensation.
Collapse
Affiliation(s)
- S. Gargani
- Université Lille Nord de France, Lille, France
- UDSL, IMPRT, Lille, France
- Inserm U859 Biotherapies of Diabetes, Faculty of Medicine, 1 Place de Verdun, F59045 Lille, France
| | - J. Thévenet
- Université Lille Nord de France, Lille, France
- UDSL, IMPRT, Lille, France
- Inserm U859 Biotherapies of Diabetes, Faculty of Medicine, 1 Place de Verdun, F59045 Lille, France
| | - J. E. Yuan
- Université Lille Nord de France, Lille, France
| | - B. Lefebvre
- Université Lille Nord de France, Lille, France
- UDSL, IMPRT, Lille, France
- Present Address: Inserm U837, Alzheimer and Tauopathies, Lille, France
| | - N. Delalleau
- Université Lille Nord de France, Lille, France
- UDSL, IMPRT, Lille, France
- Inserm U859 Biotherapies of Diabetes, Faculty of Medicine, 1 Place de Verdun, F59045 Lille, France
| | - V. Gmyr
- Université Lille Nord de France, Lille, France
- UDSL, IMPRT, Lille, France
- Inserm U859 Biotherapies of Diabetes, Faculty of Medicine, 1 Place de Verdun, F59045 Lille, France
| | - T. Hubert
- Université Lille Nord de France, Lille, France
- UDSL, IMPRT, Lille, France
- Inserm U859 Biotherapies of Diabetes, Faculty of Medicine, 1 Place de Verdun, F59045 Lille, France
| | - A. Duhamel
- Université Lille Nord de France, Lille, France
- UDSL, IMPRT, Lille, France
- Centre Hospitalier Régional Universitaire de Lille, Lille, France
- CERIM, Faculty of Medicine, Lille, France
| | - F. Pattou
- Université Lille Nord de France, Lille, France
- UDSL, IMPRT, Lille, France
- Inserm U859 Biotherapies of Diabetes, Faculty of Medicine, 1 Place de Verdun, F59045 Lille, France
- Centre Hospitalier Régional Universitaire de Lille, Lille, France
| | - J. Kerr-Conte
- Université Lille Nord de France, Lille, France
- UDSL, IMPRT, Lille, France
- Inserm U859 Biotherapies of Diabetes, Faculty of Medicine, 1 Place de Verdun, F59045 Lille, France
- Centre Hospitalier Régional Universitaire de Lille, Lille, France
| |
Collapse
|
94
|
Saxagliptin Improves Glucose Tolerance but not Survival in a Murine Model of Dilated Cardiomyopathy. Cardiovasc Endocrinol 2012; 1:74-82. [PMID: 23795310 DOI: 10.1097/xce.0b013e32835bfb24] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Glucagon-like peptide 1 (GLP-1) agonists improve myocardial function and insulin sensitivity in the setting of chronic heart failure. Endogenously produced GLP-1 peptide (7-36) is rapidly cleaved by dipeptidyl peptidase 4 (DPP4) to the 9-36 peptide, which lacks anti-hyperglycemic activity. To elucidate the effect of increased endogenous GLP-1 during heart failure progression, the DPP4 inhibitor saxagliptin or vehicle was administered by daily oral gavage to female TG9 mice, a transgenic model of dilated cardiomyopathy, starting at day of life 42, just prior to the development of detectable contractile dysfunction. Saxagliptin treatment inhibited DPP4 activity >90% and increased GLP-1 levels 4-fold following a 2 gm/kg glucose load but did not affect fasting GLP-1 levels. There was no difference in food intake or body weight between groups. At 56 days of age, oral glucose tolerance was improved in saxagliptin-versus vehicle-treated animals (AUC0-120 1340 ± 46 and 1501 ± 43 min·mmol/L, respectively, p<0.015). In contrast to the effect of a GLP-1 agonist in TG9 mice, saxagliptin had no effect on survival (80.7 ± 4.3 days) compared to vehicle-treated mice (79.6 ± 3.6 days, p = 0.46). Taken together, these data indicate that improvement in glucose tolerance is not sufficient to improve survival. Future efforts to confirm these findings in additional models of heart failure are warranted.
Collapse
|
95
|
Kelley DS, Adkins Y, Woodhouse LR, Swislocki A, Mackey BE, Siegel D. Docosahexaenoic Acid Supplementation Improved Lipocentric but Not Glucocentric Markers of Insulin Sensitivity in Hypertriglyceridemic Men. Metab Syndr Relat Disord 2012; 10:32-8. [DOI: 10.1089/met.2011.0081] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- Darshan S. Kelley
- Western Human Nutrition Research Center, Agricultural Research Service, U.S. Department of Agriculture, and Department of Nutrition, University of California, Davis, California
| | - Yuriko Adkins
- Western Human Nutrition Research Center, Agricultural Research Service, U.S. Department of Agriculture, and Department of Nutrition, University of California, Davis, California
| | - Leslie R. Woodhouse
- Western Human Nutrition Research Center, Agricultural Research Service, U.S. Department of Agriculture, and Department of Nutrition, University of California, Davis, California
| | - Arthur Swislocki
- Veteran Affairs Northern California Health Care System, Martinez, California
- Department of Medicine, School of Medicine, University of California Davis, Sacramento, California
| | - Bruce E. Mackey
- Western Regional Research Center, Agricultural Research Service, U.S. Department of Agriculture, Albany, California
| | - David Siegel
- Department of Medicine, School of Medicine, University of California Davis, Sacramento, California
- Veteran Affairs Northern California Health Care System, Sacramento, California
| |
Collapse
|
96
|
De Benedictis L, Potenza MA, Gagliardi S, Zigrino A, Montagnani M, De Salvia MA. Rosiglitazone reverses increased duodenal inhibitory response in spontaneously hypertensive rats. Neurogastroenterol Motil 2012; 24:e56-66. [PMID: 21995307 DOI: 10.1111/j.1365-2982.2011.01798.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND Thiazolidinediones (TZDs) including rosiglitazone (ROSI) are insulin sensitizing agents with beneficial gastrointestinal effects. However, no studies are available on TZDs effect in gastrointestinal motility. We evaluated the effects of ROSI on gastrointestinal inhibitory neurotransmission focusing on the modulatory roles of nitric oxide synthase/nitric oxide (NOS/NO) and heme oxygenase/carbon monoxide (HO/CO) pathways. METHODS Spontaneously hypertensive rats (SHR) were used as model of insulin resistance. Duodenal strips were obtained from vehicle-treated SHR, ROSI-treated SHR (5 mg kg(-1) by gavage daily per 6 weeks), and Wistar Kyoto (WKY). Inhibitory responses to electrical field stimulation (EFS) were evaluated in the presence of HO inhibitor zinc protoporphyrin IX (ZnPPIX, 10 μmol L(-1)) or NOS inhibitor N(G)-nitro-L-arginine (L-NNA, 100 μmol L(-1)), alone and in combination. Protein levels of HO and NOS isoforms were evaluated by immunohistochemistry and western blot analysis. KEY RESULTS Basal responses to EFS were significantly increased in duodenum strips from vehicle-treated SHR vs WKY. This effect was reversed in ROSI-treated SHR. The EFS-mediated relaxation was comparably reduced by ZnPPIX in WKY and SHR, but not in ROSI-treated SHR animals. The L-NNA reduced EFS response to a similar extent in WKY and ROSI -treated SHR, but its effect was significantly higher in vehicle-treated SHR. Expression of HO-1 protein was significantly lower, whereas HO-2 protein levels were unchanged in ROSI-treated SHR with respect to vehicle-treated SHR. Finally, increased levels of nNOS in vehicle-treated SHR were reduced in ROSI-treated SHR. CONCLUSIONS & INFERENCES Chronic ROSI treatment reverses increased SHR duodenal inhibitory response acting on CO and NO components.
Collapse
Affiliation(s)
- L De Benedictis
- Department of Biomedical Sciences and Human Oncology, Medical School, University of Bari Aldo Moro, Bari, Italy
| | | | | | | | | | | |
Collapse
|
97
|
Clemmensen C, Madsen AN, Smajilovic S, Holst B, Bräuner-Osborne H. L-Arginine improves multiple physiological parameters in mice exposed to diet-induced metabolic disturbances. Amino Acids 2011; 43:1265-75. [PMID: 22200933 DOI: 10.1007/s00726-011-1199-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2011] [Accepted: 12/06/2011] [Indexed: 12/16/2022]
Abstract
L-Arginine (L-Arg) is a conditionally essential amino acid and a natural constituent of dietary proteins. Studies in obese rats and type 2 diabetic humans have indicated that dietary supplementation with L-Arg can diminish gain in white adipose tissue (WAT) and improve insulin sensitivity. However, the effects of L-Arg on glucose homeostasis, body composition and energy metabolism remain unclear. In addition, no studies have, to our knowledge, examined whether L-Arg has beneficial effects as a dietary supplement in the mouse model. In the present study, we investigated the effects of L-Arg supplementation to male C57BL/6 mice on an array of physiological parameters. L-Arg supplemented mice were maintained on a low-protein diet and body composition, appetite regulation, glucose tolerance, insulin sensitivity and energy expenditure were evaluated. A significant reduction in epididymal WAT was observed in L-Arg supplemented mice compared with mice fed an isocaloric control diet. Surprisingly, the L-Arg supplemented animals were hyperphagic corresponding to a highly significant decrease in feed efficiency, as body weight developed in a similar pattern in both experimental groups. Glucose homeostasis experiments revealed a major effect of L-Arg supplementation on glucose tolerance and insulin sensitivity, interestingly, independent of a parallel regulation in whole-body adiposity. Increased L-Arg ingestion also raised energy expenditure; however, no concurrent effect on locomotor activity, substrate metabolism or expression of uncoupling proteins (UCP1 and UCP2) in adipose tissues was displayed. In conclusion, dietary L-Arg supplementation substantially affects an array of metabolic-associated parameters including a reduction in WAT, hyperphagia, improved insulin sensitivity and increased energy expenditure in mice fed a low-protein diet.
Collapse
Affiliation(s)
- Christoffer Clemmensen
- Department of Medicinal Chemistry, Faculty of Pharmaceutical Sciences, University of Copenhagen, Universitetsparken 2, Copenhagen, Denmark
| | | | | | | | | |
Collapse
|
98
|
Gahete MD, Córdoba-Chacón J, Anadumaka CV, Lin Q, Brüning JC, Kahn CR, Luque RM, Kineman RD. Elevated GH/IGF-I, due to somatotrope-specific loss of both IGF-I and insulin receptors, alters glucose homeostasis and insulin sensitivity in a diet-dependent manner. Endocrinology 2011; 152:4825-37. [PMID: 21990313 PMCID: PMC3230054 DOI: 10.1210/en.2011-1447] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
A unique mouse model was developed with elevated endogenous GH (2- to 3-fold) and IGF-I (1.2- to 1.4-fold), due to somatotrope-specific Cre-mediated inactivation of IGF-I receptor (IgfIr) and insulin receptor (Insr) genes (IgfIr,Insr(rGHpCre), referred to as HiGH mice). We demonstrate that the metabolic phenotype of HiGH mice is diet dependent and differs from that observed in other mouse models of GH excess due to ectopic heterologous transgene expression or pituitary tumor formation. Elevated endogenous GH promotes lean mass and whole-body lipid oxidation but has minimal effects on adiposity, even in response to diet-induced obesity. When caloric intake is moderated, elevated GH improves glucose clearance, despite low/normal insulin sensitivity, which may be explained in part by enhanced IGF-I and insulin output. However, when caloric intake is in excess, elevated GH promotes hepatic lipid accumulation, insulin resistance, hyperglycemia, and ketosis. The HiGH mouse model represents a useful tool to study the role endogenous circulating GH levels play in regulating health and disease.
Collapse
Affiliation(s)
- Manuel D Gahete
- Jesse Brown Veterans Affairs Medical Center, Research and Development Division, 820 South Damen Avenue, Chicago, Illinois 60612, USA
| | | | | | | | | | | | | | | |
Collapse
|
99
|
Ait-Omar A, Monteiro-Sepulveda M, Poitou C, Le Gall M, Cotillard A, Gilet J, Garbin K, Houllier A, Château D, Lacombe A, Veyrie N, Hugol D, Tordjman J, Magnan C, Serradas P, Clément K, Leturque A, Brot-Laroche E. GLUT2 accumulation in enterocyte apical and intracellular membranes: a study in morbidly obese human subjects and ob/ob and high fat-fed mice. Diabetes 2011; 60:2598-607. [PMID: 21852673 PMCID: PMC3178286 DOI: 10.2337/db10-1740] [Citation(s) in RCA: 110] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVE In healthy rodents, intestinal sugar absorption in response to sugar-rich meals and insulin is regulated by GLUT2 in enterocyte plasma membranes. Loss of insulin action maintains apical GLUT2 location. In human enterocytes, apical GLUT2 location has not been reported but may be revealed under conditions of insulin resistance. RESEARCH DESIGN AND METHODS Subcellular location of GLUT2 in jejunal enterocytes was analyzed by confocal and electron microscopy imaging and Western blot in 62 well-phenotyped morbidly obese subjects and 7 lean human subjects. GLUT2 locations were assayed in ob/ob and ob/+ mice receiving oral metformin or in high-fat low-carbohydrate diet-fed C57Bl/6 mice. Glucose absorption and secretion were respectively estimated by oral glucose tolerance test and secretion of [U-(14)C]-3-O-methyl glucose into lumen. RESULTS In human enterocytes, GLUT2 was consistently located in basolateral membranes. Apical GLUT2 location was absent in lean subjects but was observed in 76% of obese subjects and correlated with insulin resistance and glycemia. In addition, intracellular accumulation of GLUT2 with early endosome antigen 1 (EEA1) was associated with reduced MGAT4a activity (glycosylation) in 39% of obese subjects on a low-carbohydrate/high-fat diet. Mice on a low-carbohydrate/high-fat diet for 12 months also exhibited endosomal GLUT2 accumulation and reduced glucose absorption. In ob/ob mice, metformin promoted apical GLUT2 and improved glucose homeostasis. Apical GLUT2 in fasting hyperglycemic ob/ob mice tripled glucose release into intestinal lumen. CONCLUSIONS In morbidly obese insulin-resistant subjects, GLUT2 was accumulated in apical and/or endosomal membranes of enterocytes. Functionally, apical GLUT2 favored and endosomal GLUT2 reduced glucose transepithelial exchanges. Thus, altered GLUT2 locations in enterocytes are a sign of intestinal adaptations to human metabolic pathology.
Collapse
Affiliation(s)
- Amal Ait-Omar
- INSERM, U872, Team 9, Paris, France; Centre de Recherche des Cordeliers, Université Pierre et Marie Curie-Paris 6, UMR S 872, Paris, France
| | - Milena Monteiro-Sepulveda
- INSERM, U872, Team 9, Paris, France; Centre de Recherche des Cordeliers, Université Pierre et Marie Curie-Paris 6, UMR S 872, Paris, France
| | - Christine Poitou
- INSERM, U872, Team 7 Nutriomique, Paris, France; Centre de Recherche des Cordeliers, Université Pierre et Marie Curie-Paris 6, UMR S 872, Paris, France
- Nutrition and Endocrinology Department, Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Paris, France; Centre Recherche en Nutrition Humaine (CRNH) Ile de France, Paris, France
| | - Maude Le Gall
- INSERM, U872, Team 9, Paris, France; Centre de Recherche des Cordeliers, Université Pierre et Marie Curie-Paris 6, UMR S 872, Paris, France
| | - Aurélie Cotillard
- INSERM, U872, Team 7 Nutriomique, Paris, France; Centre de Recherche des Cordeliers, Université Pierre et Marie Curie-Paris 6, UMR S 872, Paris, France
| | - Jules Gilet
- INSERM, U872, Team 9, Paris, France; Centre de Recherche des Cordeliers, Université Pierre et Marie Curie-Paris 6, UMR S 872, Paris, France
| | - Kevin Garbin
- INSERM, U872, Team 9, Paris, France; Centre de Recherche des Cordeliers, Université Pierre et Marie Curie-Paris 6, UMR S 872, Paris, France
| | - Anne Houllier
- INSERM, U872, Team 9, Paris, France; Centre de Recherche des Cordeliers, Université Pierre et Marie Curie-Paris 6, UMR S 872, Paris, France
| | - Danièle Château
- INSERM, U872, Team 9, Paris, France; Centre de Recherche des Cordeliers, Université Pierre et Marie Curie-Paris 6, UMR S 872, Paris, France
| | - Amélie Lacombe
- Centre National de la Recherche Scientifique (EAC4413), Université Paris-Diderot, Paris, France
| | - Nicolas Veyrie
- INSERM, U872, Team 7 Nutriomique, Paris, France; Centre de Recherche des Cordeliers, Université Pierre et Marie Curie-Paris 6, UMR S 872, Paris, France
- Surgery Department, Assistance Publique-Hôpitaux de Paris, Hôtel-Dieu Hospital, Paris, France
| | - Danielle Hugol
- Pathology Department, Assistance Publique-Hôpitaux de Paris, Hôtel-Dieu Hospital, Paris, France
| | - Joan Tordjman
- INSERM, U872, Team 7 Nutriomique, Paris, France; Centre de Recherche des Cordeliers, Université Pierre et Marie Curie-Paris 6, UMR S 872, Paris, France
| | - Christophe Magnan
- Centre National de la Recherche Scientifique (EAC4413), Université Paris-Diderot, Paris, France
| | - Patricia Serradas
- INSERM, U872, Team 9, Paris, France; Centre de Recherche des Cordeliers, Université Pierre et Marie Curie-Paris 6, UMR S 872, Paris, France
| | - Karine Clément
- INSERM, U872, Team 7 Nutriomique, Paris, France; Centre de Recherche des Cordeliers, Université Pierre et Marie Curie-Paris 6, UMR S 872, Paris, France
- Nutrition and Endocrinology Department, Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Paris, France; Centre Recherche en Nutrition Humaine (CRNH) Ile de France, Paris, France
| | - Armelle Leturque
- INSERM, U872, Team 9, Paris, France; Centre de Recherche des Cordeliers, Université Pierre et Marie Curie-Paris 6, UMR S 872, Paris, France
| | - Edith Brot-Laroche
- INSERM, U872, Team 9, Paris, France; Centre de Recherche des Cordeliers, Université Pierre et Marie Curie-Paris 6, UMR S 872, Paris, France
- Corresponding author: Edith Brot-Laroche,
| |
Collapse
|
100
|
Petersen PS, Jin C, Madsen AN, Rasmussen M, Kuhre R, Egerod KL, Nielsen LB, Schwartz TW, Holst B. Deficiency of the GPR39 receptor is associated with obesity and altered adipocyte metabolism. FASEB J 2011; 25:3803-14. [DOI: 10.1096/fj.11-184531] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Pia Steen Petersen
- Laboratory for Molecular Pharmacology, Department of Neuroscience and PharmacologyUniversity of Copenhagen Copenhagen Denmark
| | - Chunyu Jin
- Laboratory for Molecular Pharmacology, Department of Neuroscience and PharmacologyUniversity of Copenhagen Copenhagen Denmark
| | - Andreas Nygaard Madsen
- Laboratory for Molecular Pharmacology, Department of Neuroscience and PharmacologyUniversity of Copenhagen Copenhagen Denmark
| | - Maria Rasmussen
- Laboratory for Molecular Pharmacology, Department of Neuroscience and PharmacologyUniversity of Copenhagen Copenhagen Denmark
| | - Rune Kuhre
- Laboratory for Molecular Pharmacology, Department of Neuroscience and PharmacologyUniversity of Copenhagen Copenhagen Denmark
| | - Kristoffer L. Egerod
- Laboratory for Molecular Pharmacology, Department of Neuroscience and PharmacologyUniversity of Copenhagen Copenhagen Denmark
| | - Lars Bo Nielsen
- Department of Clinical BiochemistryUniversity of Copenhagen Copenhagen Denmark
| | - Thue W. Schwartz
- Laboratory for Molecular Pharmacology, Department of Neuroscience and PharmacologyUniversity of Copenhagen Copenhagen Denmark
- Novo Nordisk Foundation Center for Basic Metabolic ResearchFaculty of Health Sciences, University of Copenhagen Copenhagen Denmark
| | - Birgitte Holst
- Laboratory for Molecular Pharmacology, Department of Neuroscience and PharmacologyUniversity of Copenhagen Copenhagen Denmark
- Novo Nordisk Foundation Center for Basic Metabolic ResearchFaculty of Health Sciences, University of Copenhagen Copenhagen Denmark
| |
Collapse
|