1
|
Sutton Hickey AK, Becker J, Karolczak EO, Lutas A, Krashes MJ. Dietary fat content and absorption shape standard diet devaluation through hunger circuits. Mol Metab 2024; 89:102021. [PMID: 39216534 PMCID: PMC11415638 DOI: 10.1016/j.molmet.2024.102021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 08/27/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024] Open
Abstract
OBJECTIVE Exposure to 60% high fat diet (HFD) leads to a robust consummatory preference over well-balanced chow standard diet (SD) when mice are presented with a choice. This passive HFD-induced SD devaluation following HFD challenge and withdrawal is highlighted by the significant reduction in SD food intake even in states of caloric deprivation. The elements of HFD that lead to this SD depreciation remains unclear. Possibly important factors include the amount and type of fat contained in a diet as well as past eating experiences dependent on sensory properties including taste and post ingestive feedback. We aimed to explore the role of these components to HFD-induced SD devaluation. METHODS Wildtype mice were longitudinally presented discrete HFDs in conjunction with SD and feeding and metabolic parameters were analyzed. A separate cohort of animals were assessed for acute HFD preference in 3 conditions: 1) ad libitum fed (sated), 2) overnight fasted (physiologically hungry), and 3) ad libitum fed (artificially hungry), elicited through chemogenetic Agouti-related peptide (AgRP) neuron activation. Population dynamics of AgRP neurons were recorded to distinct inaccessible and accessible diets both before and after consummatory experience. Transient receptor potential channel type M5 (TRPM5) knockout mice were used to investigate the role of fat taste perception and preference to HFD-induced SD devaluation. The clinically approved lipase inhibitor orlistat was used to test the contribution of fat absorption to HFD-induced SD devaluation. RESULTS HFD-induced SD devaluation is dependent on fat content, composition, and preference. This effect scaled both in strength and latency with higher percentages of animal fat. 60% HFD was preferred and almost exclusively consumed in preference to other diets across hours and days, but this was not as evident upon initial introduction over seconds and minutes, suggesting ingestive experience is critical. Optical fiber photometry recordings of AgRP activity supported this notion as neuronal suppression by the different diets was contingent on prior intake. While taste transduced via TRPM5 influenced HFD-evoked weight gain, it failed to impact either HFD preference or HFD-induced SD devaluation. Perturbation of post ingestive feedback through orlistat-mediated diminishment of fat absorption prevented HFD-evoked weight gain and abolished HFD-induced SD devaluation. CONCLUSIONS Post ingestive feedback via fat digestion is vital for expression of HFD-induced SD devaluation.
Collapse
Affiliation(s)
- Ames K Sutton Hickey
- Department of Psychology and Neuroscience, Temple University, Philadelphia, PA, USA
| | - Jordan Becker
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA; NIH-Brown University Graduate Program in Neuroscience, Bethesda, MD, USA
| | - Eva O Karolczak
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Andrew Lutas
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Michael J Krashes
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
2
|
Howard EJ, Meyer RK, Weninger SN, Martinez T, Wachsmuth HR, Pignitter M, Auñon-Lopez A, Kangath A, Duszka K, Gu H, Schiro G, Laubtiz D, Duca FA. Impact of Plant-Based Dietary Fibers on Metabolic Homeostasis in High-Fat Diet Mice via Alterations in the Gut Microbiota and Metabolites. J Nutr 2024; 154:2014-2028. [PMID: 38735572 PMCID: PMC11282473 DOI: 10.1016/j.tjnut.2024.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/23/2024] [Accepted: 05/08/2024] [Indexed: 05/14/2024] Open
Abstract
BACKGROUND The gut microbiota contributes to metabolic disease, and diet shapes the gut microbiota, emphasizing the need to better understand how diet impacts metabolic disease via gut microbiota alterations. Fiber intake is linked with improvements in metabolic homeostasis in rodents and humans, which is associated with changes in the gut microbiota. However, dietary fiber is extremely heterogeneous, and it is imperative to comprehensively analyze the impact of various plant-based fibers on metabolic homeostasis in an identical setting and compare the impact of alterations in the gut microbiota and bacterially derived metabolites from different fiber sources. OBJECTIVES The objective of this study was to analyze the impact of different plant-based fibers (pectin, β-glucan, wheat dextrin, resistant starch, and cellulose as a control) on metabolic homeostasis through alterations in the gut microbiota and its metabolites in high-fat diet (HFD)-fed mice. METHODS HFD-fed mice were supplemented with 5 different fiber types (pectin, β-glucan, wheat dextrin, resistant starch, or cellulose as a control) at 10% (wt/wt) for 18 wk (n = 12/group), measuring body weight, adiposity, indirect calorimetry, glucose tolerance, and the gut microbiota and metabolites. RESULTS Only β-glucan supplementation during HFD-feeding decreased adiposity and body weight gain and improved glucose tolerance compared with HFD-cellulose, whereas all other fibers had no effect. This was associated with increased energy expenditure and locomotor activity in mice compared with HFD-cellulose. All fibers supplemented into an HFD uniquely shifted the intestinal microbiota and cecal short-chain fatty acids; however, only β-glucan supplementation increased cecal butyrate concentrations. Lastly, all fibers altered the small-intestinal microbiota and portal bile acid composition. CONCLUSIONS These findings demonstrate that β-glucan consumption is a promising dietary strategy for metabolic disease, possibly via increased energy expenditure through alterations in the gut microbiota and bacterial metabolites in mice.
Collapse
Affiliation(s)
- Elizabeth J Howard
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, United States
| | - Rachel K Meyer
- School of Nutritional Sciences and Wellness, University of Arizona, Tucson, AZ, United States
| | - Savanna N Weninger
- Department of Physiology, University of Arizona, Tucson, AZ, United States
| | - Taylor Martinez
- Department of Physiology, University of Arizona, Tucson, AZ, United States
| | - Hallie R Wachsmuth
- Department of Physiology, University of Arizona, Tucson, AZ, United States
| | - Marc Pignitter
- Institute of Physiological Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Arturo Auñon-Lopez
- Institute of Physiological Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria; Vienna Doctoral School in Chemistry (DoSChem), Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Archana Kangath
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, United States
| | - Kalina Duszka
- Department of Nutritional Sciences, University of Vienna, Vienna, Austria
| | - Haiwei Gu
- College of Health Solutions, Arizona State University, Phoenix, AZ, United States
| | - Gabriele Schiro
- PANDA Core for Genomics and Microbiome Research, Steele Children's Research Center, University of Arizona, Tucson, AZ, United States
| | - Daniel Laubtiz
- PANDA Core for Genomics and Microbiome Research, Steele Children's Research Center, University of Arizona, Tucson, AZ, United States
| | - Frank A Duca
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, United States; BIO5 Institute, University of Arizona, Tucson, AZ, United States.
| |
Collapse
|
3
|
Wang F, Ko CW, Qu J, Wu D, Zhu Q, Liu M, Tso P. Apolipoprotein A-IV-Deficient Mice in 129/SvJ Background Are Susceptible to Obesity and Glucose Intolerance. Nutrients 2023; 15:4840. [PMID: 38004234 PMCID: PMC10674380 DOI: 10.3390/nu15224840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 11/06/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
Apolipoprotein A-IV (apoA-IV), synthesized by enterocytes, is potentially involved in regulating lipid absorption and metabolism, food intake, and glucose metabolism. In this study, we backcrossed apoA-IV knockout (apoA-IV-/-) mice onto the 129/SvJ background for eight generations. Compared to the wild-type (WT) mice, the 129/SvJ apoA-IV-/- mice gained more weight and exhibited delayed glucose clearance even on the chow diet. During a 16-week high-fat diet (20% by weight of fat) study, apoA-IV-/- mice were more obese than the WT mice, which was associated with their increased food intake as well as reduced energy expenditure and physical activity. In addition, apoA-IV-/- mice developed significant insulin resistance (indicated by HOMA-IR) with severe glucose intolerance even though their insulin levels were drastically higher than the WT mice. In conclusion, we have established a model of apoA-IV-/- mice onto the 129/SvJ background. Unlike in the C57BL/6J strain, apoA-IV-/- 129/SvJ mice become significantly more obese and insulin-resistant than WT mice. Our current investigations of apoA-IV in the 129/SvJ strain and our previous studies in the C57BL/6J strain underline the impact of genetic background on apoA-IV metabolic effects.
Collapse
Affiliation(s)
- Fei Wang
- Norton Healthcare, 4910 Chamberlain Lane, Louisville, KY 40202, USA;
| | - Chih-Wei Ko
- Chroma Medicine, 201 Brookine Ave, Suite 1101, Boston, MA 02215, USA;
| | - Jie Qu
- Medpace Reference Laboratories, LLC., 5365 Medpace Way, Cincinnati, OH 45227, USA;
| | - Dong Wu
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan 250012, China;
| | - Qi Zhu
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Institute, University of Cincinnati, 2180 E Galbraith Road, Cincinnati, OH 45237, USA; (Q.Z.); (M.L.)
| | - Min Liu
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Institute, University of Cincinnati, 2180 E Galbraith Road, Cincinnati, OH 45237, USA; (Q.Z.); (M.L.)
| | - Patrick Tso
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Institute, University of Cincinnati, 2180 E Galbraith Road, Cincinnati, OH 45237, USA; (Q.Z.); (M.L.)
| |
Collapse
|
4
|
Bramati G, Stauffer P, Nigri M, Wolfer DP, Amrein I. Environmental enrichment improves hippocampus-dependent spatial learning in female C57BL/6 mice in novel IntelliCage sweet reward-based behavioral tests. Front Behav Neurosci 2023; 17:1256744. [PMID: 37791111 PMCID: PMC10543696 DOI: 10.3389/fnbeh.2023.1256744] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 08/11/2023] [Indexed: 10/05/2023] Open
Abstract
The IntelliCage is an automated home-cage system that allows researchers to investigate the spontaneous behavior and learning abilities of group-housed mice. The IntelliCage enables us to increase the standardization and reproducibility of behavioral outcomes by the omission of experimenter-mouse interactions. Although the IntelliCage provides a less stressful environment for animals, standard IntelliCage protocols use controlled water access as the motivational driver for learning. To overcome possible water restrictions in slow learners, we developed a series of novel protocols based on appetitive learning, in which mice had permanent access to plain water but were additionally rewarded with sweetened water upon solving the task. C57BL/6NCrl female mice were used to assess the efficacy of these sweet reward-based protocols in a series of learning tasks. Compared to control mice tested with standard protocols, mice motivated with a sweet reward did equal to or better in operant performance and place learning tasks. Learning of temporal rules was slower than that in controls. When faced with a combined temporal x spatial working memory task, sweet-rewarded mice learned little and chose plain water. In a second set of experiments, the impact of environmental enrichment on appetitive learning was tested. Mice kept under enriched environment (EE) or standard housing (SH) conditions prior to the IntelliCage experiments performed similarly in the sweet-rewarded place learning task. EE mice performed better in the hippocampus-dependent spatial working memory task. The improved performance of EE mice in the hippocampus-dependent spatial working memory task might be explained by the observed larger volume of their mossy fibers. Our results confirm that environmental enrichment increases complex spatial learning abilities and leads to long-lasting morphological changes in the hippocampus. Furthermore, simple standard IntelliCage protocols could easily be adapted to sweet rewards, which improve animal welfare by removing the possibility of water restriction. However, complex behavioral tasks motivated by sweet reward-based learning need further adjustments to reach the same efficacy as standard protocols.
Collapse
Affiliation(s)
- Giulia Bramati
- Division Functional Neuroanatomy, Institute of Anatomy, University Zurich, Zürich, Switzerland
| | - Pia Stauffer
- Division Functional Neuroanatomy, Institute of Anatomy, University Zurich, Zürich, Switzerland
| | - Martina Nigri
- Division Functional Neuroanatomy, Institute of Anatomy, University Zurich, Zürich, Switzerland
- Department of Health Sciences and Technology, ETH, Zürich, Switzerland
| | - David P. Wolfer
- Division Functional Neuroanatomy, Institute of Anatomy, University Zurich, Zürich, Switzerland
- Department of Health Sciences and Technology, ETH, Zürich, Switzerland
| | - Irmgard Amrein
- Division Functional Neuroanatomy, Institute of Anatomy, University Zurich, Zürich, Switzerland
- Department of Health Sciences and Technology, ETH, Zürich, Switzerland
| |
Collapse
|
5
|
McCloskey AG, Miskelly MG, Lafferty RA, Flatt PR, McKillop AM. Antidiabetic actions of GPR55 agonist Abn-CBD and sitagliptin in obese-diabetic high fat fed mice. Biochem Pharmacol 2023; 208:115398. [PMID: 36581052 DOI: 10.1016/j.bcp.2022.115398] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/19/2022] [Accepted: 12/20/2022] [Indexed: 12/27/2022]
Abstract
GPR55 has been recognized as a novel anti-diabetic target exerting positive effects on beta cell function and mass. This study evaluated the metabolic actions and therapeutic efficacy of GPR55 agonist abnormal cannabidiol (Abn-CBD) administered alone and in combination with sitagliptin in diet-induced obese-diabetic mice. Chronic effects of 21-day oral administration of Abn-CBD (0.1 µmol/kg BW) monotherapy and in combination with sitagliptin (50 mg/kg BW) were assessed in obese-diabetic HFF mice (n = 8). Assessments of plasma glucose, circulating insulin, DPP-IV activity, CRP, amylase, lipids, body weight and food intake were undertaken. Glucose tolerance, insulin sensitivity, DEXA scanning and islet morphology analysis were performed at 21-days. Sitagliptin, Abn-CBD alone and in combination with sitagliptin attenuated plasma glucose by 37-53 % (p < 0.01 - p < 0.001) and enhanced circulating insulin concentrations by 23-31 % (p < 0.001). Abn-CBD alone and with sitagliptin reduced bodyweight by 9-10 % (p < 0.05). After 21-days, Abn-CBD in combination with sitagliptin (44 %; p < 0.01) improved glucose tolerance, whilst enhancing insulin sensitivity by 79 % (p < 0.01). Abn-CBD increased islet area (86 %; p < 0.05), beta cell mass (p < 0.05) and beta cell proliferation (164 %; p < 0.001), whilst in combination with sitagliptin islet area was decreased (50 %; p < 0.01). Abn-CBD alone, in combination with sitagliptin or sitagliptin alone decreased triglycerides by 34-65 % (p < 0.001) and total cholesterol concentrations by 15-25 % (p < 0.001). In addition, Abn-CBD in combination with sitagliptin reduced fat mass by 19 % (p < 0.05) and reduced CRP concentrations (39 %; p < 0.05). These findings advocate Abn-CBD monotherapy and in combination with sitagliptin as a novel and effective approach for bodyweight control and the treatment of glucose intolerance and dyslipidaemia in type-2-diabetes.
Collapse
Affiliation(s)
- Andrew G McCloskey
- School of Biomedical Sciences, Ulster University, Cromore Road, Coleraine, BT52 1SA, Northern Ireland, U.K; Health and Biomedical Research Centre (HEAL), Atlantic Technological University, ATU Sligo, Ash Lane, Sligo, F91 YW50, Ireland
| | - Michael G Miskelly
- School of Biomedical Sciences, Ulster University, Cromore Road, Coleraine, BT52 1SA, Northern Ireland, U.K
| | - Ryan A Lafferty
- School of Biomedical Sciences, Ulster University, Cromore Road, Coleraine, BT52 1SA, Northern Ireland, U.K
| | - Peter R Flatt
- School of Biomedical Sciences, Ulster University, Cromore Road, Coleraine, BT52 1SA, Northern Ireland, U.K
| | - Aine M McKillop
- School of Biomedical Sciences, Ulster University, Cromore Road, Coleraine, BT52 1SA, Northern Ireland, U.K.
| |
Collapse
|
6
|
Adar T, Ya'acov AB, Shabat Y, Mizrahi M, Zolotarov L, Lichtenstein Y, Ilan Y. Steroid-mediated liver steatosis is CD1d-dependent, while steroid-induced liver necrosis, inflammation, and metabolic changes are CD1d-independent. BMC Gastroenterol 2022; 22:169. [PMID: 35392825 PMCID: PMC8991564 DOI: 10.1186/s12876-022-02242-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 03/24/2022] [Indexed: 11/23/2022] Open
Abstract
Introduction Glucocorticoids contribute to the pathogenesis of non-alcoholic fatty liver disease (NAFLD). Natural killer T cells play a role in the pathogenesis of NAFLD and response to steroids. The present study aimed to determine the role of CD1d in steroid-mediated metabolic derangement and the steroid-protective effect of glycosphingolipids. Methods Ten groups of mice were studied. Steroids were orally administered to C57BL/6 mice to assess the therapeutic effect of β-glucosylceramide (GC) on the development of steroid-mediated liver damage and metabolic derangements. The role of CD1d in the pathogenesis of steroid-induced liver damage and in mediating the hepatoprotective effect of GC was studied in CD1d−/− mice. Results A model of oral administration of steroids was established, resulting in insulin resistance, hyperinsulinemia, hypertriglyceridemia, liver steatosis, and hepatocellular injury. Steroid administration to CD1d−/− mice was associated with hyperglycemia and hypertriglyceridemia. However, CD1d−/− mice did not manifest marked steroid-induced steatosis. GC treatment alleviated steroid-associated metabolic derangements and liver injury independent of CD1d expression. Conclusion A steroid-mediated model of NAFLD and metabolic derangements was established in which steroid-mediated steatosis was CD1d-dependent while steroid-induced liver necrosis, inflammation, and metabolic changes were CD1d-independent, which may support a dichotomy between steatosis and steatohepatitis in NAFLD. Supplementary Information The online version contains supplementary material available at 10.1186/s12876-022-02242-9.
Collapse
Affiliation(s)
- Tomer Adar
- Faculty of Medicine, Department of Medicine, Hadassah Medical Center, Hebrew University, POB 1200, 91120, Jerusalem, Israel.,Digestive Disease Institute, Shaare-Zedek Medical Center, Jerusalem, Israel
| | - Ami Ben Ya'acov
- Faculty of Medicine, Department of Medicine, Hadassah Medical Center, Hebrew University, POB 1200, 91120, Jerusalem, Israel.,Digestive Disease Institute, Shaare-Zedek Medical Center, Jerusalem, Israel
| | - Yehudit Shabat
- Faculty of Medicine, Department of Medicine, Hadassah Medical Center, Hebrew University, POB 1200, 91120, Jerusalem, Israel
| | - Meir Mizrahi
- Faculty of Medicine, Department of Medicine, Hadassah Medical Center, Hebrew University, POB 1200, 91120, Jerusalem, Israel
| | - Lida Zolotarov
- Faculty of Medicine, Department of Medicine, Hadassah Medical Center, Hebrew University, POB 1200, 91120, Jerusalem, Israel
| | - Yoav Lichtenstein
- Faculty of Medicine, Department of Medicine, Hadassah Medical Center, Hebrew University, POB 1200, 91120, Jerusalem, Israel
| | - Yaron Ilan
- Faculty of Medicine, Department of Medicine, Hadassah Medical Center, Hebrew University, POB 1200, 91120, Jerusalem, Israel.
| |
Collapse
|
7
|
Cohen DJ, Giaccagli MM, Herzfeld JD, González LN, Cuasnicú PS, Da Ros VG. Metabolic syndrome and male fertility disorders: Is there a causal link? Rev Endocr Metab Disord 2021; 22:1057-1071. [PMID: 34037916 DOI: 10.1007/s11154-021-09659-9] [Citation(s) in RCA: 1] [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] [Accepted: 05/03/2021] [Indexed: 12/12/2022]
Abstract
Infertility is a global health problem affecting 10-15% of couples in reproductive age. Recent studies have provided growing evidence supporting that lifestyle factors can affect male fertility through alterations in endocrine profiles, spermatogenesis and/or sperm function. One of these critical factors could be the change in the food intake behavior in modern societies that produces metabolic alterations. Regarding this, metabolic syndrome (MetS) prevalence has increased in epidemic in the last 40-50 years. Although MetS is associated with advanced age, changes in lifestyles have accelerated the appearance of symptoms in the reproductive age. We review herein the current understanding of the relationship between MetS and the male reproductive status. For this purpose, in this narrative review a comprehensive literature search was made in both animal models and men, allowing us to evaluate such relationship. This analysis showed a high variability in the reproductive phenotypes observed in patients and mice suffering MetS, including sperm parameters, fertility and offspring health. In view of this, we proposed that the reproductive effects, which are diverse and not robust, observed among MetS-affected males, might depend on additional factors not associated with the metabolic condition and contributed not only by the affected male but also by his partner. With this perspective, this review provides a more accurate insight of this syndrome critical for the identification of specific diagnostic indicators and treatment of MetS-induced fertility disorders.
Collapse
Affiliation(s)
- Débora Juana Cohen
- Instituto de Biología y Medicina Experimental (IByME-CONICET), Ciudad Autónoma de Buenos Aires, Argentina.
| | - María Milagros Giaccagli
- Instituto de Biología y Medicina Experimental (IByME-CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - Jael Dafne Herzfeld
- Instituto de Biología y Medicina Experimental (IByME-CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - Lucas Nicolás González
- Instituto de Biología y Medicina Experimental (IByME-CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - Patricia Sara Cuasnicú
- Instituto de Biología y Medicina Experimental (IByME-CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - Vanina Gabriela Da Ros
- Instituto de Biología y Medicina Experimental (IByME-CONICET), Ciudad Autónoma de Buenos Aires, Argentina.
| |
Collapse
|
8
|
Gissler MC, Anto-Michel N, Pennig J, Scherrer P, Li X, Marchini T, Pfeiffer K, Härdtner C, Abogunloko T, Mwinyella T, Sol Mitre L, Spiga L, Koentges C, Smolka C, von Elverfeldt D, Hoppe N, Stachon P, Dufner B, Heidt T, Piepenburg S, Hilgendorf I, Bjune JI, Dankel SN, Mellgren G, Seifert G, Eisenhardt SU, Bugger H, von Zur Muhlen C, Bode C, Zirlik A, Wolf D, Willecke F. Genetic Deficiency of TRAF5 Promotes Adipose Tissue Inflammation and Aggravates Diet-Induced Obesity in Mice. Arterioscler Thromb Vasc Biol 2021; 41:2563-2574. [PMID: 34348490 DOI: 10.1161/atvbaha.121.316677] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective: The accumulation of inflammatory leukocytes is a prerequisite of adipose tissue inflammation during cardiometabolic disease. We previously reported that a genetic deficiency of the intracellular signaling adaptor TRAF5 (TNF [tumor necrosis factor] receptor-associated factor 5) accelerates atherosclerosis in mice by increasing inflammatory cell recruitment. Here, we tested the hypothesis that an impairment of TRAF5 signaling modulates adipose tissue inflammation and its metabolic complications in a model of diet-induced obesity in mice. Approach and Results: To induce diet-induced obesity and adipose tissue inflammation, wild-type or Traf5-/- mice consumed a high-fat diet for 18 weeks. Traf5-/- mice showed an increased weight gain, impaired insulin tolerance, and increased fasting blood glucose. Weight of livers and peripheral fat pads was increased in Traf5-/- mice, whereas lean tissue weight and growth were not affected. Flow cytometry of the stromal vascular fraction of visceral adipose tissue from Traf5-/- mice revealed an increase in cytotoxic T cells, CD11c+ macrophages, and increased gene expression of proinflammatory cytokines and chemokines. At the level of cell types, expression of TNF[alpha], MIP (macrophage inflammatory protein)-1[alpha], MCP (monocyte chemoattractant protein)-1, and RANTES (regulated on activation, normal T-cell expressed and secreted) was significantly upregulated in Traf5-deficient adipocytes but not in Traf5-deficient leukocytes from visceral adipose tissue. Finally, Traf5 expression was lower in adipocytes from obese patients and mice and recovered in adipose tissue of obese patients one year after bariatric surgery. Conclusions: We show that a genetic deficiency of TRAF5 in mice aggravates diet-induced obesity and its metabolic derangements by a proinflammatory response in adipocytes. Our data indicate that TRAF5 may promote anti-inflammatory and obesity-preventing signaling events in adipose tissue.
Collapse
Affiliation(s)
- Mark Colin Gissler
- Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Germany (M.C.G., J.P., P.S., X.L., T. Marchini, K.P., C.H., T.A., T. Mwinyella, L.S.M., L.S., C.K., C.S., N.H., P.S., B.D., T.H., S.P., I.H., C.v.z.M., C.B., D.W., F.W.)
| | - Nathaly Anto-Michel
- Department of Cardiology, Medical University of Graz, Austria (N.A.M., H.B., A.Z.)
| | - Jan Pennig
- Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Germany (M.C.G., J.P., P.S., X.L., T. Marchini, K.P., C.H., T.A., T. Mwinyella, L.S.M., L.S., C.K., C.S., N.H., P.S., B.D., T.H., S.P., I.H., C.v.z.M., C.B., D.W., F.W.)
| | - Philipp Scherrer
- Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Germany (M.C.G., J.P., P.S., X.L., T. Marchini, K.P., C.H., T.A., T. Mwinyella, L.S.M., L.S., C.K., C.S., N.H., P.S., B.D., T.H., S.P., I.H., C.v.z.M., C.B., D.W., F.W.)
| | - Xiaowei Li
- Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Germany (M.C.G., J.P., P.S., X.L., T. Marchini, K.P., C.H., T.A., T. Mwinyella, L.S.M., L.S., C.K., C.S., N.H., P.S., B.D., T.H., S.P., I.H., C.v.z.M., C.B., D.W., F.W.)
| | - Timoteo Marchini
- Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Germany (M.C.G., J.P., P.S., X.L., T. Marchini, K.P., C.H., T.A., T. Mwinyella, L.S.M., L.S., C.K., C.S., N.H., P.S., B.D., T.H., S.P., I.H., C.v.z.M., C.B., D.W., F.W.)
| | - Katharina Pfeiffer
- Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Germany (M.C.G., J.P., P.S., X.L., T. Marchini, K.P., C.H., T.A., T. Mwinyella, L.S.M., L.S., C.K., C.S., N.H., P.S., B.D., T.H., S.P., I.H., C.v.z.M., C.B., D.W., F.W.)
| | - Carmen Härdtner
- Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Germany (M.C.G., J.P., P.S., X.L., T. Marchini, K.P., C.H., T.A., T. Mwinyella, L.S.M., L.S., C.K., C.S., N.H., P.S., B.D., T.H., S.P., I.H., C.v.z.M., C.B., D.W., F.W.)
| | - Tijani Abogunloko
- Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Germany (M.C.G., J.P., P.S., X.L., T. Marchini, K.P., C.H., T.A., T. Mwinyella, L.S.M., L.S., C.K., C.S., N.H., P.S., B.D., T.H., S.P., I.H., C.v.z.M., C.B., D.W., F.W.)
| | - Timothy Mwinyella
- Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Germany (M.C.G., J.P., P.S., X.L., T. Marchini, K.P., C.H., T.A., T. Mwinyella, L.S.M., L.S., C.K., C.S., N.H., P.S., B.D., T.H., S.P., I.H., C.v.z.M., C.B., D.W., F.W.)
| | - Lucia Sol Mitre
- Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Germany (M.C.G., J.P., P.S., X.L., T. Marchini, K.P., C.H., T.A., T. Mwinyella, L.S.M., L.S., C.K., C.S., N.H., P.S., B.D., T.H., S.P., I.H., C.v.z.M., C.B., D.W., F.W.)
| | - Lisa Spiga
- Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Germany (M.C.G., J.P., P.S., X.L., T. Marchini, K.P., C.H., T.A., T. Mwinyella, L.S.M., L.S., C.K., C.S., N.H., P.S., B.D., T.H., S.P., I.H., C.v.z.M., C.B., D.W., F.W.)
| | - Christoph Koentges
- Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Germany (M.C.G., J.P., P.S., X.L., T. Marchini, K.P., C.H., T.A., T. Mwinyella, L.S.M., L.S., C.K., C.S., N.H., P.S., B.D., T.H., S.P., I.H., C.v.z.M., C.B., D.W., F.W.)
- Institute of Neuropathology (C.K.), Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Christian Smolka
- Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Germany (M.C.G., J.P., P.S., X.L., T. Marchini, K.P., C.H., T.A., T. Mwinyella, L.S.M., L.S., C.K., C.S., N.H., P.S., B.D., T.H., S.P., I.H., C.v.z.M., C.B., D.W., F.W.)
| | - Dominik von Elverfeldt
- Department of Radiology, Medical Physics (D.v.E.), Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Natalie Hoppe
- Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Germany (M.C.G., J.P., P.S., X.L., T. Marchini, K.P., C.H., T.A., T. Mwinyella, L.S.M., L.S., C.K., C.S., N.H., P.S., B.D., T.H., S.P., I.H., C.v.z.M., C.B., D.W., F.W.)
| | - Peter Stachon
- Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Germany (M.C.G., J.P., P.S., X.L., T. Marchini, K.P., C.H., T.A., T. Mwinyella, L.S.M., L.S., C.K., C.S., N.H., P.S., B.D., T.H., S.P., I.H., C.v.z.M., C.B., D.W., F.W.)
| | - Bianca Dufner
- Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Germany (M.C.G., J.P., P.S., X.L., T. Marchini, K.P., C.H., T.A., T. Mwinyella, L.S.M., L.S., C.K., C.S., N.H., P.S., B.D., T.H., S.P., I.H., C.v.z.M., C.B., D.W., F.W.)
| | - Timo Heidt
- Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Germany (M.C.G., J.P., P.S., X.L., T. Marchini, K.P., C.H., T.A., T. Mwinyella, L.S.M., L.S., C.K., C.S., N.H., P.S., B.D., T.H., S.P., I.H., C.v.z.M., C.B., D.W., F.W.)
| | - Sven Piepenburg
- Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Germany (M.C.G., J.P., P.S., X.L., T. Marchini, K.P., C.H., T.A., T. Mwinyella, L.S.M., L.S., C.K., C.S., N.H., P.S., B.D., T.H., S.P., I.H., C.v.z.M., C.B., D.W., F.W.)
| | - Ingo Hilgendorf
- Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Germany (M.C.G., J.P., P.S., X.L., T. Marchini, K.P., C.H., T.A., T. Mwinyella, L.S.M., L.S., C.K., C.S., N.H., P.S., B.D., T.H., S.P., I.H., C.v.z.M., C.B., D.W., F.W.)
| | - Jan-Inge Bjune
- Center for Diabetes Research (J.-I.B., S.N.D., G.M.), University of Bergen, Norway
- Mohn Nutrition Research Laboratory, Department of Clinical Science (J.-I.B., S.N.D., G.M.), University of Bergen, Norway
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway (J.-I.B., S.N.D., G.M.)
| | - Simon N Dankel
- Center for Diabetes Research (J.-I.B., S.N.D., G.M.), University of Bergen, Norway
- Mohn Nutrition Research Laboratory, Department of Clinical Science (J.-I.B., S.N.D., G.M.), University of Bergen, Norway
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway (J.-I.B., S.N.D., G.M.)
| | - Gunnar Mellgren
- Center for Diabetes Research (J.-I.B., S.N.D., G.M.), University of Bergen, Norway
- Mohn Nutrition Research Laboratory, Department of Clinical Science (J.-I.B., S.N.D., G.M.), University of Bergen, Norway
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway (J.-I.B., S.N.D., G.M.)
| | - Gabriel Seifert
- Department of General and Visceral Surgery (G.S.), Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Steffen U Eisenhardt
- Department of Plastic and Hand Surgery, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Breisgau, Germany (S.U.E.)
| | - Heiko Bugger
- Department of Cardiology, Medical University of Graz, Austria (N.A.M., H.B., A.Z.)
| | - Constantin von Zur Muhlen
- Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Germany (M.C.G., J.P., P.S., X.L., T. Marchini, K.P., C.H., T.A., T. Mwinyella, L.S.M., L.S., C.K., C.S., N.H., P.S., B.D., T.H., S.P., I.H., C.v.z.M., C.B., D.W., F.W.)
| | - Christoph Bode
- Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Germany (M.C.G., J.P., P.S., X.L., T. Marchini, K.P., C.H., T.A., T. Mwinyella, L.S.M., L.S., C.K., C.S., N.H., P.S., B.D., T.H., S.P., I.H., C.v.z.M., C.B., D.W., F.W.)
| | - Andreas Zirlik
- Department of Cardiology, Medical University of Graz, Austria (N.A.M., H.B., A.Z.)
| | - Dennis Wolf
- Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Germany (M.C.G., J.P., P.S., X.L., T. Marchini, K.P., C.H., T.A., T. Mwinyella, L.S.M., L.S., C.K., C.S., N.H., P.S., B.D., T.H., S.P., I.H., C.v.z.M., C.B., D.W., F.W.)
| | - Florian Willecke
- Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Germany (M.C.G., J.P., P.S., X.L., T. Marchini, K.P., C.H., T.A., T. Mwinyella, L.S.M., L.S., C.K., C.S., N.H., P.S., B.D., T.H., S.P., I.H., C.v.z.M., C.B., D.W., F.W.)
- Clinic for General and Interventional Cardiology/Angiology, Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany (F.W.)
| |
Collapse
|
9
|
Duan Y, Gao H, Su H, Liu A, Liu Y, Yuan H, Xie C. Exploring the Protective Effect of ShenQi Compound on Skeletal Muscle in Diabetic Macrovasculopathy Mice. Endocr Metab Immune Disord Drug Targets 2021; 20:943-951. [PMID: 32096754 DOI: 10.2174/1871530320666200225094756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 01/21/2020] [Accepted: 01/23/2020] [Indexed: 11/22/2022]
Abstract
OBJECTIVE ShenQi compound (SQC) is a traditional herbal formula, which has been used to treat Type 2 diabetes mellitus (T2DM) and complications for years. The aim of this study was to explore the preventive and protective effects of SQC recipe on the skeletal muscle of diabetic macrovasculopathy mice, which provides a theoretical basis for the clinical use of this formula. METHODS We evaluated the effect of SQC in a diabetic vasculopathy mouse model by detecting a series of blood indicators (blood glucose, lipids and insulin) and performing histological observations. Meanwhile, we explored the molecular mechanism of SQC treatment on skeletal muscle by genome expression profiles. RESULTS The results indicated that SQC could effectively improve blood glucose, serum lipids (total cholesterol (TC), Triglyceride (TG), high-density lipoprotein cholesterol (HDL-C) and low-density lipoprotein cholesterol (LDL-C)) and insulin (INS) levels in diabetic vasculopathy mice, as well as alleviating skeletal muscle tissue damage for diabetic macrovasculopathy. Meanwhile, compared with rosiglitazone, SQC showed a better effect on blood glucose fluctuation. Moreover, the gene microarray analysis indicated that SQC might improve T2DM by affecting biological functions related to cell death and cell adhesion. Moreover, 7 genes (Celsr2, Rilpl1, Dlx6as, 2010004M13Rik, Anapc13, Gm6097, Ddx39b) might be potential therapeutic targets of SQC. CONCLUSION All these results indicate that SQC is an effective preventive and protective drug for skeletal muscle in diabetic macrovasculopathy, and could alleviate skeletal muscle tissue damage through affecting biological functions related to cell death and cell adhesion.
Collapse
Affiliation(s)
- Yuhong Duan
- Department Two of Endocrinology, Affiliated Hospital of Shaanxi University of Chinese medicine, Xianyang, China
| | - Hong Gao
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Hongxia Su
- Department Two of Endocrinology, Affiliated Hospital of Shaanxi University of Chinese medicine, Xianyang, China
| | - Aixia Liu
- Department Two of Endocrinology, Affiliated Hospital of Shaanxi University of Chinese medicine, Xianyang, China
| | - Ya Liu
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Haipo Yuan
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Chunguang Xie
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| |
Collapse
|
10
|
Chen G, Fan XY, Zheng XP, Jin YL, Liu Y, Liu SC. Human umbilical cord-derived mesenchymal stem cells ameliorate insulin resistance via PTEN-mediated crosstalk between the PI3K/Akt and Erk/MAPKs signaling pathways in the skeletal muscles of db/db mice. Stem Cell Res Ther 2020; 11:401. [PMID: 32938466 PMCID: PMC7493876 DOI: 10.1186/s13287-020-01865-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 07/23/2020] [Accepted: 07/30/2020] [Indexed: 12/21/2022] Open
Abstract
Background Globally, 1 in 11 adults have diabetes mellitus, and 90% of the cases are type 2 diabetes mellitus. Insulin resistance is a central defect in type 2 diabetes mellitus, and although multiple drugs have been developed to ameliorate insulin resistance, the limitations and accompanying side effects cannot be ignored. Thus, more effective methods are required to improve insulin resistance. Methods In the current study, db/m and db/db mice were injected with human umbilical cord-derived mesenchymal stem cells (HUC-MSCs) via tail vein injection, intraperitoneal injection, and skeletal muscle injection. Body weight, fasting blood glucose, and the survival rates were monitored. Furthermore, the anti-insulin resistance effects and potential mechanisms of transplanted HUC-MSCs were investigated in db/db mice in vivo. Results The results showed that HUC-MSC transplantation by skeletal muscle injection was safer compared with tail vein injection and intraperitoneal injection, and the survival rate reached 100% in the skeletal muscle injection transplanted mice. HUC-MSCs can stabilize localization and differentiation in skeletal muscle tissue and significantly ameliorate insulin resistance. Potential regulatory mechanisms are associated with downregulation of inflammation, regulating the balance between PI3K/Akt and ERK/MAPK signaling pathway via PTEN, but was not associated with the IGF-1/IGF-1R signaling pathway. Conclusions These results suggest HUC-MSC transplantation may be a novel therapeutic direction to prevent insulin resistance and increase insulin sensitivity, and skeletal muscle injection was the safest and most effective way.
Collapse
Affiliation(s)
- Guang Chen
- Department of Basic Medical Sciences, Taizhou University Hospital, Taizhou University, No 1139 Shifu Road, Jiaojiang District, Taizhou, 318000, China.,Department of Basic Medical Sciences, Jiamusi University, No 148 Xuefu road, Xiangyang District, Jiamusi, 154007, China
| | - Xiao-Yan Fan
- Department of Basic Medical Sciences, Taizhou University Hospital, Taizhou University, No 1139 Shifu Road, Jiaojiang District, Taizhou, 318000, China
| | - Xiao-Peng Zheng
- Department of Basic Medical Sciences, Taizhou University Hospital, Taizhou University, No 1139 Shifu Road, Jiaojiang District, Taizhou, 318000, China
| | - Yue-Lei Jin
- Department of Basic Medical Sciences, Taizhou University Hospital, Taizhou University, No 1139 Shifu Road, Jiaojiang District, Taizhou, 318000, China
| | - Ying Liu
- Jilin Tuhua Bioengineering Company Limited, Shiling Town, Tiedong District, Siping, Jilin, 136000, China
| | - Shuang-Chun Liu
- Municipal Hospital Affiliated to Medical School of Taizhou University, No 381, Zhongshan east road, Jiaojiang district, Taizhou, 318000, China.
| |
Collapse
|
11
|
Zhu Y, Zhang JY, Wei YL, Hao JY, Lei YQ, Zhao WB, Xiao YH, Sun AD. The polyphenol-rich extract from chokeberry ( Aronia melanocarpa L .) modulates gut microbiota and improves lipid metabolism in diet-induced obese rats. Nutr Metab (Lond) 2020; 17:54. [PMID: 32655675 PMCID: PMC7339576 DOI: 10.1186/s12986-020-00473-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 06/24/2020] [Indexed: 12/15/2022] Open
Abstract
The gut microbiota plays a critical role in obesity and lipid metabolism disorder. Chokeberry (Aronia melanocarpa L.) are rich in polyphenols with various physiological and pharmacological activities. We determined serum physiological parameters and fecal microbial components by using related kits, liquid chromatography-mass spectrometry (LC-MS) and 16S rRNA gene sequencing every 10 days. Real-time PCR analysis was used to measure gene expression of bile acids (BAs) and lipid metabolism in liver and adipose tissues. We analyzed the effects of different Chokeberry polyphenol (CBPs) treatment time on obesity and lipid metabolism in high fat diet (HFD)-fed rats. The results indicated that CBPs treatment prevents obesity, liver steatosis and improves dyslipidemia in HFD-fed rats. CBPs modulated the composition of the gut microbiota with the extended treatment time, reducing the Firmicutes/Bacteroidetes ratio (F/B ratio) and increasing the relative abundance of Bacteroides, Prevotella, Akkermansia and other bacterial species associated with anti-obesity properties. We found that CBPs treatment gradually decreased the total BAs pool and particularly reduced the relative content of cholic acid (CA), deoxycholic acid (DCA) and enhanced the relative content of chenodeoxycholic acid (CDCA). These changes were positively correlated Bacteroides, Prevotella and negatively correlated with Clostridium, Eubacterium, Ruminococcaceae. In liver and white adipose tissues, the gene expression of lipogenesis, lipolysis and BAs metabolism were regulated after CBPs treatment in HFD-fed rats, which was most likely mediated through FXR and TGR-5 signaling pathway to improve lipid metabolism. In addition, the mRNA expression of PPARγ, UCP1 and PGC-1α were upregulated markedly in interscapular brown adipose tissue (iBAT) after CBPs treatment. We confirmed that CBPs could reduce the body weight of HFD-fed rats by accelerating energy homeostasis and thermogenesis in iBAT. Finally, the fecal microbiota transplantation (FMT) experiment results demonstrated that FMT from CBPs-treated rats failed to reduce the weight of HFD-fed rats. However, FMT from CBPs-treated rats improved dyslipidemia and reshaped gut microbiota in HFD-fed rats. In conclusion, CBPs treatment improved obesity and complications by regulating gut microbiota in HFD-fed rats. The gut microbiota plays an important role in BAs metabolism after CBPs treatment, and BAs have therefore emerged as major effectors in microbe-host signaling events that influence host lipid metabolism, energy metabolism and thermogenesis.
Collapse
Affiliation(s)
- Yue Zhu
- College of Biological Sciences and Technology, Beijing Forestry University, Beijing, 100083 China
| | - Jia-ying Zhang
- College of Biological Sciences and Technology, Beijing Forestry University, Beijing, 100083 China
| | - Yu-long Wei
- College of Biological Sciences and Technology, Beijing Forestry University, Beijing, 100083 China
| | - Jing-yi Hao
- College of Biological Sciences and Technology, Beijing Forestry University, Beijing, 100083 China
| | - Yu-qing Lei
- College of Biological Sciences and Technology, Beijing Forestry University, Beijing, 100083 China
| | - Wan-bin Zhao
- College of Biological Sciences and Technology, Beijing Forestry University, Beijing, 100083 China
| | - Yu-hang Xiao
- College of Biological Sciences and Technology, Beijing Forestry University, Beijing, 100083 China
| | - Ai-dong Sun
- College of Biological Sciences and Technology, Beijing Forestry University, Beijing, 100083 China
| |
Collapse
|
12
|
Abstract
In order to better understand the events that precede and precipitate the onset of type 2 diabetes (T2DM), several nutritional animal models have been developed. These models are generated by manipulating the diet of either the animal itself, or its mother during her pregnancy, and in comparison to traditional genetic and knock out models, have the advantage that they more accurately reflect the etiology of human T2DM. This chapter will discuss some of the most widely used nutritional models of T2DM: Diet-induced obesity (DIO) in adult rodents, and studies of offspring of mothers fed a low-protein, high-fat and/or high-sugar diet during pregnancy and/or lactation. Several common mechanisms have been identified through which these nutritional manipulations can lead to metabolic disease, including pancreatic beta-cell dysfunction, impaired insulin signaling in skeletal muscle, and the excess accumulation of visceral adipose tissue and consequent deposition of nonesterified fatty acids in peripheral tissues. In addition, there is an emerging concept that obesity/poor quality diets result in increased production and release of pro-inflammatory cytokines from adipose tissue leading to a state of chronic low-grade inflammation, and that this is likely to represent an important link between obesity/diet and metabolic dysfunction. The following chapter will discuss the most common nutritional models of T2DM in experimental animals, their application, and relationship to human etiology, and will highlight the important insights these models have provided into the pathogenesis of T2DM.
Collapse
Affiliation(s)
- Beverly Sara Mühlhäusler
- Food and Nutrition Research Group, Department of Food and Wine Sciences, School of Agriculture, Food and Wine, The University of Adelaide, Adelaide, SA, Australia.
- FOODplus Research Centre, School of Agriculture, Food and Wine, The University of Adelaide, Adelaide, SA, Australia.
- CSIRO, Health and Biosecurity, Adelaide, SA, Australia.
| | - Carla Toop
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| | - Sheridan Gentili
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| |
Collapse
|
13
|
Pharmacological potential of novel agonists for FFAR4 on islet and enteroendocrine cell function and glucose homeostasis. Eur J Pharm Sci 2019; 142:105104. [PMID: 31669388 DOI: 10.1016/j.ejps.2019.105104] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 09/27/2019] [Accepted: 10/10/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND To investigate the metabolic effects of FFAR4-selective agonists on islet and enteroendocrine cell hormone release and the combined therapeutic effectiveness with DPP-IV inhibitors. METHODS Insulinotropic activity and specificity of FFAR4 agonists were determined in clonal pancreatic BRIN-BD11 cells. Expression of FFAR4 was assessed by qPCR and western blotting following agonist treatment in BRIN-BD11 cells and by immunohistochemistry in mouse islets. Acute in-vivo effects of agonists was investigated after intraperitoneal (i.p.) or oral administration in lean and HFF-obese diabetic mice. RESULTS GSK137647 (10-11-10-4 M) and Compound-A (10-10-10-4 M) stimulated insulin secretion at 5.6 mM (p < 0.05-p < 0.001) and 16.7 mM (p < 0.05-p < 0.001) glucose in BRIN-BD11 cells, with no cytotoxicity effects as assessed by MTT. FFAR4 antagonist (AH-7614) abolished the insulintropic effect of GSK137647 (p < 0.05-p < 0.001), whilst FFAR1 antagonist (GW1100) had no effect. Incubation of BRIN-BD11 cells with GSK137647 and Compound-A increased FFAR4 (p < 0.01) gene expression at 16.7 mM glucose, with a corresponding increase in FFAR4 (p < 0.01) protein concentrations. FFAR4 upregulation was attenuated under normoglycaemic conditions. Immunohistochemistry demonstrated co-localisation of FFAR4 and insulin in mouse islets. Orally administered GSK137647 or Compound-A (0.1 µmol/kgBW) improved glucose tolerance (p < 0.001), increased plasma insulin (p < 0.001), GLP-1 (p < 0.05), GIP (p < 0.05) and induced satiety (p < 0.001) in HFF mice, with glucose-lowering effects enhanced in combination with DPP-IV inhibitor (Sitagliptin) (p < 0.05). CONCLUSIONS Specific FFAR4 agonism improves glucose tolerance through insulin and incretin secretion, with enhanced DPP-IV inhibition in combination with Sitagliptin. GENERAL SIGNIFICANCE These findings have for the first time demonstrated that selective FFAR4 activation regulates both islet and enteroendocrine hormone function with agonist combinational therapy, presenting a promising strategy for the treatment of type-2-diabetes.
Collapse
|
14
|
Schlecht SH, Martin CT, Ochocki DN, Nolan BT, Wojtys EM, Ashton-Miller JA. Morphology of Mouse Anterior Cruciate Ligament-Complex Changes Following Exercise During Pubertal Growth. J Orthop Res 2019; 37:1910-1919. [PMID: 31042312 PMCID: PMC6700741 DOI: 10.1002/jor.24328] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 04/08/2019] [Indexed: 02/04/2023]
Abstract
Postnatal development and the physiological loading response of the anterior cruciate ligament (ACL) complex (ACL proper, entheses, and bony morphology) is not well understood. We tested whether the ACL-complex of two inbred mouse strains that collectively encompass the musculoskeletal variation observed in humans would demonstrate significant morphological differences following voluntary cage-wheel running during puberty compared with normal cage activity controls. Female A/J and C57BL/6J (B6) 6-week-old mice were provided unrestricted access to a standard cage-wheel for 4 weeks. A/J-exercise mice showed a 6.3% narrower ACL (p = 0.64), and a 20.1% more stenotic femoral notch (p < 0.01) while B6-exercise mice showed a 12.3% wider ACL (p = 0.10), compared with their respective controls. Additionally, A/J-exercise mice showed a 5.3% less steep posterior medial tibial slope (p = 0.07) and an 8.8% less steep posterior lateral tibial slope (p = 0.07), while B6-exercise mice showed a 9.8% more steep posterior medial tibial slope (p < 0.01) than their respective controls. A/J-exercise mice also showed more reinforcement of the ACL tibial enthesis with a 20.4% larger area (p < 0.01) of calcified fibrocartilage distributed at a 29.2% greater depth (p = 0.02) within the tibial enthesis, compared with their controls. These outcomes suggest exercise during puberty significantly influences ACL-complex morphology and that inherent morphological differences between these mice, as observed in their less active genetically similar control groups, resulted in a divergent phenotypic outcome between mouse strains. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:1910-1919, 2019.
Collapse
Affiliation(s)
- Stephen H. Schlecht
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, Michigan
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Colin T. Martin
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan
| | | | - Bonnie T. Nolan
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, Michigan
| | - Edward M. Wojtys
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, Michigan
| | | |
Collapse
|
15
|
Basal Diet Determined Long-Term Composition of the Gut Microbiome and Mouse Phenotype to a Greater Extent than Fecal Microbiome Transfer from Lean or Obese Human Donors. Nutrients 2019; 11:nu11071630. [PMID: 31319545 PMCID: PMC6682898 DOI: 10.3390/nu11071630] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 07/11/2019] [Accepted: 07/15/2019] [Indexed: 12/23/2022] Open
Abstract
The Western dietary pattern can alter the gut microbiome and cause obesity and metabolic disorders. To examine the interactions between diet, the microbiome, and obesity, we transplanted gut microbiota from lean or obese human donors into mice fed one of three diets for 22 weeks: (1) a control AIN93G diet; (2) the total Western diet (TWD), which mimics the American diet; or (3) a 45% high-fat diet-induced obesity (DIO) diet. We hypothesized that a fecal microbiome transfer (FMT) from obese donors would lead to an obese phenotype and aberrant glucose metabolism in recipient mice that would be exacerbated by consumption of the TWD or DIO diets. Prior to the FMT, the native microbiome was depleted using an established broad-spectrum antibiotic protocol. Interestingly, the human donor body type microbiome did not significantly affect final body weight or body composition in mice fed any of the experimental diets. Beta diversity analysis and linear discriminant analysis with effect size (LEfSe) showed that mice that received an FMT from obese donors had a significantly different microbiome compared to mice that received an FMT from lean donors. However, after 22 weeks, diet influenced the microbiome composition irrespective of donor body type, suggesting that diet is a key variable in the shaping of the gut microbiome after FMT.
Collapse
|
16
|
Rivera P, Ramírez‐López MT, Vargas A, Decara J, Vázquez M, Arco R, Gómez de Heras R, Argente J, Rodríguez de Fonseca F, Chowen JA, Suárez J. Perinatal free-choice of a high-calorie low-protein diet affects leptin signaling through IRS1 and AMPK dephosphorylation in the hypothalami of female rat offspring in adulthood. Acta Physiol (Oxf) 2019; 226:e13244. [PMID: 30589509 DOI: 10.1111/apha.13244] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 12/07/2018] [Accepted: 12/17/2018] [Indexed: 12/17/2022]
Abstract
AIM We aimed to investigate whether a dysregulated maternal diet during gestation and lactation induces long-lasting changes in the hypothalamic control of feeding behavior in the offspring and whether this effect is sex specific. METHODS The study included an analysis of appetite-regulating metabolic hormones and hypothalamic signaling in male and female offspring in adulthood after exposure to a free-choice high-calorie palatable low-protein (P) diet or standard chow (C) during (pre)gestation/lactation (maternal) and/or postweaning (offspring). RESULTS Maternal exposure to the P diet resulted in decreased protein intake and body weight gain in dams and decreased body weight gain in offspring during lactation. The maternal P diet (PC) specifically increased feed efficacy and decreased body weight and cholesterol levels in the female offspring in adulthood, but no changes in adiposity or leptin levels were found. In contrast, P diet exposure after weaning (CP and PP) increased caloric intake, adiposity and circulating levels of leptin in the male and female offspring in adulthood. The hypothalami of the female offspring exposed to the maternal P diet (PC and PP) expressed high levels of the phospho-leptin receptor and low levels of SOCS3, phospho-IRS1 and phospho-AMPK, regardless of the postweaning diet. The hypothalami of the female rats in the PC group also showed increased levels of STAT3 and the orexigenic neuropeptide Agrp. CONCLUSIONS Maternal exposure to a free-choice high-calorie low-protein diet induces a long-term feed efficacy associated with changes in leptin signaling through IRS-1 and AMPK dephosphorylation in the hypothalami of female offspring in adulthood.
Collapse
Affiliation(s)
- Patricia Rivera
- Department of Endocrinology, Fundación Investigación Biomédica del Hospital Infantil Universitario Niño Jesús Instituto de Investigación Biomédica la Princesa Madrid Spain
| | - María T. Ramírez‐López
- Departamento de Psicobiología, Facultad de Psicología Universidad Complutense de Madrid Pozuelo de Alarcón Spain
- Hospital Universitario de Getafe Getafe (Madrid) Spain
| | - Antonio Vargas
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga Universidad de Málaga Málaga Spain
| | - Juan Decara
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga Universidad de Málaga Málaga Spain
| | - Mariam Vázquez
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga Universidad de Málaga Málaga Spain
| | - Rocío Arco
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga Universidad de Málaga Málaga Spain
| | - Raquel Gómez de Heras
- Departamento de Psicobiología, Facultad de Psicología Universidad Complutense de Madrid Pozuelo de Alarcón Spain
| | - Jesús Argente
- Department of Endocrinology, Fundación Investigación Biomédica del Hospital Infantil Universitario Niño Jesús Instituto de Investigación Biomédica la Princesa Madrid Spain
- Centro de Investigación Biomédica en Red Fisiología de la Obesidad y Nutrición (CIBEROBN) Madrid Spain
- IMDEA Food Institute Campus of International Excellence (CEI) UAM + CSIC Madrid Spain
- Department of PediatricsUniversity Autonoma de Madrid Madrid Spain
| | - Fernando Rodríguez de Fonseca
- Departamento de Psicobiología, Facultad de Psicología Universidad Complutense de Madrid Pozuelo de Alarcón Spain
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga Universidad de Málaga Málaga Spain
| | - Julie A. Chowen
- Department of Endocrinology, Fundación Investigación Biomédica del Hospital Infantil Universitario Niño Jesús Instituto de Investigación Biomédica la Princesa Madrid Spain
- Centro de Investigación Biomédica en Red Fisiología de la Obesidad y Nutrición (CIBEROBN) Madrid Spain
- IMDEA Food Institute Campus of International Excellence (CEI) UAM + CSIC Madrid Spain
| | - Juan Suárez
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga Universidad de Málaga Málaga Spain
| |
Collapse
|
17
|
Emond C, DeVito MJ, Diliberto JJ, Birnbaum LS. The Influence of Obesity on the Pharmacokinetics of Dioxin in Mice: An Assessment Using Classical and PBPK Modeling. Toxicol Sci 2019; 164:218-228. [PMID: 29596651 DOI: 10.1093/toxsci/kfy078] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The effects of body fat mass on the elimination of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) was examined in mice. When male C57BL/6J mice are fed a high-fat, simple carbohydrate diet (HFD) for 13 weeks, they develop an obese phenotype. In contrast, A/J mice fed an HFD do not become obese. After 13 weeks on a normal diet (ND) or HFD, male C57BL/6J and A/J mice received a single dose by gavage of 0.1 or 5.0 µg of 2,3,7,8-tetrachloro[1,6-3H] dibenzo-p-dioxin per kg body weight. Using classical pharmacokinetics, the blood elimination half-life of TCDD was approximately 10 and 2 times longer in the C57BL/6J on the HFD compared with the mice on the ND at 0.1 and 5.0 μg/kg doses, respectively. The diet did not increase the blood half-life of TCDD in the A/J mice, which did not get obese. Using a physiologically based pharmacokinetic model for TCDD that incorporated experimentally derived percent body fat mass and tissue partition coefficients, as well as data on hepatic sequestration, did not provide accurate predictions to the data and could not explain the increase in half-life of TCDD in the HFD groups. This work demonstrates that obesity influences the half-life of TCDD, but other undetermined factors are involved in its elimination because the increase in body fat mass, decreases in cytochrome P4501A2, and altered partition coefficients could not completely explain the prolonged half-life.
Collapse
Affiliation(s)
- Claude Emond
- BioSimulation Consulting Inc., Newark, DE, USA, 19713.,Department of Environmental and Occupational Health, University of Montreal, Quebec, Canada H3N 1X9
| | - Michael J DeVito
- National Institute of Environmental Health Sciences, National Toxicology Program, Research Triangle Park, NC, USA
| | - Janet J Diliberto
- National Health and Environmental Effects Research Laboratory, U.S. Environmental protection Agency, Research Triangle Park, NC, USA, 27711
| | - Linda S Birnbaum
- National Cancer Institute, Research Triangle Park, NC, USA, 27709
| |
Collapse
|
18
|
Arboatti AS, Lambertucci F, Sedlmeier MG, Pisani G, Monti J, Álvarez MDL, Francés DEA, Ronco MT, Carnovale CE. Diethylnitrosamine enhances hepatic tumorigenic pathways in mice fed with high fat diet (Hfd). Chem Biol Interact 2019; 303:70-78. [PMID: 30826251 DOI: 10.1016/j.cbi.2019.02.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 01/28/2019] [Accepted: 02/24/2019] [Indexed: 12/21/2022]
Abstract
Obesity has been implicated in the genesis of metabolic syndromes including insulin resistance and Type 2 Diabetes Mellitus (T2DM). Given the association between T2DM and the risk of hepatocellular carcinoma (HCC), our specific goal was to determine whether the liver of HFD-induced T2DM mice is more sensitive to the carcinogen diethylnitrosamine (DEN), due to a modification of the molecular pathways implicated in the early stages of HCC pathogenesis. C57BL/6 male mice (five-week-old) were divided into 4 groups: C, C + DEN, HFD and HFD + DEN. Mice were euthanized twenty-five weeks after DEN-injection. Livers of HDF-fed mice showed a higher proliferative index than Control groups. In line with this, HFD groups showed an increase of nuclear β-catenin, and interestingly, DEN treatment led to a slight increase in the expression of this protein in HFD group. Based on these results, and to confirm this effect, we analyzed β-catenin target genes, finding that DEN treatment in HFD group led to a significant increase of Vegf, c-myc, c-jun and cyclin D1 expression levels. According to our results, the expression of TCF4 showed to be significantly increased in HFD + DEN vs. HFD. In this regard, the β-catenin/TCF4 complex enhanced its association with pSmads 2/3, as we observed an increase of nuclear Smads expression in HFD + DEN, suggesting a possible role of TGF-β1/Smads signaling pathway in this phenomenon. Our results show that the liver of HFD fed model that resembles early T2DM pathology in mice, is more sensitive to DEN, by inducing both Wnt/β-catenin and TGF β1/Smads tumorigenic pathways.
Collapse
Affiliation(s)
- A S Arboatti
- Instituto de Fisiología Experimental (IFISE-CONICET), Cátedra de Fisiología, Facultad de Ciencias Bioquímicas y Farmacéuticas- UNR, Suipacha 570, 2000, Rosario, Argentina
| | - F Lambertucci
- Instituto de Fisiología Experimental (IFISE-CONICET), Cátedra de Fisiología, Facultad de Ciencias Bioquímicas y Farmacéuticas- UNR, Suipacha 570, 2000, Rosario, Argentina
| | - M G Sedlmeier
- Instituto de Fisiología Experimental (IFISE-CONICET), Cátedra de Fisiología, Facultad de Ciencias Bioquímicas y Farmacéuticas- UNR, Suipacha 570, 2000, Rosario, Argentina
| | - G Pisani
- Cátedra de Morfología, Facultad de Ciencias Bioquímicas y Farmacéuticas, UNR, Suipacha 570, 2000, Rosario, Argentina
| | - J Monti
- Instituto de Fisiología Experimental (IFISE-CONICET), Cátedra de Fisiología, Facultad de Ciencias Bioquímicas y Farmacéuticas- UNR, Suipacha 570, 2000, Rosario, Argentina
| | - M de L Álvarez
- Instituto de Fisiología Experimental (IFISE-CONICET), Cátedra de Fisiología, Facultad de Ciencias Bioquímicas y Farmacéuticas- UNR, Suipacha 570, 2000, Rosario, Argentina; Cátedra de Morfología, Facultad de Ciencias Bioquímicas y Farmacéuticas, UNR, Suipacha 570, 2000, Rosario, Argentina
| | - D E A Francés
- Instituto de Fisiología Experimental (IFISE-CONICET), Cátedra de Fisiología, Facultad de Ciencias Bioquímicas y Farmacéuticas- UNR, Suipacha 570, 2000, Rosario, Argentina
| | - M T Ronco
- Instituto de Fisiología Experimental (IFISE-CONICET), Cátedra de Fisiología, Facultad de Ciencias Bioquímicas y Farmacéuticas- UNR, Suipacha 570, 2000, Rosario, Argentina
| | - C E Carnovale
- Instituto de Fisiología Experimental (IFISE-CONICET), Cátedra de Fisiología, Facultad de Ciencias Bioquímicas y Farmacéuticas- UNR, Suipacha 570, 2000, Rosario, Argentina.
| |
Collapse
|
19
|
O’Harte FPM, Parthsarathy V, Hogg C, Flatt PR. Long-term treatment with acylated analogues of apelin-13 amide ameliorates diabetes and improves lipid profile of high-fat fed mice. PLoS One 2018; 13:e0202350. [PMID: 30157220 PMCID: PMC6114795 DOI: 10.1371/journal.pone.0202350] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 08/01/2018] [Indexed: 01/22/2023] Open
Abstract
Previous studies have shown that modified apelin analogues exhibited enzyme resistance in plasma and improved circulating half-life compared to apelin-13. This study investigated the antidiabetic effects of chronic administration of stable long acting fatty acid modified apelin analogues, namely, (Lys8GluPAL)apelin-13 amide and pGlu(Lys8GluPAL)apelin-13 amide, in high-fat fed obese-diabetic mice. Male NIH Swiss mice (groups n = 8) were maintained either on a high-fat diet (45% fat) from 8 to 28 weeks old, or control mice were fed a normal diet (10% fat). When diet induced obesity-diabetes was established after high-fat feeding, mice were injected i.p. once daily with apelin analogues, liraglutide (25 nmol/kg) or saline (controls). Administration of (Lys8GluPAL)apelin-13 amide and pGlu(Lys8GluPAL)apelin-13 amide for 28 days significantly reduced food intake and decreased body weight. Non-fasting glucose was reduced (p<0.01 to p<0.001) and plasma insulin concentrations increased (p<0.01 to p<0.001). This was accompanied by enhanced insulin responses (p<0.01 to p<0.001) and significant reductions in glucose excursion after oral (p<0.01) or i.p. (p<0.01) glucose challenges and feeding. Apelin analogues also significantly improved HbA1c (p<0.01), enhanced insulin sensitivity (p<0.01), reduced triglycerides (p<0.001), increased HDL-cholesterol (p<0.01) and decreased LDL-cholesterol (p<0.01), compared to high-fat fed saline treated control mice. Cholesterol levels were decreased (p<0.01) by pGlu(Lys8GluPAL)apelin-13 amide and both apelin treated groups showed improved bone mineral content, reduced fat deposits and increased plasma GLP-1. Daily treatment with liraglutide mirrored many of these changes (not on bone or adipose tissue), but unlike apelin analogues increased plasma amylase. Consumption of O2, production of CO2, respiratory exchange ratio and energy expenditure were improved by apelin analogues. These results indicate that long-term treatment with acylated analogues (Lys8GluPAL)apelin-13 amide and particularly pGlu(Lys8GluPAL)apelin-13 amide resulted in similar or enhanced therapeutic responses to liraglutide in high-fat fed mice. Fatty acid derived apelin analogues represent a new and exciting development in the treatment of obesity-diabetes.
Collapse
Affiliation(s)
- Finbarr P. M. O’Harte
- School of Biomedical Sciences, SAAD Centre for Pharmacy and Diabetes, Ulster University, Coleraine, Co. Londonderry, Northern Ireland, United Kingdom
| | - Vadivel Parthsarathy
- School of Biomedical Sciences, SAAD Centre for Pharmacy and Diabetes, Ulster University, Coleraine, Co. Londonderry, Northern Ireland, United Kingdom
| | - Christopher Hogg
- School of Biomedical Sciences, SAAD Centre for Pharmacy and Diabetes, Ulster University, Coleraine, Co. Londonderry, Northern Ireland, United Kingdom
| | - Peter R. Flatt
- School of Biomedical Sciences, SAAD Centre for Pharmacy and Diabetes, Ulster University, Coleraine, Co. Londonderry, Northern Ireland, United Kingdom
| |
Collapse
|
20
|
Parra-Vargas M, Sandoval-Rodriguez A, Rodriguez-Echevarria R, Dominguez-Rosales JA, Santos-Garcia A, Armendariz-Borunda J. Delphinidin Ameliorates Hepatic Triglyceride Accumulation in Human HepG2 Cells, but Not in Diet-Induced Obese Mice. Nutrients 2018; 10:E1060. [PMID: 30103390 PMCID: PMC6115893 DOI: 10.3390/nu10081060] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 08/06/2018] [Accepted: 08/08/2018] [Indexed: 02/06/2023] Open
Abstract
Anthocyanin consumption is linked to benefits in obesity-related metabolic alterations and non-alcoholic fatty liver disease (NAFLD), though the functional role of delphinidin (Dp) is yet to be established. Therefore, this study examined the effects of Dp on metabolic alterations associated with NAFLD, and molecular mechanisms in HepG2 cells and diet-induced obese mice. Cells incubated with palmitate to induce lipid accumulation, concomitantly treated with Dp, reduced triglyceride accumulation by ~53%, and downregulated gene expression of CPT1A, SREBF1, and FASN without modifying AMP-activated protein kinase (AMPK) levels. C57BL/6Nhsd mice were fed a standard diet (control) or a high-fat/high-carbohydrate diet (HFHC) for 16 weeks. Mice in the HFHC group were subdivided and treated with Dp (HFHC-Dp, 15 mg/kg body weight/day) or a vehicle for four weeks. Dp did not affect body weight, energy intake, hyperglycemia, insulin resistance, or histological abnormalities elicited by the HFHC diet. Furthermore, the messenger RNA (mRNA) expressions of Acaca, and Fasn in hepatic or epididymal adipose tissue, and the hepatic sirtuin 1 (SIRT1)/liver kinase B1 (LKB1)/AMPK and proliferator-activated receptor alpha (PPARα) signaling axis did not significantly change due to the HFHC diet or Dp. In summary, Dp effectively reduced triglyceride accumulation in vitro through the modulation of lipid metabolic gene expression. However, a dose of Dp administrated in mice simulating the total daily anthocyanin intake in humans had no effect on either metabolic alterations or histological abnormalities associated with HFHC diets.
Collapse
Affiliation(s)
- Marcela Parra-Vargas
- Institute for Molecular Biology in Medicine and Gene Therapy, Department of Molecular Biology and Genomics, CUCS, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico.
| | - Ana Sandoval-Rodriguez
- Institute for Molecular Biology in Medicine and Gene Therapy, Department of Molecular Biology and Genomics, CUCS, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico.
| | - Roberto Rodriguez-Echevarria
- Institute for Molecular Biology in Medicine and Gene Therapy, Department of Molecular Biology and Genomics, CUCS, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico.
| | - Jose Alfredo Dominguez-Rosales
- Chronic-Degenerative Diseases Institute, Department of Molecular Biology and Genomics, CUCS, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico.
| | - Arturo Santos-Garcia
- Tecnologico de Monterrey, Campus Guadalajara, Guadalajara 45138, Jalisco, Mexico.
| | - Juan Armendariz-Borunda
- Institute for Molecular Biology in Medicine and Gene Therapy, Department of Molecular Biology and Genomics, CUCS, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico.
- Tecnologico de Monterrey, Campus Guadalajara, Guadalajara 45138, Jalisco, Mexico.
| |
Collapse
|
21
|
Wellberg EA, Kabos P, Gillen AE, Jacobsen BM, Brechbuhl HM, Johnson SJ, Rudolph MC, Edgerton SM, Thor AD, Anderson SM, Elias A, Zhou XK, Iyengar NM, Morrow M, Falcone DJ, El-Hely O, Dannenberg AJ, Sartorius CA, MacLean PS. FGFR1 underlies obesity-associated progression of estrogen receptor-positive breast cancer after estrogen deprivation. JCI Insight 2018; 3:120594. [PMID: 30046001 PMCID: PMC6124402 DOI: 10.1172/jci.insight.120594] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 06/19/2018] [Indexed: 01/07/2023] Open
Abstract
Obesity increases breast cancer mortality by promoting resistance to therapy. Here, we identified regulatory pathways in estrogen receptor-positive (ER-positive) tumors that were shared between patients with obesity and those with resistance to neoadjuvant aromatase inhibition. Among these was fibroblast growth factor receptor 1 (FGFR1), a known mediator of endocrine therapy resistance. In a preclinical model with patient-derived ER-positive tumors, diet-induced obesity promoted a similar gene expression signature and sustained the growth of FGFR1-overexpressing tumors after estrogen deprivation. Tumor FGFR1 phosphorylation was elevated with obesity and predicted a shorter disease-free and disease-specific survival for patients treated with tamoxifen. In both human and mouse mammary adipose tissue, FGF1 ligand expression was associated with metabolic dysfunction, weight gain, and adipocyte hypertrophy, implicating the impaired response to a positive energy balance in growth factor production within the tumor niche. In conjunction with these studies, we describe a potentially novel graft-competent model that can be used with patient-derived tissue to elucidate factors specific to extrinsic (host) and intrinsic (tumor) tissue that are critical for obesity-associated tumor promotion. Taken together, we demonstrate that obesity and excess energy establish a tumor environment with features of endocrine therapy resistance and identify a role for ligand-dependent FGFR1 signaling in obesity-associated breast cancer progression.
Collapse
Affiliation(s)
| | - Peter Kabos
- Division of Medical Oncology, Department of Medicine, University of Colorado Denver, Aurora, Colorado, USA
| | - Austin E. Gillen
- University of Colorado School of Medicine, RNA Bioscience Initiative, Aurora, Colorado, USA
| | - Britta M. Jacobsen
- Department of Pathology and
- Division of Endocrinology, Metabolism, & Diabetes, Department of Medicine, University of Colorado Denver, Aurora, Colorado, USA
| | - Heather M. Brechbuhl
- Division of Medical Oncology, Department of Medicine, University of Colorado Denver, Aurora, Colorado, USA
| | | | - Michael C. Rudolph
- Division of Endocrinology, Metabolism, & Diabetes, Department of Medicine, University of Colorado Denver, Aurora, Colorado, USA
| | | | | | | | - Anthony Elias
- Division of Medical Oncology, Department of Medicine, University of Colorado Denver, Aurora, Colorado, USA
| | - Xi Kathy Zhou
- Department of Healthcare Policy and Research, Weill Cornell Medical College, New York, New York, USA
| | - Neil M. Iyengar
- Department of Medicine, Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York, USA
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Monica Morrow
- Department of Surgery, MSKCC, New York, New York, USA
| | - Domenick J. Falcone
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical Center, New York, New York, USA
| | - Omar El-Hely
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | | | | | - Paul S. MacLean
- Division of Endocrinology, Metabolism, & Diabetes, Department of Medicine, University of Colorado Denver, Aurora, Colorado, USA
| |
Collapse
|
22
|
Dalby MJ, Ross AW, Walker AW, Morgan PJ. Dietary Uncoupling of Gut Microbiota and Energy Harvesting from Obesity and Glucose Tolerance in Mice. Cell Rep 2018; 21:1521-1533. [PMID: 29117558 PMCID: PMC5695904 DOI: 10.1016/j.celrep.2017.10.056] [Citation(s) in RCA: 143] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 09/05/2017] [Accepted: 10/13/2017] [Indexed: 12/27/2022] Open
Abstract
Evidence suggests that altered gut microbiota composition may be involved in the development of obesity. Studies using mice made obese with refined high-fat diets have supported this; however, these have commonly used chow as a control diet, introducing confounding factors from differences in dietary composition that have a key role in shaping microbiota composition. We compared the effects of feeding a refined high-fat diet with those of feeding either a refined low-fat diet or a chow diet on gut microbiota composition and host physiology. Feeding both refined low- or high-fat diets resulted in large alterations in the gut microbiota composition, intestinal fermentation, and gut morphology, compared to a chow diet. However, body weight, body fat, and glucose intolerance only increased in mice fed the refined high-fat diet. The choice of control diet can dissociate broad changes in microbiota composition from obesity, raising questions about the previously proposed relationship between gut microbiota and obesity. High-fat diet changes in mouse gut microbiota composition due to diet, not obesity Glucose intolerance in mice linked to high-fat diet, not changes in gut microbiota Cecal fermentation (energy harvest) decreased, not increased, by high-fat diet Choice of control diet is vitally important to studies of microbiota composition
Collapse
Affiliation(s)
- Matthew J Dalby
- Rowett Institute, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Alexander W Ross
- Rowett Institute, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Alan W Walker
- Rowett Institute, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Peter J Morgan
- Rowett Institute, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK.
| |
Collapse
|
23
|
Mehanna ET, Barakat BM, ElSayed MH, Tawfik MK. An optimized dose of raspberry ketones controls hyperlipidemia and insulin resistance in male obese rats: Effect on adipose tissue expression of adipocytokines and Aquaporin 7. Eur J Pharmacol 2018; 832:81-89. [PMID: 29787773 DOI: 10.1016/j.ejphar.2018.05.028] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 04/26/2018] [Accepted: 05/18/2018] [Indexed: 01/04/2023]
Abstract
Obesity constitutes a major worldwide problem in which hyperlipidemia and insulin resistance represents adverse metabolic consequences of it. The present study was conducted to elucidate the role of raspberry ketones (RKs) in controlling body weight gain, hyperlipidemia and insulin resistance in male obese rats through affecting the expression of various adipocytokines. As Aquaporin-7 is co-related with the expression of various adipocytokines and has recently emerged as a modulator of adipocyte metabolism, the present study evaluated the effect of RKs on adipose tissue expression of aquaporin-7(AQP7) in high-fat (HF) diet-fed rats. Groups of male rats were assigned to normal, HF diet-fed control rats and RKs-treated (250 and 500 mg/kg) groups. RKs administration effectively abrogated hyperlipidemia and oxidative burden and enhanced insulin sensitivity. In addition, treatment with RKs ameliorated adipose tissue and liver indices and the reduced adipocyte diameters. Moreover, administration of the low dose of RKs ameliorated the expression of apelin and its receptor, and visfatin with upregulating adiponectin expression compared to HF diet control rats. However, both doses effectively downregulated leptin expression. It was obvious that both RKs doses revealed effectiveness in upregulating the AQP7 expression. The present data suggest the promising therapeutic role of RKs in HF diet-induced obesity that is likely attributable, at least in part, to upregulation of AQP7 expression.
Collapse
Affiliation(s)
- Eman T Mehanna
- Department of Biochemistry, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt
| | - Bassant M Barakat
- Department of Pharmacology & Toxicology, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt; Department of Clinical Pharmacy, College of Clinical Pharmacy, Al-Baha University, Al-Baha, Saudi Arabia
| | - Mohamed H ElSayed
- Department of Physiology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Mona K Tawfik
- Department of Pharmacology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt.
| |
Collapse
|
24
|
Haissaguerre M, Ferriere A, Clark S, Guzman-Quevedo O, Tabarin A, Cota D. NPV-BSK805, an Antineoplastic Jak2 Inhibitor Effective in Myeloproliferative Disorders, Causes Adiposity in Mice by Interfering With the Action of Leptin. Front Pharmacol 2018; 9:527. [PMID: 29867515 PMCID: PMC5962752 DOI: 10.3389/fphar.2018.00527] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 05/02/2018] [Indexed: 11/23/2022] Open
Abstract
The pathophysiology of body weight gain that is observed in patients suffering from myeloproliferative neoplasms treated with inhibitors of the janus kinase (Jak) 1 and 2 pathway remains unknown. Here we hypothesized that this class of drugs interferes with the metabolic actions of leptin, as this hormone requires functional Jak2 signaling. To test this, C57BL/6J chow-fed mice received either chronic intraperitoneal (ip) or repeated intracerebroventricular (icv) administration of the selective Jak2 inhibitor NVP-BSK805, which was proven efficacious in treating polycythemia in rodents. Changes in food intake, body weight and body composition were recorded. Icv NVP-BSK805 was combined with ip leptin to evaluate ability to interfere with the action of this hormone on food intake and on induction of hypothalamic phosphorylation of signal transducer and activator of transcription 3 (STAT3). We found that chronic peripheral administration of NVP-BSK805 did not alter food intake, but increased fat mass and feed efficiency. The increase in fat mass was more pronounced during repeated icv administration of the compound, suggesting that metabolic effects were related to molecular interference in brain structures regulating energy balance. Accordingly, acute icv administration of NVP-BSK805 prevented the ability of leptin to decrease food intake and body weight by impeding STAT3 phosphorylation within the hypothalamus. Consequently, acute icv administration of NVP-BSK805 at higher dose induced hyperphagia and body weight gain. Our results provide evidence for a specific anabolic effect exerted by antineoplastic drugs targeting the Jak2 pathway, which is due to interference with the actions of leptin. Consequently, assessment of metabolic variables related to increased fat mass gain should be performed in patients treated with Jak2 inhibitors.
Collapse
Affiliation(s)
- Magalie Haissaguerre
- INSERM, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, Bordeaux, France
- University of Bordeaux, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, Bordeaux, France
- Service d’Endocrinologie, Diabétologie et Nutrition, Hôpital Haut-Lévêque, CHU de Bordeaux, Pessac, France
| | - Amandine Ferriere
- INSERM, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, Bordeaux, France
- University of Bordeaux, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, Bordeaux, France
- Service d’Endocrinologie, Diabétologie et Nutrition, Hôpital Haut-Lévêque, CHU de Bordeaux, Pessac, France
| | - Samantha Clark
- INSERM, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, Bordeaux, France
- University of Bordeaux, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, Bordeaux, France
| | - Omar Guzman-Quevedo
- INSERM, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, Bordeaux, France
- University of Bordeaux, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, Bordeaux, France
- Facultad de Químico-Farmacobiología, Universidad Michoacána de San Nicolás de Hidalgo, Morelia, Mexico
| | - Antoine Tabarin
- INSERM, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, Bordeaux, France
- University of Bordeaux, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, Bordeaux, France
- Service d’Endocrinologie, Diabétologie et Nutrition, Hôpital Haut-Lévêque, CHU de Bordeaux, Pessac, France
- *Correspondence: Antoine Tabarin, Daniela Cota,
| | - Daniela Cota
- INSERM, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, Bordeaux, France
- University of Bordeaux, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, Bordeaux, France
- *Correspondence: Antoine Tabarin, Daniela Cota,
| |
Collapse
|
25
|
Panagiotou M, Meijer JH, Deboer T. Chronic high-caloric diet modifies sleep homeostasis in mice. Eur J Neurosci 2018; 47:1339-1352. [PMID: 29737605 DOI: 10.1111/ejn.13932] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 03/13/2018] [Accepted: 03/15/2018] [Indexed: 11/29/2022]
Abstract
Obesity prevalence and sleep habit changes are commonplace nowadays, due to modern lifestyle. A bidirectional relationship likely exists between sleep quality and metabolic disruptions, which could impact quality of life. In our study, we investigated the effects of a chronic high-caloric diet on sleep architecture and sleep regulation in mice. We studied the effect of 3 months high-caloric diet (HCD, 45% fat) on sleep and the sleep electroencephalogram (EEG) in C57BL/6J mice during 24-hr baseline (BL) recordings, and after 6-hr sleep deprivation (SD). We examined the effect of HCD on sleep homeostasis, by performing parameter estimation analysis and simulations of the sleep homeostatic Process S, a measure of sleep pressure, which is reflected in the non-rapid-eye-movement (NREM) sleep slow-wave-activity (SWA, EEG power density between 0.5 and 4.0 Hz). Compared to controls (n = 11, 30.7 ± 0.8 g), mice fed with HCD (n = 9, 47.6 ± 0.8 g) showed an increased likelihood of consecutive NREM-REM sleep cycles, increased REM sleep and decreased NREM sleep EEG SWA. After SD, these effects were more pronounced. The simulation resulted in a close fit between the time course of SWA and Process S in both groups. HCD fed mice had a slower time constant (Ti = 15.98 hr) for the increase in homeostatic sleep pressure compared with controls (5.95 hr) indicating a reduced effect of waking on the increase in sleep pressure. Our results suggest that chronic HCD consumption impacts sleep regulation.
Collapse
Affiliation(s)
- Maria Panagiotou
- Laboratory for Neurophysiology, Department of Cell and Chemical Biology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Johanna H Meijer
- Laboratory for Neurophysiology, Department of Cell and Chemical Biology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Tom Deboer
- Laboratory for Neurophysiology, Department of Cell and Chemical Biology, Leiden University Medical Centre, Leiden, The Netherlands
| |
Collapse
|
26
|
Barrington WT, Wulfridge P, Wells AE, Rojas CM, Howe SYF, Perry A, Hua K, Pellizzon MA, Hansen KD, Voy BH, Bennett BJ, Pomp D, Feinberg AP, Threadgill DW. Improving Metabolic Health Through Precision Dietetics in Mice. Genetics 2018; 208:399-417. [PMID: 29158425 PMCID: PMC5753872 DOI: 10.1534/genetics.117.300536] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 11/10/2017] [Indexed: 12/30/2022] Open
Abstract
The incidence of diet-induced metabolic disease has soared over the last half-century, despite national efforts to improve health through universal dietary recommendations. Studies comparing dietary patterns of populations with health outcomes have historically provided the basis for healthy diet recommendations. However, evidence that population-level diet responses are reliable indicators of responses across individuals is lacking. This study investigated how genetic differences influence health responses to several popular diets in mice, which are similar to humans in genetic composition and the propensity to develop metabolic disease, but enable precise genetic and environmental control. We designed four human-comparable mouse diets that are representative of those eaten by historical human populations. Across four genetically distinct inbred mouse strains, we compared the American diet's impact on metabolic health to three alternative diets (Mediterranean, Japanese, and Maasai/ketogenic). Furthermore, we investigated metabolomic and epigenetic alterations associated with diet response. Health effects of the diets were highly dependent on genetic background, demonstrating that individualized diet strategies improve health outcomes in mice. If similar genetic-dependent diet responses exist in humans, then a personalized, or "precision dietetics," approach to dietary recommendations may yield better health outcomes than the traditional one-size-fits-all approach.
Collapse
Affiliation(s)
- William T Barrington
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, Texas A&M University, College Station, Texas 77843
- Department of Biological Sciences, Genetics Program, North Carolina State University, Raleigh, North Carolina 27695
| | - Phillip Wulfridge
- Center for Epigenetics, Institute for Basic Biomedical Sciences, Johns Hopkins University, Baltimore, Maryland 21205
| | - Ann E Wells
- UT-ORNL Graduate School of Genome Science and Technology, Department of Animal Science, University of Tennessee, Knoxville, Tennessee 37996
| | - Carolina Mantilla Rojas
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, Texas A&M University, College Station, Texas 77843
| | - Selene Y F Howe
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, Texas A&M University, College Station, Texas 77843
| | - Amie Perry
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas 77843
| | - Kunjie Hua
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina 27559
| | | | - Kasper D Hansen
- Center for Epigenetics, Institute for Basic Biomedical Sciences, Johns Hopkins University, Baltimore, Maryland 21205
- Department of Biostatistics, Johns Hopkins University, Baltimore, Maryland 21205
- Nathan-McKusick Institute of Genetic Medicine, Johns Hopkins University, Baltimore, Maryland 21205
| | - Brynn H Voy
- UT-ORNL Graduate School of Genome Science and Technology, Department of Animal Science, University of Tennessee, Knoxville, Tennessee 37996
| | - Brian J Bennett
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina 27559
| | - Daniel Pomp
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina 27559
| | - Andrew P Feinberg
- Center for Epigenetics, Institute for Basic Biomedical Sciences, Johns Hopkins University, Baltimore, Maryland 21205
| | - David W Threadgill
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, Texas A&M University, College Station, Texas 77843
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas 77843
- Faculty of Nutrition, Texas A&M University, College Station, Texas 77843
- Faculty of Genetics, Texas A&M University, College Station, Texas 77843
- Faculty of Toxicology, Texas A&M University, College Station, Texas 77843
| |
Collapse
|
27
|
Zhang SB, Yang S, Zhang Z, Zhang A, Zhang M, Yin L, Casey-Sawicki K, Swarts S, Vidyasagar S, Zhang L, Okunieff P. Thoracic gamma irradiation-induced obesity in C57BL/6 female mice. Int J Radiat Biol 2017; 93:1334-1342. [DOI: 10.1080/09553002.2017.1385871] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Steven B. Zhang
- Department of Radiation Oncology, University of Florida Health Cancer Center, Gainesville, FL, USA
| | - Shanmin Yang
- Department of Radiation Oncology, University of Florida Health Cancer Center, Gainesville, FL, USA
| | - Zhenhuan Zhang
- Department of Radiation Oncology, University of Florida Health Cancer Center, Gainesville, FL, USA
| | - Amy Zhang
- Department of Radiation Oncology, University of Florida Health Cancer Center, Gainesville, FL, USA
| | - Mei Zhang
- Department of Radiation Oncology, University of Florida Health Cancer Center, Gainesville, FL, USA
| | - Liangjie Yin
- Department of Radiation Oncology, University of Florida Health Cancer Center, Gainesville, FL, USA
| | - Katherine Casey-Sawicki
- Department of Radiation Oncology, University of Florida Health Cancer Center, Gainesville, FL, USA
| | - Steven Swarts
- Department of Radiation Oncology, University of Florida Health Cancer Center, Gainesville, FL, USA
| | - Sadasivan Vidyasagar
- Department of Radiation Oncology, University of Florida Health Cancer Center, Gainesville, FL, USA
| | - Lurong Zhang
- Department of Radiation Oncology, University of Florida Health Cancer Center, Gainesville, FL, USA
| | - Paul Okunieff
- Department of Radiation Oncology, University of Florida Health Cancer Center, Gainesville, FL, USA
| |
Collapse
|
28
|
Thaete LG, Qu XW, Neerhof MG, Hirsch E, Jilling T. Fetal Growth Restriction Induced by Transient Uterine Ischemia-Reperfusion: Differential Responses in Different Mouse Strains. Reprod Sci 2017; 25:1083-1092. [PMID: 28946817 DOI: 10.1177/1933719117732160] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
We characterized fetal and placental growth and uterine and placental inflammation in pregnant C3H/HeOuJ and C57BL/6J mice (strains with different sensitivities to metabolic and circulatory pathologies), using different uterine ischemia/reperfusion (I/R) protocols, to establish and refine a murine model of I/R-induced fetal growth restriction (FGR). Pregnant C3H/HeOuJ mice on gestation day 15 were subjected to unilateral uterine I/R by (1) total blood flow restriction (TFR) by occlusion of the right ovarian and uterine arteries for 30 minutes, (2) partial flow restriction (PFR) by occlusion of only the right ovarian artery for 30 minutes, or (3) sham surgery. Pregnant C57BL/6J mice were treated the same, but on gestation day 14 and with TFR for only 5 minutes due to high sensitivity of C57BL/6J mice to I/R. Four days post-I/R, the animals were euthanized to determine fetal and placental weight and fetal loss and to assay placental myeloperoxidase (MPO) activity. In C3H/HeOuJ mice, TFR/30 minutes induced significantly ( P < .05) lower fetal and placental weights and higher placental MPO activity, compared to controls. The PFR/30 minutes produced the same effects except placental weights were not reduced. In contrast, in C57BL/6J mice, TFR for only 5 minutes was sufficient to induce FGR and increase fetal loss; while PFR/30 minutes lowered fetal but not placental weights and increased fetal loss but not placental MPO activity. In summary, we present the first published model of I/R-induced FGR in mice. We find that mice of different strains have differing sensitivities to uterine I/R, therefore differing I/R response mechanisms.
Collapse
Affiliation(s)
- Larry G Thaete
- 1 Department of Obstetrics and Gynecology, NorthShore University HealthSystem, Evanston, IL, USA.,2 Department of Obstetrics and Gynecology, The University of Chicago, Pritzker School of Medicine, Chicago, IL, USA
| | - Xiao-Wu Qu
- 1 Department of Obstetrics and Gynecology, NorthShore University HealthSystem, Evanston, IL, USA
| | - Mark G Neerhof
- 1 Department of Obstetrics and Gynecology, NorthShore University HealthSystem, Evanston, IL, USA.,2 Department of Obstetrics and Gynecology, The University of Chicago, Pritzker School of Medicine, Chicago, IL, USA
| | - Emmet Hirsch
- 1 Department of Obstetrics and Gynecology, NorthShore University HealthSystem, Evanston, IL, USA.,2 Department of Obstetrics and Gynecology, The University of Chicago, Pritzker School of Medicine, Chicago, IL, USA
| | - Tamas Jilling
- 3 Division of Neonatology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
29
|
Zhao J, Li SW, Gong QQ, Ding LC, Jin YC, Zhang J, Gao JG, Sun XY. A disputed evidence on obesity: comparison of the effects of Rcan2(-/-) and Rps6kb1(-/-) mutations on growth and body weight in C57BL/6J mice. J Zhejiang Univ Sci B 2017; 17:657-71. [PMID: 27604858 DOI: 10.1631/jzus.b1600276] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
It is widely accepted that body weight and adipose mass are tightly regulated by homeostatic mechanisms, in which leptin plays a critical role through hypothalamic pathways, and obesity is a result of homeostatic disorder. However, in C57BL/6J mice, we found that Rcan2 increases food intake and plays an important role in the development of age- and diet-induced obesity through a leptin-independent mechanism. RCAN2 was initially identified as a thyroid hormone (T3)-responsive gene in human fibroblasts. Expression of RCAN2 is regulated by T3 through the PI3K-Akt/PKB-mTOR-Rps6kb1 signaling pathway. Intriguingly, both Rcan2(-/-) and Rps6kb1(-/-) mutations were reported to result in lean phenotypes in mice. In this study we compared the effects of these two mutations on growth and body weight in C57BL/6J mice. We observed reduced body weight and lower fat mass in both Rcan2(-/-) and Rps6kb1(-/-) mice compared to the wild-type mice, and we reported other differences unique to either the Rcan2(-/-) or Rps6kb1(-/-) mice. Firstly, loss of Rcan2 does not directly alter body length; however, Rcan2(-/-) mice exhibit reduced food intake. In contrast, Rps6kb1(-/-) mice exhibit abnormal embryonic development, which leads to smaller body size and reduced food intake in adulthood. Secondly, when fed a normal chow diet, Rcan2(-/-) mice weigh significantly more than Rps6kb1(-/-) mice, but both Rcan2(-/-) and Rps6kb1(-/-) mice develop similar amounts of epididymal fat. On a high-fat diet, Rcan2(-/-) mice gain body weight and fat mass at slower rates than Rps6kb1(-/-) mice. Finally, using the double-knockout mice (Rcan2(-/-) Rps6kb1(-/-)), we demonstrate that concurrent loss of Rcan2 and Rps6kb1 has an additive effect on body weight reduction in C57BL/6J mice. Our data suggest that Rcan2 and Rps6kb1 mutations both affect growth and body weight of mice, though likely through different mechanisms.
Collapse
Affiliation(s)
- Jing Zhao
- Institute of Developmental Biology, School of Life Science, Shandong University, Jinan 250100, China
| | - Shi-Wei Li
- Institute of Developmental Biology, School of Life Science, Shandong University, Jinan 250100, China
| | - Qian-Qian Gong
- Institute of Developmental Biology, School of Life Science, Shandong University, Jinan 250100, China
| | - Ling-Cui Ding
- Institute of Developmental Biology, School of Life Science, Shandong University, Jinan 250100, China
| | - Ye-Cheng Jin
- Institute of Developmental Biology, School of Life Science, Shandong University, Jinan 250100, China
| | - Jian Zhang
- Institute of Developmental Biology, School of Life Science, Shandong University, Jinan 250100, China
| | - Jian-Gang Gao
- Institute of Developmental Biology, School of Life Science, Shandong University, Jinan 250100, China
| | - Xiao-Yang Sun
- Institute of Developmental Biology, School of Life Science, Shandong University, Jinan 250100, China
| |
Collapse
|
30
|
Weiner J, Rohde K, Krause K, Zieger K, Klöting N, Kralisch S, Kovacs P, Stumvoll M, Blüher M, Böttcher Y, Heiker JT. Brown adipose tissue (BAT) specific vaspin expression is increased after obesogenic diets and cold exposure and linked to acute changes in DNA-methylation. Mol Metab 2017; 6:482-493. [PMID: 28580279 PMCID: PMC5444018 DOI: 10.1016/j.molmet.2017.03.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 03/10/2017] [Accepted: 03/15/2017] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE Several studies have demonstrated anti-diabetic and anti-obesogenic properties of visceral adipose tissue-derived serine protease inhibitor (vaspin) and so evoked its potential use for treatment of obesity-related diseases. The aim of the study was to unravel physiological regulators of vaspin expression and secretion with a particular focus on its role in brown adipose tissue (BAT) biology. METHODS We analyzed the effects of obesogenic diets and cold exposure on vaspin expression in liver and white and brown adipose tissue (AT) and plasma levels. Vaspin expression was analyzed in isolated white and brown adipocytes during adipogenesis and in response to adrenergic stimuli. DNA-methylation within the vaspin promoter was analyzed to investigate acute epigenetic changes after cold-exposure in BAT. RESULTS Our results demonstrate a strong induction of vaspin mRNA and protein expression specifically in BAT of both cold-exposed and high-fat (HF) or high-sugar (HS) fed mice. While obesogenic diets also upregulated hepatic vaspin mRNA levels, cold exposure tended to increase vaspin gene expression of inguinal white adipose tissue (iWAT) depots. Concomitantly, vaspin plasma levels were decreased upon obesogenic or thermogenic triggers. Vaspin expression was increased during adipogenesis but unaffected by sympathetic activation in brown adipocytes. Analysis of vaspin promoter methylation in AT revealed lowest methylation levels in BAT, which were acutely reduced after cold exposure. CONCLUSIONS Our data demonstrate a novel BAT-specific regulation of vaspin gene expression upon physiological stimuli in vivo with acute epigenetic changes that may contribute to cold-induced expression in BAT. We conclude that these findings indicate functional relevance and potentially beneficial effects of vaspin in BAT function.
Collapse
Affiliation(s)
- Juliane Weiner
- Divisions of Endocrinology and Nephrology, University of Leipzig, Leipzig, Germany
- Institute of Biochemistry, Faculty of Biosciences, Pharmacy and Psychology, University of Leipzig, Leipzig, Germany
| | - Kerstin Rohde
- Leipzig University Medical Center, IFB Adiposity Diseases, University of Leipzig, Leipzig, Germany
- Department of Clinical Molecular Biology, University of Oslo, Oslo, Norway
| | - Kerstin Krause
- Divisions of Endocrinology and Nephrology, University of Leipzig, Leipzig, Germany
| | - Konstanze Zieger
- Institute of Biochemistry, Faculty of Biosciences, Pharmacy and Psychology, University of Leipzig, Leipzig, Germany
| | - Nora Klöting
- Divisions of Endocrinology and Nephrology, University of Leipzig, Leipzig, Germany
- Leipzig University Medical Center, IFB Adiposity Diseases, University of Leipzig, Leipzig, Germany
| | - Susan Kralisch
- Divisions of Endocrinology and Nephrology, University of Leipzig, Leipzig, Germany
- Leipzig University Medical Center, IFB Adiposity Diseases, University of Leipzig, Leipzig, Germany
| | - Peter Kovacs
- Leipzig University Medical Center, IFB Adiposity Diseases, University of Leipzig, Leipzig, Germany
| | - Michael Stumvoll
- Divisions of Endocrinology and Nephrology, University of Leipzig, Leipzig, Germany
| | - Matthias Blüher
- Divisions of Endocrinology and Nephrology, University of Leipzig, Leipzig, Germany
- Leipzig University Medical Center, IFB Adiposity Diseases, University of Leipzig, Leipzig, Germany
| | - Yvonne Böttcher
- Leipzig University Medical Center, IFB Adiposity Diseases, University of Leipzig, Leipzig, Germany
- Department of Clinical Molecular Biology, University of Oslo, Oslo, Norway
| | - John T. Heiker
- Divisions of Endocrinology and Nephrology, University of Leipzig, Leipzig, Germany
- Institute of Biochemistry, Faculty of Biosciences, Pharmacy and Psychology, University of Leipzig, Leipzig, Germany
- Leipzig University Medical Center, IFB Adiposity Diseases, University of Leipzig, Leipzig, Germany
| |
Collapse
|
31
|
Long-term consumption of sugar-sweetened beverage during the growth period promotes social aggression in adult mice with proinflammatory responses in the brain. Sci Rep 2017; 7:45693. [PMID: 28393871 PMCID: PMC5385878 DOI: 10.1038/srep45693] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 03/06/2017] [Indexed: 12/29/2022] Open
Abstract
Overconsumption of sugar-sweetened beverages (SSBs) is known to be a key contributor to the obesity epidemic; however, its effects on behavioral changes are yet to be fully studied. In the present study, we examined the long-term effects of SSB on social aggression in mice. Three-week-old weaned mice started to drink either a 30 w/v% sucrose solution (S30), plain water (CT), or an aspartame solution with sweetness equivalent to the sucrose solution (A30) and continued to drink until they were 11-week-old adults. Aggressive behaviors were assessed by the resident-intruder test. We found that SSB significantly promoted social aggression, accompanied by heightened serum corticosterone and reduced body weight. To understand the underlying mechanism, we performed transcriptome analyses of brain. The profiles of mice on S30 were dramatically different from those on CT or A30. Transcriptional networks related to immunological function were significantly dysregulated by SSB. FACS analysis of mice on S30 revealed increased numbers of inflammatory cells in peripheral blood. Interestingly, the artificial sweetener failed to mimic the effects of sugar on social aggression and inflammatory responses. These results demonstrate that SSB promotes aggressive behaviors and provide evidence that sugar reduction strategies may be useful in efforts to prevent social aggression.
Collapse
|
32
|
Differences in Photic Entrainment of Circadian Locomotor Activity Between Lean and Obese Volcano Mice ( Neotomodon alstoni). J Circadian Rhythms 2017; 15:1. [PMID: 30210555 PMCID: PMC5356206 DOI: 10.5334/jcr.145] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Obesity is a growing problem worldwide with a clear impact on health status. It is also a condition that negatively affects circadian rhythms. When the mouse Neotomodon alstoni is fed a regular rodent chow, some individuals develop obesity, representing an opportunity to compare the effects of spontaneous obesity upon the circadian organization in this species with that observed in other rodents with induced obesity. We report differences in the free running circadian locomotor activity rhythm and in the effects of light pulses between lean and obese mice. Also, the photo-induced expression of the c-Fos protein and vasoactive intestinal peptide (VIP) in the suprachiasmatic nucleus (SCN) were examined at circadian time (CT) 14 and 22. We show that obese mice have a larger dispersion of the period of circadian locomotor rhythm in constant darkness. Photic induced phase shifts are nearly 50% shorter at CT 14, and 50% larger at CT 22 than in lean mice. The photoinduction of VIP in the SCN at CT 22 was larger in obese mice, which may be related to the differences observed in photic phase shifting. Our work indicates that the obesity in Neotomodon has effects on the neural mechanisms that regulate the circadian system.
Collapse
|
33
|
Mamounis KJ, Yasrebi A, Roepke TA. Linoleic acid causes greater weight gain than saturated fat without hypothalamic inflammation in the male mouse. J Nutr Biochem 2016; 40:122-131. [PMID: 27886622 DOI: 10.1016/j.jnutbio.2016.10.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 10/27/2016] [Accepted: 10/27/2016] [Indexed: 01/22/2023]
Abstract
A significant change in the Western diet, concurrent with the obesity epidemic, was a substitution of saturated fatty acids with polyunsaturated, specifically linoleic acid (LA). Despite increasing investigation on type as well as amount of fat, it is unclear which fatty acids are most obesogenic. The objective of this study was to determine the obesogenic potency of LA vs. saturated fatty acids and the involvement of hypothalamic inflammation. Forty-eight mice were divided into four groups: low-fat or three high-fat diets (HFDs, 45% kcals from fat) with LA comprising 1%, 15% and 22.5% of kilocalories, the balance being saturated fatty acids. Over 12 weeks, bodyweight, body composition, food intake, calorimetry, and glycemia assays were performed. Arcuate nucleus and blood were collected for mRNA and protein analysis. All HFD-fed mice were heavier and less glucose tolerant than control. The diet with 22.5% LA caused greater bodyweight gain, decreased activity, and insulin resistance compared to control and 1% LA. All HFDs elevated leptin and decreased ghrelin in plasma. Neuropeptides gene expression was higher in 22.5% HFD. The inflammatory gene Ikk was suppressed in 1% and 22.5% LA. No consistent pattern of inflammatory gene expression was observed, with suppression and augmentation of genes by one or all of the HFDs relative to control. These data indicate that, in male mice, LA induces obesity and insulin resistance and reduces activity more than saturated fat, supporting the hypothesis that increased LA intake may be a contributor to the obesity epidemic.
Collapse
Affiliation(s)
- Kyle J Mamounis
- Department of Animal Sciences, School of Environmental & Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA; Nutritional Sciences Graduate Program, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Ali Yasrebi
- Department of Animal Sciences, School of Environmental & Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Troy A Roepke
- Department of Animal Sciences, School of Environmental & Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA; Nutritional Sciences Graduate Program, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA; New Jersey Institute for Food, Nutrition, and Health, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA.
| |
Collapse
|
34
|
Knight BS, Pennell CE, Shah R, Lye SJ. Strain Differences in the Impact of Dietary Restriction on Fetal Growth and Pregnancy in Mice. Reprod Sci 2016; 14:81-90. [PMID: 17636220 DOI: 10.1177/1933719106298217] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The association between suboptimal intrauterine environment and developmental origins of adult health and disease is variable, suggesting that genotype may contribute to eventual outcome. The objective of this study was to characterize maternal and fetal responses to maternal dietary restriction during pregnancy in 2 phylogenetically distant strains of mice. Pregnant A/J (n=35) and C57BL/6J (B6) (n=36) mice underwent either a 30% dietary restriction (DR) from day 6.5 until day 17.5 of gestation or were fed ad libitum. Seven mothers from each strain and diet were randomly selected for dissection on day 18.5 to assess fetal body and organ weights and maternal endocrine status through the collection of serum to measure progesterone, corticosterone, cortisol, and estradiol levels. The remaining mice were allowed to deliver spontaneously to assess gestational effects. Both strains showed similar responses to maternal DR during pregnancy in terms of reductions in maternal weight gain during pregnancy, reductions in fetal body weight, increased pup death within 24 hours of birth, and decreased placental 11beta-HSD2 protein expression. The impact of maternal DR was greater in B6 mice than A/J when assessing reductions in fetal kidney weight, embryo-placenta ratio, increases in placental weight, fetal brain-liver ratio, and maternal corticosterone and cortisol levels. Moreover, preterm delivery was significantly increased in DR B6 mice compared to DR A/J mice. The observed strain variations in response to dietary restriction may offer a unique opportunity to investigate gene-environment interactions associated with developmental origins of adult health and disease.
Collapse
Affiliation(s)
- Brian S Knight
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | | | | | | |
Collapse
|
35
|
Li X, Lu J, Wang Y, Huo X, Li Z, Zhang S, Li C, Guo M, Du X, Chen Z. Establishment and Characterization of a Newly Established Diabetic Gerbil Line. PLoS One 2016; 11:e0159420. [PMID: 27427908 PMCID: PMC4948894 DOI: 10.1371/journal.pone.0159420] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 07/01/2016] [Indexed: 01/09/2023] Open
Abstract
Objectives We aimed to selectively breed a spontaneous diabetic gerbil when a sub-line of inbred gerbil showed increased blood glucose levels was found recently. Then we investigated the characteristics including the serum insulin, triglyceride, cholesterol, leptin, adiponectin and explored the underlying molecular mechanism for the diabetic phenotype. Methods The spontaneous diabetic line of gerbils was selectively inbreed the sub-line of gerbil by monitoring blood glucose of each animal. The serum insulin, adiponectin, and leptin levels were tested using an ELISA kit. The expression levels of GLUT4, Akt, leptin, adiponectin, and calpain 10 (CAPN10) were tested by western blot and Quantitative Real-time PCR (qPCR) in liver, skeletal muscle, and white adipose. Results Our results show that the percentages of animals with FPG≥5.2 (mmol/l), PG2h≥6.8 (mmol/l) and both FPG≥5.2 and PG2h≥6.8 (mmol/l) were increased with the number of breeding generations from F0 (21.33%) to F6 (38.46%). These diabetic gerbils exhibited insulin resistance and leptin resistance as well as decreased adiponectin level in the serum. We also observed decreased expression of adiponectin and increased expression of leptin in the skeletal muscle, respectively. Conclusions These results indicate that we have primarily established a spontaneous diabetic gerbil line, and the diabetic phenotypes may have been accounted for by altered expression of leptin and adiponectin.
Collapse
Affiliation(s)
- Xiaohong Li
- School of Basic Medical Science, Capital Medical University, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Beijing 100069, China
| | - Jing Lu
- School of Basic Medical Science, Capital Medical University, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Beijing 100069, China
| | - Ying Wang
- School of Basic Medical Science, Capital Medical University, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Beijing 100069, China
| | - Xueyun Huo
- School of Basic Medical Science, Capital Medical University, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Beijing 100069, China
| | - Zhenkun Li
- School of Basic Medical Science, Capital Medical University, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Beijing 100069, China
| | - Shuangyue Zhang
- School of Basic Medical Science, Capital Medical University, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Beijing 100069, China
| | - Changlong Li
- School of Basic Medical Science, Capital Medical University, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Beijing 100069, China
| | - Meng Guo
- School of Basic Medical Science, Capital Medical University, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Beijing 100069, China
| | - Xiaoyan Du
- School of Basic Medical Science, Capital Medical University, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Beijing 100069, China
- * E-mail: (ZC); (XD)
| | - Zhenwen Chen
- School of Basic Medical Science, Capital Medical University, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Beijing 100069, China
- * E-mail: (ZC); (XD)
| |
Collapse
|
36
|
Mouse Strain Impacts Fatty Acid Uptake and Trafficking in Liver, Heart, and Brain: A Comparison of C57BL/6 and Swiss Webster Mice. Lipids 2016; 51:549-60. [PMID: 26797754 DOI: 10.1007/s11745-015-4117-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 12/10/2015] [Indexed: 01/21/2023]
Abstract
C57BL/6 and Swiss Webster mice are used to study lipid metabolism, although differences in fatty acid uptake between these strains have not been reported. Using a steady state kinetic model, [1-(14)C]16:0, [1-(14)C]20:4n-6, or [1-(14)C]22:6n-3 was infused into awake, adult male mice and uptake into liver, heart, and brain determined. The integrated area of [1-(14)C]20:4n-6 in plasma was significantly increased in C57BL/6 mice, but [1-(14)C]16:0 and [1-(14)C]22:6n-3 were not different between groups. In heart, uptake of [1-(14)C]20:4n-6 was increased 1.7-fold in C57BL/6 mice. However, trafficking of [1-(14)C]22:6n-3 into the organic fraction of heart was significantly decreased 33 % in C57BL/6 mice. Although there were limited differences in fatty acid tracer trafficking in liver or brain, [1-(14)C]16:0 incorporation into liver neutral lipids was decreased 18 % in C57BL/6 mice. In heart, the amount of [1-(14)C]16:0 and [1-(14)C]22:6n-3 incorporated into total phospholipids were decreased 45 and 49 %, respectively, in C57BL/6 mice. This was accounted for by a 53 and 37 % decrease in [1-(14)C]16:0 and 44 and 52 % decrease in [1-(14)C]22:6n-3 entering ethanolamine glycerophospholipids and choline glycerophospholipids, respectively. In contrast, there was a significant increase in [1-(14)C]20:4n-6 esterification into all heart phospholipids of C57BL/6 mice. Although changes in uptake were limited to heart, several significant differences were found in fatty acid trafficking into heart, liver, and brain phospholipids. In summary, our data demonstrates differences in tissue fatty acid uptake and trafficking between mouse strains is an important consideration when carrying out fatty acid metabolic studies.
Collapse
|
37
|
Buchner DA, Nadeau JH. Contrasting genetic architectures in different mouse reference populations used for studying complex traits. Genome Res 2015; 25:775-91. [PMID: 25953951 PMCID: PMC4448675 DOI: 10.1101/gr.187450.114] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 03/31/2015] [Indexed: 01/14/2023]
Abstract
Quantitative trait loci (QTLs) are being used to study genetic networks, protein functions, and systems properties that underlie phenotypic variation and disease risk in humans, model organisms, agricultural species, and natural populations. The challenges are many, beginning with the seemingly simple tasks of mapping QTLs and identifying their underlying genetic determinants. Various specialized resources have been developed to study complex traits in many model organisms. In the mouse, remarkably different pictures of genetic architectures are emerging. Chromosome Substitution Strains (CSSs) reveal many QTLs, large phenotypic effects, pervasive epistasis, and readily identified genetic variants. In contrast, other resources as well as genome-wide association studies (GWAS) in humans and other species reveal genetic architectures dominated with a relatively modest number of QTLs that have small individual and combined phenotypic effects. These contrasting architectures are the result of intrinsic differences in the study designs underlying different resources. The CSSs examine context-dependent phenotypic effects independently among individual genotypes, whereas with GWAS and other mouse resources, the average effect of each QTL is assessed among many individuals with heterogeneous genetic backgrounds. We argue that variation of genetic architectures among individuals is as important as population averages. Each of these important resources has particular merits and specific applications for these individual and population perspectives. Collectively, these resources together with high-throughput genotyping, sequencing and genetic engineering technologies, and information repositories highlight the power of the mouse for genetic, functional, and systems studies of complex traits and disease models.
Collapse
Affiliation(s)
- David A Buchner
- Department of Genetics and Genome Sciences, Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Joseph H Nadeau
- Pacific Northwest Diabetes Research Institute, Seattle, Washington 98122, USA
| |
Collapse
|
38
|
Sims EK, Hatanaka M, Morris DL, Tersey SA, Kono T, Chaudry ZZ, Day KH, Moss DR, Stull ND, Mirmira RG, Evans-Molina C. Divergent compensatory responses to high-fat diet between C57BL6/J and C57BLKS/J inbred mouse strains. Am J Physiol Endocrinol Metab 2013; 305:E1495-511. [PMID: 24169046 PMCID: PMC3882376 DOI: 10.1152/ajpendo.00366.2013] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Impaired glucose tolerance (IGT) and type 2 diabetes (T2DM) are polygenic disorders with complex pathophysiologies; recapitulating them with mouse models is challenging. Despite 70% genetic homology, C57BL/6J (BL6) and C57BLKS/J (BLKS) inbred mouse strains differ in response to diet- and genetic-induced obesity. We hypothesized these differences would yield insight into IGT and T2DM susceptibility and response to pharmacological therapies. To this end, male 8-wk-old BL6 and BLKS mice were fed normal chow (18% kcal from fat), high-fat diet (HFD; 42% kcal from fat), or HFD supplemented with the PPARγ agonist pioglitazone (PIO; 140 mg PIO/kg diet) for 16 wk. Assessments of body composition, glucose homeostasis, insulin production, and energy metabolism, as well as histological analyses of pancreata were undertaken. BL6 mice gained weight and adiposity in response to HFD, leading to peripheral insulin resistance that was met with increased β-cell proliferation and insulin production. By contrast, BLKS mice responded to HFD by restricting food intake and increasing activity. These behavioral responses limited weight gain and protected against HFD-induced glucose intolerance, which in this strain was primarily due to β-cell dysfunction. PIO treatment did not affect HFD-induced weight gain in BL6 mice, and decreased visceral fat mass, whereas in BLKS mice PIO increased total fat mass without improving visceral fat mass. Differences in these responses to HFD and effects of PIO reflect divergent human responses to a Western lifestyle and underscore the careful consideration needed when choosing mouse models of diet-induced obesity and diabetes treatment.
Collapse
Affiliation(s)
- Emily K Sims
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Dowling AR, Nedorezov LB, Qiu X, Marino JS, Hill JW. Genetic factors modulate the impact of pubertal androgen excess on insulin sensitivity and fertility. PLoS One 2013; 8:e79849. [PMID: 24278193 PMCID: PMC3835926 DOI: 10.1371/journal.pone.0079849] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Accepted: 09/25/2013] [Indexed: 01/31/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is the most common endocrine disorder of reproductive age women. The syndrome is caused by a combination of environmental influences and genetic predisposition. Despite extensive efforts, the heritable factors contributing to PCOS development are not fully understood. The objective of this study was to test the hypothesis that genetic background contributes to the development of a PCOS-like reproductive and metabolic phenotype in mice exposed to excess DHEA during the pubertal transition. We tested whether the PCOS phenotype would be more pronounced on the diabetes-prone C57BL/6 background than the previously used strain, BALB/cByJ. In addition, we examined strain-dependent upregulation of the expression of ovarian and extra-ovarian candidate genes implicated in human PCOS, genes containing known strain variants, and genes involved with steroidogenesis or insulin sensitivity. These studies show that there are significant strain-related differences in metabolic response to excess androgen exposure during puberty. Additionally, our results suggest the C57BL/6J strain provides a more robust and uniform experimental platform for PCOS research than the BALB/cByJ strain.
Collapse
Affiliation(s)
- Abigail R. Dowling
- University of Toledo Medical Center, Center for Diabetes and Endocrine Research, Department of Physiology and Pharmacology, University of Toledo Medical Center, Toledo, Ohio, United States of America
| | - Laura B. Nedorezov
- University of Toledo Medical Center, Center for Diabetes and Endocrine Research, Department of Physiology and Pharmacology, University of Toledo Medical Center, Toledo, Ohio, United States of America
| | - Xiaoliang Qiu
- University of Toledo Medical Center, Center for Diabetes and Endocrine Research, Department of Physiology and Pharmacology, University of Toledo Medical Center, Toledo, Ohio, United States of America
| | - Joseph S. Marino
- Department of Kinesiology, University of North Carolina, Charlotte, North Carolina, United States of America
| | - Jennifer W. Hill
- University of Toledo Medical Center, Center for Diabetes and Endocrine Research, Department of Physiology and Pharmacology, University of Toledo Medical Center, Toledo, Ohio, United States of America
- Dept. of Obstetrics-Gynecology, University of Toledo Medical Center, Toledo, Ohio, United States of America
| |
Collapse
|
40
|
Tan JTM, McLennan SV, Williams PF, Rezaeizadeh A, Lo LWY, Bonner JG, Twigg SM. Connective tissue growth factor/CCN-2 is upregulated in epididymal and subcutaneous fat depots in a dietary-induced obesity model. Am J Physiol Endocrinol Metab 2013; 304:E1291-302. [PMID: 23571711 DOI: 10.1152/ajpendo.00654.2012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Connective tissue growth factor (CTGF), also known as CCN-2, is a cysteine-rich secreted protein that is involved in a range of biological processes, including regulation of cell growth and differentiation. Our previous in vitro studies have shown that CCN-2 inhibits adipocyte differentiation, although whether CCN-2 is regulated in vivo in adipogenesis is undetermined and was investigated in this study. C57BL/6 male mice were fed either standard laboratory chow (ND) or a diet high in fat (HFD; 45% fat) for 15 or 24 wk. HFD animals that gained >5 g in weight (termed HFD-fat) were insulin resistant and were compared with HFD-fed animals, which failed to gain weight (termed HFD-lean). HFD-fat mice had significantly increased CCN-2 mRNA levels in both the subcutaneous and epididymal fat pads, whereas CCN-2 mRNA was not induced in the epididymal site in HFD-lean mice. Also in HFD-fed animals, epididymal CCN-2 mRNA correlated positively with key genes involved in adipocyte differentiation, adiponectin and PPARγ (P < 0.001 and P < 0.002, respectively). Additionally, epididymal CCN-2 mRNA correlated positively with two markers of tissue turnover, PAI-1 in HFD-fat mice only and TIMP-1, but only in the HFD-lean mice. Collectively, these findings suggest that CCN-2 plays a role in adipocyte differentiation in vivo and thus in the pathogenesis of obesity linked with insulin resistance.
Collapse
Affiliation(s)
- Joanne T M Tan
- Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia; and
| | | | | | | | | | | | | |
Collapse
|
41
|
Mice long-term high-fat diet feeding recapitulates human cardiovascular alterations: an animal model to study the early phases of diabetic cardiomyopathy. PLoS One 2013; 8:e60931. [PMID: 23593350 PMCID: PMC3623942 DOI: 10.1371/journal.pone.0060931] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Accepted: 03/04/2013] [Indexed: 11/30/2022] Open
Abstract
Background/Aim Hypercaloric diet ingestion and sedentary lifestyle result in obesity. Metabolic syndrome is a cluster of clinical features secondary to obesity, considered as a pre-diabetic condition and recognized as an independent risk factor for cardiovascular diseases. To better understand the relationship between obesity, metabolic syndrome and cardiovascular disease as well as for the development of novel therapeutic strategies, animal models that reproduce the etiology, course and outcomes of these pathologies are required. The aim of this work was to characterize the long-term effects of high-fat diet-induced obesity on the mice cardiovascular system, in order to make available a new animal model for diabetic cardiomyopathy. Methods/Results Male C57BL/6 mice were fed with a standardized high-fat diet (obese) or regular diet (normal) for 16 months. Metabolic syndrome was evaluated testing plasma glucose, triglycerides, cholesterol, insulin, and glucose tolerance. Arterial pressure was measured using a sphygmomanometer (non invasive method) and by hemodynamic parameters (invasive method). Cardiac anatomy was described based on echocardiography and histological studies. Cardiac function was assessed by cardiac catheterization under a stress test. Cardiac remodelling and metabolic biomarkers were assessed by RT-qPCR and immunoblotting. As of month eight, the obese mice were overweight, hyperglycaemic, insulin resistant, hyperinsulinemic and hypercholesterolemic. At month 16, they also presented normal arterial pressure but altered vascular reactivity (vasoconstriction), and cardiac contractility reserve reduction, heart mass increase, cardiomyocyte hypertrophy, cardiac fibrosis, and heart metabolic compensations. By contrast, the normal mice remained healthy throughout the study. Conclusions Mice fed with a high-fat diet for prolonged time recapitulates the etiology, course and outcomes of the early phases of human diabetic cardiomyopathy.
Collapse
|
42
|
Pereira CD, Azevedo I, Monteiro R, Martins MJ. 11β-Hydroxysteroid dehydrogenase type 1: relevance of its modulation in the pathophysiology of obesity, the metabolic syndrome and type 2 diabetes mellitus. Diabetes Obes Metab 2012; 14:869-81. [PMID: 22321826 DOI: 10.1111/j.1463-1326.2012.01582.x] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Recent evidence strongly argues for a pathogenic role of glucocorticoids and 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) in obesity and the metabolic syndrome, a cluster of risk factors for atherosclerotic cardiovascular disease and type 2 diabetes mellitus (T2DM) that includes insulin resistance (IR), dyslipidaemia, hypertension and visceral obesity. This has been partially prompted not only by the striking clinical resemblances between the metabolic syndrome and Cushing's syndrome (a state characterized by hypercortisolism that associates with metabolic syndrome components) but also from monogenic rodent models for the metabolic syndrome (e.g. the leptin-deficient ob/ob mouse or the leptin-resistant Zucker rat) that display overall increased secretion of glucocorticoids. However, systemic circulating glucocorticoids are not elevated in obese patients and/or patients with metabolic syndrome. The study of the role of 11β-HSD system shed light on this conundrum, showing that local glucocorticoids are finely regulated in a tissue-specific manner at the pre-receptor level. The system comprises two microsomal enzymes that either activate cortisone to cortisol (11β-HSD1) or inactivate cortisol to cortisone (11β-HSD2). Transgenic rodent models, knockout (KO) for HSD11B1 or with HSD11B1 or HSD11B2 overexpression, specifically targeted to the liver or adipose tissue, have been developed and helped unravel the currently undisputable role of the enzymes in metabolic syndrome pathophysiology, in each of its isolated components and in their prevention. In the transgenic HSD11B1 overexpressing models, different features of the metabolic syndrome and obesity are replicated. HSD11B1 gene deficiency or HSD11B2 gene overexpression associates with improvements in the metabolic profile. In face of these demonstrations, research efforts are now being turned both into the inhibition of 11β-HSD1 as a possible pharmacological target and into the role of dietary habits on the establishment or the prevention of the metabolic syndrome, obesity and T2DM through 11β-HSD1 modulation. We intend to review and discuss 11β-HSD1 and obesity, the metabolic syndrome and T2DM and to highlight the potential of its inhibition for therapeutic or prophylactic approaches in those metabolic diseases.
Collapse
Affiliation(s)
- C D Pereira
- Department of Biochemistry (U38/FCT), Faculty of Medicine, University of Porto, Portugal
| | | | | | | |
Collapse
|
43
|
Kerley-Hamilton JS, Trask HW, Ridley CJA, Dufour E, Ringelberg CS, Nurinova N, Wong D, Moodie KL, Shipman SL, Moore JH, Korc M, Shworak NW, Tomlinson CR. Obesity is mediated by differential aryl hydrocarbon receptor signaling in mice fed a Western diet. ENVIRONMENTAL HEALTH PERSPECTIVES 2012; 120:1252-9. [PMID: 22609946 PMCID: PMC3440132 DOI: 10.1289/ehp.1205003] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Accepted: 05/18/2012] [Indexed: 05/03/2023]
Abstract
BACKGROUND Obesity is a growing worldwide problem with genetic and environmental causes, and it is an underlying basis for many diseases. Studies have shown that the toxicant-activated aryl hydrocarbon receptor (AHR) may disrupt fat metabolism and contribute to obesity. The AHR is a nuclear receptor/transcription factor that is best known for responding to environmental toxicant exposures to induce a battery of xenobiotic-metabolizing genes. OBJECTIVES The intent of the work reported here was to test more directly the role of the AHR in obesity and fat metabolism in lieu of exogenous toxicants. METHODS We used two congenic mouse models that differ at the Ahr gene and encode AHRs with a 10-fold difference in signaling activity. The two mouse strains were fed either a low-fat (regular) diet or a high-fat (Western) diet. RESULTS The Western diet differentially affected body size, body fat:body mass ratios, liver size and liver metabolism, and liver mRNA and miRNA profiles. The regular diet had no significant differential effects. CONCLUSIONS The results suggest that the AHR plays a large and broad role in obesity and associated complications, and importantly, may provide a simple and effective therapeutic strategy to combat obesity, heart disease, and other obesity-associated illnesses.
Collapse
Affiliation(s)
- Joanna S Kerley-Hamilton
- Dartmouth-Hitchcock Medical Center, Lebanon, Norris Cotton Cancer Center, Lebanon, New Hampshire, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Octreotide alleviates obesity by reducing intestinal glucose absorption and inhibiting low-grade inflammation. Eur J Nutr 2012; 52:1067-75. [DOI: 10.1007/s00394-012-0413-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Accepted: 06/27/2012] [Indexed: 11/25/2022]
|
45
|
Pamir N, McMillen TS, Edgel KA, Kim F, LeBoeuf RC. Deficiency of lymphotoxin-α does not exacerbate high-fat diet-induced obesity but does enhance inflammation in mice. Am J Physiol Endocrinol Metab 2012; 302:E961-71. [PMID: 22318945 PMCID: PMC3330720 DOI: 10.1152/ajpendo.00447.2011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2011] [Accepted: 01/30/2012] [Indexed: 12/24/2022]
Abstract
Lymphotoxin-α (LTα) is secreted by lymphocytes and acts through tumor necrosis factor-α receptors and the LTβ receptor. Our goals were to determine whether LT has a role in obesity and investigate whether LT contributes to the link between obesity and adipose tissue lymphocyte accumulation. LT deficient (LT(-/-)) and wild-type (WT) mice were fed standard pelleted rodent chow or a high-fat/high-sucrose diet (HFHS) for 13 wk. Body weight, body composition, and food intake were measured. Glucose tolerance was assessed. Systemic and adipose tissue inflammatory statuses were evaluated by quantifying plasma adipokine levels and tissue macrophage and T cell-specific gene expression in abdominal fat. LT(-/-) mice were smaller (20%) and leaner (25%) than WT controls after 13 wk of HFHS diet feeding. LT(-/-) mice showed improved glucose tolerance, suggesting that, in WT mice, LT may impair glucose metabolism. Surprisingly, adipose tissue from rodent chow- and HFHS-fed LT(-/-) mice exhibited increased T lymphocyte and macrophage infiltration compared with WT mice. Despite the fact that LT(-/-) mice exhibited an enhanced inflammatory status at the systemic and tissue level even when fed rodent chow, they were protected from enhanced diet-induced obesity and insulin resistance. Thus, LT contributes to body weight and adiposity and is required to modulate the accumulation of immune cells in adipose tissue.
Collapse
Affiliation(s)
- Nathalie Pamir
- Department of Medicine, Div. of Metabolism, Endocrinology, and Nutrition, Univ. of Washington, Seattle, WA 98109-8050, USA
| | | | | | | | | |
Collapse
|
46
|
Kim SF. Animal models of eating disorders. Neuroscience 2012; 211:2-12. [PMID: 22465439 DOI: 10.1016/j.neuroscience.2012.03.024] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2011] [Revised: 03/16/2012] [Accepted: 03/16/2012] [Indexed: 01/30/2023]
Abstract
Feeding is a fundamental process for basic survival and is influenced by genetics and environmental stressors. Recent advances in our understanding of behavioral genetics have provided a profound insight on several components regulating eating patterns. However, our understanding of eating disorders, such as anorexia nervosa, bulimia nervosa, and binge eating, is still poor. The animal model is an essential tool in the investigation of eating behaviors and their pathological forms, yet development of an appropriate animal model for eating disorders still remains challenging due to our limited knowledge and some of the more ambiguous clinical diagnostic measures. Therefore, this review will serve to focus on the basic clinical features of eating disorders and the current advances in animal models of eating disorders.
Collapse
Affiliation(s)
- S F Kim
- Center for Neurobiology and Behavior, Department of Psychiatry and Pharmacology, The Perelman School of Medicine University of Pennsylvania, 125 S 31st St., TRL Rm 2207, Philadelphia, PA 19104, USA.
| |
Collapse
|
47
|
Uchida A, Whitsitt MC, Eustaquio T, Slipchenko MN, Leary JF, Cheng JX, Buhman KK. Reduced triglyceride secretion in response to an acute dietary fat challenge in obese compared to lean mice. Front Physiol 2012; 3:26. [PMID: 22375122 PMCID: PMC3285805 DOI: 10.3389/fphys.2012.00026] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2011] [Accepted: 02/03/2012] [Indexed: 11/13/2022] Open
Abstract
Obesity results in abnormally high levels of triglyceride (TG) storage in tissues such as liver, heart, and muscle, which disrupts their normal functions. Recently, we found that lean mice challenged with high levels of dietary fat store TGs in cytoplasmic lipid droplets in the absorptive cells of the intestine, enterocytes, and that this storage increases and then decreases over time after an acute dietary fat challenge. The goal of this study was to investigate the effects of obesity on intestinal TG metabolism. More specifically we asked whether TG storage in and secretion from the intestine are altered in obesity. We investigated these questions in diet-induced obese (DIO) and leptin-deficient (ob/ob) mice. We found greater levels of TG storage in the intestine of DIO mice compared to lean mice in the fed state, but similar levels of TG storage after a 6-h fast. In addition, we found similar TG storage in the intestine of lean and DIO mice at multiple time points after an acute dietary fat challenge. Surprisingly, we found remarkably lower TG secretion from both DIO and ob/ob mice compared to lean controls in response to an acute dietary fat challenge. Furthermore, we found altered mRNA levels for genes involved in regulation of intestinal TG metabolism in lean and DIO mice at 6 h fasting and in response to an acute dietary fat challenge. More specifically, we found that many of the genes related to TG synthesis, chylomicron synthesis, TG storage, and lipolysis were induced in response to an acute dietary fat challenge in lean mice, but this induction was not observed in DIO mice. In fact, we found a significant decrease in intestinal mRNA levels of genes related to lipolysis and fatty acid oxidation in DIO mice in response to an acute dietary fat challenge. Our findings demonstrate altered TG handling by the small intestine of obese compared to lean mice.
Collapse
Affiliation(s)
- Aki Uchida
- Interdisciplinary Life Science Program, Purdue University West Lafayette, IN, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
Mavanji V, Billington CJ, Kotz CM, Teske JA. Sleep and obesity: a focus on animal models. Neurosci Biobehav Rev 2012; 36:1015-29. [PMID: 22266350 DOI: 10.1016/j.neubiorev.2012.01.001] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2011] [Revised: 11/25/2011] [Accepted: 01/07/2012] [Indexed: 12/19/2022]
Abstract
The rapid rise in obesity prevalence in the modern world parallels a significant reduction in restorative sleep (Agras et al., 2004; Dixon et al., 2007, 2001; Gangwisch and Heymsfield, 2004; Gupta et al., 2002; Sekine et al., 2002; Vioque et al., 2000; Wolk et al., 2003). Reduced sleep time and quality increases the risk for obesity, but the underlying mechanisms remain unclear (Gangwisch et al., 2005; Hicks et al., 1986; Imaki et al., 2002; Jennings et al., 2007; Moreno et al., 2006). A majority of the theories linking human sleep disturbances and obesity rely on self-reported sleep. However, studies with objective measurements of sleep/wake parameters suggest a U-shaped relationship between sleep and obesity. Studies in animal models are needed to improve our understanding of the association between sleep disturbances and obesity. Genetic and experimenter-induced models mimicking characteristics of human obesity are now available and these animal models will be useful in understanding whether sleep disturbances determine propensity for obesity, or result from obesity. These models exhibit weight gain profiles consistently different from control animals. Thus a careful evaluation of animal models will provide insight into the relationship between sleep disturbances and obesity in humans. In this review we first briefly consider the fundamentals of sleep and key sleep disturbances, such as sleep fragmentation and excessive daytime sleepiness (EDS), observed in obese individuals. Then we consider sleep deprivation studies and the role of circadian alterations in obesity. We describe sleep/wake changes in various rodent models of obesity and obesity resistance. Finally, we discuss possible mechanisms linking sleep disturbances with obesity.
Collapse
Affiliation(s)
- Vijayakumar Mavanji
- Minnesota Obesity Prevention Training Program, School of Public Health, University of Minnesota, Minneapolis, MN, USA.
| | | | | | | |
Collapse
|
49
|
Pierson CR, Dulin-Smith AN, Durban AN, Marshall ML, Marshall JT, Snyder AD, Naiyer N, Gladman JT, Chandler DS, Lawlor MW, Buj-Bello A, Dowling JJ, Beggs AH. Modeling the human MTM1 p.R69C mutation in murine Mtm1 results in exon 4 skipping and a less severe myotubular myopathy phenotype. Hum Mol Genet 2011; 21:811-25. [PMID: 22068590 DOI: 10.1093/hmg/ddr512] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
X-linked myotubular myopathy (MTM) is a severe neuromuscular disease of infancy caused by mutations of MTM1, which encodes the phosphoinositide lipid phosphatase, myotubularin. The Mtm1 knockout (KO) mouse has a severe phenotype and its short lifespan (8 weeks) makes it a challenge to use as a model in the testing of certain preclinical therapeutics. Many MTM patients succumb early in life, but some have a more favorable prognosis. We used human genotype-phenotype correlation data to develop a myotubularin-deficient mouse model with a less severe phenotype than is seen in Mtm1 KO mice. We modeled the human c.205C>T point mutation in Mtm1 exon 4, which is predicted to introduce the p.R69C missense change in myotubularin. Hemizygous male Mtm1 p.R69C mice develop early muscle atrophy prior to the onset of weakness at 2 months. The median survival period is 66 weeks. Histopathology shows small myofibers with centrally placed nuclei. Myotubularin protein is undetectably low because the introduced c.205C>T base change induced exon 4 skipping in most mRNAs, leading to premature termination of myotubularin translation. Some full-length Mtm1 mRNA bearing the mutation is present, which provides enough myotubularin activity to account for the relatively mild phenotype, as Mtm1 KO and Mtm1 p.R69C mice have similar muscle phosphatidylinositol 3-phosphate levels. These data explain the basis for phenotypic variability among human patients with MTM1 p.R69C mutations and establish the Mtm1 p.R69C mouse as a valuable model for the disease, as its less severe phenotype will expand the scope of testable preclinical therapies.
Collapse
|
50
|
Cation exchange HPLC analysis of desmosines in elastin hydrolysates. Anal Bioanal Chem 2011; 401:2473-9. [PMID: 21887606 PMCID: PMC3184222 DOI: 10.1007/s00216-011-5346-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2011] [Revised: 07/18/2011] [Accepted: 08/17/2011] [Indexed: 10/25/2022]
Abstract
Desmosine crosslinks are responsible for the elastic properties of connective tissues in lungs and cardiovascular system and are often compromised in disease states. We developed a new, fast, and simple cation exchange HPLC assay for the analysis of desmosine and isodesmosine in animal elastin. The method was validated by determining linearity, accuracy, precision, and desmosines stability and was applied to measure levels of desmosines in porcine and murine organs. The detection and quantification limits were 2 and 4 pmol, respectively. The run-time was 8 min. Our cation exchange column does not separate desmosine and isodesmosine, but their level can be quantified from absorbance at different wavelengths. Using this assay, we found that desmosines levels were significantly lower in elastin isolated from various organs of immunodeficient severe combined immunodeficiency mice compared with wild-type animals. We also found that desmosines levels were lower in lung elastin isolated from hyperhomocysteinemic Pcft(-/-) mice deficient in intestinal folate transport compared with wild-type Pcft(+/+) animals.
Collapse
|