1
|
Alsherif DA, Hussein MA, Abuelkasem SS. Salvia officinalis Improves Glycemia and Suppresses Pro-inflammatory Features in Obese Rats with Metabolic Syndrome. Curr Pharm Biotechnol 2024; 25:623-636. [PMID: 37581324 DOI: 10.2174/1389201024666230811104740] [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: 12/28/2022] [Revised: 06/12/2023] [Accepted: 06/26/2023] [Indexed: 08/16/2023]
Abstract
OBJECTIVES Obesity is regarded as the main cause of metabolic diseases and a core factor for all-cause mortality in the general population, notably from cardiovascular disease. The majority of people with type 2 diabetes have obesity and insulin resistance. Some evidence indicates that an individual with obesity is approximately 10 times more likely to develop type 2 diabetes than someone with moderate body weight. One of the most significant therapeutic herbs, Salvia officinalis (Lamiaceae) (SAGE), possesses potent medicinal importance. The aim of this article was to evaluate the anti-diabetic and antiobesity activity of SAGEAE against HFD-induced obesity in rats. METHODS Thirty adult albino rats were randomly divided into five equal groups: control, High-fat Diet (HFD) administrated rats, HFD + Salvia officinalis Aqueous Extract (SAGEAE) (150 mg/kg.bw.), HFD + SAGEAE (300 mg/kg.bw.) and HFD + metformin (500 mg/kg.bw.). Body weight, plasma biochemical parameters, oxidative stress, inflammatory indicators, hepatic Phosphoenolpyruvate Carboxykinase 1 (PCK1), Glucokinase (GK), brain Leptin Receptor (LepRb), Glucose Transporter-4 (GLUT4), Sirtuin 1 (SIRT1) and mRNA33-5P gene signalling mRNA levels were all assessed after 8 weeks. A histological examination of the liver was also performed to check for lipid accumulation. RESULTS The administration of HFD resulted in increased body weight, glucose, insulin, leptin, Total Cholesterol (TC), Triglycerides (TG), Thiobarbaturic Acid Reactive Substances (TBARS), Monocyte Chemoattractant Protein-1 (MCP1), Interleukine-6 (IL-6) and tumor necrosis factor-α (TNF- α) as well as hepatic PCK1, brain LepRb and adipose tissue mRNA33-5P gene expression. However, our findings revealed a significant reduction in adiponectin, High-density Lipoproteincholesterol (HDL-C), reduced glutathione (GSH) and Superoxide Dismutase (SOD) levels as well as the expression of hepatic GK and adipose tissue SIRT1 and GLUT4 genes. Also, administration of SAGEAE significantly normalized body weight, glucose, insulin, leptin, adiponectin, TC, TG, HDL-C, TBARs, SOD, IL-6, MCP-1 and TNF-α in plasma and liver tissue of HFD-treated rats. On the other hand, PCK1, GK, LepRb, SIRT1, GLUT4 and mRNA33-5P gene expression was enhanced in obese rats when administrated with SAGEAE. Histological and US studies support the biochemical, PCR and electrophoretic results. CONCLUSION The findings imply that SAGEAE could be used as a new pharmaceutical formula in the treatment of obesity.
Collapse
Affiliation(s)
- Diana A Alsherif
- Department of Radiology and Medical Imaging, Faculty of Applied Health Science Technology, October 6th University, October 6th City, Egypt
| | - Mohammed A Hussein
- Department of Biotechnology, Faculty of Applied Health Science Technology, October 6th University, October 6th City, Egypt
| | - Suzan S Abuelkasem
- Department of Biochemistry, Faculty of Applied Health Science Technology, October 6th University, October 6th City, Egypt
| |
Collapse
|
2
|
Abdulazeez I, Ismail IS, Mohd Faudzi SM, Christianus A, Chong SG. Study on the acute toxicity of sodium taurocholate via zebrafish mortality, behavioral response, and NMR-metabolomics analysis. Drug Chem Toxicol 2024; 47:115-130. [PMID: 37548163 DOI: 10.1080/01480545.2023.2242005] [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: 03/06/2023] [Accepted: 07/20/2023] [Indexed: 08/08/2023]
Abstract
Sodium taurocholate (NaT) is a hydrophobic bile salt that exhibits varying toxicity and antimicrobial activity. The accumulation of BSs during their entero-hepatic cycle causes cytotoxicity in the liver and intestine and could also alter the intestinal microbiome leading to various diseases. In this research, the acute toxicity of sodium taurocholate in different concentrations (3000 mg/L, 1500 mg/L, 750 mg/L, 375 mg/L, and 0 mg/L) was investigated on four months old zebrafish by immersion in water for 96 h. The results were determined based on the fish mortality, behavioral response, and NMR metabolomics analysis which revealed LC50 of 1760.32 mg/L and 1050.42 mg/L after 72 and 96 h treatment, respectively. However, the non-lethal NaT concentrations of 750 mg/L and 375 mg/L at 96 h exposure significantly (p ≤ 0.05) decreased the total distance traveled and the activity duration, also caused surface respiration on the zebrafish. Orthogonal Projections to Latent Structures Discriminant Analysis (OPLS-DA) revealed that the metabolome of the fish treated with 750 mg/L was discriminated from that of the control by PC1. Major significantly downregulated metabolites by NaT-induction include valine, isoleucine, 2-hydroxyvalerate, glycine, glycerol, choline, glucose, pyruvate, anserine, threonine, carnitine and homoserine. On the contrary, taurine, creatine, lactate, acetate and 3-hydroxybutyrate were upregulated suggesting cellular consumption of lipids, glucose and amino acids for adenosine triphosphate (ATP) generation during immune and inflammatory response. whereby these metabolites were released in the process. In conclusion, the research revealed the toxic effect of NaT and its potential to trigger changes in zebrafish metabolism.
Collapse
Affiliation(s)
- Isah Abdulazeez
- Department of Chemistry, Faculty of Science, Universiti Putra Malaysia, UPM Serdang Selangor, Malaysia
| | - Intan Safinar Ismail
- Department of Chemistry, Faculty of Science, Universiti Putra Malaysia, UPM Serdang Selangor, Malaysia
- Natural Medicines and Product Research Laboratory (NaturMeds), Institute of Bioscience (IBS), Universiti Putra Malaysia, UPM Serdang Selangor, Malaysia
| | - Siti Munirah Mohd Faudzi
- Department of Chemistry, Faculty of Science, Universiti Putra Malaysia, UPM Serdang Selangor, Malaysia
| | - Annie Christianus
- Department of Aquaculture, Faculty of Agricultural Sciences, Universiti Putra Malaysia, UPM Serdang Selangor, Malaysia
| | - Seok-Giok Chong
- Natural Medicines and Product Research Laboratory (NaturMeds), Institute of Bioscience (IBS), Universiti Putra Malaysia, UPM Serdang Selangor, Malaysia
| |
Collapse
|
3
|
Yong GJM, Porsche CE, Sitarik AR, Fujimura KE, McCauley K, Nguyen DT, Levin AM, Woodcroft KJ, Ownby DR, Rundle AG, Johnson CC, Cassidy-Bushrow A, Lynch SV. Precocious infant fecal microbiome promotes enterocyte barrier dysfuction, altered neuroendocrine signaling and associates with increased childhood obesity risk. Gut Microbes 2024; 16:2290661. [PMID: 38117587 PMCID: PMC10761186 DOI: 10.1080/19490976.2023.2290661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 11/29/2023] [Indexed: 12/22/2023] Open
Abstract
Early life gut microbiome composition has been correlated with childhood obesity, though microbial functional contributions to disease origins remain unclear. Here, using an infant birth cohort (n = 349) we identify a distinct fecal microbiota composition in 1-month-old infants with the lowest rate of exclusive breastfeeding, that relates with higher relative risk for obesity and overweight phenotypes at two years. Higher-risk infant fecal microbiomes exhibited accelerated taxonomic and functional maturation and broad-ranging metabolic reprogramming, including reduced concentrations of neuro-endocrine signals. In vitro, exposure of enterocytes to fecal extracts from higher-risk infants led to upregulation of genes associated with obesity and with expansion of nutrient sensing enteroendocrine progenitor cells. Fecal extracts from higher-risk infants also promoted enterocyte barrier dysfunction. These data implicate dysregulation of infant microbiome functional development, and more specifically promotion of enteroendocrine signaling and epithelial barrier impairment in the early-life developmental origins of childhood obesity.
Collapse
Affiliation(s)
- Germaine J. M. Yong
- Division of Gastroenterology, Department of Medicine, University of California, San Francisco, CA, USA
- Asian Microbiome Library Pte Ltd, Singapore and Singapore Institute of Food and Biotechnology Innovation, Singapore, Singapore
| | - Cara E. Porsche
- Division of Gastroenterology, Department of Medicine, University of California, San Francisco, CA, USA
| | - Alexandra R. Sitarik
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI, USA
| | - Kei E. Fujimura
- Division of Gastroenterology, Department of Medicine, University of California, San Francisco, CA, USA
- Genetic Disease Laboratory, California Department of Public Health, San Francisco, CA, USA
| | - Kathryn McCauley
- Division of Gastroenterology, Department of Medicine, University of California, San Francisco, CA, USA
| | - Dat T. Nguyen
- Division of Gastroenterology, Department of Medicine, University of California, San Francisco, CA, USA
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Albert M. Levin
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI, USA
| | | | - Dennis R. Ownby
- Division of Allergy and Clinical Immunology, Department of Pediatrics, Augusta University, Augusta, GA, USA
| | - Andrew G. Rundle
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Christine C. Johnson
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI, USA
| | | | - Susan V. Lynch
- Division of Gastroenterology, Department of Medicine, University of California, San Francisco, CA, USA
| |
Collapse
|
4
|
Wang Y, Zhou W, Liu D, Zhang Z, Xu Y, Wan X, Yu H, Yan S. Exploration of the molecular mechanism of insulin resistance in adipose tissue of patients with type 2 diabetes mellitus through a bioinformatic analysis. Minerva Endocrinol (Torino) 2023; 48:440-446. [PMID: 37534872 DOI: 10.23736/s2724-6507.22.03771-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/04/2023]
Abstract
BACKGROUND We aimed to determine the cis-expression Quantitative Trait Loci (cis-eQTL) and trans-eQTL of differentially expressed genes (DEGs) in insulin resistance (IR) related pathways. METHODS The expression profile data for insulin sensitivity (IS) and IR in the adipose tissue of patients with type 2 diabetes mellitus (T2DM) were acquired from the Gene Expression Omnibus databases. Then, the Gene set enrichment analysis (GSEA) and Gene set variation analysis (GSVA) methods were performed to identify the significant enrichment of potential Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways between IS and IR groups, and the Wilcoxon rank sum test was carried out to identify the DEGs related to KEGG pathways. Finally, the cis-eQTLs and trans-eQTLs that can affect the expression of DEGs were screened from the eQTLGen database. RESULTS The GSEA and GSVA analysis indicated that the mTOR signaling pathway, insulin signaling pathway and T2DM had a strong correlation with the pathological process of T2DM. Furthermore, six genes (ACACA, GYS2, PCK1, PRKAR1A, SLC2A4, and VEGFA) were found to be significantly differentially expressed in IR-related pathways. Finally, we have identified a total of 1073 cis-eQTLs and 24 trans-eQTLs. CONCLUSIONS We screened out six genes that were significantly differentially expressed in IR-related pathways, including ACACA, GYS2, PCK1, PRKAR1A, SLC2A4, and VEGFA. Moreover, we discovered that these six genes were affected by 1073 cis-eQTLs and 24 trans-eQTLs.
Collapse
Affiliation(s)
- Yujing Wang
- Department of Endocrinology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Weiyu Zhou
- Department of Endocrinology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Dana Liu
- Department of Endocrinology, The First Hospital, Harbin, China
| | - Zhiying Zhang
- Department of Endocrinology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yuanxin Xu
- Department of Endocrinology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaojing Wan
- Department of Endocrinology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Haiqiao Yu
- Department of Endocrinology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shuang Yan
- Department of Endocrinology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China -
| |
Collapse
|
5
|
Hershberger CE, Raj R, Mariam A, Aykun N, Allende DS, Brown M, Aucejo F, Rotroff DM. Characterization of Salivary and Plasma Metabolites as Biomarkers for HCC: A Pilot Study. Cancers (Basel) 2023; 15:4527. [PMID: 37760495 PMCID: PMC10527521 DOI: 10.3390/cancers15184527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/24/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023] Open
Abstract
(1) Background: The incidence of hepatocellular carcinoma (HCC) is rising, and current screening methods lack sensitivity. This study aimed to identify distinct and overlapping metabolites in saliva and plasma that are significantly associated with HCC. (2) Methods: Saliva samples were collected from 42 individuals (HCC = 16, cirrhosis = 12, healthy = 14), with plasma samples from 22 (HCC = 14, cirrhosis = 2, healthy = 6). We performed untargeted mass spectrometry on blood and plasma, tested metabolites for associations with HCC or cirrhosis using a logistic regression, and identified enriched pathways with Metaboanalyst. Pearson's correlation was employed to test for correlations between salivary and plasma metabolites. (3) Results: Six salivary metabolites (1-hexadecanol, isooctanol, malonic acid, N-acetyl-valine, octadecanol, and succinic acid) and ten plasma metabolites (glycine, 3-(4-hydroxyphenyl)propionic acid, aconitic acid, isocitric acid, tagatose, cellobiose, fucose, glyceric acid, isocitric acid, isothreonic acid, and phenylacetic acid) were associated with HCC. Malonic acid was correlated between the paired saliva and plasma samples. Pathway analysis highlighted deregulation of the 'The Citric Acid Cycle' in both biospecimens. (4) Conclusions: Our study suggests that salivary and plasma metabolites may serve as independent sources for HCC detection. Despite the lack of correlation between individual metabolites, they converge on 'The Citric Acid Cycle' pathway, implicated in HCC pathogenesis.
Collapse
Affiliation(s)
- Courtney E Hershberger
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Quantitative Metabolic Research, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Roma Raj
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Quantitative Metabolic Research, Cleveland Clinic, Cleveland, OH 44195, USA
- Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Arshiya Mariam
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Quantitative Metabolic Research, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Nihal Aykun
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Quantitative Metabolic Research, Cleveland Clinic, Cleveland, OH 44195, USA
- Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Daniela S Allende
- Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Mark Brown
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Microbiome and Human Health, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Federico Aucejo
- Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Daniel M Rotroff
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Quantitative Metabolic Research, Cleveland Clinic, Cleveland, OH 44195, USA
- Endocrinology and Metabolism Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| |
Collapse
|
6
|
Wiseman RW, Brown CM, Beck TW, Brault JJ, Reinoso TR, Shi Y, Chase PB. Creatine Kinase Equilibration and ΔG ATP over an Extended Range of Physiological Conditions: Implications for Cellular Energetics, Signaling, and Muscle Performance. Int J Mol Sci 2023; 24:13244. [PMID: 37686064 PMCID: PMC10487889 DOI: 10.3390/ijms241713244] [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: 07/31/2023] [Revised: 08/22/2023] [Accepted: 08/24/2023] [Indexed: 09/10/2023] Open
Abstract
In this report, we establish a straightforward method for estimating the equilibrium constant for the creatine kinase reaction (CK Keq″) over wide but physiologically and experimentally relevant ranges of pH, Mg2+ and temperature. Our empirical formula for CK Keq″ is based on experimental measurements. It can be used to estimate [ADP] when [ADP] is below the resolution of experimental measurements, a typical situation because [ADP] is on the order of micromolar concentrations in living cells and may be much lower in many in vitro experiments. Accurate prediction of [ADP] is essential for in vivo studies of cellular energetics and metabolism and for in vitro studies of ATP-dependent enzyme function under near-physiological conditions. With [ADP], we were able to obtain improved estimates of ΔGATP, necessitating the reinvestigation of previously reported ADP- and ΔGATP-dependent processes. Application to actomyosin force generation in muscle provides support for the hypothesis that, when [Pi] varies and pH is not altered, the maximum Ca2+-activated isometric force depends on ΔGATP in both living and permeabilized muscle preparations. Further analysis of the pH studies introduces a novel hypothesis around the role of submicromolar ADP in force generation.
Collapse
Affiliation(s)
- Robert Woodbury Wiseman
- Departments of Physiology and Radiology, Michigan State University, East Lansing, MI 48824, USA;
| | - Caleb Micah Brown
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Thomas Wesley Beck
- Department of Radiology, University of Washington, Seattle, WA 98195, USA
| | - Jeffrey John Brault
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA;
| | - Tyler Robert Reinoso
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA
| | - Yun Shi
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA
| | - Prescott Bryant Chase
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA
| |
Collapse
|
7
|
Lloyd EM, Pinniger GJ, Murphy RM, Grounds MD. Slow or fast: Implications of myofibre type and associated differences for manifestation of neuromuscular disorders. Acta Physiol (Oxf) 2023; 238:e14012. [PMID: 37306196 DOI: 10.1111/apha.14012] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 05/30/2023] [Accepted: 06/06/2023] [Indexed: 06/13/2023]
Abstract
Many neuromuscular disorders can have a differential impact on a specific myofibre type, forming the central premise of this review. The many different skeletal muscles in mammals contain a spectrum of slow- to fast-twitch myofibres with varying levels of protein isoforms that determine their distinctive contractile, metabolic, and other properties. The variations in functional properties across the range of classic 'slow' to 'fast' myofibres are outlined, combined with exemplars of the predominantly slow-twitch soleus and fast-twitch extensor digitorum longus muscles, species comparisons, and techniques used to study these properties. Other intrinsic and extrinsic differences are discussed in the context of slow and fast myofibres. These include inherent susceptibility to damage, myonecrosis, and regeneration, plus extrinsic nerves, extracellular matrix, and vasculature, examined in the context of growth, ageing, metabolic syndrome, and sexual dimorphism. These many differences emphasise the importance of carefully considering the influence of myofibre-type composition on manifestation of various neuromuscular disorders across the lifespan for both sexes. Equally, understanding the different responses of slow and fast myofibres due to intrinsic and extrinsic factors can provide deep insight into the precise molecular mechanisms that initiate and exacerbate various neuromuscular disorders. This focus on the influence of different myofibre types is of fundamental importance to enhance translation for clinical management and therapies for many skeletal muscle disorders.
Collapse
Affiliation(s)
- Erin M Lloyd
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, The University of Western Australia, Perth, Western Australia, Australia
- Curtin Health Innovation Research Institute, Curtin Medical School, Curtin University, Bentley, Western Australia, Australia
| | - Gavin J Pinniger
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Robyn M Murphy
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, Victoria, Australia
| | - Miranda D Grounds
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, The University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
8
|
Paoli A, Bianco A, Moro T, Mota JF, Coelho-Ravagnani CF. The Effects of Ketogenic Diet on Insulin Sensitivity and Weight Loss, Which Came First: The Chicken or the Egg? Nutrients 2023; 15:3120. [PMID: 37513538 PMCID: PMC10385501 DOI: 10.3390/nu15143120] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 07/05/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023] Open
Abstract
The ketogenic diet (KD) is, nowadays, considered an interesting nutritional approach for weight loss and improvement in insulin resistance. Nevertheless, most of the studies available in the literature do not allow a clear distinction between its effects on insulin sensitivity per se, and the effects of weight loss induced by KDs on insulin sensitivity. In this review, we discuss the scientific evidence on the direct and weight loss mediated effects of KDs on glycemic status in humans, describing the KD's biochemical background and the underlying mechanisms.
Collapse
Affiliation(s)
- Antonio Paoli
- Department of Biomedical Sciences, University of Padua, 35127 Padua, Italy
- Research Center for High Performance Sport, UCAM, Catholic University of Murcia, 30107 Murcia, Spain
| | - Antonino Bianco
- Sport and Exercise Sciences Research Unit, University of Palermo, 90144 Palermo, Italy
| | - Tatiana Moro
- Department of Biomedical Sciences, University of Padua, 35127 Padua, Italy
| | - Joao Felipe Mota
- School of Nutrition, Federal University of Goiás, Goiânia 74605-080, Brazil
- APC Microbiome Ireland, Department of Medicine, School of Microbiology, University College Cork, T12 YT20 Cork, Ireland
| | - Christianne F Coelho-Ravagnani
- Research in Exercise and Nutrition in Health and Sports Performance-PENSARE, Post-Graduate Program in Movement Sciences, Institute of Health (INISA), Federal University of Mato Grosso do Sul, Campo Grande 79070-900, Brazil
| |
Collapse
|
9
|
Dong Q, Sidra S, Gieger C, Wang-Sattler R, Rathmann W, Prehn C, Adamski J, Koenig W, Peters A, Grallert H, Sharma S. Metabolic Signatures Elucidate the Effect of Body Mass Index on Type 2 Diabetes. Metabolites 2023; 13:metabo13020227. [PMID: 36837846 PMCID: PMC9965667 DOI: 10.3390/metabo13020227] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/26/2023] [Accepted: 01/31/2023] [Indexed: 02/09/2023] Open
Abstract
Obesity plays an important role in the development of insulin resistance and diabetes, but the molecular mechanism that links obesity and diabetes is still not completely understood. Here, we used 146 targeted metabolomic profiles from the German KORA FF4 cohort consisting of 1715 participants and associated them with obesity and type 2 diabetes. In the basic model, 83 and 51 metabolites were significantly associated with body mass index (BMI) and T2D, respectively. Those metabolites are branched-chain amino acids, acylcarnitines, lysophospholipids, or phosphatidylcholines. In the full model, 42 and 3 metabolites were significantly associated with BMI and T2D, respectively, and replicate findings in the previous studies. Sobel mediation testing suggests that the effect of BMI on T2D might be mediated via lipids such as sphingomyelin (SM) C16:1, SM C18:1 and diacylphosphatidylcholine (PC aa) C38:3. Moreover, mendelian randomization suggests a causal relationship that BMI causes the change of SM C16:1 and PC aa C38:3, and the change of SM C16:1, SM C18:1, and PC aa C38:3 contribute to T2D incident. Biological pathway analysis in combination with genetics and mice experiments indicate that downregulation of sphingolipid or upregulation of phosphatidylcholine metabolism is a causal factor in early-stage T2D pathophysiology. Our findings indicate that metabolites like SM C16:1, SM C18:1, and PC aa C38:3 mediate the effect of BMI on T2D and elucidate their role in obesity related T2D pathologies.
Collapse
Affiliation(s)
- Qiuling Dong
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Faculty of Medicine, Ludwig-Maximilians-University München, 81377 Munich, Germany
| | - Sidra Sidra
- Institute for Medical Information Processing, Biometry and Epidemiology (IBE), Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Christian Gieger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- German Center for Diabetes Research (DZD), 85764 München-Neuherberg, Germany
| | - Rui Wang-Sattler
- Institute of Translational Genomics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Wolfgang Rathmann
- Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Cornelia Prehn
- Metabolomics and Proteomics Core Facility, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Jerzy Adamski
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 8 Medical Drive, Singapore 117597, Singapore
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia
| | - Wolfgang Koenig
- German Research Center for Cardiovascular Disease (DZHK), Partner site Munich Heart Alliance, 81377 Munich, Germany
- Deutsches Herzzentrum München, Technische Universität München, 81377 Munich, Germany
- Institute of Epidemiology and Medical Biometry, University of Ulm, 89069 Ulm, Germany
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- German Center for Diabetes Research (DZD), 85764 München-Neuherberg, Germany
- Chair of Epidemiology, Faculty of Medicine, Ludwig-Maximilians-University München, 81377 Munich, Germany
| | - Harald Grallert
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- German Center for Diabetes Research (DZD), 85764 München-Neuherberg, Germany
- Correspondence: (H.G.); (S.S.)
| | - Sapna Sharma
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Chair of Food Chemistry and Molecular Sensory Science, Technical University of Munich, 85354 Freising-Weihenstephan, Germany
- Correspondence: (H.G.); (S.S.)
| |
Collapse
|
10
|
Nomiyama K, Yamamoto Y, Eguchi A, Nishikawa H, Mizukawa H, Yokoyama N, Ichii O, Takiguchi M, Nakayama SMM, Ikenaka Y, Ishizuka M. Health impact assessment of pet cats caused by organohalogen contaminants by serum metabolomics and thyroid hormone analysis. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 842:156490. [PMID: 35667425 DOI: 10.1016/j.scitotenv.2022.156490] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/24/2022] [Accepted: 06/01/2022] [Indexed: 06/15/2023]
Abstract
Companion animals are in close contact with the human surroundings, and there is growing concern about the effects of harmful substances on the health of pet cats. In this study, we investigated the potential health effects of organohalogen compounds (OHCs) on thyroid hormone (TH) homeostasis and metabolomics in Japanese pet cats. There was a significant negative correlation between concentrations of several contaminants, such as polychlorinated biphenyls (PCBs), polybrominated diphenyl ethers (PBDEs), hydroxylated PCBs (OH-PCBs), hydroxylated PBDEs (OH-PBDEs), and THs in cat serum samples. These results suggested that exposure to OHCs causes a decrease in serum TH levels in pet cats. In this metabolomics study, each exposure level of parent compounds (PCBs and PBDEs) and their hydroxylated compounds (OH-PCBs and OH-PBDEs) were associated with their own unique primary metabolic pathways, suggesting that parent and phenolic compounds exhibit different mechanisms of action and biological effects. PCBs were associated with many metabolic pathways, including glutathione and purine metabolism, and the effects were replicated in in-vivo cat PCB administration studies. These results demonstrated that OHC exposure causes chronic oxidative stress in pet cats. PBDEs were positively associated with alanine, aspartate, and glutamate metabolism. Due to the chronic exposure of cats to mixtures of these contaminants, the combination of their respective metabolic pathways may have a synergistic effect.
Collapse
Affiliation(s)
- Kei Nomiyama
- Center for Marine Environmental Studies (CMES), Ehime University, Bunkyo-cho 2-5, Matsuyama, Ehime 790-8577, Japan.
| | - Yasuo Yamamoto
- Center for Marine Environmental Studies (CMES), Ehime University, Bunkyo-cho 2-5, Matsuyama, Ehime 790-8577, Japan
| | - Akifumi Eguchi
- Center for Preventive Medical Sciences, Chiba University, Inage-ku Yayoi-cho 1-33, Chiba-city 263-8522, Japan
| | - Hiroyuki Nishikawa
- Center for Marine Environmental Studies (CMES), Ehime University, Bunkyo-cho 2-5, Matsuyama, Ehime 790-8577, Japan
| | - Hazuki Mizukawa
- Department of Science and Technology for Biological Resources and Environment, Graduate School of Agriculture, Ehime University, Tarumi 3-5-7, Matsuyama, Ehime 790-8566, Japan
| | - Nozomu Yokoyama
- Veterinary Teaching Hospital, Graduate School of Veterinary Medicine, Hokkaido University, N18 W9, Sapporo, Hokkaido 060-0818, Japan
| | - Osamu Ichii
- Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo, Hokkaido 060-0818, Japan
| | - Mitsuyoshi Takiguchi
- Laboratory of Veterinary Internal Medicine, Graduate School of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo, Hokkaido 060-0818, Japan
| | - Shouta M M Nakayama
- Department of Environmental Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo, Hokkaido 060-0818, Japan
| | - Yoshinori Ikenaka
- Veterinary Teaching Hospital, Graduate School of Veterinary Medicine, Hokkaido University, N18 W9, Sapporo, Hokkaido 060-0818, Japan; Department of Environmental Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo, Hokkaido 060-0818, Japan; Water Research Group, Unit for Environmental Sciences and Management, North-West University, Potchefstroom Campus, X6001, Potchefstroom 2520, South Africa; One Health Research Center, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo 060-0818, Japan
| | - Mayumi Ishizuka
- Department of Environmental Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo, Hokkaido 060-0818, Japan
| |
Collapse
|
11
|
Hernández-Saavedra D, Markunas C, Takahashi H, Baer LA, Harris JE, Hirshman MF, Ilkayeva O, Newgard CB, Stanford KI, Goodyear LJ. Maternal Exercise and Paternal Exercise Induce Distinct Metabolite Signatures in Offspring Tissues. Diabetes 2022; 71:2094-2105. [PMID: 35838316 PMCID: PMC9501651 DOI: 10.2337/db22-0341] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 07/12/2022] [Indexed: 01/19/2023]
Abstract
That maternal and paternal exercise improve the metabolic health of adult offspring is well established. Tissue and serum metabolites play a fundamental role in the health of an organism, but how parental exercise affects offspring tissue and serum metabolites has not yet been investigated. Here, male and female breeders were fed a high-fat diet and housed with or without running wheels before breeding (males) and before and during gestation (females). Offspring were sedentary and chow fed, with parents as follows: sedentary (Sed), maternal exercise (MatEx), paternal exercise (PatEx), or maternal+paternal exercise (Mat+PatEx). Adult offspring from all parental exercise groups had similar improvement in glucose tolerance and hepatic glucose production. Targeted metabolomics was performed in offspring serum, liver, and triceps muscle. Offspring from MatEx, PatEx, and Mat+PatEx each had a unique tissue metabolite signature, but Mat+PatEx offspring had an additive phenotype relative to MatEx or PatEx alone in a subset of liver and muscle metabolites. Tissue metabolites consistently indicated that the metabolites altered with parental exercise contribute to enhanced fatty acid oxidation. These data identify distinct tissue-specific adaptations and mechanisms for parental exercise-induced improvement in offspring metabolic health. Further mining of this data set could aid the development of novel therapeutic targets to combat metabolic diseases.
Collapse
Affiliation(s)
- Diego Hernández-Saavedra
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH
- Department of Kinesiology and Community Health, University of Illinois Urbana-Champaign, Urbana, IL
| | - Christina Markunas
- Departments of Pharmacology and Cancer Biology and Medicine, Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute, Durham, NC
| | - Hirokazu Takahashi
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - Lisa A. Baer
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Johan E. Harris
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Michael F. Hirshman
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - Olga Ilkayeva
- Departments of Pharmacology and Cancer Biology and Medicine, Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute, Durham, NC
| | - Christopher B. Newgard
- Departments of Pharmacology and Cancer Biology and Medicine, Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute, Durham, NC
| | - Kristin I. Stanford
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Laurie J. Goodyear
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| |
Collapse
|
12
|
Lee MC, Hsu YJ, Sung HC, Wen YT, Wei L, Huang CC. Low Aerobic Capacity Accelerates Lipid Accumulation and Metabolic Abnormalities Caused by High-Fat Diet-Induced Obesity in Postpartum Mice. Nutrients 2022; 14:nu14183746. [PMID: 36145123 PMCID: PMC9502809 DOI: 10.3390/nu14183746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/02/2022] [Accepted: 09/06/2022] [Indexed: 11/26/2022] Open
Abstract
Women during pregnancy and postpartum show high rates of obesity and metabolic diseases, especially women with excessive caloric intake. In the past, it was proved that individuals with high intrinsic aerobic exercise capacities showed higher lipid metabolism and lower fat production than those with low intrinsic aerobic exercise capacities. The purpose of this study was to determine whether mice with the low-fitness phenotype (LAEC) were more likely to develop metabolic abnormalities and obesity under dietary induction after delivery, and if mice with a high-fitness phenotype (HAEC) had a protective mechanism. After parturition and weaning, postpartum Institute of Cancer Research (ICR) mice received dietary induction for 12 weeks and were divided into four groups (n = 8 per group): high-exercise capacity postpartum mice with a normal chow diet (HAEC-ND); high-exercise capacity postpartum mice with a high-fat diet (HAEC-HFD); low-exercise capacity postpartum mice with a normal chow diet (LAEC-ND); and low-exercise capacity postpartum mice with a high-fat diet (LAEC-HFD). Obesity caused by a high-fat diet led to decreased exercise performance (p < 0.05). Although there were significant differences in body posture under congenital conditions, the LAEC mice gained more weight and body fat after high-fat-diet intake (p < 0.05). Compared with HAEC-HFD, LAEC-HFD significantly increased blood lipids, such as total cholesterol (TC), triacylglycerol (TG), low-density lipoprotein (LDL) and other parameters (p < 0.05), and the content of TG in the liver, as well as inducing poor glucose tolerance (p < 0.05). In addition, after HFD intake, excessive energy significantly increased glycogen storage (p < 0.05), but the LAEC mice showed significantly lower muscle glycogen storage (p < 0.05). In conclusion, although we observed significant differences in intrinsic exercise capacity, and body posture and metabolic ability were also different, high-fat-diet intake caused weight gain and a risk of metabolic disorders, especially in postpartum low-fitness mice. However, HAEC mice still showed better lipid metabolism and protection mechanisms. Conversely, LAEC mice might accumulate more fat and develop metabolic diseases compared with their normal rodent chow diet (ND) control counterparts.
Collapse
Affiliation(s)
- Mon-Chien Lee
- Graduate Institute of Sports Science, National Taiwan Sport University, Taoyuan 333325, Taiwan
| | - Yi-Ju Hsu
- Graduate Institute of Sports Science, National Taiwan Sport University, Taoyuan 333325, Taiwan
| | - Hsin-Ching Sung
- Department of Anatomy, College of Medicine, Chang Gung University, Taoyuan 333323, Taiwan
- Aesthetic Medical Center, Department of Dermatology, Chang Gung Memorial Hospital, Taoyuan 333423, Taiwan
| | - Ya-Ting Wen
- Division of Neurosurgery, Department of Surgery, Wan Fang Hospital, Taipei Medical University, Taipei 116081, Taiwan
| | - Li Wei
- Division of Neurosurgery, Department of Surgery, Wan Fang Hospital, Taipei Medical University, Taipei 116081, Taiwan
- Graduate Institute of Injury Prevention and Control, Taipei Medical University, Taipei 110301, Taiwan
- Taipei Neuroscience Institute, Taipei Medical University, Taipei 110301, Taiwan
- Correspondence: (L.W.); (C.-C.H.); Tel.: +886-2-27361661 (ext. 6579) (L.W.); +886-3-328-3201 (ext. 2619) (C.-C.H.)
| | - Chi-Chang Huang
- Graduate Institute of Sports Science, National Taiwan Sport University, Taoyuan 333325, Taiwan
- Correspondence: (L.W.); (C.-C.H.); Tel.: +886-2-27361661 (ext. 6579) (L.W.); +886-3-328-3201 (ext. 2619) (C.-C.H.)
| |
Collapse
|
13
|
Zhang J, Wang W, Yang G, Ha J, Tan X, Shang X, Zhu Z, Han X, Liu Z, Zhang L, He M, Luo L. Body mass index is not associated with early onset cataract in the 45 and Up cohort study. ANNALS OF TRANSLATIONAL MEDICINE 2022; 9:1640. [PMID: 34988149 PMCID: PMC8667097 DOI: 10.21037/atm-21-2775] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 10/12/2021] [Indexed: 12/03/2022]
Abstract
Background Body mass index (BMI) has been reported to be associated with age-related cataract, whereas its impact on early onset cataract (EOC) remains unknown. Methods A total of 73,007 individuals aged 45–55 years who had no previous cataract surgeries at baseline were enrolled from the population-based 45 and Up Study. BMI was calculated based on self-reported height and weight from the baseline questionnaire. Data on cataract surgeries were obtained from the Medicare Benefits Schedule database. EOC was defined as cataract surgically treated prior to 65 years of age. A Cox proportional hazards regression was used to assess the association between BMI and the incidence of EOC during the follow-up. Results A total of 1,764 participants underwent cataract surgery over 643,717 person-years of follow-up. No significant association was observed between BMI and EOC (P for trend 0.35). Among participants who drank 5 to 7 alcoholic drinks per week, a 73% and 27% reduction in the risk of EOC was observed in participants with a BMI of 18.5–19.99 and 25.0–27.49 kg/m2, respectively, compared to those with a BMI of 20.0–22.49 kg/m2. Conclusions No association was identified between BMI and the incidence of EOC. Moderate alcohol intake may be protective against EOC.
Collapse
Affiliation(s)
- Jiaqing Zhang
- State Key Laboratory of Ophthalmology, National Clinical Research Center, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Wei Wang
- State Key Laboratory of Ophthalmology, National Clinical Research Center, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Guangyao Yang
- Department of Ophthalmology, Municipal Hospital of Chifeng, Inner Mongolia Autonomous Region, Chifeng, China
| | - Jason Ha
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia
| | - Xuhua Tan
- State Key Laboratory of Ophthalmology, National Clinical Research Center, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Xianwen Shang
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia
| | - Zhuoting Zhu
- Department of Ophthalmology, Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, Guangzhou, China
| | - Xiaotong Han
- State Key Laboratory of Ophthalmology, National Clinical Research Center, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Zhenzhen Liu
- State Key Laboratory of Ophthalmology, National Clinical Research Center, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Lei Zhang
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia.,Artificial Intelligence and Modelling in Epidemiology Program, Melbourne Sexual Health Centre, Alfred Health, Melbourne, Australia.,Central Clinical School, Faculty of Medicine, Monash University, Melbourne, Australia.,Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Mingguang He
- State Key Laboratory of Ophthalmology, National Clinical Research Center, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China.,Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia.,Ophthalmology, Department of Surgery, University of Melbourne, Melbourne, Australia
| | - Lixia Luo
- State Key Laboratory of Ophthalmology, National Clinical Research Center, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
14
|
He Y, Yang X, Li M, Zhang A, Sun N. Vitamin D supplementation and energy and metabolic homoeostasis in obese and overweight subjects: a protocol for a systematic review. BMJ Open 2021; 11:e051230. [PMID: 34593501 PMCID: PMC8487182 DOI: 10.1136/bmjopen-2021-051230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
INTRODUCTION Obesity and vitamin D deficiency are major public health problems. According to the pathophysiological mechanism of obesity as well as the bidirectional relationship between obesity and vitamin D metabolism and storage, vitamin D supplementation in obese and overweight subjects could have beneficial effects on the energy and metabolic homoeostasis. This review will assess the efficacy of vitamin D supplementation on the energy and metabolic homoeostasis in overweight and obese subjects. METHODS AND ANALYSIS In accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses Protocols, we retrieved the relevant literature from the following electronic bibliographic databases: MEDLINE/PubMed, EMBASE and the Cochrane Central Register of Controlled Trials, from inception to June 2021. A manual search of the reference lists of all the relevant research articles will be performed to identify additional studies. We will include randomised controlled trials (RCTs) published in English that examine the effects of vitamin D supplementation on energy and metabolic homoeostasis in overweight and obese subjects. RCTs with multiple vitamin D groups will also be included. Two reviewers will independently complete the article selection, data extraction and rating. The bias tool from the Cochrane Handbook for Systematic Reviews of Interventions was used to assess the methodological quality of the included studies. A narrative or quantitative synthesis will be performed based on the available data. The planned start and end dates for the study were 1 February 2021 and 1 March 2022. ETHICS AND DISSEMINATION Ethical approval will not be required for this review. The results of this review will be disseminated in a peer-reviewed journal. PROSPERO REGISTRATION NUMBER CRD42021228981.
Collapse
Affiliation(s)
- Yu He
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiao Yang
- Department of Rehabilitation, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Min Li
- Department of Rehabilitation, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Anren Zhang
- Department of Rehabilitation, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Nianyi Sun
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
- Department of Rehabilitation, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
15
|
Ahmed HMS, Mohamed SG, Ibrahim WS, Rezk AM, Mahmoud AAA, Mahmoud MF, Ibrahim IAAEH. Acute and chronic metabolic effects of carvedilol in high-fructose, high-fat diet-fed mice: implication of β-arrestin2 pathway. Can J Physiol Pharmacol 2021; 100:68-77. [PMID: 34570983 DOI: 10.1139/cjpp-2021-0299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We aimed to investigate the acute and chronic effects of carvedilol on insulin resistance in high-fructose, high-fat diet (HFrHFD) - fed mice and the implication of the β-arrestin2 pathway. The acute effect of carvedilol (10 mg/kg, i.p.) on glucose tolerance and hepatic lipid signaling in normal and insulin resistant mice was investigated. Then, the chronic effect of carvedilol on insulin resistance and dyslipidemia in HFrHFD-fed mice was examined. Changes in β-arrestin2 and its downstream signals in liver, skeletal muscle, and adipose tissue were measured. This involved measuring phosphatidylinositol 4,5-bisphosphate (PIP2) and diacylglycerol (DAG) levels and protein kinase B (AKT) activity. Carvedilol acutely reduced fasting blood glucose levels in both normal and insulin resistant mice without significantly affecting the glucose tolerance. These acute effects were associated with increased hepatic PIP2 but decreased hepatic DAG levels. Chronic administration of carvedilol significantly ameliorated insulin resistance and dyslipidemia in HFrHFD-fed mice. These chronic effects were associated with increased β-arrestin2, PIP2, and AKT activity levels but decreased DAG levels in the classical insulin target tissues. In conclusion, carvedilol acutely maintains glucose homeostasis and chronically ameliorates insulin resistance and dyslipidemia in HFrHFD-fed mice. The insulin sensitizing effects of carvedilol are highly correlated with the upregulation of β-arrestin2 pathway.
Collapse
Affiliation(s)
- Hoda M S Ahmed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Egypt.,Medical Supply Chain, Abo-Hammad Health Administration, Ministry of Health, Egypt
| | - Samar G Mohamed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Egypt.,Department of Toxic and Narcotic Drugs, Forensic Medicine, Cairo Laboratory, Medicolegal Organization, Ministry of Justice, Cairo, Egypt
| | - Wael S Ibrahim
- Department of Pharmacology and Toxicology, School of Pharmacy, Badr University in Cairo (BUC), Cairo, Egypt
| | - Asmaa M Rezk
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Egypt.,Department of Pharmacy, Benha University Hospitals, Benha, Egypt
| | - Amr A A Mahmoud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Egypt
| | - Mona F Mahmoud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Egypt
| | - Islam A A E-H Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Egypt
| |
Collapse
|
16
|
Eibl G, Rozengurt E. Metformin: review of epidemiology and mechanisms of action in pancreatic cancer. Cancer Metastasis Rev 2021; 40:865-878. [PMID: 34142285 DOI: 10.1007/s10555-021-09977-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 05/27/2021] [Indexed: 12/15/2022]
Abstract
Pancreatic ductal adenocarcinoma continues to be a lethal disease, for which efficient treatment options are very limited. Increasing efforts have been taken to understand how to prevent or intercept this disease at an early stage. There is convincing evidence from epidemiologic and preclinical studies that the antidiabetic drug metformin possesses beneficial effects in pancreatic cancer, including reducing the risk of developing the disease and improving survival in patients with early-stage disease. This review will summarize the current literature about the epidemiological data on metformin and pancreatic cancer as well as describe the preclinical evidence illustrating the anticancer effects of metformin in pancreatic cancer. Underlying mechanisms and targets of metformin will also be discussed. These include direct effects on transformed pancreatic epithelial cells and indirect, systemic effects on extra-pancreatic tissues.
Collapse
Affiliation(s)
- Guido Eibl
- Department of Surgery, David Geffen School of Medicine At UCLA, Los Angeles, CA, USA.
| | - Enrique Rozengurt
- Department of Medicine, David Geffen School of Medicine At UCLA, Los Angeles, CA, USA
| |
Collapse
|
17
|
Chen C, Yang X, Liu S, Zhang M, Wang C, Xia X, Lou Y, Xu H. The effect of lipid metabolism regulator anthocyanins from Aronia melanocarpa on 3T3-L1 preadipocytes and C57BL/6 mice via activating AMPK signaling and gut microbiota. Food Funct 2021; 12:6254-6270. [PMID: 34114580 DOI: 10.1039/d1fo00907a] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
This study investigated lipid metabolism regulation by anthocyanins from Aronia melanocarpa (AAM) in 3T3-L1 preadipocytes and high fat diet (HFD) mice. Ultra-performance liquid chromatography/ion mobility quadrupole time-of-flight mass spectrometry analysis identified the constituents of AAM, which decreased lipid content and inflammation in 3T3-L1 cells without cytotoxicity. Meanwhile, taking normal diet and orlistat mice as references, AAM supplementation improved blood lipid levels and adipocyte degeneration, promoted beneficial gut microbial growth, and maintained lipid metabolism in HFD mice. Furthermore, AAM activated the AMP-activated protein kinase (AMPK) signaling pathway, accompanied by the regulation of adipogenic transcription factors and their target genes in vitro and in vivo. Collectively, our data demonstrated that AAM exhibits anti-adipogenic activities that were partially mediated by the AMPK pathway and gut microbiota regulation. This study provides new insight into the regulation of lipid metabolism by AAM and suggests that AAM has potential therapeutic effects on hyperlipidemia.
Collapse
Affiliation(s)
- Chunping Chen
- Department of Food Science and Engineering, College of Agriculture, Yanbian University, Yanji 133000, Jilin, China.
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Secoisolariciresinol Diglucoside Regulates Adipose Tissue Metabolic Disorder in Obese Mice Induced by a Western Diet. J FOOD QUALITY 2021. [DOI: 10.1155/2021/5580772] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Secoisolariciresinol diglucoside (SDG) is the main component of flax lignans. Current studies have reported a positive effect of SDG on obesity and metabolic diseases. SDG has strong blood fat- and blood sugar-lowering, anti-inflammatory, and antioxidant effects and prevents heart disease and other chronic diseases. In this study, we explored the effects of SDG on Western diet-induced obesity and lipid metabolic disorder. Supplementing Western diet-induced obese mice with 40 mg kg1 d1, SDG for 12 weeks significantly reduced body and tissue weights. Increased adiponectin levels and decreased serum leptin and resistin levels were observed in obese mice orally administered SDG. Proliferation of adipose tissue was observed by hematoxylin and eosin staining, and cell size was quantitatively analyzed. As a result, SDG inhibited the proliferation of adipose tissue. In addition, SDG suppressed the mRNA expression of lipid synthetic genes and upregulated the mRNA expression of lipolytic genes. Overall, these results indicate that SDG inhibits obesity induced by a Western diet and regulates adipose tissue metabolic disorder. These results provide a theoretical basis for further study on the regulation of obesity and lipid metabolic disorder caused by SDG.
Collapse
|
19
|
Rehman N, Varghese J. Larval nutrition influences adult fat stores and starvation resistance in Drosophila. PLoS One 2021; 16:e0247175. [PMID: 33606785 PMCID: PMC7895371 DOI: 10.1371/journal.pone.0247175] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 02/02/2021] [Indexed: 12/16/2022] Open
Abstract
Insulin plays a major role in connecting nutrient availability to energy homeostasis by regulating metabolic pathways. Defects in insulin signalling is the primary cause for diabetes, obesity and various metabolic disorders. Nutritional status during growth and developmental stages play a crucial role in determining adult size, fecundity and ageing. However, the association between developmental nutrition and adult metabolic disorders has not been fully explored. Here, we address the effects of nutrient status during the larval growth phase on adult metabolism in Drosophila. We report that restricted food supply in larvae led to higher fat reserves and starvation resistance in mature adult flies, which we attribute to low insulin signalling. A lesser amount of stored fat was mobilised during early adult stages and during acute starvation, which accounts for the metabolic effects. Furthermore, larval diet influenced the expression of fat mobilisation genes brummer and lipid storage droplet-2 in adult flies, which led to the metabolic phenotypes reported here. Thus, the restricted nutrient environment in developing larvae led to adaptive changes that entrain the adult flies for scarce food availability.
Collapse
Affiliation(s)
- Niyas Rehman
- School of Biology, Indian Institute of Science Education and Research (IISER TVM), Thiruvananthapuram, Kerala, India
| | - Jishy Varghese
- School of Biology, Indian Institute of Science Education and Research (IISER TVM), Thiruvananthapuram, Kerala, India
- * E-mail:
| |
Collapse
|
20
|
Romeiro NC, Ferreira CM, Oliveira MF. Assessment of mitochondrial physiology of murine white adipose tissue by mechanical permeabilization and lipid depletion. Anal Biochem 2020; 611:113935. [PMID: 32898480 DOI: 10.1016/j.ab.2020.113935] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 08/25/2020] [Accepted: 08/31/2020] [Indexed: 11/20/2022]
Abstract
White adipose tissue (WAT) represents a major site of triacylglycerol energy storage and is directly associated with metabolic disorders. Mitochondria regulate cellular energy expenditure and are active in WAT. Although isolated mitochondria have been classically used to assess their functions, several artifacts can be introduced by this approach. Furthermore, important limitations exist in the available methods to determine mitochondrial physiology in permeabilized WAT. Here, we established and validated a method for functional evaluation of mice mesenteric WAT (mWAT) mitochondria by using MEchanical Permeabilization and LIpid DEpletion (MEPLIDE) coupled to high-resolution respirometry. We observed that mild stirring of mWAT for 20 min at room temperature with 4% fatty acid-free albumin (FAF-BSA) followed by 50 min without FAF-BSA selectively permeabilized white adipocytes plasma membrane. In these conditions, mWAT mitochondria were intact, exhibiting succinate-induced respiratory rates that were sensitive to classical oxidative phosphorylation modulators. Finally, the respiratory capacity of mWAT in female mice was significantly higher than in males, an observation that agrees with reported data. Therefore, the functional assessment of mWAT mitochondria through MEPLIDE coupled to high resolution respirometry proposed here will contribute to a better understanding of WAT biology in several pathophysiological contexts.
Collapse
Affiliation(s)
- Natália C Romeiro
- Laboratório de Bioquímica de Resposta Ao Estresse, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal Do Rio de Janeiro, Cidade Universitária, Rio de Janeiro, RJ, Brazil
| | - Caroline M Ferreira
- Laboratório de Bioquímica de Resposta Ao Estresse, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal Do Rio de Janeiro, Cidade Universitária, Rio de Janeiro, RJ, Brazil
| | - Marcus F Oliveira
- Laboratório de Bioquímica de Resposta Ao Estresse, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal Do Rio de Janeiro, Cidade Universitária, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
21
|
The Influence of Physical Activity on the Bioactive Lipids Metabolism in Obesity-Induced Muscle Insulin Resistance. Biomolecules 2020; 10:biom10121665. [PMID: 33322719 PMCID: PMC7764345 DOI: 10.3390/biom10121665] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 12/08/2020] [Accepted: 12/11/2020] [Indexed: 12/14/2022] Open
Abstract
High-fat diet consumption and lack of physical activity are important risk factors for metabolic disorders such as insulin resistance and cardiovascular diseases. Insulin resistance is a state of a weakened response of tissues such as skeletal muscle, adipose tissue, and liver to insulin, which causes an increase in blood glucose levels. This condition is the result of inhibition of the intracellular insulin signaling pathway. Skeletal muscle is an important insulin-sensitive tissue that accounts for about 80% of insulin-dependent glucose uptake. Although the exact mechanism by which insulin resistance is induced has not been thoroughly understood, it is known that insulin resistance is most commonly associated with obesity. Therefore, it is believed that lipids may play an important role in inducing insulin resistance. Among lipids, researchers’ attention is mainly focused on biologically active lipids: diacylglycerols (DAG) and ceramides. These lipids are able to regulate the activity of intracellular enzymes, including those involved in insulin signaling. Available data indicate that physical activity affects lipid metabolism and has a positive effect on insulin sensitivity in skeletal muscles. In this review, we have presented the current state of knowledge about the impact of physical activity on insulin resistance and metabolism of biologically active lipids.
Collapse
|
22
|
Warren JL, Hunter GR, Gower BA, Bamman MM, Windham ST, Moellering DR, Fisher G. Exercise Effects on Mitochondrial Function and Lipid Metabolism during Energy Balance. Med Sci Sports Exerc 2020; 52:827-834. [PMID: 31652245 DOI: 10.1249/mss.0000000000002190] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
INTRODUCTION/PURPOSE Aerobic exercise training (AET) has been shown to improve mitochondrial bioenergetics and upregulate proteins related to lipid metabolism. However, it remains to be determined if these alterations associated with AET persist when measured in energy balance (EB) in the days after the last bout of training. The purpose of the study was to test the hypothesis that improvements in skeletal muscle mitochondrial function induced by AET observed in previous literature would persist when measured after restoring EB conditions 72 h removed from the last exercise bout. METHODS Participants were 14 premenopausal women (age = 31.2 ± 6.7 yr, BMI = 26.6 ± 5.1 kg·m). The AET program required three monitored training sessions per week for 8-16 wk. Skeletal muscle biopsies were obtained at baseline and after 8-16 wk of AET (≥72 h after the last exercise bout). All food was provided for 72 h before biopsies, and EB was managed 24 h before testing within ±100 kcal of measured energy requirements using a whole-room calorimeter. Mitochondrial oxidative capacity was quantified in permeabilized muscle fibers from the vastus lateralis. RESULTS We found that AET increased coupled respiration (154%) and uncoupled respiration (90%) rates using a fatty acid substrate (palmitoyl carnitine) (P < 0.05). However, when rates were normalized to complex IV activity (a marker of mitochondrial content), no significant differences were observed. In addition, there were no changes in proteins known to mediate mitochondrial biogenesis or lipid transport and metabolism after AET. CONCLUSION Eight to 16 wk of AET improved mitochondrial capacity under fatty acid substrate when assessed in EB, which appears to be due to mitochondrial biogenesis.
Collapse
Affiliation(s)
- Jonathan L Warren
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL
| | - Gary R Hunter
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL
| | - Barbara A Gower
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL
| | - Marcas M Bamman
- Department of Cell Development and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL
| | - Samuel T Windham
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL
| | - Douglas R Moellering
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL
| | - Gordon Fisher
- Department of Human Studies, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
23
|
Morgan PT, Smeuninx B, Breen L. Exploring the Impact of Obesity on Skeletal Muscle Function in Older Age. Front Nutr 2020; 7:569904. [PMID: 33335909 PMCID: PMC7736105 DOI: 10.3389/fnut.2020.569904] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 11/06/2020] [Indexed: 12/16/2022] Open
Abstract
Sarcopenia is of important clinical relevance for loss of independence in older adults. The prevalence of obesity in combination with sarcopenia ("sarcopenic-obesity") is increasing at a rapid rate. However, whilst the development of sarcopenia is understood to be multi-factorial and harmful to health, the role of obesity from a protective and damaging perspective on skeletal muscle in aging, is poorly understood. Specifically, the presence of obesity in older age may be accompanied by a greater volume of skeletal muscle mass in weight-bearing muscles compared with lean older individuals, despite impaired physical function and resistance to anabolic stimuli. Collectively, these findings support a potential paradox in which obesity may protect skeletal muscle mass in older age. One explanation for these paradoxical findings may be that the anabolic response to weight-bearing activity could be greater in obese vs. lean older individuals due to a larger mechanical stimulus, compensating for the heightened muscle anabolic resistance. However, it is likely that there is a complex interplay between muscle, adipose, and external influences in the aging process that are ultimately harmful to health in the long-term. This narrative briefly explores some of the potential mechanisms regulating changes in skeletal muscle mass and function in aging combined with obesity and the interplay with sarcopenia, with a particular focus on muscle morphology and the regulation of muscle proteostasis. In addition, whilst highly complex, we attempt to provide an updated summary for the role of obesity from a protective and damaging perspective on muscle mass and function in older age. We conclude with a brief discussion on treatment of sarcopenia and obesity and a summary of future directions for this research field.
Collapse
Affiliation(s)
- Paul T. Morgan
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Benoit Smeuninx
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, United Kingdom
- Cellular & Molecular Metabolism Laboratory, Monash Institute of Pharmacological Sciences, Monash University, Parkville, VIC, Australia
| | - Leigh Breen
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
24
|
Angiotensin-(1-7) Improves Integrated Cardiometabolic Function in Aged Mice. Int J Mol Sci 2020; 21:ijms21145131. [PMID: 32698498 PMCID: PMC7403973 DOI: 10.3390/ijms21145131] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 07/14/2020] [Accepted: 07/18/2020] [Indexed: 01/07/2023] Open
Abstract
Angiotensin (Ang)-(1-7) is a beneficial renin–angiotensin system (RAS) hormone that elicits protective cardiometabolic effects in young animal models of hypertension, obesity, and metabolic syndrome. The impact of Ang-(1-7) on cardiovascular and metabolic outcomes during aging, however, remains unexplored. This study tested the hypothesis that Ang-(1-7) attenuates age-related elevations in blood pressure and insulin resistance in mice. Young adult (two-month-old) and aged (16-month-old) male C57BL/6J mice received Ang-(1-7) (400 ng/kg/min) or saline for six-weeks via a subcutaneous osmotic mini-pump. Arterial blood pressure and metabolic function indices (body composition, insulin sensitivity, and glucose tolerance) were measured at the end of treatment. Adipose and cardiac tissue masses and cardiac RAS, sympathetic and inflammatory marker gene expression were also measured. We found that chronic Ang-(1-7) treatment decreased systolic and mean blood pressure, with a similar trend for diastolic blood pressure. Ang-(1-7) also improved insulin sensitivity in aged mice to levels in young mice, without effects on glucose tolerance or body composition. The blood pressure–lowering effects of Ang-(1-7) in aged mice were associated with reduced sympathetic outflow to the heart. These findings suggest Ang-(1-7) may provide a novel pharmacological target to improve age-related cardiometabolic risk.
Collapse
|
25
|
Movahedian M, Thomas J, Rahmani J, Clark CCT, Rashidkhani B, Ghanavati M. Association between dietary glycemic index and glycemic load, insulin index and load with incidence of age-related cataract: Results from a case-control study. Diabetes Metab Syndr 2020; 14:199-204. [PMID: 32155574 DOI: 10.1016/j.dsx.2020.02.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 02/24/2020] [Accepted: 02/24/2020] [Indexed: 12/13/2022]
Abstract
AIM To identify the association between the dietary carbohydrate indexes, such as dietary glycemic index (DGI) and load (DGL), dietary insulin index (DII) and load (DIL), with the possibility of cataract. METHOD This case-control study consisted of 101 new cases of cataract and 202 controls. DGI and DGL were computed through DGI values previously published. DII was also calculated based on dietary insulin index data published previously. RESULTS There was a significant positive association between the highest quartiles of DGI (OR = 6.56; 95% CI = 2.67-16.06; P < 0.001), DGL (OR = 6.17; 95% CI = 1.93-19.37; P = 0.002) and DIL (OR = 4.17; 95% CI = 1.41-12.27; P = 0.004) with risk of cataract, compared to those on the lowest quartile, but not for DII (OR = 0.85; 95% CI = 0.39-1.86; P = 0.82). Furthermore, after stratifying groups by BMI, a significant direct association between highest quartile of DGI (OR = 6.76; 95% CI = 2.49-18.38; P < 0.001) and DGL (OR = 3.45; 95% CI = 0.96-12.37; P = 0.05) with risk of cataract was evident in individuals with elevated BMI (BMI≥25). CONCLUSION We found a significant, direct, relationship between DGI, DGL and DIL with risk of cataract. However, the association between DII and the risk of cataract was not significant, even after adjusting for related confounders.
Collapse
Affiliation(s)
- Mina Movahedian
- Department of Clinical Nutrition, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Jessica Thomas
- South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Jamal Rahmani
- Department of Community Nutrition, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Cain C T Clark
- Centre for Sport, Exercise, and Life Sciences, Coventry University, CV1 5FB, UK
| | - Bahram Rashidkhani
- Department of Community Nutrition, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Matin Ghanavati
- Department of Clinical Nutrition, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
26
|
Bretschneider H, Quade M, Lode A, Gelinsky M, Rammelt S, Zwingenberger S, Schaser KD, Vater C. Characterization of Naturally Occurring Bioactive Factor Mixtures for Bone Regeneration. Int J Mol Sci 2020; 21:ijms21041412. [PMID: 32093051 PMCID: PMC7073126 DOI: 10.3390/ijms21041412] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 02/11/2020] [Accepted: 02/13/2020] [Indexed: 12/13/2022] Open
Abstract
In this study, the bone-regenerative potential of bioactive factors derived from adipose tissue, platelet-rich plasma (PRP) and conditioned medium from hypoxia-treated human telomerase immortalized bone-marrow-derived mesenchymal stem cells (hTERT-MSC) was investigated in vitro with the aim to develop cost-effective and efficient bone substitutes for optimized regeneration of bone defects. Adipose tissue was harvested from human donors undergoing reconstructive surgery, and adipose tissue extract (ATE) was prepared. Platelet lysates (PL) were produced by repeated freeze-thaw cycles of PRP, and hypoxia-conditioned medium (HCM) was obtained by culturing human telomerase immortalized bone-marrow-derived mesenchymal stromal cells for 5 days with 1% O2. Besides analysis by cytokine and angiogenesis arrays, ELISA was performed. Angiogenic potential was investigated in cocultures of bone-marrow-derived (BM)-MSC and human umbilical vein endothelial cells. Multiple angiogenic proteins and cytokines were detected in all growth factor mixtures. HCM and ATE contained high amounts of angiogenin and CCL2/MCP-1, whereas PL contained high amounts of IGFBP-1. Culturing cells with HCM and ATE significantly increased specific ALP activity of BM-MSC as well as tubule length and junctions of endothelial networks, indicating osteogenic and angiogenic stimulation. To achieve a synergism between chemoattractive potential and osteogenic and angiogenic differentiation capacity, a combination of different growth factors appears promising for potential clinical applications.
Collapse
Affiliation(s)
- Henriette Bretschneider
- University Center of Orthopaedics and Traumatology, University Hospital Carl Gustav Carus of Technische, Universität Dresden, Fetscherstraße 74, 01307 Dresden, Germany
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus and Faculty of Medicine of Technische, Universität Dresden, Fetscherstraße 74, 01307 Dresden, Germany
| | - Mandy Quade
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus and Faculty of Medicine of Technische, Universität Dresden, Fetscherstraße 74, 01307 Dresden, Germany
| | - Anja Lode
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus and Faculty of Medicine of Technische, Universität Dresden, Fetscherstraße 74, 01307 Dresden, Germany
| | - Michael Gelinsky
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus and Faculty of Medicine of Technische, Universität Dresden, Fetscherstraße 74, 01307 Dresden, Germany
| | - Stefan Rammelt
- University Center of Orthopaedics and Traumatology, University Hospital Carl Gustav Carus of Technische, Universität Dresden, Fetscherstraße 74, 01307 Dresden, Germany
| | - Stefan Zwingenberger
- University Center of Orthopaedics and Traumatology, University Hospital Carl Gustav Carus of Technische, Universität Dresden, Fetscherstraße 74, 01307 Dresden, Germany
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus and Faculty of Medicine of Technische, Universität Dresden, Fetscherstraße 74, 01307 Dresden, Germany
| | - Klaus-Dieter Schaser
- University Center of Orthopaedics and Traumatology, University Hospital Carl Gustav Carus of Technische, Universität Dresden, Fetscherstraße 74, 01307 Dresden, Germany
| | - Corina Vater
- University Center of Orthopaedics and Traumatology, University Hospital Carl Gustav Carus of Technische, Universität Dresden, Fetscherstraße 74, 01307 Dresden, Germany
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus and Faculty of Medicine of Technische, Universität Dresden, Fetscherstraße 74, 01307 Dresden, Germany
- Correspondence:
| |
Collapse
|
27
|
Elshenawy S, Pinney SE, Stuart T, Doulias PT, Zura G, Parry S, Elovitz MA, Bennett MJ, Bansal A, Strauss JF, Ischiropoulos H, Simmons RA. The Metabolomic Signature of the Placenta in Spontaneous Preterm Birth. Int J Mol Sci 2020; 21:ijms21031043. [PMID: 32033212 PMCID: PMC7037776 DOI: 10.3390/ijms21031043] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 01/21/2020] [Accepted: 01/30/2020] [Indexed: 12/16/2022] Open
Abstract
The placenta is metabolically active and supports the growth of the fetus. We hypothesize that deficits in the capacity of the placenta to maintain bioenergetic and metabolic stability during pregnancy may result in spontaneous preterm birth (SPTB). To explore this hypothesis, we performed a nested cased control study of metabolomic signatures in placentas from women with SPTB (<36 weeks gestation) compared to normal pregnancies (≥38 weeks gestation). To control for the effects of gestational age on placenta metabolism, we also studied a subset of metabolites in non-laboring preterm and term Rhesus monkeys. Comprehensive quantification of metabolites demonstrated a significant elevation in the levels of amino acids, prostaglandins, sphingolipids, lysolipids, and acylcarnitines in SPTB placenta compared to term placenta. Additional quantification of placental acylcarnitines by tandem mass spectrometry confirmed the significant elevation in SPTB human, with no significant differences between midgestation and term placenta in Rhesus macaque. Fatty acid oxidation as measured by the flux of 3H-palmitate in SPTB placenta was lower than term. Collectively, significant and biologically relevant alterations in the placenta metabolome were identified in SPTB placenta. Altered acylcarnitine levels and fatty acid oxidation suggest that disruption in normal substrate metabolism is associated with SPTB.
Collapse
Affiliation(s)
- Summer Elshenawy
- Division of Neonatology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA; (S.E.); (T.S.); (P.-T.D.); (G.Z.); (H.I.)
| | - Sara E. Pinney
- Division of Endocrinology and Diabetes, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA;
- Center for Research on Reproduction and Women’s Health, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; (S.P.); (M.A.E.); (A.B.)
| | - Tami Stuart
- Division of Neonatology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA; (S.E.); (T.S.); (P.-T.D.); (G.Z.); (H.I.)
| | - Paschalis-Thomas Doulias
- Division of Neonatology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA; (S.E.); (T.S.); (P.-T.D.); (G.Z.); (H.I.)
| | - Gabriella Zura
- Division of Neonatology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA; (S.E.); (T.S.); (P.-T.D.); (G.Z.); (H.I.)
| | - Samuel Parry
- Center for Research on Reproduction and Women’s Health, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; (S.P.); (M.A.E.); (A.B.)
- Department of Obstetrics and Gynecology, Maternal and Child Health Research Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michal A. Elovitz
- Center for Research on Reproduction and Women’s Health, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; (S.P.); (M.A.E.); (A.B.)
- Department of Obstetrics and Gynecology, Maternal and Child Health Research Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael J. Bennett
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania and Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA;
| | - Amita Bansal
- Center for Research on Reproduction and Women’s Health, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; (S.P.); (M.A.E.); (A.B.)
| | - Jerome F. Strauss
- Center for Research on Reproduction and Women’s Health, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; (S.P.); (M.A.E.); (A.B.)
- Department of Obstetrics and Gynecology, Maternal and Child Health Research Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Harry Ischiropoulos
- Division of Neonatology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA; (S.E.); (T.S.); (P.-T.D.); (G.Z.); (H.I.)
| | - Rebecca A. Simmons
- Division of Neonatology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA; (S.E.); (T.S.); (P.-T.D.); (G.Z.); (H.I.)
- Center for Research on Reproduction and Women’s Health, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; (S.P.); (M.A.E.); (A.B.)
- Department of Obstetrics and Gynecology, Maternal and Child Health Research Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
- Correspondence:
| |
Collapse
|
28
|
Stevens VL, Carter BD, McCullough ML, Campbell PT, Wang Y. Metabolomic Profiles Associated with BMI, Waist Circumference, and Diabetes and Inflammation Biomarkers in Women. Obesity (Silver Spring) 2020; 28:187-196. [PMID: 31777189 DOI: 10.1002/oby.22670] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 09/06/2019] [Indexed: 12/18/2022]
Abstract
OBJECTIVE This study was undertaken to identify metabolites associated with BMI and waist circumference (WC) in women and to determine whether these metabolites are associated with biomarkers of metabolic health. METHODS Untargeted metabolomic analysis was done on serum from 1,534 women. Metabolites associated with BMI and WC were identified using linear regression with a Bonferroni-corrected P value. Clustered blocks of these metabolites were then defined whose association with the anthropometric measures could be represented by a single metabolite. The association of these representative metabolites with biomarkers for diabetes and inflammation was then determined. RESULTS About one-third of 781 metabolites included in the analyses were associated with BMI and/or WC. Associations were found for some novel metabolites, including several sphingolipids, nucleotides, and modified fatty acids. Among metabolites most strongly inversely associated with BMI, the choline-containing plasmalogen (O-16:0/18:1) (β = -0.30, P = 6.62 × 10-32 ) was also inversely associated with c-peptide and positively associated with adiponectin. Adjustment for BMI attenuated the metabolite-biomarker associations more for hemoglobin A1c (> 100%) and c-peptide (58.8% to > 100%) than for C-reactive protein (10.5%-40.0%) and adiponectin (7.0%-30.4%). CONCLUSIONS These results add to the list of metabolites associated with adiposity and indicate that some may influence processes that contribute to the development of obesity-related diseases.
Collapse
Affiliation(s)
- Victoria L Stevens
- Behavioral and Epidemiology Research Group, American Cancer Society, Atlanta, Georgia, USA
| | - Brian D Carter
- Behavioral and Epidemiology Research Group, American Cancer Society, Atlanta, Georgia, USA
| | - Marjorie L McCullough
- Behavioral and Epidemiology Research Group, American Cancer Society, Atlanta, Georgia, USA
| | - Peter T Campbell
- Behavioral and Epidemiology Research Group, American Cancer Society, Atlanta, Georgia, USA
| | - Ying Wang
- Behavioral and Epidemiology Research Group, American Cancer Society, Atlanta, Georgia, USA
| |
Collapse
|
29
|
Wang G, Du K, Xie Z, Tang R, Jia X, Chen S, Lai S. Screening and Identification of Differentially Expressed and Adipose Growth-Related Protein-Coding Genes During the Deposition of Perirenal Adipose Tissue in Rabbits. Diabetes Metab Syndr Obes 2020; 13:4669-4680. [PMID: 33293841 PMCID: PMC7719053 DOI: 10.2147/dmso.s284246] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 11/12/2020] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Rabbit is a good model for genetic and medical studies in other livestock species. The rabbit shows low adipose tissue deposition, and the phenomena indicates that there is some specificity of adipose deposition during the rabbit growth. However, little is known about genes that regulate the growth of adipose tissue in rabbits. MATERIALS AND METHODS Deep RNA-seq and comprehensive bioinformatics analyses were used to characterize the genes of rabbit visceral adipose tissue (VAT) at 35, 85 and 120 days after birth. Differentially expressed genes (DEGs) were identified at the three growth stages by DESeq. To explore the function of the candidate genes, Gene Ontology (GO) enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were performed. Six DEGs were randomly selected, and their expression profiles were validated by q-PCR. RESULTS A total of 20,303 known transcripts and 99,199 new transcripts from 8 RNA sequencing libraries were identified, and 34 differentially expressed genes (DEGs) were screened. GO enrichment and KEGG pathway analyses revealed that the DEGs were mainly involved in lipid metabolism regulation including acylglycerol metabolic process and mobilization, and decomposition of lipids to generate ATP in adipocytes and fatty acid metabolism, included LOC100342322 and LOC100342572. In addition, 133 protein-coding genes that play a role in adipose growth and development were screened, including acyl-CoA synthetase long-chain family member 5 (ACSL5) and fatty acid-binding protein 2 (FABP2). The validation results of six DEGs by q-PCR showed similar trends with the results of RNA-seq. CONCLUSION In summary, this study provides the first report of the coding genes profiles of rabbit adipose tissue during different growth stages. These data allow for the identification of candidate genes for subsequent studies on rabbit genetics and regulation of adipose cells, and provide an animal model for studying obesity in humans.
Collapse
Affiliation(s)
- Guoze Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu611130, People’s Republic of China
- College of Food Science, Guizhou Medical University, Guiyang550025, People’s Republic of China
| | - Kun Du
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu611130, People’s Republic of China
| | - Zhenjian Xie
- College of Pharmacy and Biological Engineering, Chengdu University, Chengdu610106, People’s Republic of China
| | - Renyong Tang
- College of Pharmacy and Biological Engineering, Chengdu University, Chengdu610106, People’s Republic of China
| | - Xianbo Jia
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu611130, People’s Republic of China
| | - Shiyi Chen
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu611130, People’s Republic of China
| | - Songjia Lai
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu611130, People’s Republic of China
- Correspondence: Songjia Lai Email
| |
Collapse
|
30
|
White MC, Miller AJ, Loloi J, Bingaman SS, Shen B, Wang M, Silberman Y, Lindsey SH, Arnold AC. Sex differences in metabolic effects of angiotensin-(1-7) treatment in obese mice. Biol Sex Differ 2019; 10:36. [PMID: 31315689 PMCID: PMC6637512 DOI: 10.1186/s13293-019-0251-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 07/09/2019] [Indexed: 12/19/2022] Open
Abstract
Background Angiotensin-(1-7) is a beneficial hormone of the renin-angiotensin system known to play a positive role in regulation of blood pressure and glucose homeostasis. Previous studies have shown that in high-fat diet (HFD)-induced obese male mice, circulating angiotensin-(1-7) levels are reduced and chronic restoration of this hormone reverses diet-induced insulin resistance; however, this has yet to be examined in female mice. We hypothesized angiotensin-(1-7) would improve insulin sensitivity and glucose tolerance in obese female mice, to a similar extent as previously observed in male mice. Methods Five-week-old male and female C57BL/6J mice (8–12/group) were placed on control diet or HFD (16% or 59% kcal from fat, respectively) for 11 weeks. After 8 weeks of diet, mice were implanted with an osmotic pump for 3-week subcutaneous delivery of angiotensin-(1-7) (400 ng/kg/min) or saline vehicle. During the last week of treatment, body mass and composition were measured and intraperitoneal insulin and glucose tolerance tests were performed to assess insulin sensitivity and glucose tolerance, respectively. Mice were euthanized at the end of the study for blood and tissue collection. Results HFD increased body mass and adiposity in both sexes. Chronic angiotensin-(1-7) infusion significantly decreased body mass and adiposity and increased lean mass in obese mice of both sexes. While both sexes tended to develop mild hyperglycemia in response to HFD, female mice developed less marked hyperinsulinemia. There was no effect of angiotensin-(1-7) on fasting glucose or insulin levels among diet and sex groups. Male and female mice similarly developed insulin resistance and glucose intolerance in response to HFD feeding. Angiotensin-(1-7) improved insulin sensitivity in both sexes but corrected glucose intolerance only in obese female mice. There were no effects of sex or angiotensin-(1-7) treatment on any of the study outcomes in control diet-fed mice. Conclusions This study provides new evidence for sex differences in the impact of chronic angiotensin-(1-7) in obese mice, with females having greater changes in glucose tolerance with treatment. These findings improve understanding of sex differences in renin-angiotensin mechanisms in obesity and illustrate the potential for targeting angiotensin-(1-7) for treatment of this condition.
Collapse
Affiliation(s)
- Melissa C White
- Department of Comparative Medicine, Penn State College of Medicine, 500 University Drive, Hershey, PA, USA
| | - Amanda J Miller
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, 500 University Drive Mail Code H109, Hershey, PA, 17033, USA
| | - Justin Loloi
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, 500 University Drive Mail Code H109, Hershey, PA, 17033, USA
| | - Sarah S Bingaman
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, 500 University Drive Mail Code H109, Hershey, PA, 17033, USA
| | - Biyi Shen
- Department of Public Health Sciences, Penn State College of Medicine, 500 University Drive, Hershey, PA, USA
| | - Ming Wang
- Department of Public Health Sciences, Penn State College of Medicine, 500 University Drive, Hershey, PA, USA
| | - Yuval Silberman
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, 500 University Drive Mail Code H109, Hershey, PA, 17033, USA
| | - Sarah H Lindsey
- Department of Pharmacology, Tulane University, 1430 Tulane Avenue, New Orleans, LA, #8683, USA
| | - Amy C Arnold
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, 500 University Drive Mail Code H109, Hershey, PA, 17033, USA.
| |
Collapse
|
31
|
White MC, Fleeman R, Arnold AC. Sex differences in the metabolic effects of the renin-angiotensin system. Biol Sex Differ 2019; 10:31. [PMID: 31262355 PMCID: PMC6604144 DOI: 10.1186/s13293-019-0247-5] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 06/18/2019] [Indexed: 02/07/2023] Open
Abstract
Obesity is a global epidemic that greatly increases risk for developing cardiovascular disease and type II diabetes. Sex differences in the obese phenotype are well established in experimental animal models and clinical populations. While having higher adiposity and obesity prevalence, females are generally protected from obesity-related metabolic and cardiovascular complications. This protection is, at least in part, attributed to sex differences in metabolic effects of hormonal mediators such as the renin-angiotensin system (RAS). Previous literature has predominantly focused on the vasoconstrictor arm of the RAS and shown that, in contrast to male rodent models of obesity and diabetes, females are protected from metabolic and cardiovascular derangements produced by angiotensinogen, renin, and angiotensin II. A vasodilator arm of the RAS has more recently emerged which includes angiotensin-(1-7), angiotensin-converting enzyme 2 (ACE2), mas receptors, and alamandine. While accumulating evidence suggests that activation of components of this counter-regulatory axis produces positive effects on glucose homeostasis, lipid metabolism, and energy balance in male animal models, female comparison studies and clinical data related to metabolic outcomes are lacking. This review will summarize current knowledge of sex differences in metabolic effects of the RAS, focusing on interactions with gonadal hormones and potential clinical implications.
Collapse
Affiliation(s)
- Melissa C White
- Department of Comparative Medicine, Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA, USA
| | - Rebecca Fleeman
- Department of Neural and Behavioral Sciences, Pennsylvania State University College of Medicine, 500 University Drive, Mail Code H109, Hershey, PA, 17033, USA
| | - Amy C Arnold
- Department of Neural and Behavioral Sciences, Pennsylvania State University College of Medicine, 500 University Drive, Mail Code H109, Hershey, PA, 17033, USA.
| |
Collapse
|
32
|
Saito H, Tanaka T, Sugahara M, Tanaka S, Fukui K, Wakashima T, Nangaku M. Inhibition of prolyl hydroxylase domain (PHD) by JTZ-951 reduces obesity-related diseases in the liver, white adipose tissue, and kidney in mice with a high-fat diet. J Transl Med 2019; 99:1217-1232. [PMID: 30952940 DOI: 10.1038/s41374-019-0239-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 01/16/2019] [Accepted: 01/28/2019] [Indexed: 12/11/2022] Open
Abstract
The epidemic of obesity and its complications is rapidly increasing worldwide. Recent drug discoveries established the utility of prolyl hydroxylase domain (PHD) inhibitors as stabilizers of hypoxia-inducible factors (HIFs) in vivo, which are currently in human clinical studies for the treatment of anemia in chronic kidney disease (CKD). These studies suggest a role for PHD inhibitors in ameliorating obesity and hyperlipidemia. We hypothesized that HIF activation using a PHD inhibitor, JTZ-951, protects from obesity-related diseases in the white adipose tissue (WAT), liver, and kidney in mice fed with high-fat diet (HFD). Eight-week-old, C57BL/6J mice were fed with HFD for 20 weeks with or without JTZ-951(0.005%; mixed in chow). Body weight and plasma non-high-density lipoprotein (HDL) cholesterol levels were significantly lower in the JTZ-951 group as compared with the vehicle group. PHD inhibition improved liver steatosis, macrophage infiltration into WAT and adipocyte fibrosis. In the kidney, PHD inhibition reduced albuminuria. Histologically, the number of F4/80- positive infiltrating macrophages and mesangial expansion were milder in the JTZ-951 group. Relative mRNA expression of adiponectin in WAT was higher in the JTZ-951-treated group and inversely correlated with hepatic steatosis score, adipocyte macrophage aggregation, and albuminuria. Activation of HIF ameliorates multiple obesity-related consequences in mice with HFD. The results of the present study offer the promising view that pharmacological PHD inhibition may be beneficial for the treatment of obesity-related diseases that can be ameliorated by weight loss.
Collapse
Affiliation(s)
- Hisako Saito
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Tetsuhiro Tanaka
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan.
| | - Mai Sugahara
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Shinji Tanaka
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Kenji Fukui
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan.,Biological and Pharmacological Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc, Osaka, Japan
| | - Takeshi Wakashima
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan.,Biological and Pharmacological Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc, Osaka, Japan
| | - Masaomi Nangaku
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan.
| |
Collapse
|
33
|
Elashry MI, Eldaey A, Glenske K, Matsakas A, Wenisch S, Arnhold S, Patel K. The effect of high-fat diet on the morphological properties of the forelimb musculature in hypertrophic myostatin null mice. J Anat 2019; 235:825-835. [PMID: 31198988 DOI: 10.1111/joa.13025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2019] [Indexed: 12/20/2022] Open
Abstract
Obesity is a worldwide nutritional disorder affecting body performance, including skeletal muscle. Inhibition of myostatin not only increases the muscle mass but also it reduces body fat accumulation. We examined the effect of high-fat diet on the phenotypic properties of forelimb muscles from myostatin null mice. Male wild-type and myostatin null mice were fed on either a normal diet or a high-fat diet (45% fat) for 10 weeks. Musculus triceps brachii Caput longum; M. triceps brachii Caput laterale; M. triceps brachii Caput mediale; M. extensor carpi ulnaris and M. flexor carpi ulnaris were processed for fiber type composition using immunohistochemistry and morphometric analysis. Although the muscle mass revealed no change under a high-fat diet, there were morphometric alterations in the absence of myostatin. We show that high-fat diet reduces the cross-sectional area of the fast (IIB and IIX) fibers in M. triceps brachii Caput longum and M. triceps brachii Caput laterale of both genotypes. In contrast, increases of fast fiber areas were observed in both M. extensor carpi ulnaris of wild-type and M. flexor carpi ulnaris of myostatin null mice. Meanwhile, a high-fat diet increased the area of the fast IIA fibers in wild-type mice; myostatin null mice display a muscle-dependent alteration in the area of the same fiber type. The combined high-fat diet and myostatin deletion shows no effect on the area of slow type I fibers. Although a high-fat diet causes a reduction in the area of the peripheral IIB fibers in both genotypes, only myostatin null mice show an increase in the area of the central IIB fibers. We provide evidence that a high-fat diet induces a muscle-dependent fast to slow myofiber shift in the absence of myostatin. The data suggest that the morphological alterations of muscle fibers under a combined high-fat diet and myostatin deletion reflect a functional adaptation of the muscle to utilize the high energy intake.
Collapse
Affiliation(s)
- Mohamed I Elashry
- Anatomy and Embryology Department, Faculty of Veterinary Medicine, University of Mansoura, Mansoura, Egypt.,Institute of Veterinary Anatomy, Histology and Embryology, Justus Liebig University of Giessen, Giessen, Germany
| | - Asmaa Eldaey
- Anatomy and Embryology Department, Faculty of Veterinary Medicine, University of Mansoura, Mansoura, Egypt.,Clinic of Small Animals, c/o Institute of Veterinary Anatomy, Histology and Embryology, Justus Liebig University of Giessen, Giessen, Germany
| | - Kristina Glenske
- Clinic of Small Animals, c/o Institute of Veterinary Anatomy, Histology and Embryology, Justus Liebig University of Giessen, Giessen, Germany
| | - Antonios Matsakas
- Molecular Physiology Laboratory, Centre for Atherothrombotic and Metabolic Disease, Hull York Medical School, University of Hull, Hull, UK
| | - Sabine Wenisch
- Clinic of Small Animals, c/o Institute of Veterinary Anatomy, Histology and Embryology, Justus Liebig University of Giessen, Giessen, Germany
| | - Stefan Arnhold
- Institute of Veterinary Anatomy, Histology and Embryology, Justus Liebig University of Giessen, Giessen, Germany
| | - Ketan Patel
- School of Biological Sciences, University of Reading, Reading, UK
| |
Collapse
|
34
|
Quines CB, Jardim NS, Araujo PCO, Cechella JL, Prado VC, Nogueira CW. Resistance training restores metabolic alterations induced by monosodium glutamate in a sex-dependent manner in male and female rats. J Cell Biochem 2019; 120:13426-13440. [PMID: 30916837 DOI: 10.1002/jcb.28617] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Revised: 01/14/2019] [Accepted: 01/28/2019] [Indexed: 01/07/2023]
Abstract
Despite resistance exercises being associated with health outcomes, numerous issues are still unresolved and further research is required before the exercise can faithfully be prescribed as medicine. The goal of this study was to investigate whether there are sex differences in resistance training effects on metabolic alterations induced by monosodium glutamate (MSG), a model of obesity, in male and female rats. Male and female Wistar rats received MSG (4 g/kg body weight/day, s.c.) from postnatal day 1 to 10. After 10 days from MSG administration, the rats were separated into two groups: MSG-sedentary and MSG-exercised. At postnatal day 60, the animals started a resistance training protocol in an 80 degrees inclined vertical ladder apparatus and performed it for 7 weeks. Control rats received saline solution and were divided in saline-sedentary and saline-exercised. Resistance training restored all plasma biochemical parameters (glucose, cholesterol, triglycerides, aspartate aminotransferase, and alanine aminotransferase) increased in male and female rats treated with MSG. The MSG administration induced hyperglycemia associated with a decrease in the skeletal muscle glucose transporter 4 (GLUT4) levels and accompanied by deregulation in proteins, G-6Pase, and tyrosine aminotransferase, involved in hepatic glucose metabolism of male and female rats. MSG induced dyslipidemia and lipotoxicity in the liver and skeletal muscle of male rats. Regarding female rats, lipotoxicity was found only in the skeletal muscle. The resistance training had beneficial effects against metabolic alterations induced by MSG in male and female rats, through regulation of proteins (GLUT2, protein kinase B, and GLUT4) involved in glucose and lipid pathways in the liver and skeletal muscle.
Collapse
Affiliation(s)
- Caroline B Quines
- Departamento de Bioquímica e Biologia Molecular, Laboratório de Síntese, Reatividade e Avaliação Farmacológica e Toxicológica de Organocalcogênios, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Natália S Jardim
- Departamento de Bioquímica e Biologia Molecular, Laboratório de Síntese, Reatividade e Avaliação Farmacológica e Toxicológica de Organocalcogênios, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Paulo Cesar O Araujo
- Departamento de Bioquímica e Biologia Molecular, Laboratório de Síntese, Reatividade e Avaliação Farmacológica e Toxicológica de Organocalcogênios, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - José Luiz Cechella
- Departamento de Bioquímica e Biologia Molecular, Laboratório de Síntese, Reatividade e Avaliação Farmacológica e Toxicológica de Organocalcogênios, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Vinicius C Prado
- Departamento de Bioquímica e Biologia Molecular, Laboratório de Síntese, Reatividade e Avaliação Farmacológica e Toxicológica de Organocalcogênios, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Cristina W Nogueira
- Departamento de Bioquímica e Biologia Molecular, Laboratório de Síntese, Reatividade e Avaliação Farmacológica e Toxicológica de Organocalcogênios, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| |
Collapse
|
35
|
Guo J, Tao H, Alasadi A, Huang Q, Jin S. Niclosamide piperazine prevents high-fat diet-induced obesity and diabetic symptoms in mice. Eat Weight Disord 2019; 24:91-96. [PMID: 28780747 DOI: 10.1007/s40519-017-0424-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Accepted: 07/21/2017] [Indexed: 12/27/2022] Open
Abstract
PURPOSE Obesity and type 2 diabetes (T2D) have become the major public health challenges globally. Mitochondrial uncoupling, which reduces intracellular lipid loads and corrects the underlying cause of insulin resistance, has emerged as a promising anti-obese and anti-diabetic intervention. Niclosamide is an anthelmintic drug approved by the US FDA with the mechanism of action that uncouples mitochondria of parasitic worms. Recently, niclosamide ethanolamine salt (NEN) was found to be a safe and effective hepatic mitochondrial uncoupler for the prevention and treatment of obesity and T2D in mouse models. The striking features of NEN prompt us to examine the anti-obese and anti-diabetic efficacy of other salt forms of niclosamide, with the ultimate goal to identify a suitable salt formulation for future clinical development. Here, we report the study with niclosamide piperazine salt (NPP), another salt form of niclosamide with documented safety profile. METHODS Mitochondrial uncoupling activity of NEN and NPP were determined by oxygen consumption assay with Seahorse XF24e Analyzer, as well as by mitochondrial membrane potential measurement in cultured cells. The in vivo anti-diabetic and anti-obesity activities were determined in C57BL/6J mice fed high-fat diet (HFD) or HFD containing 2000 ppm. NPP for 11 weeks. RESULTS Niclosamide piperazine salt showed a comparable mitochondrial uncoupling activity to NEN. Oral administration of NPP significantly reduced HFD-induced obesity, hyperglycemia and hepatic steatosis, and sensitized the insulin responses in mice. CONCLUSIONS Niclosamide piperazine salt may hold the promise to become an alternative to NEN as a drug lead for the treatment of obesity and T2D. No level of evidence Animal study.
Collapse
Affiliation(s)
- Jingjing Guo
- Department of Food Science, Rutgers University, New Brunswick, NJ, USA
| | - Hanlin Tao
- Department of Pharmacology, Rutgers University-Robert Wood Johnson Medical School, Piscataway, NJ, USA
| | - Amer Alasadi
- Department of Physiology, Rutgers University-Robert Wood Johnson Medical School, Piscataway, NJ, USA
| | - Qingrong Huang
- Department of Food Science, Rutgers University, New Brunswick, NJ, USA
| | - Shengkan Jin
- Department of Pharmacology, Rutgers University-Robert Wood Johnson Medical School, Piscataway, NJ, USA.
| |
Collapse
|
36
|
Berdeaux R, Hutchins C. Anabolic and Pro-metabolic Functions of CREB-CRTC in Skeletal Muscle: Advantages and Obstacles for Type 2 Diabetes and Cancer Cachexia. Front Endocrinol (Lausanne) 2019; 10:535. [PMID: 31428057 PMCID: PMC6688074 DOI: 10.3389/fendo.2019.00535] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 07/18/2019] [Indexed: 12/31/2022] Open
Abstract
cAMP is one of the earliest described mediators of hormone action in response to physiologic stress that allows acute stress responses and adaptation in every tissue. The classic role of cAMP signaling in metabolic tissues is to regulate nutrient partitioning. In response to acute stress, such as epinephrine released during strenuous exercise or fasting, intramuscular cAMP liberates glucose from glycogen and fatty acids from triglycerides. In the long-term, activation of Gs-coupled GPCRs stimulates muscle growth (hypertrophy) and metabolic adaptation through multiple pathways that culminate in a net increase of protein synthesis, mitochondrial biogenesis, and improved metabolic efficiency. This review focuses on regulation, function, and transcriptional targets of CREB (cAMP response element binding protein) and CRTCs (CREB regulated transcriptional coactivators) in skeletal muscle and the potential for targeting this pathway to sustain muscle mass and metabolic function in type 2 diabetes and cancer. Although the muscle-autonomous roles of these proteins might render them excellent targets for both conditions, pharmacologic targeting must be approached with caution. Gain of CREB-CRTC function is associated with excess liver glucose output in type 2 diabetes, and growing evidence implicates CREB-CRTC activation in proliferation and invasion of different types of cancer cells. We conclude that deeper investigation to identify skeletal muscle specific regulatory mechanisms that govern CREB-CRTC transcriptional activity is needed to safely take advantage of their potent effects to invigorate skeletal muscle to potentially improve health in people with type 2 diabetes and cancer.
Collapse
Affiliation(s)
- Rebecca Berdeaux
- Department of Integrative Biology and Pharmacology, Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center Houston, Houston, TX, United States
- Graduate Program in Biochemistry and Cell Biology, The MD Anderson-UTHealth Graduate School of Biomedical Sciences, Houston, TX, United States
- *Correspondence: Rebecca Berdeaux
| | - Chase Hutchins
- Department of Integrative Biology and Pharmacology, Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center Houston, Houston, TX, United States
| |
Collapse
|
37
|
Srivastava RAK. Life-style-induced metabolic derangement and epigenetic changes promote diabetes and oxidative stress leading to NASH and atherosclerosis severity. J Diabetes Metab Disord 2018; 17:381-391. [PMID: 30918873 DOI: 10.1007/s40200-018-0378-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 11/20/2018] [Indexed: 12/15/2022]
Abstract
Energy imbalance resulting from high calorie food intake and insufficient metabolic activity leads to increased body mass index (BMI) and sets the stage for metabolic derangement influencing lipid and carbohydrate metabolism and ultimately leading to insulin resistance, dyslipidemia, and type 2 diabetes. 70% of cardiovascular disease (CVD) deaths occur in patients with diabetes. Environment-induced physiological perturbations trigger epigenetic changes through chromatin modification and leads to type 2 diabetes and progression of nonalcoholic fatty liver disease (NAFLD) and CVD. Thus, in terms of disease progression and pathogenesis, energy homeostasis, metabolic dysregulation, diabetes, fatty liver, and CVD are interlinked. Since advanced glycation end products (AGEs) and low-grade inflammation in type 2 diabetes play definitive roles in the pathogenesis of liver and vascular diseases, a natural checkpoint to prevent diabetes and associated complications appears to be the identification and management of prediabetes together with weight management, since 70% of prediabetic individuals develop diabetes during their life time, and every kg of weight increase is associated with up to 9% increase in diabetes risk. A good proportion of diabetes and obesity population have fatty liver that progresses to non-alcoholic steatohepatitis (NASH) and cirrhosis, and increased risk of hepatocellular carcinoma. Diabetes and NASH both have elevated oxidative stress, impaired cholesterol elimination, and increased inflammation that leads to CVD risk. This review addresses life-style-induced metabolic pathway derangement and how it contributes to epigenetic changes, type 2 diabetes and NASH progression, which collectively lead to increased risk of CVD.
Collapse
Affiliation(s)
- Rai Ajit K Srivastava
- Integrated Pharma Solutions, Philadelphia, PA USA.,2Department of Nutrition, Wayne State University, Detroit, MI USA
| |
Collapse
|
38
|
Bódis K, Roden M. Energy metabolism of white adipose tissue and insulin resistance in humans. Eur J Clin Invest 2018; 48:e13017. [PMID: 30107041 DOI: 10.1111/eci.13017] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 07/22/2018] [Accepted: 08/12/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Insulin resistance not only occurs in obesity, but also in lipodystrophy. Although adipose tissue mass affects metabolic fluxes and participates in interorgan crosstalk, the role of energy metabolism within white adipose tissue for insulin resistance is less clear. MATERIALS AND METHODS A Medline search identified in vivo studies in humans on energy and lipid metabolism in subcutaneous (SAT) and visceral adipose tissue (VAT). Studies in adipocyte cultures and transgenic animal models were included for the better understanding of the link between abnormal energy metabolism in adipose tissue and insulin resistance. RESULTS The current literature indicates that higher lipolysis and lower lipogenesis in VAT compared to SAT enhance portal delivery of lipid metabolites (glycerol and fatty acids) to the liver. Thus, the lower lipolysis and higher lipogenesis in SAT favour storage of excess lipids and allow for protection of insulin-sensitive tissues from lipotoxic effects. In insulin-resistant humans, enhanced lipolysis and impaired lipogenesis in adipose tissue lead to release of cytokines and lipid metabolites, ultimately promoting insulin resistance. Adipose tissue of insulin-resistant humans also displays lower expression of proteins involved in mitochondrial function. In turn, this leads to lower availability of mitochondria-derived energy sources for lipogenesis in adipose tissue. CONCLUSIONS Abnormal mitochondrial function in human white adipose tissue likely contributes to the secretion of lipid metabolites and lactate, which are linked to insulin resistance in peripheral tissues. However, the relevance of adipose tissue energy metabolism for the regulation of human insulin sensitivity remains to be further elucidated.
Collapse
Affiliation(s)
- Kálmán Bódis
- Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany.,Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Michael Roden
- Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany.,Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| |
Collapse
|
39
|
Yang Y, Sadri H, Prehn C, Adamski J, Rehage J, Dänicke S, Saremi B, Sauerwein H. Acylcarnitine profiles in serum and muscle of dairy cows receiving conjugated linoleic acids or a control fat supplement during early lactation. J Dairy Sci 2018; 102:754-767. [PMID: 30343917 DOI: 10.3168/jds.2018-14685] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 08/24/2018] [Indexed: 12/21/2022]
Abstract
Acylcarnitines (ACC) are formed when fatty acid (FA)-coenzyme A enters the mitochondria for β-oxidation and the tricarboxylic acid cycle through the carnitine shuttle. Concentrations of ACC may vary depending on the metabolic conditions, but can accumulate when rates of β-oxidation exceed those of tricarboxylic acid. This study aimed to characterize muscle and blood serum acylcarnitine profiles, to determine the mRNA abundance of muscle carnitine acyltransferases, and to test whether dietary supplementation (from d 1 in milk) with conjugated linoleic acids (CLA; 100 g/d; each 12% of trans-10,cis-12 and cis-9,trans-11 CLA; n = 11) altered these compared with control fat-supplemented cows (CTR; n = 10). Blood samples and biopsies from the semitendinosus musclewere collected on d -21, 1, 21, and 70 relative to parturition. Serum and muscle ACC profiles were quantified using a targeted metabolomics approach. The CLA supplement did not affect the variables examined. The serum concentration of free carnitine decreased with the onset of lactation. The concentrations of acetylcarnitine, hydroxybutyrylcarnitine, and the sum of short-chain ACC in serum were greater from d -21 to 21 than thereafter. The serum concentrations of long-chain ACC tetradecenoylcarnitine (C14:1) and octadecenoylcarnitine (C18:1) concentrations were greater on d 1 and 21 compared with d -21. Muscle carnitine remained unchanged, whereas short- and medium-chain ACC, including propenoylcarnitine (C3:1), hydroxybutyrylcarnitine, hydroxyhexanoylcarnitine, hexenoylcarnitine (C6:1), and pimelylcarnitine were increased on d 21 compared with d -21 and decreased thereafter. In muscle, the concentrations of long-chain ACC (from C14 to C18) were elevated on d 1. The mRNA abundance of carnitine palmitoyltransferase 1, muscle isoform (CPT1B) increased 2.8-fold from d -21 to 1, followed by a decline to nearly prepartum values by d 70, whereas that of CPT2 did not change over time. The majority of serum and muscle short- and long-chain ACC were positively correlated with the FA concentrations in serum, whereas serum carnitine and C5 were negatively correlated with FA. Time-related changes in the serum and muscle ACC profiles were demonstrated that were not affected by the CLA supplement at the dosage used in the present study. The elevated concentrations of long-chain ACC species in muscle and of serum acetylcarnitine around parturition point to incomplete FA oxidation were likely due to insufficient metabolic adaptation in response to the load of FA around parturition.
Collapse
Affiliation(s)
- Y Yang
- Institute of Animal Science, Physiology and Hygiene Unit, University of Bonn, 53115 Bonn, Germany
| | - H Sadri
- Department of Clinical Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz 5166616471, Iran.
| | - C Prehn
- Institute of Experimental Genetics, Genome Analysis Center, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - J Adamski
- Institute of Experimental Genetics, Genome Analysis Center, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany; Lehrstuhl für Experimentelle Genetik, Technische Universität München, Freising-Weihenstephan 85350, Germany; German Center for Diabetes Research (DZD), München-Neuherberg 85764, Germany
| | - J Rehage
- Clinic for Cattle, University for Veterinary Medicine, Foundation, 30173 Hannover, Germany
| | - S Dänicke
- Institute of Animal Nutrition, Friedrich-Loeffler-Institute (FLI), 38116 Braunschweig, Germany
| | - B Saremi
- Evonik Nutrition & Care GmbH, Rodenbacher Chaussee 4, 63457 Hanau, Germany
| | - H Sauerwein
- Institute of Animal Science, Physiology and Hygiene Unit, University of Bonn, 53115 Bonn, Germany
| |
Collapse
|
40
|
Bódis K, Kahl S, Simon MC, Zhou Z, Sell H, Knebel B, Tura A, Strassburger K, Burkart V, Müssig K, Markgraf D, Al-Hasani H, Szendroedi J, Roden M. Reduced expression of stearoyl-CoA desaturase-1, but not free fatty acid receptor 2 or 4 in subcutaneous adipose tissue of patients with newly diagnosed type 2 diabetes mellitus. Nutr Diabetes 2018; 8:49. [PMID: 30190473 PMCID: PMC6127327 DOI: 10.1038/s41387-018-0054-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Revised: 07/23/2018] [Accepted: 08/03/2018] [Indexed: 12/27/2022] Open
Abstract
Background In subcutaneous adipose tissue (SAT), higher stearoyl-CoA desaturase-1 (SCD1) expression has been related to improved insulin sensitivity in thiazolidinedione-treated type 2 diabetes mellitus patients. In animal models, deficiency of the free fatty acid receptor (FFAR) 2 associated with higher and FFAR4-deficiency with lower insulin sensitivity. We hypothesized that increased FFAR2 expression and reductions in FFAR4 and SCD1 expression in SAT of type 2 diabetes mellitus patients associate positively with insulin resistance and impaired beta cell function. Methods Twenty-five type 2 diabetes mellitus patients and 25 glucose-tolerant humans (CON) matched for sex, age, and BMI underwent mixed-meal tests to assess insulin sensitivity (OGIS) and beta cell function (ΔAUC(C-peptide)0–180 min/ΔAUC(glucose)0–180 min) in a cross-sectional study. Gene and protein expression of SCD1 and FFAR2/4 were quantified in SAT biopsies. Results Insulin sensitivity was 14% and beta cell function 71% (both p < 0.001) lower in type 2 diabetes mellitus patients. In type 2 diabetes mellitus, SCD1 mRNA was fivefold (p < 0.001) and protein expression twofold (p < 0.01) lower. While FFAR2/4 mRNA and protein expression did not differ between groups, FFAR2 protein levels correlated negatively with beta cell function only in CON (r = −0.74, p < 0.01). However, neither SCD1 nor FFAR2/4 protein expression correlated with insulin sensitivity in both groups. Conclusions Type 2 diabetes patients have lower SCD1, which does not associate with insulin resistance. Only in non-diabetic humans, FFAR2 associated with impaired beta cell function.
Collapse
Affiliation(s)
- Kálmán Bódis
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Sabine Kahl
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Marie-Christine Simon
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Zhou Zhou
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany.,Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at the Heinrich Heine University, Düsseldorf, Germany
| | - Henrike Sell
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Birgit Knebel
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany.,Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at the Heinrich Heine University, Düsseldorf, Germany
| | - Andrea Tura
- Metabolic Unit, Institute of Biomedical Engineering, National Research Council, Padua, Italy
| | - Klaus Strassburger
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany.,Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
| | - Volker Burkart
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Karsten Müssig
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany.,Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Daniel Markgraf
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Hadi Al-Hasani
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany.,Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at the Heinrich Heine University, Düsseldorf, Germany
| | - Julia Szendroedi
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany.,Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Michael Roden
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany. .,German Center for Diabetes Research (DZD), München-Neuherberg, Germany. .,Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany.
| | | |
Collapse
|
41
|
Yuan F, Song B, Huang Z, Xia C, Liu X. Quantification of pancreatic fat with dual-echo imaging at 3.0-T MR in clinical application: how do the corrections for T1 and T2* relaxation effect work and simplified correction strategy. Acta Radiol 2018; 59:1021-1028. [PMID: 29260576 DOI: 10.1177/0284185117745908] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Background Dual-echo imaging is a routine clinical magnetic resonance (MR) sequence affected by T1 and T2* relaxation effect in fat quantification. The separate impacts of T1 and T2* relaxation effect in pancreatic fat quantification using dual-echo imaging at 3.0-T MR have not been reported in detail. Purpose To demonstrate the separate T1 and T2* relaxation effect on pancreatic fat quantification by dual-echo imaging at 3.0-T MR and the simplified correction strategy is discussed for convenient clinical application. Material and Methods Twenty-one non-alcoholic fatty liver disease (NAFLD) participants with high risk of pancreatic steatosis were included. Pancreatic fat fractions (FF) by dual-echo imaging with different corrections were compared to that of proton magnetic resonance spectroscopy (1H-MRS). Correlation analysis and Bland-Altman analysis were applied. Results The FF by 1H-MRS was 5.9 ± 1.7%. Significant positive correlation (all P < 0.01) was found between FF by 1H-MRS and each dual-echo imaging, in which T1 and T2* correction showed the best correlation (r = 0.95, FF = 6.2 ± 1.7%) and no correction showed the worst correlation (r = 0.86, FF = 5.2 ± 2.0%), and the simplified T1 and T2* correction manifested as r = 0.93 and FF = 6.3 ± 1.8%. FF by T1 and T2* correction showed the best agreement, while T1 correction showed the worst agreement as compared to that of 1H-MRS. Conclusion T1 and T2* correction shows the best performance while no correction dual-echo imaging remains clinical available which may benefit from prior OP echo. Simplified correction using single T2* (32.6 ms) of water and fat is recommended for convenient clinical application in absence of obvious pancreatic iron overload.
Collapse
Affiliation(s)
- Fang Yuan
- Department of Radiology, West China Hospital of Sichuan University, Chengdu, PR China
| | - Bin Song
- Department of Radiology, West China Hospital of Sichuan University, Chengdu, PR China
| | - Zixing Huang
- Department of Radiology, West China Hospital of Sichuan University, Chengdu, PR China
| | - Chunchao Xia
- Department of Radiology, West China Hospital of Sichuan University, Chengdu, PR China
| | - Xijiao Liu
- Department of Radiology, West China Hospital of Sichuan University, Chengdu, PR China
| |
Collapse
|
42
|
Tallis J, James RS, Seebacher F. The effects of obesity on skeletal muscle contractile function. ACTA ACUST UNITED AC 2018; 221:221/13/jeb163840. [PMID: 29980597 DOI: 10.1242/jeb.163840] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Obesity can cause a decline in contractile function of skeletal muscle, thereby reducing mobility and promoting obesity-associated health risks. We reviewed the literature to establish the current state-of-knowledge of how obesity affects skeletal muscle contraction and relaxation. At a cellular level, the dominant effects of obesity are disrupted calcium signalling and 5'-adenosine monophosphate-activated protein kinase (AMPK) activity. As a result, there is a shift from slow to fast muscle fibre types. Decreased AMPK activity promotes the class II histone deacetylase (HDAC)-mediated inhibition of the myocyte enhancer factor 2 (MEF2). MEF2 promotes slow fibre type expression, and its activity is stimulated by the calcium-dependent phosphatase calcineurin. Obesity-induced attenuation of calcium signalling via its effects on calcineurin, as well as on adiponectin and actinin affects excitation-contraction coupling and excitation-transcription coupling in the myocyte. These molecular changes affect muscle contractile function and phenotype, and thereby in vivo and in vitro muscle performance. In vivo, obesity can increase the absolute force and power produced by increasing the demand on weight-supporting muscle. However, when normalised to body mass, muscle performance of obese individuals is reduced. Isolated muscle preparations show that obesity often leads to a decrease in force produced per muscle cross-sectional area, and power produced per muscle mass. Obesity and ageing have similar physiological consequences. The synergistic effects of obesity and ageing on muscle function may exacerbate morbidity and mortality. Important future research directions include determining: the relationship between time course of weight gain and changes in muscle function; the relative effects of weight gain and high-fat diet feeding per se; the effects of obesity on muscle function during ageing; and if the effects of obesity on muscle function are reversible.
Collapse
Affiliation(s)
- Jason Tallis
- Center for Sport, Exercise and Life Sciences, Science and Health Building, Coventry University, Priory Street, Coventry CV1 5FB, UK
| | - Rob S James
- Center for Sport, Exercise and Life Sciences, Science and Health Building, Coventry University, Priory Street, Coventry CV1 5FB, UK
| | - Frank Seebacher
- School of Life and Environmental Sciences, Heydon Laurence Building A08, University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
43
|
Xu J, Zhang M, Zhang X, Yang H, Sun B, Wang Z, Zhou Y, Wang S, Liu X, Liu L. Contribution of Hepatic Retinaldehyde Dehydrogenase Induction to Impairment of Glucose Metabolism by High-Fat-Diet Feeding in C57BL/6J Mice. Basic Clin Pharmacol Toxicol 2018; 123:539-548. [PMID: 29753302 DOI: 10.1111/bcpt.13039] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Accepted: 04/27/2018] [Indexed: 11/27/2022]
Abstract
Obesity and insulin resistance are associated with overexpression of retinaldehyde dehydrogenase 1 (RALDH1). We aimed to investigate the roles of hepatic RALDH1 induction in glucose metabolism impairment using mice fed with high-fat-diet (HFD). Mice were fed with HFD for 8 weeks and treated with RALDH inhibitor citral for another 4 weeks. Oral glucose tolerance test (OGTT), pyruvate tolerance test (PTT) and insulin tolerance test were performed. Expressions of phosphoenolpyruvate carboxykinase 1 (PCK1), glucokinase (GCK) and RALDH1 were measured. Therapeutic effects of citral were also documented in diabetic rats. Effects of retinaldehyde on PCK1 and GCK expressions were examined in rat primary hepatocytes and HepG2 cells. The results showed that HFD mice were characterized by hyperlipidaemia and insulin resistance, accompanied by significantly increased RALDH1 activity and expression. Citral (10 and 50 mg/kg) ameliorated HFD-induced hyperlipidaemia and insulin resistance, as demonstrated by the improved fasting glucose, insulin levels and lipid profiles. OGTT and PTT demonstrated that citral reversed HFD-induced glucose disposal impairment and glucose production enhancement. Citral also reversed the increased PCK1 expression and decreased GCK expression by HFD. Citral therapeutic effects were reconfirmed in diabetic rats. In vitro data indicated that retinaldehyde had the strongest PCK1 induction in primary hepatocytes of diabetic rats compared with HFD rats and control rats, in line with the increased RALDH1 expression. Citral reversed the retinaldehyde-induced PCK1 expression in primary rat hepatocytes and HepG2 cells. In conclusion, RALDH1 induction impaired glucose metabolism partly via modulating PCK1 and GCK expressions. Citral improved glucose metabolism through inhibiting RALDH activity.
Collapse
Affiliation(s)
- Jiong Xu
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Mian Zhang
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xiangping Zhang
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Hanyu Yang
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Binbin Sun
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Zhongjian Wang
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yaqian Zhou
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Shuting Wang
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xiaodong Liu
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Li Liu
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
44
|
Lu FB, Hu ED, Xu LM, Chen L, Wu JL, Li H, Chen DZ, Chen YP. The relationship between obesity and the severity of non-alcoholic fatty liver disease: systematic review and meta-analysis. Expert Rev Gastroenterol Hepatol 2018; 12:491-502. [PMID: 29609501 DOI: 10.1080/17474124.2018.1460202] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
A number of researches have explored the association between obesity and nonalcoholic fatty liver disease (NAFLD) liver function, histopathology, complications, genetic factors and prognosis, but the results were conflicting and inconclusive. Areas covered: In this meta-analysis, the liver function, histopathology, metabolic complications, patatin-like phospholipase domain-containing protein 3 (PNPLA3) genetic polymorphism and prognosis were compared between non-obese and obese NAFLD. Pubmed, EMBASE, Cochrane databases were searched to identify eligible studies. The odds ratio (OR) or standardized mean difference (SMD) with 95% confidence intervals (CI) were pooled using fixed- or random-effects models. Expert commentary: This meta-analysis indicated that for NAFLD patients, obesity (according to ethnic-specific BMI cut-off points to define obesity) could predict a worse long-term prognosis. However, obesity may not be an independent factor for the development of NASH or advanced fibrosis in NAFLD patients and NAFLD should be considered as potential population for pharmacologic treatment regardless of obesity. In addition, PNPLA3 rs738409 may be more relevant to the progression of non-obese NAFLD when compared to obese NAFLD. Importantly, large-sample, long-term follow-up cohort studies based on liver biopsy are highly needed due to limited liver pathology and long-term follow-up data at present.
Collapse
Affiliation(s)
- Feng-Bin Lu
- a Department of Infectious Diseases , the First Affiliated Hospital of Wenzhou Medical University, Hepatology Institute of Wenzhou Medical University, Wenzhou Key Laboratory of Hepatology , Wenzhou , China
| | - En-De Hu
- a Department of Infectious Diseases , the First Affiliated Hospital of Wenzhou Medical University, Hepatology Institute of Wenzhou Medical University, Wenzhou Key Laboratory of Hepatology , Wenzhou , China
| | - Lan-Man Xu
- a Department of Infectious Diseases , the First Affiliated Hospital of Wenzhou Medical University, Hepatology Institute of Wenzhou Medical University, Wenzhou Key Laboratory of Hepatology , Wenzhou , China
| | - Lu Chen
- a Department of Infectious Diseases , the First Affiliated Hospital of Wenzhou Medical University, Hepatology Institute of Wenzhou Medical University, Wenzhou Key Laboratory of Hepatology , Wenzhou , China
| | - Jin-Lu Wu
- a Department of Infectious Diseases , the First Affiliated Hospital of Wenzhou Medical University, Hepatology Institute of Wenzhou Medical University, Wenzhou Key Laboratory of Hepatology , Wenzhou , China
| | - Hui Li
- a Department of Infectious Diseases , the First Affiliated Hospital of Wenzhou Medical University, Hepatology Institute of Wenzhou Medical University, Wenzhou Key Laboratory of Hepatology , Wenzhou , China
| | - Da-Zhi Chen
- b State Key Laboratory of Infectious Diseases , Medicine School of Zhejiang University , Hangzhou , China
| | - Yong-Ping Chen
- a Department of Infectious Diseases , the First Affiliated Hospital of Wenzhou Medical University, Hepatology Institute of Wenzhou Medical University, Wenzhou Key Laboratory of Hepatology , Wenzhou , China
| |
Collapse
|
45
|
Goedecke JH, Mendham AE, Clamp L, Nono Nankam PA, Fortuin-de Smidt MC, Phiri L, Micklesfield LK, Keswell D, Woudberg NJ, Lecour S, Alhamud A, Kaba M, Lutomia FM, van Jaarsveld PJ, de Villiers A, Kahn SE, Chorell E, Hauksson J, Olsson T. An Exercise Intervention to Unravel the Mechanisms Underlying Insulin Resistance in a Cohort of Black South African Women: Protocol for a Randomized Controlled Trial and Baseline Characteristics of Participants. JMIR Res Protoc 2018; 7:e75. [PMID: 29669711 PMCID: PMC5932332 DOI: 10.2196/resprot.9098] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 11/20/2017] [Accepted: 11/20/2017] [Indexed: 11/13/2022] Open
Abstract
Background The pathogenesis of type 2 diabetes (T2D) in black African women is complex and differs from that in their white counterparts. However, earlier studies have been cross-sectional and provide little insight into the causal pathways. Exercise training is consistently used as a model to examine the mechanisms underlying insulin resistance and risk for T2D. Objective The objective of the study was to examine the mechanisms underlying the changes in insulin sensitivity and secretion in response to a 12-week exercise intervention in obese black South African (SA) women. Methods A total of 45 obese (body mass index, BMI: 30-40 kg/m2) black SA women were randomized into a control (n=22) or experimental (exercise; n=23) group. The exercise group completed 12 weeks of supervised combined aerobic and resistance training (40-60 min, 4 days/week), while the control group maintained their typical physical activity patterns, and both groups were requested not to change their dietary patterns. Before and following the 12-week intervention period, insulin sensitivity and secretion (frequently sampled intravenous glucose tolerance test) and its primary and secondary determinants were measured. Dietary intake, sleep quality and quantity, physical activity, and sedentary behaviors were measured every 4 weeks. Results The final sample included 20 exercise and 15 control participants. Baseline sociodemographics, cardiorespiratory fitness, anthropometry, cardiometabolic risk factors, physical activity, and diet did not differ between the groups (P>.05). Conclusions The study describes a research protocol for an exercise intervention to understand the mechanisms underlying insulin sensitivity and secretion in obese black SA women and aims to identify causal pathways underlying the high prevalence of insulin resistance and risk for T2D in black SA women, targeting specific areas for therapeutic intervention. Trial Registration Pan African Clinical Trial Registry PACTR201711002789113; http://www.pactr.org/ATMWeb/ appmanager/atm/atmregistry?_nfpb=true&_pageLabel=portals_app_atmregistry_portal_page_13 (Archived by WebCite at http://www.webcitation.org/6xLEFqKr0)
Collapse
Affiliation(s)
- Julia H Goedecke
- Non-Communicable Diseases Research Unit, South African Medical Research Council, Cape Town, South Africa.,Division of Exercise Science and Sports Medicine, Department of Human Biology, University of Cape Town, Cape Town, South Africa
| | - Amy E Mendham
- Non-Communicable Diseases Research Unit, South African Medical Research Council, Cape Town, South Africa.,Division of Exercise Science and Sports Medicine, Department of Human Biology, University of Cape Town, Cape Town, South Africa
| | - Louise Clamp
- Division of Exercise Science and Sports Medicine, Department of Human Biology, University of Cape Town, Cape Town, South Africa
| | - Pamela A Nono Nankam
- Division of Exercise Science and Sports Medicine, Department of Human Biology, University of Cape Town, Cape Town, South Africa
| | - Melony C Fortuin-de Smidt
- Division of Exercise Science and Sports Medicine, Department of Human Biology, University of Cape Town, Cape Town, South Africa
| | - Lindokuhle Phiri
- Division of Exercise Science and Sports Medicine, Department of Human Biology, University of Cape Town, Cape Town, South Africa
| | - Lisa K Micklesfield
- Division of Exercise Science and Sports Medicine, Department of Human Biology, University of Cape Town, Cape Town, South Africa.,South African Medical Research Council / University of the Witwatersrand Developmental Pathways for Health Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Gauteng, South Africa
| | - Dheshnie Keswell
- Division of Exercise Science and Sports Medicine, Department of Human Biology, University of Cape Town, Cape Town, South Africa
| | - Nicholas J Woudberg
- Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Sandrine Lecour
- Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Ali Alhamud
- Division of Biomedical Engineering, Department of Human Biology, University of Cape Town, Cape Town, South Africa
| | - Mamadou Kaba
- Division of Medical Microbiology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Faith M Lutomia
- Division of Medical Microbiology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Paul J van Jaarsveld
- Non-Communicable Diseases Research Unit, South African Medical Research Council, Cape Town, South Africa.,Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Anniza de Villiers
- Non-Communicable Diseases Research Unit, South African Medical Research Council, Cape Town, South Africa
| | - Steven E Kahn
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, Veterans Affairs Puget Sound Health Care System and University of Washington, Seattle, WA, United States
| | - Elin Chorell
- Department of Public Health and Clinical Medicine, Umeå University, Umea, Sweden
| | - Jon Hauksson
- Department of Radiation Sciences, Umeå University, Umeå, Sweden
| | - Tommy Olsson
- Department of Public Health and Clinical Medicine, Umeå University, Umea, Sweden
| |
Collapse
|
46
|
Wu W, Xu Z, Zhang L, Liu J, Feng J, Wang X, Shan T, Wang Y. Muscle-specific deletion of Prkaa1 enhances skeletal muscle lipid accumulation in mice fed a high-fat diet. J Physiol Biochem 2017; 74:195-205. [DOI: 10.1007/s13105-017-0604-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 12/20/2017] [Indexed: 12/31/2022]
|
47
|
Associations of Mitochondrial Fatty Acid Oxidation with Body Fat in Premenopausal Women. J Nutr Metab 2017; 2017:7832057. [PMID: 29204295 PMCID: PMC5674507 DOI: 10.1155/2017/7832057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 09/07/2017] [Accepted: 09/17/2017] [Indexed: 11/24/2022] Open
Abstract
Higher in vivo fatty acid (FA) oxidation rates have been reported in obese individuals compared to lean counterparts; however whether this reflects a shift in substrate-specific oxidative capacity at the level of the skeletal muscle mitochondria has not been examined. The purpose of this study was to test the hypothesis that in situ measures of skeletal muscle mitochondria FA oxidation would be positively associated with total body fat. Participants were 38 premenopausal women (BMI = 26.5 ± 4.3 kg/m2). Total and regional fat were assessed by dual-energy X-ray absorptiometry (DXA). Mitochondrial FA oxidation was assessed in permeabilized myofibers using high-resolution respirometry and a palmitoyl carnitine substrate. We found positive associations of total fat mass with State 3 (ADP-stimulated respiration) (r = 0.379, p < 0.05) and the respiratory control ratio (RCR, measure of mitochondrial coupling) (r = 0.348, p < 0.05). When participants were dichotomized by high or low body fat percent, participants with high total body fat displayed a higher RCR compared to those with low body fat (p < 0.05). There were no associations between any measure of regional fat and mitochondrial FA oxidation independent of total fat mass. In conclusion, greater FA oxidation in obesity may reflect molecular processes that enhance FA oxidation capacity at the mitochondrial level.
Collapse
|
48
|
Ribel-Madsen A, Ribel-Madsen R, Brøns C, Newgard CB, Vaag AA, Hellgren LI. Plasma acylcarnitine profiling indicates increased fatty acid oxidation relative to tricarboxylic acid cycle capacity in young, healthy low birth weight men. Physiol Rep 2017; 4:4/19/e12977. [PMID: 27694528 PMCID: PMC5064135 DOI: 10.14814/phy2.12977] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 08/24/2016] [Indexed: 01/07/2023] Open
Abstract
We hypothesized that an increased, incomplete fatty acid beta‐oxidation in mitochondria could be part of the metabolic events leading to insulin resistance and thereby an increased type 2 diabetes risk in low birth weight (LBW) compared with normal birth weight (NBW) individuals. Therefore, we measured fasting plasma levels of 45 acylcarnitine species in 18 LBW and 25 NBW men after an isocaloric control diet and a 5‐day high‐fat, high‐calorie diet. We demonstrated that LBW men had higher C2 and C4‐OH levels after the control diet compared with NBW men, indicating an increased fatty acid beta‐oxidation relative to the tricarboxylic acid cycle flux. Also, they had higher C6‐DC, C10‐OH/C8‐DC, and total hydroxyl‐/dicarboxyl‐acylcarnitine levels, which may suggest an increased fatty acid omega‐oxidation in the liver. Furthermore, LBW and NBW men decreased several acylcarnitine levels in response to overfeeding, which is likely a result of an upregulation of fatty acid oxidation due to the dietary challenge. Moreover, C10‐OH/C8‐DC and total hydroxyl‐/dicarboxyl‐acylcarnitine levels tended to be negatively associated with the serum insulin level, and the total hydroxyl‐/dicarboxyl‐acylcarnitine level additionally tended to be negatively associated with the hepatic insulin resistance index. This indicates that an increased fatty acid omega‐oxidation could be a compensatory mechanism to prevent an accumulation of lipid species that impair insulin signaling.
Collapse
Affiliation(s)
- Amalie Ribel-Madsen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark Department of Endocrinology, Diabetes and Metabolism, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Rasmus Ribel-Madsen
- Department of Endocrinology, Diabetes and Metabolism, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark Danish Diabetes Academy, Odense, Denmark
| | - Charlotte Brøns
- Department of Endocrinology, Diabetes and Metabolism, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Christopher B Newgard
- Sarah W. Stedman Nutrition and Metabolism, Center and Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| | - Allan A Vaag
- Department of Endocrinology, Diabetes and Metabolism, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Lars I Hellgren
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| |
Collapse
|
49
|
Park JE, Lee JH, Han JS. Sargassum yezoense Extract Inhibits Carbohydrate Digestive Enzymes In Vitro and Alleviates Postprandial Hyperglycemia in Diabetic Mice. Prev Nutr Food Sci 2017; 22:166-171. [PMID: 29043213 PMCID: PMC5642797 DOI: 10.3746/pnf.2017.22.3.166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 04/21/2017] [Indexed: 11/28/2022] Open
Abstract
In this study, we investigated whether Sargassum yezoense extract (SYE) could inhibit α-glucosidase and α-amylase activities, and alleviate postprandial hyperglycemia in streptozotocin (STZ)-induced diabetic mice. Freeze-dried S. yezoense was extracted with 80% ethanol and concentrated for use in this study. The hypoglycemic effect was determined by evaluating the inhibitory activities of SYE against α-glucosidase and α-amylase as well as its ability to decrease postprandial blood glucose levels. The half-maximal inhibitory concentrations of SYE against α-glucosidase and α-amylase were 0.078±0.004 and 0.212±0.064 mg/mL, respectively. SYE was a more effective inhibitor of α-glucosidase and α-amylase activities than the positive control, acarbose. The increase in postprandial blood glucose levels was significantly alleviated in the SYE group compared with that in the control group of STZ-induced diabetic mice. Furthermore, the area under the curves significantly decreased with SYE administration in STZ-induced diabetic mice. These results suggest that SYE is a potent inhibitor of α-glucosidase and α-amylase activities and alleviates postprandial hyperglycemia caused by dietary carbohydrates.
Collapse
Affiliation(s)
- Jae-Eun Park
- Department of Food Science and Nutrition, Pusan National University, Busan 46241, Korea
| | - Ji-Hee Lee
- Department of Food Science and Nutrition, Pusan National University, Busan 46241, Korea
| | - Ji-Sook Han
- Department of Food Science and Nutrition, Pusan National University, Busan 46241, Korea
| |
Collapse
|
50
|
Bhaskaran S, Unnikrishnan A, Ranjit R, Qaisar R, Pharaoh G, Matyi S, Kinter M, Deepa SS. A fish oil diet induces mitochondrial uncoupling and mitochondrial unfolded protein response in epididymal white adipose tissue of mice. Free Radic Biol Med 2017; 108:704-714. [PMID: 28455142 DOI: 10.1016/j.freeradbiomed.2017.04.028] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 04/19/2017] [Accepted: 04/20/2017] [Indexed: 01/14/2023]
Abstract
White adipose tissue (WAT) mitochondrial dysfunction is linked to the pathogenesis of obesity driven insulin resistance. Dietary conditions that alter fat mass are known to affect white adipocyte mitochondrial function, however, the impact of high calorie diets on white adipocyte mitochondria is not fully understood. The aim of this study is to assess the effect of a diet rich in saturated or polyunsaturated fat on mitochondrial unfolded protein response (UPRmt), a retrograde signaling response that maintains mitochondrial homeostasis, in epididymal WAT (eWAT). Mice were fed a low fat diet (LFD), saturated fat diet (SFD) or fish oil (unsaturated fat diet, UFD) and assessed changes in eWAT mitochondria. Compared to mice fed a LFD, SFD-fed mice have reduced mitochondrial biogenesis markers, mitochondrial fatty acid oxidation enzymes and TCA cycle enzymes, suggesting an impaired mitochondrial function that could contribute to increased fat mass. In contrast, isocaloric UFD-fed mice have increased expression of mitochondrial uncoupling protein 1 (UCP1) and peroxisomal fatty acid oxidation enzymes suggesting that elevated mitochondrial uncoupling and peroxisomal fatty acid oxidation could contribute to the reduction in fat mass. Interestingly, expression of UPRmt-associated proteins caseinolytic peptidase (ClpP) and heat shock protein 60 (Hsp60) are induced by UFD, whereas SFD reduced the expression of ClpP. Based on our data, we propose that induction of UPRmt helps to preserve a functional mitochondria and efficient utilization of fat by UFD whereas a dampened UPRmt response might impair mitochondrial function and promote fat accumulation by SFD. Thus, our findings suggest a potential role of UPRmt in mediating the beneficial effects of fish oil.
Collapse
Affiliation(s)
- Shylesh Bhaskaran
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Archana Unnikrishnan
- Department of Geriatric Medicine and the Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Rojina Ranjit
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Rizwan Qaisar
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Gavin Pharaoh
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Stephanie Matyi
- Department of Geriatric Medicine and the Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Michael Kinter
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Sathyaseelan S Deepa
- Department of Geriatric Medicine and the Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA.
| |
Collapse
|