1
|
Lian X, Tang X. Use of a ferroptosis-related gene signature to construct diagnostic and prognostic models for assessing immune infiltration in metabolic dysfunction-associated fatty liver disease. Front Cell Dev Biol 2023; 11:1199846. [PMID: 37928903 PMCID: PMC10622674 DOI: 10.3389/fcell.2023.1199846] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 09/22/2023] [Indexed: 11/07/2023] Open
Abstract
Introduction: Metabolic dysfunction-associated fatty liver disease (MAFLD), a serious health problem worldwide, can involve ferroptosis. This study aimed to comprehensively analyze the ferroptosis-related genes associated with MAFLD. Methods: Ferroptosis-related differentially expressed genes (FRDEGs) were identified in patients with MAFLD and healthy individuals. Gene ontology functional enrichment analysis, Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis, and gene set enrichment analysis (GSEA) were used to analyze the relevant action pathways of the FRDEGs. The Encyclopedia of RNA Interactomes, CHIPBase, and comparative toxicogenomics databases were used to build mRNA-miRNA, mRNA-transcription factor (TF), and mRNA-drug interaction networks, respectively. A diagnostic model was constructed and bioinformatics analysis methods, such as least absolute shrinkage and selection operator regression analysis, Cox regression analysis, nomogram-based analysis, consensus clustering analysis, and single-sample GSEA, were used to systematically investigate the prognostic values and immunologic characteristics. Results: A total of 13 FRDEGs were obtained and eight were used to construct a diagnostic model and perform a prognostic analysis. Hub genes were also used to construct mRNA-miRNA and mRNA-TF interaction networks and potential drug or molecular compounds. Two MAFLD subtypes were identified: cluster2, which represents an "immunoactive" type, and cluster1, which represents an "immunosuppressive" type; a significant correlation was observed between the immune cell contents and the expression of three FRDEGs (NR4A1, FADS2, and SCD). Conclusion: A ferroptosis-related gene signature was constructed to diagnose MAFLD-associated steatohepatitis, predict the prognosis of MAFLD patients, and analyze the immunologic characteristics of MAFLD. Our findings may provide insights into developing innovative MAFLD treatment techniques.
Collapse
Affiliation(s)
- Xin Lian
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Xulei Tang
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
2
|
Saengnipanthkul S, Noh HL, Friedline RH, Suk S, Choi S, Acosta NK, Tran DA, Hu X, Inashima K, Kim AM, Lee KW, Kim JK. Maternal exposure to high-fat diet during pregnancy and lactation predisposes normal weight offspring mice to develop hepatic inflammation and insulin resistance. Physiol Rep 2021; 9:e14811. [PMID: 33769706 PMCID: PMC7995551 DOI: 10.14814/phy2.14811] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/01/2021] [Accepted: 03/04/2021] [Indexed: 11/24/2022] Open
Abstract
Increasing evidence shows a potential link between the perinatal nutrient environment and metabolic outcome in offspring. Here, we investigated the effects of maternal feeding of a high-fat diet (HFD) during the perinatal period on hepatic metabolism and inflammation in male offspring mice at weaning and in early adulthood. Female C57BL/6 J mice were fed HFD or normal chow (NC) for 4 weeks before mating and during pregnancy and lactation. The male offspring mice were weaned onto an NC diet, and metabolic and molecular experiments were performed in early adulthood. At postnatal day 21, male offspring mice from HFD-fed dams (Off-HFD) showed significant increases in whole body fat mass and fasting levels of glucose, insulin, and cholesterol compared to male offspring mice from NC-fed dams (Off-NC). The RT-qPCR analysis showed two- to fivefold increases in hepatic inflammatory markers (MCP-1, IL-1β, and F4/80) in Off-HFD mice. Hepatic expression of G6Pase and PEPCK was elevated by fivefold in the Off-HFD mice compared to the Off-NC mice. Hepatic expression of GLUT4, IRS-1, and PDK4, as well as lipid metabolic genes, CD36, SREBP1c, and SCD1 were increased in the Off-HFD mice compared to the Off-NC mice. In contrast, CPT1a mRNA levels were reduced by 60% in the Off-HFD mice. At postnatal day 70, despite comparable body weights to the Off-NC mice, Off-HFD mice developed hepatic inflammation with increased expression of MCP-1, CD68, F4/80, and CD36 compared to the Off-NC mice. Despite normal body weight, Off-HFD mice developed insulin resistance with defects in hepatic insulin action and insulin-stimulated glucose uptake in skeletal muscle and brown fat, and these metabolic effects were associated with hepatic inflammation in Off-HFD mice. Our findings indicate hidden, lasting effects of maternal exposure to HFD during pregnancy and lactation on metabolic homeostasis of normal weight offspring mice.
Collapse
Affiliation(s)
- Suchaorn Saengnipanthkul
- Division of NutritionDepartment of PediatricsFaculty of MedicineKhon Kaen UniversityKhon KaenThailand
- Program in Molecular MedicineUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Hye Lim Noh
- Program in Molecular MedicineUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Randall H. Friedline
- Program in Molecular MedicineUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Sujin Suk
- Program in Molecular MedicineUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Stephanie Choi
- Program in Molecular MedicineUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Nicholas K. Acosta
- Program in Molecular MedicineUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Duy A. Tran
- Program in Molecular MedicineUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Xiaodi Hu
- Program in Molecular MedicineUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Kunikazu Inashima
- Program in Molecular MedicineUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Allison M. Kim
- Program in Molecular MedicineUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Ki Won Lee
- Department of Agricultural BiotechnologyCollege of Agricultural and Life SciencesSeoul National UniversitySeoulSouth Korea
| | - Jason K. Kim
- Program in Molecular MedicineUniversity of Massachusetts Medical SchoolWorcesterMAUSA
- Division of Endocrinology, Metabolism, and DiabetesDepartment of MedicineUniversity of Massachusetts Medical SchoolWorcesterMAUSA
- Department of Agricultural BiotechnologyCollege of Agricultural and Life SciencesSeoul National UniversitySeoulSouth Korea
| |
Collapse
|
3
|
Banerjee S, Ghoshal S, Stevens JR, McCommis KS, Gao S, Castro-Sepulveda M, Mizgier ML, Girardet C, Kumar KG, Galgani JE, Niehoff ML, Farr SA, Zhang J, Butler AA. Hepatocyte expression of the micropeptide adropin regulates the liver fasting response and is enhanced by caloric restriction. J Biol Chem 2020; 295:13753-13768. [PMID: 32727846 DOI: 10.1074/jbc.ra120.014381] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/22/2020] [Indexed: 12/16/2022] Open
Abstract
The micropeptide adropin encoded by the clock-controlled energy homeostasis-associated gene is implicated in the regulation of glucose metabolism. However, its links to rhythms of nutrient intake, energy balance, and metabolic control remain poorly defined. Using surveys of Gene Expression Omnibus data sets, we confirm that fasting suppresses liver adropin expression in lean C57BL/6J (B6) mice. However, circadian rhythm data are inconsistent. In lean mice, caloric restriction (CR) induces bouts of compulsive binge feeding separated by prolonged fasting intervals, increasing NAD-dependent deacetylase sirtuin-1 signaling important for glucose and lipid metabolism regulation. CR up-regulates adropin expression and induces rhythms correlating with cellular stress-response pathways. Furthermore, adropin expression correlates positively with phosphoenolpyruvate carboxokinase-1 (Pck1) expression, suggesting a link with gluconeogenesis. Our previous data suggest that adropin suppresses gluconeogenesis in hepatocytes. Liver-specific adropin knockout (LAdrKO) mice exhibit increased glucose excursions following pyruvate injections, indicating increased gluconeogenesis. Gluconeogenesis is also increased in primary cultured hepatocytes derived from LAdrKO mice. Analysis of circulating insulin levels and liver expression of fasting-responsive cAMP-dependent protein kinase A (PKA) signaling pathways also suggests enhanced responses in LAdrKO mice during a glucagon tolerance test (250 µg/kg intraperitoneally). Fasting-associated changes in PKA signaling are attenuated in transgenic mice constitutively expressing adropin and in fasting mice treated acutely with adropin peptide. In summary, hepatic adropin expression is regulated by nutrient- and clock-dependent extrahepatic signals. CR induces pronounced postprandial peaks in hepatic adropin expression. Rhythms of hepatic adropin expression appear to link energy balance and cellular stress to the intracellular signal transduction pathways that drive the liver fasting response.
Collapse
Affiliation(s)
- Subhashis Banerjee
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Sarbani Ghoshal
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Joseph R Stevens
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Kyle S McCommis
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, Missouri, USA; Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, Missouri, USA; Saint Louis University Liver Center, Saint Louis University School of Medicine, St. Louis, Missouri USA
| | - Su Gao
- Department of Metabolism and Aging, Scripps Research Institute, Jupiter, Florida, USA
| | - Mauricio Castro-Sepulveda
- Laboratorio de Ciencias del Ejercicio. Facultad de Medicina, Universidad Finis Terrae, Santiago, Chile
| | - Maria L Mizgier
- Departamento de Ciencias de la SaludCarrera de Nutrición y Dietética and Departamento de Nutrición, Diabetes y Metabolismo, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Clemence Girardet
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - K Ganesh Kumar
- Department of Metabolism and Aging, Scripps Research Institute, Jupiter, Florida, USA
| | - Jose E Galgani
- Departamento de Ciencias de la SaludCarrera de Nutrición y Dietética and Departamento de Nutrición, Diabetes y Metabolismo, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Michael L Niehoff
- Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, Missouri, USA; Division of Geriatric Medicine, Saint Louis University School of Medicine, St. Louis, Saint Louis University School of Medicine; Research Service, John Cochran Division, Saint Louis Veterans Affairs Medical Center, Missouri, USA
| | - Susan A Farr
- Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, Missouri, USA; Division of Geriatric Medicine, Saint Louis University School of Medicine, St. Louis, Saint Louis University School of Medicine; Research Service, John Cochran Division, Saint Louis Veterans Affairs Medical Center, Missouri, USA
| | - Jinsong Zhang
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Andrew A Butler
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri, USA; Department of Metabolism and Aging, Scripps Research Institute, Jupiter, Florida, USA; Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, Missouri, USA.
| |
Collapse
|
4
|
Duan X, Cai L, Schmidt EJ, Shen J, Tycksen ED, O’Keefe R, Cheverud JM, Farooq Rai M. RNA-seq analysis of chondrocyte transcriptome reveals genetic heterogeneity in LG/J and SM/J murine strains. Osteoarthritis Cartilage 2020; 28:516-527. [PMID: 31945456 PMCID: PMC7108965 DOI: 10.1016/j.joca.2020.01.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 01/02/2020] [Accepted: 01/03/2020] [Indexed: 02/07/2023]
Abstract
OBJECTIVE To investigate the transcriptomic differences in chondrocytes obtained from LG/J (large, healer) and SM/J (small, non-healer) murine strains in an attempt to discern the molecular pathways implicated in cartilage regeneration and susceptibility to osteoarthritis (OA). DESIGN We performed RNA-sequencing on chondrocytes derived from LG/J (n = 16) and SM/J (n = 16) mice. We validated the expression of candidate genes and compared single nucleotide polymorphisms (SNPs) between the two mouse strains. We also examined gene expression of positional candidates for ear pinna regeneration and long bone length quantitative trait loci (QTLs) that display differences in cartilaginous expression. RESULTS We observed a distinct genetic heterogeneity between cells derived from LG/J and SM/J mouse strains. We found that gene ontologies representing cell development, cartilage condensation, and regulation of cell differentiation were enriched in LG/J chondrocytes. In contrast, gene ontologies enriched in the SM/J chondrocytes were mainly related to inflammation and degeneration. Moreover, SNP analysis revealed that multiple validated genes vary in sequence between LG/J and SM/J in coding and highly conserved noncoding regions. Finally, we showed that most QTLs have 20-30% of their positional candidates displaying differential expression between the two mouse strains. CONCLUSIONS While the enrichment of pathways related to cell differentiation, cartilage development and cartilage condensation infers superior healing potential of LG/J strain, the enrichment of pathways related to cytokine production, immune cell activation and inflammation entails greater susceptibility of SM/J strain to OA. These data provide novel insights into chondrocyte transcriptome and aid in identification of the quantitative trait genes and molecular differences underlying the phenotypic differences associated with individual QTLs.
Collapse
Affiliation(s)
- Xin Duan
- Department of Orthopedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Lei Cai
- Department of Orthopedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Eric J. Schmidt
- School of Physician Assistant Medicine, College of Health Sciences, University of Lynchburg, Lynchburg, VA, United States
| | - Jie Shen
- Department of Orthopedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Eric D. Tycksen
- Genome Technology Access Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Regis O’Keefe
- Department of Orthopedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, United States
| | - James M. Cheverud
- Department of Biology, Loyola University, Chicago, IL, United States
| | - Muhammad Farooq Rai
- Department of Orthopedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, United States, Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
5
|
Hernandez Cordero AI, Gonzales NM, Parker CC, Sokolof G, Vandenbergh DJ, Cheng R, Abney M, Sko A, Douglas A, Palmer AA, Gregory JS, Lionikas A. Genome-wide Associations Reveal Human-Mouse Genetic Convergence and Modifiers of Myogenesis, CPNE1 and STC2. Am J Hum Genet 2019; 105:1222-1236. [PMID: 31761296 PMCID: PMC6904802 DOI: 10.1016/j.ajhg.2019.10.014] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 10/28/2019] [Indexed: 12/11/2022] Open
Abstract
Muscle bulk in adult healthy humans is highly variable even after height, age, and sex are accounted for. Low muscle mass, due to fewer and/or smaller constituent muscle fibers, would exacerbate the impact of muscle loss occurring in aging or disease. Genetic variability substantially influences muscle mass differences, but causative genes remain largely unknown. In a genome-wide association study (GWAS) on appendicular lean mass (ALM) in a population of 85,750 middle-aged (aged 38-49 years) individuals from the UK Biobank (UKB), we found 182 loci associated with ALM (p < 5 × 10-8). We replicated associations for 78% of these loci (p < 5 × 10-8) with ALM in a population of 181,862 elderly (aged 60-74 years) individuals from UKB. We also conducted a GWAS on hindlimb skeletal muscle mass of 1,867 mice from an advanced intercross between two inbred strains (LG/J and SM/J); this GWAS identified 23 quantitative trait loci. Thirty-eight positional candidates distributed across five loci overlapped between the two species. In vitro studies of positional candidates confirmed CPNE1 and STC2 as modifiers of myogenesis. Collectively, these findings shed light on the genetics of muscle mass variability in humans and identify targets for the development of interventions for treatment of muscle loss. The overlapping results between humans and the mouse model GWAS point to shared genetic mechanisms across species.
Collapse
Affiliation(s)
- Ana I Hernandez Cordero
- School of Medicine, Medical Sciences, and Nutrition, College of Life Sciences and Medicine, University of Aberdeen, Aberdeen, UK AB24 3FX, UK
| | - Natalia M Gonzales
- Department of Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Clarissa C Parker
- Department of Psychology, Middlebury College, Middlebury, VT 05753, USA; Program in Neuroscience, Middlebury College, Middlebury, VT, 05753, USA
| | - Greta Sokolof
- Department of Psychological and Brain Sciences, The University of Iowa, Iowa City, IA 52242, USA
| | - David J Vandenbergh
- Department of Biobehavioral Health, Penn State Institute for the Neurosciences, and Molecular, Cellular, and Integrative Sciences Program, Pennsylvania State University, University Park, PA 16802, USA
| | - Riyan Cheng
- Department of Health Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Mark Abney
- Department of Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Andrew Sko
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Alex Douglas
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, AB24 3FX, UK
| | - Abraham A Palmer
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA; Institute for Genomic Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Jennifer S Gregory
- School of Medicine, Medical Sciences, and Nutrition, College of Life Sciences and Medicine, University of Aberdeen, Aberdeen, UK AB24 3FX, UK
| | - Arimantas Lionikas
- School of Medicine, Medical Sciences, and Nutrition, College of Life Sciences and Medicine, University of Aberdeen, Aberdeen, UK AB24 3FX, UK.
| |
Collapse
|
6
|
Butler AA, Zhang J, Price CA, Stevens JR, Graham JL, Stanhope KL, King S, Krauss RM, Bremer AA, Havel PJ. Low plasma adropin concentrations increase risks of weight gain and metabolic dysregulation in response to a high-sugar diet in male nonhuman primates. J Biol Chem 2019; 294:9706-9719. [PMID: 30988006 PMCID: PMC6597842 DOI: 10.1074/jbc.ra119.007528] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 03/30/2019] [Indexed: 12/15/2022] Open
Abstract
Mouse studies linking adropin, a peptide hormone encoded by the energy homeostasis-associated (ENHO) gene, to biological clocks and to glucose and lipid metabolism suggest a potential therapeutic target for managing diseases of metabolism. However, adropin's roles in human metabolism are unclear. In silico expression profiling in a nonhuman primate diurnal transcriptome atlas (GSE98965) revealed a dynamic and diurnal pattern of ENHO expression. ENHO expression is abundant in brain, including ventromedial and lateral hypothalamic nuclei regulating appetite and autonomic function. Lower ENHO expression is present in liver, lung, kidney, ileum, and some endocrine glands. Hepatic ENHO expression associates with genes involved in glucose and lipid metabolism. Unsupervised hierarchical clustering identified 426 genes co-regulated with ENHO in liver, ileum, kidney medulla, and lung. Gene Ontology analysis of this cluster revealed enrichment for epigenetic silencing by histone H3K27 trimethylation and biological processes related to neural function. Dietary intervention experiments with 59 adult male rhesus macaques indicated low plasma adropin concentrations were positively correlated with fasting glucose, plasma leptin, and apolipoprotein C3 (APOC3) concentrations. During consumption of a high-sugar (fructose) diet, which induced 10% weight gain, animals with low adropin had larger increases of plasma leptin and more severe hyperglycemia. Declining adropin concentrations were correlated with increases of plasma APOC3 and triglycerides. In summary, peripheral ENHO expression associates with pathways related to epigenetic and neural functions, and carbohydrate and lipid metabolism, suggesting co-regulation in nonhuman primates. Low circulating adropin predicts increased weight gain and metabolic dysregulation during consumption of a high-sugar diet.
Collapse
Affiliation(s)
- Andrew A Butler
- From the Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri 63104,
- The Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, St. Louis, Missouri 63104
| | - Jinsong Zhang
- From the Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri 63104
- The Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, St. Louis, Missouri 63104
| | - Candice A Price
- the Department of Molecular Biosciences, School of Veterinary Medicine and Department of Nutrition, University of California, Davis, Davis, California 95616
| | - Joseph R Stevens
- From the Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri 63104
| | - James L Graham
- the Department of Molecular Biosciences, School of Veterinary Medicine and Department of Nutrition, University of California, Davis, Davis, California 95616
| | - Kimber L Stanhope
- the Department of Molecular Biosciences, School of Veterinary Medicine and Department of Nutrition, University of California, Davis, Davis, California 95616
| | - Sarah King
- the Children's Hospital Oakland Research Institute, Oakland, California 94609, and
| | - Ronald M Krauss
- the Children's Hospital Oakland Research Institute, Oakland, California 94609, and
| | - Andrew A Bremer
- the Department of Pediatrics, Vanderbilt University, Nashville, Tennessee 37232
| | - Peter J Havel
- the Department of Molecular Biosciences, School of Veterinary Medicine and Department of Nutrition, University of California, Davis, Davis, California 95616,
| |
Collapse
|
7
|
Moshtaq MA, Rahimi MH, Mollahosseini M, Khorrami-Nezhad L, Maghbooli Z, Mirzaei K, Pooyan S, Setayesh L. Association between dietary glycemic index and liver enzymes level among apparently healthy adults. Diabetes Metab Syndr 2019; 13:1597-1602. [PMID: 31336527 DOI: 10.1016/j.dsx.2019.03.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 03/05/2019] [Indexed: 12/31/2022]
Abstract
OBJECTIVE The previous studies have revealed that there is a link between dietary glycemic index and lipid profile in overweight and obesity. The aim of study was to investigate whether the glycemic index is associated with liver enzymes. METHOD Anthropometric and biochemical parameters were measured in 265 participants. Dietary glycemic index (GI) was assessed by using a validated food-frequency questionnaire. With adjusting confounder variable, Binary logistic regression was also used to predict the relationship between liver enzymes and quartile of intake. RESULTS There was a significant difference between low and high GI diet for BMR (P = 0.01), FFM (P = 0.03), TG (P = 0.02), HDL (P = 0.002). The association between HDL and glycemic index remained significant after adjustment of sex and age (P = 0.03). Using the regression model following adjustment revealed that for each 1% increase in the degree of the GI, there was 11% elevation in liver enzyme abnormalities. In both groups of men and women, enzyme abnormalities positively correlated with GI, while only men showed remarkable correlation in all models (crude model: β = 0.07, OR = 1.07, CI = 0.98to 1.16). Additionally, an increase in the degree of GI caused an elevation in enzyme abnormalities by 7%. With adjusting sex, age, BMI, and Physical activity, a significance correlation was found between GI and Enzyme abnormalities (p-value = 0.03, OR = 1.115). CONCLUSION Our study indicated that high glycemic index diet led to the elevated levels of the liver enzymes, while being significant only in men.
Collapse
Affiliation(s)
- Mohammad Ali Moshtaq
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Mohammad Hossein Rahimi
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Mehdi Mollahosseini
- Department of Nutrition, School of Public Health, Shahid Sadoughi University of Medical Sciences, Iran
| | - Leila Khorrami-Nezhad
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Zhila Maghbooli
- Multiple Sclerosis Research Center, Sina hospital, Tehran University of Medical Sciences, Tehran, Iran.
| | - Khadijeh Mirzaei
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran.
| | - Sara Pooyan
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Leila Setayesh
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| |
Collapse
|
8
|
Chen X, Xue H, Fang W, Chen K, Chen S, Yang W, Shen T, Chen X, Zhang P, Ling W. Adropin protects against liver injury in nonalcoholic steatohepatitis via the Nrf2 mediated antioxidant capacity. Redox Biol 2019; 21:101068. [PMID: 30684890 PMCID: PMC6351233 DOI: 10.1016/j.redox.2018.101068] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 11/23/2018] [Accepted: 12/05/2018] [Indexed: 12/20/2022] Open
Abstract
Adropin, a secretory signal peptide, has shown beneficial effects on improving glucose homeostasis and dyslipidemia. However, whether this peptide affects nonalcoholic steatohepatitis (NASH) has remained unclear. In this study, the serum adropin levels, liver injury and oxidative stress were measured in diet-induced NASH mice. Adropin knock-out mice and palmitate treated primary hepatic cells were used to investigate the influence of adropin on liver injury. Our results show that serum adropin levels were decreased and negatively correlated with liver injury in NASH mice. Knockout of adropin significantly exacerbated hepatic steatosis, inflammatory responses and fibrosis in mice after either methionine-choline deficient diet (MCD) or western diet (WD) feeding. And the treatment with adropin bioactive peptides ameliorated NASH progression in mice. Adropin alleviated hepatocyte injury by upregulating the expression of Gclc, Gclm, and Gpx1 in a manner dependent on Nrf2 transcriptional activity and by increasing the glutathione (GSH) levels. And adropin significantly increased CBP expression and promoted its binding with Nrf2, which enhanced Nrf2 transcriptional activity. Furthermore, AAV8-mediated overexpression of hepatic Nrf2 expression functionally restored the liver injury induced by adropin-deficiency MCD-fed mice. These findings provide evidence that adropin activates Nrf2 signaling and plays a protective role in liver injury of NASH and therefore might represent a novel target for the prevention and treatment of NASH.
Collapse
Affiliation(s)
- Xu Chen
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, PR China; Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou 510080, PR China.
| | - Hongliang Xue
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, PR China; Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou 510080, PR China.
| | - Wanjun Fang
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, PR China; Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou 510080, PR China.
| | - Ke Chen
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, PR China; Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou 510080, PR China.
| | - Shen Chen
- Department of Toxicology, School of Public Health, Sun Yat-sen University, 510080, PR China; Guangzhou Key Laboratory of Environmental Pollution and Health Risk Assessment, Guangzhou 510080, PR China.
| | - Wenqi Yang
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, PR China; Laboratory Center for Sport Science and Medicine, Guangzhou Institute of Physical Education, Guangzhou 510080, PR China.
| | - Tianran Shen
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, PR China; Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou 510080, PR China.
| | - Xuechen Chen
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, PR China; Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou 510080, PR China.
| | - Peiwen Zhang
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, PR China; Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou 510080, PR China.
| | - Wenhua Ling
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, PR China; Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou 510080, PR China.
| |
Collapse
|
9
|
Chinzei N, Rai MF, Hashimoto S, Schmidt EJ, Takebe K, Cheverud JM, Sandell LJ. Evidence for Genetic Contribution to Variation in Posttraumatic Osteoarthritis in Mice. Arthritis Rheumatol 2019; 71:370-381. [PMID: 30225954 DOI: 10.1002/art.40730] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 09/13/2018] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Recombinant inbred mouse strains generated from an LG/J and SM/J intercross offer a unique resource to study complex genetic traits such as osteoarthritis (OA). We undertook this study to determine the susceptibility of 14 strains to various phenotypes characteristic of posttraumatic OA. We hypothesized that phenotypic variability is associated with genetic variability. METHODS Ten-week-old male mice underwent surgical destabilization of the medial meniscus (DMM) to induce posttraumatic OA. Mice were killed 8 weeks after surgery, and knee joints were processed for histology to score cartilage degeneration and synovitis. Micro-computed tomography was used to analyze trabecular bone parameters including subchondral bone plate thickness and synovial ectopic calcifications. Gene expression in the knees was assessed using a QuantiGene Plex assay. RESULTS Broad-sense heritability ranged from 0.18 to 0.58, which suggested that the responses to surgery were moderately heritable. The LGXSM-33, LGXSM-5, LGXSM-46, and SM/J strains were highly susceptible to OA, while the LGXSM-131b, LGXSM-163, LGXSM-35, LGXSM-128a, LGXSM-6, and LG/J strains were relatively OA resistant. This study was the first to accomplish measurement of genetic correlations of phenotypes that are characteristic of posttraumatic OA. Cartilage degeneration was significantly positively associated with synovitis (r = 0.83-0.92), and subchondral bone plate thickness was negatively correlated with ectopic calcifications (r = -0.59). Moreover, we showed that 40 of the 78 genes tested were significantly correlated with various OA phenotypes. However, unlike the OA phenotypes, there was no evidence for genetic variation in differences in gene expression levels between DMM-operated and sham-operated knees. CONCLUSION For these mouse strains, various characteristics of posttraumatic OA varied with genetic composition, which demonstrated a genetic basis for susceptibility to posttraumatic OA. The heritability of posttraumatic OA was established. Phenotypes exhibited various degrees of correlations; cartilage degeneration was positively correlated with synovitis, but not with the formation of ectopic calcifications. Further investigation of the genome regions that contain genes implicated in OA, as well as further investigation of gene expression data, will be useful for studying mechanisms of OA and identifying therapeutic targets.
Collapse
Affiliation(s)
| | | | | | | | - Ken Takebe
- Konan Kakogawa Hospital, Kakogawa, Japan
| | | | | |
Collapse
|
10
|
Keleher MR, Zaidi R, Hicks L, Shah S, Xing X, Li D, Wang T, Cheverud JM. A high-fat diet alters genome-wide DNA methylation and gene expression in SM/J mice. BMC Genomics 2018; 19:888. [PMID: 30526554 PMCID: PMC6286549 DOI: 10.1186/s12864-018-5327-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 11/28/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND While the genetics of obesity has been well defined, the epigenetics of obesity is poorly understood. Here, we used a genome-wide approach to identify genes with differences in both DNA methylation and expression associated with a high-fat diet in mice. RESULTS We weaned genetically identical Small (SM/J) mice onto a high-fat or low-fat diet and measured their weights weekly, tested their glucose and insulin tolerance, assessed serum biomarkers, and weighed their organs at necropsy. We measured liver gene expression with RNA-seq (using 21 total libraries, each pooled with 2 mice of the same sex and diet) and DNA methylation with MRE-seq and MeDIP-seq (using 8 total libraries, each pooled with 4 mice of the same sex and diet). There were 4356 genes with expression differences associated with diet, with 184 genes exhibiting a sex-by-diet interaction. Dietary fat dysregulated several pathways, including those involved in cytokine-cytokine receptor interaction, chemokine signaling, and oxidative phosphorylation. Over 7000 genes had differentially methylated regions associated with diet, which occurred in regulatory regions more often than expected by chance. Only 5-10% of differentially methylated regions occurred in differentially expressed genes, however this was more often than expected by chance (p = 2.2 × 10- 8). CONCLUSIONS Discovering the gene expression and methylation changes associated with a high-fat diet can help to identify new targets for epigenetic therapies and inform about the physiological changes in obesity. Here, we identified numerous genes with altered expression and methylation that are promising candidates for further study.
Collapse
Affiliation(s)
- Madeline Rose Keleher
- Department of Evolution, Ecology, and Population Biology, Washington University in St. Louis, St. Louis, MO 63105 USA
- Biology Department, 1032 W. Sheridan Road, Chicago, IL 60660 USA
| | - Rabab Zaidi
- Department of Biology, Loyola University, Chicago, IL 60660 USA
| | - Lauren Hicks
- Department of Biology, Loyola University, Chicago, IL 60660 USA
| | - Shyam Shah
- Department of Biology, Loyola University, Chicago, IL 60660 USA
| | - Xiaoyun Xing
- Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110 USA
| | - Daofeng Li
- Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110 USA
| | - Ting Wang
- Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110 USA
| | | |
Collapse
|
11
|
Keleher MR, Zaidi R, Patel K, Ahmed A, Bettler C, Pavlatos C, Shah S, Cheverud JM. The effect of dietary fat on behavior in mice. J Diabetes Metab Disord 2018; 17:297-307. [PMID: 30918865 DOI: 10.1007/s40200-018-0373-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 11/09/2018] [Indexed: 10/27/2022]
Abstract
Purpose Obesity is linked to cognitive dysfunction in humans and rodents, and its effects can be passed on to the next generation. However, the extent of these effects is not well understood. The purpose of this study was to determine the effect of a prenatal maternal high-fat diet and an individual high-fat diet in inbred mice. Methods We varied maternal diet and offspring diet to test the hypothesis that a high-fat diet would increase anxiety, reduce activity levels, and impair nest-building. First, we fed a high-fat (HF) or low-fat (LF) diet to genetically identical female Small (SM/J) mice and mated them with LF males. We cross-fostered all offspring to LF-fed SM/J nurses and weaned them onto an HF or LF diet. We weighed the mice weekly and we tested anxiety with the Open Field Test, activity levels with instantaneous scan sampling, and nest building using the Deacon Scale. Results Diet significantly affected weight, with HF females weighing 28.2 g (± 1.4 g SE) and LF females weighing 15.1 g (± 1.6 g SE) at 17 weeks old. The offspring's own diet had major behavioral effects. HF mice produced more fecal boli and urinations in the Open Field Test, built lower-quality nests, and had lower activity in adulthood than LF mice. The only trait that a prenatal maternal diet significantly affected was whether the offspring built their nests inside or outside of a hut. Conclusions Offspring diet, but not prenatal maternal diet, affected a wide range of behaviors in these mice.
Collapse
Affiliation(s)
- Madeline Rose Keleher
- 1Department of Evolution, Ecology, and Population Biology, Washington University in St. Louis, St. Louis, MO USA.,2Department of Biology, Loyola University, 1032 W. Sheridan Road, Chicago, IL 60660 USA
| | - Rabab Zaidi
- 2Department of Biology, Loyola University, 1032 W. Sheridan Road, Chicago, IL 60660 USA
| | - Kayna Patel
- 2Department of Biology, Loyola University, 1032 W. Sheridan Road, Chicago, IL 60660 USA
| | - Amer Ahmed
- 2Department of Biology, Loyola University, 1032 W. Sheridan Road, Chicago, IL 60660 USA
| | - Carlee Bettler
- 2Department of Biology, Loyola University, 1032 W. Sheridan Road, Chicago, IL 60660 USA
| | - Cassondra Pavlatos
- 2Department of Biology, Loyola University, 1032 W. Sheridan Road, Chicago, IL 60660 USA
| | - Shyam Shah
- 2Department of Biology, Loyola University, 1032 W. Sheridan Road, Chicago, IL 60660 USA
| | - James M Cheverud
- 2Department of Biology, Loyola University, 1032 W. Sheridan Road, Chicago, IL 60660 USA
| |
Collapse
|
12
|
Zang H, Jiang F, Cheng X, Xu H, Hu X. Serum adropin levels are decreased in Chinese type 2 diabetic patients and negatively correlated with body mass index. Endocr J 2018; 65:685-691. [PMID: 29669965 DOI: 10.1507/endocrj.ej18-0060] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Adropin has been identified as potent regulatory hormone implicated in insulin sensitivity and the maintenance of energy homeostasis. The aim of current study was to investigate serum adropin concentrations of type 2 diabetes mellitus (T2DM) patients in the fasting status, especially those overweight/obese and evaluate the relationships between adropin levels and metabolic parameters. A total of 116 T2DM patients and 60 controls with normal glucose tolerance (NGT) were recruited to the study. Adropin concentration was determined using commercial ELISA kits. Anthropometric characteristics were collected and biochemistry, glycosylated hemoglobin A1c (HbA1c) and fasting insulin (FIns) were detected by clinical laboratory. Insulin resistance was estimated by homeostasis model 2 assessment of insulin resistance (HOMA2-IR). Serum adropin levels in Chinese T2DM patients were decreased compared with the controls [3.8 (3.0-5.5) vs. 5.5 (3.7-7.9) ng/mL, p < 0.01]. Meanwhile, overweight/obese patients had more considerably reduced levels of adropin. Adropin level was negatively correlated with body mass index (BMI), high-sensitive C reactive protein (hs-CRP), triglycerides (TG), fasting plasma glucose (FPG), FIns, HOMA2-IR and HbA1c, while positively with high-density lipoprotein cholesterol (HDL-C) in study participants (p < 0.01). The correlations of adropin with glucolipid variables (TG, HDL-C, FPG, FIns, HOMA2-IR, HbA1c) still existed after adjusting the effect of BMI. Besides, HOMA2-IR and HbA1c were independent factors associated with serum adropin levels. Binary logistic regression analyses showed that adropin was significantly associated with T2DM after removing confounding factors (p < 0.01). Receiver operating characteristic (ROC) curve demonstrated adropin concentration of 5.8 ng/mL could be used as a possible optimal cut-off value to identify T2DM from non-T2DM with sensitivity of 81.9% and specificity of 46.7%. Serum adropin concentrations are decreased in Chinese T2DM patients, especially those overweight/obese. Adropin, associated with glucolipid homeostasis and insulin sensitivity, may implicate in the pathogenesis of T2DM.
Collapse
Affiliation(s)
- Huanhuan Zang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Fusong Jiang
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Xingbo Cheng
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Hong Xu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Xingna Hu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| |
Collapse
|
13
|
Maternal high-fat diet associated with altered gene expression, DNA methylation, and obesity risk in mouse offspring. PLoS One 2018; 13:e0192606. [PMID: 29447215 PMCID: PMC5813940 DOI: 10.1371/journal.pone.0192606] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 01/28/2018] [Indexed: 12/18/2022] Open
Abstract
We investigated maternal obesity in inbred SM/J mice by assigning females to a high-fat diet or a low-fat diet at weaning, mating them to low-fat-fed males, cross-fostering the offspring to low-fat-fed SM/J nurses at birth, and weaning the offspring onto a high-fat or low-fat diet. A maternal high-fat diet exacerbated obesity in the high-fat-fed daughters, causing them to weigh more, have more fat, and have higher serum levels of leptin as adults, accompanied by dozens of gene expression changes and thousands of DNA methylation changes in their livers and hearts. Maternal diet particularly affected genes involved in RNA processing, immune response, and mitochondria. Between one-quarter and one-third of differentially expressed genes contained a differentially methylated region associated with maternal diet. An offspring high-fat diet reduced overall variation in DNA methylation, increased body weight and organ weights, increased long bone lengths and weights, decreased insulin sensitivity, and changed the expression of 3,908 genes in the liver. Although the offspring were more affected by their own diet, their maternal diet had epigenetic effects lasting through adulthood, and in the daughters these effects were accompanied by phenotypic changes relevant to obesity and diabetes.
Collapse
|
14
|
Ghoshal S, Stevens JR, Billon C, Girardet C, Sitaula S, Leon AS, Rao DC, Skinner JS, Rankinen T, Bouchard C, Nuñez MV, Stanhope KL, Howatt DA, Daugherty A, Zhang J, Schuelke M, Weiss EP, Coffey AR, Bennett BJ, Sethupathy P, Burris TP, Havel PJ, Butler AA. Adropin: An endocrine link between the biological clock and cholesterol homeostasis. Mol Metab 2017; 8:51-64. [PMID: 29331507 PMCID: PMC5985041 DOI: 10.1016/j.molmet.2017.12.002] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 11/28/2017] [Accepted: 12/02/2017] [Indexed: 01/13/2023] Open
Abstract
Objective Identify determinants of plasma adropin concentrations, a secreted peptide translated from the Energy Homeostasis Associated (ENHO) gene linked to metabolic control and vascular function. Methods Associations between plasma adropin concentrations, demographics (sex, age, BMI) and circulating biomarkers of lipid and glucose metabolism were assessed in plasma obtained after an overnight fast in humans. The regulation of adropin expression was then assessed in silico, in cultured human cells, and in animal models. Results In humans, plasma adropin concentrations are inversely related to atherogenic LDL-cholesterol (LDL-C) levels in men (n = 349), but not in women (n = 401). Analysis of hepatic Enho expression in male mice suggests control by the biological clock. Expression is rhythmic, peaking during maximal food consumption in the dark correlating with transcriptional activation by RORα/γ. The nadir in the light phase coincides with the rest phase and repression by Rev-erb. Plasma adropin concentrations in nonhuman primates (rhesus monkeys) also exhibit peaks coinciding with feeding times (07:00 h, 15:00 h). The ROR inverse agonists SR1001 and the 7-oxygenated sterols 7-β-hydroxysterol and 7-ketocholesterol, or the Rev-erb agonist SR9009, suppress ENHO expression in cultured human HepG2 cells. Consumption of high-cholesterol diets suppress expression of the adropin transcript in mouse liver. However, adropin over expression does not prevent hypercholesterolemia resulting from a high cholesterol diet and/or LDL receptor mutations. Conclusions In humans, associations between plasma adropin concentrations and LDL-C suggest a link with hepatic lipid metabolism. Mouse studies suggest that the relationship between adropin and cholesterol metabolism is unidirectional, and predominantly involves suppression of adropin expression by cholesterol and 7-oxygenated sterols. Sensing of fatty acids, cholesterol and oxysterols by the RORα/γ ligand-binding domain suggests a plausible functional link between adropin expression and cellular lipid metabolism. Furthermore, the nuclear receptors RORα/γ and Rev-erb may couple adropin synthesis with circadian rhythms in carbohydrate and lipid metabolism. In male humans, plasma adropin concentrations are inversely related to low-density circulating cholesterol (LDL-C) levels. Adropin expression is regulated by core elements of the biological clock (RORA/G, Rev-Erb). Sterol-sensing by the ROR ligand-binding domain provides a plausible link between adropin expression and lipid metabolism. In mouse liver, adropin expression is rhythmic and suppressed by exogenous (dietary) cholesterol.
Collapse
Affiliation(s)
- Sarbani Ghoshal
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis University, St. Louis, MO, USA
| | - Joseph R Stevens
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis University, St. Louis, MO, USA
| | - Cyrielle Billon
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis University, St. Louis, MO, USA
| | - Clemence Girardet
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis University, St. Louis, MO, USA
| | - Sadichha Sitaula
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis University, St. Louis, MO, USA
| | - Arthur S Leon
- School of Kinesiology and Leisure Studies, University of Minnesota, Minneapolis, MN, USA
| | - D C Rao
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - James S Skinner
- Department of Kinesiology, Indiana University, Bloomington, IN, USA
| | - Tuomo Rankinen
- Human Genomics Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA
| | - Claude Bouchard
- Human Genomics Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA
| | - Marinelle V Nuñez
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California-Davis, Davis, CA, USA; Department of Nutrition, School of Medicine, University of California-Davis, Davis, CA, USA
| | - Kimber L Stanhope
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California-Davis, Davis, CA, USA; Department of Nutrition, School of Medicine, University of California-Davis, Davis, CA, USA
| | - Deborah A Howatt
- Saha Cardiovascular Research Center, Department of Physiology, University of Kentucky, KY, USA
| | - Alan Daugherty
- Saha Cardiovascular Research Center, Department of Physiology, University of Kentucky, KY, USA
| | - Jinsong Zhang
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis University, St. Louis, MO, USA
| | - Matthew Schuelke
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis University, St. Louis, MO, USA
| | - Edward P Weiss
- Department of Nutrition and Dietetics, Doisy College of Health Sciences, Saint Louis University, St. Louis, MO, USA
| | - Alisha R Coffey
- Curriculum in Genetics and Molecular Biology, University of North Carolina, Chapel Hill, NC, USA
| | - Brian J Bennett
- Obesity and Metabolism Unit, Western Human Nutrition Center, USDA-ARS, Davis, CA, USA
| | - Praveen Sethupathy
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Thomas P Burris
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis University, St. Louis, MO, USA
| | - Peter J Havel
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California-Davis, Davis, CA, USA; Department of Nutrition, School of Medicine, University of California-Davis, Davis, CA, USA
| | - Andrew A Butler
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis University, St. Louis, MO, USA.
| |
Collapse
|
15
|
Salleh MS, Mazzoni G, Höglund JK, Olijhoek DW, Lund P, Løvendahl P, Kadarmideen HN. RNA-Seq transcriptomics and pathway analyses reveal potential regulatory genes and molecular mechanisms in high- and low-residual feed intake in Nordic dairy cattle. BMC Genomics 2017; 18:258. [PMID: 28340555 PMCID: PMC5366136 DOI: 10.1186/s12864-017-3622-9] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 03/11/2017] [Indexed: 11/24/2022] Open
Abstract
Background The selective breeding of cattle with high-feed efficiencies (FE) is an important goal of beef and dairy cattle producers. Global gene expression patterns in relevant tissues can be used to study the functions of genes that are potentially involved in regulating FE. In the present study, high-throughput RNA sequencing data of liver biopsies from 19 dairy cows were used to identify differentially expressed genes (DEGs) between high- and low-FE groups of cows (based on Residual Feed Intake or RFI). Subsequently, a profile of the pathways connecting the DEGs to FE was generated, and a list of candidate genes and biomarkers was derived for their potential inclusion in breeding programmes to improve FE. Results The bovine RNA-Seq gene expression data from the liver was analysed to identify DEGs and, subsequently, identify the molecular mechanisms, pathways and possible candidate biomarkers of feed efficiency. On average, 57 million reads (short reads or short mRNA sequences < ~200 bases) were sequenced, 52 million reads were mapped, and 24,616 known transcripts were quantified according to the bovine reference genome. A comparison of the high- and low-RFI groups revealed 70 and 19 significantly DEGs in Holstein and Jersey cows, respectively. The interaction analysis (high vs. low RFI x control vs. high concentrate diet) showed no interaction effects in the Holstein cows, while two genes showed interaction effects in the Jersey cows. The analyses showed that DEGs act through certain pathways to affect or regulate FE, including steroid hormone biosynthesis, retinol metabolism, starch and sucrose metabolism, ether lipid metabolism, arachidonic acid metabolism and drug metabolism cytochrome P450. Conclusion We used RNA-Seq-based liver transcriptomic profiling of high- and low-RFI dairy cows in two breeds and identified significantly DEGs, their molecular mechanisms, their interactions with other genes and functional enrichments of different molecular pathways. The DEGs that were identified were the CYP’s and GIMAP genes for the Holstein and Jersey cows, respectively, which are related to the primary immunodeficiency pathway and play a major role in feed utilization and the metabolism of lipids, sugars and proteins. Electronic supplementary material The online version of this article (doi:10.1186/s12864-017-3622-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- M S Salleh
- Animal Breeding, Quantitative Genetics and Systems Biology Group, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, DK-1870, Frederiksberg C, Denmark
| | - G Mazzoni
- Animal Breeding, Quantitative Genetics and Systems Biology Group, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, DK-1870, Frederiksberg C, Denmark
| | - J K Höglund
- Department of Molecular Biology and Genetics - Center for Quantitative Genetics and Genomics, Aarhus University, AU Foulum, DK-8830, Tjele, Denmark
| | - D W Olijhoek
- Department of Molecular Biology and Genetics - Center for Quantitative Genetics and Genomics, Aarhus University, AU Foulum, DK-8830, Tjele, Denmark.,Department of Animal Science - Animal Nutrition and Physiology, Aarhus University, AU Foulum, DK-8830, Tjele, Denmark
| | - P Lund
- Department of Animal Science - Animal Nutrition and Physiology, Aarhus University, AU Foulum, DK-8830, Tjele, Denmark
| | - P Løvendahl
- Department of Molecular Biology and Genetics - Center for Quantitative Genetics and Genomics, Aarhus University, AU Foulum, DK-8830, Tjele, Denmark
| | - H N Kadarmideen
- Department of Bio and Health Informatics, Technical University of Denmark, DK-2800, Kgs. Lyngby, Denmark.
| |
Collapse
|
16
|
Lawson HA, Zayed M, Wayhart JP, Fabbrini E, Love-Gregory L, Klein S, Semenkovich CF. Physiologic and genetic evidence links hemopexin to triglycerides in mice and humans. Int J Obes (Lond) 2017; 41:631-638. [PMID: 28119529 DOI: 10.1038/ijo.2017.19] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 12/21/2016] [Accepted: 01/11/2017] [Indexed: 01/28/2023]
Abstract
BACKGROUND/OBJECTIVES Elevated triglycerides predict insulin resistance and vascular disease in obesity, but how the inert triglyceride molecule is related to development of metabolic disease is unknown. To pursue novel potential mediators of triglyceride-associated metabolic disease, we used a forward genetics approach involving inbred mice and translated our findings to human subjects. SUBJECTS/METHODS Hemopexin (HPX) was identified as a differentially expressed gene within a quantitative trait locus associated with serum triglycerides in an F16 advanced intercross between the LG/J and SM/J strains of mice. Hpx expression was evaluated in both the reproductive fat pads and livers of mice representing three strains, LG/J (n=25), SM/J (n=27) and C57Bl/6J (n=19), on high- and low-fat diets. The effect of altered Hpx expression on adipogenesis was studied in 3T3-L1 cells. Circulating HPX protein along with HPX expression were characterized in subcutaneous white adipose tissue samples obtained from a cohort of metabolically abnormal (n=18) and of metabolically normal (n=24) obese human subjects. We further examined the relationship between HPX and triglycerides in human atherosclerotic plaques (n=18). RESULTS HPX expression in mouse adipose tissue, but not in liver, was regulated by dietary fat regardless of genetic background. HPX increased in concert with adipogenesis in 3T3-L1 cells, and disruption of its expression impaired adipocyte differentiation. RNAseq data from the adipose tissue of obese humans showed differential expression of HPX based on metabolic disease status (P<0.05), and circulating HPX levels were correlated with serum triglycerides in these subjects (r=0.33; P=0.03). HPX was also found in an unbiased proteomic screen of human atherosclerotic plaques and shown to display differential abundance based on the extent of disease and triglyceride content (P<0.05). CONCLUSIONS Our findings suggest that HPX is associated with triglycerides and provide a framework for understanding mechanisms underlying lipid metabolism and metabolic disease.
Collapse
Affiliation(s)
- H A Lawson
- Department of Genetics, Washington University School of Medicine, St Louis, MO, USA
| | - M Zayed
- Department of Surgery, Section of Vascular Surgery, Washington University School of Medicine, St Louis, MO, USA
| | - J P Wayhart
- Department of Genetics, Washington University School of Medicine, St Louis, MO, USA
| | - E Fabbrini
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - L Love-Gregory
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - S Klein
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - C F Semenkovich
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| |
Collapse
|
17
|
Ishikawa A. Identification of a Putative Quantitative Trait Gene for Resistance to Obesity in Mice Using Transcriptome Analysis and Causal Inference Tests. PLoS One 2017; 12:e0170652. [PMID: 28114323 PMCID: PMC5256930 DOI: 10.1371/journal.pone.0170652] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 01/09/2017] [Indexed: 11/19/2022] Open
Abstract
It is still challenging to identify causal genes governing obesity. Pbwg1.5, a quantitative trait locus (QTL) for resistance to obesity, was previously discovered from wild Mus musculus castaneus mice and was fine-mapped to a 2.1-Mb genomic region of mouse chromosome 2, where no known gene with an effect on white adipose tissue (WAT) has been reported. The aim of this study was to identify a strong candidate gene for Pbwg1.5 by an integration approach of transcriptome analysis (RNA-sequencing followed by real-time PCR analysis) and the causal inference test (CIT), a statistical method to infer causal relationships between diplotypes, gene expression and trait values. Body weight, body composition and biochemical traits were measured in F2 mice obtained from an intercross between the C57BL/6JJcl strain and a congenic strain carrying Pbwg1.5 on the C57BL/6JJcl background. The F2 mice showed significant diplotype differences in 12 traits including body weight, WAT weight and serum cholesterol/triglyceride levels. The transcriptome analysis revealed that Ly75, Pla2r1, Fap and Gca genes were differentially expressed in the liver and that Fap, Ifih1 and Grb14 were differentially expressed in WAT. However, CITs indicated statistical evidence that only the liver Ly75 gene mediated between genotype and WAT. Ly75 expression was negatively associated with WAT weight. The results suggested that Ly75 is a putative quantitative trait gene for the obesity-resistant Pbwg1.5 QTL discovered from the wild M. m. castaneus mouse. The finding provides a novel insight into a better understanding of the genetic basis for prevention of obesity.
Collapse
Affiliation(s)
- Akira Ishikawa
- Laboratory of Animal Genetics, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Aichi, Japan
- * E-mail:
| |
Collapse
|
18
|
Butler AA, St-Onge MP, Siebert EA, Medici V, Stanhope KL, Havel PJ. Differential Responses of Plasma Adropin Concentrations To Dietary Glucose or Fructose Consumption In Humans. Sci Rep 2015; 5:14691. [PMID: 26435060 PMCID: PMC4592955 DOI: 10.1038/srep14691] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 09/04/2015] [Indexed: 12/16/2022] Open
Abstract
Adropin is a peptide hormone encoded by the Energy Homeostasis Associated (ENHO) gene whose physiological role in humans remains incompletely defined. Here we investigated the impact of dietary interventions that affect systemic glucose and lipid metabolism on plasma adropin concentrations in humans. Consumption of glucose or fructose as 25% of daily energy requirements (E) differentially affected plasma adropin concentrations (P < 0.005) irrespective of duration, sex or age. Glucose consumption reduced plasma adropin from 3.55 ± 0.26 to 3.28 ± 0.23 ng/ml (N = 42). Fructose consumption increased plasma adropin from 3.63 ± 0.29 to 3.93 ± 0.34 ng/ml (N = 45). Consumption of high fructose corn syrup (HFCS) as 25% E had no effect (3.43 ± 0.32 versus 3.39 ± 0.24 ng/ml, N = 26). Overall, the effect of glucose, HFCS and fructose on circulating adropin concentrations were similar to those observed on postprandial plasma triglyceride concentrations. Furthermore, increases in plasma adropin levels with fructose intake were most robust in individuals exhibiting hypertriglyceridemia. Individuals with low plasma adropin concentrations also exhibited rapid increases in plasma levels following consumption of breakfasts supplemented with lipids. These are the first results linking plasma adropin levels with dietary sugar intake in humans, with the impact of fructose consumption linked to systemic triglyceride metabolism. In addition, dietary fat intake may also increase circulating adropin concentrations.
Collapse
Affiliation(s)
- Andrew A Butler
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, 1402 South Grand Boulevard, St Louis, Missouri 63104, USA
| | | | - Emily A Siebert
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, 1402 South Grand Boulevard, St Louis, Missouri 63104, USA
| | - Valentina Medici
- Division of Gastroenterology, School of Medicine , University of California at Davis, 1 Shields Ave, Davis, CA 95616
| | - Kimber L Stanhope
- Departments of Nutrition and Molecular Biosciences, School of Veterinary Medicine, University of California at Davis, 1 Shields Ave, Davis, CA 95616
| | - Peter J Havel
- Departments of Nutrition and Molecular Biosciences, School of Veterinary Medicine, University of California at Davis, 1 Shields Ave, Davis, CA 95616
| |
Collapse
|
19
|
Nikolskiy I, Conrad DF, Chun S, Fay JC, Cheverud JM, Lawson HA. Using whole-genome sequences of the LG/J and SM/J inbred mouse strains to prioritize quantitative trait genes and nucleotides. BMC Genomics 2015; 16:415. [PMID: 26016481 PMCID: PMC4445795 DOI: 10.1186/s12864-015-1592-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 04/28/2015] [Indexed: 12/04/2022] Open
Abstract
Background The laboratory mouse is the most commonly used model for studying variation in complex traits relevant to human disease. Here we present the whole-genome sequences of two inbred strains, LG/J and SM/J, which are frequently used to study variation in complex traits as diverse as aging, bone-growth, adiposity, maternal behavior, and methamphetamine sensitivity. Results We identified small nucleotide variants (SNVs) and structural variants (SVs) in the LG/J and SM/J strains relative to the reference genome and discovered novel variants in these two strains by comparing their sequences to other mouse genomes. We find that 39% of the LG/J and SM/J genomes are identical-by-descent (IBD). We characterized amino-acid changing mutations using three algorithms: LRT, PolyPhen-2 and SIFT. We also identified polymorphisms between LG/J and SM/J that fall in regulatory regions and highly informative transcription factor binding sites (TFBS). We intersected these functional predictions with quantitative trait loci (QTL) mapped in advanced intercrosses of these two strains. We find that QTL are both over-represented in non-IBD regions and highly enriched for variants predicted to have a functional impact. Variants in QTL associated with metabolic (231 QTL identified in an F16 generation) and developmental (41 QTL identified in an F34 generation) traits were interrogated and we highlight candidate quantitative trait genes (QTG) and nucleotides (QTN) in a QTL on chr13 associated with variation in basal glucose levels and in a QTL on chr6 associated with variation in tibia length. Conclusions We show how integrating genomic sequence with QTL reduces the QTL search space and helps researchers prioritize candidate genes and nucleotides for experimental follow-up. Additionally, given the LG/J and SM/J phylogenetic context among inbred strains, these data contribute important information to the genomic landscape of the laboratory mouse. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-1592-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Igor Nikolskiy
- Department of Genetics, Washington University School of Medicine, Campus Box 8108, 660 S Euclid Ave, St Louis, MO, 63110, USA.
| | - Donald F Conrad
- Department of Genetics, Washington University School of Medicine, Campus Box 8108, 660 S Euclid Ave, St Louis, MO, 63110, USA.
| | - Sung Chun
- Division of Genetics, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| | - Justin C Fay
- Department of Genetics, Washington University School of Medicine, Campus Box 8108, 660 S Euclid Ave, St Louis, MO, 63110, USA.
| | | | - Heather A Lawson
- Department of Genetics, Washington University School of Medicine, Campus Box 8108, 660 S Euclid Ave, St Louis, MO, 63110, USA.
| |
Collapse
|
20
|
Gao S, McMillan RP, Zhu Q, Lopaschuk GD, Hulver MW, Butler AA. Therapeutic effects of adropin on glucose tolerance and substrate utilization in diet-induced obese mice with insulin resistance. Mol Metab 2015; 4:310-24. [PMID: 25830094 PMCID: PMC4354928 DOI: 10.1016/j.molmet.2015.01.005] [Citation(s) in RCA: 123] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 01/06/2015] [Accepted: 01/09/2015] [Indexed: 12/11/2022] Open
Abstract
Objective The peptide hormone adropin regulates fuel selection preferences in skeletal muscle under fed and fasted conditions. Here, we investigated whether adropin treatment can ameliorate the dysregulation of fuel substrate metabolism, and improve aspects of glucose homeostasis in diet-induced obesity (DIO) with insulin resistance. Methods DIO C57BL/6 mice maintained on a 60% kcal fat diet received five intraperitoneal (i.p.) injections of the bioactive peptide adropin34-76 (450 nmol/kg/i.p.). Following treatment, glucose tolerance and whole body insulin sensitivity were assessed and indirect calorimetry was employed to analyze whole body substrate oxidation preferences. Biochemical assays performed in skeletal muscle samples analyzed insulin signaling action and substrate oxidation. Results Adropin treatment improved glucose tolerance, enhanced insulin action and augmented metabolic flexibility towards glucose utilization. In muscle, adropin treatment increased insulin-induced Akt phosphorylation and cell-surface expression of GLUT4 suggesting sensitization of insulin signaling pathways. Reduced incomplete fatty acid oxidation and increased CoA/acetyl-CoA ratio suggested improved mitochondrial function. The underlying mechanisms appear to involve suppressions of carnitine palmitoyltransferase-1B (CPT-1B) and CD36, two key enzymes in fatty acid utilization. Adropin treatment activated pyruvate dehydrogenase (PDH), a rate-limiting enzyme in glucose oxidation, and downregulated PDH kinase-4 (PDK-4) that inhibits PDH. Along with these changes, adropin treatment downregulated peroxisome proliferator-activated receptor-gamma coactivator-1α that regulates expression of Cpt1b, Cd36 and Pdk4. Conclusions Adropin treatment of DIO mice enhances glucose tolerance, ameliorates insulin resistance and promotes preferential use of carbohydrate over fat in fuel selection. Skeletal muscle is a key organ in mediating adropin's whole-body effects, sensitizing insulin signaling pathways and altering fuel selection preference to favor glucose while suppressing fat oxidation.
Collapse
Affiliation(s)
- Su Gao
- Department of Metabolism and Aging, Scripps Research Institute, Jupiter, FL, USA
| | - Ryan P. McMillan
- Department of Human Nutrition, Foods and Exercise, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Qingzhang Zhu
- Department of Metabolism and Aging, Scripps Research Institute, Jupiter, FL, USA
| | - Gary D. Lopaschuk
- Department of Pediatrics, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, AB, Canada
| | - Matthew W. Hulver
- Department of Human Nutrition, Foods and Exercise, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Andrew A. Butler
- Department of Metabolism and Aging, Scripps Research Institute, Jupiter, FL, USA
- Department of Pharmacological & Physiological Science, Saint Louis University School of Medicine, Saint Louis, MO, USA
- Corresponding author. Pharmacological & Physiological Science, Saint Louis University School of Medicine, 1402 S Grand Blvd, St Louis, MO 63104, USA. Tel.: +1 314 977 6425; fax: +1 314 977 6410.
| |
Collapse
|
21
|
Sáinz N, Barrenetxe J, Moreno-Aliaga MJ, Martínez JA. Leptin resistance and diet-induced obesity: central and peripheral actions of leptin. Metabolism 2015; 64:35-46. [PMID: 25497342 DOI: 10.1016/j.metabol.2014.10.015] [Citation(s) in RCA: 290] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 10/15/2014] [Accepted: 10/20/2014] [Indexed: 12/15/2022]
Abstract
Obesity is a chronic disease that represents one of the most serious global health burdens associated to an excess of body fat resulting from an imbalance between energy intake and expenditure, which is regulated by environmental and genetic interactions. The adipose-derived hormone leptin acts via a specific receptor in the brain to regulate energy balance and body weight, although this protein can also elicit a myriad of actions in peripheral tissues. Obese individuals, rather than be leptin deficient, have in most cases, high levels of circulating leptin. The failure of these high levels to control body weight suggests the presence of a resistance process to the hormone that could be partly responsible of disturbances on body weight regulation. Furthermore, leptin resistance can impair physiological peripheral functions of leptin such as lipid and carbohydrate metabolism and nutrient intestinal utilization. The present document summarizes those findings regarding leptin resistance development and the role of this hormone in the development and maintenance of an obese state. Thus, we focused on the effect of the impaired leptin action on adipose tissue, liver, skeletal muscle and intestinal function and the accompanying relationships with diet-induced obesity. The involvement of some inflammatory mediators implicated in the development of obesity and their roles in leptin resistance development are also discussed.
Collapse
Affiliation(s)
- Neira Sáinz
- Department of Nutrition, Food Sciences and Physiology, University of Navarra, C/Irunlarrea 1, 31008 Pamplona, Spain
| | - Jaione Barrenetxe
- Department of Nutrition, Food Sciences and Physiology, University of Navarra, C/Irunlarrea 1, 31008 Pamplona, Spain
| | - María J Moreno-Aliaga
- Department of Nutrition, Food Sciences and Physiology, University of Navarra, C/Irunlarrea 1, 31008 Pamplona, Spain; CIBER Fisiopatología de la Obesidad y la Nutrición (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - José Alfredo Martínez
- Department of Nutrition, Food Sciences and Physiology, University of Navarra, C/Irunlarrea 1, 31008 Pamplona, Spain; CIBER Fisiopatología de la Obesidad y la Nutrición (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain.
| |
Collapse
|
22
|
Ishikawa A, Okuno SI. Fine mapping and candidate gene search of quantitative trait loci for growth and obesity using mouse intersubspecific subcongenic intercrosses and exome sequencing. PLoS One 2014; 9:e113233. [PMID: 25398139 PMCID: PMC4232600 DOI: 10.1371/journal.pone.0113233] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 10/26/2014] [Indexed: 12/20/2022] Open
Abstract
Although growth and body composition traits are quantitative traits of medical and agricultural importance, the genetic and molecular basis of those traits remains elusive. Our previous genome-wide quantitative trait locus (QTL) analyses in an intersubspecific backcross population between C57BL/6JJcl (B6) and wild Mus musculus castaneus mice revealed a major growth QTL (named Pbwg1) on a proximal region of mouse chromosome 2. Using the B6.Cg-Pbwg1 intersubspecific congenic strain created, we revealed 12 closely linked QTLs for body weight and body composition traits on an approximately 44.1-Mb wild-derived congenic region. In this study, we narrowed down genomic regions harboring three (Pbwg1.12, Pbwg1.3 and Pbwg1.5) of the 12 linked QTLs and searched for possible candidate genes for the QTLs. By phenotypic analyses of F2 intercross populations between B6 and each of four B6.Cg-Pbwg1 subcongenic strains with overlapping and non-overlapping introgressed regions, we physically defined Pbwg1.12 affecting body weight to a 3.8-Mb interval (61.5-65.3 Mb) on chromosome 2. We fine-mapped Pbwg1.3 for body length to an 8.0-Mb interval (57.3-65.3) and Pbwg1.5 for abdominal white fat weight to a 2.1-Mb interval (59.4-61.5). The wild-derived allele at Pbwg1.12 and Pbwg1.3 uniquely increased body weight and length despite the fact that the wild mouse has a smaller body size than that of B6, whereas it decreased fat weight at Pbwg1.5. Exome sequencing and candidate gene prioritization suggested that Gcg and Grb14 are putative candidate genes for Pbwg1.12 and that Ly75 and Itgb6 are putative candidate genes for Pbwg1.5. These genes had nonsynonymous SNPs, but the SNPs were predicted to be not harmful to protein functions. These results provide information helpful to identify wild-derived quantitative trait genes causing enhanced growth and resistance to obesity.
Collapse
Affiliation(s)
- Akira Ishikawa
- Laboratory of Animal Genetics, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Aichi, Japan
- * E-mail:
| | - Sin-ichiro Okuno
- Laboratory of Animal Genetics, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Aichi, Japan
| |
Collapse
|
23
|
Kalafati IP, Borsa D, Dedoussis GVZ. The Genetics of Nonalcoholic Fatty Liver Disease: Role of Diet as a Modifying Factor. Curr Nutr Rep 2014. [DOI: 10.1007/s13668-014-0085-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|