1
|
Tesema B, Liu GQ, Jiang XP. Active kisspeptin DNA vaccines oral immunization disrupt mRNA hormone receptors expression in ram lambs. Anim Biotechnol 2023; 34:2285-2294. [PMID: 35714982 DOI: 10.1080/10495398.2022.2087665] [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] [Indexed: 11/01/2022]
Abstract
To evaluate the efficacy of oral immunization with active kisspeptin DNA vaccine on the expression of hormone receptor mRNA. For this study, ten 56-day-old Hu breed ram lambs were randomly assigned to the treatment and control groups (n = 5). Treatment Experimental group received C500/pKS-asd and the control group received C500/pVAX-asd (aspartate-β semialdehyde dehydrogenase orally on days 0, 28, and 56, and blood samples were taken at each immunization interval (14-day) and tissues samples were collected at the end of the experimental period (day 98). The collected samples were stored in the refrigerator at -20 °C and liquid nitrogen, respectively, for laboratory examination. Total RNA was extracted from samples using TRIzol reagent and quantitative real-time polymerase chain reaction (QPCR) was used to quantify the levels of KISS1, G protein-coupled receptor-54 (Kiss1r), and gonadotrophin-releasing hormone (GnRH) mRNA in the hypothalamus. Levels of luteinizing hormone receptor (LHR) and luteinizing hormone beta (LHβ) mRNA, and follicle-stimulating hormone receptor (FSHR) and follicle-stimulating hormone beta (FSHβ) mRNA in the testes and pituitary were analyzed, respectively. Further, gonadotropin-releasing hormone receptor (GnRHR) mRNA expression level in the pituitary was measured. Moreover, the Kiss1r concentration level in the blood was measured using an indirect ELISA. The concentration of Kiss1r in the blood was lower in the treatment group than in the control group (p < 0.05). The levels of testicular FSHR and LHR mRNA were significantly lower in the treatment group (p < 0.05) when compared to the control group. Furthermore, the treatment group's levels of hypothalamic KISS1, Kiss1r, and GnRH mRNA were significantly lower (p < 0.05) than the controls. LH, FSH, and GnRHR mRNA expression in the pituitary were also significantly lower in the treatment group (p < 0.01 and p < 0.05, respectively). These findings imply that oral immunization with active kisspeptin DNA vaccine suppresses hormone receptor mRNA expression in the ram lambs.
Collapse
Affiliation(s)
- Birhanu Tesema
- Key Laboratory of Agricultural - Animal Genetics, Breeding, and Reproduction, Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, People's Republic of China
- Department of Animal Science, College of Agricultural Sciences, Bule Hora University, Bule Hora, Ethiopia
| | - Gui-Qiang Liu
- Key Laboratory of Agricultural - Animal Genetics, Breeding, and Reproduction, Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Xun-Ping Jiang
- Key Laboratory of Agricultural - Animal Genetics, Breeding, and Reproduction, Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, People's Republic of China
| |
Collapse
|
2
|
The role of leptin and low testosterone in obesity. Int J Impot Res 2022; 34:704-713. [DOI: 10.1038/s41443-022-00534-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 01/21/2022] [Indexed: 12/29/2022]
|
3
|
LeDuc CA, Skowronski AA, Rosenbaum M. The Role of Leptin in the Development of Energy Homeostatic Systems and the Maintenance of Body Weight. Front Physiol 2021; 12:789519. [PMID: 34955895 PMCID: PMC8703217 DOI: 10.3389/fphys.2021.789519] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 11/17/2021] [Indexed: 12/12/2022] Open
Abstract
LEP is a pleiotropic gene and the actions of leptin extend well beyond simply acting as the signal of the size of adipose tissue stores originally proposed. This is a discussion of the multi-system interactions of leptin with the development of the neural systems regulating energy stores, and the subsequent maintenance of energy stores throughout the lifespan. The prenatal, perinatal, and later postnatal effects of leptin on systems regulating body energy stores and on the energy stores themselves are heavily influenced by the nutritional environment which leptin exposure occurs. This review discusses the prenatal and perinatal roles of leptin in establishing the neuronal circuitry and other systems relevant to the adiposity set-point (or “threshold”) and the role of leptin in maintaining weight homeostasis in adulthood. Therapeutic manipulation of the intrauterine environment, use of leptin sensitizing agents, and identification of specific cohorts who may be more responsive to leptin or other means of activating the leptin signaling pathway are ripe areas for future research.
Collapse
Affiliation(s)
- Charles A LeDuc
- Division of Molecular Genetics, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, United States
| | - Alicja A Skowronski
- Division of Molecular Genetics, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, United States
| | - Michael Rosenbaum
- Division of Molecular Genetics, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, United States
| |
Collapse
|
4
|
Dadousis C, Somavilla A, Ilska JJ, Johnsson M, Batista L, Mellanby RJ, Headon D, Gottardo P, Whalen A, Wilson D, Dunn IC, Gorjanc G, Kranis A, Hickey JM. A genome-wide association analysis for body weight at 35 days measured on 137,343 broiler chickens. Genet Sel Evol 2021; 53:70. [PMID: 34496773 PMCID: PMC8424881 DOI: 10.1186/s12711-021-00663-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 08/23/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Body weight (BW) is an economically important trait in the broiler (meat-type chickens) industry. Under the assumption of polygenicity, a "large" number of genes with "small" effects is expected to control BW. To detect such effects, a large sample size is required in genome-wide association studies (GWAS). Our objective was to conduct a GWAS for BW measured at 35 days of age with a large sample size. METHODS The GWAS included 137,343 broilers spanning 15 pedigree generations and 392,295 imputed single nucleotide polymorphisms (SNPs). A false discovery rate of 1% was adopted to account for multiple testing when declaring significant SNPs. A Bayesian ridge regression model was implemented, using AlphaBayes, to estimate the contribution to the total genetic variance of each region harbouring significant SNPs (1 Mb up/downstream) and the combined regions harbouring non-significant SNPs. RESULTS GWAS revealed 25 genomic regions harbouring 96 significant SNPs on 13 Gallus gallus autosomes (GGA1 to 4, 8, 10 to 15, 19 and 27), with the strongest associations on GGA4 at 65.67-66.31 Mb (Galgal4 assembly). The association of these regions points to several strong candidate genes including: (i) growth factors (GGA1, 4, 8, 13 and 14); (ii) leptin receptor overlapping transcript (LEPROT)/leptin receptor (LEPR) locus (GGA8), and the STAT3/STAT5B locus (GGA27), in connection with the JAK/STAT signalling pathway; (iii) T-box gene (TBX3/TBX5) on GGA15 and CHST11 (GGA1), which are both related to heart/skeleton development); and (iv) PLAG1 (GGA2). Combined together, these 25 genomic regions explained ~ 30% of the total genetic variance. The region harbouring significant SNPs that explained the largest portion of the total genetic variance (4.37%) was on GGA4 (~ 65.67-66.31 Mb). CONCLUSIONS To the best of our knowledge, this is the largest GWAS that has been conducted for BW in chicken to date. In spite of the identified regions, which showed a strong association with BW, the high proportion of genetic variance attributed to regions harbouring non-significant SNPs supports the hypothesis that the genetic architecture of BW35 is polygenic and complex. Our results also suggest that a large sample size will be required for future GWAS of BW35.
Collapse
Affiliation(s)
| | | | - Joanna J. Ilska
- The Roslin Institute, University of Edinburgh, Midlothian, UK
| | - Martin Johnsson
- The Roslin Institute, University of Edinburgh, Midlothian, UK
- Department of Animal Breeding and Genetics, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Lorena Batista
- The Roslin Institute, University of Edinburgh, Midlothian, UK
| | | | - Denis Headon
- The Roslin Institute, University of Edinburgh, Midlothian, UK
| | - Paolo Gottardo
- Italian Brown Breeders Association, Loc. Ferlina 204, 37012 Bussolengo, Italy
| | - Andrew Whalen
- The Roslin Institute, University of Edinburgh, Midlothian, UK
| | - David Wilson
- The Roslin Institute, University of Edinburgh, Midlothian, UK
| | - Ian C. Dunn
- The Roslin Institute, University of Edinburgh, Midlothian, UK
| | - Gregor Gorjanc
- The Roslin Institute, University of Edinburgh, Midlothian, UK
| | - Andreas Kranis
- The Roslin Institute, University of Edinburgh, Midlothian, UK
- Aviagen Ltd, Midlothian, UK
| | - John M. Hickey
- The Roslin Institute, University of Edinburgh, Midlothian, UK
| |
Collapse
|
5
|
Yaghootkar H, Zhang Y, Spracklen CN, Karaderi T, Huang LO, Bradfield J, Schurmann C, Fine RS, Preuss MH, Kutalik Z, Wittemans LBL, Lu Y, Metz S, Willems SM, Li-Gao R, Grarup N, Wang S, Molnos S, Sandoval-Zárate AA, Nalls MA, Lange LA, Haesser J, Guo X, Lyytikäinen LP, Feitosa MF, Sitlani CM, Venturini C, Mahajan A, Kacprowski T, Wang CA, Chasman DI, Amin N, Broer L, Robertson N, Young KL, Allison M, Auer PL, Blüher M, Borja JB, Bork-Jensen J, Carrasquilla GD, Christofidou P, Demirkan A, Doege CA, Garcia ME, Graff M, Guo K, Hakonarson H, Hong J, Ida Chen YD, Jackson R, Jakupović H, Jousilahti P, Justice AE, Kähönen M, Kizer JR, Kriebel J, LeDuc CA, Li J, Lind L, Luan J, Mackey DA, Mangino M, Männistö S, Martin Carli JF, Medina-Gomez C, Mook-Kanamori DO, Morris AP, de Mutsert R, Nauck M, Prokic I, Pennell CE, Pradhan AD, Psaty BM, Raitakari OT, Scott RA, Skaaby T, Strauch K, Taylor KD, Teumer A, Uitterlinden AG, Wu Y, Yao J, Walker M, North KE, Kovacs P, Ikram MA, van Duijn CM, Ridker PM, Lye S, Homuth G, Ingelsson E, Spector TD, McKnight B, Province MA, Lehtimäki T, Adair LS, Rotter JI, Reiner AP, Wilson JG, Harris TB, Ripatti S, Grallert H, Meigs JB, Salomaa V, Hansen T, Willems van Dijk K, Wareham NJ, Grant SFA, Langenberg C, Frayling TM, Lindgren CM, Mohlke KL, Leibel RL, Loos RJF, Kilpeläinen TO. Genetic Studies of Leptin Concentrations Implicate Leptin in the Regulation of Early Adiposity. Diabetes 2020; 69:2806-2818. [PMID: 32917775 PMCID: PMC7679778 DOI: 10.2337/db20-0070] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 09/09/2020] [Indexed: 02/02/2023]
Abstract
Leptin influences food intake by informing the brain about the status of body fat stores. Rare LEP mutations associated with congenital leptin deficiency cause severe early-onset obesity that can be mitigated by administering leptin. However, the role of genetic regulation of leptin in polygenic obesity remains poorly understood. We performed an exome-based analysis in up to 57,232 individuals of diverse ancestries to identify genetic variants that influence adiposity-adjusted leptin concentrations. We identify five novel variants, including four missense variants, in LEP, ZNF800, KLHL31, and ACTL9, and one intergenic variant near KLF14. The missense variant Val94Met (rs17151919) in LEP was common in individuals of African ancestry only, and its association with lower leptin concentrations was specific to this ancestry (P = 2 × 10-16, n = 3,901). Using in vitro analyses, we show that the Met94 allele decreases leptin secretion. We also show that the Met94 allele is associated with higher BMI in young African-ancestry children but not in adults, suggesting that leptin regulates early adiposity.
Collapse
Affiliation(s)
- Hanieh Yaghootkar
- Genetics of Complex Traits, University of Exeter Medical School, Royal Devon & Exeter Hospital, Exeter, U.K.
- Division of Medical Sciences, Department of Health Sciences, Luleå University of Technology, Luleå, Sweden
- Research Centre for Optimal Health, School of Life Sciences, University of Westminster, London, U.K
| | - Yiying Zhang
- Division of Molecular Genetics, Department of Pediatrics, Columbia University, New York, NY
| | - Cassandra N Spracklen
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Biostatistics and Epidemiology, University of Massachusetts-Amherst, Amherst, MA
| | - Tugce Karaderi
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, U.K
- Department of Biological Sciences, Faculty of Arts and Sciences, Eastern Mediterranean University, Famagusta, Cyprus
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- DTU Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Lam Opal Huang
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jonathan Bradfield
- Center for Applied Genomics, Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, PA
- Quantinuum Research LLC, San Diego, CA
| | - Claudia Schurmann
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Rebecca S Fine
- Department of Genetics, Harvard Medical School, Boston, MA
- Division of Endocrinology and Center for Basic and Translational Obesity Research, Boston Children's Hospital, Boston, MA
- Broad Institute of MIT and Harvard, Cambridge, MA
| | - Michael H Preuss
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Zoltan Kutalik
- Genetics of Complex Traits, University of Exeter Medical School, Royal Devon & Exeter Hospital, Exeter, U.K
- Center for Primary Care and Public Health, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Laura B L Wittemans
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, U.K
- MRC Epidemiology Unit, University of Cambridge, Cambridge, U.K
| | - Yingchang Lu
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
- Division of Epidemiology, Department of Medicine, Vanderbilt-Ingram Cancer Center, and Vanderbilt Epidemiology Center, Vanderbilt University School of Medicine, Nashville, TN
| | - Sophia Metz
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sara M Willems
- MRC Epidemiology Unit, University of Cambridge, Cambridge, U.K
| | - Ruifang Li-Gao
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Niels Grarup
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Shuai Wang
- Department of Biostatistics, Boston University School of Public Health, Boston, MA
| | - Sophie Molnos
- German Center for Diabetes Research, München-Neuherberg, Germany
- Research Unit of Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München Research Center for Environmental Health, München-Neuherberg, Germany
| | | | - Mike A Nalls
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD
- Data Tecnica International, Glen Echo, MD
| | - Leslie A Lange
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado-Denver, Denver, CO
| | - Jeffrey Haesser
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
- Department of Clinical Chemistry, Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Mary F Feitosa
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO
| | - Colleen M Sitlani
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA
| | - Cristina Venturini
- Department of Twin Research and Genetic Epidemiology, Kings College London, London, U.K
| | - Anubha Mahajan
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, U.K
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, U.K
| | - Tim Kacprowski
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
| | - Carol A Wang
- Department of Biostatistics, Boston University School of Public Health, Boston, MA
| | - Daniel I Chasman
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Najaf Amin
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Linda Broer
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Neil Robertson
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, U.K
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, U.K
| | - Kristin L Young
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Matthew Allison
- Department of Family Medicine and Public Health, University of California, San Diego, La Jolla, CA
| | - Paul L Auer
- Joseph J. Zilber School of Public Health, University of Wisconsin-Milwaukee, Milwaukee, WI
| | - Matthias Blüher
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
| | - Judith B Borja
- Office of Population Studies Foundation, Inc., Cebu City, Philippines
- Department of Nutrition and Dietetics, University of San Carlos, Cebu City, Philippines
| | - Jette Bork-Jensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Germán D Carrasquilla
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Ayse Demirkan
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Claudia A Doege
- Department of Pathology and Cell Biology, Columbia University, New York, NY
| | - Melissa E Garcia
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Bethesda, MD
| | - Mariaelisa Graff
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Carolina Center for Genome Sciences, Chapel Hill, NC
| | - Kaiying Guo
- Division of Molecular Genetics, Department of Pediatrics, Columbia University, New York, NY
| | - Hakon Hakonarson
- Center for Applied Genomics, Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, PA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Jaeyoung Hong
- Department of Biostatistics, Boston University School of Public Health, Boston, MA
| | - Yii-Der Ida Chen
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
| | - Rebecca Jackson
- Division of Endocrinology, Diabetes, and Metabolism, Ohio State University, Columbus, OH
| | - Hermina Jakupović
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Pekka Jousilahti
- Department of Public Health Solutions, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Anne E Justice
- Center for Biomedical and Translational Informatics, Geisinger, Danville, PA
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital, Tampere, Finland
- Department of Clinical Physiology, Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Jorge R Kizer
- Cardiology Section, San Francisco Veterans Affairs Health Care System, University of California San Francisco, San Francisco, CA
- Departments of Medicine and Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA
| | - Jennifer Kriebel
- German Center for Diabetes Research, München-Neuherberg, Germany
- Research Unit of Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München Research Center for Environmental Health, München-Neuherberg, Germany
| | - Charles A LeDuc
- Division of Molecular Genetics, Department of Pediatrics, Columbia University, New York, NY
| | - Jin Li
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Palo Alto, CA
| | - Lars Lind
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Jian'an Luan
- MRC Epidemiology Unit, University of Cambridge, Cambridge, U.K
| | - David A Mackey
- Centre for Ophthalmology and Visual Science, Lions Eye Institute, The University of Western Australia, Perth, West Australia, Australia
| | - Massimo Mangino
- Department of Twin Research and Genetic Epidemiology, Kings College London, London, U.K
- NIHR Biomedical Research Centre at Guy's and St Thomas' Foundation Trust, London, U.K
| | - Satu Männistö
- Department of Public Health Solutions, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Jayne F Martin Carli
- Division of Molecular Genetics, Department of Pediatrics, Columbia University, New York, NY
| | - Carolina Medina-Gomez
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, the Netherlands
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Dennis O Mook-Kanamori
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
- Department of Public Health and Primary Care, Leiden University Medical Center, Leiden, the Netherlands
| | - Andrew P Morris
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, U.K
- Department of Biostatistics, University of Liverpool, Liverpool, U.K
- Division of Musculoskeletal and Dermatological Sciences, University of Manchester, Manchester, U.K
| | - Renée de Mutsert
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Matthias Nauck
- DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Ivana Prokic
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Craig E Pennell
- Department of Biostatistics, Boston University School of Public Health, Boston, MA
| | - Arund D Pradhan
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Departments of Epidemiology, Medicine, and Health Services, University of Washington, Seattle, WA
- Kaiser Permanente Washington Health Research Institute, Seattle, WA
| | - Olli T Raitakari
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
- Research Centre of Applied and Preventive Cardiovascular Medicine, Turku University Hospital, Turku, Finland
| | - Robert A Scott
- MRC Epidemiology Unit, University of Cambridge, Cambridge, U.K
| | - Tea Skaaby
- Center for Clinical Research and Disease Prevention, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
| | - Konstantin Strauch
- Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Chair of Genetic Epidemiology, Insitute of Medical Information Processing, Biometry, and Epidemiology (IBE), Faculty of Medicine, Ludwig Maximilian University Munich, München, Germany
| | - Kent D Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
| | - Alexander Teumer
- DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Andre G Uitterlinden
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, the Netherlands
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Ying Wu
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Jie Yao
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
| | - Mark Walker
- Institute of Cellular Medicine (Diabetes), Newcastle University, Newcastle upon Tyne, U.K
| | - Kari E North
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Peter Kovacs
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, the Netherlands
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Cornelia M van Duijn
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Paul M Ridker
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Stephen Lye
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Georg Homuth
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Erik Ingelsson
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Palo Alto, CA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Palo Alto, CA
- Stanford Diabetes Research Center, Stanford University, Stanford, CA
- Molecular Epidemiology and Science for Life Laboratory, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Tim D Spector
- Department of Twin Research and Genetic Epidemiology, Kings College London, London, U.K
| | - Barbara McKnight
- Department of Biostatistics, University of Washington, Seattle, WA
| | - Michael A Province
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
- Department of Clinical Chemistry, Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Linda S Adair
- Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
| | - Alexander P Reiner
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - James G Wilson
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS
| | - Tamara B Harris
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Bethesda, MD
| | - Samuli Ripatti
- Broad Institute of MIT and Harvard, Cambridge, MA
- Institute for Molecular Medicine Finland, Helsinki, Finland
- Public Health, University of Helsinki, Helsinki, Finland
| | - Harald Grallert
- German Center for Diabetes Research, München-Neuherberg, Germany
- Research Unit of Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München Research Center for Environmental Health, München-Neuherberg, Germany
| | - James B Meigs
- Division of General Internal Medicine, Massachusetts General Hospital, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
- Program in Population and Medical Genetics, Broad Institute of MIT and Harvard, Cambridge, MA
| | - Veikko Salomaa
- Department of Public Health Solutions, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ko Willems van Dijk
- Division of Endocrinology, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden, the Netherlands
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Struan F A Grant
- Center for Applied Genomics, Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, PA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Center for Spatial and Functional Genomics, Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, PA
- Division of Endocrinology and Diabetes, The Children's Hospital of Philadelphia, Philadelphia, PA
- Institute of Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | | | - Timothy M Frayling
- Genetics of Complex Traits, University of Exeter Medical School, Royal Devon & Exeter Hospital, Exeter, U.K
| | - Cecilia M Lindgren
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, U.K
- Broad Institute of MIT and Harvard, Cambridge, MA
- Big Data Institute, Nuffield Department of Medicine, University of Oxford, Oxford, U.K
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Rudolph L Leibel
- Division of Molecular Genetics, Department of Pediatrics, Columbia University, New York, NY
| | - Ruth J F Loos
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Tuomas O Kilpeläinen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
6
|
Wang L, Sinnott-Armstrong N, Wagschal A, Wark AR, Camporez JP, Perry RJ, Ji F, Sohn Y, Oh J, Wu S, Chery J, Moud BN, Saadat A, Dankel SN, Mellgren G, Tallapragada DSP, Strobel SM, Lee MJ, Tewhey R, Sabeti PC, Schaefer A, Petri A, Kauppinen S, Chung RT, Soukas A, Avruch J, Fried SK, Hauner H, Sadreyev RI, Shulman GI, Claussnitzer M, Näär AM. A MicroRNA Linking Human Positive Selection and Metabolic Disorders. Cell 2020; 183:684-701.e14. [PMID: 33058756 PMCID: PMC8092355 DOI: 10.1016/j.cell.2020.09.017] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 05/08/2020] [Accepted: 09/03/2020] [Indexed: 01/09/2023]
Abstract
Positive selection in Europeans at the 2q21.3 locus harboring the lactase gene has been attributed to selection for the ability of adults to digest milk to survive famine in ancient times. However, the 2q21.3 locus is also associated with obesity and type 2 diabetes in humans, raising the possibility that additional genetic elements in the locus may have contributed to evolutionary adaptation to famine by promoting energy storage, but which now confer susceptibility to metabolic diseases. We show here that the miR-128-1 microRNA, located at the center of the positively selected locus, represents a crucial metabolic regulator in mammals. Antisense targeting and genetic ablation of miR-128-1 in mouse metabolic disease models result in increased energy expenditure and amelioration of high-fat-diet-induced obesity and markedly improved glucose tolerance. A thrifty phenotype connected to miR-128-1-dependent energy storage may link ancient adaptation to famine and modern metabolic maladaptation associated with nutritional overabundance.
Collapse
Affiliation(s)
- Lifeng Wang
- Massachusetts General Hospital Cancer Center, Charlestown, MA 02129, USA,Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA,These authors contributed equally,Present address: Cardiovascular & Metabolism, Janssen Pharmaceutical Companies of Johnson & Johnson, Spring House, PA 19477, USA
| | - Nasa Sinnott-Armstrong
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA,Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA,These authors contributed equally
| | - Alexandre Wagschal
- Massachusetts General Hospital Cancer Center, Charlestown, MA 02129, USA,Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA,Present address: Vertex Pharmaceuticals, Watertown, MA 02472, USA
| | - Abigail R. Wark
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Joao-Paulo Camporez
- Departments of Internal Medicine and Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT 06510, USA,Present address: Ribeirao Preto School of Medicine, University of Sao Paulo, Sao Paulo 14049-90, Brazil
| | - Rachel J. Perry
- Departments of Internal Medicine and Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Fei Ji
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Yoojin Sohn
- Massachusetts General Hospital Cancer Center, Charlestown, MA 02129, USA,Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA,Present address: Vanderbilt University, Nashville, TN 37235, USA
| | - Justin Oh
- Massachusetts General Hospital Cancer Center, Charlestown, MA 02129, USA,Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA,Present address: Vertex Pharmaceuticals, Watertown, MA 02472, USA
| | - Su Wu
- Massachusetts General Hospital Cancer Center, Charlestown, MA 02129, USA,Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA,Present address: Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Jessica Chery
- Massachusetts General Hospital Cancer Center, Charlestown, MA 02129, USA,Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA,Present address: Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Bahareh Nemati Moud
- Else Kroener-Fresenius-Center of Nutritional Medicine, School of Life Sciences, Technical University of Munich, 85354 Freising, Germany
| | - Alham Saadat
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Simon N. Dankel
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, 5021 Bergen, Norway,Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, 5020 Bergen, Norway
| | - Gunnar Mellgren
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, 5021 Bergen, Norway,Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, 5020 Bergen, Norway
| | - Divya Sri Priyanka Tallapragada
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, 5021 Bergen, Norway,Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, 5020 Bergen, Norway
| | - Sophie Madlen Strobel
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA,Institute of Nutritional Medicine, School of Medicine, Technical University of Munich, 80992 Munich, Germany
| | - Mi-Jeong Lee
- Obesity Center, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA,Present address: Department of Human Nutrition, Food and Animal Sciences, University of Hawaii, Honolulu, HI 96822, USA
| | - Ryan Tewhey
- Center for Systems Biology, Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA,Present address: The Jackson Laboratory, Bar Harbor, ME 04609, USA
| | - Pardis C. Sabeti
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA,Center for Systems Biology, Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA
| | - Anne Schaefer
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School ofMedicine atMount Sinai, New York, New York 10029, USA
| | - Andreas Petri
- Center for RNA Medicine, Department of Clinical Medicine, Aalborg University, 2450 Copenhagen, Denmark
| | - Sakari Kauppinen
- Center for RNA Medicine, Department of Clinical Medicine, Aalborg University, 2450 Copenhagen, Denmark
| | - Raymond T. Chung
- Liver Center, Gastrointestinal Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Alexander Soukas
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA,Department of Medicine, Center for Genomic Medicine and Diabetes Unit, Massachusetts General Hospital, Boston, MA 02114, USA,Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Joseph Avruch
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA,Department of Medicine, Harvard Medical School, Boston, MA 02114, USA,Diabetes unit, Medical Services, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Susan K. Fried
- Obesity Center, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA,Present address: Diabetes, Obesity and Metabolism Institute, Mt. Sinai School of Medicine, New York, NY 10029, USA
| | - Hans Hauner
- Else Kroener-Fresenius-Center of Nutritional Medicine, School of Life Sciences, Technical University of Munich, 85354 Freising, Germany,Institute of Nutritional Medicine, School of Medicine, Technical University of Munich, 80992 Munich, Germany
| | - Ruslan I. Sadreyev
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Gerald I. Shulman
- Departments of Internal Medicine and Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Melina Claussnitzer
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA,Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Anders M. Näär
- Massachusetts General Hospital Cancer Center, Charlestown, MA 02129, USA,Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA,Present address: Department of Nutritional Sciences & Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA,Lead Contact,Correspondence: https://doi.org/10.1016/j.cell.2020.09.017
| |
Collapse
|
7
|
Fonseca PAS, Suárez-Vega A, Cánovas A. Weighted Gene Correlation Network Meta-Analysis Reveals Functional Candidate Genes Associated with High- and Sub-Fertile Reproductive Performance in Beef Cattle. Genes (Basel) 2020; 11:genes11050543. [PMID: 32408659 PMCID: PMC7290847 DOI: 10.3390/genes11050543] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 05/04/2020] [Accepted: 05/06/2020] [Indexed: 12/13/2022] Open
Abstract
Improved reproductive efficiency could lead to economic benefits for the beef industry, once the intensive selection pressure has led to a decreased fertility. However, several factors limit our understanding of fertility traits, including genetic differences between populations and statistical limitations. In the present study, the RNA-sequencing data from uterine samples of high-fertile (HF) and sub-fertile (SF) animals was integrated using co-expression network meta-analysis, weighted gene correlation network analysis, identification of upstream regulators, variant calling, and network topology approaches. Using this pipeline, top hub-genes harboring fixed variants (HF × SF) were identified in differentially co-expressed gene modules (DcoExp). The functional prioritization analysis identified the genes with highest potential to be key-regulators of the DcoExp modules between HF and SF animals. Consequently, 32 functional candidate genes (10 upstream regulators and 22 top hub-genes of DcoExp modules) were identified. These genes were associated with the regulation of relevant biological processes for fertility, such as embryonic development, germ cell proliferation, and ovarian hormone regulation. Additionally, 100 candidate variants (single nucleotide polymorphisms (SNPs) and insertions and deletions (INDELs)) were identified within those genes. In the long-term, the results obtained here may help to reduce the frequency of subfertility in beef herds, reducing the associated economic losses caused by this condition.
Collapse
Affiliation(s)
- Pablo A. S. Fonseca
- Correspondence: (P.A.S.F.); (A.C.); Tel.: +1-519-824-4120 (ext. 56295) (A.C.)
| | | | - Angela Cánovas
- Correspondence: (P.A.S.F.); (A.C.); Tel.: +1-519-824-4120 (ext. 56295) (A.C.)
| |
Collapse
|
8
|
Shrestha N, Cuffe JSM, Holland OJ, Bulmer AC, Hill M, Perkins AV, Muhlhausler BS, McAinch AJ, Hryciw DH. Elevated maternal linoleic acid reduces circulating leptin concentrations, cholesterol levels and male fetal survival in a rat model. J Physiol 2019; 597:3349-3361. [PMID: 31124126 DOI: 10.1113/jp277583] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 05/09/2019] [Indexed: 01/28/2023] Open
Abstract
KEY POINTS Linoleic acid consumption is increasing in Western populations. We investigated whether elevated linoleic acid in pregnancy was deleterious to mothers or offspring. Maternal and fetal body and organ weights were not affected by elevated linoleic acid consumption. Maternal lipids and leptin were altered following elevated linoleic acid consumption. Male offspring numbers were reduced following elevated linoleic acid consumption. ABSTRACT Dietary intakes of linoleic acid (LA) have increased dramatically in Western populations, including in women of reproductive age. Pro-inflammatory effects of LA may have detrimental effects on maternal and offspring outcomes. We aimed to investigate whether consumption of a maternal diet with elevated LA altered maternal inflammatory or metabolic markers during pregnancy, fetal growth and/or the sex ratio of the offspring. Female Wistar Kyoto rats consumed a diet high in LA (HLA) (6.21% of energy) or a diet low in LA (LLA) (1.44% of energy) for 10 weeks prior to mating and during pregnancy. Pregnant rats were killed at embryonic day 20 (E20). There were no differences in maternal or fetal body weights or organ weights in the HLA group compared to the LLA group. There was no difference in maternal circulating cytokine concentrations between dietary groups. In the maternal liver, IL-1α concentrations were significantly lower, and TNF-α and IL-7 significantly higher in the HLA group. Total plasma cholesterol, LDL-cholesterol, HDL cholesterol and the total:HDL cholesterol ratio were lower in dams fed the HLA diet. mRNA expression of sterol regulatory element binding transcription factor 1 (SREBF-1) and leptin in maternal adipose tissue was lower in the HLA group, as were circulating leptin concentrations. The proportion of male fetuses was lower and circulating prostaglandin E metabolite concentrations were increased in the HLA group. In conclusion, consumption of a maternal diet high in linoleic acid alters cholesterol metabolism and prostaglandin E metabolite concentrations, which may contribute to the reduced proportion of male offspring.
Collapse
Affiliation(s)
- Nirajan Shrestha
- School of Medical Science, Griffith University, Southport, QLD, Australia
| | - James S M Cuffe
- School of Medical Science, Griffith University, Southport, QLD, Australia.,School of Biomedical Sciences, The University of Queensland, St Lucia, QLD, Australia
| | - Olivia J Holland
- School of Medical Science, Griffith University, Southport, QLD, Australia
| | - Andrew C Bulmer
- School of Medical Science, Griffith University, Southport, QLD, Australia
| | - Melissa Hill
- School of Environment and Science, Griffith University, Nathan, QLD, Australia
| | - Anthony V Perkins
- School of Medical Science, Griffith University, Southport, QLD, Australia
| | - Beverly S Muhlhausler
- Food and Nutrition Research Group, Department of Wine and Food Science, School of Agriculture, Food and Wine, University of Adelaide, Adelaide, SA, Australia
| | - Andrew J McAinch
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia.,Australian Institute for Musculoskeletal Science (AIMSS), Victoria University, St Albans, VIC, Australia
| | - Deanne H Hryciw
- School of Environment and Science, Griffith University, Nathan, QLD, Australia.,Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
| |
Collapse
|
9
|
Harter CJL, Kavanagh GS, Smith JT. The role of kisspeptin neurons in reproduction and metabolism. J Endocrinol 2018; 238:R173-R183. [PMID: 30042117 DOI: 10.1530/joe-18-0108] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Accepted: 06/13/2018] [Indexed: 02/06/2023]
Abstract
Kisspeptin is a neuropeptide with a critical role in the function of the hypothalamic-pituitary-gonadal (HPG) axis. Kisspeptin is produced by two major populations of neurons located in the hypothalamus, the rostral periventricular region of the third ventricle (RP3V) and arcuate nucleus (ARC). These neurons project to and activate gonadotrophin-releasing hormone (GnRH) neurons (acting via the kisspeptin receptor, Kiss1r) in the hypothalamus and stimulate the secretion of GnRH. Gonadal sex steroids stimulate kisspeptin neurons in the RP3V, but inhibit kisspeptin neurons in the ARC, which is the underlying mechanism for positive- and negative feedback respectively, and it is now commonly accepted that the ARC kisspeptin neurons act as the GnRH pulse generator. Due to kisspeptin's profound effect on the HPG axis, a focus of recent research has been on afferent inputs to kisspeptin neurons and one specific area of interest has been energy balance, which is thought to facilitate effects such as suppressing fertility in those with under- or severe over-nutrition. Alternatively, evidence is building for a direct role for kisspeptin in regulating energy balance and metabolism. Kiss1r-knockout (KO) mice exhibit increased adiposity and reduced energy expenditure. Although the mechanisms underlying these observations are currently unknown, Kiss1r is expressed in adipose tissue and potentially brown adipose tissue (BAT) and Kiss1rKO mice exhibit reduced energy expenditure. Recent studies are now looking at the effects of kisspeptin signalling on behaviour, with clinical evidence emerging of kisspeptin affecting sexual behaviour, further investigation of potential neuronal pathways are warranted.
Collapse
Affiliation(s)
- Campbell J L Harter
- School of Human SciencesThe University of Western Australia, Perth, Western Australia, Australia
| | - Georgia S Kavanagh
- School of Human SciencesThe University of Western Australia, Perth, Western Australia, Australia
| | - Jeremy T Smith
- School of Human SciencesThe University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
10
|
Piaggi P, Vinales KL, Basolo A, Santini F, Krakoff J. Energy expenditure in the etiology of human obesity: spendthrift and thrifty metabolic phenotypes and energy-sensing mechanisms. J Endocrinol Invest 2018; 41:83-89. [PMID: 28741280 PMCID: PMC5756119 DOI: 10.1007/s40618-017-0732-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Accepted: 07/11/2017] [Indexed: 11/26/2022]
Abstract
The pathogenesis of human obesity is the result of dysregulation of the reciprocal relationship between food intake and energy expenditure (EE), which influences daily energy balance and ultimately leads to weight gain. According to principles of energy homeostasis, a relatively lower EE in a setting of energy balance may lead to weight gain; however, results from different study groups are contradictory and indicate a complex interaction between EE and food intake which may differentially influence weight change in humans. Recently, studies evaluating the adaptive response of one component to perturbations of the other component of energy balance have revealed both the existence of differing metabolic phenotypes ("spendthrift" and "thrifty") resulting from overeating or underfeeding, as well as energy-sensing mechanisms linking EE to food intake, which might explain the propensity of an individual to weight gain. The purpose of this review is to debate the role that human EE plays on body weight regulation and to discuss the physiologic mechanisms linking EE and food intake. An increased understanding of the complex interplay between human metabolism and food consumption may provide insight into pathophysiologic mechanisms underlying weight gain, which may eventually lead to prevention and better treatment of human obesity.
Collapse
Affiliation(s)
- P Piaggi
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), 4212 North 16th Street, Phoenix, AZ, 85016, USA.
- Endocrinology Unit, Obesity Research Center, University Hospital of Pisa, Pisa, Italy.
| | - K L Vinales
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), 4212 North 16th Street, Phoenix, AZ, 85016, USA
| | - A Basolo
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), 4212 North 16th Street, Phoenix, AZ, 85016, USA
- Endocrinology Unit, Obesity Research Center, University Hospital of Pisa, Pisa, Italy
| | - F Santini
- Endocrinology Unit, Obesity Research Center, University Hospital of Pisa, Pisa, Italy
| | - J Krakoff
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), 4212 North 16th Street, Phoenix, AZ, 85016, USA
| |
Collapse
|
11
|
Comparative Study of Reproductive Development in Wild and Captive-Reared Greater Amberjack Seriola dumerili (Risso, 1810). PLoS One 2017; 12:e0169645. [PMID: 28056063 PMCID: PMC5215828 DOI: 10.1371/journal.pone.0169645] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 12/20/2016] [Indexed: 01/06/2023] Open
Abstract
The greater amberjack Seriola dumerili is a large teleost fish with rapid growth and excellent flesh quality, whose domestication represents an ambitious challenge for aquaculture. The occurrence of reproductive dysfunctions in greater amberjack reared in captivity was investigated by comparing reproductive development of wild and captive-reared individuals. Wild and captive-reared breeders were sampled in the Mediterranean Sea during three different phases of the reproductive cycle: early gametogenesis (EARLY, late April-early May), advanced gametogenesis (ADVANCED, late May-early June) and spawning (SPAWNING, late June-July). Fish reproductive state was evaluated using the gonado-somatic index (GSI), histological analysis of the gonads and determination of sex steroid levels in the plasma, and correlated with leptin expression in the liver and gonad biochemical composition. The GSI and sex steroid levels were lower in captive-reared than in wild fish. During the ADVANCED period, when the wild greater amberjack breeders were already in spawning condition, ovaries of captive-reared breeders showed extensive atresia of late vitellogenic oocytes and spermatogenic activity ceased in the testes of half of the examined males. During the SPAWNING period, all captive-reared fish had regressed gonads, while wild breeders still displayed reproductive activity. Liver leptin expression and gonad proximate composition of wild and captive greater amberjack were similar. However, the gonads of captive-reared fish showed different total polar lipid contents, as well as specific lipid classes and fatty acid profiles with respect to wild individuals. This study underlines the need for an improvement in rearing technology for this species, which should include minimum handling during the reproductive season and the formulation of a specific diet to overcome the observed gonadal decrements of phospholipids, DHA (22:6n-3) and ARA (20:4n-6), compared to wild breeders.
Collapse
|
12
|
Heinrich G, Ghadieh HE, Ghanem SS, Muturi HT, Rezaei K, Al-Share QY, Bowman TA, Zhang D, Garofalo RS, Yin L, Najjar SM. Loss of Hepatic CEACAM1: A Unifying Mechanism Linking Insulin Resistance to Obesity and Non-Alcoholic Fatty Liver Disease. Front Endocrinol (Lausanne) 2017; 8:8. [PMID: 28184213 PMCID: PMC5266688 DOI: 10.3389/fendo.2017.00008] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 01/10/2017] [Indexed: 12/25/2022] Open
Abstract
The pathogenesis of human non-alcoholic fatty liver disease (NAFLD) remains unclear, in particular in the context of its relationship to insulin resistance and visceral obesity. Work on the carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) in mice has resolved some of the related questions. CEACAM1 promotes insulin clearance by enhancing the rate of uptake of the insulin-receptor complex. It also mediates a negative acute effect of insulin on fatty acid synthase activity. This positions CEACAM1 to coordinate the regulation of insulin and lipid metabolism. Fed a regular chow diet, global null mutation of Ceacam1 manifest hyperinsulinemia, insulin resistance, obesity, and steatohepatitis. They also develop spontaneous chicken-wire fibrosis, characteristic of non-alcoholic steatohepatitis. Reduction of hepatic CEACAM1 expression plays a significant role in the pathogenesis of diet-induced metabolic abnormalities, as bolstered by the protective effect of hepatic CEACAM1 gain-of-function against the metabolic response to dietary fat. Together, this emphasizes that loss of hepatic CEACAM1 links NAFLD to insulin resistance and obesity.
Collapse
Affiliation(s)
- Garrett Heinrich
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
- Heritage College of Osteopathic Medicine, Diabetes Institute, Ohio University, Athens, OH, USA
| | - Hilda E. Ghadieh
- Center for Diabetes and Endocrine Research (CeDER), College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Simona S. Ghanem
- Center for Diabetes and Endocrine Research (CeDER), College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Harrison T. Muturi
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Khadijeh Rezaei
- Center for Diabetes and Endocrine Research (CeDER), College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Qusai Y. Al-Share
- Center for Diabetes and Endocrine Research (CeDER), College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Thomas A. Bowman
- Center for Diabetes and Endocrine Research (CeDER), College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Deqiang Zhang
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | | | - Lei Yin
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Sonia M. Najjar
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
- Heritage College of Osteopathic Medicine, Diabetes Institute, Ohio University, Athens, OH, USA
- *Correspondence: Sonia M. Najjar,
| |
Collapse
|
13
|
Russo L, Ghadieh HE, Ghanem SS, Al-Share QY, Smiley ZN, Gatto-Weis C, Esakov EL, McInerney MF, Heinrich G, Tong X, Yin L, Najjar SM. Role for hepatic CEACAM1 in regulating fatty acid metabolism along the adipocyte-hepatocyte axis. J Lipid Res 2016; 57:2163-2175. [PMID: 27777319 DOI: 10.1194/jlr.m072066] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 10/17/2016] [Indexed: 12/15/2022] Open
Abstract
Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) regulates insulin sensitivity by promoting hepatic insulin clearance and mediating suppression of fatty acid synthase activity. Feeding C57BL/6J male mice with a high-fat (HF) diet for 3-4 weeks triggered a >60% decrease in hepatic CEACAM1 levels to subsequently impair insulin clearance and cause systemic insulin resistance and hepatic steatosis. This study aimed at investigating whether lipolysis drives reduction in hepatic CEACAM1 and whether this constitutes a key mechanism leading to diet-induced metabolic abnormalities. Blocking lipolysis with a daily intraperitoneal injection of nicotinic acid in the last two days of a 30-day HF feeding regimen demonstrated that white adipose tissue (WAT)-derived fatty acids repressed hepatic CEACAM1-dependent regulation of insulin and lipid metabolism in 3-month-old male C57BL/6J mice. Adenoviral-mediated CEACAM1 redelivery countered the adverse metabolic effect of the HF diet on insulin resistance, hepatic steatosis, visceral obesity, and energy expenditure. It also reversed the effect of HF diet on inflammation and fibrosis in WAT and liver. This assigns a causative role for lipolysis-driven decrease in hepatic CEACAM1 level and its regulation of insulin and lipid metabolism in sustaining systemic insulin resistance, hepatic steatosis, and other abnormalities associated with excessive energy supply.
Collapse
Affiliation(s)
- Lucia Russo
- Center for Diabetes and Endocrine Research, University of Toledo, Toledo, OH 43614
| | - Hilda E Ghadieh
- Center for Diabetes and Endocrine Research, University of Toledo, Toledo, OH 43614
| | - Simona S Ghanem
- Center for Diabetes and Endocrine Research, University of Toledo, Toledo, OH 43614
| | - Qusai Y Al-Share
- Center for Diabetes and Endocrine Research, University of Toledo, Toledo, OH 43614
| | - Zachary N Smiley
- Center for Diabetes and Endocrine Research, University of Toledo, Toledo, OH 43614
| | - Cara Gatto-Weis
- Center for Diabetes and Endocrine Research, University of Toledo, Toledo, OH 43614.,Department of Pathology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614
| | - Emily L Esakov
- Center for Diabetes and Endocrine Research, University of Toledo, Toledo, OH 43614.,Department of Medicinal and Biological Chemistry at the College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614
| | - Marcia F McInerney
- Center for Diabetes and Endocrine Research, University of Toledo, Toledo, OH 43614.,Department of Medicinal and Biological Chemistry at the College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614
| | - Garrett Heinrich
- Center for Diabetes and Endocrine Research, University of Toledo, Toledo, OH 43614.,Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701
| | - Xin Tong
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48019
| | - Lei Yin
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48019
| | - Sonia M Najjar
- Center for Diabetes and Endocrine Research, University of Toledo, Toledo, OH 43614 .,Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701
| |
Collapse
|
14
|
Behavioural and physiological responses of wood mice (Apodemus sylvaticus) to experimental manipulations of predation and starvation risk. Physiol Behav 2015; 149:331-9. [DOI: 10.1016/j.physbeh.2015.06.037] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 05/20/2015] [Accepted: 06/28/2015] [Indexed: 11/17/2022]
|
15
|
Saber H, Himali JJ, Shoamanesh A, Beiser A, Pikula A, Harris TB, Roubenoff R, Romero JR, Kase CS, Vasan RS, Seshadri S. Serum Leptin Levels and the Risk of Stroke: The Framingham Study. Stroke 2015; 46:2881-5. [PMID: 26337973 DOI: 10.1161/strokeaha.115.009463] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Accepted: 08/05/2015] [Indexed: 01/14/2023]
Abstract
BACKGROUND AND PURPOSE Leptin is a major adipokine that regulates weight balance and energy homeostasis. There is inconsistent evidence linking circulating leptin levels to risk of stroke. We tested the hypothesis that leptin levels are associated with risk of incident stroke in an elderly community based sample. METHODS Serum leptin levels were assayed in 757 stroke free individuals (mean age, 79 years; 62% women) from the Framingham Original Cohort at the 22nd examination cycle (1990-1994). Incidence of all -stroke and ischemic stroke were prospectively ascertained. RESULTS During a mean follow up of 10 years, 119 individuals developed stroke (99 ischemic strokes). In multivariable Cox regression models, log leptin levels were not associated with incidence of all -stroke or ischemic stroke (hazard ratios per SD increment in log leptin 0.90 [0.73-1.09] and 0.89 [0.72-1.11], respectively). The results were suggestive for potential effect modification by waist/hip ratio for the association between leptin and stroke (P=0.03). Adjusting for age, sex, and established stroke risk factors, analysis stratified by waist/hip ratio quartiles revealed a lower incidence of first-ever all-stroke and ischemic stroke associated with higher leptin levels among only subjects in the top waist/hip ratio quartile (hazard ratio, 0.64 [0.43, 0.95] versus 0.98 [0.77, 1.25] for incident all-stroke and 0.61 [0.39, 0.95] versus 0.96 [0.74, 1.26] for ischemic stroke). CONCLUSIONS Leptin levels were not directly related to the risk of incident stroke overall but there was an inverse association with stroke in the top waist/hip ratio quartile. Further investigations are required to confirm these findings and explore possible mechanisms for the observed association.
Collapse
Affiliation(s)
- Hamidreza Saber
- From the Framingham Heart Study, MA (H.S., A.B., J.R.R., C.S.K., R.S.V., S.S.); Department of Biostatistics, Boston University School of Public Health, MA (A.B.); Division of Neurology, Department of Medicine, McMaster University/Population Health Research Institute, Hamilton, ON, Canada (A.S.); Department of Neurology, University of Toronto, Toronto, ON, Canada (A.P.); Geriatric Epidemiology section, National Institute on Aging, National Institute of Health, Bethesda, MD (T.B.H.); Friedman School of Nutrition Science and Policy, Tufts University School of Medicine, Boston, MA (R.R.); and Department of Neurology, Boston University School of Medicine, MA (H.S., J.J.H., A.B., J.R.R., C.S.K., R.S.V., S.S.).
| | - Jayandra J Himali
- From the Framingham Heart Study, MA (H.S., A.B., J.R.R., C.S.K., R.S.V., S.S.); Department of Biostatistics, Boston University School of Public Health, MA (A.B.); Division of Neurology, Department of Medicine, McMaster University/Population Health Research Institute, Hamilton, ON, Canada (A.S.); Department of Neurology, University of Toronto, Toronto, ON, Canada (A.P.); Geriatric Epidemiology section, National Institute on Aging, National Institute of Health, Bethesda, MD (T.B.H.); Friedman School of Nutrition Science and Policy, Tufts University School of Medicine, Boston, MA (R.R.); and Department of Neurology, Boston University School of Medicine, MA (H.S., J.J.H., A.B., J.R.R., C.S.K., R.S.V., S.S.)
| | - Ashkan Shoamanesh
- From the Framingham Heart Study, MA (H.S., A.B., J.R.R., C.S.K., R.S.V., S.S.); Department of Biostatistics, Boston University School of Public Health, MA (A.B.); Division of Neurology, Department of Medicine, McMaster University/Population Health Research Institute, Hamilton, ON, Canada (A.S.); Department of Neurology, University of Toronto, Toronto, ON, Canada (A.P.); Geriatric Epidemiology section, National Institute on Aging, National Institute of Health, Bethesda, MD (T.B.H.); Friedman School of Nutrition Science and Policy, Tufts University School of Medicine, Boston, MA (R.R.); and Department of Neurology, Boston University School of Medicine, MA (H.S., J.J.H., A.B., J.R.R., C.S.K., R.S.V., S.S.)
| | - Alexa Beiser
- From the Framingham Heart Study, MA (H.S., A.B., J.R.R., C.S.K., R.S.V., S.S.); Department of Biostatistics, Boston University School of Public Health, MA (A.B.); Division of Neurology, Department of Medicine, McMaster University/Population Health Research Institute, Hamilton, ON, Canada (A.S.); Department of Neurology, University of Toronto, Toronto, ON, Canada (A.P.); Geriatric Epidemiology section, National Institute on Aging, National Institute of Health, Bethesda, MD (T.B.H.); Friedman School of Nutrition Science and Policy, Tufts University School of Medicine, Boston, MA (R.R.); and Department of Neurology, Boston University School of Medicine, MA (H.S., J.J.H., A.B., J.R.R., C.S.K., R.S.V., S.S.)
| | - Aleksandra Pikula
- From the Framingham Heart Study, MA (H.S., A.B., J.R.R., C.S.K., R.S.V., S.S.); Department of Biostatistics, Boston University School of Public Health, MA (A.B.); Division of Neurology, Department of Medicine, McMaster University/Population Health Research Institute, Hamilton, ON, Canada (A.S.); Department of Neurology, University of Toronto, Toronto, ON, Canada (A.P.); Geriatric Epidemiology section, National Institute on Aging, National Institute of Health, Bethesda, MD (T.B.H.); Friedman School of Nutrition Science and Policy, Tufts University School of Medicine, Boston, MA (R.R.); and Department of Neurology, Boston University School of Medicine, MA (H.S., J.J.H., A.B., J.R.R., C.S.K., R.S.V., S.S.)
| | - Tamara B Harris
- From the Framingham Heart Study, MA (H.S., A.B., J.R.R., C.S.K., R.S.V., S.S.); Department of Biostatistics, Boston University School of Public Health, MA (A.B.); Division of Neurology, Department of Medicine, McMaster University/Population Health Research Institute, Hamilton, ON, Canada (A.S.); Department of Neurology, University of Toronto, Toronto, ON, Canada (A.P.); Geriatric Epidemiology section, National Institute on Aging, National Institute of Health, Bethesda, MD (T.B.H.); Friedman School of Nutrition Science and Policy, Tufts University School of Medicine, Boston, MA (R.R.); and Department of Neurology, Boston University School of Medicine, MA (H.S., J.J.H., A.B., J.R.R., C.S.K., R.S.V., S.S.)
| | - Ronenn Roubenoff
- From the Framingham Heart Study, MA (H.S., A.B., J.R.R., C.S.K., R.S.V., S.S.); Department of Biostatistics, Boston University School of Public Health, MA (A.B.); Division of Neurology, Department of Medicine, McMaster University/Population Health Research Institute, Hamilton, ON, Canada (A.S.); Department of Neurology, University of Toronto, Toronto, ON, Canada (A.P.); Geriatric Epidemiology section, National Institute on Aging, National Institute of Health, Bethesda, MD (T.B.H.); Friedman School of Nutrition Science and Policy, Tufts University School of Medicine, Boston, MA (R.R.); and Department of Neurology, Boston University School of Medicine, MA (H.S., J.J.H., A.B., J.R.R., C.S.K., R.S.V., S.S.)
| | - Jose Rafael Romero
- From the Framingham Heart Study, MA (H.S., A.B., J.R.R., C.S.K., R.S.V., S.S.); Department of Biostatistics, Boston University School of Public Health, MA (A.B.); Division of Neurology, Department of Medicine, McMaster University/Population Health Research Institute, Hamilton, ON, Canada (A.S.); Department of Neurology, University of Toronto, Toronto, ON, Canada (A.P.); Geriatric Epidemiology section, National Institute on Aging, National Institute of Health, Bethesda, MD (T.B.H.); Friedman School of Nutrition Science and Policy, Tufts University School of Medicine, Boston, MA (R.R.); and Department of Neurology, Boston University School of Medicine, MA (H.S., J.J.H., A.B., J.R.R., C.S.K., R.S.V., S.S.)
| | - Carlos S Kase
- From the Framingham Heart Study, MA (H.S., A.B., J.R.R., C.S.K., R.S.V., S.S.); Department of Biostatistics, Boston University School of Public Health, MA (A.B.); Division of Neurology, Department of Medicine, McMaster University/Population Health Research Institute, Hamilton, ON, Canada (A.S.); Department of Neurology, University of Toronto, Toronto, ON, Canada (A.P.); Geriatric Epidemiology section, National Institute on Aging, National Institute of Health, Bethesda, MD (T.B.H.); Friedman School of Nutrition Science and Policy, Tufts University School of Medicine, Boston, MA (R.R.); and Department of Neurology, Boston University School of Medicine, MA (H.S., J.J.H., A.B., J.R.R., C.S.K., R.S.V., S.S.)
| | - Ramachandran S Vasan
- From the Framingham Heart Study, MA (H.S., A.B., J.R.R., C.S.K., R.S.V., S.S.); Department of Biostatistics, Boston University School of Public Health, MA (A.B.); Division of Neurology, Department of Medicine, McMaster University/Population Health Research Institute, Hamilton, ON, Canada (A.S.); Department of Neurology, University of Toronto, Toronto, ON, Canada (A.P.); Geriatric Epidemiology section, National Institute on Aging, National Institute of Health, Bethesda, MD (T.B.H.); Friedman School of Nutrition Science and Policy, Tufts University School of Medicine, Boston, MA (R.R.); and Department of Neurology, Boston University School of Medicine, MA (H.S., J.J.H., A.B., J.R.R., C.S.K., R.S.V., S.S.)
| | - Sudha Seshadri
- From the Framingham Heart Study, MA (H.S., A.B., J.R.R., C.S.K., R.S.V., S.S.); Department of Biostatistics, Boston University School of Public Health, MA (A.B.); Division of Neurology, Department of Medicine, McMaster University/Population Health Research Institute, Hamilton, ON, Canada (A.S.); Department of Neurology, University of Toronto, Toronto, ON, Canada (A.P.); Geriatric Epidemiology section, National Institute on Aging, National Institute of Health, Bethesda, MD (T.B.H.); Friedman School of Nutrition Science and Policy, Tufts University School of Medicine, Boston, MA (R.R.); and Department of Neurology, Boston University School of Medicine, MA (H.S., J.J.H., A.B., J.R.R., C.S.K., R.S.V., S.S.)
| |
Collapse
|
16
|
Al-Share QY, DeAngelis AM, Lester SG, Bowman TA, Ramakrishnan SK, Abdallah SL, Russo L, Patel PR, Kaw MK, Raphael CK, Kim AJ, Heinrich G, Lee AD, Kim JK, Kulkarni RN, Philbrick WM, Najjar SM. Forced Hepatic Overexpression of CEACAM1 Curtails Diet-Induced Insulin Resistance. Diabetes 2015; 64:2780-90. [PMID: 25972571 PMCID: PMC4512217 DOI: 10.2337/db14-1772] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 03/16/2015] [Indexed: 12/18/2022]
Abstract
Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) regulates insulin sensitivity by promoting hepatic insulin clearance. Liver-specific inactivation or global null-mutation of Ceacam1 impairs hepatic insulin extraction to cause chronic hyperinsulinemia, resulting in insulin resistance and visceral obesity. In this study we investigated whether diet-induced insulin resistance implicates changes in hepatic CEACAM1. We report that feeding C57/BL6J mice a high-fat diet reduced hepatic CEACAM1 levels by >50% beginning at 21 days, causing hyperinsulinemia, insulin resistance, and elevation in hepatic triacylglycerol content. Conversely, liver-specific inducible CEACAM1 expression prevented hyperinsulinemia and markedly limited insulin resistance and hepatic lipid accumulation that were induced by prolonged high-fat intake. This was partly mediated by increased hepatic β-fatty acid oxidation and energy expenditure. The data demonstrate that the high-fat diet reduced hepatic CEACAM1 expression and that overexpressing CEACAM1 in liver curtailed diet-induced metabolic abnormalities by protecting hepatic insulin clearance.
Collapse
Affiliation(s)
- Qusai Y Al-Share
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH Department of Physiology and Pharmacology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH
| | - Anthony M DeAngelis
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH Department of Physiology and Pharmacology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH
| | - Sumona Ghosh Lester
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH Department of Physiology and Pharmacology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH
| | - Thomas A Bowman
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH Department of Physiology and Pharmacology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH
| | - Sadeesh K Ramakrishnan
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH Department of Physiology and Pharmacology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH
| | - Simon L Abdallah
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH Department of Physiology and Pharmacology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH
| | - Lucia Russo
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH Department of Physiology and Pharmacology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH
| | - Payal R Patel
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH Department of Physiology and Pharmacology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH
| | - Meenakshi K Kaw
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH Department of Physiology and Pharmacology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH
| | - Christian K Raphael
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH Department of Physiology and Pharmacology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH
| | - Andrea Jung Kim
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH Department of Rehabilitation Sciences, College of Health Sciences, The University of Toledo, Toledo, OH
| | - Garrett Heinrich
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH Department of Rehabilitation Sciences, College of Health Sciences, The University of Toledo, Toledo, OH
| | - Abraham D Lee
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH Department of Rehabilitation Sciences, College of Health Sciences, The University of Toledo, Toledo, OH
| | - Jason K Kim
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Rohit N Kulkarni
- Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - William M Philbrick
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT
| | - Sonia M Najjar
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH Department of Physiology and Pharmacology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH
| |
Collapse
|
17
|
Aguilar-Moreno M, Galicia-Castillo OR, Aguilera-Reyes U, Varea-González C, Bernis-Carro C, García-López GI. Hormonal State Comparison (Progesterone, Estradiol, and Leptin) of Body Fat and Body Mass Indices in Mexican Women as a Risk Factor for Neonatal Physiologic Condition. J Pediatr Adolesc Gynecol 2015; 28:149-56. [PMID: 26046604 DOI: 10.1016/j.jpag.2014.06.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 05/30/2014] [Accepted: 06/30/2014] [Indexed: 10/25/2022]
Abstract
STUDY OBJECTIVE Describe the impact of teen pregnancy on later ovarian activity and metabolic hormones considering the concentration of current levels of ovarian steroids and leptin in a sample of Mexican females. DESIGN Cross-sectional study in the maternity of the General Hospital of Atlacomulco and campus of the Autonomous University of the State of Mexico. PARTICIPANTS 71 women between the ages of 18 and 24, and 160 neonates seen between March 2010 and June 2012. MAIN OUTCOME MEASURES The measurements obtained included anthropometric body composition (bioelectrical impedance), serum hormone quantification of ovarian steroids and leptin (immunoassays), and the Apgar scores, height, and weight in neonates. Statistical analysis included ANOVA, Student, and chi-square for P < .05. RESULTS Adolescent mothers showed significantly lower concentrations of estradiol (P = .001) and progesterone (P = .001). However, higher levels of leptin in adolescent mothers were not statistically different compared with older mothers (P = .84). Also, leptin was correlated with all measures of adiposity. The mean birth weights (P = .001) and Apgar scores (P = .001) were lower in neonates of adolescent mothers than in neonates of adult mothers. There was no association between maternal age with the anthropometric variables studied. CONCLUSIONS Early reproduction represents a metabolic stress condition that modifies the long term ovarian activity and metabolic hormones, and impacts the morbidity-mortality of the mother and offspring in a later vital life cycle stage.
Collapse
Affiliation(s)
| | | | - Ulises Aguilera-Reyes
- Laboratory of Animal Behavior, Faculty of Sciences, Autonomous University of the State of Mexico, Atlacomulco, Mexico
| | - Carlos Varea-González
- Department of Biology, Faculty of Sciences, Autonomous University of Madrid, Madrid, Spain
| | - Cristina Bernis-Carro
- Department of Biology, Faculty of Sciences, Autonomous University of Madrid, Madrid, Spain
| | | |
Collapse
|
18
|
Khorram O, Keen-Rinehart E, Chuang TD, Ross MG, Desai M. Maternal undernutrition induces premature reproductive senescence in adult female rat offspring. Fertil Steril 2014; 103:291-8.e2. [PMID: 25439841 DOI: 10.1016/j.fertnstert.2014.09.026] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 09/18/2014] [Accepted: 09/19/2014] [Indexed: 12/11/2022]
Abstract
OBJECTIVE To determine the effects of maternal undernutrition (MUN) on the reproductive axis of aging offspring. DESIGN Animal (rat) study. SETTING Research laboratory. ANIMAL(S) Female Sprague-Dawley rats. INTERVENTION(S) Food restriction during the second half of pregnancy in rats. MAIN OUTCOME MEASURE(S) Circulating gonadotropins, antimüllerian hormone (AMH), ovarian morphology, estrous cyclicity, and gene expression studies in the hypothalamus and ovary in 1-day-old (P1) and aging adult offspring. RESULT(S) Offspring of MUN dams had low birth weight (LBW) and by adult age developed obesity. In addition, 80% of adult LBW offspring had disruption of estrous cycle by 8 months of age, with the majority of animals in persistent estrous. Ovarian morphology was consistent with acyclicity, with ovaries exhibiting large cystic structures and reduced corpora lutea. There was an elevation in circulating T, increased ovarian expression of enzymes involved in androgen synthesis, an increase in plasma LH/FSH levels, a reduction in E2 levels, and no changes in AMH in adult LBW offspring compared with in control offspring. Hypothalamic expression of leptin receptor (ObRb), estrogen receptor-α (ER-α), and GnRH protein was altered in an age-dependent manner with increased ObRb and ER-α expression in P1 LBW hypothalami and a reversal of this expression pattern in adult LBW hypothalami. CONCLUSION(S) Our data indicate that the maternal nutritional environment programs the reproductive potential of the offspring through alteration of the hypothalamic-pituitary-gonadal axis. The premature reproductive senescence in LBW offspring could be secondary to the development of obesity and hyperleptinemia in these animals in adult life.
Collapse
Affiliation(s)
- Omid Khorram
- Department of Obstetrics and Gynecology, Harbor-UCLA Medical Center, and LA Biomedical Institute, Torrance, California.
| | - Erin Keen-Rinehart
- Department of Obstetrics and Gynecology, Harbor-UCLA Medical Center, and LA Biomedical Institute, Torrance, California
| | - Tsai-Der Chuang
- Department of Obstetrics and Gynecology, Harbor-UCLA Medical Center, and LA Biomedical Institute, Torrance, California
| | - Michael G Ross
- Department of Obstetrics and Gynecology, Harbor-UCLA Medical Center, and LA Biomedical Institute, Torrance, California
| | - Mina Desai
- Department of Obstetrics and Gynecology, Harbor-UCLA Medical Center, and LA Biomedical Institute, Torrance, California
| |
Collapse
|
19
|
Abstract
The hyperphagia, low sympathetic nervous system tone, and decreased circulating concentrations of bioactive thyroid hormones that are common to states of congenital leptin deficiency and hypoleptinemia following and during weight loss suggest that the major physiological function of leptin is to signal states of negative energy balance and decreased energy stores. In weight-reduced humans, these phenotypes together with pronounced hypometabolism and increased parasympathetic nervous system tone create the optimal circumstance for weight regain. Based on the weight loss induced by leptin administration in states of leptin deficiency (obese) and observed similarity of phenotypes in states of congenital and dietary-induced states of hypoleptinemia (reduced obese), it has been suggested that exogenous leptin could potentially be useful in initiating, promoting, and sustaining weight reduction. However, the responses of human beings to exogenous leptin administration are dependent not only on extant energy stores but also on energy balance. Leptin administration to humans at usual weight has little, if any, effect on body weight while leptin administration during weight loss mitigates hunger, especially if given in supraphysiological doses during severe caloric restriction. Leptin repletion is most effective following weight loss by dietary restriction. In this state of weight stability but reduced energy stores, leptin at least partially reverses many of the metabolic, autonomic, neuroendocrine, and behavioral adaptations that favor weight regain. The major physiological function of leptin is to signal states of negative energy balance and decreased energy stores. Leptin, and pharmacotherapies affecting leptin signaling pathways, is likely to be most useful in sustaining weight loss.
Collapse
Affiliation(s)
- Michael Rosenbaum
- Division of Molecular GeneticsDepartments of Pediatrics and Medicine, College of Physicians and Surgeons, Columbia University, Russ Berrie Medical Science Pavilion, 6th Floor, 1150 St Nicholas Avenue, New York, New York 10032, USA
| | - Rudolph L Leibel
- Division of Molecular GeneticsDepartments of Pediatrics and Medicine, College of Physicians and Surgeons, Columbia University, Russ Berrie Medical Science Pavilion, 6th Floor, 1150 St Nicholas Avenue, New York, New York 10032, USA
| |
Collapse
|
20
|
Effects of chronic leptin infusion on subsequent body weight and composition in mice: Can body weight set point be reset? Mol Metab 2014; 3:432-40. [PMID: 24944902 PMCID: PMC4060284 DOI: 10.1016/j.molmet.2014.02.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Revised: 02/20/2014] [Accepted: 02/24/2014] [Indexed: 11/22/2022] Open
Abstract
Circulating leptin concentrations correlate with fat mass and signal the status of somatic energy stores to the brain. Previous studies suggest that diet-induced elevations of body weight increase body weight “set-point”. To assess whether chronic hyperleptinemia is responsible for this shift in defended body weight, we elevated circulating leptin concentrations in lean mice to those comparable to diet-induced obese mice for eighteen weeks. We hypothesized that following cessation of leptin infusion, a higher body weight would be defended. Compared to saline-infused controls, leptin-infused mice had elevated circulating leptin concentrations, gained less weight, yet had similar metabolic rates. Following cessation of leptin administration, leptin-infused mice gained some weight yet plateaued at 5–10% below controls. These results suggest that, unlike mice rendered hyperleptinemic by diet-induced weight gain, leptin-infused mice do not subsequently “defend” a higher body weight, suggesting that hyperleptinemia per se does not mimic the CNS consequences of chronic weight gain.
Collapse
|
21
|
Gill R, Cheung YH, Shen Y, Lanzano P, Mirza NM, Ten S, Maclaren NK, Motaghedi R, Han JC, Yanovski JA, Leibel RL, Chung WK. Whole-exome sequencing identifies novel LEPR mutations in individuals with severe early onset obesity. Obesity (Silver Spring) 2014; 22:576-84. [PMID: 23616257 PMCID: PMC3791145 DOI: 10.1002/oby.20492] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Accepted: 04/02/2013] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Obesity is a major public health problem that increases the risk for a broad spectrum of co-morbid conditions. Despite evidence for a strong genetic contribution to susceptibility to obesity, previous efforts to discover the relevant genes using positional cloning have failed to account for most of the apparent genetic risk variance. DESIGN AND METHODS Deploying a strategy combining analysis of exome sequencing data in extremely obese members of four consanguineous families with segregation analysis, we screened for causal genetic variants. Filter-based analysis and homozygosity mapping were used to identify and prioritize putative functional variants. RESULTS Two novel frameshift mutations in the leptin receptor in two of the families were identified. CONCLUSIONS These results provide proof-of-principle that whole-exome sequencing of families segregating for extreme obesity can identify causal pathogenic mutations. The methods described here can be extended to additional families segregating for extreme obesity and should enable the identification of mutations in novel genes that predispose to obesity.
Collapse
Affiliation(s)
- Richard Gill
- Division of Molecular Genetics, Department of Pediatrics, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY, USA
- Department of Epidemiology, Mailman School of Public Health, Columbia University Medical Center, New York, NY, USA
| | - Yee Him Cheung
- Division of Molecular Genetics, Department of Pediatrics, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY, USA
| | - Yufeng Shen
- Department of Biomedical Informatics, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY, USA
| | - Patricia Lanzano
- Division of Molecular Genetics, Department of Pediatrics, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY, USA
| | - Nazrat M. Mirza
- Section on Growth and Obesity, Program in Developmental Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health, Bethesda, MD, USA
- Children’s National Medical Center, Washington, DC, USA
| | - Svetlana Ten
- Division of Pediatric Endocrinology at Maimonides Infants and Children’s Hospital of Brooklyn and SUNY Downstate Medical Center, Brooklyn, NY, USA
| | - Noel K. Maclaren
- Department of Pediatrics, Weill Medical College of Cornell University, New York, NY, USA
| | - Roja Motaghedi
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
| | - Joan C. Han
- Section on Growth and Obesity, Program in Developmental Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health, Bethesda, MD, USA
| | - Jack A. Yanovski
- Section on Growth and Obesity, Program in Developmental Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health, Bethesda, MD, USA
| | - Rudolph L. Leibel
- Division of Molecular Genetics, Department of Pediatrics, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY, USA
| | - Wendy K. Chung
- Division of Molecular Genetics, Department of Pediatrics, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
22
|
Yan H, Zhang HW, Fu P, Liu BL, Jin WZ, Duan SB, Xue J, Liu K, Sun ZM, Zeng XW. Leptin's effect on accelerated fracture healing after traumatic brain injury. Neurol Res 2013; 35:537-44. [PMID: 23594570 DOI: 10.1179/1743132813y.0000000201] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
OBJECTIVE To investigate mechanisms behind the faster rehabilitation of limb fractures when associated with traumatic brain injury (TBI). METHODS New Zealand rabbits were divided into TBI group and sham-operation group for four studies as follows: (1) blood and cerebrospinal fluid (CSF) were drawn on days 1, 3, and 7 to demonstrate changes in serum leptin, growth hormone (GH), insulin-like growth factor 1 (IGF-1), and CSF leptin; (2) bone defection was created by drilling in the tibial bone and either leptin or normal saline was injected into rabbit's cerebellomedullary cistern. X-ray was taken at 1 days, 2 weeks, and 5 weeks and evaluated by criteria to determine rate of bone healing; (3) FITC-labeled rabbit leptin was injected into TBI and sham-operation groups, and frozen sections of rabbit brain were observed to identify differences in central nervous system (CNS) leptin by fluorescence; (4) polymerase chain reaction (PCR) was used to evaluate the expression of leptin production by brain tissue. RESULTS Serum and CSF leptin, GH, and IGF-1 concentrations were found to be higher in the TBI group than the sham-operation group at days 1, 3, and 7 (P<0·05). CSF leptin of the TBI group was positively correlated with serum leptin on day 1 (P<0·05), and positively correlated with GH and IGF-1 on days 3 and 7 (P<0·05). X-ray criteria demonstrated that leptin administration caused significantly faster healing calluses at 3 and 5 weeks as compared to control animals (P<0·05). FITC-labeled leptin study demonstrated that TBI animals had stronger expression of leptin in the brain than sham-operated animals. However, PCR of brain tissue leptin showed no significant differences between TBI and sham-operated animals in the expression of leptin. CONCLUSIONS Our study suggests that increased CSF leptin, likely from blood-brain barrier breakdown, combined with elevated serum GH and IGF-1 after TBI, leads to accelerated fracture healing.
Collapse
Affiliation(s)
- Hua Yan
- Department of Neurosurgery, Tianjin Huanhu Hospital, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Garcia-Garcia RM. Integrative control of energy balance and reproduction in females. ISRN VETERINARY SCIENCE 2012; 2012:121389. [PMID: 23762577 PMCID: PMC3671732 DOI: 10.5402/2012/121389] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Accepted: 09/04/2012] [Indexed: 11/23/2022]
Abstract
There is a strong association between nutrition and reproduction. Chronic dietary energy deficits as well as energy surpluses can impair reproductive capacity. Metabolic status impacts reproductive function at systemic level, modulating the hypothalamic GnRH neuronal network and/or the pituitary gonadotropin secretion through several hormones and neuropeptides, and at the ovarian level, acting through the regulation of follicle growth and steroidogenesis by means of the growth hormone-IGF-insulin system and local ovarian mediators. In the past years, several hormones and neuropeptides have been emerging as important mediators between energy balance and reproduction. The present review goes over the main sites implicated in the control of energy balance linked to reproductive success and summarizes the most important metabolic and neuroendocrine signals that participate in reproductive events with special emphasis on the role of recently discovered neuroendocrine peptides. Also, a little overview about the effects of maternal nutrition, affecting offspring reproduction, has been presented.
Collapse
Affiliation(s)
- R M Garcia-Garcia
- Physiology Department (Animal Physiology), Complutense University, Avenida Puerta de Hierro S/N, 28040 Madrid, Spain
| |
Collapse
|
24
|
Angel-Chávez LI, Tene-Pérez CE, Castro E. Leptin receptor gene K656N polymorphism is associated with low body fat levels and elevated high-density cholesterol levels in Mexican children and adolescents. Endocr Res 2012; 37:124-34. [PMID: 22746211 DOI: 10.3109/07435800.2011.648360] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION This study evaluated the association between obesity and the K109R, Q223R, and K656N polymorphisms of the leptin receptor (LEPR) locus. Such polymorphisms cause changes in the extracellular extreme of the LEPR gene product and appear to be related to signal transduction toward the cell. SUBJECTS AND METHODS A total of 128 participants between 6 and 17 years of age from a Mexican Mestizo population were included in the study. Individuals were classified as overweight-obese (n = 76) and normal (n = 52), based on anthropomorphic measurements. The K109R, Q223R, and K656N polymorphisms of the LEPR were determined by the size of restriction fragments obtained from polymorphic fragment amplification (polymerase chain reaction-restriction fragment length polymorphism) obtained from genomic DNA. Allele frequency was compared using the chi-square test. Odds ratio was calculated to determine allele obesity risk factor. RESULTS Variant allele frequency was 109R = 0.35, 223R = 0.49, and 656N = 0.11 for the K109R, Q223R, and K656N polymorphisms, respectively. No statistically significant association with obesity was found in any of the alleles. The N allele of the K656N polymorphism was associated with nonobesity markers, such as high concentrations of high-density lipoproteins, normal body mass index, less thickness of skinfolds, and body perimeters. None of the alleles studied were shown to be obesity risk factors. CONCLUSIONS Our results suggest that there is no association between the K109R, Q223R, and K656N polymorphisms of the LEPR gene with obesity, and none of the alleles of the LEPR gene K109R, Q223R, and K656N polymorphisms are an obesity risk factor.
Collapse
|
25
|
Cabrera CP, Dunn IC, Fell M, Wilson PW, Burt DW, Waddington D, Talbot R, Hocking PM, Law A, Knott S, Haley CS, de Koning DJ. Complex traits analysis of chicken growth using targeted genetical genomics. Anim Genet 2011; 43:163-71. [PMID: 22404352 DOI: 10.1111/j.1365-2052.2011.02223.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Dissecting the genetic control of complex trait variation remains very challenging, despite many advances in technology. The aim of this study was to use a major growth quantitative trait locus (QTL) in chickens mapped to chromosome 4 as a model for a targeted approach to dissect the QTL. We applied a variant of the genetical genomics approach to investigate genome-wide gene expression differences between two contrasting genotypes of a marked QTL. This targeted approach allows the direct quantification of the link between the genotypes and the genetic responses, thus narrowing the QTL-phenotype gap using fewer samples (i.e. microarrays) compared with the genome-wide genetical genomics studies. Four differentially expressed genes were localized under the region of the QTL. One of these genes is a potential positional candidate gene (AADAT) that affects lysine and tryptophan metabolism and has alternative splicing variants between the two genotypes. In addition, the lysine and glycolysis metabolism pathways were significantly enriched for differentially expressed genes across the genome. The targeted approach provided a complementary route to fine mapping of QTL by characterizing the local and the global downstream effects of the QTL and thus generating further hypotheses about the action of that QTL.
Collapse
Affiliation(s)
- C P Cabrera
- Medical Research Council, Human Genetics Unit, Edinburgh, UK.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Ravussin Y, Gutman R, Diano S, Shanabrough M, Borok E, Sarman B, Lehmann A, LeDuc CA, Rosenbaum M, Horvath TL, Leibel RL. Effects of chronic weight perturbation on energy homeostasis and brain structure in mice. Am J Physiol Regul Integr Comp Physiol 2011; 300:R1352-62. [PMID: 21411766 DOI: 10.1152/ajpregu.00429.2010] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Maintenance of reduced body weight in lean and obese human subjects results in the persistent decrease in energy expenditure below what can be accounted for by changes in body mass and composition. Genetic and developmental factors may determine a central nervous system (CNS)-mediated minimum threshold of somatic energy stores below which behavioral and metabolic compensations for weight loss are invoked. A critical question is whether this threshold can be altered by environmental influences and by what mechanisms such alterations might be achieved. We examined the bioenergetic, behavioral, and CNS structural responses to weight reduction of diet-induced obese (DIO) and never-obese (CON) C57BL/6J male mice. We found that weight-reduced (WR) DIO-WR and CON-WR animals showed reductions in energy expenditure, adjusted for body mass and composition, comparable (-10-15%) to those seen in human subjects. The proportion of excitatory synapses on arcuate nucleus proopiomelanocortin neurons was decreased by ∼50% in both DIO-WR and CON-WR mice. These data suggest that prolonged maintenance of an elevated body weight (fat) alters energy homeostatic systems to defend a higher level of body fat. The synaptic changes could provide a neural substrate for the disproportionate decline in energy expenditure in weight-reduced individuals. This response to chronic weight elevation may also occur in humans. The mouse model described here could help to identify the molecular/cellular mechanisms underlying both the defense mechanisms against sustained weight loss and the upward resetting of those mechanisms following sustained weight gain.
Collapse
Affiliation(s)
- Y Ravussin
- 1Department of Pediatrics, Division of Molecular Genetics, Columbia University, College of Physicians and Surgeons, New York, New York 10032, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Donato J, Elias CF. The ventral premammillary nucleus links metabolic cues and reproduction. Front Endocrinol (Lausanne) 2011; 2:57. [PMID: 22649378 PMCID: PMC3355867 DOI: 10.3389/fendo.2011.00057] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2011] [Accepted: 10/04/2011] [Indexed: 11/15/2022] Open
Abstract
The amount of body fat and the energy balance are important factors that influence the timing of puberty and the normal reproductive function. Leptin is a key hormone that conveys to the central nervous system information about the individual energy reserve and modulates the hypothalamus-pituitary-gonad (HPG) axis. Recent findings suggest that the ventral premammillary nucleus (PMV) mediates the effects of leptin as a permissive factor for the onset of puberty and the coordinated secretion of luteinizing hormone during conditions of negative energy balance. In this review, we will summarize the existing literature about the potential role played by PMV neurons in the regulation of the HPG axis.
Collapse
Affiliation(s)
- Jose Donato
- Division of Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical CenterDallas, TX, USA
- *Correspondence: Jose Donato Jr., Division of Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Y6.206, Dallas, TX 75390, USA. e-mail:
| | - Carol Fuzeti Elias
- Division of Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical CenterDallas, TX, USA
| |
Collapse
|
28
|
El-Eshmawy MM, Abdel Aal IA, El hawary AK. Association of ghrelin and leptin with reproductive hormones in constitutional delay of growth and puberty. Reprod Biol Endocrinol 2010; 8:153. [PMID: 21176234 PMCID: PMC3022842 DOI: 10.1186/1477-7827-8-153] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2010] [Accepted: 12/22/2010] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Constitutional delay of growth and puberty (CDGP) is a variation of the onset and timing of pubertal development without a defined endocrine abnormality. Recently published studies indicate that leptin and ghrelin play a role in puberty initiation and progress. They have been implicated in regulation of GnRH secretion, with ghrelin having inhibitory and leptin, facilitatory effects. We hypothesized that elevated ghrelin and reduced leptin concentrations could be implicated in altering the tempo of puberty in adolescents with CDGP. So in the current study we evaluate variations in leptin and ghrelin levels in adolescent boys with CDGP, the relationships between both hormones and reproductive hormones including LH, FSH and testosterone were also evaluated. METHODS The study enrolled 23 adolescent boys with CDGP and 20 healthy controls matched for age and sex. Weight, height, BMI, testicular volume, bone age, bone age delay, serum FSH, LH, testosterone, leptin and ghrelin were assessed. RESULTS Adolescent boys with CDGP had significantly lower leptin and higher ghrelin than normal controls. Leptin was positively correlated with BMI, bone age, testicular volume, FSH, LH and testosterone and negatively correlated with delayed bone age and ghrelin. Ghrelin was negatively correlated with BMI, bone age, testicular volume, FSH, LH and testosterone. With multiple regression analysis BMI, FSH, LH, testosterone and ghrelin remained independently correlated with leptin while BMI, LH and testosterone remained independently correlated with ghrelin. CONCLUSION Elevated serum ghrelin and decreased leptin concentrations and their associations with reproductive hormones may explain the sexual immaturity in adolescent boys with CDGP.
Collapse
Affiliation(s)
- Mervat M El-Eshmawy
- Internal Medicine Department, Mansoura Specialized Medical Hospital, Faculty of Medicine, Mansoura University, Egypt
| | - Ibrahim A Abdel Aal
- Clinical Pathology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Amany K El hawary
- Pediatric Department, Mansoura pediatric Hospital, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
29
|
Heinrich G, Ghosh S, DeAngelis AM, Schroeder-Gloeckler JM, Patel PR, Castaneda TR, Jeffers S, Lee AD, Jung DY, Zhang Z, Opland DM, Myers MG, Kim JK, Najjar SM. Carcinoembryonic antigen-related cell adhesion molecule 2 controls energy balance and peripheral insulin action in mice. Gastroenterology 2010; 139:644-52, 652.e1. [PMID: 20381490 PMCID: PMC2910848 DOI: 10.1053/j.gastro.2010.03.056] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2010] [Revised: 03/04/2010] [Accepted: 03/25/2010] [Indexed: 01/21/2023]
Abstract
BACKGROUND & AIMS The carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) is a transmembrane glycoprotein with pleotropic functions, including clearance of hepatic insulin. We investigated the functions of the related protein CEACAM2, which has tissue-specific distribution (kidney, uterus, and crypt epithelia of intestinal tissues), in genetically modified mice. METHODS Ceacam2-null mice (Cc2-/-) were generated from a 129/SvxC57BL/6J background. Female mice were assessed by hyperinsulinemic-euglycemic clamp analysis and indirect calorimetry and body fat composition was measured. Cc2-/- mice and controls were fed as pairs, given insulin tolerance tests, and phenotypically characterized. RESULTS Female, but not male Cc2-/- mice exhibited obesity that resulted from hyperphagia and reduced energy expenditure. Pair feeding experiments showed that hyperphagia led to peripheral insulin resistance. Insulin action was normal in liver but compromised in skeletal muscle of female Cc2-/- mice; the mice had incomplete fatty acid oxidation and impaired glucose uptake and disposal. The mechanism of hyperphagia in Cc2-/- mice is not clear, but appears to result partly from increased hyperinsulinemia-induced hypothalamic fatty acid synthase levels and activity. Hyperinsulinemia was caused by increased insulin secretion. CONCLUSIONS In mice, CEACAM2 is expressed by the hypothalamus. Cc2-/- mice develop obesity from hyperphagia and reduced energy expenditure, indicating its role in regulating energy balance and insulin sensitivity.
Collapse
Affiliation(s)
- Garrett Heinrich
- Center for Diabetes and Endocrine Research at the College of Medicine at the University of Toledo, Health Science Campus, Toledo, Ohio, 43614,Department of Physiology & Pharmacology at the College of Medicine at the University of Toledo, Health Science Campus, Toledo, Ohio, 43614
| | - Sumona Ghosh
- Center for Diabetes and Endocrine Research at the College of Medicine at the University of Toledo, Health Science Campus, Toledo, Ohio, 43614,Department of Physiology & Pharmacology at the College of Medicine at the University of Toledo, Health Science Campus, Toledo, Ohio, 43614
| | - Anthony M. DeAngelis
- Center for Diabetes and Endocrine Research at the College of Medicine at the University of Toledo, Health Science Campus, Toledo, Ohio, 43614,Department of Physiology & Pharmacology at the College of Medicine at the University of Toledo, Health Science Campus, Toledo, Ohio, 43614
| | - Jill M. Schroeder-Gloeckler
- Center for Diabetes and Endocrine Research at the College of Medicine at the University of Toledo, Health Science Campus, Toledo, Ohio, 43614,Department of Physiology & Pharmacology at the College of Medicine at the University of Toledo, Health Science Campus, Toledo, Ohio, 43614
| | - Payal R. Patel
- Center for Diabetes and Endocrine Research at the College of Medicine at the University of Toledo, Health Science Campus, Toledo, Ohio, 43614,Department of Physiology & Pharmacology at the College of Medicine at the University of Toledo, Health Science Campus, Toledo, Ohio, 43614
| | - Tamara R. Castaneda
- Center for Diabetes and Endocrine Research at the College of Medicine at the University of Toledo, Health Science Campus, Toledo, Ohio, 43614,Department of Physiology & Pharmacology at the College of Medicine at the University of Toledo, Health Science Campus, Toledo, Ohio, 43614
| | - Shane Jeffers
- Center for Diabetes and Endocrine Research at the College of Medicine at the University of Toledo, Health Science Campus, Toledo, Ohio, 43614,Department of Physiology & Pharmacology at the College of Medicine at the University of Toledo, Health Science Campus, Toledo, Ohio, 43614
| | - Abraham D. Lee
- Center for Diabetes and Endocrine Research at the College of Medicine at the University of Toledo, Health Science Campus, Toledo, Ohio, 43614,Department of Physical Therapy at the College of Medicine at the University of Toledo, Health Science Campus, Toledo, Ohio, 43614
| | - Dae Young Jung
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, PA, 17033, Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605
| | - Zhiyou Zhang
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, PA, 17033
| | - Darren M. Opland
- Department of Medicine, University of Michigan, Ann Arbor, MI, 48109
| | - Martin G. Myers
- Department of Medicine, University of Michigan, Ann Arbor, MI, 48109
| | - Jason K. Kim
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, PA, 17033, Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605
| | - Sonia M. Najjar
- Center for Diabetes and Endocrine Research at the College of Medicine at the University of Toledo, Health Science Campus, Toledo, Ohio, 43614,Department of Physiology & Pharmacology at the College of Medicine at the University of Toledo, Health Science Campus, Toledo, Ohio, 43614,Address correspondence to: Sonia M. Najjar, Ph.D. College of Medicine University of Toledo Health Science Campus 3000 Arlington Avenue, Mail stop 1008 Toledo, Ohio, 43614 Tel: (419) 383-4059 FAX: (419) 383-2871
| |
Collapse
|
30
|
Leibel RL, Rosenbaum M. Metabolic Responses to Weight Perturbation. RESEARCH AND PERSPECTIVES IN ENDOCRINE INTERACTIONS 2010. [DOI: 10.1007/978-3-642-13517-0_12] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
31
|
Fernandes RM, Abreu AV, Silva RB, Silva DF, Martinez GL, Babinski MA, Ramos CF. Maternal malnutrition during lactation reduces skull growth in weaned rat pups: Experimental and morphometric investigation. Anat Sci Int 2008; 83:123-30. [DOI: 10.1111/j.1447-073x.2007.00212.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
32
|
Martin B, Golden E, Carlson OD, Egan JM, Mattson MP, Maudsley S. Caloric restriction: impact upon pituitary function and reproduction. Ageing Res Rev 2008; 7:209-24. [PMID: 18329344 DOI: 10.1016/j.arr.2008.01.002] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2007] [Revised: 01/25/2008] [Accepted: 01/28/2008] [Indexed: 01/05/2023]
Abstract
Reduced energy intake, or caloric restriction (CR), is known to extend life span and to retard age-related health decline in a number of different species, including worms, flies, fish, mice and rats. CR has been shown to reduce oxidative stress, improve insulin sensitivity, and alter neuroendocrine responses and central nervous system (CNS) function in animals. CR has particularly profound and complex actions upon reproductive health. At the reductionist level the most crucial physiological function of any organism is its capacity to reproduce. For a successful species to thrive, the balance between available energy (food) and the energy expenditure required for reproduction must be tightly linked. An ability to coordinate energy balance and fecundity involves complex interactions of hormones from both the periphery and the CNS and primarily centers upon the master endocrine gland, the anterior pituitary. In this review article we review the effects of CR on pituitary gonadotrope function and on the male and female reproductive axes. A better understanding of how dietary energy intake affects reproductive axis function and endocrine pulsatility could provide novel strategies for the prevention and management of reproductive dysfunction and its associated comorbidities.
Collapse
Affiliation(s)
- Bronwen Martin
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, 5600 Nathan Shock Drive, Baltimore, MD 21224, USA.
| | | | | | | | | | | |
Collapse
|
33
|
Tena-Sempere M. Ghrelin and reproduction: ghrelin as novel regulator of the gonadotropic axis. VITAMINS AND HORMONES 2008; 77:285-300. [PMID: 17983861 DOI: 10.1016/s0083-6729(06)77012-1] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Identification of ghrelin in late 1999, as the endogenous ligand of the growth hormone secretagogue receptor (GHSR), opened up a new era in our understanding of the regulatory mechanisms of several neuroendocrine systems, including growth and energy homeostasis. Based on similarities with other endocrine integrators and its proposed role as signal for energy insufficiency, it appeared tempting to hypothesize that ghrelin might also operate as regulator of reproductive function. Yet, contrary to other of its biological actions the reproductive "dimension" of ghrelin has remained largely unexplored. Nonetheless, experimental evidence, coming mostly from animal studies, have been gathered during the last years suggesting that ghrelin may actually function as a metabolic modulator of the gonadotropic axis, with predominant inhibitory effects in line with its role as signal of energy deficit. These effects likely include inhibition of luteinizing hormone (LH) secretion (which has been reported in different species and developmental stages), as well as partial suppression of normal puberty onset. In addition, expression and/or direct gonadal actions of ghrelin have been reported in the human, rat, and chicken. Altogether, those findings document a novel reproductive facet of ghrelin, which may cooperate with other neuroendocrine integrators, as leptin, in the joint control of energy balance and reproduction.
Collapse
Affiliation(s)
- Manuel Tena-Sempere
- Physiology Section, Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14004, Córdoba, Spain
| |
Collapse
|
34
|
Luque RM, Kineman RD, Tena-Sempere M. Regulation of hypothalamic expression of KiSS-1 and GPR54 genes by metabolic factors: analyses using mouse models and a cell line. Endocrinology 2007; 148:4601-11. [PMID: 17595226 DOI: 10.1210/en.2007-0500] [Citation(s) in RCA: 200] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
It is well established that reproductive function is metabolically gated. However, the mechanisms whereby energy stores and metabolic cues influence fertility are yet to be completely deciphered. Recently, the hypothalamic KiSS-1/GPR54 system has emerged as a fundamental regulator of the gonadotropic axis, which conveys the modulatory actions of sex steroids to GnRH neurons. Evidence is also mounting that KiSS-1 neurons may also represent the link between systemic metabolic signals and central control of reproduction. To further explore this possibility, we examined the impact of changes in energy status and key metabolic regulators on the hypothalamic expression of KiSS-1 and GPR54 genes, using different mouse models and the hypothalamic cell line N6. Time-course analysis of the effects of short-term fasting revealed a rapid (12- and 24-h) decline in KiSS-1 and GPR54 mRNA levels, which preceded that of GnRH (48 h). In contrast, diet-induced obesity or obesity associated with leptin deficiency (ob/ob vs. wild-type mice) failed to induce overt changes in hypothalamic expression of KiSS-1 and GPR54 genes. However, leptin infusion of ob/ob mice evoked a significant increase in KiSS-1 and GPR54 mRNA levels compared with pair-fed controls. Moreover, leptin, but not insulin or IGF-I, stimulated KiSS-1 mRNA expression in the mouse hypothalamic cell line N6. In addition, neuropeptide Y (NPY) null mice showed decreased KiSS-1 mRNA levels at the hypothalamus, whereas exposure to NPY increased expression of KiSS-1 in hypothalamic N6 cells. In sum, our present data further characterize the functional relevance and putative key mediators (such as leptin and NPY) of the metabolic regulation of the hypothalamic KiSS-1 system in the mouse.
Collapse
Affiliation(s)
- Raul M Luque
- Physiology Section, Department of Cell Biology, Physiology, and Immunology, University of Cordoba, Avenida Menendez Pidal, 14004 Cordoba, Spain
| | | | | |
Collapse
|
35
|
Tena-Sempere M, Barreiro ML, Lage M, Dieguez C, Casanueva FF. Role of leptin and ghrelin in the regulation of gonadal function. Expert Rev Endocrinol Metab 2007; 2:239-249. [PMID: 30754186 DOI: 10.1586/17446651.2.2.239] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Gonadal development and function is sustained by the complex interaction of an array of regulatory signals that operate directly on the gonads and/or indirectly via modulation of gonadotropin secretion. During the last decade, different factors primarily involved in the control of food intake and energy balance have been demonstrated as putative modulators of different elements of the reproductive axis, including the gonads, thus helping to define the neuroendocrine basis for the link between body energy stores and fertility. These factors include not only the adipocyte-derived hormone leptin, which is indispensable for proper energy balance and reproduction, but also a number of neuropeptides and hormones of central and peripheral origin. In the latter, growing evidence strongly suggests the involvement of the stomach-secreted peptide ghrelin in the control of several aspects of gonadal function. Interestingly, leptin and ghrelin have been proposed as reciprocally related regulators of energy homeostasis; however, their potential interplay in the control of reproduction remains ill defined. This work will summarize the most salient findings concerning the potential roles of leptin and ghrelin in the functional control of the gonads. In addition, open issues regarding the reproductive facets of these metabolic signals will be highlighted. Overall, the authors propose that through complementary or antagonistic actions, leptin and ghrelin may jointly cooperate to modulate a wide set of reproductive functions, thereby contributing to the physiologic integration of energy balance and reproduction.
Collapse
Affiliation(s)
- M Tena-Sempere
- a University of Córdoba, Physiology Section, Department of Cell Biology, Physiology & Immunology, 14004 Córdoba, Spain.
| | - M L Barreiro
- b University of Córdoba, Physiology Section, Department of Cell Biology, Physiology & Immunology, 14004 Córdoba, Spain.
| | - M Lage
- c University of Santiago de Compostela, Department of Medicine, Faculty of Medicine, Complejo Hospitalario Universitario de Santiago, 15705 Santiago de Compostela, Spain.
| | - C Dieguez
- d University of Santiago de Compostela, Department of Physiology, Faculty of Medicine, Complejo Hospitalario Universitario de Santiago, 15705 Santiago de Compostela, Spain.
| | - F F Casanueva
- e University of Santiago de Compostela, Department of Medicine, Faculty of Medicine, Complejo Hospitalario Universitario de Santiago, 15705 Santiago de Compostela, Spain.
| |
Collapse
|
36
|
Navarro VM, Castellano JM, García-Galiano D, Tena-Sempere M. Neuroendocrine factors in the initiation of puberty: the emergent role of kisspeptin. Rev Endocr Metab Disord 2007; 8:11-20. [PMID: 17340172 DOI: 10.1007/s11154-007-9028-2] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Puberty is the end-point of a complex series of developmental events, defined by the dynamic interaction between genetic factors and environmental cues, ultimately leading to the attainment of reproductive capacity. The neuroendocrine basis of puberty has been the subject of extensive investigation in the last decades, and identification of the trigger(s) of puberty onset has drawn considerable attention. In this context, recognition of the fundamental role of kisspeptin (encoded by the KiSS-1 gene) and its receptor GPR54 as major gatekeepers of gonadotropic function in general, and puberty onset in particular, has been a major breakthrough in contemporary Neuroendocrinology. Indeed, during the last 3 years, the so-called KiSS-1/GPR54 system has been substantiated as pivotal regulator of puberty in mammals; the lack of GPR54 signaling being coupled to sexual immaturity (impuberism) in mice and humans. In this review, we will summarize the most salient experimental data (mostly obtained in laboratory animals) demonstrating the key roles of hypothalamic KiSS-1 neurons in the activation of the reproductive axis at puberty, and its regulation by metabolic and, eventually, environmental factors. Whether the KiSS-1 system is the trigger for puberty onset and/or it operates as integrator and effector of up-stream regulatory factors warrants further investigation.
Collapse
Affiliation(s)
- Victor M Navarro
- Physiology Section, Department of Cell Biology, Physiology and Immunology, University of Córdoba, Avda. Menéndez Pidal s/n, 14004, Córdoba, Spain
| | | | | | | |
Collapse
|
37
|
Tena-Sempere M. Roles of ghrelin and leptin in the control of reproductive function. Neuroendocrinology 2007; 86:229-41. [PMID: 17851226 DOI: 10.1159/000108410] [Citation(s) in RCA: 107] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2006] [Accepted: 11/27/2006] [Indexed: 01/17/2023]
Abstract
Reproductive function in mammals, defined as the capacity to generate viable male and female gametes, and to support pregnancy and lactation selectively in the female, is sensitive to the metabolic state of the organism. This contention, long assumed on the basis of intuitive knowledge, became formulated on a scientific basis only recently, with the identification of a number of neuroendocrine signals which crucially participate in the joint control of energy balance and reproduction. A paradigmatic example in this context is the adipocyte-derived hormone, leptin; a satiety factor which signals the amount of body energy (fat) stores not only to the circuits controlling food intake but also to a number of neuroendocrine axes, including the reproductive system. More recently, the reproductive dimension of another metabolic hormone, namely the orexigenic stomach-secreted peptide, ghrelin, has been disclosed by observations on its putative roles in the control of gonadal function and gonadotropin secretion. Of note, leptin and ghrelin have been proposed to act as reciprocal regulators of energy homeostasis. However, their potential interplay in the control of reproduction remains largely unexplored. Based on the comparison of the biological actions of leptin and ghrelin at different levels of the hypothalamic-pituitary-gonadal axis, reviewed in detail herein, we propose that, through concurrent or antagonistic actions, the leptin-ghrelin pair is likely to operate also as modulator of different reproductive functions, thereby contributing to the physiological integration of reproduction and energy balance.
Collapse
Affiliation(s)
- Manuel Tena-Sempere
- Physiology Section, Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain.
| |
Collapse
|
38
|
Brasil FB, Faria TS, Costa WS, Sampaio FJB, Ramos CF. The pups’ endometrium morphology is affected by maternal malnutrition during suckling. Maturitas 2005; 51:405-12. [PMID: 16039415 DOI: 10.1016/j.maturitas.2004.10.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2004] [Revised: 10/19/2004] [Accepted: 10/21/2004] [Indexed: 10/26/2022]
Abstract
OBJECTIVES This study aims to determine the effects of maternal protein and energy malnutrition during lactation on the endometrial structures of the offspring at puberty. METHODS At parturition, dams were randomly assigned to the following groups: control group (C), with free access to a standard laboratory diet containing 23% protein; protein-restricted (PR) group, with free access to an isoenergy and protein-restricted diet containing 8% protein; and energy-restricted (ER) group, receiving standard laboratory diet in restricted quantities. After weaning, all female pups had free access to standard laboratory diet. At puberty, the animals were sacrificed with pentobarbital and only females on the diestrum stage were used for the analyses. The stereological method used for quantifying the uterine endometrium was the M42 test system. RESULTS When compared to C group, both PR and ER groups presented a significant reduction in the length density of the glands (PR=53%, ER=35.7%, p<0.001), in the volumetric density of the epithelium (PR=49%, ER=38%, p<0.001) and lumen (PR=42.7%, p<0.001; ER=23.8%, p<0.001) and in the surface density of the inner (PR=22%, ER=13.8%, p<0.001) and outer (PR=55.4%, p<0.01; ER=40.6%, p<0.001) glands. The volumetric density of the stroma was significantly higher in both PR (114%, p<0.001) and ER (117%, p<0.001) groups. In all parameters studied, there was no significant difference between PR and ER groups. CONCLUSIONS Our results show that the protein and energy restriction during lactation leads to an atrophy of the uterine endometrial glands of the offspring at puberty.
Collapse
Affiliation(s)
- Flávia B Brasil
- Urogenital Research Unit, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | | | | | | |
Collapse
|
39
|
Guilherme C, Bianchini A, Martinez PE, Robaldo RB, Colares EP. Serum leptin concentration during the terrestrial phase of the Southern elephant seal Mirounga leonina (Carnivora: Phocidae). Gen Comp Endocrinol 2004; 139:137-42. [PMID: 15504391 DOI: 10.1016/j.ygcen.2004.09.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2004] [Revised: 08/28/2004] [Accepted: 09/03/2004] [Indexed: 11/30/2022]
Abstract
This study aimed to verify the serum leptin concentration during the terrestrial phase of the Southern elephant seal (Mirounga leonina) on Elephant Island (South Shetlands, Antarctica). Sera from 25 adult Southern elephant seals were analyzed. Leptin concentration was determined using the 125I radioimmunoassay method. Total protein, triglycerides, and cholesterol concentration were measured by spectrophotometric methods. Seals were grouped by sex and their physiological status (reproducing or molting seals). In reproducing seals, serum concentrations of leptin, total protein, triglycerides, and cholesterol were, respectively, 9.33+/-1.97 ng/ml, 6.87+/-0.09 g/dl, 98.26+/-2.12 mg/dl, and 232.17+/-41.18 mg/dl in males and 5.30+/-1.36 ng/ml, 6.44+/-0.29 g/dl, 109.01+/-3.34 mg/dl, and 219.20+/-26.65 in females (mean+/-1 SD). In molting seals, these values were 2.35+/-1.51 ng/ml, 7.42+/-0.25 mg/dl, 321.10+/-20.01 mg/dl, and 244.66+/-22.24 mg/dl in males and 2.94+/-1.89 ng/ml, 7.88+/-0.46 mg/dl, 197.54 mg/dl, and 224.55+/-16.70 mg/dl in females. In both males and females, there were no significant differences (P>0.05) in total protein and cholesterol concentration between reproducing and molting seals. However, both males and females showed higher leptin concentration (P<0.05) and lower triglyceride concentration (P<0.05) in the reproductive period than in the molt period. In the reproductive period, it was expected that the Southern elephant seal shows an inhibited hunger sensation, mobilizes stored energy, and stimulates the HPG axis. Results from the present study support this hypothesis and suggest that this strategy may be effected by the hormonal stimulation of leptin.
Collapse
Affiliation(s)
- C Guilherme
- Programa de Pós-Graduação em Ciências Fisiológicas--Fisiologia Animal Comparada--FURG, Campus Carreiros, Av. Itália km 8, Rio Grande, RS 96201-900, Brazil
| | | | | | | | | |
Collapse
|
40
|
Teixeira RJ, Ginzbarg D, Rodrigues Freitas J, Fucks G, Silva CM, Bordallo MAN. Serum leptin levels in premature pubarche and prepubertal girls with and without obesity. J Pediatr Endocrinol Metab 2004; 17:1393-8. [PMID: 15526717 DOI: 10.1515/jpem.2004.17.10.1393] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Leptin can be regarded as a marker of the nutritional status of the body. This study was performed to determine the correlation of leptin levels with insulin (I) and androgens in girls with premature pubarche (PP) and prepubertal controls (C) with (OB) or without (nOB) obesity. We studied 25 girls with PP and 14 C; girls were dived into two subgroups according to body mass index (BMI): OB (18 PP and 8 C) and nOB (7 PP and 6 C). Obesity was defined as BMI >95th percentile for chronological age. Serum levels of leptin, I, glucose (G), DHEAS, testosterone, androstenedione (A), cortisol, SHBG, IGFBP-1 and lipid profile were measured. The fasting G to I ratio (FGIR) was calculated and FGIR <7 was considered as suggestive of I resistance (IR). Data were analyzed comparing PP vs C and OB vs nOB. Serum DHEAS (0.60 +/- 0.45 vs 0.18 +/- 0.22 microg/ml) and A (895.5 +/- 420.4 vs 457.0 +/- 352.1 pg/ml) levels were significantly higher in PP than C. Other hormonal and metabolic parameters were similar. Serum leptin (30.8 +/- 18.3 vs 8.1 +/- 5.9 ng/ml), A (841.8 +/- 471.1 vs 522.5 +/- 317.2 pg/ml), DHEAS (0.53 +/- 0.44 vs 0.31 +/- 0.39 microg/ml), G (88.4 +/- 8.8 vs 80.2 +/- 8.1 mg/dl), I (13.5 +/- 7.7 vs 5.1 +/- 3.7 microU/ml) and total cholesterol (TC) (180.5 +/- 30.9 vs 161.8 +/- 29.5 mg/dl) levels were greater in the OB than in the nOB group. IR was observed in 10 girls with OB and in one with nOB. Leptin was correlated with BMI (r = 0.83), SHBG (r = -0.44), IGFBP-1 (r = -0.47), I (r = 0.37), A (r = 0.48) and TC (r = 0.36), but in multiple regression analysis only with BMI (r2 = 0.72, p < 0.001). Girls with PP and prepubertal OB girls showed elevated leptin levels independent of I and androgen levels. Girls with OB had a greater degree of hyperandrogenism and IR. As obesity, IR and hyperandrogenism are common findings in polycystic ovary syndrome (PCOS), which is more prevalent in young women with a history of PP, a role of leptin in PCOS can be suggested. In addition, girls with PP could be considered a population at risk for plurimetabolic syndrome.
Collapse
Affiliation(s)
- Rosimere J Teixeira
- Department of Endocrinology, Hospital Universitario Pedro Ernesto (HUPE), State University of Rio de Janeiro, Rio de Janeiro, Brazil.
| | | | | | | | | | | |
Collapse
|
41
|
Barreiro ML, Pineda R, Navarro VM, Lopez M, Suominen JS, Pinilla L, Señaris R, Toppari J, Aguilar E, Diéguez C, Tena-Sempere M. Orexin 1 receptor messenger ribonucleic acid expression and stimulation of testosterone secretion by orexin-A in rat testis. Endocrinology 2004; 145:2297-306. [PMID: 14764632 DOI: 10.1210/en.2003-1405] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Orexins are hypothalamic neuropeptides primarily involved in the regulation of food intake and arousal states. In addition, a role for orexins as central neuroendocrine modulators of reproductive function has recently emerged. Prepro-orexin and orexin type-1 receptor mRNAs have been detected in the rat testis. This raises the possibility of additional peripheral actions of orexins in the control of reproductive axis, which remains so far unexplored. To analyze the biological effects and mechanisms of action of orexins in the male gonad, we evaluated testicular expression of orexin receptor 1 (OX(1)R) and orexin receptor 2 (OX(2)R) mRNAs in different experimental settings and the effect of orexin-A on testicular testosterone (T) secretion. Persistent expression of OX(1)R mRNA was demonstrated in the rat testis throughout postnatal development. In contrast, OX(2)R transcript was not detected at any developmental stage. Expression of OX(1)R mRNA persisted after selective elimination of mature Leydig cells and was detected in isolated seminiferous tubules at defined stages of the seminiferous epithelial cycle. In addition, testicular OX(1)R mRNA expression appeared to be under hormonal regulation; it was reduced by long-term hypophysectomy and partially restored by FSH replacement, whereas down-regulation was observed after exposure to increasing doses of the ligand in vitro. Moreover, OX(1)R mRNA expression was sensitive to neonatal imprinting by estrogen. Finally, orexin-A, in a dose-dependent manner, significantly increased basal, but not human choriogonadotropin-stimulated, T secretion in vitro. A similar stimulatory effect was observed in vivo after intratesticular administration of orexin-A. In conclusion, our present results provide the first evidence for the regulated expression of OX(1)R mRNA and functional role of orexin-A in the rat testis. Overall, our data are suggestive of a novel site of action of orexins in the control of male reproductive axis.
Collapse
Affiliation(s)
- M L Barreiro
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14004 Córdoba, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Amills M, Jiménez N, Villalba D, Tor M, Molina E, Cubiló D, Marcos C, Francesch A, Sànchez A, Estany J. Identification of three single nucleotide polymorphisms in the chicken insulin-like growth factor 1 and 2 genes and their associations with growth and feeding traits. Poult Sci 2003; 82:1485-93. [PMID: 14601723 DOI: 10.1093/ps/82.10.1485] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The chicken insulin-like growth factor (IGF)1 and IGF2 genes have been partially sequenced in six individuals of each of two chicken strains of the Black Penedesenca breed (PN and MN). These two strains are genetically diverse for growth traits. Sequence alignment revealed the existence of three single nucleotide polymorphisms (SNP) (IGF1-SNP1, IGF2-SNP2, and IGF2-SNP3). These three SNP and a fourth IGF1 polymorphism (IGF1-SNP4) were typed in 60 individuals from each strain by using PCR-RFLP or primer extension analysis. No significant associations among these four SNP, growth traits, and plasma IGF1 concentration were identified. In contrast, suggestive associations (P < or = 0.05) were found between IGF1-SNP1 and average daily gain at 107 d and feed efficiency at 44, 73, and 107 d. However, these associations were not simultaneously found in both strains suggesting that they might have been produced by linkage disequilibrium with another mutation located in the IGF1 locus or another linked gene. Since the PN and MN strains differ very markedly on their feed intake, the chicken leptin gene was included in the sequence analysis. Unfortunately, attempts to amplify several regions of this gene were unsuccessful. Even when primers complementary to highly conserved regions were used, the PCR consistently failed. Other authors have reported similar problems when trying to amplify avian leptin sequences.
Collapse
Affiliation(s)
- M Amills
- Unitat de Ciència Animal, Departament de Ciència Animal i dels Aliments, Facultat de Veterinària, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Watanobe H, Habu S. Manipulation of neonatal gonadal steroid milieu and leptin secretion in later life in male and female rats. REGULATORY PEPTIDES 2003; 110:219-24. [PMID: 12573803 DOI: 10.1016/s0167-0115(02)00215-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The mechanism underlying the gender-based difference in circulating leptin levels (females>males) is still uncertain, because the difference persists even after adjustment for fat mass and sex steroid concentrations. In this study, we tested the possibility that the neonatal sex steroid milieu, which is critical for the sexual differentiation of the brain, may permanently affect leptin secretion in rats of both sexes. Male rats were neonatally castrated (NC), and females were neonatally androgenized (NA) by testosterone (T). Two subsets of the NC males were given T on postnatal day 1 or 29. Appropriate controls for all these groups were prepared. The animals were sacrificed on postnatal day 57, and at this age, the percent body fat was similar among all the male and female groups. NC males had a two-fold, significantly higher level of leptin than intact males. This hyperleptinemia induced by NC was prevented by T when it was given neonatally, but not on the day 29. By contrast, NA for females was without effect on leptin titers in later life. These results suggest that neonatal T in male rats may, at least in part, mediate the sex-related difference in leptin secretion that becomes apparent in later life. However, as intact females still had significantly higher leptin titers than NC males, it is very likely that additional factors may also be responsible for the sexually dimorphic leptin secretion in rats.
Collapse
Affiliation(s)
- Hajime Watanobe
- Division of Internal Medicine, Clinical Research Center, International University of Health and Welfare, 2600-1 Kitakanemaru, Otawara, Tochigi 324-8501, Japan.
| | | |
Collapse
|
44
|
Léonhardt M, Lesage J, Croix D, Dutriez-Casteloot I, Beauvillain JC, Dupouy JP. Effects of perinatal maternal food restriction on pituitary-gonadal axis and plasma leptin level in rat pup at birth and weaning and on timing of puberty. Biol Reprod 2003; 68:390-400. [PMID: 12533401 DOI: 10.1095/biolreprod.102.003269] [Citation(s) in RCA: 112] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
The effects of maternal 50% food restriction (FR) during the last week of gestation and/or lactation on pituitary-gonadal axis (at birth and weaning), on circulating levels of leptin (at weaning), and on the onset of puberty have been determined in rats at birth and at weaning. Maternal FR during pregnancy has no effect at term on the litter size, on the basal level of testosterone in male pups, and on the drastic surge of circulating testosterone that occurs 2 h after birth. At weaning, similar retardation of body growth is observed in male and female pups from mothers exposed to FR. This undernutrition induces the most drastic effects when it is performed during both gestation and lactation or during lactation alone. Drastic retardation of testicle growth with reduction of cross-sectional area and intratubular lumen of the seminiferous tubules is observed in male pups from mothers exposed to undernutrition during both gestation and lactation or during lactation alone. Maternal FR during the perinatal period reduces circulating levels of FSH in male pups without affecting LH and testosterone concentrations. Maternal FR does not affect circulating levels of LH, estradiol, and progesterone in female pups. Female pups from mothers exposed to FR during both gestation and lactation show a significant increase of plasma FSH as well as a drastic retardation of ovarian growth. The follicular population was also altered. The number of antral follicles of small size (vesicular follicles) was increased, although the number of antral follicles of large size (graafian follicles) was reduced. Maternal FR occurring during both late gestation and lactation (male and female pups), during lactation alone (male and female pups), or during late gestation (female pups) induces a drastic reduction of plasma leptin and fat mass in pups at weaning. The onset of puberty is delayed in pups of both sexes from mothers exposed to FR during lactation and during both gestation and lactation. In conclusion, these data demonstrate that a perinatal growth retardation induced by maternal FR has long-term consequences on both size and histology of the genitals, on plasma gonadotropins and leptin levels, on fat stores at weaning, and on the onset of puberty.
Collapse
Affiliation(s)
- Marion Léonhardt
- Laboratoire de Neuroendocrinologie du Développement, UPRES 2701, Université de Lille 1, 59655 Villeneuve d'Ascq, France
| | | | | | | | | | | |
Collapse
|
45
|
Zaman N, Hall CM, Gill MS, Jones J, Tillmann V, Westwood M, Whatmore AJ, Clayton PE. Leptin measurement in urine in children and its relationship to other growth peptides in serum and urine. Clin Endocrinol (Oxf) 2003; 58:78-85. [PMID: 12519416 DOI: 10.1046/j.1365-2265.2003.01677.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE Leptin has been implicated in the interaction between nutrition, energy balance and sexual maturation in humans. A non-invasive method of measuring leptin would greatly facilitate longitudinal studies of changes in leptin in normal children. The aim of this study was to evaluate the use of urinary leptin as a surrogate for serum leptin measurements. DESIGN We have modified and validated a serum immunoradiometric assay (IRMA) kit for the measurement of leptin in urine, and subsequently investigated the relationship between urinary leptin and other growth-related proteins. METHODS Cross-sectional study: urinary leptin, measured in the first morning urine voided and expressed as ng excreted overnight, and serum concentrations of leptin, IGF-I, IGF-II, IGFBP-3 and IGFBP-1 were determined in a cohort of 188 healthy schoolchildren aged 5-19 years (88 males, 100 females). Height, weight and pubertal status were assessed in all children. Longitudinal study: urinary levels of leptin, IGF-I and GH were measured daily in two adults (one male, one female) over a period of 6 weeks. RESULTS The detection limit of this modified assay was 0.59 ng/L. The intra- and interassay coefficients of variation range was 4-8% and 4-9%, respectively. The recovery of recombinant leptin added to urine was 98-108%, and the assay had a recovery rate for serial dilution in the range of 106-112%. Urinary leptin correlated significantly with serum leptin (r = +0.65, P < 0.01). Urinary leptin showed similar changes through puberty to those of serum leptin, with levels rising in females throughout puberty, whereas in males levels peaked at G2/G3 then decreased. BMI SDS was the main determinant of urinary leptin, as it was for serum leptin. In the cross-sectional study urinary leptin correlated significantly with serum IGF-I (r = +0.41, P = 0.001), IGF-II (r = +0.19, P = 0.05), IGFBP-3 (r = +0.29, P = 0.001) and IGFBP-1 (r = -0.25, P = 0.001). In the adult study, leptin was also detected in urine with similar night-to-night variability to that found for IGF-I and GH. CONCLUSION Urinary leptin is a valid marker of serum leptin concentrations, and therefore this non-invasive assay would be a useful tool for longitudinal assessment of changes in leptin in children.
Collapse
Affiliation(s)
- Nasra Zaman
- Endocrine Sciences Research Group, University of Manchester, UK
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
Leptin, the adipocyte-derived hormone that plays a key role in body weight homeostasis, has recently emerged as a relevant neuroendocrine mediator in different systems, including the reproductive axis. Thus, compelling evidence points out a major role of leptin in the regulation of female pubertal development and fertility, both in humans and experimental animals. The contribution of leptin to the proper functioning of the male reproductive system has been less clear. However, data gathered in recent years, from independent groups and through a variety of experimental approaches, strongly suggest that leptin is able to act at different levels of the hypothalamic-pituitary-testicular axis. Herein, we review the biological effects and potential mechanisms of action of leptin upon rodent testis. Leptin appears to act as a direct inhibitory signal for testicular steroidogenesis, which may be relevant to explain the link between decreased testosterone secretion and hyperleptinaemia in obese men. Analysis of the molecular basis for leptin-induced inhibition of testosterone secretion revealed the potential involvement of decreased gene expression of several up-stream factors (e.g. SF-1, StAR and P450scc) in the steroidogenic pathway. In this context, testicular expression of leptin receptor (Ob-R) gene shows a complex pattern of alternative splicing with generation of multiple variants, including the functional leptin receptor type-b (Ob-Rb) and several short isoforms. Moreover, Ob-R mRNA expression in rat testis was regulated by homologous (leptin) as well as heterologous (gonadotropins) signals. Overall, the current data indicate that the testis is a direct target for leptin actions. Furthermore, the available evidence is suggestive of a tightly regulated, complex mode of action of leptin at different levels of the male gonadal axis that involves not only stimulatory but also inhibitory effects.
Collapse
Affiliation(s)
- M Tena-Sempere
- Department of Cell Biology, Physiology and Immunology (Physiology Section), Faculty of Medicine, University of Córdoba, Avda. Menéndez Pidal s/n, 14004 Cordoba, Spain.
| | | |
Collapse
|
47
|
Benini ZL, Camilloni MA, Scordato C, Lezzi G, Savia G, Oriani G, Bertoli S, Balzola F, Liuzzi A, Petroni ML. Contribution of weight cycling to serum leptin in human obesity. Int J Obes (Lond) 2001; 25:721-6. [PMID: 11360156 DOI: 10.1038/sj.ijo.0801587] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2000] [Revised: 10/11/2000] [Accepted: 11/06/2000] [Indexed: 11/08/2022]
Abstract
OBJECTIVE To investigate to what extent serum leptin concentrations in obese humans are influenced by a history of weight cycling. DESIGN Cross-sectional study on serum leptin concentrations and body composition in a cohort of obese subjects in whom a retrospective recall of weight and diet history was made. SUBJECTS One hundred and twenty-eight obese patients (89 females and 39 males), aged 18-61 y, body mass index (BMI) 31.2-63.4 kg/m(2). MEASUREMENTS Serum leptin; various fatness and fat distribution parameters (by anthropometry and bioelectrical impedance analysis); history of overweight at puberty; number, magnitude and timing of previous diet episodes and of consequent weight regain by interview. RESULTS By univariate analysis, serum leptin concentrations were significantly correlated with weight, waist-hip ratio, percentage body fat, maximal percentage weight loss in a single diet episode, cumulative percentage weight loss in all diet episodes, cumulative weight regained in all diet episodes, but not with the number of diet episodes. All correlations related to anthropometric and body composition parameters were stronger for men, compared to women, although the male subgroup was smaller. On the contrary, there was a strong positive correlation between weight cycling parameters and serum leptin in women but not in men. Leptin concentrations were significantly higher in patients who were overweight at puberty than in those who were not overweight at puberty. After correction for percentage body fat, presence of overweight at puberty did not correlate any longer with leptin concentrations in either gender. In women, cumulative percentage weight loss in all diet episodes contributed an additional 5% to the variance of serum leptin in the overall model. CONCLUSION The positive correlation between weight cycling and leptin concentration in obesity is mainly accounted for the higher percentage body fat in obese weight cyclers, although in women weight cycling per se independently contributes to the variance of serum leptin.
Collapse
Affiliation(s)
- Z L Benini
- Clinical Nutrition Laboratory, Istituto Auxologico Italiano, Piancavallo, Verbania, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Graham KS, Leibel RL. Yellow mice, red hair, and childhood obesity: the melanocortinergic pathway in energy homeostasis. J Pediatr 2001; 139:177-81. [PMID: 11487740 DOI: 10.1067/mpd.2001.117574] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
49
|
Dhillon H, Kalra SP, Prima V, Zolotukhin S, Scarpace PJ, Moldawer LL, Muzyczka N, Kalra PS. Central leptin gene therapy suppresses body weight gain, adiposity and serum insulin without affecting food consumption in normal rats: a long-term study. REGULATORY PEPTIDES 2001; 99:69-77. [PMID: 11384767 DOI: 10.1016/s0167-0115(01)00237-3] [Citation(s) in RCA: 83] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The weight-reducing effects of leptin are predominantly mediated through the hypothalamus in the brain. Gene therapy strategies designed for weight control have so far tested the short-term effect of peripherally delivered viral vectors encoding the leptin gene. In order to circumvent the multiple peripheral effects of hyperleptinemia and to overcome the age-related development of leptin resistance due to multiple factors, including defective leptin transport across the blood brain barrier, we determined whether delivery of viral vectors directly into the brain is a viable therapeutic strategy for long-term weight control in normal wild-type rats. A recombinant adeno-associated virus (rAAV) vector encoding rat leptin (Ob) cDNA was generated (rAAV-betaOb). When administered once intracerebroventricularly (i.c.v.), rAAV-betaOb suppressed the normal time-related weight gain for extended periods of time in adult Sprague-Dawley rats. The vector expression was confirmed by immunocytochemical localization of GFP and RT-PCR analysis of leptin in the hypothalamus. This sustained restraint on weight gain was not due to shifts in caloric consumption because food-intake was similar in rAAV-betaOb-treated and rAAV-GFP-treated control rats throughout the experiment. Weight gain suppression, first apparent after 2 weeks, was a result of reduced white fat depots and was accompanied by drastically reduced serum leptin and insulin concentrations in conjunction with normoglycemia. Additionally, there was a marked increase in uncoupling protein-1 (UCP1) mRNA expression in brown adipose tissue, thereby indicating increased energy expenditure through thermogenesis. Seemingly, a selective enhancement in energy expenditure following central delivery of the leptin gene is a viable therapeutic strategy to control the age-related weight gain and provide protection from the accompanying multiple peripheral effects of hyperleptinemia and hyperinsulinemia.
Collapse
Affiliation(s)
- H Dhillon
- Department of Physiology, College of Medicine, Box 100274, University of Florida, Gainesville, FL 32610-0274, USA
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Tena-Sempere M, Pinilla L, Zhang FP, González LC, Huhtaniemi I, Casanueva FF, Dieguez C, Aguilar E. Developmental and hormonal regulation of leptin receptor (Ob-R) messenger ribonucleic acid expression in rat testis. Biol Reprod 2001; 64:634-43. [PMID: 11159367 DOI: 10.1095/biolreprod64.2.634] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
In target tissues, leptin receptor (Ob-R) gene expression results in an array of alternatively spliced isoforms (Ob-Ra to Ob-Rf) with different functional features. Recent evidence has pointed to a direct role of leptin in the control of testicular function. However, complete elucidation of the pattern of Ob-R gene expression in the male gonad is still pending. The focus of this study was to characterize in detail the developmental pattern of expression and hormonal regulation of Ob-R gene in rat testis. To this end, the overall expression of Ob-R mRNA was compared to that of the fully functional, long Ob-Rb isoform in different experimental settings, using semiquantitative reverse transcription-polymerase chain reaction. Expression of Ob-R mRNA was detected in testes from 15-, 30-, 45-, and 75-day-old rats at rather constant relative levels. In contrast, testicular expression of Ob-Rb mRNA was higher in pubertal testes (15- to 30-day-old rats) and declined in adulthood. In testes from 30-day-old animals, analysis of isoform distribution revealed that, in addition to abundant Ob-Rb mRNA levels, expression of Ob-Ra, Ob-Rf, and, to a lesser extent, Ob-Rc and Ob-Re messages is detected. Testicular Ob-R mRNA expression appeared sensitive to neonatal imprinting as neonatal treatment with estradiol benzoate (500 microg/rat; Day 1 postpartum) resulted in a persistent increase (P: < 0.01) in the relative expression level of Ob-R mRNA, a phenomenon only partially mimicked by neonatal suppression of serum gonadotropins by means of LHRH-antagonist administration. In addition, neonatal estrogenization differentially altered the pattern of expression of Ob-R isoforms in adult rat testis, as expression of Ob-Rb mRNA was decreased to undetectable levels, whereas that of Ob-Rc remained unaltered, and Ob-Ra, Ob-Rf, and, to a lesser extent, Ob-Re mRNA levels were significantly increased (P: < 0.01) by neonatal exposure to estrogen. Finally, down-regulation of testicular Ob-R gene expression by homologous and heterologous signals was demonstrated as relative levels of Ob-R and Ob-Rb mRNAs were significantly decreased (P: < 0.01), in a coordinate manner, in rat testis after exposure to human recombinant leptin in vitro, and after stimulation with hCG and FSH in vivo. In conclusion, our results indicate that testicular Ob-R gene expression is developmentally regulated, imprinted by the neonatal endocrine milieu, and sensitive to regulation by leptin and gonadotropins. The ability of pivotal signals in testicular function to regulate Ob-R gene expression further supports the contention of a direct role of leptin in functional control of the rat testis.
Collapse
Affiliation(s)
- M Tena-Sempere
- Department of Physiology, Faculty of Medicine, University of Córdoba, Avda Menéndez Pidal s/n, 14004 Córdoba, Spain.
| | | | | | | | | | | | | | | |
Collapse
|