1
|
Young JA, Hinrichs A, Bell S, Geitgey DK, Hume-Rivera D, Bounds A, Soneson M, Laron Z, Yaron-Shaminsky D, Wolf E, List EO, Kopchick JJ, Berryman DE. Growth hormone insensitivity and adipose tissue: tissue morphology and transcriptome analyses in pigs and humans. Pituitary 2023; 26:660-674. [PMID: 37747600 PMCID: PMC10956721 DOI: 10.1007/s11102-023-01355-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/18/2023] [Indexed: 09/26/2023]
Abstract
PURPOSE Growth hormone receptor knockout (GHR-KO) pigs have recently been developed, which serve as a large animal model of Laron syndrome (LS). GHR-KO pigs, like individuals with LS, are obese but lack some comorbidities of obesity. The purpose of this study was to examine the histological and transcriptomic phenotype of adipose tissue (AT) in GHR-KO pigs and humans with LS. METHODS Intraabdominal (IA) and subcutaneous (SubQ) AT was collected from GHR-KO pigs and examined histologically for adipocyte size and collagen content. RNA was isolated and cDNA sequenced, and the results were analyzed to determine differentially expressed genes that were used for enrichment and pathway analysis in pig samples. For comparison, we also performed limited analyses on human AT collected from a single individual with and without LS. RESULTS GHR-KO pigs have increased adipocyte size, while the LS AT had a trend towards an increase. Transcriptome analysis revealed 55 differentially expressed genes present in both depots of pig GHR-KO AT. Many significant terms in the enrichment analysis of the SubQ depot were associated with metabolism, while in the IA depot, IGF and longevity pathways were negatively enriched. In pathway analysis, multiple expected and novel pathways were significantly affected by genotype, i.e. KO vs. controls. When GH related gene expression was analyzed, SOCS3 and CISH showed species-specific changes. CONCLUSION AT of GHR-KO pigs has several similarities to that of humans with LS in terms of adipocyte size and gene expression profile that help describe the depot-specific adipose phenotype of both groups.
Collapse
Affiliation(s)
- Jonathan A Young
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Arne Hinrichs
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany
- Center for Innovative Medical Models (CiMM), Department of Veterinary Sciences, LMU Munich, Oberschleißheim, Germany
| | - Stephen Bell
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | | | | | - Addison Bounds
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
| | - Maggie Soneson
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
| | - Zvi Laron
- Endocrinology and Diabetes Research Unit, Schneider Children's Medical Center, Petah Tikva, Israel
| | - Danielle Yaron-Shaminsky
- Endocrinology and Diabetes Research Unit, Schneider Children's Medical Center, Petah Tikva, Israel
| | - Eckhard Wolf
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany
- Center for Innovative Medical Models (CiMM), Department of Veterinary Sciences, LMU Munich, Oberschleißheim, Germany
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, Munich, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany
| | - Edward O List
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - John J Kopchick
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Darlene E Berryman
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA.
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA.
| |
Collapse
|
2
|
List EO, Duran-Ortiz S, Kulkarni P, Davis E, Mora-Criollo P, Berryman DE, Kopchick JJ. Growth hormone receptor gene disruption. VITAMINS AND HORMONES 2023; 123:109-149. [PMID: 37717983 PMCID: PMC11462719 DOI: 10.1016/bs.vh.2022.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2023]
Abstract
Much of our understanding of growth hormone's (GH)'s numerous activities stems from studies utilizing GH receptor (GHR) knockout mice. More recently, the role of GH action has been examined by creating mice with tissue-specific or temporal GHR disruption. To date, 37 distinct GHR knockout mouse lines have been created. Targeted tissues include fat, liver, muscle, heart, bone, brain, macrophage, intestine, hematopoietic stem cells, pancreatic β cells, and inducible multi-tissue "global" disruption at various ages. In this chapter, a summary of each mouse line is provided with background information on the generation of the mouse line as well as important physiological outcomes resulting from GHR gene disruption. Collectively, these mouse lines provide unique insights into GH action and have resulted in the development of new hypotheses about the functions ascribed to GH action in particular tissues.
Collapse
Affiliation(s)
- Edward O List
- The Edison Biotechnology Institute, and the Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, United States
| | - Silvana Duran-Ortiz
- The Edison Biotechnology Institute, and the Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, United States
| | - Prateek Kulkarni
- The Edison Biotechnology Institute, and the Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, United States
| | - Emily Davis
- The Edison Biotechnology Institute, and the Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, United States
| | - Patricia Mora-Criollo
- The Edison Biotechnology Institute, and the Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, United States
| | - Darlene E Berryman
- The Edison Biotechnology Institute, and the Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, United States
| | - John J Kopchick
- The Edison Biotechnology Institute, and the Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, United States.
| |
Collapse
|
3
|
Backe MB, Andersen RC, Jensen M, Jin C, Hundahl C, Dmytriyeva O, Treebak JT, Hansen JB, Gerhart-Hines Z, Madsen KL, Holst B. PICK1-Deficient Mice Maintain Their Glucose Tolerance During Diet-Induced Obesity. J Endocr Soc 2023; 7:bvad057. [PMID: 37200849 PMCID: PMC10185814 DOI: 10.1210/jendso/bvad057] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Indexed: 05/20/2023] Open
Abstract
Context Metabolic disorders such as obesity represent a major health challenge. Obesity alone has reached epidemic proportions, with at least 2.8 million people worldwide dying annually from diseases caused by overweight or obesity. The brain-metabolic axis is central to maintain homeostasis under metabolic stress via an intricate signaling network of hormones. Protein interacting with C kinase 1 (PICK1) is important for the biogenesis of various secretory vesicles, and we have previously shown that PICK1-deficient mice have impaired secretion of insulin and growth hormone. Objective The aim was to investigate how global PICK1-deficient mice respond to high-fat diet (HFD) and assess its role in insulin secretion in diet-induced obesity. Methods We characterized the metabolic phenotype through assessment of body weight, composition, glucose tolerance, islet morphology insulin secretion in vivo, and glucose-stimulated insulin secretion ex vivo. Results PICK1-deficient mice displayed similar weight gain and body composition as wild-type (WT) mice following HFD. While HFD impaired glucose tolerance of WT mice, PICK1-deficient mice were resistant to further deterioration of their glucose tolerance compared with already glucose-impaired chow-fed PICK1-deficient mice. Surprisingly, mice with β-cell-specific knockdown of PICK1 showed impaired glucose tolerance both on chow and HFD similar to WT mice. Conclusion Our findings support the importance of PICK1 in overall hormone regulation. However, importantly, this effect is independent of the PICK1 expression in the β-cell, whereby global PICK1-deficient mice resist further deterioration of their glucose tolerance following diet-induced obesity.
Collapse
Affiliation(s)
- Marie Balslev Backe
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Department of Clinical Epidemiology, Steno Diabetes Center Copenhagen, 2730 Herlev, Denmark
| | - Rita Chan Andersen
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Morten Jensen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Chunyu Jin
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Cecilie Hundahl
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Oksana Dmytriyeva
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Jonas T Treebak
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Jakob Bondo Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Zach Gerhart-Hines
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Kenneth L Madsen
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Birgitte Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
4
|
Delpisheh A, Safarzade A. The effect of high-intensity interval training on serum and adipose tissues vaspin levels in rats fed a high-fat high-sucrose diet. Horm Mol Biol Clin Investig 2022; 43:449-453. [PMID: 35796296 DOI: 10.1515/hmbci-2021-0107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 06/11/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Vaspin is an adipocytokine with insulin-sensitizing and anti-inflammatory traits. OBJECTIVES The purpose of this study was to evaluate the effect of high-intensity interval training (HIIT) on serum, visceral and subcutaneous adipose tissue vaspin levels in rats exposed to a diet high in fat and sugar (HFS). MATERIALS AND METHODS Thirty-two male Wistar rats were randomly divided into HFS and standard diet (SD) groups. After 12 weeks, each group was divided into sedentary and HIIT groups. HIIT program was performed 3 times/week for 8 weeks. Retroperitoneal adipose tissue, inguinal adipose tissue and serum were collected to analyze vaspin levels. Also, serum glucose and insulin levels, insulin resistance index (HOMA-IR) and retroperitoneal and inguinal fat weights were measured. RESULTS HFS significantly increased weight gain, weight of inguinal (p=0.001) and retroperitoneal fat depots (p<0.001), serum glucose levels (p<0.001) and HOMA-IR (p<0.001). The HIIT was able to decline weight gain and fat mass (p<0.05) but did not affect inguinal and retroperitoneal fat depots' vaspin levels. Eight weeks' HIIT significantly increased serum vaspin (p=0.002) and decreased insulin (p=0.001) levels only in rats fed with SD. CONCLUSIONS Although the HIIT program can cause significantly reducing effects on weight gain and fat depots' weights, it does not effect on circulating and fat depots' vaspin levels in rats fed an HFS.
Collapse
Affiliation(s)
- Ali Delpisheh
- Department of Exercise Physiology, Faculty of Sports Sciences, University of Mazandaran, Babolsar, Iran
| | - Alireza Safarzade
- Department of Exercise Physiology, Faculty of Sports Sciences, University of Mazandaran, Babolsar, Iran.,Athletic Performance and Health Research Center, University of Mazandaran, Babolsar, Iran
| |
Collapse
|
5
|
Al-Massadi O, Parini P, Fernø J, Luquet S, Quiñones M. Metabolic actions of the growth hormone-insulin growth factor-1 axis and its interaction with the central nervous system. Rev Endocr Metab Disord 2022; 23:919-930. [PMID: 35687272 DOI: 10.1007/s11154-022-09732-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/02/2022] [Indexed: 10/18/2022]
Abstract
The growth hormone/insulin growth factor-1 axis is a key endocrine system that exerts profound effects on metabolism by its actions on different peripheral tissues but also in the brain. Growth hormone together with insulin growth factor-1 perform metabolic adjustments, including regulation of food intake, energy expenditure, and glycemia. The dysregulation of this hepatic axis leads to different metabolic disorders including obesity, type 2 diabetes or liver disease. In this review, we discuss how the growth hormone/insulin growth factor-1 axis regulates metabolism and its interactions with the central nervous system. Finally, we state our vision for possible therapeutic uses of compounds based in the components of this hepatic axis.
Collapse
Affiliation(s)
- Omar Al-Massadi
- Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain.
- CIBER de Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, 15706, Santiago de Compostela, Spain.
| | - Paolo Parini
- Department of Laboratory Medicine, Division of Clinical Chemistry, Karolinska Institute, Stockholm, Sweden
- Department of Medicine, Metabolism Unit, Karolinska Institute at Karolinska University Hospital Huddinge, Stockholm, Sweden
- Patient Area Nephrology and Endocrinology, Inflammation and Infection Theme, Karolinska University Hospital, Stockholm, Sweden
| | - Johan Fernø
- Hormone Laboratory, Haukeland University Hospital, Bergen, Norway
| | - Serge Luquet
- Unité de Biologie Fonctionnelle et Adaptative, Univ Paris Diderot, Sorbonne Paris Cité, CNRS UMR 8251, F-75205, Paris, France
| | - Mar Quiñones
- Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain.
- CIBER de Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, 15706, Santiago de Compostela, Spain.
- Unité de Biologie Fonctionnelle et Adaptative, Univ Paris Diderot, Sorbonne Paris Cité, CNRS UMR 8251, F-75205, Paris, France.
| |
Collapse
|
6
|
Brown-Borg HM. Growth hormone, not IGF-1 is the key longevity regulator in mammals. J Gerontol A Biol Sci Med Sci 2022; 77:1719-1723. [PMID: 35436323 PMCID: PMC9434454 DOI: 10.1093/gerona/glac092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Holly M Brown-Borg
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND
| |
Collapse
|
7
|
Poklukar K, Čandek-Potokar M, Vrecl M, Batorek-Lukač N, Fazarinc G, Kress K, Weiler U, Stefanski V, Škrlep M. The effect of immunocastration on adipose tissue deposition and composition in pigs. Animal 2020; 15:100118. [PMID: 33712216 DOI: 10.1016/j.animal.2020.100118] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 10/08/2020] [Accepted: 10/12/2020] [Indexed: 12/29/2022] Open
Abstract
Immunocastrated pigs (IC) exhibit intensive fat deposition after immunisation, but the underlying mechanisms of intensified fat metabolism and deposition are not yet fully understood. Moreover, there is also a lack of comparative studies performed on IC, entire males (EM) and surgical castrates (SC). The main objective of our research was, therefore, to characterise the adipose tissue from the quantitative, histo-morphological and biochemical perspectives in IC 5 weeks after their immunisation in comparison to EM and SC. Immunocastrated pigs had an intermediate position in carcass fatness traits between EM (the leanest) and SC (the fattest). The histo-morphological traits of the subcutaneous adipose tissue of IC were similar to those of SC and differed from those of EM; i.e., they exhibited larger adipocytes in the outer backfat and a larger lobulus surface area in both backfat layers than EM. Intensive fat tissue development in IC was corroborated with higher activities of lipogenic enzymes (i.e., fatty acid synthase, malic enzyme, glucose 6-phosphate dehydrogenase, citrate cleavage enzyme), which was especially pronounced in the subcutaneous adipose tissue of IC (1.5- to 2.7-fold higher activity than in EM or SC). The fatty acid composition of the backfat in IC was similar to that in EM pigs. Both IC and EM exhibited less saturated and more polyunsaturated fatty acids than SC. In contrast, the fatty acid composition of the intramuscular fat of longissimus dorsi muscle in IC pigs was more similar to SC than to EM (higher monounsaturated and lower polyunsaturated fatty acid content in IC and SC than EM). In this study, it was demonstrated that immunocastration notably influenced lipid metabolism. This was shown by increased quantity of lipid depots and with changes in adipose tissue cellularity compared to EM, with changes in the fatty acid composition of the intramuscular fat and enhanced lipogenic activity compared to both EM and SC. These results provide new insights into the specificity of adipose tissue development and deposition in IC compared to EM and SC.
Collapse
Affiliation(s)
- K Poklukar
- Animal Science Department, Agricultural Institute of Slovenia, Hacquetova 17, 1000 Ljubljana, Slovenia
| | - M Čandek-Potokar
- Animal Science Department, Agricultural Institute of Slovenia, Hacquetova 17, 1000 Ljubljana, Slovenia; Faculty of Agriculture and Life Sciences, University of Maribor, Pivola 10, 2311 Hoče, Slovenia
| | - M Vrecl
- Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, Gerbičeva 60, 1000 Ljubljana, Slovenia
| | - N Batorek-Lukač
- Animal Science Department, Agricultural Institute of Slovenia, Hacquetova 17, 1000 Ljubljana, Slovenia
| | - G Fazarinc
- Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, Gerbičeva 60, 1000 Ljubljana, Slovenia
| | - K Kress
- Behavioral Physiology of Livestock, Institute of Animal Science, University of Hohenheim, Garbenstr. 17, 70599 Stuttgart, Germany
| | - U Weiler
- Behavioral Physiology of Livestock, Institute of Animal Science, University of Hohenheim, Garbenstr. 17, 70599 Stuttgart, Germany
| | - V Stefanski
- Behavioral Physiology of Livestock, Institute of Animal Science, University of Hohenheim, Garbenstr. 17, 70599 Stuttgart, Germany
| | - M Škrlep
- Animal Science Department, Agricultural Institute of Slovenia, Hacquetova 17, 1000 Ljubljana, Slovenia.
| |
Collapse
|
8
|
Duran-Ortiz S, Young JA, Jara A, Jensen EA, Basu R, List EO, Qian Y, Kopchick JJ, Berryman DE. Differential gene signature in adipose tissue depots of growth hormone transgenic mice. J Neuroendocrinol 2020; 32:e12893. [PMID: 33043505 PMCID: PMC7606825 DOI: 10.1111/jne.12893] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 05/18/2020] [Accepted: 07/15/2020] [Indexed: 02/05/2023]
Abstract
Bovine growth hormone (bGH) transgenic mice mimic the clinical condition of acromegaly, having high circulating growth hormone (GH) levels. These mice are giant, have decreased adipose tissue (AT) mass, impaired glucose metabolism and a shortened lifespan. The detrimental effects of excess GH have been suggested, in part, to be a result of its depot-specific actions on AT. To investigate this relationship, we evaluated gene expression, biological mechanisms, cellular pathways and predicted microRNA (miRNA) in two AT depots (subcutaneous [Subq] and epididymal [Epi]) from bGH and littermate controls using RNA sequencing analysis. Two analyses on the differentially expressed genes (DEG) were performed: (i) comparison of the same AT depot between bGH and wild-type (WT) mice (genotype comparison) and (ii) comparison of Subq and Epi AT depots within the same genotype (depot comparison). For the genotype comparison, we found a higher number of significant DEG in the Subq AT depot of bGH mice compared to WT controls, corroborating previous reports that GH has a greater impact on the Subq depot. Furthermore, most of the DEG in bGH mice were not shared by WT mice, suggesting that excess GH induces the expression of genes not commonly present in AT. Through gene ontology and pathway analysis, the genotype comparison revealed that the DEG of the Subq depot of bGH mice relate to fatty acid oxidation, branched-chain amino acid degradation and the immune system. Additionally, the AT depot comparison showed that the immune cell activation and T-cell response appear up-regulated in the Subq compared to the Epi AT depot. The miRNA prediction also suggested a modulation of T-cell-related biological process in Subq. In summary, the present study provides a unique resource for understanding the specific differences in gene expression that are driven by both excess GH action and AT depot location.
Collapse
Affiliation(s)
- Silvana Duran-Ortiz
- Edison Biotechnology Institute, Athens, OH
- Molecular and Cellular Biology Program, Ohio University, Athens, OH
- Department of Biological Sciences, College of Arts and Sciences, Ohio University, Athens, OH
| | - Jonathan A. Young
- Edison Biotechnology Institute, Athens, OH
- Molecular and Cellular Biology Program, Ohio University, Athens, OH
- Department of Biological Sciences, College of Arts and Sciences, Ohio University, Athens, OH
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH
| | - Adam Jara
- Edison Biotechnology Institute, Athens, OH
- Molecular and Cellular Biology Program, Ohio University, Athens, OH
- Department of Biological Sciences, College of Arts and Sciences, Ohio University, Athens, OH
| | | | | | | | | | - John J. Kopchick
- Edison Biotechnology Institute, Athens, OH
- Molecular and Cellular Biology Program, Ohio University, Athens, OH
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH
| | - Darlene E. Berryman
- Edison Biotechnology Institute, Athens, OH
- Molecular and Cellular Biology Program, Ohio University, Athens, OH
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH
- Corresponding Author at: Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA.
| |
Collapse
|
9
|
Škrlep M, Tomašević I, Mörlein D, Novaković S, Egea M, Garrido MD, Linares MB, Peñaranda I, Aluwé M, Font-i-Furnols M. The Use of Pork from Entire Male and Immunocastrated Pigs for Meat Products-An Overview with Recommendations. Animals (Basel) 2020; 10:E1754. [PMID: 32993171 PMCID: PMC7601181 DOI: 10.3390/ani10101754] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/05/2020] [Accepted: 09/09/2020] [Indexed: 01/09/2023] Open
Abstract
Due to the strong public initiative in Europe and increased regulator focus to mitigate pain, surgical castration of pigs is being gradually abandoned, while the importance of other sex categories like entire males (EM) and immunocastrates (IC) increases. Although beneficial for animal welfare and economics, their use also brings forward several quality problems. Besides the occurrence of boar taint in EM, these include excessive carcass leanness, softer fat, meat color and pH deviations, inferior water holding capacity and increased meat toughness. In this paper, the raw material differences between the male sex categories and their influence on product quality are reviewed, and possible solutions are presented. Using EM for dried or thermally processed products may result in lower processing yields and inferior sensory quality, which may partially be prevented by applying specific processing adaptations. Immunocastration is a viable solution, especially when prolonging the vaccination to slaughter interval. Low to medium levels of boar taint can be effectively managed in most of the meat products, applying procedures like cooking, microbial inoculation or masking (by spices and especially smoking), while highly tainted material can be valorized only by combining various methods and/or with dilution of the tainted meat.
Collapse
Affiliation(s)
- Martin Škrlep
- Agricultural Institute of Slovenia, Hacquetova ulica 17, SI-1000 Ljubljana, Slovenia
| | - Igor Tomašević
- Faculty of Agriculture, University of Belgrade, Nemanjina 6, 11080 Belgrade, Serbia; (I.T.); (S.N.)
| | - Daniel Mörlein
- Department of Animal Sciences, University of Göttingen, Albrecht-Thaer-Weg 3, 37075 Göttingen, Germany;
| | - Saša Novaković
- Faculty of Agriculture, University of Belgrade, Nemanjina 6, 11080 Belgrade, Serbia; (I.T.); (S.N.)
| | - Macarena Egea
- Department of Food Science and Technology, Veterinary Faculty, University of Murcia, Espinardo, 30071 Murcia, Spain; (M.E.); (M.D.G.); (M.B.L.); (I.P.)
| | - María Dolores Garrido
- Department of Food Science and Technology, Veterinary Faculty, University of Murcia, Espinardo, 30071 Murcia, Spain; (M.E.); (M.D.G.); (M.B.L.); (I.P.)
| | - María Belén Linares
- Department of Food Science and Technology, Veterinary Faculty, University of Murcia, Espinardo, 30071 Murcia, Spain; (M.E.); (M.D.G.); (M.B.L.); (I.P.)
| | - Irene Peñaranda
- Department of Food Science and Technology, Veterinary Faculty, University of Murcia, Espinardo, 30071 Murcia, Spain; (M.E.); (M.D.G.); (M.B.L.); (I.P.)
| | - Marijke Aluwé
- Flanders Research Institute for Agriculture, Fisheries and Food (ILVO), Scheldeweg 68, 9090 Melle, Belgium;
| | | |
Collapse
|
10
|
Duran-Ortiz S, Noboa V, Kopchick JJ. Tissue-specific disruption of the growth hormone receptor (GHR) in mice: An update. Growth Horm IGF Res 2020; 51:1-5. [PMID: 31923746 PMCID: PMC9704042 DOI: 10.1016/j.ghir.2019.12.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 12/02/2019] [Accepted: 12/21/2019] [Indexed: 01/01/2023]
Abstract
The Growth hormone receptor (GHR) is expressed in many cells/tissues in the body. To investigate the specific metabolic effects of GH action in distinct tissues, several tissue-specific GHR gene disrupted or knockout (KO) mouse lines have been generated. Previously, we have described the effects of GHRKO in several known insulin sensitive tissues, namely liver, muscle and adipose tissue. In this review, we further explore and summarize the main findings of recently published GHRKO results in liver, adipocytes, intestine, bone, brain and heart.
Collapse
Affiliation(s)
- Silvana Duran-Ortiz
- Edison Biotechnology Institute, United States of America; Department of Biological Sciences, College of Arts and Sciences, United States of America; Molecular and Cellular Biology Program, United States of America.
| | - Vanessa Noboa
- School of Medicine, Universidad San Francisco de Quito, United States of America.
| | - John J Kopchick
- Edison Biotechnology Institute, United States of America; Molecular and Cellular Biology Program, United States of America; Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, United States of America.
| |
Collapse
|
11
|
Pinet K, McLaughlin KA. Mechanisms of physiological tissue remodeling in animals: Manipulating tissue, organ, and organism morphology. Dev Biol 2019; 451:134-145. [DOI: 10.1016/j.ydbio.2019.04.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 03/29/2019] [Accepted: 04/03/2019] [Indexed: 12/21/2022]
|
12
|
Yurekli BS, Kutbay NO, Aksit M, Suner A, Simsir IY, Seckiner S, Kocabas GU, Bozkaya G, Saygili F. Acromegaly is associated with high fibroblast growth factor-21 levels. J Endocrinol Invest 2019; 42:53-60. [PMID: 29754168 DOI: 10.1007/s40618-018-0885-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 04/03/2018] [Indexed: 11/11/2022]
Abstract
PURPOSE Fibroblast growth factor-21 (FGF-21) is a member of fibroblast growth factor family. Both growth hormone (GH) and FGF-21 take place in the regulation of glucose and lipid metabolism. We aimed to investigate FGF-21 levels in acromegaly which is characterized by excess GH levels and is associated with comorbidities and altered body composition. METHODS We studied 43 subjects (21 females and 22 males, mean age of 50.0 ± 12.8) with acromegaly. The control group consisted of 40 gender- and age-matched subjects (25 females and 15 males, mean age of 48.8 ± 8.8). Acromegaly patients were classified into two groups; active acromegaly (AA; n = 26) and controlled acromegaly (CA; n = 17). Metabolic, anthropometric and laboratory values of subjects were recorded. FGF-21 level was measured by ELISA assay. RESULTS Median FGF-21 levels were significantly higher in acromegaly group compared to control group (85.5 vs. 59.0 pg/mL, p = 0.02, respectively). In the multiple regression model, FPG, A1c, HOMA-IR, glucose intolerance, BMI, visceral fat, hs-CRP, presence of hypertension, dyslipidemia and acromegaly were included as independent variables to explain variability of plasma FGF-21 levels in whole study group. The presence of acromegaly was the only determinant of increased FGF-21 levels in the whole study group (β coefficient = 0.253, p = 0.006). CONCLUSION FGF-21 levels were increased significantly in acromegaly group. Increased FGF-21 levels were significantly and independently associated with the state of acromegaly. Acromegaly may also be a FGF-21 resistance state independent from insulin resistance, glucose intolerance, obesity, hypertension and dyslipidemia.
Collapse
Affiliation(s)
- B S Yurekli
- Division of Endocrinology, Ege University Faculty of Medicine, Ankara Street, Bornova, Izmir, Turkey.
| | - N O Kutbay
- Division of Endocrinology, Ege University Faculty of Medicine, Ankara Street, Bornova, Izmir, Turkey
| | - M Aksit
- Department of Biochemistry, Izmir Bozyaka Education and Research Hospital, Izmir, Turkey
| | - A Suner
- Department of Biostatistics, Ege University Faculty of Medicine, Izmir, Turkey
| | - I Y Simsir
- Division of Endocrinology, Ege University Faculty of Medicine, Ankara Street, Bornova, Izmir, Turkey
| | - S Seckiner
- Department of Nutrition and Dietetics, Ege University Faculty of Medicine, Izmir, Turkey
| | - G U Kocabas
- Division of Endocrinology, Izmir Bozyaka Education and Research Hospital, Izmir, Turkey
| | - G Bozkaya
- Department of Biochemistry, Izmir Bozyaka Education and Research Hospital, Izmir, Turkey
| | - F Saygili
- Division of Endocrinology, Ege University Faculty of Medicine, Ankara Street, Bornova, Izmir, Turkey
| |
Collapse
|
13
|
Castillo AR, de Souza AL, Alegre SM, Atala YB, Zantut-Wittmann DE, Garmes HM. Insulin Sensitivity Is Not Decreased in Adult Patients With Hypopituitarism Without Growth Hormone Replacement. Front Endocrinol (Lausanne) 2019; 10:534. [PMID: 31447781 PMCID: PMC6692434 DOI: 10.3389/fendo.2019.00534] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 07/18/2019] [Indexed: 11/13/2022] Open
Abstract
Decreased insulin sensitivity in patients with hypopituitarism without GH replacement (pHP-WGHR) remains conflicting in literature. It is known that these patients present a decrease in free fat mass and an increase in fat mass. Typically, these kinds of alterations in body composition are associated with a decrease in insulin sensitivity; however, there is no consensus if this association is found in pHP-WGHR. Thus, we investigated pHP-WGHR regarding insulin sensitivity by euglycemic hyperinsulinemic clamp, the gold standard method, and body composition. In a cross-sectional study, we evaluated 15 pHP-WGHR followed up in a Service of Neuroendocrinology and 15 individuals with normal pituitary function as a control group with similar age, gender and body mass index. Insulin sensitivity was evaluated by euglycemic hyperinsulinemic clamp and homeostatic model assessment insulin resistance (HOMA-IR). Kappa coefficient evaluated the agreement between these two methods. Percentage of fat mass, percentage of free fat mass, fat mass weight and free fat mass weight were assessed by electrical bioimpedance. The pHP-WGHR presented similar insulin sensitivity to control group by euglycemic hyperinsulinemic clamp, both by the M-value, (p = 0.0913) and by the area under the glucose infusion rate curve, (p = 0.0628). These patients showed lower levels of fasting glycemia (p = 0.0128), insulin (p = 0.0007), HOMA-IR (p = 0.009). HOMA-IR shows poor concordance with euglycemic hyperinsulinemic clamp (Kappa = 0.16) in pHP-WGHR, while in the control group the agreement was good (Kappa = 0.53). The pHP-WGHR presented higher values of percentage of fat mass (p = 0.0381) and lower values of percentage of free fat mass (p = 0.0464) and free fat mass weight (0.0421) than the control group. This study demonstrated that the insulin sensitivity evaluated by euglycemic hyperinsulinemic clamp in pHP-WGHR was similar to individuals with normal pituitary function, despite the pHP-WGHR presenting higher fat mass percentage. HOMA-IR was not a good method for assessing insulin sensitivity in pHP-WGHR.
Collapse
Affiliation(s)
- Alejandro Rosell Castillo
- Endocrinology Division, Department of Clinical Medicine, Faculty of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Aglecio Luiz de Souza
- Department of Internal Medicine, Faculty of Medical Sciences, State University of Campinas, Campinas, Brazil
| | - Sarah Monte Alegre
- Department of Internal Medicine, Faculty of Medical Sciences, State University of Campinas, Campinas, Brazil
| | - Yeelen Ballesteros Atala
- Endocrinology Division, Department of Clinical Medicine, Faculty of Medical Sciences, University of Campinas, Campinas, Brazil
| | | | - Heraldo Mendes Garmes
- Endocrinology Division, Department of Clinical Medicine, Faculty of Medical Sciences, University of Campinas, Campinas, Brazil
- *Correspondence: Heraldo Mendes Garmes
| |
Collapse
|
14
|
Xie T, Ding H, Xia M, Zhang X, Sun W, Liu T, Gu Y, Sun C, Hu F. Dynamic changes in the distribution of facial and abdominal adipose tissue correlated with surgical treatment in acromegaly. Endocrine 2018; 62:552-559. [PMID: 30203120 DOI: 10.1007/s12020-018-1742-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 08/29/2018] [Indexed: 12/31/2022]
Abstract
PURPOSE Acromegaly is a systemic metabolic disease. Growth hormone (GH) have a significant impact on adipose tissue (AT). A huge reduction of serum GH after surgical treatment may cause substantial AT redistribution. The objective of this study was to illustrate the dynamic changes in distribution of facial and abdominal AT correlated with surgical treatment in patients with acromegaly. METHODS Abdominal AT in 17 acromegaly patients (group 1) was studied longitudinally preoperatively and 1 month to 1 year postoperatively. The facial and abdominal subcutaneous AT (fSAT and aSAT) of another 17 acromegaly patients (group 2) were compared with 7 nonfunctional pituitary adenoma (NFPA) controls. The areas of fSAT, aSAT, and visceral adipose tissue (VAT) were obtained by MRI and quantified by image analysis software, and intrahepatic lipid (IHL) was assessed by 1H magnetic resonance spectroscopy (MRS). RESULTS Abdominal adipose tissue (aSAT, VAT, and IHL) increased overall after surgical treatment. However, IHL first decreased and then continuously increased during the follow-up. Compared with the increased amount of aSAT, the fSAT amount decreased after surgical treatment. The inconsistency of this phenomenon did not appear in the NFPA control subjects. CONCLUSION The perioperative dynamic distribution of the facial and abdominal fat in acromegaly revealed regional differences in the intricate effect of GH on adipose tissue. Reduction of serum GH after surgical treatment of acromegaly was associated with dynamic increases of IHL, abdominal visceral, and subcutaneous fat, but a reduction of facial subcutaneous fat.
Collapse
Affiliation(s)
- Tao Xie
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hailin Ding
- Department of Emergency, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Mingfeng Xia
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaobiao Zhang
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai, China.
- Digital Medical Research Center, Zhongshan Hospital, Fudan University, Shanghai, China.
- Shanghai Key Laboratory of Medical Image Computing and Computer-Assisted Intervention, Shanghai, China.
| | - Wei Sun
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tengfei Liu
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ye Gu
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chongjing Sun
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Fan Hu
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
15
|
Rojas JM, Bolze F, Thorup I, Nowak J, Dalsgaard CM, Skydsgaard M, Berthelsen LO, Keane KA, Søeborg H, Sjögren I, Jensen JT, Fels JJ, Offenberg HK, Andersen LW, Dalgaard M. The Effect of Diet-induced Obesity on Toxicological Parameters in the Polygenic Sprague-Dawley Rat Model. Toxicol Pathol 2018; 46:777-798. [DOI: 10.1177/0192623318803557] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
| | - Florian Bolze
- Toxicology Development Projects, Novo Nordisk A/S, Måløv, Denmark
| | - Inger Thorup
- Toxicopathology, Novo Nordisk A/S, Måløv, Denmark
| | - Jette Nowak
- Toxicopathology, Novo Nordisk A/S, Måløv, Denmark
| | | | | | | | | | | | | | | | | | | | | | - Majken Dalgaard
- Early Regulatory Toxicology, Novo Nordisk A/S, Måløv, Denmark
| |
Collapse
|
16
|
Darcy J, McFadden S, Fang Y, Berryman DE, List EO, Milcik N, Bartke A. Increased environmental temperature normalizes energy metabolism outputs between normal and Ames dwarf mice. Aging (Albany NY) 2018; 10:2709-2722. [PMID: 30334813 PMCID: PMC6224234 DOI: 10.18632/aging.101582] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 09/26/2018] [Indexed: 06/08/2023]
Abstract
Ames dwarf (Prop1df) mice possess a loss-of-function mutation that results in deficiency of growth hormone, prolactin, and thyroid-stimulating hormone, as well as exceptional longevity. Work in other laboratories suggests that increased respiration and lipid utilization are important for maximizing mammalian longevity. Interestingly, these phenotypes are observed in Ames dwarf mice. We recently demonstrated that Ames dwarf mice have hyperactive brown adipose tissue (BAT), and hypothesized that this may in part be due to their increased surface to mass ratio leading to increased heat loss and an increased demand for thermogenesis. Here, we used increased environmental temperature (eT) to interrogate this hypothesis. We found that increased eT diminished BAT activity in Ames dwarf mice, and led to the normalization of both VO2 and respiratory quotient between dwarf and normal mice, as well as partial normalization (i.e. impairment) of glucose homeostasis in Ames dwarf mice housed at an increased eT. Together, these data suggest that an increased demand for thermogenesis is partially responsible for the improved energy metabolism and glucose homeostasis which are observed in Ames dwarf mice.
Collapse
Affiliation(s)
- Justin Darcy
- Division of Geriatric Research, Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL 62702, USA
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL 62702, USA
- Current Affiliation: Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02115, USA
| | - Samuel McFadden
- Division of Geriatric Research, Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL 62702, USA
| | - Yimin Fang
- Division of Geriatric Research, Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL 62702, USA
| | - Darlene E. Berryman
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
- The Diabetes Institute at Ohio University, Ohio University, Athens, OH 45701, USA
| | - Edward O. List
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA
| | - Nicholas Milcik
- Division of Geriatric Research, Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL 62702, USA
| | - Andrzej Bartke
- Division of Geriatric Research, Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL 62702, USA
| |
Collapse
|
17
|
Hjortebjerg R. IGFBP-4 and PAPP-A in normal physiology and disease. Growth Horm IGF Res 2018; 41:7-22. [PMID: 29864720 DOI: 10.1016/j.ghir.2018.05.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 05/15/2018] [Accepted: 05/29/2018] [Indexed: 02/07/2023]
Abstract
Insulin-like growth factor (IGF) binding protein-4 (IGFBP-4) is a modulator of the IGF system, exerting both inhibitory and stimulatory effects on IGF-induced cellular growth. IGFBP-4 is the principal substrate for the enzyme pregnancy-associated plasma protein-A (PAPP-A). Through IGF-dependent cleavage of IGFBP-4 in the vicinity of the IGF receptor, PAPP-A is able to increase IGF bioavailability and stimulate IGF-mediated growth. Recently, the stanniocalcins (STCs) were identified as novel inhibitors of PAPP-A proteolytic activity, hereby adding additional members to the seemingly endless list of proteins belonging to the IGF family. Our understanding of these proteins has advanced throughout recent years, and there is evidence to suggest that the role of IGFBP-4 and PAPP-A in defining the relationship between total IGF and IGF bioactivity can be linked to a number of pathological conditions. This review provides an overview of the experimental and clinical findings on the IGFBP-4/PAPP-A/STC axis as a regulator of IGF activity and examines the conundrum surrounding extrapolation of circulating concentrations to tissue action of these proteins. The primary focus will be on the biological significance of IGFBP-4 and PAPP-A in normal physiology and in pathophysiology with emphasis on metabolic disorders, cardiovascular diseases, and cancer. Finally, the review assesses current new trajectories of IGFBP-4 and PAPP-A research.
Collapse
Affiliation(s)
- Rikke Hjortebjerg
- Medical Research Laboratory, Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark; The Danish Diabetes Academy, Odense, Denmark.
| |
Collapse
|
18
|
Jia D, Zheng W, Jiang H. Growth hormone facilitates 5'-azacytidine-induced myogenic but inhibits 5'-azacytidine-induced adipogenic commitment in C3H10T1/2 mesenchymal stem cells. Growth Horm IGF Res 2018; 40:9-16. [PMID: 29626795 DOI: 10.1016/j.ghir.2018.03.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 02/19/2018] [Accepted: 03/27/2018] [Indexed: 11/20/2022]
Abstract
The C3H10T1/2 cells are considered mesenchymal stem cells (MSCs) because they can be induced to become the progenitor cells for myocytes, adipocytes, osteoblasts, and chondrocytes by the DNA methyltransferase inhibitor 5'-azacytidine. In this study, we determined the effect of growth hormone (GH) on the myogenic and adipogenic lineage commitment in C3H10T1/2 cells. The C3H10T1/2 cells were treated with recombinant bovine GH in the presence or absence of 5'-azacytidine for 4 days. The myogenic commitment in C3H10T1/2 cells was assessed by immunostaining them for MyoD, the marker for myoblasts, and by determining their capacity to differentiate into the multinucleated myotubes. The adipogenic commitment in C3H10T1/2 cells was assessed by determining their ability to differentiate into adipocytes. Myotubes and adipocyteswere identified by immunocytochemistry and Oil Red O staining, respectively. C3H10T1/2 cells treated with 5'-azacytidine and GH for 4 days contained a greater percentage of MyoD-positive cells than those treated with 5'-axacytidine alone (P < 0.05). The former generated more myotubes than the latter upon induced myoblast differentiation (P < 0.05). However, C3H10T1/2 cells treated with GH alone did not form any myotubes. C3H10T1/2 cells treated with 5'-azacytidine formed adipocytes upon adipocyte differentiation induction, whereas C3H10T1/2 cells treated with GH alone did not form any adipocytes. C3H10T1/2 cells treated with both 5'-azacytidine and GH formed fewer adipocytes than those treated with 5'-azacytidine alone (P < 0.05). Both GHR and IGF-I mRNA expression in C3H10T1/2 cells were increased by 5'-azacytidine (P < 0.05), but neither was affected by GH. Overall, this study showed that GH enhanced 5'-azacytidine-induced commitment in C3H10T1/2 cells to myoblasts but inhibited 5'-azacytidine-induced commitment to preadipocytes. These results support the possibility that GH stimulates skeletal muscle growth and inhibits adipose tissue growth in part by stimulating the myogenic commitment and inhibiting the adipogenic commitment, respectively, in mesenchymal stem cells.
Collapse
Affiliation(s)
- Dan Jia
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA, United States
| | - Weijiang Zheng
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA, United States; College of Animal Sciences, Nanjing Agricultural University, Nanjing, China
| | - Honglin Jiang
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA, United States.
| |
Collapse
|
19
|
Chae HS, You BH, Kim DY, Lee H, Ko HW, Ko HJ, Choi YH, Choi SS, Chin YW. Sauchinone controls hepatic cholesterol homeostasis by the negative regulation of PCSK9 transcriptional network. Sci Rep 2018; 8:6737. [PMID: 29712938 PMCID: PMC5928089 DOI: 10.1038/s41598-018-24935-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 04/12/2018] [Indexed: 12/15/2022] Open
Abstract
Whole-transcriptome analysis and western blotting of sauchinone-treated HepG2 cells demonstrated that sauchinone regulated genes relevant to cholesterol metabolism and synthesis. In particular, it was found that the expression of proprotein convertase subtilisin/kexin type 9 (PCSK9) was downregulated, and the expression of low density lipoprotein receptor (LDLR) was upregulated in sauchinone-treated HepG2 cells. Consequently, LDL-cholesterol (LDL-C) uptake was increased. As a transcriptional regulator of PCSK9 expression, sterol regulatory elements binding protein-2 (SREBP-2) was proposed by transcriptome analysis and western blotting. Oral administration of sauchinone increased hepatic LDLR through PCSK9 inhibition in obese mice and showed the reduced serum LDL-C levels and downstream targets of SREBP-2. Thus, it is evident that sauchinone reduces hepatic steatosis by downregulating the expression of hepatic PCSK9 via SREBP-2.
Collapse
Affiliation(s)
- Hee-Sung Chae
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang, Gyeonggi-do, 10326, Republic of Korea
| | - Byoung Hoon You
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang, Gyeonggi-do, 10326, Republic of Korea
| | - Dong-Yeop Kim
- Division of Biomedical Convergence, College of Biomedical Science, and Institute of Bioscience & Biotechnology, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Hankyu Lee
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang, Gyeonggi-do, 10326, Republic of Korea
| | - Hyuk Wan Ko
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang, Gyeonggi-do, 10326, Republic of Korea
| | - Hyun-Jeong Ko
- Laboratory of Microbiology and Immunology, College of Pharmacy, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Young Hee Choi
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang, Gyeonggi-do, 10326, Republic of Korea
| | - Sun Shim Choi
- Division of Biomedical Convergence, College of Biomedical Science, and Institute of Bioscience & Biotechnology, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Young-Won Chin
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang, Gyeonggi-do, 10326, Republic of Korea.
| |
Collapse
|
20
|
Householder LA, Comisford R, Duran-Ortiz S, Lee K, Troike K, Wilson C, Jara A, Harberson M, List EO, Kopchick JJ, Berryman DE. Increased fibrosis: A novel means by which GH influences white adipose tissue function. Growth Horm IGF Res 2018; 39:45-53. [PMID: 29279183 PMCID: PMC5858978 DOI: 10.1016/j.ghir.2017.12.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 12/18/2017] [Accepted: 12/19/2017] [Indexed: 01/10/2023]
Abstract
OBJECTIVE White adipose tissue (WAT) fibrosis - the buildup of extracellular matrix (ECM) proteins, primarily collagen - is now a recognized hallmark of tissue dysfunction and is increased with obesity and lipodystrophy. While growth hormone (GH) is known to increase collagen in several tissues, no previous research has addressed its effect on ECM in WAT. Thus, the purpose of this study is to determine if GH influences WAT fibrosis. DESIGN This study examined WAT from four distinct strains of GH-altered mice (bGH and GHA transgenic mice as well as two tissue specific GH receptor gene disrupted lines, fat growth hormone receptor knockout or FaGHRKO and liver growth hormone receptor knockout or LiGHRKO mice). Collagen content and adipocyte size were studied in all cohorts and compared to littermate controls. In addition, mRNA expression of fibrosis-associated genes was assessed in one cohort (6month old male bovine GH transgenic and WT mice) and cultured 3T3-L1 adipocytes treated with GH. RESULTS Collagen stained area was increased in WAT from bGH mice, was depot-dependent, and increased with age. Furthermore, increased collagen content was associated with decreased adipocyte size in all depots but more dramatic changes in the subcutaneous fat pad. Notably, the increase in collagen was not associated with an increase in collagen gene expression or other genes known to promote fibrosis in WAT, but collagen gene expression was increased with acute GH administration in 3T3-LI cells. In contrast, evaluation of 6month old GH antagonist (GHA) male mice showed significantly decreased collagen in the subcutaneous depot. Lastly, to assess if GH induced collagen deposition directly or indirectly (via IGF-1), fat (Fa) and liver (Li) specific GHRKO mice were evaluated. Decreased fibrosis in FaGHRKO and increased fibrosis in LiGHRKO mice suggest GH is primarily responsible for the alterations in collagen. CONCLUSIONS Our results show that GH action is positively associated with an increase in WAT collagen content as well as a decrease in adipocyte size, particularly in the subcutaneous depot. This effect appears to be due to GH and not IGF-1 and reveals a novel means by which GH regulates WAT accumulation.
Collapse
Affiliation(s)
- Lara A Householder
- The Diabetes Institute, Ohio University, Athens, OH, United States; Edison Biotechnology Institute, Ohio University, Athens, OH, United States; School of Applied Health Sciences and Wellness, College of Health Sciences and Professions, United States
| | - Ross Comisford
- The Diabetes Institute, Ohio University, Athens, OH, United States; Edison Biotechnology Institute, Ohio University, Athens, OH, United States
| | - Silvana Duran-Ortiz
- Edison Biotechnology Institute, Ohio University, Athens, OH, United States; School of Applied Health Sciences and Wellness, College of Health Sciences and Professions, United States
| | - Kevin Lee
- The Diabetes Institute, Ohio University, Athens, OH, United States; Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, United States
| | - Katie Troike
- The Diabetes Institute, Ohio University, Athens, OH, United States; Edison Biotechnology Institute, Ohio University, Athens, OH, United States; School of Applied Health Sciences and Wellness, College of Health Sciences and Professions, United States
| | - Cody Wilson
- The Diabetes Institute, Ohio University, Athens, OH, United States; Edison Biotechnology Institute, Ohio University, Athens, OH, United States
| | - Adam Jara
- Edison Biotechnology Institute, Ohio University, Athens, OH, United States
| | - Mitchell Harberson
- The Diabetes Institute, Ohio University, Athens, OH, United States; Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, United States
| | - Edward O List
- Edison Biotechnology Institute, Ohio University, Athens, OH, United States
| | - John J Kopchick
- The Diabetes Institute, Ohio University, Athens, OH, United States; Edison Biotechnology Institute, Ohio University, Athens, OH, United States; Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, United States
| | - Darlene E Berryman
- The Diabetes Institute, Ohio University, Athens, OH, United States; Edison Biotechnology Institute, Ohio University, Athens, OH, United States; Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, United States.
| |
Collapse
|
21
|
Hjortebjerg R, Berryman DE, Comisford R, List EO, Oxvig C, Bjerre M, Frystyk J, Kopchick JJ. Depot-specific and GH-dependent regulation of IGF binding protein-4, pregnancy-associated plasma protein-A, and stanniocalcin-2 in murine adipose tissue. Growth Horm IGF Res 2018; 39:54-61. [PMID: 29398370 DOI: 10.1016/j.ghir.2018.01.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 12/21/2017] [Accepted: 01/15/2018] [Indexed: 01/23/2023]
Abstract
INTRODUCTION Pregnancy-associated plasma protein-A (PAPP-A) stimulates insulin-like growth factor (IGF)-I action through proteolytic cleavage of IGF binding protein-4 (IGFBP-4). Recently, stanniocalcin-2 (STC2) was discovered as an inhibitor of PAPP-A. Most members of the IGF system are expressed in adipose tissue (AT), but there is a relative paucity of information on the distribution of IGFBP-4, PAPP-A, and STC2 in different AT depots. Since IGF-I expression in AT is highly GH-dependent, we used bovine GH transgenic (bGH) and GH receptor knockout (GHR-/-) mice to investigate AT depot-specific expression patterns of IGFBP-4, PAPP-A, and STC2, and whether the regulation is GH-dependent. METHODS Seven-month-old male bGH, GHR-/- and wild type (WT) control mice were used. Body composition was determined, and subcutaneous, epididymal, retroperitoneal, mesenteric and brown adipose tissue (BAT) depots were collected. RNA expression of Igfbp4, Pappa, and Stc2 was assessed by reverse transcription quantitative PCR and IGFBP-4 protein by Western blotting. RESULTS Igfbp4, Pappa, and Stc2 RNA levels were differentially expressed in an AT depot-dependent manner in WT mice. Igfbp4 RNA levels were significantly higher in all white AT depots than in BAT. Pappa was most highly expressed in the mesenteric depot: levels were 7.5-fold higher in mesenteric than in subcutaneous AT (p < .001). Although intraabdominal in origin, epididymal and retroperitoneal Pappa expression levels were 69% and 68% lower, respectively, as compared to mesenteric levels (p < .001). Stc2 RNA expression was significantly higher in all intraabdominal white AT as compared to subcutaneous AT and BAT; levels in epididymal, retroperitoneal, and mesenteric were all more than three-fold higher than in subcutaneous AT (p < .001) and 12-fold higher than in BAT (p < .001). Gene expression patterns in bGH and GHR-/- mice mimicked those in WT mice, suggesting that GH does not affect the transcription of the STC2-PAPP-A-IGFBP-4-axis in AT. However, proteins levels of intact IGFBP-4 were significantly increased in bGH mice and decreased in GHR-/- mice, whereas the PAPP-A-generated IGFBP-4 fragment level was unaltered. CONCLUSION Expression of Igfbp4, Pappa, and Stc2 differ between AT depots and is generally higher in white AT than in BAT. The transcription appears to occur in a GH-independent manner, whereas IGFBP-4 protein levels are highly influenced by altered GH activity.
Collapse
Affiliation(s)
- Rikke Hjortebjerg
- Medical Research Laboratory, Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark; Danish Diabetes Academy, Odense, Denmark; Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA.
| | - Darlene E Berryman
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA; The Diabetes Institute, Ohio University, Athens, OH 45701, USA.
| | - Ross Comisford
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; The Diabetes Institute, Ohio University, Athens, OH 45701, USA.
| | - Edward O List
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; The Diabetes Institute, Ohio University, Athens, OH 45701, USA.
| | - Claus Oxvig
- Department of Molecular Biology and Genetics, Faculty of Science and Technology, Aarhus University, Aarhus, Denmark.
| | - Mette Bjerre
- Medical Research Laboratory, Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark.
| | - Jan Frystyk
- Medical Research Laboratory, Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark; Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark.
| | - John J Kopchick
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA; The Diabetes Institute, Ohio University, Athens, OH 45701, USA.
| |
Collapse
|
22
|
Duran-Ortiz S, Noboa V, Kopchick JJ. Disruption of the GH receptor gene in adult mice and in insulin sensitive tissues. Growth Horm IGF Res 2018; 38:3-7. [PMID: 29198419 DOI: 10.1016/j.ghir.2017.11.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 11/13/2017] [Accepted: 11/23/2017] [Indexed: 01/31/2023]
Abstract
To elucidate whether a specific tissue is responsible for the beneficial health and longevity phenotype seen in growth hormone (GH) receptor (R) knockout (GHRKO) mice, the GHR gene was disrupted specifically in insulin sensitive tissues; namely, liver, adipose, and muscle. Furthermore, to investigate if the health- and life-span effects seen in the germline GHRKO mice were replicated when GH action was ablated after puberty; young, adult onset GHRKO mice were produced and characterized. In this review, we summarized the main findings derived from these mouse lines.
Collapse
Affiliation(s)
- Silvana Duran-Ortiz
- Edison Biotechnology Institute, United States; Department of Biological Sciences, College of Arts and Sciences, United States; Molecular and Cellular Biology Program, United States.
| | - Vanessa Noboa
- School of Medicine, Universidad San Francisco de Quito, Ecuador.
| | - John J Kopchick
- Edison Biotechnology Institute, United States; Molecular and Cellular Biology Program, United States; Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, 45701, United States.
| |
Collapse
|
23
|
|
24
|
Berryman DE, List EO. Growth Hormone's Effect on Adipose Tissue: Quality versus Quantity. Int J Mol Sci 2017; 18:ijms18081621. [PMID: 28933734 PMCID: PMC5578013 DOI: 10.3390/ijms18081621] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 07/10/2017] [Accepted: 07/17/2017] [Indexed: 02/07/2023] Open
Abstract
Obesity is an excessive accumulation or expansion of adipose tissue (AT) due to an increase in either the size and/or number of its characteristic cell type, the adipocyte. As one of the most significant public health problems of our time, obesity and its associated metabolic complications have demanded that attention be given to finding effective therapeutic options aimed at reducing adiposity or the metabolic dysfunction associated with its accumulation. Growth hormone (GH) has therapeutic potential due to its potent lipolytic effect and resultant ability to reduce AT mass while preserving lean body mass. However, AT and its resident adipocytes are significantly more dynamic and elaborate than once thought and require one not to use the reduction in absolute mass as a readout of efficacy alone. Paradoxically, therapies that reduce GH action may ultimately prove to be healthier, in part because GH also possesses potent anti-insulin activities along with concerns that GH may promote the growth of certain cancers. This review will briefly summarize some of the newer complexities of AT relevant to GH action and describe the current understanding of how GH influences this tissue using data from both humans and mice. We will conclude by considering the therapeutic use of GH or GH antagonists in obesity, as well as important gaps in knowledge regarding GH and AT.
Collapse
Affiliation(s)
- Darlene E Berryman
- The Diabetes Institute at Ohio University, 108 Konneker Research Labs, Ohio University, Athens, OH 45701, USA.
- Edison Biotechnology Institute, 218 Konneker Research Labs, Ohio University, Athens, OH 45701, USA.
| | - Edward O List
- The Diabetes Institute at Ohio University, 108 Konneker Research Labs, Ohio University, Athens, OH 45701, USA.
- Edison Biotechnology Institute, 218 Konneker Research Labs, Ohio University, Athens, OH 45701, USA.
| |
Collapse
|
25
|
Duran-Ortiz S, Brittain AL, Kopchick JJ. The impact of growth hormone on proteomic profiles: a review of mouse and adult human studies. Clin Proteomics 2017; 14:24. [PMID: 28670222 PMCID: PMC5492507 DOI: 10.1186/s12014-017-9160-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 06/20/2017] [Indexed: 12/17/2022] Open
Abstract
Growth hormone (GH) is a protein that is known to stimulate postnatal growth, counter regulate insulin's action and induce expression of insulin-like growth factor-1. GH exerts anabolic or catabolic effects depending upon on the targeted tissue. For instance, GH increases skeletal muscle and decreases adipose tissue mass. Our laboratory has spent the past two decades studying these effects, including the effects of GH excess and depletion, on the proteome of several mouse and human tissues. This review first discusses proteomic techniques that are commonly used for these types of studies. We then examine the proteomic differences found in mice with excess circulating GH (bGH mice) or mice with disruption of the GH receptor gene (GHR-/-). We also describe the effects of increased and decreased GH action on the proteome of adult patients with either acromegaly, GH deficiency or patients after short-term GH treatment. Finally, we explain how these proteomic studies resulted in the discovery of potential biomarkers for GH action, particularly those related with the effects of GH on aging, glucose metabolism and body composition.
Collapse
Affiliation(s)
- Silvana Duran-Ortiz
- Edison Biotechnology Institute, Ohio University, Athens, OH USA.,Department of Biological Sciences, College of Arts and Sciences, Ohio University, Athens, OH USA.,Molecular and Cellular Biology Program, Ohio University, Athens, OH USA
| | - Alison L Brittain
- Edison Biotechnology Institute, Ohio University, Athens, OH USA.,Department of Biological Sciences, College of Arts and Sciences, Ohio University, Athens, OH USA.,Molecular and Cellular Biology Program, Ohio University, Athens, OH USA.,Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701 USA
| | - John J Kopchick
- Edison Biotechnology Institute, Ohio University, Athens, OH USA.,Molecular and Cellular Biology Program, Ohio University, Athens, OH USA.,Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701 USA
| |
Collapse
|
26
|
Troike KM, Henry BE, Jensen EA, Young JA, List EO, Kopchick JJ, Berryman DE. Impact of Growth Hormone on Regulation of Adipose Tissue. Compr Physiol 2017. [PMID: 28640444 DOI: 10.1002/cphy.c160027] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Increasing prevalence of obesity and obesity-related conditions worldwide has necessitated a more thorough understanding of adipose tissue (AT) and expanded the scope of research in this field. AT is now understood to be far more complex and dynamic than previously thought, which has also fueled research to reevaluate how hormones, such as growth hormone (GH), alter the tissue. In this review, we will introduce properties of AT important for understanding how GH alters the tissue, such as anatomical location of depots and adipokine output. We will provide an overview of GH structure and function and define several human conditions and cognate mouse lines with extremes in GH action that have helped shape our understanding of GH and AT. A detailed discussion of the GH/AT relationship will be included that addresses adipokine production, immune cell populations, lipid metabolism, senescence, differentiation, and fibrosis, as well as brown AT and beiging of white AT. A brief overview of how GH levels are altered in an obese state, and the efficacy of GH as a therapeutic option to manage obesity will be given. As we will reveal, the effects of GH on AT are numerous, dynamic and depot-dependent. © 2017 American Physiological Society. Compr Physiol 7:819-840, 2017.
Collapse
Affiliation(s)
- Katie M Troike
- The Diabetes Institute at Ohio University, 108 Konneker Research Labs, Ohio University, Athens, Ohio, USA.,School of Applied Health Sciences and Wellness, College of Health Sciences and Professions, Ohio University, Athens, Ohio, USA
| | - Brooke E Henry
- The Diabetes Institute at Ohio University, 108 Konneker Research Labs, Ohio University, Athens, Ohio, USA.,School of Applied Health Sciences and Wellness, College of Health Sciences and Professions, Ohio University, Athens, Ohio, USA
| | - Elizabeth A Jensen
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA.,Edison Biotechnology Institute, Konneker Research Labs, Ohio University, Athens, Ohio, USA
| | - Jonathan A Young
- Department of Biological Sciences, College of Arts and Sciences, Ohio University, Athens, Ohio, USA.,Edison Biotechnology Institute, Konneker Research Labs, Ohio University, Athens, Ohio, USA
| | - Edward O List
- The Diabetes Institute at Ohio University, 108 Konneker Research Labs, Ohio University, Athens, Ohio, USA.,Edison Biotechnology Institute, Konneker Research Labs, Ohio University, Athens, Ohio, USA
| | - John J Kopchick
- The Diabetes Institute at Ohio University, 108 Konneker Research Labs, Ohio University, Athens, Ohio, USA.,Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA.,Edison Biotechnology Institute, Konneker Research Labs, Ohio University, Athens, Ohio, USA
| | - Darlene E Berryman
- The Diabetes Institute at Ohio University, 108 Konneker Research Labs, Ohio University, Athens, Ohio, USA.,Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA
| |
Collapse
|
27
|
Hjortebjerg R, Berryman DE, Comisford R, Frank SJ, List EO, Bjerre M, Frystyk J, Kopchick JJ. Insulin, IGF-1, and GH Receptors Are Altered in an Adipose Tissue Depot-Specific Manner in Male Mice With Modified GH Action. Endocrinology 2017; 158:1406-1418. [PMID: 28323915 PMCID: PMC5460824 DOI: 10.1210/en.2017-00084] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 02/22/2017] [Indexed: 12/28/2022]
Abstract
Growth hormone (GH) is a determinant of glucose homeostasis and adipose tissue (AT) function. Using 7-month-old transgenic mice expressing the bovine growth hormone (bGH) gene and growth hormone receptor knockout (GHR-/-) mice, we examined whether changes in GH action affect glucose, insulin, and pyruvate tolerance and AT expression of proteins involved in the interrelated signaling pathways of GH, insulinlike growth factor 1 (IGF-1), and insulin. Furthermore, we searched for AT depot-specific differences in control mice. Glycated hemoglobin levels were reduced in bGH and GHR-/- mice, and bGH mice displayed impaired gluconeogenesis as judged by pyruvate tolerance testing. Serum IGF-1 was elevated by 90% in bGH mice, whereas IGF-1 and insulin were reduced by 97% and 61% in GHR-/- mice, respectively. Igf1 RNA was increased in subcutaneous, epididymal, retroperitoneal, and brown adipose tissue (BAT) depots in bGH mice (mean increase ± standard error of the mean in all five depots, 153% ± 27%) and decreased in all depots in GHR-/- mice (mean decrease, 62% ± 4%). IGF-1 receptor expression was decreased in all AT depots of bGH mice (mean decrease, 49% ± 6%) and increased in all AT depots of GHR-/- mice (mean increase, 94% ± 8%). Insulin receptor expression was reduced in retroperitoneal, mesenteric, and BAT depots in bGH mice (mean decrease in all depots, 56% ± 4%) and augmented in subcutaneous, retroperitoneal, mesenteric, and BAT depots in GHR-/- mice (mean increase: 51% ± 1%). Collectively, our findings indicate a role for GH in influencing hormone signaling in AT in a depot-dependent manner.
Collapse
Affiliation(s)
- Rikke Hjortebjerg
- Medical Research Laboratory, Department of Clinical Medicine, Faculty of Health, Aarhus University, 8000 Aarhus, Denmark
- Danish Diabetes Academy, 5000 Odense, Denmark
- Edison Biotechnology Institute, Ohio University, Athens, Ohio 45701
| | - Darlene E. Berryman
- Edison Biotechnology Institute, Ohio University, Athens, Ohio 45701
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio 45701
- The Diabetes Institute at Ohio University, Ohio University, Athens, Ohio 45701
| | - Ross Comisford
- Edison Biotechnology Institute, Ohio University, Athens, Ohio 45701
- The Diabetes Institute at Ohio University, Ohio University, Athens, Ohio 45701
| | - Stuart J. Frank
- Division of Endocrinology, Diabetes and Metabolism, University of Alabama at Birmingham, Birmingham, Alabama 35924
- Medical Service, Birmingham Veterans Affairs Medical Center, Birmingham, Alabama 35233
| | - Edward O. List
- Edison Biotechnology Institute, Ohio University, Athens, Ohio 45701
| | - Mette Bjerre
- Medical Research Laboratory, Department of Clinical Medicine, Faculty of Health, Aarhus University, 8000 Aarhus, Denmark
| | - Jan Frystyk
- Medical Research Laboratory, Department of Clinical Medicine, Faculty of Health, Aarhus University, 8000 Aarhus, Denmark
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, 8000 Aarhus, Denmark
| | - John J. Kopchick
- Edison Biotechnology Institute, Ohio University, Athens, Ohio 45701
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio 45701
| |
Collapse
|
28
|
Darcy J, McFadden S, Bartke A. Altered structure and function of adipose tissue in long-lived mice with growth hormone-related mutations. Adipocyte 2017; 6:69-75. [PMID: 28425851 DOI: 10.1080/21623945.2017.1308990] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
A major focus of biogerontology is elucidating the role(s) of the endocrine system in aging and the accumulation of age-related diseases. Endocrine control of mammalian longevity was first reported in Ames dwarf (Prop1df) mice, which are long-lived due to a recessive Prop1 loss-of-function mutation resulting in deficiency of growth hormone (GH), thyroid-stimulating hormone, and prolactin. Following this report, several other GH-related mutants with altered longevity have been described including long-lived Snell dwarf and growth hormone receptor knockout mice, and short-lived GH overexpressing transgenic mice. One of the emerging areas of interest in these mutant mice is the role of adipose tissue in their altered healthspan and lifespan. Here, we provide an overview of the alterations in body composition of GH-related mutants, as well as the altered thermogenic potential of their brown adipose tissue and the altered cellular senescence and adipokine production of their white adipose tissue.
Collapse
Affiliation(s)
- Justin Darcy
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, Illinois, USA
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, Illinois, USA
| | - Samuel McFadden
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, Illinois, USA
| | - Andrzej Bartke
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, Illinois, USA
| |
Collapse
|
29
|
Jeong JY, Ibrahim M, Kim MJ, So K, Jeong YD, Park S, Kim M, Lee HJ. Comparisons of extracellular matrix-related gene expression levels in different adipose tissues from Korean cattle. Livest Sci 2017. [DOI: 10.1016/j.livsci.2017.02.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
30
|
Kaltenecker D, Mueller KM, Benedikt P, Feiler U, Themanns M, Schlederer M, Kenner L, Schweiger M, Haemmerle G, Moriggl R. Adipocyte STAT5 deficiency promotes adiposity and impairs lipid mobilisation in mice. Diabetologia 2017; 60:296-305. [PMID: 27858140 PMCID: PMC6518368 DOI: 10.1007/s00125-016-4152-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 10/12/2016] [Indexed: 01/04/2023]
Abstract
AIMS/HYPOTHESIS Dysfunction of lipid metabolism in white adipose tissue can substantially interfere with health and quality of life, for example in obesity and associated metabolic diseases. Therefore, it is important to characterise pathways that regulate lipid handling in adipocytes and determine how they affect metabolic homeostasis. Components of the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway are involved in adipocyte physiology and pathophysiology. However, the exact physiological importance of the STAT family member STAT5 in white adipose tissue is yet to be determined. Here, we aimed to delineate adipocyte STAT5 functions in the context of lipid metabolism in white adipose tissue. METHODS We generated an adipocyte specific knockout of Stat5 in mice using the Adipoq-Cre recombinase transgene followed by in vivo and in vitro biochemical and molecular studies. RESULTS Adipocyte-specific deletion of Stat5 resulted in increased adiposity, while insulin resistance and gluconeogenic capacity was decreased, indicating that glucose metabolism can be improved by interfering with adipose STAT5 function. Basal lipolysis and fasting-induced lipid mobilisation were diminished upon STAT5 deficiency, which coincided with reduced levels of the rate-limiting lipase of triacylglycerol hydrolysis, adipose triglyceride lipase (ATGL, encoded by Pnpla2) and its coactivator comparative gene identification 58 (CGI-58). In a mechanistic analysis, we identified a functional STAT5 response element within the Pnpla2 promoter, indicating that Pnpla2 is transcriptionally regulated by STAT5. CONCLUSIONS/INTERPRETATION Our findings reveal an essential role for STAT5 in maintaining lipid homeostasis in white adipose tissue and provide a rationale for future studies into the potential of STAT5 manipulation to improve outcomes in metabolic diseases.
Collapse
Affiliation(s)
- Doris Kaltenecker
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210, Vienna, Austria
| | - Kristina M Mueller
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210, Vienna, Austria
| | - Pia Benedikt
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Ursula Feiler
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Madeleine Themanns
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210, Vienna, Austria
| | - Michaela Schlederer
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| | - Lukas Kenner
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
- Unit of Pathology of Laboratory Animals, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Martina Schweiger
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Guenter Haemmerle
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Richard Moriggl
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria.
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210, Vienna, Austria.
- Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
31
|
Gude MF, Hjortebjerg R, Oxvig C, Thyø AA, Magnusson NE, Bjerre M, Pedersen SB, Frystyk J. PAPP-A, IGFBP-4 and IGF-II are secreted by human adipose tissue cultures in a depot-specific manner. Eur J Endocrinol 2016; 175:509-519. [PMID: 27585595 DOI: 10.1530/eje-16-0569] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 08/16/2016] [Accepted: 09/01/2016] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Adipose tissue secretes pregnancy-associated plasma protein-A (PAPP-A), which may increase local IGF action through cleavage of IGF-binding protein-4 (IGFBP-4). We tested whether this mechanism was operational in human visceral and subcutaneous adipose tissue (i.e. VAT and SAT). DESIGN Explants of VAT and SAT from 26 obese subjects (hereof 17 women, BMI 39.5 (37.2; 42.8) kg/m2 (median (25%; 75% confidence interval) and SAT from eight lean, age-matched women (BMI 23.6 (22.4; 24.9) kg/m2) were incubated with or without GH (100 µg/L) and the media were harvested. METHODS Media were assessed for concentrations of PAPP-A, intact and PAPP-A-cleaved IGFBP-4, IGF-I and IGF-II, and IGF-I receptor (IGF-IR) activation by bioassay. RESULTS In obese subjects, VAT media contained higher concentrations than SAT of PAPP-A (4.4-fold) and both PAPP-A-generated IGFBP-4 fragments (C-terminal: 3.3-fold, N-terminal: 1.5-fold) (all P < 0.0005). Intact IGFBP-4 levels were similar in SAT and VAT. VAT media contained elevated IGF-II (1.4-fold; P < 0.005), but similar IGF-I concentrations compared with SAT. Still, VAT media contained a 1.8-fold increased ability to stimulate the IGF-IR (P < 0.005). IGF-I protein concentration and IGF-IR activation increased more in VAT media than SAT media following GH stimulation (both P < 0.05). At baseline, SAT media protein levels from lean and obese women were similar, with the exception of PAPP-A being 1.8-fold elevated in VAT media (P < 0.05). GH induced a similar increase in IGF-I media levels in SAT from obese and lean women. CONCLUSION Human adipose tissue cultures secrete enzymatically active PAPP-A, IGFBP-4 and IGF-II in a depot-specific manner, suggesting differential regulation of IGF activity. Further, IGF-II appears to be more prominent than IGF-I. Finally, VAT appears more GH responsive than SAT.
Collapse
Affiliation(s)
- Mette Faurholdt Gude
- Medical Research LaboratoryDepartment of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus C, Denmark
| | - Rikke Hjortebjerg
- Medical Research LaboratoryDepartment of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus C, Denmark
- Danish Diabetes AcademyOdense, Denmark
| | - Claus Oxvig
- Department of Molecular Biology and GeneticsFaculty of Science and Technology, Aarhus University, Aarhus C, Denmark
| | | | - Nils Erik Magnusson
- Medical Research LaboratoryDepartment of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus C, Denmark
| | - Mette Bjerre
- Medical Research LaboratoryDepartment of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus C, Denmark
| | - Steen Bønløkke Pedersen
- Medical Research LaboratoryDepartment of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus C, Denmark
- Endocrinology and Internal MedicineAarhus University Hospital, Aarhus C, Denmark
| | - Jan Frystyk
- Medical Research LaboratoryDepartment of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus C, Denmark
- Endocrinology and Internal MedicineAarhus University Hospital, Aarhus C, Denmark
| |
Collapse
|
32
|
Brooks NE, Hjortebjerg R, Henry BE, List EO, Kopchick JJ, Berryman DE. Fibroblast growth factor 21, fibroblast growth factor receptor 1, and β-Klotho expression in bovine growth hormone transgenic and growth hormone receptor knockout mice. Growth Horm IGF Res 2016; 30-31:22-30. [PMID: 27585733 DOI: 10.1016/j.ghir.2016.08.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 08/19/2016] [Accepted: 08/22/2016] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Although growth hormone (GH) and fibroblast growth factor 21 (FGF21) have a reported relationship, FGF21 and its receptor, fibroblast growth factor receptor 1 (FGFR1) and cofactor β-Klotho (KLB), have not been analyzed in chronic states of altered GH action. The objective of this study was to quantify circulating FGF21 and tissue specific expression of Fgf21, Fgfr1, and Klb in mice with modified GH action. Based on previous studies, we hypothesized that bovine GH transgenic (bGH) mice will be FGF21 resistant and GH receptor knockout (GHR-/-) mice will have normal FGF21 action. DESIGN Seven-month-old male bGH mice (n=9) and wild type (WT) controls (n=10), and GHR-/- mice (n=8) and WT controls (n=8) were used for all measurements. Body composition was determined before dissection, and tissue weights were measured at the time of dissection. Serum FGF21 levels were evaluated by ELISA. Expression of Fgf21, Fgfr1, and Klb mRNA in white adipose tissue (AT), brown AT, and liver were evaluated by reverse transcription quantitative PCR. RESULTS As expected, bGH mice had increased body weight (p=3.70E-8) but decreased percent fat mass (p=4.87E-4). Likewise, GHR-/- mice had decreased body weight (p=1.78E-10) but increased percent fat mass (p=1.52E-9), due to increased size of the subcutaneous AT depot when normalized to body weight (p=1.60E-10). Serum FGF21 levels were significantly elevated in bGH mice (p=0.041) and unchanged in GHR-/- mice (p=0.88). Expression of Fgf21, Fgfr1, and Klb mRNA in white AT and liver were downregulated or unchanged in both bGH and GHR-/- mice. The only exception was Fgf21 expression in brown AT of GHR-/-, which trended toward increased expression (p=0.075). CONCLUSIONS In accordance with our hypothesis, we provide evidence that circulating FGF21 is increased in bGH animals, but remains unchanged in GHR-/- mice. Downregulation or no change in Fgf21, Fgfr1, and Klb expression are seen in white AT, brown AT, and liver of bGH and GHR-/- mice when compared to their respective controls, except for an increase in brown AT Fgf21 expression in GHR-/- mice, which could suggest a possible link to increased thermogenic potential in these mice. Overall, these results suggest possible modulation of FGF21 by GH resulting in FGF21 resistance or changes in FGF21 levels due to GH induced changes in liver size or kidney function.
Collapse
Affiliation(s)
- Nicole E Brooks
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; Honors Tutorial College, Ohio University, Athens, OH 45701, USA
| | - Rikke Hjortebjerg
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; Danish Diabetes Academy, Odense, Denmark
| | - Brooke E Henry
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; School of Applied Health Sciences and Wellness, College of Health Sciences and Professions, Ohio University, Athens, OH 45701, USA; The Diabetes Institute at Ohio University, Ohio University, Athens, OH 45701, USA
| | - Edward O List
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA
| | - John J Kopchick
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Darlene E Berryman
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA; School of Applied Health Sciences and Wellness, College of Health Sciences and Professions, Ohio University, Athens, OH 45701, USA; The Diabetes Institute at Ohio University, Ohio University, Athens, OH 45701, USA.
| |
Collapse
|
33
|
Chusyd DE, Wang D, Huffman DM, Nagy TR. Relationships between Rodent White Adipose Fat Pads and Human White Adipose Fat Depots. Front Nutr 2016; 3:10. [PMID: 27148535 PMCID: PMC4835715 DOI: 10.3389/fnut.2016.00010] [Citation(s) in RCA: 222] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 03/26/2016] [Indexed: 01/09/2023] Open
Abstract
The objective of this review was to compare and contrast the physiological and metabolic profiles of rodent white adipose fat pads with white adipose fat depots in humans. Human fat distribution and its metabolic consequences have received extensive attention, but much of what has been tested in translational research has relied heavily on rodents. Unfortunately, the validity of using rodent fat pads as a model of human adiposity has received less attention. There is a surprisingly lack of studies demonstrating an analogous relationship between rodent and human adiposity on obesity-related comorbidities. Therefore, we aimed to compare known similarities and disparities in terms of white adipose tissue (WAT) development and distribution, sexual dimorphism, weight loss, adipokine secretion, and aging. While the literature supports the notion that many similarities exist between rodents and humans, notable differences emerge related to fat deposition and function of WAT. Thus, further research is warranted to more carefully define the strengths and limitations of rodent WAT as a model for humans, with a particular emphasis on comparable fat depots, such as mesenteric fat.
Collapse
Affiliation(s)
- Daniella E Chusyd
- Department of Nutrition Science, University of Alabama at Birmingham , Birmingham, AL , USA
| | - Donghai Wang
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA; Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Derek M Huffman
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA; Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Tim R Nagy
- Department of Nutrition Science, University of Alabama at Birmingham , Birmingham, AL , USA
| |
Collapse
|
34
|
Olarescu NC, Bollerslev J. The Impact of Adipose Tissue on Insulin Resistance in Acromegaly. Trends Endocrinol Metab 2016; 27:226-237. [PMID: 26948712 DOI: 10.1016/j.tem.2016.02.005] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Revised: 02/09/2016] [Accepted: 02/11/2016] [Indexed: 01/11/2023]
Abstract
Adipose tissue (AT) is recognized as key contributor to the systemic insulin resistance and overt diabetes seen in metabolic syndrome. Acromegaly is a disease characterized by excessive secretion of growth hormone (GH) and insulin-like growth factor I (IGF-I). GH is known both for its action on AT and for its detrimental effect on glucose metabolism and insulin signaling. In active acromegaly, while body fat deports are diminished, insulin resistance is increased. Early studies have demonstrated defects in insulin action, both at the hepatic and extrahepatic (i.e., muscle and fat) levels, in active disease. This review discusses recent data suggesting that AT inflammation, altered AT distribution, and impaired adipogenesis are potential mechanisms contributing to systemic insulin resistance in acromegaly.
Collapse
Affiliation(s)
- Nicoleta Cristina Olarescu
- Section of Specialized Endocrinology, Department of Endocrinology, Oslo University Hospital, Rikshospitalet, Oslo, Norway; Faculty of Medicine, University of Oslo, Norway.
| | - Jens Bollerslev
- Section of Specialized Endocrinology, Department of Endocrinology, Oslo University Hospital, Rikshospitalet, Oslo, Norway; Faculty of Medicine, University of Oslo, Norway
| |
Collapse
|
35
|
Berryman DE, Henry B, Hjortebjerg R, List EO, Kopchick JJ. Developments in our understanding of the effects of growth hormone on white adipose tissue from mice: implications to the clinic. Expert Rev Endocrinol Metab 2016; 11:197-207. [PMID: 28435436 PMCID: PMC5397118 DOI: 10.1586/17446651.2016.1147950] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Adipose tissue (AT) is a well-established target of growth hormone (GH) and is altered in clinical conditions associated with excess, deficiency and absence of GH action. Due to the difficulty in collecting AT from clinical populations, genetically modified mice have been useful in better understanding how GH affects this tissue. Recent findings in mice would suggest that the impact of GH on AT is beyond alterations of lipolysis, lipogenesis or proliferation/ differentiation. AT depot-specific alterations in immune cells, extracellular matrix, adipokines, and senescence indicate an expanded role for GH in AT physiology. This mouse data will guide additional studies necessary to evaluate the therapeutic potential and safety of GH for conditions associated with altering AT, such as obesity. In this review, we introduce several relatively new intricacies of GH's effect on AT, focusing on recent studies in mice. Finally, we summarize the clinical implications of these findings.
Collapse
Affiliation(s)
- Darlene E Berryman
- Executive Director, The Diabetes Institute at Ohio University, 108 Konneker Research Labs, Ohio University, (740) 593-9661 - phone, (740) 593-4795 - fax
| | - Brooke Henry
- 108 Konneker Research Labs, Ohio University, (740) 593-9665
| | - Rikke Hjortebjerg
- Medical Research Laboratory, Department of Clinical Medicine, Aarhus University, Noerrebrogade 44, 8000 Aarhus C, Denmark, +45 6166 8045 - phone, +45 7846 2150 - fax
| | - Edward O List
- Senior Scientist, 218 Konneker Research Labs, Edison Biotechnology Institute, Ohio University, (740) 593-4620 - phone, (740) 593-4795 - fax
| | - John J Kopchick
- Distinguished Professor, Goll Ohio Eminent Scholar, 172 Water Tower Drive, Ohio University, (740) 593-4534 - phone, (740) 593-4795 - fax
| |
Collapse
|
36
|
Dal J, List EO, Jørgensen JOL, Berryman DE. Glucose and Fat Metabolism in Acromegaly: From Mice Models to Patient Care. Neuroendocrinology 2016; 103:96-105. [PMID: 25925240 DOI: 10.1159/000430819] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 04/20/2015] [Indexed: 11/19/2022]
Abstract
Patients with active acromegaly are frequently insulin resistant, glucose intolerant, and at risk for developing overt type 2 diabetes. At the same time, these patients have a relatively lean phenotype associated with mobilization and oxidation of free fatty acids. These features are reversed by curative surgical removal of the growth hormone (GH)-producing adenoma. Mouse models of acromegaly share many of these characteristics, including a lean phenotype and proneness to type 2 diabetes. There are, however, also species differences with respect to oxidation rates of glucose and fat as well as the specific mechanisms underlying GH-induced insulin resistance. The impact of acromegaly treatment on insulin sensitivity and glucose tolerance depends on the treatment modality (e.g. somatostatin analogs also suppress insulin secretion, whereas the GH antagonist restores insulin sensitivity). The interplay between animal research and clinical studies has proven useful in the field of acromegaly and should be continued in order to understand the metabolic actions of GH.
Collapse
Affiliation(s)
- Jakob Dal
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | | | | | | |
Collapse
|
37
|
Choi YH, Bae JK, Chae HS, Kim YM, Sreymom Y, Han L, Jang HY, Chin YW. α-Mangostin Regulates Hepatic Steatosis and Obesity through SirT1-AMPK and PPARγ Pathways in High-Fat Diet-Induced Obese Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2015; 63:8399-8406. [PMID: 26368128 DOI: 10.1021/acs.jafc.5b01637] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Previous studies have shown that α-mangostin (α-MG) suppresses intracellular fat accumulation and stimulation of lipolysis in in vitro systems. Together with the relatively high distribution of α-MG in liver and fat, these observations made it possible to propose a plausible hypothesis that an α-MG supplement may regulate hepatic steatosis and obesity. An α-MG supplement (50 mg/kg) reduced the body weight gain (13.8%) and epidymal and retroperitoneal fat mass accumulation (15.0 and 11.3%, respectively), as well as the biochemical serum profiles such as cholesterol [TC (26.9%), LDL-C (39.1%), and HDL-C (15.3%)], glucose (30.2%), triglyceride (29.7%), and fatty acid (30.3%) levels in high-fat fed mice compared with the high-fat diet-treated group, indicating that α-MG may regulate lipid metabolism. In addition, an α-MG supplement up-regulated hepatic AMPK, SirT1, and PPARγ levels compared with the high-fat diet states, suggesting that α-MG regulates hepatic steatosis and obesity through the SirT1-AMPK and PPARγ pathways in high-fat diet-induced obese mice.
Collapse
Affiliation(s)
- Young Hee Choi
- College of Pharmacy and BK21Plus R-Find Team, Dongguk University-Seoul , 32 Dongguk-lo, Ilsandong-gu, Goyang, Gyeonggi-do 410-820, South Korea
| | - Jin Kyung Bae
- College of Pharmacy and BK21Plus R-Find Team, Dongguk University-Seoul , 32 Dongguk-lo, Ilsandong-gu, Goyang, Gyeonggi-do 410-820, South Korea
| | - Hee-Sung Chae
- College of Pharmacy and BK21Plus R-Find Team, Dongguk University-Seoul , 32 Dongguk-lo, Ilsandong-gu, Goyang, Gyeonggi-do 410-820, South Korea
| | - Young-Mi Kim
- College of Pharmacy and BK21Plus R-Find Team, Dongguk University-Seoul , 32 Dongguk-lo, Ilsandong-gu, Goyang, Gyeonggi-do 410-820, South Korea
| | - Yim Sreymom
- College of Pharmacy and BK21Plus R-Find Team, Dongguk University-Seoul , 32 Dongguk-lo, Ilsandong-gu, Goyang, Gyeonggi-do 410-820, South Korea
| | - Ling Han
- College of Pharmacy and BK21Plus R-Find Team, Dongguk University-Seoul , 32 Dongguk-lo, Ilsandong-gu, Goyang, Gyeonggi-do 410-820, South Korea
| | - Ha Young Jang
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation , 80 Dongnae-ro, Dong-gu, Daegu 701-310, South Korea
| | - Young-Won Chin
- College of Pharmacy and BK21Plus R-Find Team, Dongguk University-Seoul , 32 Dongguk-lo, Ilsandong-gu, Goyang, Gyeonggi-do 410-820, South Korea
| |
Collapse
|
38
|
Comisford R, Lubbers ER, Householder LA, Suer O, Tchkonia T, Kirkland JL, List EO, Kopchick JJ, Berryman DE. Growth Hormone Receptor Antagonist Transgenic Mice Have Increased Subcutaneous Adipose Tissue Mass, Altered Glucose Homeostasis and No Change in White Adipose Tissue Cellular Senescence. Gerontology 2015; 62:163-72. [PMID: 26372907 DOI: 10.1159/000439050] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 07/29/2015] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Growth hormone (GH)-resistant/deficient mice experience improved glucose homeostasis and substantially increased lifespan. Recent evidence suggests that long-lived GH-resistant/deficient mice are protected from white adipose tissue (WAT) dysfunction, including WAT cellular senescence, impaired adipogenesis and loss of subcutaneous WAT in old age. This preservation of WAT function has been suggested to be a potential mechanism for the extended lifespan of these mice. OBJECTIVE The objective of this study was to examine WAT senescence, WAT distribution and glucose homeostasis in dwarf GH receptor antagonist (GHA) transgenic mice, a unique mouse strain having decreased GH action but normal longevity. METHODS 18-month-old female GHA mice and wild-type (WT) littermate controls were used. Prior to dissection, body composition, fasting blood glucose as well as glucose and insulin tolerance tests were performed. WAT distribution was determined by weighing four distinct WAT depots at the time of dissection. Cellular senescence in four WAT depots was assessed using senescence-associated β-galactosidase staining to quantify the senescent cell burden, and real-time qPCR to quantify gene expression of senescence markers p16 and IL-6. RESULTS GHA mice had a 22% reduction in total body weight, a 33% reduction in lean mass and a 10% increase in body fat percentage compared to WT controls. GHA mice had normal fasting blood glucose and improved insulin sensitivity; however, they exhibited impaired glucose tolerance. Moreover, GHA mice displayed enhanced lipid storage in the inguinal subcutaneous WAT depot (p < 0.05) and a 1.7-fold increase in extra-/intraperitoneal WAT ratio compared to controls (p < 0.05). Measurements of WAT cellular senescence showed no difference between GHA mice and WT controls. CONCLUSIONS Similar to other mice with decreased GH action, female GHA mice display reduced age-related lipid redistribution and improved insulin sensitivity, but no change in cellular senescence. The similar abundance of WAT senescent cells in GHA and control mice suggests that any protection against generation of senescent cells afforded by decreased GH action, low insulin-like growth factor 1 and/or improved insulin sensitivity in the GHA mice may be offset by their severe adiposity, since obesity is known to increase senescence.
Collapse
Affiliation(s)
- Ross Comisford
- Edison Biotechnology Institute, Ohio University, Athens, Ohio, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Murray PG, Higham CE, Clayton PE. 60 YEARS OF NEUROENDOCRINOLOGY: The hypothalamo-GH axis: the past 60 years. J Endocrinol 2015; 226:T123-40. [PMID: 26040485 DOI: 10.1530/joe-15-0120] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/03/2015] [Indexed: 12/19/2022]
Abstract
At the time of the publication of Geoffrey Harris's monograph on 'Neural control of the pituitary gland' 60 years ago, the pituitary was recognised to produce a growth factor, and extracts administered to children with hypopituitarism could accelerate growth. Since then our understanding of the neuroendocrinology of the GH axis has included identification of the key central components of the GH axis: GH-releasing hormone and somatostatin (SST) in the 1970s and 1980s and ghrelin in the 1990s. Characterisation of the physiological control of the axis was significantly advanced by frequent blood sampling studies in the 1980s and 1990s; the pulsatile pattern of GH secretion and the factors that influenced the frequency and amplitude of the pulses have been defined. Over the same time, spontaneously occurring and targeted mutations in the GH axis in rodents combined with the recognition of genetic causes of familial hypopituitarism demonstrated the key factors controlling pituitary development. As the understanding of the control of GH secretion advanced, developments of treatments for GH axis disorders have evolved. Administration of pituitary-derived human GH was followed by the introduction of recombinant human GH in the 1980s, and, more recently, by long-acting GH preparations. For GH excess disorders, dopamine agonists were used first followed by SST analogues, and in 2005 the GH receptor blocker pegvisomant was introduced. This review will cover the evolution of these discoveries and build a picture of our current understanding of the hypothalamo-GH axis.
Collapse
Affiliation(s)
- P G Murray
- Centre for Paediatrics and Child HealthInstitute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, M13 9WL, UKDepartment of Paediatric EndocrinologyRoyal Manchester Children's Hospital, Central Manchester Foundation Hospitals NHS Trust, Manchester Academic Health Science Centre, Manchester, M13 9WL, UKDepartment of EndocrinologyThe Christie Hospital NHS Foundation Trust, Manchester, M20 4BX, UKCentre for Endocrinology and DiabetesInstitute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, M13 9WL, UK Centre for Paediatrics and Child HealthInstitute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, M13 9WL, UKDepartment of Paediatric EndocrinologyRoyal Manchester Children's Hospital, Central Manchester Foundation Hospitals NHS Trust, Manchester Academic Health Science Centre, Manchester, M13 9WL, UKDepartment of EndocrinologyThe Christie Hospital NHS Foundation Trust, Manchester, M20 4BX, UKCentre for Endocrinology and DiabetesInstitute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, M13 9WL, UK
| | - C E Higham
- Centre for Paediatrics and Child HealthInstitute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, M13 9WL, UKDepartment of Paediatric EndocrinologyRoyal Manchester Children's Hospital, Central Manchester Foundation Hospitals NHS Trust, Manchester Academic Health Science Centre, Manchester, M13 9WL, UKDepartment of EndocrinologyThe Christie Hospital NHS Foundation Trust, Manchester, M20 4BX, UKCentre for Endocrinology and DiabetesInstitute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, M13 9WL, UK Centre for Paediatrics and Child HealthInstitute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, M13 9WL, UKDepartment of Paediatric EndocrinologyRoyal Manchester Children's Hospital, Central Manchester Foundation Hospitals NHS Trust, Manchester Academic Health Science Centre, Manchester, M13 9WL, UKDepartment of EndocrinologyThe Christie Hospital NHS Foundation Trust, Manchester, M20 4BX, UKCentre for Endocrinology and DiabetesInstitute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, M13 9WL, UK
| | - P E Clayton
- Centre for Paediatrics and Child HealthInstitute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, M13 9WL, UKDepartment of Paediatric EndocrinologyRoyal Manchester Children's Hospital, Central Manchester Foundation Hospitals NHS Trust, Manchester Academic Health Science Centre, Manchester, M13 9WL, UKDepartment of EndocrinologyThe Christie Hospital NHS Foundation Trust, Manchester, M20 4BX, UKCentre for Endocrinology and DiabetesInstitute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, M13 9WL, UK Centre for Paediatrics and Child HealthInstitute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, M13 9WL, UKDepartment of Paediatric EndocrinologyRoyal Manchester Children's Hospital, Central Manchester Foundation Hospitals NHS Trust, Manchester Academic Health Science Centre, Manchester, M13 9WL, UKDepartment of EndocrinologyThe Christie Hospital NHS Foundation Trust, Manchester, M20 4BX, UKCentre for Endocrinology and DiabetesInstitute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, M13 9WL, UK
| |
Collapse
|
40
|
Olarescu NC, Berryman DE, Householder LA, Lubbers ER, List EO, Benencia F, Kopchick JJ, Bollerslev J. GH action influences adipogenesis of mouse adipose tissue-derived mesenchymal stem cells. J Endocrinol 2015; 226:13-23. [PMID: 25943560 PMCID: PMC4560118 DOI: 10.1530/joe-15-0012] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/05/2015] [Indexed: 12/15/2022]
Abstract
GH influences adipocyte differentiation, but both stimulatory and inhibitory effects have been described. Adipose tissue-derived mesenchymal stem cells (AT-MSCs) are multipotent and are able to differentiate into adipocytes, among other cells. Canonical Wnt/β-catenin signaling activation impairs adipogenesis. The aim of the present study was to elucidate the role of GH on AT-MSC adipogenesis using cells isolated from male GH receptor knockout (GHRKO), bovine GH transgenic (bGH) mice, and wild-type littermate control (WT) mice. AT-MSCs from subcutaneous (sc), epididiymal (epi), and mesenteric (mes) AT depots were identified and isolated by flow cytometry (Pdgfrα+ Sca1+ Cd45- Ter119- cells). Their in vitro adipogenic differentiation capacity was determined by cell morphology and real-time RT-PCR. Using identical in vitro conditions, adipogenic differentiation of AT-MSCs was only achieved in the sc depot, and not in epi and mes depots. Notably, we observed an increased differentiation in cells isolated from sc-GHRKO and an impaired differentiation of sc-bGH cells as compared to sc-WT cells. Axin2, a marker of Wnt/β-catenin activation, was increased in mature sc-bGH adipocytes, which suggests that activation of this pathway may be responsible for the decreased adipogenesis. Thus, the present study demonstrates that (i) adipose tissue in mice has a well-defined population of Pdgfrα+ Sca1+ MSCs; (ii) the differentiation capacity of AT-MSCs varies from depot to depot regardless of GH genotype; (iii) the lack of GH action increases adipogenesis in the sc depot; and iv) activation of the Wnt/β-catenin pathway might mediate the GH effect on AT-MSCs. Taken together, the present results suggest that GH diminishes fat mass in part by altering adipogenesis of MSCs.
Collapse
Affiliation(s)
- Nicoleta C Olarescu
- Section of Specialized EndocrinologyDepartment of Endocrinology, Oslo University Hospital, Rikshospitalet, PO Box 4950, N-0424 Oslo, NorwayFaculty of MedicineUniversity of Oslo, Oslo, NorwayEdison Biotechnology InstituteOhio University, Athens, Ohio, USAHeritage College of Osteopathic MedicineOhio University, Athens, Ohio, USA Section of Specialized EndocrinologyDepartment of Endocrinology, Oslo University Hospital, Rikshospitalet, PO Box 4950, N-0424 Oslo, NorwayFaculty of MedicineUniversity of Oslo, Oslo, NorwayEdison Biotechnology InstituteOhio University, Athens, Ohio, USAHeritage College of Osteopathic MedicineOhio University, Athens, Ohio, USA
| | - Darlene E Berryman
- Section of Specialized EndocrinologyDepartment of Endocrinology, Oslo University Hospital, Rikshospitalet, PO Box 4950, N-0424 Oslo, NorwayFaculty of MedicineUniversity of Oslo, Oslo, NorwayEdison Biotechnology InstituteOhio University, Athens, Ohio, USAHeritage College of Osteopathic MedicineOhio University, Athens, Ohio, USA Section of Specialized EndocrinologyDepartment of Endocrinology, Oslo University Hospital, Rikshospitalet, PO Box 4950, N-0424 Oslo, NorwayFaculty of MedicineUniversity of Oslo, Oslo, NorwayEdison Biotechnology InstituteOhio University, Athens, Ohio, USAHeritage College of Osteopathic MedicineOhio University, Athens, Ohio, USA
| | - Lara A Householder
- Section of Specialized EndocrinologyDepartment of Endocrinology, Oslo University Hospital, Rikshospitalet, PO Box 4950, N-0424 Oslo, NorwayFaculty of MedicineUniversity of Oslo, Oslo, NorwayEdison Biotechnology InstituteOhio University, Athens, Ohio, USAHeritage College of Osteopathic MedicineOhio University, Athens, Ohio, USA Section of Specialized EndocrinologyDepartment of Endocrinology, Oslo University Hospital, Rikshospitalet, PO Box 4950, N-0424 Oslo, NorwayFaculty of MedicineUniversity of Oslo, Oslo, NorwayEdison Biotechnology InstituteOhio University, Athens, Ohio, USAHeritage College of Osteopathic MedicineOhio University, Athens, Ohio, USA
| | - Ellen R Lubbers
- Section of Specialized EndocrinologyDepartment of Endocrinology, Oslo University Hospital, Rikshospitalet, PO Box 4950, N-0424 Oslo, NorwayFaculty of MedicineUniversity of Oslo, Oslo, NorwayEdison Biotechnology InstituteOhio University, Athens, Ohio, USAHeritage College of Osteopathic MedicineOhio University, Athens, Ohio, USA
| | - Edward O List
- Section of Specialized EndocrinologyDepartment of Endocrinology, Oslo University Hospital, Rikshospitalet, PO Box 4950, N-0424 Oslo, NorwayFaculty of MedicineUniversity of Oslo, Oslo, NorwayEdison Biotechnology InstituteOhio University, Athens, Ohio, USAHeritage College of Osteopathic MedicineOhio University, Athens, Ohio, USA
| | - Fabian Benencia
- Section of Specialized EndocrinologyDepartment of Endocrinology, Oslo University Hospital, Rikshospitalet, PO Box 4950, N-0424 Oslo, NorwayFaculty of MedicineUniversity of Oslo, Oslo, NorwayEdison Biotechnology InstituteOhio University, Athens, Ohio, USAHeritage College of Osteopathic MedicineOhio University, Athens, Ohio, USA
| | - John J Kopchick
- Section of Specialized EndocrinologyDepartment of Endocrinology, Oslo University Hospital, Rikshospitalet, PO Box 4950, N-0424 Oslo, NorwayFaculty of MedicineUniversity of Oslo, Oslo, NorwayEdison Biotechnology InstituteOhio University, Athens, Ohio, USAHeritage College of Osteopathic MedicineOhio University, Athens, Ohio, USA Section of Specialized EndocrinologyDepartment of Endocrinology, Oslo University Hospital, Rikshospitalet, PO Box 4950, N-0424 Oslo, NorwayFaculty of MedicineUniversity of Oslo, Oslo, NorwayEdison Biotechnology InstituteOhio University, Athens, Ohio, USAHeritage College of Osteopathic MedicineOhio University, Athens, Ohio, USA
| | - Jens Bollerslev
- Section of Specialized EndocrinologyDepartment of Endocrinology, Oslo University Hospital, Rikshospitalet, PO Box 4950, N-0424 Oslo, NorwayFaculty of MedicineUniversity of Oslo, Oslo, NorwayEdison Biotechnology InstituteOhio University, Athens, Ohio, USAHeritage College of Osteopathic MedicineOhio University, Athens, Ohio, USA Section of Specialized EndocrinologyDepartment of Endocrinology, Oslo University Hospital, Rikshospitalet, PO Box 4950, N-0424 Oslo, NorwayFaculty of MedicineUniversity of Oslo, Oslo, NorwayEdison Biotechnology InstituteOhio University, Athens, Ohio, USAHeritage College of Osteopathic MedicineOhio University, Athens, Ohio, USA
| |
Collapse
|
41
|
Hill CM, Arum O, Boparai RK, Wang F, Fang Y, Sun LY, Masternak MM, Bartke A. Female PAPP-A knockout mice are resistant to metabolic dysfunction induced by high-fat/high-sucrose feeding at middle age. AGE (DORDRECHT, NETHERLANDS) 2015; 37:9765. [PMID: 25953669 PMCID: PMC4424199 DOI: 10.1007/s11357-015-9765-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 03/02/2015] [Indexed: 05/19/2023]
Abstract
Longevity and aging are influenced by common intracellular signals of the insulin/insulin-like growth factor (IGF)-1 pathway. Abnormally high levels of bioactive IGF-1 increase the development of various cancers and may contribute to metabolic diseases such as insulin resistance. Enhanced availability of IGF-1 is promoted by cleavage of IGF binding proteins (IGFBPs) by proteases, including the pregnancy-associated plasma protein-A (PAPPA). In vitro, PAPP-A is regulated by pro-inflammatory cytokines (PICs) such as interleukin (IL)-6 and tumor necrosis factor (TNF). Mice born with deficiency of the Papp-a gene (PAPP-A knockout (KO) mice) live ~30-40 % longer than their normal littermates and have decreased bioactive IGF-1 on standard diets. Our objective was to elucidate how the effects of high-fat, high-sucrose diet (HFHS) promote obesity, induce metabolic dysfunction, and alter systemic cytokine expression in PAPP-A KO and normal mice. PAPP-A KO mice fed HFHS diet for 10 weeks were more glucose tolerant and had enhanced insulin sensitivity compared to normal mice fed HFHS diet. PAPP-A KO mice fed HFHS diet had lower levels of pro-inflammatory cytokines (IL-2, IL-6, and TNF-α) compared to normal mice fed the same diet. However, anti-inflammatory cytokine levels (IL-4 and adiponectin) were higher in PAPP-A KO mice fed HFHS diet compared to normal mice fed HFHS. Circulating PAPP-A levels were elevated in normal mice fed an HFHS diet compared to normal mice fed a standard, low-fat, low-sucrose (LFLS) diet. Indirect calorimetry showed, at 10 weeks of feeding HFHS diet, significantly increased oxygen consumption (VO2) in PAPP-A KO mice fed HFHS diet compared to normal mice fed the same diet. Furthermore, respiratory quotient (RQ) was significantly lower in PAPP-A KO mice fed HFHS diet compared to normal (N) mice fed HFHS diet indicating PAPP-A KO mice fed HFHS diet are able to rely on fat as their primary source of energy more so than normal controls. We conclude that PAPP-A KO mice are resistant to the HFHS diet induction of metabolic dysfunction associated with higher levels of anti-inflammatory cytokines and a remarkably metabolic flexible phenotype and that some of the effects of HFHS diet in normal animals may be due to increased levels of PAPP-A.
Collapse
Affiliation(s)
- Cristal M. Hill
- />Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL USA
- />Geriatrics Research Laboratory, Department of Internal Medicine, Southern Illinois University School of Medicine, P.O. Box 19628, Springfield, IL 62794 USA
| | - Oge Arum
- />Geriatrics Research Laboratory, Department of Internal Medicine, Southern Illinois University School of Medicine, P.O. Box 19628, Springfield, IL 62794 USA
- />631 N. 6th St., Springfield, IL 62702 USA
| | - Ravneet K. Boparai
- />Geriatrics Research Laboratory, Department of Internal Medicine, Southern Illinois University School of Medicine, P.O. Box 19628, Springfield, IL 62794 USA
| | - Feiya Wang
- />Geriatrics Research Laboratory, Department of Internal Medicine, Southern Illinois University School of Medicine, P.O. Box 19628, Springfield, IL 62794 USA
| | - Yimin Fang
- />Geriatrics Research Laboratory, Department of Internal Medicine, Southern Illinois University School of Medicine, P.O. Box 19628, Springfield, IL 62794 USA
| | - Liou Y. Sun
- />Geriatrics Research Laboratory, Department of Internal Medicine, Southern Illinois University School of Medicine, P.O. Box 19628, Springfield, IL 62794 USA
| | - Michal M. Masternak
- />Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL USA
| | - Andrzej Bartke
- />Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL USA
- />Geriatrics Research Laboratory, Department of Internal Medicine, Southern Illinois University School of Medicine, P.O. Box 19628, Springfield, IL 62794 USA
| |
Collapse
|
42
|
Benencia F, Harshman S, Duran-Ortiz S, Lubbers ER, List EO, Householder L, Al-Naeeli M, Liang X, Welch L, Kopchick JJ, Berryman DE. Male bovine GH transgenic mice have decreased adiposity with an adipose depot-specific increase in immune cell populations. Endocrinology 2015; 156:1794-803. [PMID: 25521584 PMCID: PMC4398765 DOI: 10.1210/en.2014-1794] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
White adipose tissue (WAT) is composed of mature adipocytes and a stromal vascular fraction (SVF), which contains a variety of cells, including immune cells that vary among the different WAT depots. Growth hormone (GH) impacts immune function and adiposity in an adipose depot-specific manner. However, its effects on WAT immune cell populations remain unstudied. Bovine GH transgenic (bGH) mice are commonly used to study the in vivo effects of GH. These giant mice have an excess of GH action, impaired glucose metabolism, decreased adiposity, increased lean mass, and a shortened lifespan. Therefore, the purpose of this study was to characterize the WAT depot-specific differences in immune cell populations in the presence of excess GH in vivo. Three WAT depots were assessed: inguinal (sc), epididymal (EPI), and mesenteric (MES). Subcutaneous and MES bGH WAT depots showed a significantly higher number of total SVF cells, yet only MES bGH WAT had higher leukocyte counts compared with control samples. By means of flow cytometry analysis of the SVF, we detected greater macrophage and regulatory T-cell infiltration in sc and MES bGH WAT depots compared with controls. However, no differences were observed in the EPI WAT depot. RNA-sequencing confirmed significant alterations in pathways related to T-cell infiltration and activation in the sc depot with fewer significant changes in the EPI bGH WAT depot. These findings collectively point to a previously unrecognized role for GH in influencing the distribution of WAT immune cell populations in a depot-specific manner.
Collapse
Affiliation(s)
- Fabian Benencia
- Department of Biomedical Sciences (F.B., J.J.K., D.E.B.), Heritage College of Osteopathic Medicine; Russ College of Engineering and Technology (F.B.); Diabetes Institute (F.B., E.O.L., M.A.-N., J.J.K., D.E.B.); Edison Biotechnology Institute (S.H., S.D.-O., E.R.L., E.O.L., L.H., J.J.K., D.E.B.); School of Applied Health Sciences and Wellness (S.H., S.D.-O., D.E.B.), College of Health Sciences and Professions; Department of Biological Sciences (M.A.-N.), Ohio University Zanesville; School of Electrical Engineering and Computer Science (X.L., L.W.); and Biomedical Engineering Program (L.W.), Ohio University, Athens, Ohio 45701
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Yang T, Householder LA, Lubbers ER, List EO, Troike K, Vesel C, Duran-Ortiz S, Kopchick JJ, Berryman DE. Growth hormone receptor antagonist transgenic mice are protected from hyperinsulinemia and glucose intolerance despite obesity when placed on a HF diet. Endocrinology 2015; 156:555-64. [PMID: 25406017 PMCID: PMC4298328 DOI: 10.1210/en.2014-1617] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Reduced GH levels have been associated with improved glucose metabolism and increased longevity despite obesity in multiple mouse lines. However, one mouse line, the GH receptor antagonist (GHA) transgenic mouse, defies this trend because it has reduced GH action and increased adiposity, but glucose metabolism and life span are similar to controls. Slight differences in glucose metabolism and adiposity profiles can become exaggerated on a high-fat (HF) diet. Thus, in this study, male and female GHA and wild-type (WT) mice in a C57BL/6 background were placed on HF and low-fat (LF) diets for 11 weeks, starting at 10 weeks of age, to assess how GHA mice respond to additional metabolic stress of HF feeding. On a HF diet, all mice showed significant weight gain, although GHA gained weight more dramatically than WT mice, with males gaining more than females. Most of this weight gain was due to an increase in fat mass with WT mice increasing primarily in the white adipose tissue perigonadal depots, whereas GHA mice gained in both the sc and perigonadal white adipose tissue regions. Notably, GHA mice were somewhat protected from detrimental glucose metabolism changes on a HF diet because they had only modest increases in serum glucose levels, remained glucose tolerant, and did not develop hyperinsulinemia. Sex differences were observed in many measures with males reacting more dramatically to both a reduction in GH action and HF diet. In conclusion, our findings show that GHA mice, which are already obese, are susceptible to further adipose tissue expansion with HF feeding while remaining resilient to alterations in glucose homeostasis.
Collapse
Affiliation(s)
- Tianxu Yang
- Edison Biotechnology Institute (T.Y., L.A.H., E.R.L., E.O.L., K.T., C.V., S.D.-O., J.J.K., D.E.B.), School of Applied Health Sciences and Wellness, College of Health Sciences and Professions (T.Y., L.A.H., K.T., S.D.-O., D.E.B.), and Department of Biomedical Sciences (J.K., D.E.B.), Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio 45701
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Savastano S, Di Somma C, Barrea L, Colao A. The complex relationship between obesity and the somatropic axis: the long and winding road. Growth Horm IGF Res 2014; 24:221-226. [PMID: 25315226 DOI: 10.1016/j.ghir.2014.09.002] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Revised: 09/22/2014] [Accepted: 09/22/2014] [Indexed: 01/08/2023]
Abstract
Despite the considerable body of evidence pointing to a possible relationship between the state of the adipose tissue depots and regulation of the somatotropic axis, to date the relationship between obesity and low growth hormone (GH) status remains incompletely understood. The low GH status in obesity is mainly considered as a functional condition, largely reversible after a sustained weight loss. Moreover, due to the effects of the adiposity on the regulation of the somatotropic axis, the application of GH stimulation tests in obesity may also lead to an incorrect diagnosis of GH deficieny (GHD). On the other hand, similar to patients with GHD unrelated to obesity, the reduced GH response to stimulation testing in obese individuals is associated with increased prevalence of cardiovascular risk factors and detrimental alterations of body composition, which contribute to worsening their cardio-metabolic risk profile. In addition, the reduced GH secretion may result in reduced serum insulin-like growth factor (IGF)-1 levels, and the concordance of low peak GH and low IGF-1 identifies a subset of obese individuals with high cardiovascular risk. Furthermore, after weight loss, the normalization of the GH response and IGF-1 levels may or may not occur, and in patients undergoing bariatric surgery the persistence of a low GH status may affect the post-operative outcomes. In this review, we will provide an overview on some clinically relevant aspects of the relationship between obesity axis and the somatotropic axis in the light of the recently published research.
Collapse
Affiliation(s)
- Silvia Savastano
- Dipartimento di Medicina Clinica e Chirurgia, Unità di Endocrinologia, Università degli Studi di Napoli Federico II, Via S. Pansini, 5, Naples, Italy.
| | | | | | - Annamaria Colao
- Dipartimento di Medicina Clinica e Chirurgia, Unità di Endocrinologia, Università degli Studi di Napoli Federico II, Via S. Pansini, 5, Naples, Italy
| |
Collapse
|
45
|
Gomes-Santos E, Salvatori R, Ferrão TO, Oliveira CRP, Diniz RDCA, Santana JAM, Pereira FA, Barbosa RAA, Souza AHO, Melo EV, Epitácio-Pereira CC, Oliveira-Santos AA, Oliveira IAS, Machado JA, Santana-Júnior FJ, Barreto-Filho JAS, Aguiar-Oliveira MH. Increased visceral adiposity and cortisol to cortisone ratio in adults with congenital lifetime isolated GH deficiency. J Clin Endocrinol Metab 2014; 99:3285-9. [PMID: 24926956 DOI: 10.1210/jc.2014-2132] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT Adult-onset GH deficiency (GHD) increases visceral adiposity and the activity of the enzyme 11β-hydroxysteroid dehydrogenase, which converts cortisone (E) to cortisol (F), both linked to insulin resistance and increased cardiovascular risk. Conversely, we reported that adults with congenital isolated GHD (IGHD) have increased insulin sensitivity. OBJECTIVE To assess the type of fat distribution and the amount of visceral and sc fat and to correlate them to the F/E ratio in adults with untreated IGHD due to a mutation in the GHRH receptor gene. METHODS Body composition was assessed by dual-energy x-ray absorptiometry, thickness of sc and visceral fat was measured by sonography, and serum F and E were measured in 23 IGHD subjects and 21 age-matched controls. RESULTS Waist/hip ratio (WHR), trunk fat, and trunk/extremity fat (TR/EXT) ratio were higher in IGHD subjects. Visceral fat index (VFI) (but not sc fat index [SFI]) was higher in IGHD. F and F/E ratio were also higher in IGHD. In all 44 individuals, WHR correlated with TR/EXT ratio, thickness of visceral fat, VFI/SFI ratio, F, and F/E ratio. TR/EXT ratio correlated with visceral fat thickness, VFI/SFI ratio, and F. Age had a significant effect on VFI and on F/E ratio. Body mass index SD score and WHR have a similar significant effect on TR/EXT ratio and on F/E ratio. CONCLUSIONS Lifetime congenital untreated IGHD causes increased visceral adiposity with a high F/E ratio. However, the increased insulin sensitivity suggests that visceral adiposity needs a minimal GH secretion to translate into increased insulin resistance.
Collapse
Affiliation(s)
- Elenilde Gomes-Santos
- Federal University of Sergipe, Divisions of Endocrinology (E.G.-S., C.R.P.O., R.D.C.A.D., J.A.M.S., F.A.P., R.A.A.B. A.H.O.S., E.V.M., C.C.E.-P., A.A.O.-S., I.A.S.O., J.A.M., F.J.S.-J., J.A.S.B.-F., M.H.A.-O.), and Radiology (T.O.F.), Aracaju, SE, Brazil 49060-100; and Division of Endocrinology, Diabetes, and Metabolism (R.S.), The Johns Hopkins University School of Medicine, Baltimore, Maryland 21287
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Tirabassi G, delli Muti N, Buldreghini E, Lenzi A, Balercia G. Central body fat changes in men affected by post-surgical hypogonadotropic hypogonadism undergoing testosterone replacement therapy are modulated by androgen receptor CAG polymorphism. Nutr Metab Cardiovasc Dis 2014; 24:908-913. [PMID: 24787905 DOI: 10.1016/j.numecd.2014.02.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 02/05/2014] [Accepted: 02/25/2014] [Indexed: 11/30/2022]
Abstract
BACKGROUND AND AIMS Little is known about the effect of androgen receptor (AR) gene CAG repeat polymorphism in conditioning body composition changes after testosterone replacement therapy (TRT). In this study, we aimed to clarify this aspect by focussing our attention on male post-surgical hypogonadotropic hypogonadism, a condition often associated with partial or total hypopituitarism. METHODS AND RESULTS Fourteen men affected by post-surgical hypogonadotropic hypogonadism and undergoing several replacement hormone therapies were evaluated before and after TRT. Dual-energy X-ray absorptiometry (DEXA)-derived body composition measurements, pituitary-dependent hormones and AR gene CAG repeat polymorphism were considered. While testosterone and insulin-like growth factor-1 (IGF-1) levels increased after TRT, cortisol concentration decreased. No anthropometric or body composition parameters varied significantly, except for abdominal fat decrease. The number of CAG triplets was positively and significantly correlated with this abdominal fat decrease, while the opposite occurred between the latter and Δ-testosterone. No correlation of IGF-1 or cortisol variation (Δ-) with Δ-abdominal fat was found. At multiple linear regression, after correction for Δ-testosterone, the positive association between CAG triplet number and abdominal fat change was confirmed. CONCLUSIONS In male post-surgical hypogonadotropic hypogonadism, shorter length of AR CAG repeat tract is independently associated with a more marked decrease of abdominal fat after TRT.
Collapse
Affiliation(s)
- G Tirabassi
- Division of Endocrinology, Department of Clinical and Molecular Sciences, Umberto I Hospital, Polytechnic University of Marche, Ancona, Italy
| | - N delli Muti
- Division of Endocrinology, Department of Clinical and Molecular Sciences, Umberto I Hospital, Polytechnic University of Marche, Ancona, Italy
| | - E Buldreghini
- Division of Endocrinology, Department of Clinical and Molecular Sciences, Umberto I Hospital, Polytechnic University of Marche, Ancona, Italy
| | - A Lenzi
- Andrology, Pathophysiology of Reproduction and Endocrine Diagnosis Unit, Policlinic Umberto I, University of Rome 'La Sapienza', Rome, Italy
| | - G Balercia
- Division of Endocrinology, Department of Clinical and Molecular Sciences, Umberto I Hospital, Polytechnic University of Marche, Ancona, Italy.
| |
Collapse
|
47
|
List EO, Berryman DE, Funk K, Jara A, Kelder B, Wang F, Stout MB, Zhi X, Sun L, White TA, LeBrasseur NK, Pirtskhalava T, Tchkonia T, Jensen EA, Zhang W, Masternak MM, Kirkland JL, Miller RA, Bartke A, Kopchick JJ. Liver-specific GH receptor gene-disrupted (LiGHRKO) mice have decreased endocrine IGF-I, increased local IGF-I, and altered body size, body composition, and adipokine profiles. Endocrinology 2014; 155:1793-805. [PMID: 24517230 PMCID: PMC3990850 DOI: 10.1210/en.2013-2086] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Accepted: 02/04/2014] [Indexed: 11/19/2022]
Abstract
GH is an important regulator of body growth and composition as well as numerous other metabolic processes. In particular, liver plays a key role in the GH/IGF-I axis, because the majority of circulating "endocrine" IGF-I results from GH-stimulated liver IGF-I production. To develop a better understanding of the role of liver in the overall function of GH, we generated a strain of mice with liver-specific GH receptor (GHR) gene knockout (LiGHRKO mice). LiGHRKO mice had a 90% decrease in circulating IGF-I levels, a 300% increase in circulating GH, and significant changes in IGF binding protein (IGFBP)-1, IGFBP-2, IGFBP-3, IGFBP-5, and IGFBP-7. LiGHRKO mice were smaller than controls, with body length and body weight being significantly decreased in both sexes. Analysis of body composition over time revealed a pattern similar to those found in GH transgenic mice; that is, LiGHRKO mice had a higher percentage of body fat at early ages followed by lower percentage of body fat in adulthood. Local IGF-I mRNA levels were significantly increased in skeletal muscle and select adipose tissue depots. Grip strength was increased in LiGHRKO mice. Finally, circulating levels of leptin, resistin, and adiponectin were increased in LiGHRKO mice. In conclusion, LiGHRKO mice are smaller despite increased local mRNA expression of IGF-I in several tissues, suggesting that liver-derived IGF-I is indeed important for normal body growth. Furthermore, our data suggest that novel GH-dependent cross talk between liver and adipose is important for regulation of adipokines in vivo.
Collapse
Affiliation(s)
- Edward O List
- Edison Biotechnology Institute (E.O.L., D.E.B., K.F., A.J., B.K., E.A.J., W.Z., J.J.K.), Department of Specialty Medicine, Heritage College of Osteopathic Medicine (E.O.L.), School of Applied Health Sciences and Wellness (D.E.B.), Department of Biomedical Sciences, Heritage College of Osteopathic Medicine (D.E.B., A.J., J.J.K.), Ohio University, Athens, Ohio 45701-2942; Department of Internal Medicine (F.W., A.B.), Geriatrics Research, Southern Illinois University School of Medicine, Springfield, Illinois 62794-9628; Robert and Arlene Kogod Center on Aging (M.B.S., T.A.W., N.K.L., T.P., T.T., J.L.K.), Mayo Clinic, Rochester, Minnesota 55905-0002; College of Medicine, Burnett School of Biomedical Sciences (X.Z., M.M.M.), University of Central Florida, Orlando, Florida 32827-7406; and Department of Pathology and Geriatrics Center (L.S., R.A.M.), University of Michigan, Ann Arbor, Michigan 48109-2200
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Contrasting cellularity on fat deposition in the subcutaneous adipose tissue and longissimus lumborum muscle from lean and fat pigs under dietary protein reduction. Animal 2014; 8:629-37. [DOI: 10.1017/s1751731114000160] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
49
|
Sackmann-Sala L, Berryman DE, Lubbers ER, Zhang H, Vesel CB, Troike KM, Gosney ES, List EO, Kopchick JJ. Age-related and depot-specific changes in white adipose tissue of growth hormone receptor-null mice. J Gerontol A Biol Sci Med Sci 2013; 69:34-43. [PMID: 23873966 DOI: 10.1093/gerona/glt110] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Growth hormone receptor-null (GHR(-/-)) mice are dwarf, insulin sensitive, and long-lived in spite of increased adiposity. However, their adiposity is not uniform, with select white adipose tissue (WAT) depots enlarged. To study WAT depot-specific effects on insulin sensitivity and life span, we analyzed individual WAT depots of 12- and 24-month-old GHR(-) (/-) and wild-type (WT) mice, as well as their plasma levels of selected hormones. Adipocyte sizes and plasma insulin, leptin, and adiponectin levels decreased with age in both GHR(-) (/-) and WT mice. Two-dimensional gel electrophoresis proteomes of WAT depots were similar among groups, but several proteins involved in endocytosis and/or cytoskeletal organization (Ehd2, S100A10, actin), anticoagulation (S100A10, annexin A5), and age-related conditions (alpha2-macroglobulin, apolipoprotein A-I, transthyretin) showed significant differences between genotypes. Because Ehd2 may regulate endocytosis of Glut4, we measured Glut4 levels in the WAT depots of GHR(-) (/-) and WT mice. Inguinal WAT of 12-month-old GHR(-) (/-) mice displayed lower levels of Glut4 than WT. Overall, the protein changes detected in this study offer new insights into possible mechanisms contributing to enhanced insulin sensitivity and extended life span in GHR(-) (/-) mice.
Collapse
Affiliation(s)
- Lucila Sackmann-Sala
- Edison Biotechnology Institute, Ohio University, 1 Water Tower Dr., The Ridges, Athens, OH 45701.
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Berryman DE, Glad CAM, List EO, Johannsson G. The GH/IGF-1 axis in obesity: pathophysiology and therapeutic considerations. Nat Rev Endocrinol 2013; 9:346-56. [PMID: 23568441 DOI: 10.1038/nrendo.2013.64] [Citation(s) in RCA: 155] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Obesity has become one of the most common medical problems in developed countries, and this disorder is associated with high incidences of hypertension, dyslipidaemia, cardiovascular disease, type 2 diabetes mellitus and specific cancers. Growth hormone (GH) stimulates the production of insulin-like growth factor 1 in most tissues, and together GH and insulin-like growth factor 1 exert powerful collective actions on fat, protein and glucose metabolism. Clinical trials assessing the effects of GH treatment in patients with obesity have shown consistent reductions in total adipose tissue mass, in particular abdominal and visceral adipose tissue depots. Moreover, studies in patients with abdominal obesity demonstrate a marked effect of GH therapy on body composition and on lipid and glucose homeostasis. Therefore, administration of recombinant human GH or activation of endogenous GH production has great potential to influence the onset and metabolic consequences of obesity. However, the clinical use of GH is not without controversy, given conflicting results regarding its effects on glucose metabolism. This Review provides an introduction to the role of GH in obesity and summarizes clinical and preclinical data that describe how GH can influence the obese state.
Collapse
Affiliation(s)
- Darlene E Berryman
- Edison Biotechnology Institute, Ohio University, 1 Water Tower Drive, The Ridges, Athens, OH 45701, USA
| | | | | | | |
Collapse
|