1
|
Mohamed I, Gautam M, Abosheaishaa H, Hussain S, Kumar K, Kotak A, Baugh M, Qureshi R, Jaber F, Dahiya DS, Alba L, Duong N. Growth hormone augmentation in metabolic dysfunction-associated steatotic liver disease: a systematic review and meta-analysis of randomized controlled trials. Eur J Gastroenterol Hepatol 2024; 36:1259-1266. [PMID: 38973533 DOI: 10.1097/meg.0000000000002819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/09/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is characterized by hepatic steatosis and metabolic dysregulation. Growth hormone (GH) augmentation has emerged as a potential therapeutic intervention for treating MASLD. This systematic review and meta-analysis aimed to evaluate the impact of GH augmentation on different parameters of MASLD. A systematic literature search identified randomized controlled trials investigating GH augmentation in MASLD patients. Search results were screened via Covidence and the Risk of Bias 2 tool was used to assess bias in randomized controlled trials. Statistical analysis utilized RevMan v5.3. We combined dichotomous outcomes employing odds ratios and continuous outcomes utilizing mean difference (MD), each with a 95% confidence interval (CI). Statistical significance was indicated by a P -value less than 0.05. Heterogeneity was evaluated using I2 tests. Our results showed that GH augmentation resulted in a significant reduction in both relative (MD: -46.26; 95% CI: -71.52, -21.00; P = 0.0003) and absolute (MD: -5.15; 95% CI: -7.93, -2.37; P = 0.0003) hepatic fat fraction. GH augmentation significantly reduced alanine aminotransferase (MD: -5.97; 95% CI: -10.31, -1.62; P = 0.007) and gamma-glutamyl transferase (MD: -16.18; 95% CI: -30.76, -1.59; P = 0.03) levels. No significant changes were observed in hemoglobin A1c, C-reactive protein, fasting serum glucose, BMI, triglycerides, and low-density lipoprotein cholesterol levels. Our meta-analysis highlights GH augmentation as a promising therapy for reducing liver steatosis and improving liver enzyme levels in MASLD patients. Further large-scale trials are warranted to examine the long-term effects, safety profiles, and potential impact on various measures.
Collapse
Affiliation(s)
- Islam Mohamed
- Department of Internal Medicine, University of Missouri-Kansas City, Kansas City, Missouri
| | - Misha Gautam
- Department of Internal Medicine, University of Missouri-Kansas City, Kansas City, Missouri
| | - Hazem Abosheaishaa
- Department of Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Sophia Hussain
- School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri
| | - Kopal Kumar
- School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri
| | - Anaya Kotak
- School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri
| | - Macy Baugh
- School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri
| | - Raabia Qureshi
- School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri
| | - Fouad Jaber
- Department of Internal Medicine, University of Missouri-Kansas City, Kansas City, Missouri
| | | | - Laura Alba
- Department of Gastroenterology and Hepatology, University of Missouri-Kansas City, Kansas City, Missouri
| | - Nikki Duong
- Department of Gastroenterology and Hepatology, Stanford University, Stanford, California, USA
| |
Collapse
|
2
|
Kineman RD, Del Rio-Moreno M, Waxman DJ. Liver-specific actions of GH and IGF1 that protect against MASLD. Nat Rev Endocrinol 2024:10.1038/s41574-024-01037-0. [PMID: 39322791 DOI: 10.1038/s41574-024-01037-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/29/2024] [Indexed: 09/27/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD; also known as nonalcoholic fatty liver disease) is a chronic condition associated with metabolic syndrome, a group of conditions that includes obesity, insulin resistance, hyperlipidaemia and cardiovascular disease. Primary growth hormone (GH) deficiency is associated with MASLD, and the decline in circulating levels of GH with weight gain might contribute to the development of MASLD. Raising endogenous GH secretion or administering GH replacement therapy in the context of MASLD enhances insulin-like growth factor 1 (IGF1) production and reduces steatosis and the severity of liver injury. GH and IGF1 indirectly control MASLD progression by regulating systemic metabolic function. Evidence supports the proposal that GH and IGF1 also have a direct role in regulating liver metabolism and health. This Review focuses on how GH acts on the hepatocyte in a sex-dependent manner to limit lipid accumulation, reduce stress, and promote survival and regeneration. In addition, we discuss how GH and IGF1 might regulate non-parenchymal cells of the liver to control inflammation and fibrosis, which have a major effect on hepatocyte survival and regeneration. Development of a better understanding of how GH and IGF1 coordinate the functions of specific, individual liver cell types might provide insight into the aetiology of MASLD initiation and progression and suggest novel approaches for the treatment of MASLD.
Collapse
Affiliation(s)
- Rhonda D Kineman
- Department of Medicine, Section of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, IL, USA.
- Jesse Brown VA Medical Center, Research and Development Division, Chicago, IL, USA.
| | - Mercedes Del Rio-Moreno
- Department of Medicine, Section of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, IL, USA
- Jesse Brown VA Medical Center, Research and Development Division, Chicago, IL, USA
| | - David J Waxman
- Department of Biology and Bioinformatics Program, Boston University, Boston, MA, USA
| |
Collapse
|
3
|
Fang H, Li Q, Wang H, Ren Y, Zhang L, Yang L. Maternal nutrient metabolism in the liver during pregnancy. Front Endocrinol (Lausanne) 2024; 15:1295677. [PMID: 38572473 PMCID: PMC10987773 DOI: 10.3389/fendo.2024.1295677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 03/08/2024] [Indexed: 04/05/2024] Open
Abstract
The liver plays pivotal roles in nutrient metabolism, and correct hepatic adaptations are required in maternal nutrient metabolism during pregnancy. In this review, hepatic nutrient metabolism, including glucose metabolism, lipid and cholesterol metabolism, and protein and amino acid metabolism, is first addressed. In addition, recent progress on maternal hepatic adaptations in nutrient metabolism during pregnancy is discussed. Finally, the factors that regulate hepatic nutrient metabolism during pregnancy are highlighted, and the factors include follicle-stimulating hormone, estrogen, progesterone, insulin-like growth factor 1, prostaglandins fibroblast growth factor 21, serotonin, growth hormone, adrenocorticotropic hormone, prolactin, thyroid stimulating hormone, melatonin, adrenal hormone, leptin, glucagon-like peptide-1, insulin glucagon and thyroid hormone. Our vision is that more attention should be paid to liver nutrient metabolism during pregnancy, which will be helpful for utilizing nutrient appropriately and efficiently, and avoiding liver diseases during pregnancy.
Collapse
Affiliation(s)
- Hongxu Fang
- School of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, China
| | - Qingyang Li
- College of Life Sciences, Hebei Normal University, Shijiazhuang, China
| | - Haichao Wang
- School of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, China
| | - Ying Ren
- School of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, China
| | - Leying Zhang
- School of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, China
| | - Ling Yang
- School of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, China
| |
Collapse
|
4
|
McCall KD, Walter D, Patton A, Thuma JR, Courreges MC, Palczewski G, Goetz DJ, Bergmeier S, Schwartz FL. Anti-Inflammatory and Therapeutic Effects of a Novel Small-Molecule Inhibitor of Inflammation in a Male C57BL/6J Mouse Model of Obesity-Induced NAFLD/MAFLD. J Inflamm Res 2023; 16:5339-5366. [PMID: 38026235 PMCID: PMC10658948 DOI: 10.2147/jir.s413565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 10/31/2023] [Indexed: 12/01/2023] Open
Abstract
Purpose Non-alcoholic fatty liver disease (NAFLD), recently renamed metabolic (dysfunction) associated fatty liver disease (MAFLD), is the most common chronic liver disease in the United States. Presently, there is an intense and ongoing effort to identify and develop novel therapeutics for this disease. In this study, we explored the anti-inflammatory activity of a new compound, termed IOI-214, and its therapeutic potential to ameliorate NAFLD/MAFLD in male C57BL/6J mice fed a high fat (HF) diet. Methods Murine macrophages and hepatocytes in culture were treated with lipopolysaccharide (LPS) ± IOI-214 or DMSO (vehicle), and RT-qPCR analyses of inflammatory cytokine gene expression were used to assess IOI-214's anti-inflammatory properties in vitro. Male C57BL/6J mice were also placed on a HF diet and treated once daily with IOI-214 or DMSO for 16 weeks. Tissues were collected and analyzed to determine the effects of IOI-214 on HF diet-induced NAFL D/MAFLD. Measurements such as weight, blood glucose, serum cholesterol, liver/serum triglyceride, insulin, and glucose tolerance tests, ELISAs, metabolomics, Western blots, histology, gut microbiome, and serum LPS binding protein analyses were conducted. Results IOI-214 inhibited LPS-induced inflammation in macrophages and hepatocytes in culture and abrogated HF diet-induced mesenteric fat accumulation, hepatic inflammation and steatosis/hepatocellular ballooning, as well as fasting hyperglycemia without affecting insulin resistance or fasting insulin, cholesterol or TG levels despite overall obesity in vivo in male C57BL/6J mice. IOI-214 also decreased systemic inflammation in vivo and improved gut microbiota dysbiosis and leaky gut. Conclusion Combined, these data indicate that IOI-214 works at multiple levels in parallel to inhibit the inflammation that drives HF diet-induced NAFLD/MAFLD, suggesting that it may have therapeutic potential for NAFLD/MAFLD.
Collapse
Affiliation(s)
- Kelly D McCall
- Molecular and Cellular Biology Program, Ohio University College of Arts & Sciences, Athens, OH, USA
- Department of Biological Sciences, Ohio University College of Arts & Sciences, Athens, OH, USA
- Department of Specialty Medicine, Ohio University Heritage College of Osteopathic Medicine, Athens, OH, USA
- Department of Biomedical Sciences, Ohio University Heritage College of Osteopathic Medicine, Athens, OH, USA
- Diabetes Institute, Ohio University Heritage College of Osteopathic Medicine, Athens, OH, USA
- Biomedical Engineering Program, Ohio University Russ College of Engineering and Technology, Athens, OH, USA
| | - Debra Walter
- Molecular and Cellular Biology Program, Ohio University College of Arts & Sciences, Athens, OH, USA
- Department of Biological Sciences, Ohio University College of Arts & Sciences, Athens, OH, USA
| | - Ashley Patton
- Molecular and Cellular Biology Program, Ohio University College of Arts & Sciences, Athens, OH, USA
- Department of Biological Sciences, Ohio University College of Arts & Sciences, Athens, OH, USA
| | - Jean R Thuma
- Department of Specialty Medicine, Ohio University Heritage College of Osteopathic Medicine, Athens, OH, USA
| | - Maria C Courreges
- Department of Specialty Medicine, Ohio University Heritage College of Osteopathic Medicine, Athens, OH, USA
| | | | - Douglas J Goetz
- Molecular and Cellular Biology Program, Ohio University College of Arts & Sciences, Athens, OH, USA
- Biomedical Engineering Program, Ohio University Russ College of Engineering and Technology, Athens, OH, USA
- Department of Chemical & Biomolecular Engineering, Ohio University Russ College of Engineering and Technology, Athens, OH, USA
| | - Stephen Bergmeier
- Molecular and Cellular Biology Program, Ohio University College of Arts & Sciences, Athens, OH, USA
- Biomedical Engineering Program, Ohio University Russ College of Engineering and Technology, Athens, OH, USA
- Department of Chemistry & Biochemistry, Ohio University College of Arts & Sciences, Athens, OH, USA
| | - Frank L Schwartz
- Department of Specialty Medicine, Ohio University Heritage College of Osteopathic Medicine, Athens, OH, USA
- Diabetes Institute, Ohio University Heritage College of Osteopathic Medicine, Athens, OH, USA
- Biomedical Engineering Program, Ohio University Russ College of Engineering and Technology, Athens, OH, USA
| |
Collapse
|
5
|
Vázquez-Borrego MC, Del Río-Moreno M, Pyatkov M, Sarmento-Cabral A, Mahmood M, Pelke N, Wnek M, Cordoba-Chacon J, Waxman DJ, Puchowicz MA, McGuinness OP, Kineman RD. Direct and systemic actions of growth hormone receptor (GHR)-signaling on hepatic glycolysis, de novo lipogenesis and insulin sensitivity, associated with steatosis. Metabolism 2023; 144:155589. [PMID: 37182789 PMCID: PMC10843389 DOI: 10.1016/j.metabol.2023.155589] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/28/2023] [Accepted: 05/08/2023] [Indexed: 05/16/2023]
Abstract
BACKGROUND Evidence is accumulating that growth hormone (GH) protects against the development of steatosis and progression of non-alcoholic fatty liver disease (NAFLD). GH may control steatosis indirectly by altering systemic insulin sensitivity and substrate delivery to the liver and/or by the direct actions of GH on hepatocyte function. APPROACH To better define the hepatocyte-specific role of GH receptor (GHR) signaling on regulating steatosis, we used a mouse model with adult-onset, hepatocyte-specific GHR knockdown (aHepGHRkd). To prevent the reduction in circulating insulin-like growth factor 1 (IGF1) and the subsequent increase in GH observed after aHepGHRkd, subsets of aHepGHRkd mice were treated with adeno-associated viral vectors (AAV) driving hepatocyte-specific expression of IGF1 or a constitutively active form of STAT5b (STAT5bCA). The impact of hepatocyte-specific modulation of GHR, IGF1 and STAT5b on carbohydrate and lipid metabolism was studied across multiple nutritional states and in the context of hyperinsulinemic:euglycemic clamps. RESULTS Chow-fed male aHepGHRkd mice developed steatosis associated with an increase in hepatic glucokinase (GCK) and ketohexokinase (KHK) expression and de novo lipogenesis (DNL) rate, in the post-absorptive state and in response to refeeding after an overnight fast. The aHepGHRkd-associated increase in hepatic KHK, but not GCK and steatosis, was dependent on hepatocyte expression of carbohydrate response element binding protein (ChREBP), in re-fed mice. Interestingly, under clamp conditions, aHepGHRkd also increased the rate of DNL and expression of GCK and KHK, but impaired insulin-mediated suppression of hepatic glucose production, without altering plasma NEFA levels. These effects were normalized with AAV-mediated hepatocyte expression of IGF1 or STAT5bCA. Comparison of the impact of AAV-mediated hepatocyte IGF1 versus STAT5bCA in aHepGHRkd mice across multiple nutritional states, indicated the restorative actions of IGF1 are indirect, by improving systemic insulin sensitivity, independent of changes in the liver transcriptome. In contrast, the actions of STAT5b are due to the combined effects of raising IGF1 and direct alterations in the hepatocyte gene program that may involve suppression of BCL6 and FOXO1 activity. However, the direct and IGF1-dependent actions of STAT5b cannot fully account for enhanced GCK activity and lipogenic gene expression observed after aHepGHRkd, suggesting other GHR-mediated signals are involved. CONCLUSION These studies demonstrate hepatocyte GHR-signaling controls hepatic glycolysis, DNL, steatosis and hepatic insulin sensitivity indirectly (via IGF1) and directly (via STAT5b). The relative contribution of these indirect and direct actions of GH on hepatocytes is modified by insulin and nutrient availability. These results improve our understanding of the physiologic actions of GH on regulating adult metabolism to protect against NAFLD progression.
Collapse
Affiliation(s)
- Mari C Vázquez-Borrego
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, IL, United States of America; Research and Development Division, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, United States of America
| | - Mercedes Del Río-Moreno
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, IL, United States of America; Research and Development Division, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, United States of America
| | - Maxim Pyatkov
- Department of Biology & Bioinformatics Program, Boston University, Boston, MA, United States of America
| | - André Sarmento-Cabral
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, IL, United States of America; Research and Development Division, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, United States of America
| | - Mariyah Mahmood
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, IL, United States of America; Research and Development Division, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, United States of America
| | - Natalie Pelke
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, IL, United States of America; Research and Development Division, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, United States of America
| | - Magdalena Wnek
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, IL, United States of America; Research and Development Division, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, United States of America
| | - Jose Cordoba-Chacon
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, IL, United States of America; Research and Development Division, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, United States of America
| | - David J Waxman
- Department of Biology & Bioinformatics Program, Boston University, Boston, MA, United States of America
| | - Michelle A Puchowicz
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Owen P McGuinness
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, United States of America
| | - Rhonda D Kineman
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, IL, United States of America; Research and Development Division, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, United States of America.
| |
Collapse
|
6
|
Zhao L, Jia D, Tan Z, Jiang H. Association of growth hormone deficiency with an increased number of preadipocytes in subcutaneous fat. Front Endocrinol (Lausanne) 2023; 14:1199589. [PMID: 37305046 PMCID: PMC10250704 DOI: 10.3389/fendo.2023.1199589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 05/10/2023] [Indexed: 06/13/2023] Open
Abstract
The inhibitory effect of growth hormone (GH) on adipose tissue growth is well known, but the underlying mechanism is not fully understood. In this study, we determined the possibility that GH inhibits adipose tissue growth by inhibiting adipogenesis, the process of formation of adipocytes from stem cells, in the lit/lit mice. The lit/lit mice are GH deficient because of a spontaneous mutation to the GH releasing hormone receptor (ghrhr) gene, and they have more subcutaneous fat despite being smaller than the lit/+ mice at the same age. We found that cells of the stromal vascular fraction (SVF) of subcutaneous fat from the lit/lit mice had greater adipogenic potential than those from the lit/+ mice, as evidenced by forming greater numbers of lipid droplets-containing adipocytes and having greater expression of adipocyte marker genes during induced adipocyte differentiation in culture. However, addition of GH to the culture did not reverse the superior adipogenic potential of subcutaneous SVF from the lit/lit mice. Through florescence-activated cell sorting and quantification of mRNAs of preadipocyte markers, including CD34, CD29, Sca-1, CD24, Pref-1, and PPARγ, we found that subcutaneous SVF from the lit/lit mice contained more preadipocytes than that from the lit/+ mice. These results support the notion that GH inhibits adipose tissue growth in mice at least in part by inhibiting adipogenesis. Furthermore, these results suggest that GH inhibits adipogenesis in mice not by inhibiting the terminal differentiation of preadipocytes into adipocytes, rather by inhibiting the formation of preadipocytes from stem cells or the recruitment of stem cells to the fat depot.
Collapse
|
7
|
Perone F, Pingitore A, Conte E, Halasz G, Ambrosetti M, Peruzzi M, Cavarretta E. Obesity and Cardiovascular Risk: Systematic Intervention Is the Key for Prevention. Healthcare (Basel) 2023; 11:healthcare11060902. [PMID: 36981559 PMCID: PMC10048800 DOI: 10.3390/healthcare11060902] [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: 01/29/2023] [Revised: 03/09/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
Obesity is a serious public health issue and associated with an increased risk of cardiovascular disease events and mortality. The risk of cardiovascular complications is directly related to excess body fat mass and ectopic fat deposition, but also other obesity-related complications such as pre-type 2 diabetes, obstructive sleep apnoea, and non-alcoholic fatty liver diseases. Body mass index and waist circumference are used to classify a patient as overweight or obese and to stratify cardiovascular risk. Physical activity and diet, despite being key points in preventing adverse events and reducing cardiovascular risk, are not always successful strategies. Pharmacological treatments for weight reduction are promising strategies, but are restricted by possible safety issues and cost. Nonetheless, these treatments are associated with improvements in cardiovascular risk factors, and studies are ongoing to better evaluate cardiovascular outcomes. Bariatric surgery is effective in reducing the incidence of death and cardiovascular events such as myocardial infarction and stroke. Cardiac rehabilitation programs in obese patients improve cardiovascular disease risk factors, quality of life, and exercise capacity. The aim of this review was to critically analyze the current role and future aspects of lifestyle changes, medical and surgical treatments, and cardiac rehabilitation in obese patients, to reduce cardiovascular disease risk and mortality, and to highlight the need for a multidisciplinary approach to improving cardiovascular outcomes.
Collapse
Affiliation(s)
- Francesco Perone
- Cardiac Rehabilitation Unit, Rehabilitation Clinic "Villa delle Magnolie", 81020 Castel Morrone, Caserta, Italy
| | - Annachiara Pingitore
- Department of General and Specialistic Surgery "Paride Stefanini", Sapienza University of Rome, 00161 Rome, Italy
| | - Edoardo Conte
- Department of Clinical Cardiology and Cardiovascular Imaging, Galeazzi-Sant'Ambrogio Hospital IRCCS, 20100 Milan, Lombardy, Italy
| | - Geza Halasz
- Cardiology Department, Azienda Ospedaliera San Camillo Forlanini, 00152 Rome, Italy
| | - Marco Ambrosetti
- Cardiovascular Rehabilitation Unit, ASST Crema Santa Marta Hospital, 26027 Rivolta D'Adda, Cremona, Italy
| | - Mariangela Peruzzi
- Department of Clinical Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, 00161 Rome, Italy
- Mediterranea Cardiocentro, 80122 Naples, Campania, Italy
| | - Elena Cavarretta
- Mediterranea Cardiocentro, 80122 Naples, Campania, Italy
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Latina, Italy
| |
Collapse
|
8
|
Swarbrick MM, Cox CL, Graham JL, Knudsen LB, Stanhope K, Raun K, Havel PJ. Growth hormone treatment does not augment the anti-diabetic effects of liraglutide in UCD-T2DM rats. Endocrinol Diabetes Metab 2022; 6:e392. [PMID: 36480511 PMCID: PMC9836246 DOI: 10.1002/edm2.392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 10/29/2022] [Accepted: 11/02/2022] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION The incretin hormone glucagon-like peptide-1 (GLP-1) slows gastric emptying, increases satiety and enhances insulin secretion. GLP-1 receptor agonists, such as liraglutide, are used therapeutically in humans to improve glycaemic control and delay the onset of type 2 diabetes mellitus (T2DM). In UCD-T2DM rats, a model of polygenic obesity and insulin resistance, we have previously reported that daily liraglutide administration delayed diabetes onset by >4 months. Growth hormone (GH) may exert anti-diabetic effects, including increasing β-cell mass and insulin secretion, while disrupting GH signalling in mice reduces both the size and number of pancreatic islets. We therefore hypothesized that GH supplementation would augment liraglutide's anti-diabetic effects. METHODS Male UCD-T2DM rats were treated daily with GH (0.3 mg/kg) and/or liraglutide (0.2 mg/kg) from 2 months of age. Control (vehicle) and food-restricted (with food intake matched to liraglutide-treated rats) groups were also studied. The effects of treatment on diabetes onset and weight gain were assessed, as well as measures of glucose tolerance, lipids and islet morphology. RESULTS Liraglutide treatment significantly reduced food intake and body weight and improved glucose tolerance and insulin sensitivity, relative to controls. After 4.5 months, none of the liraglutide-treated rats had developed T2DM (overall p = .019). Liraglutide-treated rats also displayed lower fasting triglyceride (TG) concentrations and lower hepatic TG content, compared to control rats. Islet morphology was improved in liraglutide-treated rats, with significantly increased pancreatic insulin content (p < .05 vs. controls). Although GH treatment tended to increase body weight (and gastrocnemius muscle weight), there were no obvious effects on diabetes onset or other diabetes-related outcomes. CONCLUSION GH supplementation did not augment the anti-diabetic effects of liraglutide.
Collapse
Affiliation(s)
- Michael M. Swarbrick
- Departments of Nutrition and Department of Molecular Biosciences, School of Veterinary MedicineUniversity of California, DavisOne Shielad AvenueDavisCaliforniaUSA,Present address:
Bone Research Program, ANZAC Research InstituteThe University of SydneyConcordNew South WalesAustralia,Present address:
Concord Clinical School, Faculty of Medicine and HealthThe University of SydneyAustralia
| | - Chad L. Cox
- Departments of Nutrition and Department of Molecular Biosciences, School of Veterinary MedicineUniversity of California, DavisOne Shielad AvenueDavisCaliforniaUSA
| | - James L. Graham
- Departments of Nutrition and Department of Molecular Biosciences, School of Veterinary MedicineUniversity of California, DavisOne Shielad AvenueDavisCaliforniaUSA
| | | | - Kimber Stanhope
- Departments of Nutrition and Department of Molecular Biosciences, School of Veterinary MedicineUniversity of California, DavisOne Shielad AvenueDavisCaliforniaUSA
| | | | - Peter J. Havel
- Departments of Nutrition and Department of Molecular Biosciences, School of Veterinary MedicineUniversity of California, DavisOne Shielad AvenueDavisCaliforniaUSA
| |
Collapse
|
9
|
Growth Hormone Improves Adipose Tissue Browning and Muscle Wasting in Mice with Chronic Kidney Disease-Associated Cachexia. Int J Mol Sci 2022; 23:ijms232315310. [PMID: 36499637 PMCID: PMC9740214 DOI: 10.3390/ijms232315310] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/30/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
Cachexia associated with chronic kidney disease (CKD) has been linked to GH resistance. In CKD, GH treatment enhances muscular performance. We investigated the impact of GH on cachexia brought on by CKD. CKD was induced by 5/6 nephrectomy in c57BL/6J mice. After receiving GH (10 mg/kg/day) or saline treatment for six weeks, CKD mice were compared to sham-operated controls. GH normalized metabolic rate, increased food intake and weight growth, and improved in vivo muscular function (rotarod and grip strength) in CKD mice. GH decreased uncoupling proteins (UCP)s and increased muscle and adipose tissue ATP content in CKD mice. GH decreased lipolysis of adipose tissue by attenuating expression and protein content of adipose triglyceride lipase and protein content of phosphorylated hormone-sensitive lipase in CKD mice. GH reversed the increased expression of beige adipocyte markers (UCP-1, CD137, Tmem26, Tbx1, Prdm16, Pgc1α, and Cidea) and molecules implicated in adipose tissue browning (Cox2/Pgf2α, Tlr2, Myd88, and Traf6) in CKD mice. Additionally, GH normalized the molecular markers of processes connected to muscle wasting in CKD, such as myogenesis and muscle regeneration. By using RNAseq, we previously determined the top 12 skeletal muscle genes differentially expressed between mice with CKD and control animals. These 12 genes' aberrant expression has been linked to increased muscle thermogenesis, fibrosis, and poor muscle and neuron regeneration. In this study, we demonstrated that GH restored 7 of the top 12 differentially elevated muscle genes in CKD mice. In conclusion, GH might be an effective treatment for muscular atrophy and browning of adipose tissue in CKD-related cachexia.
Collapse
|
10
|
Li T, Bai H, Fang H, Yang L, Yan P. Growth hormone inhibits adipogenic differentiation and induces browning in bovine subcutaneous adipocytes. Growth Horm IGF Res 2022; 66:101498. [PMID: 36007464 DOI: 10.1016/j.ghir.2022.101498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 08/12/2022] [Accepted: 08/15/2022] [Indexed: 11/17/2022]
Abstract
OBJECTIVE It is well established that growth hormone (GH) has the ability to stimulate lipolysis. The effects of GH on adipocyte differentiation and browning have not been clearly described. Therefore, the present study aimed to elucidate the role of GH in the differentiation and browning of bovine subcutaneous adipocytes as well as its underlying molecular mechanisms. METHODS We first treated bovine subcutaneous preadipocytes with different concentrations (0, 10, 100, and 500 ng/mL) of GH for 8 days and measured lipid accumulation and gene expression. Afterward, we treated preadipocytes and mature adipocytes with 500 ng/mL GH and determined differentiation and browning-related indicators. Finally, we investigated the expression of STAT5B in both preadipocytes and mature adipocytes after GH treatment. RESULTS We demonstrated that GH inhibited lipid accumulation and decreased the expression levels of adipogenic key genes (SCD1, SREBP1, PPARγ, and CEBPα) during adipocyte differentiation. Moreover, we observed that the inhibitory effect of GH on the early stage of adipocyte differentiation (0-2 days) was stronger than that on the later stage of adipocyte differentiation (2-8 days). We also found that GH promoted the expression levels of browning-related genes such as uncoupling protein 1 (UCP1) in mature adipocytes. Concurrently, GH promoted mitochondrial biogenesis and increased the expression levels of mitochondrial biogenesis-related genes. In addition, GH promoted phosphorylation of signal transducers and activator of transcription 5 b (STAT5B) and contributed to translocation of STAT5B to nucleus. After blocking the expression of STAT5B protein, GH weakened the inhibition of adipogenic key genes and reduced the promotion of browning-related genes in bovine subcutaneous adipocytes. CONCLUSIONS GH can inhibit adipocyte differentiation and promote adipocyte browning by regulating STAT5B in bovine subcutaneous adipocytes.
Collapse
Affiliation(s)
- Tingting Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Hui Bai
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Haoyuan Fang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Liang Yang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Peishi Yan
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
11
|
List EO, Berryman DE, Slyby J, Duran-Ortiz S, Funk K, Bisset ES, Howlett SE, Kopchick JJ. Disruption of Growth Hormone Receptor in Adipocytes Improves Insulin Sensitivity and Lifespan in Mice. Endocrinology 2022; 163:bqac129. [PMID: 35952979 PMCID: PMC9467438 DOI: 10.1210/endocr/bqac129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Indexed: 11/19/2022]
Abstract
Growth hormone receptor knockout (GHRKO) mice have been used for 25 years to uncover some of the many actions of growth hormone (GH). Since they are extremely long-lived with enhanced insulin sensitivity and protected from multiple age-related diseases, they are often used to study healthy aging. To determine the effect that adipose tissue has on the GHRKO phenotype, our laboratory recently created and characterized adipocyte-specific GHRKO (AdGHRKO) mice, which have increased adiposity but appear healthy with enhanced insulin sensitivity. To test the hypothesis that removal of GH action in adipocytes might partially replicate the increased lifespan and healthspan observed in global GHRKO mice, we assessed adiposity, cytokines/adipokines, glucose homeostasis, frailty, and lifespan in aging AdGHRKO mice of both sexes. Our results show that disrupting the GH receptor gene in adipocytes improved insulin sensitivity at advanced age and increased lifespan in male AdGHRKO mice. AdGHRKO mice also exhibited increased fat mass, reduced circulating levels of insulin, c-peptide, adiponectin, resistin, and improved frailty scores with increased grip strength at advanced ages. Comparison of published mean lifespan data from GHRKO mice to that from AdGHRKO and muscle-specific GHRKO mice suggests that approximately 23% of lifespan extension in male GHRKO is due to GHR disruption in adipocytes vs approximately 19% in muscle. Females benefited less from GHR disruption in these 2 tissues with approximately 19% and approximately 0%, respectively. These data indicate that removal of GH's action, even in a single tissue, is sufficient for observable health benefits that promote long-term health, reduce frailty, and increase longevity.
Collapse
Affiliation(s)
- Edward O List
- Edison Biotechnology Institute, Ohio University, Athens, Ohio 45701, USA
- Department of Specialty Medicine, Heritage College of Osteopathic Medicine, Athens, Ohio 45701, USA
| | - Darlene E Berryman
- Edison Biotechnology Institute, Ohio University, Athens, Ohio 45701, USA
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Athens, Ohio 45701, USA
| | - Julie Slyby
- Edison Biotechnology Institute, Ohio University, Athens, Ohio 45701, USA
| | | | - Kevin Funk
- Edison Biotechnology Institute, Ohio University, Athens, Ohio 45701, USA
| | - Elise S Bisset
- Department of Pharmacology Dalhousie University Halifax, Halifax , Nova Scotia , Canada
| | - Susan E Howlett
- Department of Pharmacology Dalhousie University Halifax, Halifax , Nova Scotia , Canada
- Department of Medicine (Geriatric Medicine), Dalhousie University Halifax, Halifax , Nova Scotia , Canada
| | - John J Kopchick
- Edison Biotechnology Institute, Ohio University, Athens, Ohio 45701, USA
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Athens, Ohio 45701, USA
| |
Collapse
|
12
|
Towards Understanding the Direct and Indirect Actions of Growth Hormone in Controlling Hepatocyte Carbohydrate and Lipid Metabolism. Cells 2021; 10:cells10102532. [PMID: 34685512 PMCID: PMC8533955 DOI: 10.3390/cells10102532] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 09/20/2021] [Accepted: 09/23/2021] [Indexed: 02/06/2023] Open
Abstract
Growth hormone (GH) is critical for achieving normal structural growth. In addition, GH plays an important role in regulating metabolic function. GH acts through its GH receptor (GHR) to modulate the production and function of insulin-like growth factor 1 (IGF1) and insulin. GH, IGF1, and insulin act on multiple tissues to coordinate metabolic control in a context-specific manner. This review will specifically focus on our current understanding of the direct and indirect actions of GH to control liver (hepatocyte) carbohydrate and lipid metabolism in the context of normal fasting (sleep) and feeding (wake) cycles and in response to prolonged nutrient deprivation and excess. Caveats and challenges related to the model systems used and areas that require further investigation towards a clearer understanding of the role GH plays in metabolic health and disease are discussed.
Collapse
|
13
|
Young JA, Henry BE, Benencia F, Bell S, List EO, Kopchick JJ, Berryman DE. GHR -/- Mice are protected from obesity-related white adipose tissue inflammation. J Neuroendocrinol 2020; 32:e12854. [PMID: 32350959 PMCID: PMC7554100 DOI: 10.1111/jne.12854] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 03/09/2020] [Accepted: 04/03/2020] [Indexed: 12/27/2022]
Abstract
Growth hormone (GH) excess in bovine (b)GH transgenic mice has been shown to alter white adipose tissue (WAT) immune cell populations. The present study aimed to evaluate the effects of GH resistance on WAT immune cell populations using GH receptor knockout (GHR-/- ) mice. Eight- and 24-month-old, male GHR-/- and wild-type mice were used. Body composition and tissue weights were determined, and systemic inflammation was assessed by measuring serum cytokine levels. The stromal vascular fraction (SVF) was isolated from three distinct WAT depots, and immune cell populations were quantified using flow cytometry. GHR-/- mice at both ages had decreased body weight but were obese. Although no significant changes were observed in serum levels of the measured cytokines, SVF cell alterations were seen and differed from depot to depot. Total SVF cells were decreased in epidydimal (Epi) depots, whereas SVF cells per gram adipose tissue weight were increased in mesenteric (Mes) depots of GHR-/- mice relative to controls. T cells and T helper cells were increased in Mes at 8 months old, whereas cytotoxic T cells were decreased in subcutaneous (SubQ) at 24 months old. Other cells were unchanged at both ages measured. The present study demonstrates that removal of GH action results in modest and depot-specific changes to several immune cell populations in WAT of intra-abdominal depots (Epi and Mes), which are somewhat surprising results because the SubQ has the largest change in size, whereas the Mes has no size change. Taken together with previous results from bovine GH transgenic mice, these data suggest that GH induces changes in the immune cell population of WAT in a depot-specific manner. Notably, GHR-/- mice appear to be protected from age-related WAT inflammation and immune cell infiltration despite obesity.
Collapse
Affiliation(s)
- Jonathan A. Young
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - 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
| | - Fabian Benencia
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Stephen Bell
- 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
- The Diabetes Institute at Ohio University, Ohio University, Athens, OH 45701, USA
- Corresponding Author at: Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA.
| |
Collapse
|
14
|
List EO, Berryman DE, Basu R, Buchman M, Funk K, Kulkarni P, Duran-Ortiz S, Qian Y, Jensen EA, Young JA, Yildirim G, Yakar S, Kopchick JJ. The Effects of 20-kDa Human Placental GH in Male and Female GH-deficient Mice: An Improved Human GH? Endocrinology 2020; 161:5859553. [PMID: 32556100 PMCID: PMC7375802 DOI: 10.1210/endocr/bqaa097] [Citation(s) in RCA: 6] [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: 03/26/2020] [Accepted: 06/11/2020] [Indexed: 11/19/2022]
Abstract
A rare 20K isoform of GH-V (here abbreviated as GHv) was discovered in 1998. To date, only 1 research article has characterized this isoform in vivo, observing that GHv treatment in male high-fat fed rats had several GH-like activities, but unlike GH lacked diabetogenic and lactogenic activities and failed to increase IGF-1 or body length. Therefore, the current study was conducted to further characterize the in vivo activities of GHv in a separate species and in a GH-deficient model (GH-/- mice) and with both sexes represented. GHv-treated GH-/- mice had significant increases to serum IGF-1, femur length, body length, body weight, and lean body mass and reduced body fat mass similar to mice receiving GH treatment. GH treatment increased circulating insulin levels and impaired insulin sensitivity; in contrast, both measures were unchanged in GHv-treated mice. Since GHv lacks prolactin receptor (PRLR) binding activity, we tested the ability of GH and GHv to stimulate the proliferation of human cancer cell lines and found that GHv has a decreased proliferative response in cancers with high PRLR. Our findings demonstrate that GHv can stimulate insulin-like growth factor-1 and subsequent longitudinal body growth in GH-deficient mice similar to GH, but unlike GH, GHv promoted growth without inhibiting insulin action and without promoting the growth of PRLR-positive cancers in vitro. Thus, GHv may represent improvements to current GH therapies especially for individuals at risk for metabolic syndrome or PRLR-positive cancers.
Collapse
Affiliation(s)
- Edward O List
- Edison Biotechnology Institute, Ohio University, Athens, Ohio
- Department of Specialty Medicine, Heritage College of Osteopathic Medicine, Athens, Ohio
- Correspondence: Edward O. List, PhD, Edison Biotechnology Institute, Ohio University, Athens, Ohio 45701. E-mail:
| | - Darlene E Berryman
- Edison Biotechnology Institute, Ohio University, Athens, Ohio
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Athens, Ohio
| | - Reetobrata Basu
- Edison Biotechnology Institute, Ohio University, Athens, Ohio
| | - Mathew Buchman
- Edison Biotechnology Institute, Ohio University, Athens, Ohio
| | - Kevin Funk
- Edison Biotechnology Institute, Ohio University, Athens, Ohio
| | | | | | - Yanrong Qian
- Edison Biotechnology Institute, Ohio University, Athens, Ohio
| | | | | | - Gozde Yildirim
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, New York
| | - Shoshana Yakar
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, New York
| | - John J Kopchick
- Edison Biotechnology Institute, Ohio University, Athens, Ohio
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Athens, Ohio
| |
Collapse
|
15
|
Liu H, Sun Y, Zhang X, Li S, Hu D, Xiao L, Chen Y, He L, Wang DW. Integrated Analysis of Summary Statistics to Identify Pleiotropic Genes and Pathways for the Comorbidity of Schizophrenia and Cardiometabolic Disease. Front Psychiatry 2020; 11:256. [PMID: 32425817 PMCID: PMC7212438 DOI: 10.3389/fpsyt.2020.00256] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Accepted: 03/17/2020] [Indexed: 12/31/2022] Open
Abstract
Genome-wide association studies (GWAS) have identified abundant risk loci associated with schizophrenia (SCZ), cardiometabolic disease (CMD) including body mass index, coronary artery diseases, type 2 diabetes, low- and high-density lipoprotein, total cholesterol, and triglycerides. Although recent studies have suggested that genetic risk shared between these disorders, the pleiotropic genes and biological pathways shared between them are still vague. Here we integrated comprehensive multi-dimensional data from GWAS, expression quantitative trait loci (eQTL), and gene set database to systematically identify potential pleiotropic genes and biological pathways shared between SCZ and CMD. By integrating the results from different approaches including FUMA, Sherlock, SMR, UTMOST, FOCUS, and DEPICT, we revealed 21 pleiotropic genes that are likely to be shared between SCZ and CMD. These genes include VRK2, SLC39A8, NT5C2, AMBRA1, ARL6IP4, OGFOD2, PITPNM2, CDK2AP1, C12orf65, ABCB9, SETD8, MPHOSPH9, FES, FURIN, INO80E, YPEL3, MAPK3, SREBF1, TOM1L2, GATAD2A, and TM6SF2. In addition, we also performed the gene-set enrichment analysis using the software of GSA-SNP2 and MAGMA with GWAS summary statistics and identified three biological pathways (MAPK-TRK signaling, growth hormone signaling, and regulation of insulin secretion signaling) shared between them. Our study provides insights into the pleiotropic genes and biological pathways underlying mechanisms for the comorbidity of SCZ and CMD. However, further genetic and functional studies are required to validate the role of these potential pleiotropic genes and pathways in the etiology of the comorbidity of SCZ and CMD, which should provide potential targets for future diagnostics and therapeutics.
Collapse
Affiliation(s)
- Hao Liu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Genetics and Development, Shanghai Mental Health Center, Shanghai Jiaotong University, Shanghai, China
| | - Yang Sun
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Xinxin Zhang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Genetics and Development, Shanghai Mental Health Center, Shanghai Jiaotong University, Shanghai, China
| | - Shiyang Li
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Dong Hu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Lei Xiao
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Yanghui Chen
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Lin He
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Genetics and Development, Shanghai Mental Health Center, Shanghai Jiaotong University, Shanghai, China
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| |
Collapse
|
16
|
Likitnukul S, Kalandakanond-Thongsong S, Thammacharoen S. Evidence of growth hormone effect on plasma leptin in diet-induced obesity and diet-resistant rats. ASIAN BIOMED 2019. [DOI: 10.1515/abm-2019-0023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract
Background
Plasma leptin is regulated by several factors, including growth hormone (GH), which influences the pathophysiology of obesity.
Objective
To demonstrate the short-term effect of GH on plasma leptin levels in 3 conditions in vivo with the different amount of body fat mass.
Methods
Adult male Wistar rats were fed with standard chow or hypercaloric diet (HC). The HC rats were demonstrated as HC-feeding obese (HC-O) and HC-feeding resistant (HC-R) rats. Then, they were treated with GH or saline for 3 days. Basal plasma leptin levels were measured at 24 and 32 h. For meal-induced condition, all rats were fed for 2 hand plasma leptin was measured. Further 16-h fasting period, plasma leptin, insulin, and insulin sensitivity indexes were determined.
Results
The short-term GH treatment decreased basal plasma leptin at 32 h after the first GH injection in HC-O rats. However, GH treatment had no effect on meal-induced plasma leptin in all rats. Furthermore, GH treatment attenuated fasting effect on plasma leptin in control and HC-R rats. The insulin resistance (IR) induced by the short-term GH treatment was demonstrated by higher fasting plasma insulin and the increased homeostasis model of IR in HC-R rats.
Conclusions
The study demonstrates the important role of greater fat mass in HC-O rats, which results in decreased basal plasma leptin after short-term GH treatment. For meal-induced condition, GH had no effect on plasma leptin in all rats. Interestingly, GH could attenuate fasting effect on plasma leptin in rats that have lower fat mass.
Collapse
Affiliation(s)
- Sutharinee Likitnukul
- Department of Veterinary Physiology, Faculty of Veterinary Science, Chulalongkorn University , Bangkok 10330 , Thailand
| | | | - Sumpun Thammacharoen
- Department of Veterinary Physiology, Faculty of Veterinary Science, Chulalongkorn University , Bangkok 10330 , Thailand
| |
Collapse
|
17
|
List EO, Berryman DE, Buchman M, Jensen EA, Funk K, Duran-Ortiz S, Qian Y, Young JA, Slyby J, McKenna S, Kopchick JJ. GH Knockout Mice Have Increased Subcutaneous Adipose Tissue With Decreased Fibrosis and Enhanced Insulin Sensitivity. Endocrinology 2019; 160:1743-1756. [PMID: 31099824 PMCID: PMC6760334 DOI: 10.1210/en.2019-00167] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Accepted: 05/11/2019] [Indexed: 12/31/2022]
Abstract
In 1997, our laboratory used targeted gene disruption of the GH receptor (GHR) to generate GHR knockout (GHR-/-) mice, which have been used in >127 published studies to help elucidate GH's numerous activities. However, because GH replacement studies cannot be performed using this line, a GH knockout mouse line via targeted disruption of the GH gene is needed. Therefore, we created and characterized GH gene-disrupted (GH-/-) mice. GH-/- mice have severely decreased IGF-1 levels, small body size, and altered body composition with increased adiposity. GH-/- mice are extremely insulin sensitive but glucose intolerant, with a dramatic reduction in pancreatic islet size. Importantly, disruption of the GH gene had profound and depot-specific effects on white adipose tissue (WAT). Subcutaneous WAT from male and female GH-/- mice have significantly larger adipocytes and reduced fibrosis, neither of which occurred in perigonadal WAT, suggesting that GH has a more pronounced effect on subcutaneous WAT. Comparisons of GH-/- mice to previously published data on GHR-/- mice show a remarkably similar phenotype. Finally, we demonstrate that GH-/- mice are responsive to GH treatment, as shown by changes to serum IGF-1 levels; body length, weight, and composition; and insulin sensitivity. This study not only provides characterization of the first mouse line with targeted mutation of the GH gene but also indicates that GH gene disruption dramatically influences fibrosis of subcutaneous WAT.
Collapse
Affiliation(s)
- Edward O List
- Edison Biotechnology Institute, Ohio University, Athens, 45701
- Department of Specialty Medicine, Heritage College of Osteopathic Medicine, Athens, Ohio
- Correspondence: Edward O. List, PhD, Edison Biotechnology Institute, Ohio University, Athens, Ohio 45701. E-mail:
| | - Darlene E Berryman
- Edison Biotechnology Institute, Ohio University, Athens, 45701
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Athens, Ohio
| | - Mathew Buchman
- Edison Biotechnology Institute, Ohio University, Athens, 45701
- College of Health Sciences and Professions, Ohio University, Athens, Ohio
| | | | - Kevin Funk
- Edison Biotechnology Institute, Ohio University, Athens, 45701
| | | | - Yanrong Qian
- Edison Biotechnology Institute, Ohio University, Athens, 45701
| | | | - Julie Slyby
- Edison Biotechnology Institute, Ohio University, Athens, 45701
| | | | - John J Kopchick
- Edison Biotechnology Institute, Ohio University, Athens, 45701
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Athens, Ohio
| |
Collapse
|
18
|
List EO, Berryman DE, Buchman M, Parker C, Funk K, Bell S, Duran-Ortiz S, Qian Y, Young JA, Wilson C, Slyby J, McKenna S, Jensen EA, Kopchick JJ. Adipocyte-Specific GH Receptor-Null (AdGHRKO) Mice Have Enhanced Insulin Sensitivity With Reduced Liver Triglycerides. Endocrinology 2019; 160:68-80. [PMID: 30462209 PMCID: PMC6304108 DOI: 10.1210/en.2018-00850] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 11/09/2018] [Indexed: 12/16/2022]
Abstract
Global GH receptor-null or knockout (GHRKO) mice have been extensively studied owing to their unique phenotype (dwarf and obese but remarkably insulin sensitive and long-lived). To better understand the influence of adipose tissue (AT) on the GHRKO phenotype, we previously generated fat-specific GHRKO (FaGHRKO) mice using the adipocyte protein-2 (aP2) promoter driving Cre expression. Unlike global GHRKO mice, FaGHRKO mice are larger than control mice and have an increase in white AT (WAT) mass and adipocyte size as well as an increase in brown AT mass. FaGHRKO mice also have an unexpected increase in IGF-1, decrease in adiponectin, no change in insulin sensitivity or liver triglyceride content, and a decreased lifespan. Extensive analysis of the aP2 promoter/enhancer by multiple laboratories has revealed expression in nonadipose tissues, confounding interpretation of results. In the current study, we used the adiponectin promoter/enhancer to drive Cre expression, which better targets mature adipocytes, and generated a new line of adipocyte-specific GHRKO (AdGHRKO) mice. AdGHRKO mice have an increase in adipocyte size and WAT depot mass in all depots except male perigonadal, a WAT accumulation pattern similar to FaGHRKO mice. Likewise, adiponectin levels and WAT fibrosis are decreased in both tissue-specific mouse lines. However, unlike FaGHRKO mice, AdGHRKO mice have no change in IGF-1 levels, improved glucose homeostasis, and reduced liver triglycerides. Thus, AdGHRKO mice should be valuable for future studies assessing the contribution of adipocyte GHR signaling in long-term health and lifespan.
Collapse
Affiliation(s)
- Edward O List
- Edison Biotechnology Institute, Ohio University, Athens, Ohio
- Department of Specialty Medicine, Heritage College of Osteopathic Medicine, Athens, Ohio
- Correspondence: Edward O. List, PhD, Edison Biotechnology Institute, Ohio University, 218 Konneker Research Labs, 172 Watertower Drive, Athens, Ohio 45701. E-mail:
| | - Darlene E Berryman
- Edison Biotechnology Institute, Ohio University, Athens, Ohio
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Athens, Ohio
| | - Mathew Buchman
- Edison Biotechnology Institute, Ohio University, Athens, Ohio
- College of Health Sciences and Professions, Ohio University, Athens, Ohio
| | - Caitlin Parker
- Edison Biotechnology Institute, Ohio University, Athens, Ohio
- College of Health Sciences and Professions, Ohio University, Athens, Ohio
| | - Kevin Funk
- Edison Biotechnology Institute, Ohio University, Athens, Ohio
| | - Stephen Bell
- Edison Biotechnology Institute, Ohio University, Athens, Ohio
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Athens, Ohio
| | | | - Yanrong Qian
- Edison Biotechnology Institute, Ohio University, Athens, Ohio
| | | | - Cody Wilson
- Edison Biotechnology Institute, Ohio University, Athens, Ohio
| | - Julie Slyby
- Edison Biotechnology Institute, Ohio University, Athens, Ohio
| | | | | | - John J Kopchick
- Edison Biotechnology Institute, Ohio University, Athens, Ohio
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Athens, Ohio
| |
Collapse
|
19
|
Cordoba-Chacon J, Sarmento-Cabral A, del Rio-Moreno M, Diaz-Ruiz A, Subbaiah PV, Kineman RD. Adult-Onset Hepatocyte GH Resistance Promotes NASH in Male Mice, Without Severe Systemic Metabolic Dysfunction. Endocrinology 2018; 159:3761-3774. [PMID: 30295789 PMCID: PMC6202859 DOI: 10.1210/en.2018-00669] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 10/01/2018] [Indexed: 12/20/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD), which includes nonalcoholic steatohepatitis (NASH), is associated with reduced GH input/signaling, and GH therapy is effective in the reduction/resolution of NAFLD/NASH in selected patient populations. Our laboratory has focused on isolating the direct vs indirect effects of GH in preventing NAFLD/NASH. We reported that chow-fed, adult-onset, hepatocyte-specific, GH receptor knockdown (aHepGHRkd) mice rapidly (within 7 days) develop steatosis associated with increased hepatic de novo lipogenesis (DNL), independent of changes in systemic metabolic function. In this study, we report that 6 months after induction of aHepGHRkd early signs of NASH develop, which include hepatocyte ballooning, inflammation, signs of mild fibrosis, and elevated plasma alanine aminotransferase. These changes occur in the presence of enhanced systemic lipid utilization, without evidence of white adipose tissue lipolysis, indicating that the liver injury that develops after aHepGHRkd is due to hepatocyte-specific loss of GH signaling and not due to secondary defects in systemic metabolic function. Specifically, enhanced hepatic DNL is sustained with age in aHepGHRkd mice, associated with increased hepatic markers of lipid uptake/re-esterification. Because hepatic DNL is a hallmark of NAFLD/NASH, these studies suggest that enhancing hepatocyte GH signaling could represent an effective therapeutic target to reduce DNL and treat NASH.
Collapse
Affiliation(s)
- Jose Cordoba-Chacon
- Section of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Andre Sarmento-Cabral
- Section of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
- Research and Development Division, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois
| | - Mercedes del Rio-Moreno
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC)/University of Cordoba, Cordoba, Spain
| | - Alberto Diaz-Ruiz
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
- Nutritional Interventions Group, Precision Nutrition and Aging, Institute IMDEA Food, Madrid, Spain
| | - Papasani V Subbaiah
- Section of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
- Research and Development Division, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois
| | - Rhonda D Kineman
- Section of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
- Research and Development Division, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois
| |
Collapse
|
20
|
Ha MS, Son WM. Combined exercise is a modality for improving insulin resistance and aging-related hormone biomarkers in elderly Korean women. Exp Gerontol 2018; 114:13-18. [PMID: 30359693 DOI: 10.1016/j.exger.2018.10.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 10/16/2018] [Accepted: 10/17/2018] [Indexed: 12/01/2022]
Abstract
BACKGROUND The functional decline of the endocrine and immune systems with aging causes changes in the regulation of the body's metabolism and defense functions. Although it is impossible to stop aging artificially, regular exercises reportedly delay aging and have positive effects on senile diseases. The improvements in insulin resistance values and aging-related hormones in elderly women after combined exercises have not been effectively elucidated. OBJECTIVE In this study, we aimed to examine the impact of combined exercise on insulin resistance and aging-related hormones in elderly women. METHODS Twenty elderly Korean women were randomly assigned to a "non-exercise" (n = 10) or combined exercise group (n = 10). The exercise group performed both anaerobic and aerobic exercises for 12 weeks, three times per week. Exercise intensity was increased gradually, from 40% to 70% of the heart rate reserve (HRR) every 3 weeks. Insulin resistance, growth hormone, IGF-1, DHEA-S, and estrogen were measured before and after the 12-week intervention. RESULTS The main effect was found in the glucose level for a time, which significantly decreased in the exercise group. The insulin level and HOMA-IR showed significant interaction effects and increased significantly in the control group. The GH level showed significant interaction effects and increased significantly in the exercise group. The IGF-1 level showed significant interaction effects, but not significantly within and between groups. The DHEA-S level revealed interaction effects and the main effect for a time and significantly increased in the exercise group. The estrogen level exhibited an interaction effect and increased significantly in the exercise group. After the 12-week combined exercise intervention, no significant difference was observed between groups. DISCUSSION This study has shown that 12-week combined exercise is useful for improving insulin resistance and GH, IGF-1, and DHEA-S levels in elderly women. Thus, this study provides evidence that combined exercise is a useful therapeutic method to decrease insulin resistance and stimulate the secretion of aging-related hormones in elderly women.
Collapse
Affiliation(s)
- Min-Seong Ha
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Faculty of Health and Sports Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8574, Japan.
| | - Won-Mok Son
- Department of Physical Education, Pusan National University, 2 Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Republic of Korea.
| |
Collapse
|
21
|
Patton A, Church T, Wilson C, Thuma J, Goetz DJ, Berryman DE, List EO, Schwartz F, McCall KD. Phenylmethimazole abrogates diet-induced inflammation, glucose intolerance and NAFLD. J Endocrinol 2018; 237:337-351. [PMID: 29666152 PMCID: PMC5958349 DOI: 10.1530/joe-18-0078] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 04/17/2018] [Indexed: 12/12/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the hepatic manifestation of both metabolic and inflammatory diseases and has become the leading chronic liver disease worldwide. High-fat (HF) diets promote an increased uptake and storage of free fatty acids (FFAs) and triglycerides (TGs) in hepatocytes, which initiates steatosis and induces lipotoxicity, inflammation and insulin resistance. Activation and signaling of Toll-like receptor 4 (TLR4) by FFAs induces inflammation evident in NAFLD and insulin resistance. Currently, there are no effective treatments to specifically target inflammation associated with this disease. We have established the efficacy of phenylmethimazole (C10) to prevent lipopolysaccharide and palmitate-induced TLR4 signaling. Because TLR4 is a key mediator in pro-inflammatory responses, it is a potential therapeutic target for NAFLD. Here, we show that treatment with C10 inhibits HF diet-induced inflammation in both liver and mesenteric adipose tissue measured by a decrease in mRNA levels of pro-inflammatory cytokines. Additionally, C10 treatment improves glucose tolerance and hepatic steatosis despite the development of obesity due to HF diet feeding. Administration of C10 after 16 weeks of HF diet feeding reversed glucose intolerance, hepatic inflammation, and improved hepatic steatosis. Thus, our findings establish C10 as a potential therapeutic for the treatment of NAFLD.
Collapse
Affiliation(s)
- Ashley Patton
- Department of Specialty MedicineHeritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA
- Diabetes Institute Ohio University, Athens, Ohio, USA
- Department of Biological SciencesOhio University, Athens, Ohio, USA
- Molecular & Cellular Biology ProgramCollege of Arts and Sciences, Ohio University, Athens, Ohio, USA
| | - Tyler Church
- Department of Specialty MedicineHeritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA
- Diabetes Institute Ohio University, Athens, Ohio, USA
| | - Caroline Wilson
- Department of Chemical and Biomolecular EngineeringRuss College of Engineering and Technology, Ohio University, Athens, Ohio, USA
| | - Jean Thuma
- Department of Specialty MedicineHeritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA
- Diabetes Institute Ohio University, Athens, Ohio, USA
| | - Douglas J Goetz
- Department of Chemical and Biomolecular EngineeringRuss College of Engineering and Technology, Ohio University, Athens, Ohio, USA
- Molecular & Cellular Biology ProgramCollege of Arts and Sciences, Ohio University, Athens, Ohio, USA
- Biomedical Engineering ProgramOhio University, Athens, Ohio, USA
| | - Darlene E Berryman
- Diabetes Institute Ohio University, Athens, Ohio, USA
- Department of Biomedical SciencesOhio University, Athens, Ohio, USA
- The Edison Biotechnology InstituteOhio University, Athens, Ohio, USA
| | - Edward O List
- Department of Specialty MedicineHeritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA
- Diabetes Institute Ohio University, Athens, Ohio, USA
- The Edison Biotechnology InstituteOhio University, Athens, Ohio, USA
| | - Frank Schwartz
- Department of Specialty MedicineHeritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA
- Diabetes Institute Ohio University, Athens, Ohio, USA
| | - Kelly D McCall
- Department of Specialty MedicineHeritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA
- Diabetes Institute Ohio University, Athens, Ohio, USA
- Department of Biological SciencesOhio University, Athens, Ohio, USA
- Molecular & Cellular Biology ProgramCollege of Arts and Sciences, Ohio University, Athens, Ohio, USA
- Biomedical Engineering ProgramOhio University, Athens, Ohio, USA
- Department of Biomedical SciencesOhio University, Athens, Ohio, USA
| |
Collapse
|
22
|
Cabrera D, Cabello-Verrugio C, Solís N, San Martín D, Cofré C, Pizarro M, Arab JP, Abrigo J, Campos F, Irigoyen B, Carrasco-Avino G, Bezares K, Riquelme V, Riquelme A, Arrese M, Barrera F. Somatotropic Axis Dysfunction in Non-Alcoholic Fatty Liver Disease: Beneficial Hepatic and Systemic Effects of Hormone Supplementation. Int J Mol Sci 2018; 19:ijms19051339. [PMID: 29724029 PMCID: PMC5983806 DOI: 10.3390/ijms19051339] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 03/30/2018] [Accepted: 04/10/2018] [Indexed: 02/07/2023] Open
Abstract
Background: Somatotropic axis dysfunction associated with non-alcoholic fatty liver disease (NAFLD) has potential multisystemic detrimental effects. Here, we analysed the effects of growth hormone (GH) and insulin-like growth factor-1 (IGF-1) supplementation on liver histology, adipokine profile and muscle function in an NAFLD model. Methods: C57BL/6 mice were fed with a high fat diet (HFD) for 12 weeks and were separated into three groups treated for 4 weeks with: (1) High fat diet (HFD) (n = 10); (2) HFD + GH 9 μg/g/d (n = 10); (3) HFD + IGF-1 0.02 µg/g/d (n = 9). A control group fed a chow diet was included (n = 6). Liver histology, liver triglycerides content, serum alanine aminotransferase (ALT) activity, adiponectin and leptin serum levels, in vivo muscle strength, tetanic force and muscle fibre cross-sectional area (CSA) were measured. Results: HFD + GH and HFD + IGF-1 groups showed significantly lower ALT activity compared to HFD (p < 0.01). Liver triglyceride content in HFD + GH was decreased compared to HFD (p < 0.01). Histologic steatosis score was increased in HFD and HFD + GH group (p < 0.01), whereas HFD + IGF-1 presented no difference compared to the chow group (p = 0.3). HFD + GH group presented lower serum leptin and adiponectin levels compared to HFD. GH and IGF-1 supplementation therapy reverted HFD-induced reduction in muscle strength and CSA (sarcopenia). Conclusions: GH and IGF-1 supplementation induced significant improvement in liver steatosis, aminotransferases and sarcopenia in a diet-induced NAFLD model.
Collapse
Affiliation(s)
- Daniel Cabrera
- Departament of Gastroenterology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile.
| | | | - Nancy Solís
- Departament of Gastroenterology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile.
| | - Diego San Martín
- Departament of Gastroenterology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile.
| | - Catalina Cofré
- Departament of Gastroenterology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile.
| | - Margarita Pizarro
- Departament of Gastroenterology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile.
| | - Juan Pablo Arab
- Departament of Gastroenterology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile.
| | - Johanna Abrigo
- Faculty of Biological Sciences, Universidad Andrés Bello, Santiago 8320000, Chile.
| | - Fabián Campos
- Faculty of Biological Sciences, Universidad Andrés Bello, Santiago 8320000, Chile.
| | - Betzabé Irigoyen
- Faculty of Biological Sciences, Universidad Andrés Bello, Santiago 8320000, Chile.
| | - Gonzalo Carrasco-Avino
- Departament of Pathotology, Clínica Las Condes, Santiago 8320000, Chile.
- Department of Pathology, Hospital Clínico Universidad de Chile, Santiago 8320000, Chile.
| | - Katiuska Bezares
- Department of Pathology, Hospital Clínico San Juan de Dios, Santiago 8320000, Chile.
| | - Valentina Riquelme
- Faculty of Arts, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile.
| | - Arnoldo Riquelme
- Departament of Gastroenterology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile.
- Department of Health Sciences, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile.
| | - Marco Arrese
- Departament of Gastroenterology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile.
- Centro de Envejecimiento y Regeneración (CARE), Pontificia Universidad Católica de Chile, Santiago 8320000, Chile.
| | - Francisco Barrera
- Departament of Gastroenterology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile.
| |
Collapse
|
23
|
Gong Z, Tas E, Yakar S, Muzumdar R. Hepatic lipid metabolism and non-alcoholic fatty liver disease in aging. Mol Cell Endocrinol 2017; 455:115-130. [PMID: 28017785 DOI: 10.1016/j.mce.2016.12.022] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 09/23/2016] [Accepted: 12/16/2016] [Indexed: 02/06/2023]
Abstract
Aging is associated with dysregulation of glucose and lipid metabolism. Various factors that contribute to the dysregulation include both modifiable (e.g. obesity, insulin resistance) and non-modifiable risk factors (age-associated physiologic changes). Although there is no linear relationship between aging and prevalence of non-alcoholic fatty liver disease, current data strongly suggests that advanced age leads to more severe histological changes and poorer clinical outcomes. Hepatic lipid accumulation could lead to significant hepatic and systemic consequences including steatohepatitis, cirrhosis, impairment of systemic glucose metabolism and metabolic syndrome, thereby contributing to age-related diseases. Insulin, leptin and adiponectin are key regulators of the various physiologic processes that regulate hepatic lipid metabolism. Recent advances have expanded our understanding in this field, highlighting the role of novel mediators such as FGF 21, and mitochondria derived peptides. In this review, we will summarize the mediators of hepatic lipid metabolism and how they are altered in aging.
Collapse
Affiliation(s)
- Zhenwei Gong
- Department of Pediatrics, University of Pittsburgh School of Medicine, One Children's Hospital Drive, 4401 Penn Avenue, Pittsburgh, PA 15224, USA; Children's Hospital of Pittsburgh of UPMC, One Children's Hospital Drive, 4401 Penn Avenue, Pittsburgh, PA 15224, USA
| | - Emir Tas
- Children's Hospital of Pittsburgh of UPMC, One Children's Hospital Drive, 4401 Penn Avenue, Pittsburgh, PA 15224, USA
| | - Shoshana Yakar
- David B. Kriser Dental Center, Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY 10010, USA
| | - Radhika Muzumdar
- Department of Pediatrics, University of Pittsburgh School of Medicine, One Children's Hospital Drive, 4401 Penn Avenue, Pittsburgh, PA 15224, USA; Children's Hospital of Pittsburgh of UPMC, One Children's Hospital Drive, 4401 Penn Avenue, Pittsburgh, PA 15224, USA; Department of Cell Biology, University of Pittsburgh School of Medicine, 3500 Terrace Street, 5362 Biomedical Sciences Tower, Pittsburgh, PA 15261, USA.
| |
Collapse
|
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
|
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
|
26
|
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
|
27
|
Cady G, Landeryou T, Garratt M, Kopchick JJ, Qi N, Garcia-Galiano D, Elias CF, Myers MG, Miller RA, Sandoval DA, Sadagurski M. Hypothalamic growth hormone receptor (GHR) controls hepatic glucose production in nutrient-sensing leptin receptor (LepRb) expressing neurons. Mol Metab 2017; 6:393-405. [PMID: 28462074 PMCID: PMC5404104 DOI: 10.1016/j.molmet.2017.03.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 02/28/2017] [Accepted: 03/04/2017] [Indexed: 12/22/2022] Open
Abstract
Objective The GH/IGF-1 axis has important roles in growth and metabolism. GH and GH receptor (GHR) are active in the central nervous system (CNS) and are crucial in regulating several aspects of metabolism. In the hypothalamus, there is a high abundance of GH-responsive cells, but the role of GH signaling in hypothalamic neurons is unknown. Previous work has demonstrated that the Ghr gene is highly expressed in LepRb neurons. Given that leptin is a key regulator of energy balance by acting on leptin receptor (LepRb)-expressing neurons, we tested the hypothesis that LepRb neurons represent an important site for GHR signaling to control body homeostasis. Methods To determine the importance of GHR signaling in LepRb neurons, we utilized Cre/loxP technology to ablate GHR expression in LepRb neurons (LeprEYFPΔGHR). The mice were generated by crossing the Leprcre on the cre-inducible ROSA26-EYFP mice to GHRL/L mice. Parameters of body composition and glucose homeostasis were evaluated. Results Our results demonstrate that the sites with GHR and LepRb co-expression include ARH, DMH, and LHA neurons. Leptin action was not altered in LeprEYFPΔGHR mice; however, GH-induced pStat5-IR in LepRb neurons was significantly reduced in these mice. Serum IGF-1 and GH levels were unaltered, and we found no evidence that GHR signaling regulates food intake and body weight in LepRb neurons. In contrast, diminished GHR signaling in LepRb neurons impaired hepatic insulin sensitivity and peripheral lipid metabolism. This was paralleled with a failure to suppress expression of the gluconeogenic genes and impaired hepatic insulin signaling in LeprEYFPΔGHR mice. Conclusion These findings suggest the existence of GHR-leptin neurocircuitry that plays an important role in the GHR-mediated regulation of glucose metabolism irrespective of feeding. GHR and LepRb are co-localized in the ARH, DMH and LHA neurons. GHR signaling does not regulate food intake and body weight in LepRb neurons. Diminished GHR signaling in LepRb neurons impairs hepatic glucose production.
Collapse
Key Words
- ARH, arcuate nucleus of the hypothalamus
- CNS, central nervous system
- DMH, dorsomedial hypothalamic nucleus
- GH, growth hormone
- GHR, growth hormone receptor
- Glucose production
- Growth hormone receptor
- Hypothalamus
- LHA, lateral hypothalamus
- Lepr, leptin receptor
- Leptin receptor
- Liver
- POMC, proopiomelanocortin
- PVH, paraventricular hypothalamic nucleus
- Stat3, signal transducer and activator of transcription 3
- Stat5, signal transducer and activator of transcription 5
Collapse
Affiliation(s)
- Gillian Cady
- Department of Pathology and Geriatrics Center, University of Michigan Medical School, USA
| | - Taylor Landeryou
- Department of Pathology and Geriatrics Center, University of Michigan Medical School, USA
| | - Michael Garratt
- Department of Pathology and Geriatrics Center, University of Michigan Medical School, USA
| | - John J Kopchick
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
| | - Nathan Qi
- Division of Metabolism, Endocrinology and Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - David Garcia-Galiano
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Carol F Elias
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Martin G Myers
- Division of Metabolism, Endocrinology and Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Richard A Miller
- Department of Pathology and Geriatrics Center, University of Michigan Medical School, USA
| | - Darleen A Sandoval
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Marianna Sadagurski
- Division of Geriatric and Palliative Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
28
|
Abstract
Obesity is a global epidemic that contributes to a number of health complications including cardiovascular disease, type 2 diabetes, cancer and neuropsychiatric disorders. Pharmacotherapeutic strategies to treat obesity are urgently needed. Research over the past two decades has increased substantially our knowledge of central and peripheral mechanisms underlying homeostatic energy balance. Homeostatic mechanisms involve multiple components including neuronal circuits, some originating in hypothalamus and brain stem, as well as peripherally-derived satiety, hunger and adiposity signals that modulate neural activity and regulate eating behavior. Dysregulation of one or more of these homeostatic components results in obesity. Coincident with obesity, reward mechanisms that regulate hedonic aspects of food intake override the homeostatic regulation of eating. In addition to functional interactions between homeostatic and reward systems in the regulation of food intake, homeostatic signals have the ability to alter vulnerability to drug abuse. Regarding the treatment of obesity, pharmacological monotherapies primarily focus on a single protein target. FDA-approved monotherapy options include phentermine (Adipex-P®), orlistat (Xenical®), lorcaserin (Belviq®) and liraglutide (Saxenda®). However, monotherapies have limited efficacy, in part due to the recruitment of alternate and counter-regulatory pathways. Consequently, a multi-target approach may provide greater benefit. Recently, two combination products have been approved by the FDA to treat obesity, including phentermine/topiramate (Qsymia®) and naltrexone/bupropion (Contrave®). The current review provides an overview of homeostatic and reward mechanisms that regulate energy balance, potential therapeutic targets for obesity and current treatment options, including some candidate therapeutics in clinical development. Finally, challenges in anti-obesity drug development are discussed.
Collapse
Affiliation(s)
- Vidya Narayanaswami
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, 40536, USA
| | - Linda P Dwoskin
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, 40536, USA.
| |
Collapse
|
29
|
The Association Between IGF-1 Levels and the Histologic Severity of Nonalcoholic Fatty Liver Disease. Clin Transl Gastroenterol 2017; 8:e217. [PMID: 28125073 PMCID: PMC5288606 DOI: 10.1038/ctg.2016.72] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 12/07/2016] [Indexed: 12/13/2022] Open
Abstract
Objectives: The mechanisms responsible for the development of nonalcoholic fatty liver disease (NAFLD) and progression to nonalcoholic steatohepatitis (NASH) are incompletely understood. Growing evidence suggests that growth hormone (GH) and insulin-like growth factor-1 (IGF-1) may have roles in the development and progression of NAFLD. We hypothesized that lower serum IGF-1 levels would be associated with increased liver fat accumulation, inflammation, and fibrosis in a group of meticulously phenotyped obese subjects with liver biopsies. Methods: A retrospective, cross-sectional study was performed at Massachusetts General Hospital, Boston, MA, USA and St. Mary's Hospital, Richmond, VA, USA. Liver biopsies were performed in 142 subjects during NAFLD work-up or bariatric surgery and were graded by a single, blinded pathologist. Main outcome measures included liver histology and serum IGF-1. Results: Mean age was 52±10 years and body mass index (BMI) was 43±9 kg/m2. Mean serum IGF-1 was lower in subjects with lobular inflammation (112±47 vs. 136±57 ng/ml, P=0.01), hepatocyte ballooning (115±48 vs. 135±57 ng/ml, P=0.05), higher fibrosis stage (stage 2–4 vs. 0–1; 96±40 vs. 125±51 ng/ml, P=0.005), and NASH (109±45 vs. 136±57 ng/ml, P=0.002). All results remained significant after controlling for age, BMI, and a diagnosis of diabetes, and all but hepatocyte ballooning (trend, P=0.06) remained significant after excluding individuals with cirrhosis. Steatosis was not significantly associated with mean serum IGF-1 levels. Conclusions: Low serum IGF-1 levels are associated with increased histologic severity of NAFLD when rigorously controlled for age, BMI, the presence of diabetes, and after the exclusion of subjects with cirrhosis. Further investigation is warranted to determine the differential effects of GH and IGF-1 on the development and progression of NAFLD, which could further elucidate pathophysiology and identify therapeutic targets.
Collapse
|
30
|
Liu Z, Cordoba-Chacon J, Kineman RD, Cronstein BN, Muzumdar R, Gong Z, Werner H, Yakar S. Growth Hormone Control of Hepatic Lipid Metabolism. Diabetes 2016; 65:3598-3609. [PMID: 27679560 PMCID: PMC5127251 DOI: 10.2337/db16-0649] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 09/20/2016] [Indexed: 12/15/2022]
Abstract
In humans, low levels of growth hormone (GH) and its mediator, IGF-1, associate with hepatic lipid accumulation. In mice, congenital liver-specific ablation of the GH receptor (GHR) results in reductions in circulating IGF-1 and hepatic steatosis, associated with systemic insulin resistance. Due to the intricate relationship between GH and IGF-1, the relative contribution of each hormone to the development of hepatic steatosis is unclear. Our goal was to dissect the mechanisms by which hepatic GH resistance leads to steatosis and overall insulin resistance, independent of IGF-1. We have generated a combined mouse model with liver-specific ablation of GHR in which we restored liver IGF-1 expression via the hepatic IGF-1 transgene. We found that liver GHR ablation leads to increases in lipid uptake, de novo lipogenesis, hyperinsulinemia, and hyperglycemia accompanied with severe insulin resistance and increased body adiposity and serum lipids. Restoration of IGF-1 improved overall insulin sensitivity and lipid profile in serum and reduced body adiposity, but was insufficient to protect against steatosis-induced hepatic inflammation or oxidative stress. We conclude that the impaired metabolism in states of GH resistance results from direct actions of GH on lipid uptake and de novo lipogenesis, whereas its actions on extrahepatic tissues are mediated by IGF-1.
Collapse
Affiliation(s)
- Zhongbo Liu
- Department of Basic Science & Craniofacial Biology, David B. Kriser Dental Center, NYU College of Dentistry, New York, NY
| | - Jose Cordoba-Chacon
- Research and Development, Jesse Brown VA Medical Center, Chicago, IL
- Department of Medicine, Section of Endocrinology, Diabetes and Metabolism, University of Illinois at Chicago, Chicago, IL
| | - Rhonda D Kineman
- Research and Development, Jesse Brown VA Medical Center, Chicago, IL
- Department of Medicine, Section of Endocrinology, Diabetes and Metabolism, University of Illinois at Chicago, Chicago, IL
| | | | - Radhika Muzumdar
- Division of Pediatric Endocrinology, Diabetes and Metabolism Consultation, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Zhenwei Gong
- Division of Pediatric Endocrinology, Diabetes and Metabolism Consultation, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Haim Werner
- Department of Human Molecular Genetics and Biochemistry, The Sackler School of Medicine, Tel Aviv University, Ramat Aviv, Israel
| | - Shoshana Yakar
- Department of Basic Science & Craniofacial Biology, David B. Kriser Dental Center, NYU College of Dentistry, New York, NY
| |
Collapse
|
31
|
Raider K, Ma D, Harris JL, Fuentes I, Rogers RS, Wheatley JL, Geiger PC, Yeh HW, Choi IY, Brooks WM, Stanford JA. A high fat diet alters metabolic and bioenergetic function in the brain: A magnetic resonance spectroscopy study. Neurochem Int 2016; 97:172-80. [PMID: 27125544 PMCID: PMC4900919 DOI: 10.1016/j.neuint.2016.04.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 04/18/2016] [Accepted: 04/20/2016] [Indexed: 11/18/2022]
Abstract
Diet-induced obesity and associated metabolic effects can lead to neurological dysfunction and increase the risk of developing Alzheimer's disease (AD) and Parkinson's disease (PD). Despite these risks, the effects of a high-fat diet on the central nervous system are not well understood. To better understand the mechanisms underlying the effects of high fat consumption on brain regions affected by AD and PD, we used proton magnetic resonance spectroscopy ((1)H-MRS) to measure neurochemicals in the hippocampus and striatum of rats fed a high fat diet vs. normal low fat chow. We detected lower concentrations of total creatine (tCr) and a lower glutamate-to-glutamine ratio in the hippocampus of high fat rats. Additional effects observed in the hippocampus of high fat rats included higher N-acetylaspartylglutamic acid (NAAG), and lower myo-inositol (mIns) and serine (Ser) concentrations. Post-mortem tissue analyses revealed lower phosphorylated AMP-activated protein kinase (pAMPK) in the striatum but not in the hippocampus of high fat rats. Hippocampal pAMPK levels correlated significantly with tCr, aspartate (Asp), phosphoethanolamine (PE), and taurine (Tau), indicating beneficial effects of AMPK activation on brain metabolic and energetic function, membrane turnover, and edema. A negative correlation between pAMPK and glucose (Glc) indicates a detrimental effect of brain Glc on cellular energy response. Overall, these changes indicate alterations in neurotransmission and in metabolic and bioenergetic function in the hippocampus and in the striatum of rats fed a high fat diet.
Collapse
Affiliation(s)
- Kayla Raider
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Delin Ma
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Janna L Harris
- Department of Anatomy & Cell Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA; Hoglund Brain Imaging Center, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Isabella Fuentes
- Department of Anatomy & Cell Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Robert S Rogers
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Joshua L Wheatley
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Paige C Geiger
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Hung-Wen Yeh
- Department of Biostatistics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - In-Young Choi
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA; Department of Neurology, University of Kansas Medical Center, Kansas City, KS 66160, USA; Hoglund Brain Imaging Center, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - William M Brooks
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA; Department of Neurology, University of Kansas Medical Center, Kansas City, KS 66160, USA; Hoglund Brain Imaging Center, University of Kansas Medical Center, Kansas City, KS 66160, USA; Kansas Intellectual and Developmental Disabilities Research Center, University of Kansas Medical Center, Kansas City, KS 66160, USA; University of Kansas Alzheimer's Disease Center, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - John A Stanford
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA; Kansas Intellectual and Developmental Disabilities Research Center, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| |
Collapse
|
32
|
Kineman RD, Majumdar N, Subbaiah PV, Cordoba-Chacon J. Hepatic PPARγ Is Not Essential for the Rapid Development of Steatosis After Loss of Hepatic GH Signaling, in Adult Male Mice. Endocrinology 2016; 157:1728-35. [PMID: 26950202 PMCID: PMC4870866 DOI: 10.1210/en.2015-2077] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Our group has previously reported de novo lipogenesis (DNL) and hepatic triglyceride content increases in chow-fed male mice within 7 days of hepatocyte-specific GH receptor knockdown (aLivGHRkd). Here, we report that these changes are associated with an increase in hepatic expression of peroxisome proliferator-activated receptor γ (PPARγ), consistent with previous reports showing steatosis is associated with an increase in PPARγ expression in mice with congenital loss of hepatic GH signaling. PPARγ is thought to be an important driver of steatosis by enhancing DNL, as well as increasing the uptake and esterification of extrahepatic fatty acids (FAs). In order to determine whether hepatic PPARγ is critical for the rapid development of steatosis in the aLivGHRkd mouse model, we have generated aLivGHRkd mice, with or without PPARγ (ie, adult-onset, hepatocyte-specific double knockout of GHR and PPARγ). Hepatic PPARγ was not required for the rapid increase in liver triglyceride content or FA indexes of DNL (16:0/18:2 and 16:1/16:0). However, loss of hepatic PPARγ blunted the rise in fatty acid translocase/CD36 and monoacylglycerol acyltransferase 1 expression induced by aLivGHRkd, and this was associated with a reduction in the hepatic content of 18:2. These results suggest that the major role of PPARγ is to enhance pathways critical in uptake and reesterification of extrahepatic FA. Because FAs have been reported to directly increase PPARγ expression, we speculate that in the aLivGHRkd mouse, the FA produced by DNL enhances the expression of PPARγ, which in turn increases extrahepatic FA uptake, thereby further enhancing PPARγ activity and exacerbating steatosis overtime.
Collapse
Affiliation(s)
- Rhonda D Kineman
- Research and Development Division (R.D.K., N.M., P.V.S., J.C.-C.), Jesse Brown Veterans Affairs Medical Center; and Department of Medicine (R.D.K., N.M., P.V.S., J.C.-C.), Section of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, Illinois 60612
| | - Neena Majumdar
- Research and Development Division (R.D.K., N.M., P.V.S., J.C.-C.), Jesse Brown Veterans Affairs Medical Center; and Department of Medicine (R.D.K., N.M., P.V.S., J.C.-C.), Section of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, Illinois 60612
| | - Papasani V Subbaiah
- Research and Development Division (R.D.K., N.M., P.V.S., J.C.-C.), Jesse Brown Veterans Affairs Medical Center; and Department of Medicine (R.D.K., N.M., P.V.S., J.C.-C.), Section of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, Illinois 60612
| | - Jose Cordoba-Chacon
- Research and Development Division (R.D.K., N.M., P.V.S., J.C.-C.), Jesse Brown Veterans Affairs Medical Center; and Department of Medicine (R.D.K., N.M., P.V.S., J.C.-C.), Section of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, Illinois 60612
| |
Collapse
|
33
|
Cordoba-Chacon J, Majumdar N, List EO, Diaz-Ruiz A, Frank SJ, Manzano A, Bartrons R, Puchowicz M, Kopchick JJ, Kineman RD. Growth Hormone Inhibits Hepatic De Novo Lipogenesis in Adult Mice. Diabetes 2015; 64:3093-103. [PMID: 26015548 PMCID: PMC4542445 DOI: 10.2337/db15-0370] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 05/20/2015] [Indexed: 02/07/2023]
Abstract
Patients with nonalcoholic fatty liver disease (NAFLD) are reported to have low growth hormone (GH) production and/or hepatic GH resistance. GH replacement can resolve the fatty liver condition in diet-induced obese rodents and in GH-deficient patients. However, it remains to be determined whether this inhibitory action of GH is due to direct regulation of hepatic lipid metabolism. Therefore, an adult-onset, hepatocyte-specific, GH receptor (GHR) knockdown (aLivGHRkd) mouse was developed to model hepatic GH resistance in humans that may occur after sexual maturation. Just 7 days after aLivGHRkd, hepatic de novo lipogenesis (DNL) was increased in male and female chow-fed mice, compared with GHR-intact littermate controls. However, hepatosteatosis developed only in male and ovariectomized female aLivGHRkd mice. The increase in DNL observed in aLivGHRkd mice was not associated with hyperactivation of the pathway by which insulin is classically considered to regulate DNL. However, glucokinase mRNA and protein levels as well as fructose-2,6-bisphosphate levels were increased in aLivGHRkd mice, suggesting that enhanced glycolysis drives DNL in the GH-resistant liver. These results demonstrate that hepatic GH actions normally serve to inhibit DNL, where loss of this inhibitory signal may explain, in part, the inappropriate increase in hepatic DNL observed in NAFLD patients.
Collapse
Affiliation(s)
- Jose Cordoba-Chacon
- Research and Development Division, Jesse Brown VA Medical Center, Chicago, IL Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL
| | - Neena Majumdar
- Research and Development Division, Jesse Brown VA Medical Center, Chicago, IL Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL
| | - Edward O List
- Edison Biotechnology Institute, Ohio University, Athens, OH Department of Specialty Medicine, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH
| | - Alberto Diaz-Ruiz
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD
| | - Stuart J Frank
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL Endocrinology Section Medical Service, Birmingham VA Medical Center, Birmingham, AL
| | - Anna Manzano
- Department of Physiological Sciences, University of Barcelona, L'Hospitalet, Barcelona, Spain
| | - Ramon Bartrons
- Department of Physiological Sciences, University of Barcelona, L'Hospitalet, Barcelona, Spain
| | - Michelle Puchowicz
- Department of Nutrition, Case Western Reserve University School of Medicine, Cleveland, OH
| | - John J Kopchick
- Edison Biotechnology Institute, Ohio University, Athens, OH Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH
| | - Rhonda D Kineman
- Research and Development Division, Jesse Brown VA Medical Center, Chicago, IL Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL
| |
Collapse
|
34
|
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
|
35
|
Shi SY, Luk CT, Brunt JJ, Sivasubramaniyam T, Lu SY, Schroer SA, Woo M. Adipocyte-specific deficiency of Janus kinase (JAK) 2 in mice impairs lipolysis and increases body weight, and leads to insulin resistance with ageing. Diabetologia 2014; 57:1016-26. [PMID: 24531222 DOI: 10.1007/s00125-014-3185-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2013] [Accepted: 01/21/2014] [Indexed: 12/14/2022]
Abstract
AIMS/HYPOTHESIS The growing obesity epidemic necessitates a better understanding of adipocyte biology and its role in metabolism. The Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway mediates signalling by numerous cytokines and hormones that regulate adipocyte function, illustrating the physiological importance of adipose JAK-STAT. The aim of this study was to investigate potential roles of adipocyte JAK2, an essential player in the JAK-STAT pathway, in adipocyte biology and metabolism. METHODS We generated adipocyte-specific Jak2 knockout (A-Jak2 KO) mice using the Cre-loxP system with Cre expression driven by the Ap2 (also known as Fabp4) promoter. RESULTS Starting at 2-3 months of age, male and female A-Jak2 KO mice gradually gained more body weight than control littermates primarily due to increased adiposity. This was associated with reduced energy expenditure in A-Jak2 KO mice. In perigonadal adipose tissue, the expression of numerous genes involved in lipid metabolism was differentially regulated. In addition, adipose tissue from A-Jak2 KO mice displayed impaired lipolysis in response to isoprenaline, growth hormone and leptin stimulation, suggesting that adipose JAK2 directly modulates the lipolytic program. Impaired lipid homeostasis was also associated with disrupted adipokine secretion. Accordingly, while glucose metabolism was normal at 2 months of age, by 5-6 months of age, A-Jak2 KO mice had whole-body insulin resistance. CONCLUSIONS/INTERPRETATION Our results suggest that adipocyte JAK2 plays a critical role in the regulation of adipocyte biology and whole-body metabolism. Targeting of the JAK-STAT pathway could be a novel therapeutic option for the treatment of obesity and type 2 diabetes.
Collapse
Affiliation(s)
- Sally Yu Shi
- Toronto General Research Institute, 101 College Street, MaRS Centre/TMDT, Room 10-363, Toronto, ON, Canada, M5G 1L7
| | | | | | | | | | | | | |
Collapse
|
36
|
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
|
37
|
Recinella L, Shohreh R, Salvatori R, Orlando G, Vacca M, Brunetti L. Effects of isolated GH deficiency on adipose tissue, feeding and adipokines in mice. Growth Horm IGF Res 2013; 23:237-242. [PMID: 24021480 DOI: 10.1016/j.ghir.2013.08.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Revised: 07/30/2013] [Accepted: 08/20/2013] [Indexed: 10/26/2022]
Abstract
OBJECTIVE Growth hormone deficiency (GHD) leads to growth failure and changes in body composition including increased fat accumulation and reduced lean body mass in both humans and rodents. The aim of this study was to characterize the consequences of isolated GHD (IGHD) on adiposity, total body weight (TBW), and food intake in a mouse model of autosomal recessive IGHD due to targeted ablation of the GH-releasing hormone (GHRH) gene [GHRH knockout (GHRHKO)]. Animals were also analyzed with respect to leptin, adiponectin and visfatin circulating levels and gene expression in both intra-abdominal and subcutaneous fat. DESIGN We studied 8 male mice homozygous for GHRHKO allele (-/-) and 8 heterozygous (+/-) animals as controls. Feeding and TBW data were collected weekly from 3 through 5 months of age. Animals were then euthanized for measurement of body length and intra-abdominal (epididymal and retroperitoneal) and subcutaneous (interscapular, axillary, gluteal and inguinal) fat weights, and for blood collection for leptin, adiponectin and visfatin measurement. Gene expression of leptin, adiponectin and visfatin in adipose tissue was evaluated by real-time reverse transcription polymerase chain reaction. RESULTS GHRHKO mice had significantly increased relative intra-abdominal (P<0.01) and subcutaneous (P<0.0001) fat, accompanied by significantly increased food intake per TBW (P<0.01), whereas - despite 40% higher food consumption--TBW change was not different from controls over the 2 month period. Adiponectin and visfatin mRNA levels were decreased in both intra-abdominal (P<0.001) and subcutaneous fat (P<0.0001), while leptin mRNA levels were not different from controls. Conversely, serum adiponectin levels were higher in GHRHKO mice (P<0.0001), whereas serum visfatin and leptin did not significantly differ from controls. CONCLUSIONS IGHD due to targeted ablation of the GHRH gene in mice is associated with increased relative subcutaneous and intra-abdominal fat mass and higher food consumption which is not related to changes in circulating leptin.
Collapse
Affiliation(s)
- Lucia Recinella
- Department of Pharmacy, G. d'Annunzio University, Chieti, Italy
| | | | | | | | | | | |
Collapse
|
38
|
Vijayakumar A, Buffin NJ, Gallagher EJ, Blank J, Wu Y, Yakar S, LeRoith D. Deletion of growth hormone receptors in postnatal skeletal muscle of male mice does not alter muscle mass and response to pathological injury. Endocrinology 2013; 154:3776-83. [PMID: 23861377 PMCID: PMC5398538 DOI: 10.1210/en.2013-1209] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
In this study, we investigated whether loss of GH receptor (GHR) signaling in postnatal skeletal muscle alters muscle mass and regenerative ability in adult mice and whether this was dependent on IGF-1 receptor (IGF-1R) signaling. To do so, we used mouse models with skeletal muscle-specific loss of GHR signaling (mGHRKO), IGF-1R and insulin receptor signaling (MKR), or both GHR and IGF-1R/insulin receptor signaling (mGHRKO/MKR). We did not find a reduction in muscle cross-sectional area, fiber type composition, or response to pathological muscle injury in male mGHRKO and mGHRKO/MKR mice when compared with control and MKR mice, respectively. This could potentially be explained by unchanged skeletal muscle Igf-1 expression in mGHRKO and mGHRKO/MKR mice relative to control and MKR mice, respectively. Furthermore, MKR and mGHRKO/MKR mice, but not mGHRKO mice, demonstrated reduced fiber fusion after cardiotoxin injection, suggesting that IGF-1, and not GH, promotes fiber fusion in adult mice. In summary, our data suggest that GHR signaling in postnatal skeletal muscle does not play a significant role in regulating muscle mass or muscle regeneration. Additionally, in our model, muscle Igf-1 expression is not dependent on GHR signaling in postnatal skeletal muscle.
Collapse
MESH Headings
- Animals
- Cardiotoxins/toxicity
- Diabetes Mellitus/drug therapy
- Diabetes Mellitus/metabolism
- Growth Hormone/metabolism
- Humans
- Insulin/metabolism
- Insulin/therapeutic use
- Insulin Resistance
- Insulin-Like Growth Factor I/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Muscle Development
- Muscle, Skeletal/cytology
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/pathology
- Muscle, Skeletal/physiology
- Receptor, IGF Type 1/biosynthesis
- Receptor, IGF Type 1/genetics
- Receptor, IGF Type 1/metabolism
- Receptor, Insulin/genetics
- Receptor, Insulin/metabolism
- Receptors, Somatotropin/genetics
- Receptors, Somatotropin/metabolism
- Regeneration
- Signal Transduction/drug effects
Collapse
Affiliation(s)
- Archana Vijayakumar
- MD, PhD, Division of Endocrinology, Metabolism, and Bone Diseases, Department of Medicine, Mount Sinai School of Medicine, One Gustave L. Levy Place, Box 1055, New York, New York 10029.
| | | | | | | | | | | | | |
Collapse
|
39
|
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
|
40
|
Franck SE, van der Lely AJ, Neggers S. Extra-hepatic Acromegaly. EUROPEAN ENDOCRINOLOGY 2013; 9:66-70. [PMID: 30349613 DOI: 10.17925/ee.2013.09.01.66] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Accepted: 02/14/2013] [Indexed: 02/03/2023]
Abstract
After the introduction of somatostatin analogs (LA-SMSA) and the growth hormone (GH) receptor antagonist, pegvisomant (Peg-v) normal serum insulin-like growth factor-1 (IGF-1) concentrations in virtually every patients with acromegaly is possible. The impact of these products on the GH-IGF1 axis is completely different. We advocate that LA-SMSA may normalize serum IGF1 levels in the presence of elevated GH actions in extra-hepatic tissues. This results in persistent peripheral disease activity that we call 'extra-hepatic acromegaly'. Peg-v competitively blocks systemic GH action and results in a GH serum level increase. Therefore high doses of Peg-v are necessary to control IGF-1. Since the mode of action differs between these products, it is questionable if identical IGF-1 levels, during Peg-v or LA-SMSA are really identical representations of the biochemical situation. With the traditional biomarkers medical treatment is therefore difficult to monitor with the traditional biomarkers. Additionally, Peg-v and LA-SMSA could be ideal combination since they have different mode of actions. We believe that the time has come to challenge the existing concepts of treatment and monitoring of patients with acromegaly.
Collapse
Affiliation(s)
- Sanne E Franck
- Department of Internal Medicine, Erasmus University MC, Rotterdam, the Netherlands
| | | | - Sebastian Neggers
- Department of Internal Medicine, Erasmus University MC, Rotterdam, the Netherlands
| |
Collapse
|
41
|
Lubbers ER, List EO, Jara A, Sackman-Sala L, Cordoba-Chacon J, Gahete MD, Kineman RD, Boparai R, Bartke A, Kopchick JJ, Berryman DE. Adiponectin in mice with altered GH action: links to insulin sensitivity and longevity? J Endocrinol 2013; 216:363-74. [PMID: 23261955 PMCID: PMC3756886 DOI: 10.1530/joe-12-0505] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Adiponectin is positively correlated with longevity and negatively correlated with many obesity-related diseases. While there are several circulating forms of adiponectin, the high-molecular-weight (HMW) version has been suggested to have the predominant bioactivity. Adiponectin gene expression and cognate serum protein levels are of particular interest in mice with altered GH signaling as these mice exhibit extremes in obesity that are positively associated with insulin sensitivity and lifespan as opposed to the typical negative association of these factors. While a few studies have reported total adiponectin levels in young adult mice with altered GH signaling, much remains unresolved, including changes in adiponectin levels with advancing age, proportion of total adiponectin in the HMW form, adipose depot of origin, and differential effects of GH vs IGF1. Therefore, the purpose of this study was to address these issues using assorted mouse lines with altered GH signaling. Our results show that adiponectin is generally negatively associated with GH activity, regardless of age. Further, the amount of HMW adiponectin is consistently linked with the level of total adiponectin and not necessarily with previously reported lifespan or insulin sensitivity of these mice. Interestingly, circulating adiponectin levels correlated strongly with inguinal fat mass, implying that the effects of GH on adiponectin are depot specific. Interestingly, rbGH, but not IGF1, decreased circulating total and HMW adiponectin levels. Taken together, these results fill important gaps in the literature related to GH and adiponectin and question the frequently reported associations of total and HMW adiponectin with insulin sensitivity and longevity.
Collapse
Affiliation(s)
- Ellen R. Lubbers
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701
| | - Edward O. List
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701
| | - Adam Jara
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701
- Department of Biomedical Sciences, College of Osteopathic Medicine, Ohio University, Athens, OH
| | | | | | - Manuel D. Gahete
- Jesse Brown VA Medical Center, Research and Development, Chicago IL 60612
| | - Rhonda D. Kineman
- Jesse Brown VA Medical Center, Research and Development, Chicago IL 60612
| | - Ravneet Boparai
- Southern Illinois University School of Medicine, Springfield, IL 62794
| | - Andrzej Bartke
- Southern Illinois University School of Medicine, Springfield, IL 62794
| | - John J. Kopchick
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701
- Department of Biomedical Sciences, College of Osteopathic Medicine, Ohio University, Athens, OH
| | - Darlene E. Berryman
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701
- Department of Biomedical Sciences, College of Osteopathic Medicine, Ohio University, Athens, OH
- School of Applied Health Sciences and Wellness, College of Health Sciences and Professions, Ohio University, Athens, OH 45701
| |
Collapse
|
42
|
List EO, Berryman DE, Funk K, Gosney ES, Jara A, Kelder B, Wang X, Kutz L, Troike K, Lozier N, Mikula V, Lubbers ER, Zhang H, Vesel C, Junnila RK, Frank SJ, Masternak MM, Bartke A, Kopchick JJ. The role of GH in adipose tissue: lessons from adipose-specific GH receptor gene-disrupted mice. Mol Endocrinol 2013; 27:524-35. [PMID: 23349524 DOI: 10.1210/me.2012-1330] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
GH receptor (GHR) gene-disrupted mice (GHR-/-) have provided countless discoveries as to the numerous actions of GH. Many of these discoveries highlight the importance of GH in adipose tissue. For example GHR-/- mice are insulin sensitive yet obese with preferential enlargement of the sc adipose depot. GHR-/- mice also have elevated levels of leptin, resistin, and adiponectin, compared with controls leading some to suggest that GH may negatively regulate certain adipokines. To help clarify the role that GH exerts specifically on adipose tissue in vivo, we selectively disrupted GHR in adipose tissue to produce Fat GHR Knockout (FaGHRKO) mice. Surprisingly, FaGHRKOs shared only a few characteristics with global GHR-/- mice. Like the GHR-/- mice, FaGHRKO mice are obese with increased total body fat and increased adipocyte size. However, FaGHRKO mice have increases in all adipose depots with no improvements in measures of glucose homeostasis. Furthermore, resistin and adiponectin levels in FaGHRKO mice are similar to controls (or slightly decreased) unlike the increased levels found in GHR-/- mice, suggesting that GH does not regulate these adipokines directly in adipose tissue in vivo. Other features of FaGHRKO mice include decreased levels of adipsin, a near-normal GH/IGF-1 axis, and minimal changes to a large assortment of circulating factors that were measured such as IGF-binding proteins. In conclusion, specific removal of GHR in adipose tissue is sufficient to increase adipose tissue and decrease circulating adipsin. However, removal of GHR in adipose tissue alone is not sufficient to increase levels of resistin or adiponectin and does not alter glucose metabolism.
Collapse
Affiliation(s)
- Edward O List
- Edison Biotechnology Institute, Athens, OH 45701, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
List EO, Berryman DE, Wright-Piekarski J, Jara A, Funk K, Kopchick JJ. The effects of weight cycling on lifespan in male C57BL/6J mice. Int J Obes (Lond) 2012; 37:1088-94. [PMID: 23229739 PMCID: PMC3609933 DOI: 10.1038/ijo.2012.203] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Revised: 10/02/2012] [Accepted: 11/09/2012] [Indexed: 12/30/2022]
Abstract
OBJECTIVE With the increasing rates of obesity, many people diet in an attempt to lose weight. As weight loss is seldom maintained in a single effort, weight cycling is a common occurrence. Unfortunately, reports from clinical studies that have attempted to determine the effect of weight cycling on mortality are in disagreement, and to date, no controlled animal study has been performed to assess the impact of weight cycling on longevity. Therefore, our objective was to determine whether weight cycling altered lifespan in mice that experienced repeated weight gain and weight loss throughout their lives. METHODS Male C57BL/6J mice were placed on one of three lifelong diets: a low-fat (LF) diet, a high-fat (HF) diet or a cycled diet in which the mice alternated between 4 weeks on the LF diet and 4 weeks on the HF diet. Body weight, body composition, several blood parameters and lifespan were assessed. RESULTS Cycling between the HF and LF diet resulted in large fluctuations in body weight and fat mass. These gains and losses corresponded to significant increases and decreases, respectively, in leptin, resistin, GIP, IGF-1, glucose, insulin and glucose tolerance. Surprisingly, weight cycled mice had no significant difference in lifespan (801±45 days) as compared to LF-fed controls (828±74 days), despite being overweight and eating a HF diet for half of their lives. In contrast, the HF-fed group experienced a significant decrease in lifespan (544±73 days) compared with LF-fed controls and cycled mice. CONCLUSIONS This is the first controlled mouse study to demonstrate the effect of lifelong weight cycling on longevity. The act of repeatedly gaining and losing weight, in itself, did not decrease lifespan and was more beneficial than remaining obese.
Collapse
Affiliation(s)
- E O List
- Edison Biotechnology Institute, Konneker Research Laboratories, Ohio University, Athens, OH 45701, USA.
| | | | | | | | | | | |
Collapse
|
44
|
Bielohuby M, Schaab M, Kummann M, Sawitzky M, Gebhardt R, Binder G, Frystyk J, Bjerre M, Hoeflich A, Kratzsch J, Bidlingmaier M. Serum IGF-I is not a reliable pharmacodynamic marker of exogenous growth hormone activity in mice. Endocrinology 2011; 152:4764-76. [PMID: 21971154 DOI: 10.1210/en.2011-1432] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Serum IGF-I is a well-established pharmacodynamic marker of GH administration in humans and has been used for this purpose in animal studies. However, its general suitability in wild-type laboratory mice has not been demonstrated. Here we show that treatment with recombinant human GH (rhGH) in four different strains of laboratory mice increases body weight, lean body mass, and liver weight but does not increase hepatic expression and release of IGF-I. In contrast and as expected, hypophysectomized rats show a rapid increase in serum IGF-I after rhGH administration. The lack of IGF-I up-regulation in mice occurs despite hepatic activation of the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway and is not explained by GH dose, route of administration, origin of GH (i.e. recombinant human, bovine, and murine GH), treatment duration, genetic background, sex, or formation of neutralizing antibodies. Effects on other components of the GH/IGF pathway were highly influenced by genetic background and sex but not consistently affected by rhGH treatment. We conclude that IGF-I is not a reliable indicator of the biological effects of exogenous GH treatment in genetically and pharmacologically unmodified mice. We speculate that IGF-I release is already maximal in these animals and cannot be further increased by exogenous GH treatment. This is also suggested by the observation of restored IGF-I up-regulation in isolated murine hepatocytes after rhGH treatment. Total body weight, lean body mass, and liver weight may be more reliable phenotypic indicators in these models.
Collapse
Affiliation(s)
- Maximilian Bielohuby
- Endocrine Research Unit, Medizinische Klinik-Innenstadt, Ludwig-Maximilians University, D-80336 Munich, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Ding J, List EO, Bower BD, Kopchick JJ. Differential effects of growth hormone versus insulin-like growth factor-I on the mouse plasma proteome. Endocrinology 2011; 152:3791-802. [PMID: 21791560 PMCID: PMC3176651 DOI: 10.1210/en.2011-1217] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Accepted: 07/07/2011] [Indexed: 12/23/2022]
Abstract
The GH/IGF-I axis has both pre- and postpubertal metabolic effects. However, the differential effects of GH and/or IGF-I on animal physiology or the plasma proteome are still being unraveled. In this report, we analyzed several physiological effects along with the plasma proteome after treatment of mice with recombinant bovine GH or recombinant human IGF-I. GH and IGF-I showed similar effects in increasing body length, body weight, lean and fluid masses, and organ weights including muscle, kidney, and spleen. However, GH significantly increased serum total cholesterol, whereas IGF-I had no effect on it. Both acute and longer-term effects on the plasma proteome were determined. Proteins found to be significantly changed by recombinant bovine GH and/or recombinant human IGF-I injections were identified by mass spectrometry (MS) and MS/MS. The identities of these proteins were further confirmed by Western blotting analysis. Isoforms of apolipoprotein A4, apolipoprotein E, serum amyloid protein A-1, clusterin, transthyretin, and several albumin fragments were found to be differentially regulated by GH vs. IGF-I in mouse plasma. Thus, we have identified several plasma protein biomarkers that respond specifically and differentially to GH or IGF-I and may represent new physiological targets of these hormones. These findings may lead to better understanding of the independent biological effects of GH vs. IGF-I. In addition, these novel biomarkers may be useful for the development of tests to detect illicit use of GH or IGF-I.
Collapse
Affiliation(s)
- Juan Ding
- Edison Biotechnology Institute, College of Osteopathic Medicine, Ohio University, Athens, Ohio 45701, USA
| | | | | | | |
Collapse
|
46
|
Faulhaber-Walter R, Jou W, Mizel D, Li L, Zhang J, Kim SM, Huang Y, Chen M, Briggs JP, Gavrilova O, Schnermann JB. Impaired glucose tolerance in the absence of adenosine A1 receptor signaling. Diabetes 2011; 60:2578-87. [PMID: 21831968 PMCID: PMC3178298 DOI: 10.2337/db11-0058] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
OBJECTIVE The role of adenosine (ADO) in the regulation of glucose homeostasis is not clear. In the current study, we used A1-ADO receptor (A1AR)-deficient mice to investigate the role of ADO/A1AR signaling for glucose homeostasis. RESEARCH DESIGN AND METHODS After weaning, A1AR(-/-) and wild-type mice received either a standard diet (12 kcal% fat) or high-fat diet (HFD; 45 kcal% fat). Body weight, fasting plasma glucose, plasma insulin, and intraperitoneal glucose tolerance tests were performed in 8-week-old mice and again after 12-20 weeks of subsequent observation. Body composition was quantified by magnetic resonance imaging and epididymal fat-pad weights. Glucose metabolism was investigated by hyperinsulinemic-euglycemic clamp studies. To describe pathophysiological mechanisms, adipokines and Akt phosphorylation were measured. RESULTS A1AR(-/-) mice were significantly heavier than wild-type mice because of an increased fat mass. Fasting plasma glucose and insulin were significantly higher in A1AR(-/-) mice after weaning and remained higher in adulthood. An intraperitoneal glucose challenge disclosed a significantly slower glucose clearance in A1AR(-/-) mice. An HFD enhanced this phenotype in A1AR(-/-) mice and unmasked a dysfunctional insulin secretory mechanism. Insulin sensitivity was significantly impaired in A1AR(-/-) mice on the standard diet shortly after weaning. Clamp studies detected a significant decrease of net glucose uptake in A1AR(-/-) mice and a reduced glucose uptake in muscle and white adipose tissue. Effects were not triggered by leptin deficiency but involved a decreased Akt phosphorylation. CONCLUSIONS ADO/A1AR signaling contributes importantly to insulin-controlled glucose homeostasis and insulin sensitivity in C57BL/6 mice and is involved in the metabolic regulation of adipose tissue.
Collapse
Affiliation(s)
- Robert Faulhaber-Walter
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Ding J, Kopchick JJ. Plasma biomarkers of mouse aging. AGE (DORDRECHT, NETHERLANDS) 2011; 33:291-307. [PMID: 20842533 PMCID: PMC3168609 DOI: 10.1007/s11357-010-9179-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2010] [Accepted: 08/25/2010] [Indexed: 05/24/2023]
Abstract
Normal aging is accompanied by a series of physiological changes such as gray hair, cataracts, reduced immunity, and increased susceptibility to disease. To identify novel biomarkers of normal aging, we analyzed plasma proteins of male mice longitudinally from 2 to 19 months of age. Plasma proteins were analyzed by two-dimensional gel electrophoresis and identified using mass spectrometry (MS), MS/MS and liquid chromatography MS/MS. We found that many plasma proteins exist as multiple isoforms with different masses and/or charges. Thirty-nine protein spots (corresponding to six distinct proteins) have been identified, 13 of which exhibited significant changes with age. For example, several proteins increased significantly during aging including one isoform of transthyretin, two isoforms of haptoglobin, and three isoforms of immunoglobulin kappa chain. Conversely, several proteins decreased significantly during aging including peroxiredoxin-2, serum amyloid protein A-1, and five isoforms of albumin. Identification of these proteins provides new biomarkers of normal aging in mice. If validated in humans, these biomarkers may facilitate therapeutic interventions to identify premature aging, delay aging, and/or improve healthspan of the elderly.
Collapse
Affiliation(s)
- Juan Ding
- Edison Biotechnology Institute, Ohio University, 1 Water Tower Drive, The Ridges, Athens, OH 45701 USA
| | - John J. Kopchick
- Edison Biotechnology Institute, Ohio University, 1 Water Tower Drive, The Ridges, Athens, OH 45701 USA
- Department of Biomedical Sciences, College of Osteopathic Medicine, Ohio University, Athens, OH 45701 USA
| |
Collapse
|
48
|
Berryman DE, List EO, Sackmann-Sala L, Lubbers E, Munn R, Kopchick JJ. Growth hormone and adipose tissue: beyond the adipocyte. Growth Horm IGF Res 2011; 21:113-123. [PMID: 21470887 PMCID: PMC3112270 DOI: 10.1016/j.ghir.2011.03.002] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2011] [Accepted: 03/03/2011] [Indexed: 01/04/2023]
Abstract
The last two decades have seen resurgence in research focused on adipose tissue. In part, the enhanced interest stems from an alarming increase in obesity rates worldwide. However, an understanding that this once simple tissue is significantly more intricate and interactive than previously realized has fostered additional attention. While few would argue that growth hormone (GH) radically alters fat mass, newer findings revealing the complexity of adipose tissue requires that GH's influence on this tissue be reexamined. Therefore, the objective of this review is to describe the more recent understanding of adipose tissue and to summarize our current knowledge of how GH may influence and contribute to these newer complexities of this tissue with special focus on the available data from mice with altered GH action.
Collapse
Affiliation(s)
- Darlene E Berryman
- School of Applied Health Sciences and Wellness College of Health Sciences and Human Performance, Ohio University, Athens, OH 45701, United States.
| | | | | | | | | | | |
Collapse
|
49
|
Plasma proteomic profiles of bovine growth hormone transgenic mice as they age. Transgenic Res 2011; 20:1305-20. [PMID: 21365322 DOI: 10.1007/s11248-011-9499-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2010] [Accepted: 02/20/2011] [Indexed: 12/17/2022]
Abstract
Attenuation of the growth hormone (GH)/insulin-like growth factor-1 (IGF-1) axis results in extended lifespan in many organisms including mice. Conversely, GH transgenic mice have excess GH action and die prematurely. We have studied bovine (b) GH transgenic mice (n = 9) and their wild type (WT) littermates (n = 8) longitudinally and have determined several age-related changes. Compared to WT mice, bGH mice lost fat mass, became hypoglycemic and had lower insulin levels at older ages despite being hyperinsulinemic when young. To examine plasma protein differences in bGH mice relative to controls, samples at 2, 4, 8, 12 and 16 months of age were analyzed by two-dimensional gel electrophoresis followed by identification using mass spectrometry. We found several differences in plasma proteins of bGH mice compared to controls, including increased apolipoprotein E (five isoforms), haptoglobin (four isoforms) and mannose-binding protein-C (one out of three isoforms), and decreased transthyretin (six isoforms). In addition, clusterin (two out of six isoforms) and haptoglobin (four isoforms) were up-regulated in bGH mice as a function of age. Finally, alpha-2 macroglobulin (seven isoforms) was altered in an isoform-specific manner with two isoforms increased and two decreased in bGH mouse plasma compared to controls. In conclusion, identification of these proteins suggests that bGH mice exhibit an increased inflammatory state with an adverse lipid profile, possibly contributing to their diminished life expectancy. Also, these newly discovered plasma proteins may be indicative or 'biomarkers' of a shortened lifespan.
Collapse
|
50
|
Passariello CL, Gruodytė R, Hiio K, Mäestu J, Jürimäe J, Saar M, Cicchella A, Stefanelli C, Jürimäe T. ADIPOQ SNP45 associated with lean body mass in physically active normal weight adolescent girls. Am J Hum Biol 2011; 22:813-8. [PMID: 20878967 DOI: 10.1002/ajhb.21087] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
UNLABELLED Recently, two single nucleotide polymorphisms at position 45 and 276 on the adiponectin gene (ADIPOQ) have been recognized as determinants of total adiponectin levels, insulin resistance, and risk for diabetes in various obese populations. OBJECTIVES The aim of this study was to determine whether these two polymorphisms are indeed determinants in the development of metabolic disorders or whether they are secondary to other confounding factors. METHODS To do so, we have selected 170 physically active adolescent girls (mean age, 14.03 ± 1.5 years and mean body mass index, 19.98 ± 2.5 kg/m²) devoid of any metabolic diseases or confounding factors, to better attribute any findings to genotype effects. Concentration of adiponectin, insulin, and glucose were determined from blood samples with appropriate kits. Body fat parameters were evaluated with dual-energy X-ray absorptiometry, and genotype was analyzed with DNA extracted from whole blood samples followed by polymerase chain reaction and electrophoresis to separate alleles. RESULTS Neither single nucleotide polymorphism +45T/G nor +276G/T was related to homeostasis model assessment index or adiponectin levels; however, the presence of the G allele on site 45 favored a significant decrease in lean body mass compared with those who were T homozygous (TG:36.90/TT:41.07 kg, P < 0.05). CONCLUSIONS Results suggest that the reported increase in the risk of diabetes in subjects that were G allele carriers at site 45 in obese populations compared with normal-weight populations can be linked instead to a change in muscle mass or the muscle itself present in this genotype group.
Collapse
Affiliation(s)
- Catherine L Passariello
- Faculty of Exercise and Sport Sciences, Department of Biochemistry G. Moruzzi, University of Bologna, Bologna, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|