1
|
Huang J, Siyar S, Sharma R, Herrig I, Wise L, Aidt S, List E, Kopchick JJ, Puri V, Lee KY. Adipocyte Subpopulations Mediate Growth Hormone-induced Lipolysis and Glucose Tolerance in Male Mice. Endocrinology 2023; 164:bqad151. [PMID: 37897489 DOI: 10.1210/endocr/bqad151] [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: 05/30/2023] [Revised: 10/03/2023] [Accepted: 10/09/2023] [Indexed: 10/30/2023]
Abstract
In adipose tissue, growth hormone (GH) stimulates lipolysis, leading to an increase in plasma free fatty acid levels and a reduction in insulin sensitivity. In our previous studies, we have found that GH increases lipolysis by reducing peroxisome proliferator-activated receptor γ (PPARγ) transcription activity, leading to a reduction of tat-specific protein 27 (FSP27, also known as CIDEC) expression. In previous studies, our laboratory uncovered 3 developmentally distinct subpopulations of white adipocytes. In this manuscript, we show that one of the subpopulations, termed type 2 adipocytes, has increased GH-induced signaling and lipolysis compared to other adipocyte subtypes. To assess the physiological role of GH-mediated lipolysis mediated by this adipocyte subpopulation, we specifically expressed human FSP27 (hFSP27) transgene in type 2 adipocytes (type2Ad-hFSP27tg mice). Systemically, male type2Ad-hFSP27tg mice displayed reduced serum glycerol release and nonesterified fatty acids levels after acute GH treatment, and improvement in acute, but not chronic, GH-induced glucose intolerance. Furthermore, we demonstrate that type2Ad-hFSP27tg mice displayed improved hepatic insulin signaling. Taken together, these results indicate that this adipocyte subpopulation is a critical regulator of the GH-mediated lipolytic and metabolic response. Thus, further investigation of adipocyte subpopulations may provide novel treatment strategies to regulate GH-induced glucose intolerance in patients with growth and metabolic disorders.
Collapse
Affiliation(s)
- Jun Huang
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Sohana Siyar
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Rita Sharma
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Isabella Herrig
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Lauren Wise
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Spencer Aidt
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Edward List
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
- The Diabetes Institute, Ohio University, Athens, OH 45701, USA
| | - John J Kopchick
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
- The Diabetes Institute, Ohio University, Athens, OH 45701, USA
| | - Vishwajeet Puri
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
- The Diabetes Institute, Ohio University, Athens, OH 45701, USA
| | - Kevin Y Lee
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
- The Diabetes Institute, Ohio University, Athens, OH 45701, USA
| |
Collapse
|
2
|
The role of exercise and hypoxia on glucose transport and regulation. Eur J Appl Physiol 2023; 123:1147-1165. [PMID: 36690907 DOI: 10.1007/s00421-023-05135-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 01/06/2023] [Indexed: 01/25/2023]
Abstract
Muscle glucose transport activity increases with an acute bout of exercise, a process that is accomplished by the translocation of glucose transporters to the plasma membrane. This process remains intact in the skeletal muscle of individuals with insulin resistance and type 2 diabetes mellitus (T2DM). Exercise training is, therefore, an important cornerstone in the management of individuals with T2DM. However, the acute systemic glucose responses to carbohydrate ingestion are often augmented during the early recovery period from exercise, despite increased glucose uptake into skeletal muscle. Accordingly, the first aim of this review is to summarize the knowledge associated with insulin action and glucose uptake in skeletal muscle and apply these to explain the disparate responses between systemic and localized glucose responses post-exercise. Herein, the importance of muscle glycogen depletion and the key glucoregulatory hormones will be discussed. Glucose uptake can also be stimulated independently by hypoxia; therefore, hypoxic training presents as an emerging method for enhancing the effects of exercise on glucose regulation. Thus, the second aim of this review is to discuss the potential for systemic hypoxia to enhance the effects of exercise on glucose regulation.
Collapse
|
3
|
Abstract
PURPOSE OF REVIEW Canonical growth hormone (GH)-dependent signaling is essential for growth and counterregulatory responses to hypoglycemia, but also may contribute to glucose homeostasis (even in the absence of hypoglycemia) via its impact on metabolism of carbohydrates, lipids and proteins, body composition, and cardiovascular risk profile. The aim of this review is to summarize recent data implicating GH action in metabolic control, including both IGF-1-dependent and -independent pathways, and its potential role as target for T2D therapy. RECENT FINDINGS Experimental blockade of the GHR can modulate glucose metabolism. Moreover, the soluble form of the GH receptor (GHR, or GHBP) was recently identified as a mediator of improvement in glycemic control in patients with T2D randomized to bariatric surgery vs. medical therapy. Reductions in GHR were accompanied by increases in plasma GH, but unchanged levels of both total and free IGF-1. Likewise, hepatic GHR expression is reduced following both RYGB and VSG in rodents. Emerging data indicate that GH signaling is important for regulation of long-term glucose metabolism in T2D. Future studies will be required to dissect tissue-specific GH signaling and sensitivity and their contributions to systemic glucose metabolism.
Collapse
Affiliation(s)
- Xuehong Dong
- Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Endocrinology, Diabetes & Metabolism, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lei Su
- Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Geriatrics, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Mary-Elizabeth Patti
- Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
4
|
Arlien-Søborg MC, Dal J, Madsen MA, Høgild ML, Hjelholt AJ, Pedersen SB, Møller N, Jessen N, Jørgensen JOL. Reversible insulin resistance in muscle and fat unrelated to the metabolic syndrome in patients with acromegaly. EBioMedicine 2021; 75:103763. [PMID: 34929488 PMCID: PMC8688588 DOI: 10.1016/j.ebiom.2021.103763] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/22/2021] [Accepted: 12/03/2021] [Indexed: 10/27/2022] Open
Abstract
BACKGROUND Patients with active acromegaly exhibit insulin resistance despite a lean phenotype whereas controlled disease improves insulin sensitivity and increases fat mass. The mechanisms underlying this paradox remain elusive, but growth hormone (GH)-induced lipolysis plays a central role. The aim of the study was to investigative the molecular mechanisms of insulin resistance dissociated from obesity in patients with acromegaly. METHODS In a prospective study, twenty-one patients with newly diagnosed acromegaly were studied at diagnosis and after disease control obtained by either surgery alone (n=10) or somatostatin analogue (SA) treatment (n=11) with assessment of body composition (DXA scan), whole body and tissue-specific insulin sensitivity and GH and insulin signalling in adipose tissue and skeletal muscle. FINDINGS Disease control of acromegaly significantly reduced lean body mass (p<0.001) and increased fat mass (p<0.001). At diagnosis, GH signalling (pSTAT5) was constitutively activated in fat and enhanced expression of GH-regulated genes (CISH and IGF-I) were detected in muscle and fat. Insulin sensitivity in skeletal muscle, liver and adipose tissue increased after disease control regardless of treatment modality. This was associated with enhanced insulin signalling in both muscle and fat including downregulation of phosphatase and tensin homolog (PTEN) together with reduced signalling of GH and lipolytic activators in fat. INTERPRETATION In conclusion, the study support that uncontrolled lipolysis is a major feature of insulin resistance in active acromegaly, and is characterized by upregulation of PTEN and suppression of insulin signalling in both muscle and fat. FUNDING This work was supported by a grant from the Independent Research Fund, Denmark (7016-00303A) and from the Alfred Benzon Foundation, Denmark.
Collapse
Affiliation(s)
- Mai C Arlien-Søborg
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Denmark; Medical Research Laboratory, Department of Clinical Medicine, Aarhus University Hospital, Denmark.
| | - Jakob Dal
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Denmark; Department of Endocrinology, Aalborg University Hospital, Denmark; Steno Diabetes Centre North, Aalborg University Hospital, Aalborg, Denmark
| | - Michael Alle Madsen
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Denmark; Department of Nuclear Medicine & PET Centre, Aarhus University Hospital, Denmark
| | - Morten Lyng Høgild
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Denmark; Medical Research Laboratory, Department of Clinical Medicine, Aarhus University Hospital, Denmark
| | - Astrid Johannesson Hjelholt
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Denmark; Medical Research Laboratory, Department of Clinical Medicine, Aarhus University Hospital, Denmark
| | | | - Niels Møller
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Denmark; Medical Research Laboratory, Department of Clinical Medicine, Aarhus University Hospital, Denmark
| | - Niels Jessen
- Steno Diabetes Centre, Aarhus, Denmark; Department of Clinical Pharmacology, University of Aarhus, Aarhus, Denmark; Department of Biomedicine, Aarhus University, Denmark
| | - Jens O L Jørgensen
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Denmark; Medical Research Laboratory, Department of Clinical Medicine, Aarhus University Hospital, Denmark
| |
Collapse
|
5
|
Sharma R, Kopchick JJ, Puri V, Sharma VM. Effect of growth hormone on insulin signaling. Mol Cell Endocrinol 2020; 518:111038. [PMID: 32966863 PMCID: PMC7606590 DOI: 10.1016/j.mce.2020.111038] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/31/2020] [Accepted: 09/17/2020] [Indexed: 12/21/2022]
Abstract
Growth hormone (GH) is a pleiotropic hormone that coordinates an array of physiological processes, including effects on bone, muscle, and fat, ultimately resulting in growth. Metabolically, GH promotes anabolic action in most tissues except adipose, where its catabolic action causes the breakdown of stored triglycerides into free fatty acids (FFA). GH antagonizes insulin action via various molecular pathways. Chronic GH secretion suppresses the anti-lipolytic action of insulin and increases FFA flux into the systemic circulation; thus, promoting lipotoxicity, which causes pathophysiological problems, including insulin resistance. In this review, we will provide an update on GH-stimulated adipose lipolysis and its consequences on insulin signaling in liver, skeletal muscle, and adipose tissue. Furthermore, we will discuss the mechanisms that contribute to the diabetogenic action of GH.
Collapse
Affiliation(s)
- Rita Sharma
- Department of Biomedical Sciences, Ohio University, Athens, OH, 45701, USA
| | - John J Kopchick
- Department of Biomedical Sciences, Ohio University, Athens, OH, 45701, USA; Edison Biotechnology Institute, Ohio University, Athens, OH, 45701, USA; Diabetes Institute, Ohio University, Athens, OH, 45701, USA
| | - Vishwajeet Puri
- Department of Biomedical Sciences, Ohio University, Athens, OH, 45701, USA; Diabetes Institute, Ohio University, Athens, OH, 45701, USA
| | - Vishva M Sharma
- Department of Biomedical Sciences, Ohio University, Athens, OH, 45701, USA; Diabetes Institute, Ohio University, Athens, OH, 45701, USA.
| |
Collapse
|
6
|
Hjelholt AJ, Charidemou E, Griffin JL, Pedersen SB, Gudiksen A, Pilegaard H, Jessen N, Møller N, Jørgensen JOL. Insulin resistance induced by growth hormone is linked to lipolysis and associated with suppressed pyruvate dehydrogenase activity in skeletal muscle: a 2 × 2 factorial, randomised, crossover study in human individuals. Diabetologia 2020; 63:2641-2653. [PMID: 32945898 DOI: 10.1007/s00125-020-05262-w] [Citation(s) in RCA: 10] [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: 06/08/2020] [Accepted: 07/15/2020] [Indexed: 02/07/2023]
Abstract
AIMS/HYPOTHESIS Growth hormone (GH) causes insulin resistance that is linked to lipolysis, but the underlying mechanisms are unclear. We investigated if GH-induced insulin resistance in skeletal muscle involves accumulation of diacylglycerol (DAG) and ceramide as well as impaired insulin signalling, or substrate competition between fatty acids and glucose. METHODS Nine GH-deficient male participants were randomised and examined in a 2 × 2 factorial design with and without administration of GH and acipimox (an anti-lipolytic compound). As-treated analyses were performed, wherefore data from three visits from two patients were excluded due to incorrect GH administration. The primary outcome was insulin sensitivity, expressed as the AUC of the glucose infusion rate (GIRAUC), and furthermore, the levels of DAGs and ceramides, insulin signalling and the activity of the active form of pyruvate dehydrogenase (PDHa) were assessed in skeletal muscle biopsies obtained in the basal state and during a hyperinsulinaemic-euglycaemic clamp (HEC). RESULTS Co-administration of acipimox completely suppressed the GH-induced elevation in serum levels of NEFA (GH versus GH+acipimox, p < 0.0001) and abrogated GH-induced insulin resistance (mean GIRAUC [95% CI] [mg min-1 kg-1] during the HEC: control, 595 [493, 718]; GH, 468 [382, 573]; GH+acipimox, 654 [539, 794]; acipimox, 754 [618, 921]; GH vs GH+acipimox: p = 0.004). GH did not significantly change either the accumulation of DAGs and ceramides or insulin signalling in skeletal muscle, but GH antagonised the insulin-stimulated increase in PDHa activity (mean ± SEM [% from the basal state to the HEC]: control, 47 ± 19; GH, -15 ± 21; GH+acipimox, 3 ± 21; acipimox, 57 ± 22; main effect: p = 0.02). CONCLUSIONS/INTERPRETATION GH-induced insulin resistance in skeletal muscle is: (1) causally linked to lipolysis; (2) not associated with either accumulation of DAGs and ceramides or impaired insulin signalling; (3) likely to involve substrate competition between glucose and lipid intermediates. TRIAL REGISTRATION ClinicalTrials.gov NCT02782208 FUNDING: The work was supported by the Grant for Growth Innovation (GGI), which was funded by Merck KGaA, Darmstadt, Germany. Graphical abstract.
Collapse
Affiliation(s)
- Astrid J Hjelholt
- Medical Research Laboratory, Department of Clinical Medicine, Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus N, Denmark.
- Department of Clinical Pharmacology, Aarhus University Hospital, Aarhus C, Denmark.
| | - Evelina Charidemou
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, UK
| | - Julian L Griffin
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, UK
| | - Steen B Pedersen
- Medical Research Laboratory, Department of Clinical Medicine, Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus N, Denmark
| | - Anders Gudiksen
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Henriette Pilegaard
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Niels Jessen
- Department of Clinical Pharmacology, Aarhus University Hospital, Aarhus C, Denmark
- Steno Diabetes Centre Aarhus, Aarhus University Hospital, Aarhus N, Denmark
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | - Niels Møller
- Medical Research Laboratory, Department of Clinical Medicine, Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus N, Denmark
| | - Jens O L Jørgensen
- Medical Research Laboratory, Department of Clinical Medicine, Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus N, Denmark
| |
Collapse
|
7
|
Kopchick JJ, Berryman DE, Puri V, Lee KY, Jorgensen JOL. The effects of growth hormone on adipose tissue: old observations, new mechanisms. Nat Rev Endocrinol 2020; 16:135-146. [PMID: 31780780 PMCID: PMC7180987 DOI: 10.1038/s41574-019-0280-9] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/16/2019] [Indexed: 12/18/2022]
Abstract
The ability of growth hormone (GH) to induce adipose tissue lipolysis has been known for over five decades; however, the molecular mechanisms that mediate this effect and the ability of GH to inhibit insulin-stimulated glucose uptake have scarcely been documented. In this same time frame, our understanding of adipose tissue has evolved to reveal a complex structure with distinct types of adipocyte, depot-specific differences, a biologically significant extracellular matrix and important endocrine properties mediated by adipokines. All these aforementioned features, in turn, can influence lipolysis. In this Review, we provide a historical and current overview of the lipolytic effect of GH in humans, mice and cultured cells. More globally, we explain lipolysis in terms of GH-induced intracellular signalling and its effect on obesity, insulin resistance and lipotoxicity. In this regard, findings that define molecular mechanisms by which GH induces lipolysis are described. Finally, data are presented for the differential effect of GH on specific adipose tissue depots and on distinct classes of metabolically active adipocytes. Together, these cellular, animal and human studies reveal novel cellular phenotypes and molecular pathways regulating the metabolic effects of GH on adipose tissue.
Collapse
Affiliation(s)
- John J Kopchick
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA.
- The Diabetes Institute, Ohio University Heritage College of Osteopathic Medicine, Athens, OH, USA.
- Department of Biomedical Sciences, Ohio University College of Osteopathic Medicine, Athens, OH, USA.
| | - Darlene E Berryman
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- The Diabetes Institute, Ohio University Heritage College of Osteopathic Medicine, Athens, OH, USA
- Department of Biomedical Sciences, Ohio University College of Osteopathic Medicine, Athens, OH, USA
| | - Vishwajeet Puri
- The Diabetes Institute, Ohio University Heritage College of Osteopathic Medicine, Athens, OH, USA
- Department of Biomedical Sciences, Ohio University College of Osteopathic Medicine, Athens, OH, USA
| | - Kevin Y Lee
- The Diabetes Institute, Ohio University Heritage College of Osteopathic Medicine, Athens, OH, USA
- Department of Biomedical Sciences, Ohio University College of Osteopathic Medicine, Athens, OH, USA
| | - Jens O L Jorgensen
- Department of Endocrinology and Diabetes, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
8
|
Forrest L, Sedmak C, Sikder S, Grewal S, Harman SM, Blackman MR, Muniyappa R. Effects of growth hormone on hepatic insulin sensitivity and glucose effectiveness in healthy older adults. Endocrine 2019; 63:497-506. [PMID: 30617738 PMCID: PMC6420854 DOI: 10.1007/s12020-018-01834-4] [Citation(s) in RCA: 6] [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: 11/26/2018] [Accepted: 12/23/2018] [Indexed: 01/06/2023]
Abstract
PURPOSE Growth hormone (GH) replacement decreases insulin sensitivity in healthy individuals. However, the effects of GH on organ-specific insulin sensitivity and glucose effectiveness are not well characterized. The purpose of this study was to evaluate the effects of GH administration for 26 weeks on muscle and hepatic insulin sensitivity and glucose effectiveness in healthy older individuals. METHODS This report is from a 26-week randomized, double-blind, placebo-controlled parallel-group trial in healthy, ambulatory, community-dwelling older women and men. We compared surrogate indices of insulin sensitivity [quantitative insulin-sensitivity check index (QUICKI), muscle insulin sensitivity index (MISI), hepatic insulin resistance index (HIRI)] and glucose effectiveness [oral glucose effectiveness index (oGE)] derived from oral glucose tolerance tests (OGTTs) in subjects before and after 26 weeks of administration of GH (n = 17) or placebo (n = 15) as an exploratory outcome. RESULTS GH administration for 26 weeks significantly increased fasting insulin concentrations and HIRI but did not significantly change MISI or oGE compared to placebo. CONCLUSIONS GH administration for 26 weeks in healthy older subjects impairs insulin sensitivity in the liver but not skeletal muscle and does not alter glucose effectiveness.
Collapse
Affiliation(s)
- Lala Forrest
- Clinical Endocrine Section, Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Caroline Sedmak
- Clinical Endocrine Section, Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Shanaz Sikder
- Clinical Endocrine Section, Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Shivraj Grewal
- Clinical Endocrine Section, Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - S Mitchell Harman
- Endocrinology Section, Department of Medicine, Phoenix VA Health Care System, Phoenix, AZ, USA
| | - Marc R Blackman
- Research Service, Veterans Affairs Medical Center, Washington, DC, USA
- Department of Medicine, Georgetown University School of Medicine, Washington, DC, 20007, USA
- Department of Medicine, George Washington University School of Medicine, Washington, DC, 20052, USA
| | - Ranganath Muniyappa
- Clinical Endocrine Section, Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
9
|
Høgild ML, Bak AM, Pedersen SB, Rungby J, Frystyk J, Møller N, Jessen N, Jørgensen JOL. Growth hormone signaling and action in obese versus lean human subjects. Am J Physiol Endocrinol Metab 2019; 316:E333-E344. [PMID: 30576246 DOI: 10.1152/ajpendo.00431.2018] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Growth hormone (GH) levels are blunted in obesity, but it is not known whether this relates to altered GH sensitivity and whether this influences the metabolic adaptation to fasting. Therefore, we investigated the effect of obesity on GH signal transduction and fasting-induced changes in GH action. Nine obese (BMI 35.7 kg/m2) and nine lean (BMI 21.5 kg/m2) men were studied in a randomized crossover design with 1) an intravenous GH bolus, 2) an intravenous saline bolus, and 3) 72 h of fasting. Insulin sensitivity (hyperinsulinemic, euglycemic clamp) and substrate metabolism (glucose tracer and indirect calorimetry) were measured in studies 1 and 2. In vivo GH signaling was assessed in muscle and fat biopsies. GH pharmacokinetics did not differ between obese and lean subjects, but endogenous GH levels were reduced in obesity. GH signaling (STAT5b phosphorylation and CISH mRNA transcription), and GH action (induction of lipolysis and peripheral insulin resistance) were similar in the two groups, but a GH-induced insulin antagonistic effect on endogenous glucose production only occurred in the obese. Fasting-induced IGF-I reduction was completely abrogated in obese subjects despite a comparable relative increase in GH levels (ΔIGF-I: lean, -66 ± 10 vs. obese, 27 ± 16 µg/l; P < 0.01; ΔGH: lean, 647 ± 280 vs. obese, 544 ± 220%; P = 0.76]. We conclude that 1) GH signaling is normal in obesity, 2) in the obese state, the preservation of IGF-I with fasting and the augmented GH-induced central insulin resistance indicate increased hepatic GH sensitivity, 3) blunted GH levels in obesity may protect against insulin resistance without compromising IGF-I status.
Collapse
Affiliation(s)
- Morten Lyng Høgild
- Medical Research Laboratory, Department of Clinical Medicine, Aarhus University Hospital , Denmark
- Department of Clinical Medicine, Aarhus University , Denmark
| | - Ann Mosegaard Bak
- Medical Research Laboratory, Department of Clinical Medicine, Aarhus University Hospital , Denmark
- Department of Clinical Medicine, Aarhus University , Denmark
| | - Steen Bønløkke Pedersen
- Medical Research Laboratory, Department of Clinical Medicine, Aarhus University Hospital , Denmark
- Department of Clinical Medicine, Aarhus University , Denmark
| | - Jørgen Rungby
- Department of Biomedicine, Aarhus University , Denmark
| | - Jan Frystyk
- Medical Research Laboratory, Department of Clinical Medicine, Aarhus University Hospital , Denmark
- Department of Clinical Medicine, Aarhus University , Denmark
| | - Niels Møller
- Medical Research Laboratory, Department of Clinical Medicine, Aarhus University Hospital , Denmark
- Department of Clinical Medicine, Aarhus University , Denmark
| | - Niels Jessen
- Department of Clinical Medicine, Aarhus University , Denmark
- Department of Biomedicine, Aarhus University , Denmark
- Department of Clinical Pharmacology, Aarhus University Hospital , Denmark
| | - Jens O L Jørgensen
- Medical Research Laboratory, Department of Clinical Medicine, Aarhus University Hospital , Denmark
- Department of Clinical Medicine, Aarhus University , Denmark
| |
Collapse
|
10
|
Vila G, Jørgensen JOL, Luger A, Stalla GK. Insulin Resistance in Patients With Acromegaly. Front Endocrinol (Lausanne) 2019; 10:509. [PMID: 31417493 PMCID: PMC6683662 DOI: 10.3389/fendo.2019.00509] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 07/12/2019] [Indexed: 12/20/2022] Open
Abstract
Acromegaly is characterized by chronic overproduction of growth hormone (GH) that leads to insulin resistance, glucose intolerance and, ultimately, diabetes. The GH-induced sustained stimulation of lipolysis plays a major role not only in the development of insulin resistance and prediabetes/diabetes, but also in the reduction of lipid accumulation, making acromegaly a unique case of severe insulin resistance in the presence of reduced body fat. In the present review, we elucidate the effects of GH hypersecretion on metabolic organs, describing the pathophysiology of impaired glucose tolerance in acromegaly, as well as the impact of acromegaly-specific therapies on glucose metabolism. In addition, we highlight the role of insulin resistance in the development of acromegaly-associated complications such as hypertension, cardiac disease, sleep apnea, polycystic ovaries, bone disease, and cancer. Taken together, insulin resistance is an important metabolic hallmark of acromegaly, which is strongly related to disease activity, the development of comorbidities, and might even impact the response to drugs used in the treatment of acromegaly.
Collapse
Affiliation(s)
- Greisa Vila
- Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Jens Otto L. Jørgensen
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Anton Luger
- Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Günter K. Stalla
- Max Planck Institute of Psychiatry, Munich, Germany
- *Correspondence: Günter K. Stalla ;
| |
Collapse
|
11
|
Sharma VM, Vestergaard ET, Jessen N, Kolind-Thomsen P, Nellemann B, Nielsen TS, Vendelbo MH, Møller N, Sharma R, Lee KY, Kopchick JJ, Jørgensen JOL, Puri V. Growth hormone acts along the PPARγ-FSP27 axis to stimulate lipolysis in human adipocytes. Am J Physiol Endocrinol Metab 2019; 316:E34-E42. [PMID: 30325658 PMCID: PMC6417689 DOI: 10.1152/ajpendo.00129.2018] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 10/01/2018] [Accepted: 10/03/2018] [Indexed: 12/22/2022]
Abstract
The lipolytic effects of growth hormone (GH) have been known for half a century and play an important physiological role for substrate metabolism during fasting. In addition, sustained GH-induced lipolysis is causally linked to insulin resistance. However, the underlying molecular mechanisms remain elusive. In the present study, we obtained experimental data in human subjects and used human adipose-derived stromal vascular cells (hADSCs) as a model system to elucidate GH-triggered molecular signaling that stimulates adipose tissue lipolysis and insulin resistance in human adipocytes. We discovered that GH downregulates the expression of fat-specific protein (FSP27), a negative regulator of lipolysis, by impairing the transcriptional ability of the master transcriptional regulator, peroxisome proliferator-activated receptor-γ (PPARγ) via MEK/ERK activation. Ultimately, GH treatment promotes phosphorylation of PPARγ at Ser273 and causes its translocation from nucleus to the cytosol. Surprisingly, FSP27 overexpression inhibited PPARγ Ser273 phosphorylation and promoted its nuclear retention. GH antagonist treatment had similar effects. Our study identifies a novel signaling mechanism by which GH transcriptionally induces lipolysis via the MEK/ERK pathway that acts along PPARγ-FSP27 in human adipose tissue.
Collapse
Affiliation(s)
- Vishva M Sharma
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University , Athens, Ohio
- The Diabetes Institute, Ohio University , Athens, Ohio
| | - Esben Thyssen Vestergaard
- Medical Research Laboratory, Aarhus University , Aarhus , Denmark
- Department of Pediatrics, Randers Regional Hospital, Randers, Denmark
| | - Niels Jessen
- Medical Research Laboratory, Aarhus University , Aarhus , Denmark
- Research Laboratory for Biochemical Pathology, Aarhus University Hospital , Aarhus , Denmark
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital , Aarhus , Denmark
| | - Peter Kolind-Thomsen
- Medical Research Laboratory, Aarhus University , Aarhus , Denmark
- Research Laboratory for Biochemical Pathology, Aarhus University Hospital , Aarhus , Denmark
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital , Aarhus , Denmark
| | | | - Thomas S Nielsen
- Medical Research Laboratory, Aarhus University , Aarhus , Denmark
- Faculty of Health and Medical Sciences, The Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen , Copenhagen , Denmark
| | - Mikkel Holm Vendelbo
- Medical Research Laboratory, Aarhus University , Aarhus , Denmark
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital , Aarhus , Denmark
| | - Niels Møller
- Medical Research Laboratory, Aarhus University , Aarhus , Denmark
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital , Aarhus , Denmark
| | - Rita Sharma
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University , Athens, Ohio
- The Diabetes Institute, Ohio University , Athens, Ohio
| | - Kevin Y Lee
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University , Athens, Ohio
- The Diabetes Institute, Ohio University , Athens, Ohio
| | - John J Kopchick
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University , Athens, Ohio
- The Diabetes Institute, Ohio University , Athens, Ohio
- Edison Biotechnology Institute, Ohio University , Athens, Ohio
| | - Jens Otto Lunde Jørgensen
- Medical Research Laboratory, Aarhus University , Aarhus , Denmark
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital , Aarhus , Denmark
| | - Vishwajeet Puri
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University , Athens, Ohio
- The Diabetes Institute, Ohio University , Athens, Ohio
| |
Collapse
|
12
|
Sharma R, Luong Q, Sharma VM, Harberson M, Harper B, Colborn A, Berryman DE, Jessen N, Jørgensen JOL, Kopchick JJ, Puri V, Lee KY. Growth hormone controls lipolysis by regulation of FSP27 expression. J Endocrinol 2018; 239:289-301. [PMID: 30400015 PMCID: PMC6226059 DOI: 10.1530/joe-18-0282] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 09/24/2018] [Indexed: 01/10/2023]
Abstract
Growth hormone (GH) has long been known to stimulate lipolysis and insulin resistance; however, the molecular mechanisms underlying these effects are unknown. In the present study, we demonstrate that GH acutely induces lipolysis in cultured adipocytes. This effect is secondary to the reduced expression of a negative regulator of lipolysis, fat-specific protein 27 (FSP27; aka Cidec) at both the mRNA and protein levels. These effects are mimicked in vivo as transgenic overexpression of GH leads to a reduction of FSP27 expression. Mechanistically, we show GH modulation of FSP27 expression is mediated through activation of both MEK/ERK- and STAT5-dependent intracellular signaling. These two molecular pathways interact to differentially manipulate peroxisome proliferator-activated receptor gamma activity (PPARγ) on the FSP27 promoter. Furthermore, overexpression of FSP27 is sufficient to fully suppress GH-induced lipolysis and insulin resistance in cultured adipocytes. Taken together, these data decipher a molecular mechanism by which GH acutely regulates lipolysis and insulin resistance in adipocytes.
Collapse
Affiliation(s)
- Rita Sharma
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH
- The Diabetes Institute, Ohio University, Athens, OH
| | - Quyen Luong
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH
- The Diabetes Institute, Ohio University, Athens, OH
| | - Vishva M. Sharma
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH
- The Diabetes Institute, Ohio University, Athens, OH
| | - Mitchell Harberson
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH
- The Diabetes Institute, Ohio University, Athens, OH
| | - Brian Harper
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH
- The Diabetes Institute, Ohio University, Athens, OH
| | - Andrew Colborn
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH
- The Diabetes Institute, Ohio University, Athens, OH
| | - Darlene E. Berryman
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH
- The Diabetes Institute, Ohio University, Athens, OH
| | - Niels Jessen
- Research Laboratory for Biochemical Pathology, Aarhus University Hospital, Aarhus, Denmark
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Biomedicine, Aarhus University, Denmark
| | - Jens Otto Lunde Jørgensen
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
- Medical Research Laboratory, Aarhus University, Aarhus, Denmark
| | - John J. Kopchick
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH
- The Diabetes Institute, Ohio University, Athens, OH
- Edison Biotechnology Institute, Ohio University, Athens, OH
| | - Vishwajeet Puri
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH
- The Diabetes Institute, Ohio University, Athens, OH
| | - Kevin Y. Lee
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH
- The Diabetes Institute, Ohio University, Athens, OH
| |
Collapse
|
13
|
Tang H, Yonemitsu I, Ikeda Y, Watakabe K, Shibata S, Hosomichi J, Ono T. Effects of unilateral nasal obstruction on the characteristics of jaw-closing muscles in growing rats. Angle Orthod 2018; 89:102-110. [PMID: 30221984 DOI: 10.2319/021918-132.1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
OBJECTIVES Mouth breathing caused by nasal obstruction (owing to abnormal pressure of masticatory muscles) affects craniofacial growth and development. The influence of unilateral nasal obstruction on jaw-closing muscles was investigated in rats to reveal one of the etiologic mechanisms. MATERIALS AND METHODS Forty 8-day-old male Wistar rats were used in this study. Experimental rats were subjected to left-sided nasal obstruction by burning the external nostril tissue at the age of 8 days. Pulse oxygen saturation was recorded each week. Morphologic changes were evaluated by staining with hematoxylin and eosin (to assess the cross-sectional area) and by adenosine triphosphatase activity staining (to assess the myosin heavy chain isoform composition). Immunohistochemical and reverse transcription quantitative real-time polymerase chain reaction analyses of tumor necrosis factor-α and glucose transporter 4 were carried out at 5 and 9 weeks of age. RESULTS The cross-sectional area of the jaw-closing muscles was lower in the experimental group at 9 weeks of age. The percentage of myosin heavy chain-2a in masseter muscles was increased in the experimental group compared with the control group. An increase in the tumor necrosis factor-α messenger RNA and protein levels and a decrease in the glucose transporter 4 messenger RNA and protein levels at 5 and 9 weeks of age in the jaw-closing muscles in the experimental group were noted. CONCLUSIONS Unilateral nasal obstruction could affect the morphology and contractile characteristics of jaw-closing muscles during growth in rats.
Collapse
|
14
|
Badawy AAB. Modulation of Tryptophan and Serotonin Metabolism as a Biochemical Basis of the Behavioral Effects of Use and Withdrawal of Androgenic-Anabolic Steroids and Other Image- and Performance-Enhancing Agents. Int J Tryptophan Res 2018; 11:1178646917753422. [PMID: 29487480 PMCID: PMC5821294 DOI: 10.1177/1178646917753422] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 10/29/2017] [Indexed: 01/01/2023] Open
Abstract
Modulation of tryptophan (Trp) metabolism may underpin the behavioral effects of androgenic-anabolic steroids (AAS) and associated image and performance enhancers. Euphoria, arousal, and decreased anxiety observed with moderate use and exercise may involve enhanced cerebral serotonin synthesis and function by increased release of albumin-bound Trp and estrogen-mediated liver Trp 2,3-dioxygenase (TDO) inhibition and enhancement of serotonin function. Aggression, anxiety, depression, personality disorders, and psychosis, observed on withdrawal of AAS or with use of large doses, can be caused by decreased serotonin synthesis due to TDO induction on withdrawal, excess Trp inhibiting the 2 enzymes of serotonin synthesis, and increased cerebral levels of neuroactive kynurenines. Exercise and excessive protein and branched-chain amino acid intakes may aggravate the effects of large AAS dosage. The hypothesis is testable in humans and experimental animals by measuring parameters of Trp metabolism and disposition and related metabolic processes.
Collapse
Affiliation(s)
- Abdulla A-B Badawy
- Cardiff School of Health Sciences, Cardiff Metropolitan University, Cardiff, UK
| |
Collapse
|
15
|
Dehkhoda F, Lee CMM, Medina J, Brooks AJ. The Growth Hormone Receptor: Mechanism of Receptor Activation, Cell Signaling, and Physiological Aspects. Front Endocrinol (Lausanne) 2018; 9:35. [PMID: 29487568 PMCID: PMC5816795 DOI: 10.3389/fendo.2018.00035] [Citation(s) in RCA: 167] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 01/29/2018] [Indexed: 01/02/2023] Open
Abstract
The growth hormone receptor (GHR), although most well known for regulating growth, has many other important biological functions including regulating metabolism and controlling physiological processes related to the hepatobiliary, cardiovascular, renal, gastrointestinal, and reproductive systems. In addition, growth hormone signaling is an important regulator of aging and plays a significant role in cancer development. Growth hormone activates the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) signaling pathway, and recent studies have provided a new understanding of the mechanism of JAK2 activation by growth hormone binding to its receptor. JAK2 activation is required for growth hormone-mediated activation of STAT1, STAT3, and STAT5, and the negative regulation of JAK-STAT signaling comprises an important step in the control of this signaling pathway. The GHR also activates the Src family kinase signaling pathway independent of JAK2. This review covers the molecular mechanisms of GHR activation and signal transduction as well as the physiological consequences of growth hormone signaling.
Collapse
Affiliation(s)
- Farhad Dehkhoda
- The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Christine M. M. Lee
- The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Johan Medina
- The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Andrew J. Brooks
- The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
16
|
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
|
17
|
Olarescu NC, Bollerslev J. The Impact of Adipose Tissue on Insulin Resistance in Acromegaly. Trends Endocrinol Metab 2016; 27:226-237. [PMID: 26948712 DOI: 10.1016/j.tem.2016.02.005] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Revised: 02/09/2016] [Accepted: 02/11/2016] [Indexed: 01/11/2023]
Abstract
Adipose tissue (AT) is recognized as key contributor to the systemic insulin resistance and overt diabetes seen in metabolic syndrome. Acromegaly is a disease characterized by excessive secretion of growth hormone (GH) and insulin-like growth factor I (IGF-I). GH is known both for its action on AT and for its detrimental effect on glucose metabolism and insulin signaling. In active acromegaly, while body fat deports are diminished, insulin resistance is increased. Early studies have demonstrated defects in insulin action, both at the hepatic and extrahepatic (i.e., muscle and fat) levels, in active disease. This review discusses recent data suggesting that AT inflammation, altered AT distribution, and impaired adipogenesis are potential mechanisms contributing to systemic insulin resistance in acromegaly.
Collapse
Affiliation(s)
- Nicoleta Cristina Olarescu
- Section of Specialized Endocrinology, Department of Endocrinology, Oslo University Hospital, Rikshospitalet, Oslo, Norway; Faculty of Medicine, University of Oslo, Norway.
| | - Jens Bollerslev
- Section of Specialized Endocrinology, Department of Endocrinology, Oslo University Hospital, Rikshospitalet, Oslo, Norway; Faculty of Medicine, University of Oslo, Norway
| |
Collapse
|
18
|
Dal J, List EO, Jørgensen JOL, Berryman DE. Glucose and Fat Metabolism in Acromegaly: From Mice Models to Patient Care. Neuroendocrinology 2016; 103:96-105. [PMID: 25925240 DOI: 10.1159/000430819] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 04/20/2015] [Indexed: 11/19/2022]
Abstract
Patients with active acromegaly are frequently insulin resistant, glucose intolerant, and at risk for developing overt type 2 diabetes. At the same time, these patients have a relatively lean phenotype associated with mobilization and oxidation of free fatty acids. These features are reversed by curative surgical removal of the growth hormone (GH)-producing adenoma. Mouse models of acromegaly share many of these characteristics, including a lean phenotype and proneness to type 2 diabetes. There are, however, also species differences with respect to oxidation rates of glucose and fat as well as the specific mechanisms underlying GH-induced insulin resistance. The impact of acromegaly treatment on insulin sensitivity and glucose tolerance depends on the treatment modality (e.g. somatostatin analogs also suppress insulin secretion, whereas the GH antagonist restores insulin sensitivity). The interplay between animal research and clinical studies has proven useful in the field of acromegaly and should be continued in order to understand the metabolic actions of GH.
Collapse
Affiliation(s)
- Jakob Dal
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | | | | | | |
Collapse
|
19
|
Anderwald CH, Tura A, Gessl A, Smajis S, Bieglmayer C, Marculescu R, Luger A, Pacini G, Krebs M. Whole-body insulin sensitivity rather than body-mass-index determines fasting and post-glucose-load growth hormone concentrations. PLoS One 2014; 9:e115184. [PMID: 25517727 PMCID: PMC4269423 DOI: 10.1371/journal.pone.0115184] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 11/19/2014] [Indexed: 11/25/2022] Open
Abstract
Background Obese, non-acromegalic persons show lower growth hormone (GH) concentrations at fasting and reduced GH nadir during an oral glucose tolerance test (OGTT). However, this finding has never been studied with regard to whole-body insulin-sensitivity as a possible regulator. Methods In this retrospective analysis, non-acromegalic (NonACRO, n = 161) and acromegalic (ACRO, n = 35), non-diabetic subjects were subdivided into insulin-sensitive (IS) and –resistant (IR) groups according to the Clamp-like Index (CLIX)-threshold of 5 mg·kg−1·min−1 from the OGTT. Results Non-acromegalic IS (CLIX: 8.8±0.4 mg·kg−1·min−1) persons with similar age and sex distribution, but lower (p<0.001) body-mass-index (BMI = 25±0 kg/m2, 84% females, 56±1 years) had 59% and 70%, respectively, higher (p<0.03) fasting GH and OGTT GH area under the curve concentrations than IR (CLIX: 3.5±0.1 mg·kg−1·min−1, p<0.001) subjects (BMI = 29±1 kg/m2, 73% females, 58±1 years). When comparing on average overweight non-acromegalic IS and IR with similar anthropometry (IS: BMI: 27±0 kg/m2, 82% females, 58±2 years; IR: BMI: 27±0 kg/m2, 71% females, 60±1 years), but different CLIX (IS: 8.7±0.9 vs. IR: 3.8±0.1 mg·kg−1·min−1, p<0.001), the results remained almost the same. In addition, when adjusted for OGTT-mediated glucose rise, GH fall was less pronounced in IR. In contrast, in acromegalic subjects, no difference was found between IS and IR patients with regard to fasting and post-glucose-load GH concentrations. Conclusions Circulating GH concentrations at fasting and during the OGTT are lower in non-acromegalic insulin-resistant subjects. This study seems the first to demonstrate that insulin sensitivity rather than body-mass modulates fasting and post-glucose-load GH concentrations in non-diabetic non–acromegalic subjects.
Collapse
Affiliation(s)
- Christian-Heinz Anderwald
- Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
- Metabolic Unit, Institute of Biomedical Engineering, National Research Council (ISIB-CNR), Padua, Italy
- Mariahilf Community Pharmacy, Arnoldstein, Austria
- Medical Direction, Specialized Hospital Complex Agathenhof, Micheldorf, Austria
- * E-mail:
| | - Andrea Tura
- Metabolic Unit, Institute of Biomedical Engineering, National Research Council (ISIB-CNR), Padua, Italy
| | - Alois Gessl
- Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Sabina Smajis
- Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Christian Bieglmayer
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Vienna, Vienna, Austria
| | - Rodrig Marculescu
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Vienna, Vienna, Austria
| | - Anton Luger
- Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Giovanni Pacini
- Metabolic Unit, Institute of Biomedical Engineering, National Research Council (ISIB-CNR), Padua, Italy
| | - Michael Krebs
- Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
20
|
Thankamony A, Tossavainen PH, Sleigh A, Acerini C, Elleri D, Dalton RN, Jackson NC, Umpleby AM, Williams RM, Dunger DB. Short-term administration of pegvisomant improves hepatic insulin sensitivity and reduces soleus muscle intramyocellular lipid content in young adults with type 1 diabetes. J Clin Endocrinol Metab 2014; 99:639-47. [PMID: 24423298 DOI: 10.1210/jc.2013-3264] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT Data on the metabolic effects of GH derived from studies using GH suppression by pharmacological agents may not reflect selective actions. OBJECTIVE The purpose of this study was to evaluate the effects of GH antagonism on glucose and lipid metabolism using pegvisomant, a selective GH receptor antagonist in patients with type 1 diabetes (T1D). DESIGN AND PARTICIPANTS In a randomized, placebo-controlled, crossover study, 10 young adults with T1D were evaluated at baseline and after 4 weeks of treatment with either 10 mg of pegvisomant or placebo. The assessments included an overnight euglycemic steady state followed by a hyperinsulinemic euglycemic clamp and used glucose and glycerol cold stable isotopes. OUTCOME MEASURES Hepatic and peripheral insulin sensitivity (IS), lipid turnover, and intramyocellular lipid (IMCL) were measured. RESULTS Compared with placebo, pegvisomant treatment resulted in lower IGF-I levels (P < .001). During the overnight steady state, insulin requirements for euglycemia (P = .019), insulin levels (P = .008), and glucose production rates (Ra) (P = .033) were reduced. During the clamp study, glucose infusion rates (P = .031) increased and glucose Ra (P = .015) decreased whereas glucose disposal rates were unchanged. Free fatty acid levels were similar during the steady state but were lower during the clamp (P = .040) after pegvisomant. Soleus muscle IMCL decreased after treatment (P = .024); however, no change in tibialis anterior muscle was observed. CONCLUSIONS The study demonstrates that GH antagonism in T1D results in improved hepatic insulin sensitivity. Lack of consistent changes in free fatty acid levels may suggest a direct effect of GH on IS. Unchanged peripheral IS despite reductions in IMCL indicate that GH-induced alterations in IMCL may not be causally linked to glucose metabolism.
Collapse
Affiliation(s)
- A Thankamony
- Department of Paediatrics (A.T., P.H.T., C.A., D.E., R.M.W., D.B.D.), University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 0QQ, United Kingdom; Wolfson Brain Imaging Centre (A.S.), University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 0QQ, United Kingdom; Department of Paediatrics (P.H.T.), University of Oulu, 90029 OYS, Oulu, Finland; WellChild Laboratory (R.N.D.), King's College London, Evelina Children's Hospital, London SE1 7EH, United Kingdom; Diabetes and Metabolic Medicine (N.C.J., A.M.U.), Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7WG, United Kingdom; and National Institute for Health Research Cambridge Comprehensive Biomedical Research Centre (D.B.D.), Cambridge, CB2 0QQ, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
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: 159] [Impact Index Per Article: 13.3] [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
|
22
|
Clasen BFF, Krusenstjerna-Hafstrøm T, Vendelbo MH, Thorsen K, Escande C, Møller N, Pedersen SB, Jørgensen JOL, Jessen N. Gene expression in skeletal muscle after an acute intravenous GH bolus in human subjects: identification of a mechanism regulating ANGPTL4. J Lipid Res 2013; 54:1988-97. [PMID: 23606725 DOI: 10.1194/jlr.p034520] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Growth hormone (GH) acutely stimulates lipolysis and fat oxidation, a process that operates postabsorptively and involves activation of the JAK-STAT pathway in the target tissue; no in vivo data exist regarding subsequent GH-regulated gene transcription. We obtained serum samples and muscle biopsies in human subjects before and 2 h after administration of a GH bolus. A significant (~75%) elevation in serum FFA levels was recorded post GH. Microarray identified 79 GH-regulated genes in muscle. With qRT-PCR, we then examined the expression of selected genes in the presence and absence of glucose-induced suppression of lipolysis. Four genes involved in the JAK-STAT5 signaling pathway were regulated by GH, including SOCS1-3 and CISH, in addition to three genes associated with insulin action: NFκB1A, PIK3C2B, and PRKAG2. The gene encoding ANGPTL4, a protein involved in lipolysis and suppression of LPL activity, exhibited the most pronounced upregulation (5.6-fold) after GH, which was abrogated by concomitant suppression of lipolysis. Therefore, the GH-induced stimulation of ANGPTL4 gene expression seems secondary to induction of lipolysis. This new concept implies that abundant supply of circulating FFA decreases the need for alternative triglyceride-derived FFA through distinct inhibition of LPL mediated by increased ANGPTL4 gene expression in human muscle.
Collapse
Affiliation(s)
- Berthil F F Clasen
- Institute of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Kelly AK, Waters SM, McGee M, Browne JA, Magee DA, Kenny DA. Expression of key genes of the somatotropic axis in longissimus dorsi muscle of beef heifers phenotypically divergent for residual feed intake. J Anim Sci 2013; 91:159-67. [DOI: 10.2527/jas.2012-5557] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Affiliation(s)
- A. K. Kelly
- School of Agriculture, Food Science and Veterinary Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - S. M. Waters
- Animal and Bioscience Research Department, Animal and Grassland Research and Innovation Centre, Teagasc, Dunsany, Co. Meath, Ireland
| | - M. McGee
- Teagasc, Animal & Grassland Research and Innovation Centre, Grange, Dunsany, Co. Meath, Ireland
| | - J. A. Browne
- School of Agriculture, Food Science and Veterinary Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - D. A. Magee
- School of Agriculture, Food Science and Veterinary Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - D. A. Kenny
- Animal and Bioscience Research Department, Animal and Grassland Research and Innovation Centre, Teagasc, Dunsany, Co. Meath, Ireland
| |
Collapse
|
24
|
Kaminsky P, Walker PM, Deibener J, Barbe F, Jeannesson E, Escanye JM, Dousset B, Klein M. Growth hormone potentiates thyroid hormone effects on post-exercise phosphocreatine recovery in skeletal muscle. Growth Horm IGF Res 2012; 22:240-244. [PMID: 22939217 DOI: 10.1016/j.ghir.2012.08.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Revised: 07/31/2012] [Accepted: 08/02/2012] [Indexed: 01/21/2023]
Abstract
OBJECTIVE The aim of the study was to determine the respective impact of thyroxine and growth hormone on in vivo skeletal mitochondrial function assessed via post exercise phosphocreatine recovery. DESIGN The hind leg muscles of 32 hypophysectomized rats were investigated using (31)P nuclear magnetic resonance spectroscopy at rest and during the recovery period following a non tetanic stimulation of the sciatic nerve. Each rat was supplemented with hydrocortisone and was randomly assigned to one of the 4 groups: the group Hx was maintained in hypopituitarism., the group HxT was treated with 1 μg/100g/day of thyroxine (T4), the group HxG with 0.2 IU/kg/day of recombinant human GH (rGH) and the group HxGT by both thyroxine and rGH. Inorganic phosphate (Pi), phosphocreatine (PCr) and ATP were directly measured on the spectra, permitting the calculation of the phosphorylation potential (PP). RESULTS At rest, the rats treated with rGH or T4 exhibited higher PCr levels than rats Hx. The recovery rates of PCr and PP were higher in rats treated with T4 than in T4-deprivated rats, suggesting improved mitochondrial function. The rats treated by both T4 and rGH showed higher PCr and PP recovery than those maintained in hypopituitarism or treated with T4 or rGH alone. CONCLUSIONS The study demonstrates that in contrast to T4, GH given alone in hypophysectomized rats does not improve in vivo mitochondrial oxidative metabolism. Growth hormone potentiates T4 effects on oxidative metabolism.
Collapse
Affiliation(s)
- P Kaminsky
- Pôle des Spécialités Médicales, Service de Médecine Interne, Centre Hospitalier Universitaire de Nancy, Hôpitaux de Brabois, 54500 Vandoeuvre, France.
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Stanley TL, Falutz J, Marsolais C, Morin J, Soulban G, Mamputu JC, Assaad H, Turner R, Grinspoon SK. Reduction in visceral adiposity is associated with an improved metabolic profile in HIV-infected patients receiving tesamorelin. Clin Infect Dis 2012; 54:1642-51. [PMID: 22495074 DOI: 10.1093/cid/cis251] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Tesamorelin, a growth hormone-releasing hormone analogue, decreases visceral adipose tissue (VAT) by 15%-20% over 6-12 months in individuals with human immunodeficiency virus (HIV)-associated abdominal adiposity, but it is unknown whether VAT reduction is directly associated with endocrine and metabolic changes. METHODS In 2 phase III, randomized, double-blind studies, men and women with HIV-associated abdominal fat accumulation were randomly assigned (ratio, 2:1) to receive tesamorelin or placebo for 26 weeks. At week 26, patients initially receiving tesamorelin were randomly assigned to continue receiving tesamorelin or to receive placebo for an additional 26 weeks. In per-protocol analysis of 402 subjects initially randomly assigned to receive tesamorelin, those with ≥8% reduction in VAT were defined a priori as responders per the statistical analysis plan. Post hoc analyses were performed to assess differences between responders and nonresponders. RESULTS Compared with tesamorelin nonresponders, responders experienced greater mean (±SD) reduction in triglyceride levels (26 weeks: -0.6 ± 1.7 mmol/L vs -0.1 ± 1.2 mmol/L [P = .005]; 52 weeks: -0.8 ± 1.8 mmol/L vs 0.0 ± 1.1 mmol/L [P = .003]) and attenuated changes in fasting glucose levels (26 weeks: 1 ± 16 mg/dL vs 5 ± 14 mg/dL [P = .01]; 52 weeks: -1 ± 14 mg/dL vs 8 ± 17 mg/dL [P < .001]), hemoglobin A1c levels (26 weeks: 0.1 ± 0.3% vs 0.3 ± 0.4% [P < .001]; 52 weeks: 0.0 ± 0.3% vs 0.2 ± 0.5% [P = .003]), and other parameters of glucose homeostasis. Similar patterns were seen for adiponectin levels, with significant improvement in responders vs nonresponders. Changes in lipid levels and glucose homeostasis were significantly associated with percentage change in VAT. CONCLUSIONS In contrast to nonresponders, HIV-infected patients receiving tesamorelin with ≥8% reduction in VAT have significantly improved triglyceride levels, adiponectin levels, and preservation of glucose homeostasis over 52 weeks of treatment. CLINICALTRIALS.GOV REGISTRATION: NCT00123253, NCT00435136, NCT00608023.
Collapse
Affiliation(s)
- Takara L Stanley
- Program in Nutritional Metabolism, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Zhang ZX, Liu YK, Pan H, Pan L, Zhang Q, Su HM, Zhao QL, Li H, He C. The effect of polyethylene glycol recombinant human growth hormone on growth and glucose metabolism in hypophysectomized rats. Growth Horm IGF Res 2012; 22:30-35. [PMID: 22257554 DOI: 10.1016/j.ghir.2011.12.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2011] [Revised: 08/13/2011] [Accepted: 12/14/2011] [Indexed: 01/25/2023]
Abstract
OBJECTIVE To study the effect of polyethylene glycol recombinant human growth hormone on growth and glucose metabolism in hypophysectomized rats, and compare the effect of treatment between recombinant human growth hormone (rhGH) and polyethylene glycol rhGH (PEG-rhGH). METHODS Hypophysectomy was performed in juvenile rats to build the animal model of GH deficiency. The hypophysectomized animals were randomly assigned into three groups and treated with saline (negative control, n=20), rhGH (n=20) and PEG-rhGH (n=20). A sham operation was performed to set up the normal control (n=20). Body weight, body length and tail length were recorded every 2days for a 14-day treatment and bone growth was measured at the end of therapy. Glucose infusion rate (GIR) determined by euglycemic hyperinsulinemic clamp was used to evaluate insulin sensitivity after GH treatment. We also examined plasma glucose and serum insulin levels RESULTS Compared with the negative control, the body weight, body length, tail length and bone growth increased significantly in hypophesectomized rats treated by GH (P<0.01). Although the weight gain in the first 10days was higher in the PEG-rhGH group than in the rhGH group (P<0.05), the growth promoting effect was similar between rhGH and PEG-rhGH (P>0.05). Neither rhGH nor PEG-rhGH impaired glucose tolerance of rats after hypophesectomy. Compared with negative controls, according to decreased serum insulin, reduced insulin expression in pancreatic cells and increased GIR in the clamp, both rhGH and PEG-rhGH groups had improved insulin sensitivity within 14 days (P<0.01). However, with prolonged treatment, the GIR in the rhGH-treated rats decreased significantly (P<0.05); while PEG-rhGH did not interfere with GIR, even after a doubled dose (P>0.05). CONCLUSIONS PEG-rhGH had the same linear growth promoting efficacy as unmodified rhGH. The short-term GH replacement could improve insulin sensitivity in hypophysectomized rats, especially after PEGylation.
Collapse
Affiliation(s)
- Zhi Xin Zhang
- Department of Pediatric, China-Japan Friendship Hospital, Beijing, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Koshinaka K, Toshinai K, Mohammad A, Noma K, Oshikawa M, Ueno H, Yamaguchi H, Nakazato M. Therapeutic potential of ghrelin treatment for unloading-induced muscle atrophy in mice. Biochem Biophys Res Commun 2011; 412:296-301. [DOI: 10.1016/j.bbrc.2011.07.086] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2011] [Accepted: 07/20/2011] [Indexed: 01/06/2023]
|
28
|
Krusenstjerna-Hafstrøm T, Clasen BF, Møller N, Jessen N, Pedersen SB, Christiansen JS, Jørgensen JOL. Growth hormone (GH)-induced insulin resistance is rapidly reversible: an experimental study in GH-deficient adults. J Clin Endocrinol Metab 2011; 96:2548-57. [PMID: 21613350 DOI: 10.1210/jc.2011-0273] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT It is clinically relevant and of physiological interest to investigate whether GH-induced insulin resistance depends on the timing of GH exposure relative to when insulin sensitivity is assessed. HYPOTHESIS GH-induced insulin resistance is rapidly reversible. DESIGN AND PARTICIPANTS Eight male GH-deficient patients underwent a 6-h euglycemic-hyperinsulinemic glucose clamp thrice in a randomized crossover design receiving either no GH (study 0), a 7-h GH infusion (0.2-0.3 mg in total) that terminated 5 h before the clamp (study 1), or a similar GH infusion timed to continue during the first hour of the clamp (study 2). A muscle biopsy was obtained 30 min into the clamp. The patients were compared with eight healthy untreated control subjects (study c). MAIN OUTCOME MEASURES The glucose infusion rate, indirect calorimetry, and free fatty acid metabolism were assessed. In muscle biopsies, protein phosphorylation of signal transducer and activator of transcription 5, Akt, and Akt substrate 160 (phospho-Akt substrate signal) and gene expression of IGF-I and SOCS1-3 were assessed. RESULTS Insulin sensitivity differed significantly between the GH-deficiency studies (P = 0.005) with distinct insulin resistance in study 2 and increased insulin sensitivity in study 0 [area under the glucose infusion rate curve (mg/kg · min): 1663 ± 151 (study 0) vs. 1482 ± 166 (study 1) vs. 1123 ± 136 (study 2) vs. 1492 ± 229 (control group)]. Free fatty acid levels and lipid oxidation were elevated in response to GH exposure but became suppressed during the clamp. IGF-I and SOCS3 gene expression was increased in study 2. CONCLUSIONS Very-low-dose GH exposure evokes acute insulin resistance that subsides after 5 h. This time-dependent reversibility should be considered when assessing the impact of GH on glucose homeostasis.
Collapse
Affiliation(s)
- T Krusenstjerna-Hafstrøm
- Department of Internal Medicine and Endocrinology and Medical Research Laboratories, Aarhus University Hospital, Denmark.
| | | | | | | | | | | | | |
Collapse
|
29
|
List EO, Sackmann-Sala L, Berryman DE, Funk K, Kelder B, Gosney ES, Okada S, Ding J, Cruz-Topete D, Kopchick JJ. Endocrine parameters and phenotypes of the growth hormone receptor gene disrupted (GHR-/-) mouse. Endocr Rev 2011; 32:356-86. [PMID: 21123740 PMCID: PMC3365798 DOI: 10.1210/er.2010-0009] [Citation(s) in RCA: 143] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Disruption of the GH receptor (GHR) gene eliminates GH-induced intracellular signaling and, thus, its biological actions. Therefore, the GHR gene disrupted mouse (GHR-/-) has been and is a valuable tool for helping to define various parameters of GH physiology. Since its creation in 1995, this mouse strain has been used by our laboratory and others for numerous studies ranging from growth to aging. Some of the most notable discoveries are their extreme insulin sensitivity in the presence of obesity. Also, the animals have an extended lifespan, which has generated a large number of investigations into the roles of GH and IGF-I in the aging process. This review summarizes the many results derived from the GHR-/- mice. We have attempted to present the findings in the context of current knowledge regarding GH action and, where applicable, to discuss how these mice compare to GH insensitivity syndrome in humans.
Collapse
Affiliation(s)
- Edward O List
- The Edison Biotechnology Institute, Ohio University, Athens, Ohio 45701, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Krusenstjerna-Hafstrøm T, Madsen M, Vendelbo MH, Pedersen SB, Christiansen JS, Møller N, Jessen N, Jørgensen JOL. Insulin and GH signaling in human skeletal muscle in vivo following exogenous GH exposure: impact of an oral glucose load. PLoS One 2011; 6:e19392. [PMID: 21559284 PMCID: PMC3086909 DOI: 10.1371/journal.pone.0019392] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2011] [Accepted: 03/29/2011] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION GH induces acute insulin resistance in skeletal muscle in vivo, which in rodent models has been attributed to crosstalk between GH and insulin signaling pathways. Our objective was to characterize time course changes in signaling pathways for GH and insulin in human skeletal muscle in vivo following GH exposure in the presence and absence of an oral glucose load. METHODS Eight young men were studied in a single-blinded randomized crossover design on 3 occasions: 1) after an intravenous GH bolus 2) after an intravenous GH bolus plus an oral glucose load (OGTT), and 3) after intravenous saline plus OGTT. Muscle biopsies were taken at t = 0, 30, 60, and 120. Blood was sampled at frequent intervals for assessment of GH, insulin, glucose, and free fatty acids (FFA). RESULTS GH increased AUC(glucose) after an OGTT (p<0.05) without significant changes in serum insulin levels. GH induced phosphorylation of STAT5 independently of the OGTT. Conversely, the OGTT induced acute phosphorylation of the insulin signaling proteins Akt (ser(473) and thr(308)), and AS160.The combination of OGTT and GH suppressed Akt activation, whereas the downstream expression of AS160 was amplified by GH. WE CONCLUDED THE FOLLOWING: 1) A physiological GH bolus activates STAT5 signaling pathways in skeletal muscle irrespective of ambient glucose and insulin levels 2) Insulin resistance induced by GH occurs without a distinct suppression of insulin signaling proteins 3) The accentuation of the glucose-stimulated activation of AS 160 by GH does however indicate a potential crosstalk between insulin and GH. TRIAL REGISTRATION ClinicalTrials.gov NCT00477997.
Collapse
Affiliation(s)
- Thomas Krusenstjerna-Hafstrøm
- Department of Internal Medicine and Endocrinology (MEA) and Medical Research Laboratories, Aarhus University Hospital, Aarhus, Denmark.
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Vestergaard ET, Buhl M, Gjedsted J, Madsen M, Jessen N, Nielsen S, Gaylinn BD, Liu J, Thorner MO, Moller N, Jorgensen JOL. Acute peripheral metabolic effects of intraarterial ghrelin infusion in healthy young men. J Clin Endocrinol Metab 2011; 96:468-77. [PMID: 21084392 PMCID: PMC5393419 DOI: 10.1210/jc.2010-1995] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
CONTEXT Ghrelin is the endogenous agonist for the growth hormone secretagogue receptor (GHS-R). Intravenous administration of ghrelin induces insulin resistance and hyperglycemia and increases the levels of free fatty acids (FFA). OBJECTIVE To investigate whether these effects are mediated directly by ghrelin in skeletal muscle tissue. DESIGN This study was single blinded, randomized, and placebo controlled. Eight healthy men (25.5 ± 3.1 years) received 240 min of intraarterial ghrelin infusion (4.2 ng × kg(-1) × min(-1)) into one femoral artery and intraarterial placebo infusion into the contralateral artery. Simultaneous blood samples were drawn from both femoral veins and muscle biopsies were obtained from both legs during both a basal period and during a hyperinsulinemic and euglycemic clamp period. RESULTS Ghrelin significantly elevated venous FFA levels and venous dilution of palmitate, suggestive of increased lipolysis. Glucose metabolism was unchanged, and there were no direct effects on pertinent enzymes in the insulin signaling cascade. The metabolic clearance rate of acyl ghrelin was 12.5 ± 3.3 ml × kg(-1) × min(-1). Acyl and desacyl ghrelin levels both increased. CONCLUSIONS The results of this study suggest that ghrelin may stimulate lipolysis directly in skeletal muscle.
Collapse
|
32
|
Jensen J, Ruge T, Lai YC, Svensson MK, Eriksson JW. Effects of adrenaline on whole-body glucose metabolism and insulin-mediated regulation of glycogen synthase and PKB phosphorylation in human skeletal muscle. Metabolism 2011; 60:215-26. [PMID: 20153492 DOI: 10.1016/j.metabol.2009.12.028] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2009] [Revised: 12/23/2009] [Accepted: 12/28/2009] [Indexed: 11/20/2022]
Abstract
In the present study, we investigated the effect of adrenaline on insulin-mediated regulation of glucose and fat metabolism with focus on regulation of skeletal muscle PKB, GSK-3, and glycogen synthase (GS) phosphorylation. Ten healthy subjects (5 men and 5 women) received a 240-minute intravenous infusion of adrenaline (0.05 μg/[kg min]) or saline; after 120 minutes, a hyperinsulinemic-euglycemic clamp was added. Adrenaline infusion increased blood glucose concentration by approximately 50%, but the hyperinsulinemic clamp normalized blood glucose within 30 minutes. Glucose infusion rate during the last hour was approximately 60% lower during adrenaline infusion compared with saline (4.3 ± 0.5 vs 11.2 ± 0.6 mg/kg lean body mass per minute). Insulin increased PKB Ser⁴⁷³, PKB Thr³⁰⁸, and GSK-3β Ser⁹ phosphorylation in skeletal muscles; coinfusion of adrenaline did not influence insulin-stimulated PKB and GSK-3 phosphorylation. Adrenaline alone did not influence phosphorylation of PKB and GSK-3β. Insulin increased GS fractional activity and decreased GS Ser⁶⁴¹ and Ser⁶⁴⁵,⁶⁴⁹,⁶⁵³,⁶⁵⁷ phosphorylation. In the presence of adrenaline, insulin did neither activate GS nor dephosphorylate GS Ser⁶⁴¹. Surprisingly, GS Ser⁷ phosphorylation was not influenced by adrenaline. Adrenaline increased plasma lactate concentration; and muscle glycogen content was reduced in skeletal muscle the day after adrenaline infusion, supporting that insulin does not stimulate glycogen synthesis in skeletal muscles when adrenaline is present. In conclusion, adrenaline did not influence basal or insulin-stimulated PKB and GSK-3β phosphorylation in muscles, but completely blocked insulin-mediated GS activation and Ser⁶⁴¹ dephosphorylation. Still, insulin normalized adrenaline-mediated hyperglycemia.
Collapse
Affiliation(s)
- Jørgen Jensen
- Department of Physiology, National Institute of Occupational Health, P.O. Box 8149 Dep, N-0033 Oslo, Norway.
| | | | | | | | | |
Collapse
|
33
|
Effects of GH in human muscle and fat. Pediatr Nephrol 2010; 25:705-9. [PMID: 19902270 DOI: 10.1007/s00467-009-1334-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2009] [Revised: 09/07/2009] [Accepted: 09/09/2009] [Indexed: 10/20/2022]
Abstract
Skeletal muscle is the major constituent of lean body mass and a major determinant of energy expenditure both at rest and during physical activity. Growth hormone, in turn, influences muscle mass as well as energy expenditure. Growth hormone substitution in adults increases muscle mass by 5-10%, but part of the effect is attributed to rehydration rather than protein accretion. In addition, GH regulates substrate metabolism in muscle and in particular antagonizes insulin-stimulated glucose disposal. This effect is linked to increased free fatty acid (FFA) flux but the molecular mechanisms remain unclear. During fasting, GH-induced insulin resistance may be favorable by reducing the demand of gluconeogenesis from protein. But in the postprandial phase, GH exposure may compromise glucose tolerance via the same mechanisms. Understanding the mechanisms whereby GH antagonizes insulin-stimulated glucose disposal in muscle is an important future research field with implications for a variety of clinical conditions ranging from malnutrition to obesity and type 2 diabetes.
Collapse
|
34
|
List EO, Palmer AJ, Berryman DE, Bower B, Kelder B, Kopchick JJ. Growth hormone improves body composition, fasting blood glucose, glucose tolerance and liver triacylglycerol in a mouse model of diet-induced obesity and type 2 diabetes. Diabetologia 2009; 52:1647-55. [PMID: 19468705 DOI: 10.1007/s00125-009-1402-z] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2009] [Accepted: 05/05/2009] [Indexed: 11/25/2022]
Abstract
AIMS/HYPOTHESIS Growth hormone has been used experimentally in two studies to treat individuals with type 2 diabetes, with both reporting beneficial effects on glucose metabolism. However, concerns over potential diabetogenic actions of growth hormone complicate its anticipated use to treat type 2 diabetes. Thus, an animal model of type 2 diabetes could help evaluate the effects of growth hormone for treating this condition. METHODS Male C57BL/6J mice were placed on a high-fat diet to induce obesity and type 2 diabetes. Starting at 16 weeks of age, mice were treated once daily for 6 weeks with one of four different doses of growth hormone. Body weight, body composition, fasting blood glucose, insulin, glucose tolerance, liver triacylglycerol, tissue weights and blood chemistries were determined. RESULTS Body composition measurements revealed a dose-dependent decrease in fat and an increase in lean mass. Analysis of fat loss by depot revealed that subcutaneous and mesenteric fat was the most sensitive to growth hormone treatment. In addition, growth hormone treatment resulted in improvement in glucose metabolism, with the highest dose normalising glucose, glucose tolerance and liver triacylglycerol. In contrast, insulin levels were not altered by the treatment, nor did organ weights change. However, fasting plasma leptin and resistin were significantly decreased after growth hormone treatment. CONCLUSIONS/INTERPRETATION Growth hormone therapy improves glucose metabolism in this mouse model of obesity and type 2 diabetes, providing a means to explore the molecular mechanism(s) of this treatment.
Collapse
Affiliation(s)
- E O List
- Edison Biotechnology Institute, Ohio University, 101 Konneker Research Laboratories, Athens, OH 45701, USA
| | | | | | | | | | | |
Collapse
|
35
|
Møller N, Jørgensen JOL. Effects of growth hormone on glucose, lipid, and protein metabolism in human subjects. Endocr Rev 2009; 30:152-77. [PMID: 19240267 DOI: 10.1210/er.2008-0027] [Citation(s) in RCA: 663] [Impact Index Per Article: 41.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
In evolutionary terms, GH and intracellular STAT 5 signaling is a very old regulatory system. Whereas insulin dominates periprandially, GH may be viewed as the primary anabolic hormone during stress and fasting. GH exerts anabolic effects directly and through stimulation of IGF-I, insulin, and free fatty acids (FFA). When subjects are well nourished, the GH-induced stimulation of IGF-I and insulin is important for anabolic storage and growth of lean body mass (LBM), adipose tissue, and glycogen reserves. During fasting and other catabolic states, GH predominantly stimulates the release and oxidation of FFA, which leads to decreased glucose and protein oxidation and preservation of LBM and glycogen stores. The most prominent metabolic effect of GH is a marked increase in lipolysis and FFA levels. In the basal state, the effects of GH on protein metabolism are modest and include increased protein synthesis and decreased breakdown at the whole body level and in muscle together with decreased amino acid degradation/oxidation and decreased hepatic urea formation. During fasting and stress, the effects of GH on protein metabolism become more pronounced; lack of GH during fasting increases protein loss and urea production rates by approximately 50%, with a similar increase in muscle protein breakdown. GH is a counterregulatory hormone that antagonizes the hepatic and peripheral effects of insulin on glucose metabolism via mechanisms involving the concomitant increase in FFA flux and uptake. This ability of GH to induce insulin resistance is significant for the defense against hypoglycemia, for the development of "stress" diabetes during fasting and inflammatory illness, and perhaps for the "Dawn" phenomenon (the increase in insulin requirements in the early morning hours). Adult patients with GH deficiency are insulin resistant-probably related to increased adiposity, reduced LBM, and impaired physical performance-which temporarily worsens when GH treatment is initiated. Conversely, despite increased LBM and decreased fat mass, patients with acromegaly are consistently insulin resistant and become more sensitive after appropriate treatment.
Collapse
Affiliation(s)
- Niels Møller
- Medical Department M, Aarhus University Hospital, Aarhus Sygehus, DK, Aarhus, Denmak
| | | |
Collapse
|
36
|
Park MJ, Jung SR, Jung HL, Craig BW, Lee CD, Kang HY. Effects of 4 weeks recombinant human growth hormone administration on insulin resistance of skeletal muscle in rats. Yonsei Med J 2008; 49:1008-16. [PMID: 19108026 PMCID: PMC2628033 DOI: 10.3349/ymj.2008.49.6.1008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
PURPOSE Effect of recombinant human growth hormone (rhGH) administration on lipid storage, and its subsequent effect on insulin sensitivity have not yet been adequately examined. Thus, we investigated the effects of rhGH treatment on muscle triglyceride (TG) and ceramide content, and insulin sensitivity after 4 weeks of rhGH administration in rats. MATERIALS AND METHODS Fourteen rats were randomly assigned to two groups: rhGH injection group (GH, n = 7) and saline injection group (CON, n = 7). GH received rhGH by subcutaneous injections (130 microg.kg(-1).day(-1), 6 days.week(-1)) for 4 weeks, while CON received saline injections that were equivalent in volume to GH group. Intramuscular TG and ceramide content and hepatic TG content were measured. To determine insulin sensitivity, oral glucose tolerance test (OGTT) and muscle incubation for glucose transport rate were performed in rats, and used as indicators of insulin sensitivity. We also examined plasma lipid profiles. RESULTS After 4 weeks of rhGH treatment, the GH group had higher muscle and liver TG contents than the CON (p < 0.05). Ceramide content in GH was significantly greater than that in CON (p < 0.05). GH also had higher plasma levels of FFA (p < 0.05), glucose and insulin responses during OGTT (p < 0.05), and lower glucose transport rates in submaximal insulin concentration (p < 0.05) as compared with CON. Results indicate that rhGH treatment is associated with insulin resistance in rats. CONCLUSION rhGH treatment elevated muscle TG and ceramide content, and hepatic TG content. Thus, elevation of these compounded by rhGH treatment could contribute to the development of insulin resistance in rats.
Collapse
Affiliation(s)
- Mi Jung Park
- Department of Pediatrics, Sanggye Paik Hospital, Inje University School of Medicine, Seoul, Korea
| | - Su Ryun Jung
- Department of Physical Education, Kyungpook National University, Daegu, Korea
| | - Hyun Lyung Jung
- Department of Physical Education, Kyungpook National University, Daegu, Korea
| | - Bruce W. Craig
- Human Performance Laboratory, Ball State University, Muncie, Indiana, USA
| | - Chong-Do Lee
- Department of Exercise and Wellness, Arizona State University, Mesa, Arizona, USA
| | - Ho Youl Kang
- Department of Physical Education, Kyungpook National University, Daegu, Korea
| |
Collapse
|
37
|
Krag MB, Nielsen S, Guo Z, Pedersen SB, Schmitz O, Christiansen JS, Jørgensen JOL. Peroxisome proliferator-activated receptor gamma agonism modifies the effects of growth hormone on lipolysis and insulin sensitivity. Clin Endocrinol (Oxf) 2008; 69:452-61. [PMID: 18331610 DOI: 10.1111/j.1365-2265.2008.03231.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
CONTEXT Peroxisome proliferator-activated receptor gamma (PPAR-gamma) agonists such as thiazolidinediones (TZDs) improve insulin sensitivity in type 2 diabetes mellitus (T2DM) through effects on fat metabolism whereas GH stimulates lipolysis and induces insulin resistance. OBJECTIVE To evaluate the impact of TZDs on fat metabolism and insulin sensitivity in subjects exposed to stable GH levels. DESIGN A randomized, placebo-controlled, double-blind parallel-group study including 20 GH-deficient patients on continued GH replacement therapy. The patients were studied before and after 12 weeks. INTERVENTION Patients received either pioglitazone 30 mg (N = 10) or placebo (N = 10) once daily for 12 weeks. RESULTS Adiponectin levels almost doubled during pioglitazone treatment (P = 0.0001). Pioglitazone significantly decreased basal free fatty acid (FFA) levels (P = 0.02) and lipid oxidation (P = 0.02). Basal glucose oxidation rate (P = 0.004) and insulin sensitivity (P = 0.03) improved in the patients who received pioglitazone treatment. The change in insulin-stimulated adiponectin level after pioglitazone treatment was positively correlated to the change in insulin-stimulated total glucose disposal (R = 0.69, P = 0.04). CONCLUSION The impact of GH on lipolysis and insulin sensitivity can be modified by administration of TZDs.
Collapse
Affiliation(s)
- Morten B Krag
- Medical Department M, Aarhus University Hospital, Aarhus Sygehus, Aarhus C, Denmark
| | | | | | | | | | | | | |
Collapse
|
38
|
Nielsen C, Gormsen LC, Jessen N, Pedersen SB, Møller N, Lund S, Jørgensen JOL. Growth hormone signaling in vivo in human muscle and adipose tissue: impact of insulin, substrate background, and growth hormone receptor blockade. J Clin Endocrinol Metab 2008; 93:2842-50. [PMID: 18460563 DOI: 10.1210/jc.2007-2414] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
CONTEXT GH induces insulin resistance in muscle and fat, and in vitro data indicate that this may involve cross-talk between the signaling pathways of the two hormones. OBJECTIVE Our objective was to investigate GH and insulin signaling in vivo in human muscle and fat tissue in response to GH, GH receptor blockade, and insulin stimulation. DESIGN We conducted two randomized crossover studies. PARTICIPANTS Sixteen healthy males participated. INTERVENTION GH was administered as a bolus (n = 8) and constant infusion (n = 8). The bolus study included three arms: 1) control (saline), 2) GH (0.5 mg iv), and 3) GH blockade (pegvisomant 30 mg sc), each combined with a hyperinsulinemic glucose clamp. The infusion study included two arms: 1) GH infusion (45 ng/.kg.min, 5.5 h) and 2) saline infusion (5.5 h) combined with a hyperinsulinemic glucose clamp during the final 2.5 h. MAIN OUTCOME MEASURES Muscle and fat biopsies were subjected to Western blotting for expression of Stat5/p-Stat5, Akt/p-Akt, and ERK1/2/p-ERK1/2 and to real-time RT-PCR for expression of SOCS1-3 and IGF-I mRNA. RESULTS GH significantly reduced insulin sensitivity. The GH bolus as well as GH infusion induced phosphorylation of Stat5 in muscle and fat, and SOCS3 and IGF-I mRNA expression increased after GH infusion. Hyperinsulinemia induced Akt phosphorylation in both tissues, irrespective of GH status. In muscle, ERK1/2 phosphorylation was increased by insulin, but insulin per se did not induce phosphorylation of Stat5. CONCLUSIONS GH exposure associated with insulin resistance acutely translates into GH receptor signaling in human muscle and fat without evidence of cross-talk with insulin signaling pathways. The molecular mechanisms subserving GH-induced insulin resistance in humans remain unclarified.
Collapse
Affiliation(s)
- Charlotte Nielsen
- Medical Department M, Aarhus University Hospital, Norrebrogade 44, Aarhus C, Denmark.
| | | | | | | | | | | | | |
Collapse
|
39
|
Short KR, Moller N, Bigelow ML, Coenen-Schimke J, Nair KS. Enhancement of muscle mitochondrial function by growth hormone. J Clin Endocrinol Metab 2008; 93:597-604. [PMID: 18000087 PMCID: PMC2243230 DOI: 10.1210/jc.2007-1814] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
CONTEXT Although GH promotes growth and protein anabolism, which are ATP-dependent processes, the GH effect on mitochondrial regulation remains to be determined. OBJECTIVE Our objective was to determine the acute effect of GH on mitochondrial oxidative capacity in skeletal muscle of healthy subjects. DESIGN AND SETTING The study was a randomized crossover design at an academic medical center. PARTICIPANTS Nine healthy men and women completed the study. INTERVENTION GH (150 microg/h) or saline was infused for 14 h on separate days, and muscle biopsies were obtained. MAIN OUTCOME MEASURES Outcome measures included mitochondrial function, gene expression, and protein metabolism. RESULTS The 4-fold increase in plasma GH caused elevations in plasma IGF-I, insulin, glucose, and free fatty acids and a shift in fuel selection, with less carbohydrate (-69%) and leucine (-43%) oxidation and 29% more fat oxidation. Muscle mitochondrial ATP production rate and citrate synthase activity were increased 16-35% in response to GH. GH also resulted in higher abundance of muscle mRNAs encoding IGF-I, mitochondrial proteins from the nuclear (cytochrome c oxidase subunit 4) and mitochondrial (cytochrome c oxidase subunit 3) genomes, the nuclear-derived mitochondrial transcription factor A, and glucose transporter 4. Although GH increased whole-body protein synthesis (nonoxidative disposal of leucine), no effect on synthesis rate of muscle mitochondrial proteins was observed. CONCLUSIONS These results demonstrate that acute GH action promotes an increase in mitochondrial oxidative capacity and abundance of several mitochondrial genes. These events may occur through direct or indirect effects of GH on intracellular signaling pathways but do not appear to involve a change in mitochondrial protein synthesis rate.
Collapse
Affiliation(s)
- Kevin R Short
- Endocrinology Research Unit, Mayo Clinic School of Medicine, 5-194 Joseph, 200 First Street SW, Rochester, Minnesota 55905, USA
| | | | | | | | | |
Collapse
|
40
|
Alkhateeb H, Chabowski A, Glatz JFC, Luiken JFP, Bonen A. Two phases of palmitate-induced insulin resistance in skeletal muscle: impaired GLUT4 translocation is followed by a reduced GLUT4 intrinsic activity. Am J Physiol Endocrinol Metab 2007; 293:E783-93. [PMID: 17550999 DOI: 10.1152/ajpendo.00685.2006] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We examined, in soleus muscle, the effects of prolonged palmitate exposure (0, 6, 12, 18 h) on insulin-stimulated glucose transport, intramuscular lipid accumulation and oxidation, activation of selected insulin-signaling proteins, and the insulin-stimulated translocation of GLUT4. Insulin-stimulated glucose transport was progressively reduced after 6 h (-33%), 12 h (-66%), and 18 h (-89%) of palmitate exposure. These decrements were closely associated with concurrent reductions in palmitate oxidation at 6 h (-40%), 12 h (-60%), and 18 h (-67%). In contrast, intramuscular ceramide (+24%) and diacylglycerol (+32%) concentrations, insulin-stimulated AS160 (-36%) and PRAS40 (-33%) phosphorylations, and Akt (-40%), PKCtheta (-50%), and GLUT4 translocation (-40%) to the plasma membrane were all maximally altered within the first 6 h of palmitate treatment. No further changes were observed in any of these parameters after 12 and 18 h of palmitate exposure. Thus, the intrinsic activity of GLUT4 was markedly reduced after 12 and 18 h of palmitate treatment. During this reduced GLUT4 intrinsic activity phase at 12 and 18 h, the reduction in glucose transport was twofold greater compared with the early phase (< or =6 h), when only GLUT4 translocation was impaired. Our study indicates that palmitate-induced insulin resistance is provoked by two distinct mechanisms: 1) an early phase (< or =6 h), during which lipid-mediated impairments in insulin signaling and GLUT4 translocation reduce insulin-stimulated glucose transport, followed by 2) a later phase (12 and 18 h), during which the intrinsic activity of GLUT4 is markedly reduced independently of any further alterations in intramuscular lipid accumulation, insulin signaling and GLUT4 translocation.
Collapse
Affiliation(s)
- Hakam Alkhateeb
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada
| | | | | | | | | |
Collapse
|
41
|
Xia Y, Wan X, Duan Q, He S, Wang X. Inhibition of protein kinase B by palmitate in the insulin signaling of HepG2 cells and the preventive effect of arachidonic acid on insulin resistance. ACTA ACUST UNITED AC 2007; 1:200-6. [PMID: 24557677 DOI: 10.1007/s11684-007-0038-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2006] [Accepted: 02/28/2007] [Indexed: 11/27/2022]
Abstract
Elevated plasma levels of free fatty acids (FFAs) may contribute to insulin resistance (IR) that is characteristic of type 2 diabetes mellitus. In this study, we investigated the effects of two fatty acids, palmitate (PA) and arachidonic acid (AA) on glycogenesis under insulin signaling in HepG2cells, a transformed hepatic carcinoma cell line. In the presence of 200 μmol of palmitate, insulin (10(-7) mol/L) stimulation of glycogenesis was inhibited, as evidenced by increased glucose in the medium and decreased intracellular glycogen. Wortmannin (WM), a specific inhibitor of PI3K, dramatically decreased the amount of intracellular glycogen in cells without PA incubation. However, glycogen in PA treated cells was not significantly changed by WM, indicating that PA may also act on PI3K. Interestingly, AA restored the effects of WM inhibition on glycogenesis in PA cells. Western blot analysis demonstrated that PA in the absence of WM increased phosphorylated glycogen synthase (inactive form of GS) and decreased phosphorylated protein kinase B (active form of PKB), causing a reduction of intracellular glycogen. AA, however, reversed the effects of PA on GS and PKB. Furthermore, inhibition of protein kinase C (PKC) by a specific inhibitor chelerythrine chloride (CC) abolished the inhibitory effect of PA on glycogen synthesis by decreasing phosphorylated GS and increasing phosphorylated PKB. However, the effect of CC in the presence of PA disappeared when AA was also present.Our results suggest that there is a disruption of the insulin signaling pathway between PKB and GS when the cells were exposed to PA, contributing to IR. PA may also interrupt the PKC signaling pathway. In contrast, AA could rescue glycogenesis impaired by PA.
Collapse
Affiliation(s)
- Yanzhi Xia
- Department of Medical Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | | | | | | | | |
Collapse
|
42
|
Jørgensen JOL, Møller L, Krag M, Billestrup N, Christiansen JS. Effects of growth hormone on glucose and fat metabolism in human subjects. Endocrinol Metab Clin North Am 2007; 36:75-87. [PMID: 17336735 DOI: 10.1016/j.ecl.2006.11.005] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
This article focuses on in vivo data from tests performed in normal subjects and in patients who had abnormal growth hormone (GH) status. Experimental data in human subjects demonstrate that GH acutely inhibits glucose disposal in skeletal muscle. At the same time GH stimulates the turnover and oxidation of free fatty acid (FFA), and experimental evidence suggests a causal link between elevated FFA levels and insulin resistance in skeletal muscle. Observational data in GH-deficient adults do not indicate that GH replacement is associated with significant impairment of glucose tolerance, but it is recommended that overdosing be avoided and glycemic control be monitored.
Collapse
Affiliation(s)
- Jens O L Jørgensen
- Medical Department M (Endocrinology and Diabetes) and Institute of Experimental Clinical Research, Aarhus University Hospital, Norrebrogade 44, DK-800C, Aarhus, Denmark.
| | | | | | | | | |
Collapse
|
43
|
Tarantino G, Saldalamacchia G, Conca P, Arena A. Non-alcoholic fatty liver disease: further expression of the metabolic syndrome. J Gastroenterol Hepatol 2007; 22:293-303. [PMID: 17295757 DOI: 10.1111/j.1440-1746.2007.04824.x] [Citation(s) in RCA: 124] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Non-alcoholic fatty liver disease has been associated with metabolic disorders, including central obesity, dyslipidemia, hypertension and hyperglycemia. Metabolic syndrome, obesity, and insulin resistance are major risk factors in the pathogenesis of non-alcoholic fatty liver disease. Non-alcoholic fatty liver disease refers to a wide spectrum of liver damage, ranging from simple steatosis to non-alcoholic steatohepatitis, advanced fibrosis and cirrhosis.
Collapse
Affiliation(s)
- Giovanni Tarantino
- Department of Clinical and Experimental Medicine, Federico II University Medical School, Naples, Italy.
| | | | | | | |
Collapse
|
44
|
Krag MB, Gormsen LC, Guo Z, Christiansen JS, Jensen MD, Nielsen S, Jørgensen JOL. Growth hormone-induced insulin resistance is associated with increased intramyocellular triglyceride content but unaltered VLDL-triglyceride kinetics. Am J Physiol Endocrinol Metab 2007; 292:E920-7. [PMID: 17132823 DOI: 10.1152/ajpendo.00374.2006] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The ability of growth hormone (GH) to stimulate lipolysis and cause insulin resistance in skeletal muscle may be causally linked, but the mechanisms remain obscure. We investigated the impact of GH on the turnover of FFA and VLDL-TG, intramuscular triglyceride content (IMTG), and insulin sensitivity (euglycemic clamp) in nine healthy men in a randomized double-blind placebo-controlled crossover study after 8 days treatment with (A) Placebo+Placebo, (B) GH (2 mg daily)+Placebo, and (C) GH (2 mg daily)+Acipimox (250 mgx3 daily). In the basal state, GH (B) increased FFA levels (P<0.05), palmitate turnover (P<0.05), and lipid oxidation (P=0.05), but VLDL-TG kinetics were unaffected. Administration of acipimox (C) suppressed basal lipolysis but did not influence VLDL-TG kinetics. In the basal state, IMTG content increased after GH (B; P=0.03). Insulin resistance was induced by GH irrespective of concomitant acipimox (P<0.001). The turnover of FFA and VLDL-TG was suppressed by hyperinsulinemia during placebo and GH, whereas coadministration of acipimox induced a rebound increase FFA turnover and VLDL-TG clearance. We conclude that these results show that GH-induced insulin resistance is associated with increased IMTG and unaltered VLDL-TG kinetics; we hypothesize that fat oxidation in muscle tissue is an important primary effect of GH and that circulating FFA rather than VLDL-TG constitute the major source for this process; and the role of IMTG in the development of GH-induced insulin resistance merits future research.
Collapse
Affiliation(s)
- Morten B Krag
- Medical Department M, Endocrinology and Diabetes, Aarhus University Hospital, DK-8000 Aarhus C, Denmark
| | | | | | | | | | | | | |
Collapse
|
45
|
Jørgensen JOL, Jessen N, Pedersen SB, Vestergaard E, Gormsen L, Lund SA, Billestrup N. GH receptor signaling in skeletal muscle and adipose tissue in human subjects following exposure to an intravenous GH bolus. Am J Physiol Endocrinol Metab 2006; 291:E899-905. [PMID: 16757551 DOI: 10.1152/ajpendo.00024.2006] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Growth hormone (GH) regulates muscle and fat metabolism, which impacts on body composition and insulin sensitivity, but the underlying GH signaling pathways have not been studied in vivo in humans. We investigated GH signaling in biopsies from muscle and abdominal fat obtained 30 (n = 3) or 60 (n = 3) min after an intravenous bolus of GH (0.5 mg) vs. saline in conjunction with serum sampling in six healthy males after an overnight fast. Expression of the following signal proteins were assayed by Western blotting: STAT5/p-STAT5, MAPK, and Akt/PKB. IRS-1-associated PI 3-kinase activity was measured by in vitro phosphorylation of PI. STAT5 DNA binding activity was assessed with EMSA, and the expression of IGF-I and SOCS mRNA was measured by real-time RT-PCR. GH induced a 52% increase in circulating FFA levels with peak values after 155 min (P = 0.03). Tyrosine-phosphorylated STAT5 was detected in muscle and fat of all subjects after GH. Activation of MAPK was observed in several lysates but without GH dependency. Neither PKB/Akt nor PI 3-kinase activity was affected by GH. GH-induced STAT5 DNA binding and expression of IGF-I mRNA were detected in fat, whereas expression of SOCS-1 and -3 tended to increase after GH in muscle and fat, respectively. We conclude that 1) STAT5 is acutely activated in human muscle and fat after a GH bolus, but additional downstream GH signaling was significant only in fat; 2) the direct GH effects in muscle need further characterization; and 3) this human in vivo model may be used to study the mechanisms subserving the actions of GH on substrate metabolism and insulin sensitivity in muscle and fat.
Collapse
Affiliation(s)
- Jens O L Jørgensen
- Aarhus University Hospital and Institute of Clinical Research, Aarhus University, Aarhus, and Steno Diabetes Center, Copenhagen, Denmark.
| | | | | | | | | | | | | |
Collapse
|