1
|
Rong P, Mu Y, Wang M, Chen L, Liu F, Jin Y, Feng W, Zhou K, Liang H, Wang HY, Chen S. Targeting IGF1 to alleviate obesity through regulating energy expenditure and fat deposition. SCIENCE CHINA. LIFE SCIENCES 2025; 68:1662-1675. [PMID: 39843847 DOI: 10.1007/s11427-024-2768-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 10/31/2024] [Indexed: 01/24/2025]
Abstract
Insulin-like growth factor 1 (IGF1) is a regulator of both cellular hypertrophy and lipogenesis, which are two key processes for pathogenesis of obesity. However, the in vivo role of IGF1 in the development of obesity remains unclear. Here, we show that IGF1 expression is increased in adipose tissue in obese human patients and animal models. Elevation of IGF1 is associated with increased lipogenic gene expression and decreased energy expenditure. Genetic down-regulation of IGF1 normalizes lipogenic gene expression, restores aberrant energy metabolism and alleviates obese phenotype of a genetic mouse model with IGF1-hypersecretion. Importantly, genetic down-regulation of IGF1 exerts similar effects on development of diet-induced obesity. Furthermore, berberine that is an AMP-activated protein kinase (AMPK) activator in medicinal herbs inhibits IGF1 secretion, decreases lipogenic gene expression and alleviates diet-induced adiposity. Collectively, our findings demonstrate that hypersecretion of IGF1 is a critical factor for the development of obesity and can be targeted using AMPK activators to alleviate obesity.
Collapse
Affiliation(s)
- Ping Rong
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing, 210061, China
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China
| | - Yinqiu Mu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing, 210061, China
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China
| | - Meiqin Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing, 210061, China
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China
| | - Liang Chen
- College of Life Sciences, Anhui Medical University, Hefei, 230032, China
| | - Fangtong Liu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing, 210061, China
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China
| | - Yuxin Jin
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing, 210061, China
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China
| | - Weikuan Feng
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing, 210061, China
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China
| | - Kun Zhou
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing, 210061, China
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China
| | - Hui Liang
- Department of General Surgery, First Affiliated Hospital, Nanjing Medical University, Nanjing, 210029, China
| | - Hong-Yu Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing, 210061, China.
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China.
| | - Shuai Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing, 210061, China.
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China.
| |
Collapse
|
2
|
Mohamed I, Gautam M, Abosheaishaa H, Hussain S, Kumar K, Kotak A, Baugh M, Qureshi R, Jaber F, Dahiya DS, Alba L, Duong N. Growth hormone augmentation in metabolic dysfunction-associated steatotic liver disease: a systematic review and meta-analysis of randomized controlled trials. Eur J Gastroenterol Hepatol 2024; 36:1259-1266. [PMID: 38973533 DOI: 10.1097/meg.0000000000002819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/09/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is characterized by hepatic steatosis and metabolic dysregulation. Growth hormone (GH) augmentation has emerged as a potential therapeutic intervention for treating MASLD. This systematic review and meta-analysis aimed to evaluate the impact of GH augmentation on different parameters of MASLD. A systematic literature search identified randomized controlled trials investigating GH augmentation in MASLD patients. Search results were screened via Covidence and the Risk of Bias 2 tool was used to assess bias in randomized controlled trials. Statistical analysis utilized RevMan v5.3. We combined dichotomous outcomes employing odds ratios and continuous outcomes utilizing mean difference (MD), each with a 95% confidence interval (CI). Statistical significance was indicated by a P -value less than 0.05. Heterogeneity was evaluated using I2 tests. Our results showed that GH augmentation resulted in a significant reduction in both relative (MD: -46.26; 95% CI: -71.52, -21.00; P = 0.0003) and absolute (MD: -5.15; 95% CI: -7.93, -2.37; P = 0.0003) hepatic fat fraction. GH augmentation significantly reduced alanine aminotransferase (MD: -5.97; 95% CI: -10.31, -1.62; P = 0.007) and gamma-glutamyl transferase (MD: -16.18; 95% CI: -30.76, -1.59; P = 0.03) levels. No significant changes were observed in hemoglobin A1c, C-reactive protein, fasting serum glucose, BMI, triglycerides, and low-density lipoprotein cholesterol levels. Our meta-analysis highlights GH augmentation as a promising therapy for reducing liver steatosis and improving liver enzyme levels in MASLD patients. Further large-scale trials are warranted to examine the long-term effects, safety profiles, and potential impact on various measures.
Collapse
Affiliation(s)
- Islam Mohamed
- Department of Internal Medicine, University of Missouri-Kansas City, Kansas City, Missouri
| | - Misha Gautam
- Department of Internal Medicine, University of Missouri-Kansas City, Kansas City, Missouri
| | - Hazem Abosheaishaa
- Department of Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Sophia Hussain
- School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri
| | - Kopal Kumar
- School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri
| | - Anaya Kotak
- School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri
| | - Macy Baugh
- School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri
| | - Raabia Qureshi
- School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri
| | - Fouad Jaber
- Department of Internal Medicine, University of Missouri-Kansas City, Kansas City, Missouri
| | | | - Laura Alba
- Department of Gastroenterology and Hepatology, University of Missouri-Kansas City, Kansas City, Missouri
| | - Nikki Duong
- Department of Gastroenterology and Hepatology, Stanford University, Stanford, California, USA
| |
Collapse
|
3
|
Arisaka O, Koyama S, Imataka G, Naganuma J, Arisaka T, Akatsuka S. The Unexpected Detection of Esophageal Varices Caused by Liver Cirrhosis in a 47-Year-Old Man Treated with a Growth Hormone in Childhood. Diseases 2024; 12:251. [PMID: 39452494 PMCID: PMC11507581 DOI: 10.3390/diseases12100251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 10/06/2024] [Accepted: 10/07/2024] [Indexed: 10/26/2024] Open
Abstract
Background: We report a rare case highlighting the progression of liver disease in a male patient with idiopathic childhood-onset growth hormone (GH) deficiency. Case presentation: The patient was diagnosed with hypopituitarism at six years old and was treated with thyroxine therapy and GH for his short stature, with testosterone added at the age of 15. GH therapy was discontinued when the patient was 18 years old, but thyroid and testosterone treatments continued. The patient had been taking medication for hyperlipidemia until the age of 30 and was noted to have impaired glucose tolerance at the age of 40, but HbA1c levels remained normal. At the age of 47, esophageal varices were incidentally discovered via endoscopy, revealing liver cirrhosis. Laboratory tests showed liver dysfunction and abnormal lipid levels, and hepatitis viral markers were absent. The patient had no history of drinking alcohol or smoking, and no family history of diabetes. Results: Ultimately, this case demonstrates that metabolic dysfunction-associated steatotic liver disease (MASLD/metabolic dysfunction-associated steatohepatitis (MASH)) is under-recognized in GH deficiency cases and can progress to liver cirrhosis. Conclusions: Therefore, careful evaluation of MASLD/MASH in childhood-onset GH deficiency is necessary, and GH replacement therapy should continue into adulthood, if possible.
Collapse
Affiliation(s)
- Osamu Arisaka
- Department of Pediatrics, Dokkyo Medical University, Tochigi 321-0293, Japan; (S.K.); (G.I.); (J.N.)
| | - Satomi Koyama
- Department of Pediatrics, Dokkyo Medical University, Tochigi 321-0293, Japan; (S.K.); (G.I.); (J.N.)
| | - George Imataka
- Department of Pediatrics, Dokkyo Medical University, Tochigi 321-0293, Japan; (S.K.); (G.I.); (J.N.)
| | - Junko Naganuma
- Department of Pediatrics, Dokkyo Medical University, Tochigi 321-0293, Japan; (S.K.); (G.I.); (J.N.)
| | - Takahiro Arisaka
- Department of Gastroenterology, Dokkyo Medical University, Tochigi 321-0293, Japan;
| | - Sei Akatsuka
- Department of Pediatrics, Koto Hospital, Tokyo 136-0072, Japan
| |
Collapse
|
4
|
Xie RL, Liang R, Luo YY, Ruan ZH, Li YF, Liu WS. Growth Axis Somatostatin, Growth Hormone Receptor, and Insulin-like Growth Factor-1 Genes Express and Are Affected by the Injection of Exogenous Growth Hormone in Chinemys reevesii. Genes (Basel) 2023; 14:2032. [PMID: 38002975 PMCID: PMC10671357 DOI: 10.3390/genes14112032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 10/27/2023] [Accepted: 10/30/2023] [Indexed: 11/26/2023] Open
Abstract
In this study, to explore the effect of growth hormone changes on the related genes and regulatory roles of the turtle, PCR amplification, real-time fluorescence quantitative analysis, and enzyme cutting technology were used to clone and sequence the somatostatin (SS) gene, growth hormone receptor (GHR), and insulin-like growth factor-1 (IGF-I) sequence of Chinemys reevesii. The effects of human growth hormone on the mRNA expression of growth-axis-related genes SS, GHR, and IGF-1 in different sexes were observed. The study of the SS gene in turtles using real-time fluorescence quantitative PCR showed that the SS gene was mainly expressed in the nervous system and the digestive system, with the highest expression found in the brain, while the GHR gene and the IGF-I gene were expressed in all tissues of Chinemys reevesii. The SS gene was expressed in the brain, pituitary, liver, stomach, and intestine, with the highest expression in the brain and the lowest expression in the liver. Within 4 weeks of the injection of exogenous growth hormone, the expression level of the SS gene in the brain of both sexes first increased and then decreased, showing a parabolic trend, and the expression level of the experimental group was lower than that of the control group. After the injection of growth hormone (GH), the expression of the GHR gene in the liver of both sexes showed a significant increase in the first week, decreasing to the control group level in the second week, and then gradually increasing. Finally, a significant level of difference in the expression of the GHR gene was reached at 3 and 4 weeks. In terms of the IGF-I gene, the changing trend of the expression level in the liver was the same as that of the GHR gene. After the injection of exogenous growth hormone, although the expression of the SS gene increased the inhibition of the secretion of the GHR gene by the Reeves' turtle, exogenous growth hormone could replace the synthesis of GH and GHR, accelerating the growth of the turtle. The experiments showed that the injection of recombinant human growth hormone affects the expression of SS, GHR, and IGF-1 genes, and promotes the growth of the Reeves' turtle.
Collapse
Affiliation(s)
- Rui-Lin Xie
- College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China; (R.-L.X.); (Y.-Y.L.); (Y.-F.L.)
| | - Rui Liang
- Foshan Institute of Agricultural Science, Foshan 528251, China;
| | - Yuan-Yuan Luo
- College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China; (R.-L.X.); (Y.-Y.L.); (Y.-F.L.)
| | - Zhuo-Hao Ruan
- Laboratory of Aquatic Sciences, Key Laboratory of Animal Nutrition and Feed Science in South China of Ministry of Agriculture and Rural Affairs, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510610, China;
| | - Yi-Fu Li
- College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China; (R.-L.X.); (Y.-Y.L.); (Y.-F.L.)
| | - Wen-Sheng Liu
- College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China; (R.-L.X.); (Y.-Y.L.); (Y.-F.L.)
- Guangdong Province Engineering Research Centre of Aquatic Immunization and Aquaculture Health Techniques, South China Agricultural University, Guangzhou 510642, China
- University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, Guangzhou 510642, China
| |
Collapse
|
5
|
Moustaki M, Paschou SA, Xekouki P, Kotsa K, Peppa M, Psaltopoulou T, Kalantaridou S, Vryonidou A. Secondary diabetes mellitus in acromegaly. Endocrine 2023; 81:1-15. [PMID: 36882643 PMCID: PMC10239382 DOI: 10.1007/s12020-023-03339-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 02/21/2023] [Indexed: 03/09/2023]
Abstract
Secondary diabetes mellitus (DM) is a common complication of acromegaly, encountered in up to 55% of cases. Vice versa, the prevalence of acromegaly is markedly higher in cohorts of patients with type 2 DM (T2DM). The presence of secondary DM depends primarily on acromegaly status and is associated with increased cardiovascular morbidity, malignancy rate and overall mortality. The principal pathophysiologic mechanism is increased insulin resistance due to excessive lipolysis and altered fat distribution, reflected at the presence of intermuscular fat and attenuated, dysfunctional adipose tissue. Insulin resistance is ascribed to the direct, diabetogenic effects of growth hormone (GH), which prevail over the insulin-sensitizing effects of insulin-like growth factor 1 (IGF-1), probably due to higher glucometabolic potency of GH, IGF-1 resistance, or both. Inversely, GH and IGF-1 act synergistically in increasing insulin secretion. Hyperinsulinemia in portal vein leads to enhanced responsiveness of liver GH receptors and IGF-1 production, pointing towards a mutually amplifying loop between GH-IGF-1 axis and insulin. Secondary DM occurs upon beta cell exhaustion, principally due to gluco-lipo-toxicity. Somatostatin analogues inhibit insulin secretion; especially pasireotide (PASI) impairs glycaemic profile in up to 75% of cases, establishing a separate pathophysiologic entity, PASI-induced DM. In contrast, pegvisomant and dopamine agonizts improve insulin sensitivity. In turn, metformin, pioglitazone and sodium-glucose transporters 2 inhibitors might be disease-modifying by counteracting hyperinsulinemia or acting pleiotropically. Large, prospective cohort studies are needed to validate the above notions and define optimal DM management in acromegaly.
Collapse
Affiliation(s)
- Melpomeni Moustaki
- Department of Endocrinology and Diabetes Center, Hellenic Red Cross Hospital, Athens, Greece
| | - Stavroula A Paschou
- Endocrine Unit and Diabetes Center, Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece.
| | - Paraskevi Xekouki
- Department of Endocrinology and Diabetes, University General Hospital of Heraklion, School of Medicine, University of Crete, Heraklion, Greece
| | - Kalliopi Kotsa
- Endocrine Unit and Diabetes Center, First Department of Internal Medicine, AHEPA University Hospital, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Melpomeni Peppa
- Endocrine Unit and Diabetes Center, Second Department of Internal Medicine, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Theodora Psaltopoulou
- Endocrine Unit and Diabetes Center, Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Sophia Kalantaridou
- 3rd Department of Obstetrics and Gynecology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Andromachi Vryonidou
- Department of Endocrinology and Diabetes Center, Hellenic Red Cross Hospital, Athens, Greece
| |
Collapse
|
6
|
Al-Samerria S, Radovick S. Exploring the Therapeutic Potential of Targeting GH and IGF-1 in the Management of Obesity: Insights from the Interplay between These Hormones and Metabolism. Int J Mol Sci 2023; 24:9556. [PMID: 37298507 PMCID: PMC10253584 DOI: 10.3390/ijms24119556] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/23/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023] Open
Abstract
Obesity is a growing public health problem worldwide, and GH and IGF-1 have been studied as potential therapeutic targets for managing this condition. This review article aims to provide a comprehensive view of the interplay between GH and IGF-1 and metabolism within the context of obesity. We conducted a systematic review of the literature that was published from 1993 to 2023, using MEDLINE, Embase, and Cochrane databases. We included studies that investigated the effects of GH and IGF-1 on adipose tissue metabolism, energy balance, and weight regulation in humans and animals. Our review highlights the physiological functions of GH and IGF-1 in adipose tissue metabolism, including lipolysis and adipogenesis. We also discuss the potential mechanisms underlying the effects of these hormones on energy balance, such as their influence on insulin sensitivity and appetite regulation. Additionally, we summarize the current evidence regarding the efficacy and safety of GH and IGF-1 as therapeutic targets for managing obesity, including in pharmacological interventions and hormone replacement therapy. Finally, we address the challenges and limitations of targeting GH and IGF-1 in obesity management.
Collapse
Affiliation(s)
- Sarmed Al-Samerria
- Laboratory of Human Growth and Reproductive Development, Department of Pediatrics, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA;
| | | |
Collapse
|
7
|
Backe MB, Andersen RC, Jensen M, Jin C, Hundahl C, Dmytriyeva O, Treebak JT, Hansen JB, Gerhart-Hines Z, Madsen KL, Holst B. PICK1-Deficient Mice Maintain Their Glucose Tolerance During Diet-Induced Obesity. J Endocr Soc 2023; 7:bvad057. [PMID: 37200849 PMCID: PMC10185814 DOI: 10.1210/jendso/bvad057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Indexed: 05/20/2023] Open
Abstract
Context Metabolic disorders such as obesity represent a major health challenge. Obesity alone has reached epidemic proportions, with at least 2.8 million people worldwide dying annually from diseases caused by overweight or obesity. The brain-metabolic axis is central to maintain homeostasis under metabolic stress via an intricate signaling network of hormones. Protein interacting with C kinase 1 (PICK1) is important for the biogenesis of various secretory vesicles, and we have previously shown that PICK1-deficient mice have impaired secretion of insulin and growth hormone. Objective The aim was to investigate how global PICK1-deficient mice respond to high-fat diet (HFD) and assess its role in insulin secretion in diet-induced obesity. Methods We characterized the metabolic phenotype through assessment of body weight, composition, glucose tolerance, islet morphology insulin secretion in vivo, and glucose-stimulated insulin secretion ex vivo. Results PICK1-deficient mice displayed similar weight gain and body composition as wild-type (WT) mice following HFD. While HFD impaired glucose tolerance of WT mice, PICK1-deficient mice were resistant to further deterioration of their glucose tolerance compared with already glucose-impaired chow-fed PICK1-deficient mice. Surprisingly, mice with β-cell-specific knockdown of PICK1 showed impaired glucose tolerance both on chow and HFD similar to WT mice. Conclusion Our findings support the importance of PICK1 in overall hormone regulation. However, importantly, this effect is independent of the PICK1 expression in the β-cell, whereby global PICK1-deficient mice resist further deterioration of their glucose tolerance following diet-induced obesity.
Collapse
Affiliation(s)
- Marie Balslev Backe
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Department of Clinical Epidemiology, Steno Diabetes Center Copenhagen, 2730 Herlev, Denmark
| | - Rita Chan Andersen
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Morten Jensen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Chunyu Jin
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Cecilie Hundahl
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Oksana Dmytriyeva
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Jonas T Treebak
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Jakob Bondo Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Zach Gerhart-Hines
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Kenneth L Madsen
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Birgitte Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
8
|
Gancheva S, Kahl S, Herder C, Strassburger K, Sarabhai T, Pafili K, Szendroedi J, Schlensak M, Roden M. Metabolic surgery-induced changes of the growth hormone system relate to improved adipose tissue function. Int J Obes (Lond) 2023; 47:505-511. [PMID: 36959287 DOI: 10.1038/s41366-023-01292-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 02/26/2023] [Accepted: 03/01/2023] [Indexed: 03/25/2023]
Abstract
AIMS Body weight loss improves insulin resistance and growth hormone secretion in obesity, which may be regulated by leptin according to preclinical studies. How changes in leptin, lipids and insulin sensitivity after bariatric (metabolic) surgery affect the human growth hormone system is yet unclear. PARTICIPANTS AND METHODS People with obesity (OBE, n = 79, BMI 50.8 ± 6.3 kg/m2) were studied before, 2, 12, 24 and 52 weeks after metabolic surgery and compared to lean healthy humans (control; CON, n = 24, BMI 24.3 ± 3.1 kg/m2). Tissue-specific insulin sensitivity was assessed by hyperinsulinemic-euglycemic clamps with D-[6,6-2H2]glucose. Fasting leptin, growth hormone (GH), insulin-like growth factor 1 (IGF-1) and IGF-binding proteins (IGFBP1, IGFBP3) were measured using ELISA. RESULTS At baseline, OBE exhibited higher glycemia and leptinemia as well as pronounced peripheral, adipose tissue and hepatic insulin resistance compared to CON. GH and IGFBP1 were lower, while IGF1 was comparable between groups. At 52 weeks, OBE had lost 33% body weight and doubled their peripheral insulin sensitivity, which was paralleled by continuous increases in GH, IGF-1 and IGFBP1 as well as decrease in leptin. The rise in GH correlated with reductions in free fatty acids, adipose tissue insulin resistance and insulinemia, but not with changes in body weight, peripheral insulin sensitivity, glycemia or leptinemia. The rise in IGF-1 correlated with reduction in high-sensitive C-reactive protein. CONCLUSION Reversal of alterations of the GH-IGF-1 axis after surgically-induced weight loss is unlikely related to improved leptin secretion and/or insulin sensitivity, but is rather associated with restored adipose tissue function and reduced low-grade inflammation.
Collapse
Affiliation(s)
- Sofiya Gancheva
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital, Heinrich-Heine University, Düsseldorf, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Partner Düsseldorf, Munich-Neuherberg, Düsseldorf, Germany
| | - Sabine Kahl
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Partner Düsseldorf, Munich-Neuherberg, Düsseldorf, Germany
| | - Christian Herder
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital, Heinrich-Heine University, Düsseldorf, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Partner Düsseldorf, Munich-Neuherberg, Düsseldorf, Germany
| | - Klaus Strassburger
- German Center for Diabetes Research (DZD e.V.), Partner Düsseldorf, Munich-Neuherberg, Düsseldorf, Germany
- Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
| | - Theresia Sarabhai
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital, Heinrich-Heine University, Düsseldorf, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Partner Düsseldorf, Munich-Neuherberg, Düsseldorf, Germany
| | - Kalliopi Pafili
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital, Heinrich-Heine University, Düsseldorf, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Partner Düsseldorf, Munich-Neuherberg, Düsseldorf, Germany
| | - Julia Szendroedi
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital, Heinrich-Heine University, Düsseldorf, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Partner Düsseldorf, Munich-Neuherberg, Düsseldorf, Germany
- Department of Internal Medicine I and Clinical Chemistry, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Michael Roden
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital, Heinrich-Heine University, Düsseldorf, Germany.
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany.
- German Center for Diabetes Research (DZD e.V.), Partner Düsseldorf, Munich-Neuherberg, Düsseldorf, Germany.
| |
Collapse
|
9
|
Fraenkel E, Lazurova I. IGF-1 and IGFBP3 as indirect markers of hepatic insulin resistance and their relation to metabolic syndrome parameters in liver steatosis patients. Endocr Regul 2023; 57:69-79. [PMID: 37183691 DOI: 10.2478/enr-2023-0009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/16/2023] Open
Abstract
Objective. The aim of the present study was to assess insulin-like growth factor 1 (IGF-1) and IGF-binding protein 3 (IGFBP3) as markers of insulin resistance in patients with prediabetes and type 2 diabetes mellitus (TDM2). Patients and Methods. This observational clinical study included 76 obese/overweight patients at the age of 45-75 years with T2DM on oral diabetic medication and ultrasonographically or by a computerized tomography (CT) diagnosed liver steatosis. Correlation analysis was performed between plasma levels of insulin, C-peptide, IGF-1, IGFBP3 and HOMA indexes on the one hand and between plasma levels of ALT, AST, triglyceride, cholesterol, and HDL cholesterol and body mass index (BMI) of patients on the other hand. In case of significant partial correlation coefficients, a multiple linear regression model with IGF-1 and IGFBP3 used as outcome variables adjusted for age and sex groups was calculated. According to these regression models, ROC curves were prepared with HOMA index=3 used as a classificator of insulin resistance. Results. Significant correlation was found between C-peptide and IGF-1 (r=0.24, p≤0.05), C-peptide and IGFBP3 (r=0.24, p≤0.05), IGFBP3 and cholesterol (r=0.22, p≤0.05) IGFBP3 and ALT (r=0.19, p≤0.05), HOMA index and triglycerides (r=0.22, p≤0.05), and HOMA index and ALT (r=0.23, p≤0.05). Significant correlation adjusted for age and gender was found between C-peptide and IGF-1 plasma levels (R2=0.20, p<0.05) with AUROC 0.685 (p≤0.01) and C-peptide and IGFBP3 plasma levels (R2=0.28, p<0.05) with AUROC 0.684 (p≤0.01). Significant correlation adjusted for age and gender was found between triglyceride and IGFBP3 plasma levels (R2=0.28, p<0.05) with AUROC 0.616 (p≤0.01). After the distribution of patients according to their IGFBP3 levels, we found a difference between the 1st and the 4th quartiles in terms of triglyceride levels. Conclusion. Our results demonstrate a fundamental role of IGF-1 and IGFBP3 in the patho-physiology of hepatic insulin resistance and suggest them as indirect indicators of the hepatic insulin resistance.
Collapse
Affiliation(s)
- Emil Fraenkel
- 11st Department of Internal Medicine, P.J. Safarik University, Kosice, Slovakia
| | - Ivica Lazurova
- 11st Department of Internal Medicine, P.J. Safarik University, Kosice, Slovakia
| |
Collapse
|
10
|
Tavares MR, Frazao R, Donato J. Understanding the role of growth hormone in situations of metabolic stress. J Endocrinol 2023; 256:JOE-22-0159. [PMID: 36327147 DOI: 10.1530/joe-22-0159] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/03/2022] [Indexed: 11/06/2022]
Abstract
Growth hormone (GH) is secreted by the anterior pituitary gland and plays a key role in controlling tissue and body growth. While basal GH secretion is considerably reduced along adulthood and aging, several situations of metabolic stress can lead to robust increases in circulating GH levels. The objective of the present review is to summarize and discuss the importance of GH regulating different physiological functions in situations of metabolic stress, including prolonged food restriction, hypoglycemia, exercise, pregnancy, and obesity. The presented data indicate that GH increases hunger perception/food intake, fat mobilization, blood glucose levels, and insulin resistance and produces changes in energy expenditure and neuroendocrine responses during metabolic challenges. When all these effects are considered in the context of situations of metabolic stress, they contribute to restore homeostasis by (1) helping the organism to use appropriate energy substrates, (2) preventing hypoglycemia or increasing the availability of glucose, (3) stimulating feeding to provide nutrients in response to energy-demanding activities or to accelerate the recovery of energy stores, and (4) affecting the activity of neuronal populations involved in the control of metabolism and stress response. Thus, the central and peripheral effects of GH coordinate multiple adaptations during situations of metabolic stress that ultimately help the organism restore homeostasis, increasing the chances of survival.
Collapse
Affiliation(s)
- Mariana Rosolen Tavares
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Renata Frazao
- Department of Anatomy, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Jose Donato
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
11
|
Gardner EJ, Kentistou KA, Stankovic S, Lockhart S, Wheeler E, Day FR, Kerrison ND, Wareham NJ, Langenberg C, O'Rahilly S, Ong KK, Perry JR. Damaging missense variants in IGF1R implicate a role for IGF-1 resistance in the etiology of type 2 diabetes. CELL GENOMICS 2022; 2:None. [PMID: 36530175 PMCID: PMC9750938 DOI: 10.1016/j.xgen.2022.100208] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 08/12/2022] [Accepted: 10/07/2022] [Indexed: 11/09/2022]
Abstract
Type 2 diabetes (T2D) is a heritable metabolic disorder. While population studies have identified hundreds of common genetic variants associated with T2D, the role of rare (frequency < 0.1%) protein-coding variation is less clear. We performed exome sequence analysis in 418,436 (n = 32,374 T2D cases) individuals in the UK Biobank. We identified previously reported genes (GCK, GIGYF1, HNF1A) in addition to missense variants in ZEB2 (n = 31 carriers; odds ratio [OR] = 5.5 [95% confidence interval = 2.5-12.0]; p = 6.4 × 10-7), MLXIPL (n = 245; OR = 2.3 [1.6-3.2]; p = 3.2 × 10-7), and IGF1R (n = 394; OR = 2.4 [1.8-3.2]; p = 1.3 × 10-10). Carriers of damaging missense variants within IGF1R were also shorter (-2.2 cm [-1.8 to -2.7]; p = 1.2 × 10-19) and had higher circulating insulin-like growth factor-1 (IGF-1) protein levels (2.3 nmol/L [1.7-2.9]; p = 2.8 × 10-14), indicating relative IGF-1 resistance. A likely causal role of IGF-1 resistance was supported by Mendelian randomization analyses using common variants. These results increase understanding of the genetic architecture of T2D and highlight the growth hormone/IGF-1 axis as a potential therapeutic target.
Collapse
Affiliation(s)
- Eugene J. Gardner
- MRC Epidemiology Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Katherine A. Kentistou
- MRC Epidemiology Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Stasa Stankovic
- MRC Epidemiology Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Samuel Lockhart
- MRC Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Eleanor Wheeler
- MRC Epidemiology Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Felix R. Day
- MRC Epidemiology Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Nicola D. Kerrison
- MRC Epidemiology Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Nicholas J. Wareham
- MRC Epidemiology Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Claudia Langenberg
- MRC Epidemiology Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Computational Medicine, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Stephen O'Rahilly
- MRC Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - Ken K. Ong
- MRC Epidemiology Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - John R.B. Perry
- MRC Epidemiology Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Metabolic Research Laboratory, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| |
Collapse
|
12
|
Wang W, Tesfay EB, van Klinken JB, Willems van Dijk K, Bartke A, van Heemst D, Noordam R. Clustered Mendelian randomization analyses identify distinct and opposing pathways in the association between genetically influenced insulin-like growth factor-1 and type 2 diabetes mellitus. Int J Epidemiol 2022; 51:1874-1885. [PMID: 35656699 PMCID: PMC9749721 DOI: 10.1093/ije/dyac119] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 05/17/2022] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND There is inconsistent evidence for the causal role of serum insulin-like growth factor-1 (IGF-1) concentration in the pathogenesis of human age-related diseases such as type 2 diabetes (T2D). Here, we investigated the association between IGF-1 and T2D using (clustered) Mendelian randomization (MR) analyses in the UK Biobank. METHODS We conducted Cox proportional hazard analyses in 451 232 European-ancestry individuals of the UK Biobank (55.3% women, mean age at recruitment 56.6 years), among which 13 247 individuals developed type 2 diabetes during up to 12 years of follow-up. In addition, we conducted two-sample MR analyses based on independent single nucleotide polymorphisms (SNPs) associated with IGF-1. Given the heterogeneity between the MR effect estimates of individual instruments (P-value for Q statistic = 4.03e-145), we also conducted clustered MR analyses. Biological pathway analyses of the identified clusters were performed by over-representation analyses. RESULTS In the Cox proportional hazard models, with IGF-1 concentrations stratified in quintiles, we observed that participants in the lowest quintile had the highest relative risk of type 2 diabetes [hazard ratio (HR): 1.31; 95% CI: 1.23-1.39). In contrast, in the two-sample MR analyses, higher genetically influenced IGF-1 was associated with a higher risk of type 2 diabetes. Based on the heterogeneous distribution of MR effect estimates of individual instruments, six clusters of genetically determined IGF-1 associated either with a lower or a higher risk of type 2 diabetes were identified. The main clusters in which a higher IGF-1 was associated with a lower risk of type 2 diabetes consisted of instruments mapping to genes in the growth hormone signalling pathway, whereas the main clusters in which a higher IGF-1 was associated with a higher risk of type 2 diabetes consisted of instruments mapping to genes in pathways related to amino acid metabolism and genomic integrity. CONCLUSIONS The IGF-1-associated SNPs used as genetic instruments in MR analyses showed a heterogeneous distribution of MR effect estimates on the risk of type 2 diabetes. This was likely explained by differences in the underlying molecular pathways that increase IGF-1 concentration and differentially mediate the effects of IGF-1 on type 2 diabetes.
Collapse
Affiliation(s)
- Wenyi Wang
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Ephrem Baraki Tesfay
- Department of Internal Medicine, Section of Gerontology and Geriatrics; Leiden University Medical Center, Leiden, The Netherlands
| | - Jan Bert van Klinken
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands.,Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Core Facility Metabolomics, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Ko Willems van Dijk
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands.,Department of Internal Medicine, Division Endocrinology, Leiden University Medical Center, Leiden, The Netherlands.,Leiden Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Andrzej Bartke
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Diana van Heemst
- Department of Internal Medicine, Section of Gerontology and Geriatrics; Leiden University Medical Center, Leiden, The Netherlands
| | - Raymond Noordam
- Department of Internal Medicine, Section of Gerontology and Geriatrics; Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
13
|
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
|
14
|
Kabbani M, Michailidis E, Steensels S, Fulmer CG, Luna JM, Le Pen J, Tardelli M, Razooky B, Ricardo-Lax I, Zou C, Zeck B, Stenzel AF, Quirk C, Foquet L, Ashbrook AW, Schneider WM, Belkaya S, Lalazar G, Liang Y, Pittman M, Devisscher L, Suemizu H, Theise ND, Chiriboga L, Cohen DE, Copenhaver R, Grompe M, Meuleman P, Ersoy BA, Rice CM, de Jong YP. Human hepatocyte PNPLA3-148M exacerbates rapid non-alcoholic fatty liver disease development in chimeric mice. Cell Rep 2022; 40:111321. [PMID: 36103835 PMCID: PMC11587767 DOI: 10.1016/j.celrep.2022.111321] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 05/11/2022] [Accepted: 08/16/2022] [Indexed: 11/28/2022] Open
Abstract
Advanced non-alcoholic fatty liver disease (NAFLD) is a rapidly emerging global health problem associated with pre-disposing genetic polymorphisms, most strikingly an isoleucine to methionine substitution in patatin-like phospholipase domain-containing protein 3 (PNPLA3-I148M). Here, we study how human hepatocytes with PNPLA3 148I and 148M variants engrafted in the livers of broadly immunodeficient chimeric mice respond to hypercaloric diets. As early as four weeks, mice developed dyslipidemia, impaired glucose tolerance, and steatosis with ballooning degeneration selectively in the human graft, followed by pericellular fibrosis after eight weeks of hypercaloric feeding. Hepatocytes with the PNPLA3-148M variant, either from a homozygous 148M donor or overexpressed in a 148I donor background, developed microvesicular and severe steatosis with frequent ballooning degeneration, resulting in more active steatohepatitis than 148I hepatocytes. We conclude that PNPLA3-148M in human hepatocytes exacerbates NAFLD. These models will facilitate mechanistic studies into human genetic variant contributions to advanced fatty liver diseases.
Collapse
Affiliation(s)
- Mohammad Kabbani
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA; Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Eleftherios Michailidis
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA; Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University, Atlanta, GA 30322, USA
| | - Sandra Steensels
- Division of Gastroenterology and Hepatology, Weill Cornell Medicine, 413 East 69th Street, BB626, New York, NY 10065, USA
| | - Clifton G Fulmer
- Department of Pathology, Weill Cornell Medicine, New York, NY 10065, USA; Robert J. Tomsich Pathology and Laboratory Medicine Institute, The Cleveland Clinic, Cleveland, OH 44195, USA
| | - Joseph M Luna
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - Jérémie Le Pen
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - Matteo Tardelli
- Division of Gastroenterology and Hepatology, Weill Cornell Medicine, 413 East 69th Street, BB626, New York, NY 10065, USA
| | - Brandon Razooky
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - Inna Ricardo-Lax
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - Chenhui Zou
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA; Division of Gastroenterology and Hepatology, Weill Cornell Medicine, 413 East 69th Street, BB626, New York, NY 10065, USA
| | - Briana Zeck
- Department of Pathology, NYU Langone, New York, NY 10028, USA
| | - Ansgar F Stenzel
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA; Department of Infectious Diseases, Molecular Virology, Heidelberg University, Heidelberg, Germany
| | - Corrine Quirk
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | | | - Alison W Ashbrook
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - William M Schneider
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - Serkan Belkaya
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, NY 10065, USA
| | - Gadi Lalazar
- Division of Gastroenterology and Hepatology, Weill Cornell Medicine, 413 East 69th Street, BB626, New York, NY 10065, USA; Laboratory of Cellular Biophysics, The Rockefeller University, New York, NY 10065, USA
| | - Yupu Liang
- Center for Clinical and Translational Science, The Rockefeller University, New York, NY 10065, USA
| | - Meredith Pittman
- Department of Pathology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Lindsey Devisscher
- Department of Basic and Applied Medical Sciences, Gut-Liver Immunopharmacology Unit, Ghent University, Ghent, Belgium
| | | | - Neil D Theise
- Department of Pathology, NYU Langone, New York, NY 10028, USA
| | - Luis Chiriboga
- Department of Pathology, NYU Langone, New York, NY 10028, USA
| | - David E Cohen
- Division of Gastroenterology and Hepatology, Weill Cornell Medicine, 413 East 69th Street, BB626, New York, NY 10065, USA
| | | | - Markus Grompe
- Yecuris Corporation, Tualatin, OR 97062, USA; Department of Pediatrics, Oregon Stem Cell Center, Oregon Health and Science University, Portland, OR 97239, USA
| | - Philip Meuleman
- Laboratory of Liver Infectious Diseases, Ghent University, Ghent, Belgium
| | - Baran A Ersoy
- Division of Gastroenterology and Hepatology, Weill Cornell Medicine, 413 East 69th Street, BB626, New York, NY 10065, USA
| | - Charles M Rice
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - Ype P de Jong
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA; Division of Gastroenterology and Hepatology, Weill Cornell Medicine, 413 East 69th Street, BB626, New York, NY 10065, USA.
| |
Collapse
|
15
|
Doycheva I, Erickson D, Watt KD. Growth hormone deficiency and NAFLD: An overlooked and underrecognized link. Hepatol Commun 2022; 6:2227-2237. [PMID: 35765700 PMCID: PMC9426379 DOI: 10.1002/hep4.1953] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/07/2022] [Accepted: 03/13/2022] [Indexed: 11/17/2022] Open
Abstract
Growth hormone and its mediator insulin‐like growth factor‐1 exert their effect on different organs and control various physiologic metabolic processes. Adult growth hormone deficiency (AGHD) presents with one or more components of metabolic syndrome and can be associated with nonalcoholic fatty liver disease (NAFLD). AGHD is present in spectrum of hypothalamic/pituitary disorders as well as cranial radiation of brain tumors and often remains underdiagnosed or untreated due to its nonspecific symptoms, relatively difficult diagnosis in some clinical scenarios, and various barriers to treatment. NAFLD usually develops soon after diagnosis of AGHD and might progress rapidly to nonalcoholic steatohepatitis (NASH) with advanced fibrosis, eventually requiring liver transplantation. A timely initiation of growth hormone replacement therapy might be important, although studies so far have demonstrated controversial results on NAFLD, primarily due to small sample size and different diagnostic methods of NAFLD. Increased awareness of the association between AGHD and NAFLD would facilitate early diagnosis of NAFLD and NASH if present. Therefore, a multidisciplinary approach involving hepatology and endocrinology should become a standard of care for these patients.
Collapse
Affiliation(s)
- Iliana Doycheva
- Section of Endocrinology, Diabetes, and Metabolism, University of Chicago, Chicago, Illinois, USA
| | - Dana Erickson
- Division of Endocrinology, Metabolism and Nutrition, Mayo Clinic, Rochester, Minnesota, USA
| | - Kymberly D Watt
- Gastroenterology and Hepatology Department, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
16
|
Dichtel LE, Cordoba-Chacon J, Kineman RD. Growth Hormone and Insulin-Like Growth Factor 1 Regulation of Nonalcoholic Fatty Liver Disease. J Clin Endocrinol Metab 2022; 107:1812-1824. [PMID: 35172328 PMCID: PMC9202731 DOI: 10.1210/clinem/dgac088] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Indexed: 11/19/2022]
Abstract
Patients with obesity have a high prevalence of nonalcoholic fatty liver disease (NAFLD), representing a spectrum of simple steatosis to nonalcoholic steatohepatitis (NASH), without and with fibrosis. Understanding the etiology of NAFLD is clinically relevant since NAFLD is an independent risk factor for diabetes and cardiovascular disease. In addition, NASH predisposes patients to the development of cirrhosis and hepatocellular carcinoma, and NASH cirrhosis represents the fastest growing indication for liver transplantation in the United States. It is appreciated that multiple factors are involved in the development and progression of NAFLD. Growth hormone (GH) and insulin-like growth factor 1 (IGF1) regulate metabolic, immune, and hepatic stellate cell function, and alterations in the production and function of GH is associated with obesity and NAFLD/NASH. Therefore, this review will focus on the potential role of GH and IGF1 in the regulation of hepatic steatosis, inflammation, and fibrosis.
Collapse
Affiliation(s)
- Laura E Dichtel
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Jose Cordoba-Chacon
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Illinois at Chicago, Chicago, IL, USA
| | - Rhonda D Kineman
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Illinois at Chicago, Chicago, IL, USA
- Jesse Brown VA Medical Center, Research and Development Division, Chicago, IL, USA
| |
Collapse
|
17
|
Brown-Borg HM. Growth hormone, not IGF-1 is the key longevity regulator in mammals. J Gerontol A Biol Sci Med Sci 2022; 77:1719-1723. [PMID: 35436323 PMCID: PMC9434454 DOI: 10.1093/gerona/glac092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Holly M Brown-Borg
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND
| |
Collapse
|
18
|
Dietary acid load and risk of type 2 diabetes mellitus: A case-control study. Clin Nutr ESPEN 2022; 48:308-312. [DOI: 10.1016/j.clnesp.2022.01.029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 01/15/2022] [Accepted: 01/23/2022] [Indexed: 11/23/2022]
|
19
|
Towards Understanding the Direct and Indirect Actions of Growth Hormone in Controlling Hepatocyte Carbohydrate and Lipid Metabolism. Cells 2021; 10:cells10102532. [PMID: 34685512 PMCID: PMC8533955 DOI: 10.3390/cells10102532] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 09/20/2021] [Accepted: 09/23/2021] [Indexed: 02/06/2023] Open
Abstract
Growth hormone (GH) is critical for achieving normal structural growth. In addition, GH plays an important role in regulating metabolic function. GH acts through its GH receptor (GHR) to modulate the production and function of insulin-like growth factor 1 (IGF1) and insulin. GH, IGF1, and insulin act on multiple tissues to coordinate metabolic control in a context-specific manner. This review will specifically focus on our current understanding of the direct and indirect actions of GH to control liver (hepatocyte) carbohydrate and lipid metabolism in the context of normal fasting (sleep) and feeding (wake) cycles and in response to prolonged nutrient deprivation and excess. Caveats and challenges related to the model systems used and areas that require further investigation towards a clearer understanding of the role GH plays in metabolic health and disease are discussed.
Collapse
|
20
|
Haffner D, Grund A, Leifheit-Nestler M. Renal effects of growth hormone in health and in kidney disease. Pediatr Nephrol 2021; 36:2511-2530. [PMID: 34143299 PMCID: PMC8260426 DOI: 10.1007/s00467-021-05097-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 04/27/2021] [Indexed: 12/29/2022]
Abstract
Growth hormone (GH) and its mediator insulin-like growth factor-1 (IGF-1) have manifold effects on the kidneys. GH and IGF receptors are abundantly expressed in the kidney, including the glomerular and tubular cells. GH can act either directly on the kidneys or via circulating or paracrine-synthesized IGF-1. The GH/IGF-1 system regulates glomerular hemodynamics, renal gluconeogenesis, tubular sodium and water, phosphate, and calcium handling, as well as renal synthesis of 1,25 (OH)2 vitamin D3 and the antiaging hormone Klotho. The latter also acts as a coreceptor of the phosphaturic hormone fibroblast-growth factor 23 in the proximal tubule. Recombinant human GH (rhGH) is widely used in the treatment of short stature in children, including those with chronic kidney disease (CKD). Animal studies and observations in acromegalic patients demonstrate that GH-excess can have deleterious effects on kidney health, including glomerular hyperfiltration, renal hypertrophy, and glomerulosclerosis. In addition, elevated GH in patients with poorly controlled type 1 diabetes mellitus was thought to induce podocyte injury and thereby contribute to the development of diabetic nephropathy. This manuscript gives an overview of the physiological actions of GH/IGF-1 on the kidneys and the multiple alterations of the GH/IGF-1 system and its consequences in patients with acromegaly, CKD, nephrotic syndrome, and type 1 diabetes mellitus. Finally, the impact of short- and long-term treatment with rhGH/rhIGF-1 on kidney function in patients with kidney diseases will be discussed.
Collapse
Affiliation(s)
- Dieter Haffner
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Pediatric Research Center, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
- Pediatric Research Center, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| | - Andrea Grund
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Pediatric Research Center, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
- Pediatric Research Center, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Maren Leifheit-Nestler
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Pediatric Research Center, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
- Pediatric Research Center, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| |
Collapse
|
21
|
Low levels of sulfur and cobalt during the pre- and periconceptional periods affect the oocyte yield of donors and the DNA methylome of preimplantation bovine embryos. J Dev Orig Health Dis 2021; 13:231-243. [PMID: 33941306 DOI: 10.1017/s2040174421000222] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Maternal nutrition is critical in mammalian development, influencing the epigenetic reprogramming of gametes, embryos, and fetal programming. We evaluated the effects of different levels of sulfur (S) and cobalt (Co) in the maternal diet throughout the pre- and periconceptional periods on the biochemical and reproductive parameters of the donors and the DNA methylome of the progeny in Bos indicus cattle. The low-S/Co group differed from the control with respect to homocysteine, folic acid, B12, insulin growth factor 1, and glucose. The oocyte yield was lower in heifers from the low S/Co group than that in the control heifers. Embryos from the low-S/Co group exhibited 2320 differentially methylated regions (DMRs) across the genome compared with the control embryos. We also characterized candidate DMRs linked to the DNMT1 and DNMT3B genes in the blood and sperm cells of the adult progeny. A DMR located in DNMT1 that was identified in embryos remained differentially methylated in the sperm of the progeny from the low-S/Co group. Therefore, we associated changes in specific compounds in the maternal diet with DNA methylation modifications in the progeny. Our results help to elucidate the impact of maternal nutrition on epigenetic reprogramming in livestock, opening new avenues of research to study the effect of disturbed epigenetic patterns in early life on health and fertility in adulthood. Considering that cattle are physiologically similar to humans with respect to gestational length, our study may serve as a model for studies related to the developmental origin of health and disease in humans.
Collapse
|
22
|
Gossing W, Radke L, Biering H, Diederich S, Mai K, Frohme M. The ElonginB/C-Cullin5-SOCS-Box-Complex Is a Potential Biomarker for Growth Hormone Disorders. Biomedicines 2021; 9:biomedicines9020201. [PMID: 33671326 PMCID: PMC7921923 DOI: 10.3390/biomedicines9020201] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 01/31/2021] [Accepted: 02/08/2021] [Indexed: 01/29/2023] Open
Abstract
Insulin-like growth factor 1 (IGF-1) is the standard biochemical marker for the diagnosis and treatment control of acromegaly and growth hormone deficiency (GHD). However, its limitations necessitate the screening for new specific and sensitive biomarkers. The elonginB/C-cullin5-SOCS-box-complex (ECS-complex) (an intracellular five-protein complex) is stimulated by circulating growth hormone (GH) and regulates GH receptor levels through a negative feedback loop. It mediates the cells' sensitivity for GH and therefore, represents a potent new biomarker for those diseases. In this study, individual ECS-complex proteins were measured in whole blood samples of patients with acromegaly (n = 32) or GHD (n = 12) via ELISA and compared to controls. Hierarchical clustering of the results revealed that by combining the three ECS-complex proteins suppressor of cytokine signaling 2 (SOCS2), cullin-5 and ring-box protein 2 (Rbx-2), 93% of patient samples could be separated from controls, despite many patients having a normal IGF-1 or not receiving medical treatment. SOCS2 showed the best individual diagnostic performance with an overall accuracy of 0.93, while the combination of the three proteins correctly identified all patients and controls. This resulted in perfect sensitivity and specificity for all patient groups, which demonstrates potential benefits of the ECS-complex proteins as clinical biomarkers for the diagnostics of GH-related diseases and substantiates their important role in GH metabolism.
Collapse
Affiliation(s)
- Wilhelm Gossing
- Division Molecular Biotechnology and Functional Genomics, Technical University of Applied Sciences, 15745 Wildau, Germany; (W.G.); (L.R.); (H.B.)
- Department of Endocrinology and Metabolism, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (S.D.); (K.M.)
| | - Lars Radke
- Division Molecular Biotechnology and Functional Genomics, Technical University of Applied Sciences, 15745 Wildau, Germany; (W.G.); (L.R.); (H.B.)
| | - Henrik Biering
- Division Molecular Biotechnology and Functional Genomics, Technical University of Applied Sciences, 15745 Wildau, Germany; (W.G.); (L.R.); (H.B.)
- Praxis an der Kaisereiche, Innere Medizin, Endokrinologie, Diabetologie, 12159 Berlin, Germany
| | - Sven Diederich
- Department of Endocrinology and Metabolism, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (S.D.); (K.M.)
- MVZ Medicover Berlin-Mitte, Innere Medizin, Endokrinologie, Andrologie, 10117 Berlin, Germany
| | - Knut Mai
- Department of Endocrinology and Metabolism, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (S.D.); (K.M.)
- German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, 10117 Berlin, Germany
| | - Marcus Frohme
- Division Molecular Biotechnology and Functional Genomics, Technical University of Applied Sciences, 15745 Wildau, Germany; (W.G.); (L.R.); (H.B.)
- Correspondence:
| |
Collapse
|
23
|
Deguise MO, Pileggi C, De Repentigny Y, Beauvais A, Tierney A, Chehade L, Michaud J, Llavero-Hurtado M, Lamont D, Atrih A, Wishart TM, Gillingwater TH, Schneider BL, Harper ME, Parson SH, Kothary R. SMN Depleted Mice Offer a Robust and Rapid Onset Model of Nonalcoholic Fatty Liver Disease. Cell Mol Gastroenterol Hepatol 2021; 12:354-377.e3. [PMID: 33545428 PMCID: PMC8257458 DOI: 10.1016/j.jcmgh.2021.01.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 01/26/2021] [Accepted: 01/27/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS Nonalcoholic fatty liver disease (NAFLD) is considered a health epidemic with potential devastating effects on the patients and the healthcare systems. Current preclinical models of NAFLD are invariably imperfect and generally take a long time to develop. A mouse model of survival motor neuron (SMN) depletion (Smn2B/- mice) was recently shown to develop significant hepatic steatosis in less than 2 weeks from birth. The rapid onset of fatty liver in Smn2B/- mice provides an opportunity to identify molecular markers of NAFLD. Here, we investigated whether Smn2B/- mice display typical features of NAFLD/nonalcoholic steatohepatitis (NASH). METHODS Biochemical, histologic, electron microscopy, proteomic, and high-resolution respirometry were used. RESULTS The Smn2B/- mice develop microvesicular steatohepatitis within 2 weeks, a feature prevented by AAV9-SMN gene therapy. Although fibrosis is not overtly apparent in histologic sections of the liver, there is molecular evidence of fibrogenesis and presence of stellate cell activation. The consequent liver damage arises from mitochondrial reactive oxygen species production and results in hepatic dysfunction in protein output, complement, coagulation, iron homeostasis, and insulin-like growth factor-1 metabolism. The NAFLD phenotype is likely due to non-esterified fatty acid overload from peripheral lipolysis subsequent to hyperglucagonemia compounded by reduced muscle use and insulin resistance. Despite the low hepatic mitochondrial content, isolated mitochondria show enhanced β-oxidation, likely as a compensatory response, resulting in the production of reactive oxygen species. In contrast to typical NAFLD/NASH, the Smn2B/- mice lose weight because of their associated neurological condition (spinal muscular atrophy) and develop hypoglycemia. CONCLUSIONS The Smn2B/- mice represent a good model of microvesicular steatohepatitis. Like other models, it is not representative of the complete NAFLD/NASH spectrum. Nevertheless, it offers a reliable, low-cost, early-onset model that is not dependent on diet to identify molecular players in NAFLD pathogenesis and can serve as one of the very few models of microvesicular steatohepatitis for both adult and pediatric populations.
Collapse
Affiliation(s)
- Marc-Olivier Deguise
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada,Centre for Neuromuscular Disease, University of Ottawa, Ottawa, Ontario, Canada
| | - Chantal Pileggi
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Yves De Repentigny
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Ariane Beauvais
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Alexandra Tierney
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Lucia Chehade
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada,Centre for Neuromuscular Disease, University of Ottawa, Ottawa, Ontario, Canada
| | - Jean Michaud
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Maica Llavero-Hurtado
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, United Kingdom,The Roslin Institute, Royal (Dick) School of Veterinary Studies, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Douglas Lamont
- FingerPrints Proteomics Facility, University of Dundee, Dundee, United Kingdom
| | - Abdelmadjid Atrih
- FingerPrints Proteomics Facility, University of Dundee, Dundee, United Kingdom
| | - Thomas M. Wishart
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, United Kingdom,The Roslin Institute, Royal (Dick) School of Veterinary Studies, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Thomas H. Gillingwater
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, United Kingdom,College of Medicine & Veterinary Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Bernard L. Schneider
- Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland,Bertarelli Foundation Gene Therapy Platform, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Geneva, Switzerland
| | - Mary-Ellen Harper
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Simon H. Parson
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, United Kingdom,Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Rashmi Kothary
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada,Centre for Neuromuscular Disease, University of Ottawa, Ottawa, Ontario, Canada,Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada,Correspondence Address correspondence to: Rashmi Kothary, PhD, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, Ontario, Canada K1H 8L6. fax: (613) 737-8803.
| |
Collapse
|
24
|
Stanley TL, Fourman LT, Zheng I, McClure CM, Feldpausch MN, Torriani M, Corey KE, Chung RT, Lee H, Kleiner DE, Hadigan CM, Grinspoon SK. Relationship of IGF-1 and IGF-Binding Proteins to Disease Severity and Glycemia in Nonalcoholic Fatty Liver Disease. J Clin Endocrinol Metab 2021; 106:e520-e533. [PMID: 33125080 PMCID: PMC7823253 DOI: 10.1210/clinem/dgaa792] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Indexed: 12/11/2022]
Abstract
CONTEXT Growth hormone (GH) and IGF-1 help regulate hepatic glucose and lipid metabolism, and reductions in these hormones may contribute to development of nonalcoholic fatty liver disease (NAFLD). OBJECTIVE To assess relationships between hepatic expression of IGF1 and IGF-binding proteins (IGFBPs) and measures of glycemia and liver disease in adults with NAFLD. Secondarily to assess effects of GH-releasing hormone (GHRH) on circulating IGFBPs. DESIGN Analysis of data from a randomized clinical trial of GHRH. SETTING Two US academic medical centers. PARTICIPANTS Participants were 61 men and women 18 to 70 years of age with HIV-infection, ≥5% hepatic fat fraction, including 39 with RNA-Seq data from liver biopsy. MAIN OUTCOME MEASURES Hepatic steatosis, inflammation, and fibrosis by histopathology and measures of glucose homeostasis. RESULTS Hepatic IGF1 mRNA was significantly lower in individuals with higher steatosis and NAFLD Activity Score (NAS) and was inversely related to glucose parameters, independent of circulating IGF-1. Among the IGFBPs, IGFBP2 and IGFBP4 were lower and IGFBP6 and IGFBP7 (also known as IGFBP-related protein 1) were higher with increasing steatosis. Hepatic IGFBP6 and IGFBP7 mRNA levels were positively associated with NAS. IGFBP7 mRNA increased with increasing fibrosis. Hepatic IGFBP1 mRNA was inversely associated with glycemia and insulin resistance, with opposite relationships present for IGFBP3 and IGFBP7. GHRH increased circulating IGFBP-1 and IGFBP-3, but decreased IGFBP-2 and IGFBP-6. CONCLUSIONS These data demonstrate novel relationships of IGF-1 and IGFBPs with NAFLD severity and glucose control, with divergent roles seen for different IGFBPs. Moreover, the data provide new information on the complex effects of GHRH on IGFBPs.
Collapse
Affiliation(s)
- Takara L Stanley
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Lindsay T Fourman
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Isabel Zheng
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Colin M McClure
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Meghan N Feldpausch
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Martin Torriani
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Kathleen E Corey
- Liver Center, Gastroenterology Division, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Raymond T Chung
- Liver Center, Gastroenterology Division, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Hang Lee
- Biostatistics Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - David E Kleiner
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Colleen M Hadigan
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Steven K Grinspoon
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Correspondence and Reprint Requests: Steven K. Grinspoon, MD, Professor of Medicine, Harvard Medical School, MGH Endowed Chair in Neuroendocrinology and Metabolism, Chief, Metabolism Unit, Massachusetts General Hospital, 55 Fruit Street 5LON207, Boston, MA 02114, United States. E-mail:
| |
Collapse
|
25
|
Geng T, Wang M, Li X, Zhou T, Ma H, Fonseca VA, Koh WP, Huang T, Heianza Y, Qi L. Birth weight modifies the relation between adulthood levels of insulin-like growth factor-1 and type 2 diabetes: a prospective cohort study. BMJ Open Diabetes Res Care 2021; 9:9/1/e001885. [PMID: 33648986 PMCID: PMC7925240 DOI: 10.1136/bmjdrc-2020-001885] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 11/18/2020] [Accepted: 11/25/2020] [Indexed: 11/03/2022] Open
Abstract
INTRODUCTION Insulin-like growth factor-1 (IGF-1) has been implicated in fetal and early-life growth and development of type 2 diabetes (T2D). We aimed to examine the interaction between circulating IGF-1 and birth weight in relation to risk of T2D. RESEARCH DESIGN AND METHODS We included 181 090 adults, aged 39-70 years in the UK Biobank Study, who were free of diabetes or major cardiovascular diseases at baseline. Serum IGF-1 levels were determined using chemiluminescent immunoassay method. Birth weight was self-reported; a Genetic Risk Score (GRS) was calculated to define the genetically determined birth weight. The outcome was the incidence of T2D. RESULTS We identified 3299 incident T2D cases over an average of 9.9 years of follow-up. Among the participants with birth weight of ≥2.5 kg, IGF-1 levels were inversely associated with T2D risk in a dose-dependent manner (p-trend<0.001). In contrast, the association was not significant among those with birth weight of <2.5 kg (p-interaction=0.001). The GRS of birth weight did not interact with IGF-1 levels on T2D risk. CONCLUSIONS Our results indicate that birth weight significantly modifies the relation between adulthood levels of circulating IGF-1 and the risk of T2D. Our findings highlight the importance of early-life risk factors in the development of the lifecourse prevention strategies targeting IGF-1 and T2D.
Collapse
Affiliation(s)
- Tingting Geng
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore
- Department of Epidemiology, School of Public Health and Tropical Medicine,Tulane University, New Orleans, Louisiana, USA
| | - Mengying Wang
- Department of Epidemiology, School of Public Health and Tropical Medicine,Tulane University, New Orleans, Louisiana, USA
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Xiang Li
- Department of Epidemiology, School of Public Health and Tropical Medicine,Tulane University, New Orleans, Louisiana, USA
| | - Tao Zhou
- Department of Epidemiology, School of Public Health and Tropical Medicine,Tulane University, New Orleans, Louisiana, USA
| | - Hao Ma
- Department of Epidemiology, School of Public Health and Tropical Medicine,Tulane University, New Orleans, Louisiana, USA
| | - Vivian A Fonseca
- Section of Endocrinology and Metabolism, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Woon-Puay Koh
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore
- Health Services and Systems Research, Duke-NUS Medical School, Singapore
| | - Tao Huang
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing, China
- Key Laboratory of Molecular Cardiovascular Sciences, Peking University, Beijing, China
- Center for Intelligent Public Health, Academy for Artificial Intelligence, Beijing, China
| | - Yoriko Heianza
- Department of Epidemiology, School of Public Health and Tropical Medicine,Tulane University, New Orleans, Louisiana, USA
| | - Lu Qi
- Department of Epidemiology, School of Public Health and Tropical Medicine,Tulane University, New Orleans, Louisiana, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Channing Division of Network Medicine,Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
26
|
Herrero-Labrador R, Trueba-Saiz A, Martinez-Rachadell L, Fernandez de Sevilla ME, Zegarra-Valdivia JA, Pignatelli J, Diaz-Pacheco S, Fernandez AM, Torres Aleman I. Circulating Insulin-Like Growth Factor I is Involved in the Effect of High Fat Diet on Peripheral Amyloid β Clearance. Int J Mol Sci 2020; 21:ijms21249675. [PMID: 33352990 PMCID: PMC7766006 DOI: 10.3390/ijms21249675] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 12/15/2020] [Accepted: 12/16/2020] [Indexed: 01/11/2023] Open
Abstract
Obesity is a risk factor for Alzheimer’s disease (AD), but underlying mechanisms are not clear. We analyzed peripheral clearance of amyloid β (Aβ) in overweight mice because its systemic elimination may impact brain Aβ load, a major landmark of AD pathology. We also analyzed whether circulating insulin-like growth factor I (IGF-I) intervenes in the effects of overweight as this growth factor modulates brain Aβ clearance and is increased in the serum of overweight mice. Overweight mice showed increased Aβ accumulation by the liver, the major site of elimination of systemic Aβ, but unaltered brain Aβ levels. We also found that Aβ accumulation by hepatocytes is stimulated by IGF-I, and that mice with low serum IGF-I levels show reduced liver Aβ accumulation—ameliorated by IGF-I administration, and unchanged brain Aβ levels. In the brain, IGF-I favored the association of its receptor (IGF-IR) with the Aβ precursor protein (APP), and at the same time, stimulated non-amyloidogenic processing of APP in astrocytes, as indicated by an increased sAPPα/sAPPβ ratio after IGF-I treatment. Since serum IGF-I enters into the brain in an activity-dependent manner, we analyzed in overweight mice the effect of brain activation by environmental enrichment (EE) on brain IGF-IR phosphorylation and its association to APP, as a readout of IGF-I activity. After EE, significantly reduced brain IGF-IR phosphorylation and APP/IGF-IR association were found in overweight mice as compared to lean controls. Collectively, these results indicate that a high-fat diet influences peripheral clearance of Aβ without affecting brain Aβ load. Increased serum IGF-I likely contributes to enhanced peripheral Aβ clearance in overweight mice, without affecting brain Aβ load probably because its brain entrance is reduced.
Collapse
Affiliation(s)
- Raquel Herrero-Labrador
- Cajal Institute, CSIC, 28002 Madrid, Spain; (R.H.-L.); (A.T.-S.); (L.M.-R.); (M.E.F.d.S.); (J.A.Z.-V.); (J.P.); (S.D.-P.); (A.M.F.)
- Ciberned, 28029 Madrid, Spain
| | - Angel Trueba-Saiz
- Cajal Institute, CSIC, 28002 Madrid, Spain; (R.H.-L.); (A.T.-S.); (L.M.-R.); (M.E.F.d.S.); (J.A.Z.-V.); (J.P.); (S.D.-P.); (A.M.F.)
- Ciberned, 28029 Madrid, Spain
| | - Laura Martinez-Rachadell
- Cajal Institute, CSIC, 28002 Madrid, Spain; (R.H.-L.); (A.T.-S.); (L.M.-R.); (M.E.F.d.S.); (J.A.Z.-V.); (J.P.); (S.D.-P.); (A.M.F.)
- Ciberned, 28029 Madrid, Spain
| | - Mᵃ Estrella Fernandez de Sevilla
- Cajal Institute, CSIC, 28002 Madrid, Spain; (R.H.-L.); (A.T.-S.); (L.M.-R.); (M.E.F.d.S.); (J.A.Z.-V.); (J.P.); (S.D.-P.); (A.M.F.)
- Ciberned, 28029 Madrid, Spain
| | - Jonathan A. Zegarra-Valdivia
- Cajal Institute, CSIC, 28002 Madrid, Spain; (R.H.-L.); (A.T.-S.); (L.M.-R.); (M.E.F.d.S.); (J.A.Z.-V.); (J.P.); (S.D.-P.); (A.M.F.)
- Ciberned, 28029 Madrid, Spain
- Universidad Nacional de San Agustín de Arequipa, 04001 Arequipa, Peru
| | - Jaime Pignatelli
- Cajal Institute, CSIC, 28002 Madrid, Spain; (R.H.-L.); (A.T.-S.); (L.M.-R.); (M.E.F.d.S.); (J.A.Z.-V.); (J.P.); (S.D.-P.); (A.M.F.)
- Ciberned, 28029 Madrid, Spain
| | - Sonia Diaz-Pacheco
- Cajal Institute, CSIC, 28002 Madrid, Spain; (R.H.-L.); (A.T.-S.); (L.M.-R.); (M.E.F.d.S.); (J.A.Z.-V.); (J.P.); (S.D.-P.); (A.M.F.)
| | - Ana M. Fernandez
- Cajal Institute, CSIC, 28002 Madrid, Spain; (R.H.-L.); (A.T.-S.); (L.M.-R.); (M.E.F.d.S.); (J.A.Z.-V.); (J.P.); (S.D.-P.); (A.M.F.)
- Ciberned, 28029 Madrid, Spain
| | - Ignacio Torres Aleman
- Cajal Institute, CSIC, 28002 Madrid, Spain; (R.H.-L.); (A.T.-S.); (L.M.-R.); (M.E.F.d.S.); (J.A.Z.-V.); (J.P.); (S.D.-P.); (A.M.F.)
- Ciberned, 28029 Madrid, Spain
- Correspondence:
| |
Collapse
|
27
|
Herring Milt and Herring Milt Protein Hydrolysate Are Equally Effective in Improving Insulin Sensitivity and Pancreatic Beta-Cell Function in Diet-Induced Obese- and Insulin-Resistant Mice. Mar Drugs 2020; 18:md18120635. [PMID: 33322303 PMCID: PMC7763884 DOI: 10.3390/md18120635] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 12/02/2020] [Accepted: 12/08/2020] [Indexed: 02/06/2023] Open
Abstract
Although genetic predisposition influences the onset and progression of insulin resistance and diabetes, dietary nutrients are critical. In general, protein is beneficial relative to carbohydrate and fat but dependent on protein source. Our recent study demonstrated that 70% replacement of dietary casein protein with the equivalent quantity of protein derived from herring milt protein hydrolysate (HMPH; herring milt with proteins being enzymatically hydrolyzed) significantly improved insulin resistance and glucose homeostasis in high-fat diet-induced obese mice. As production of protein hydrolysate increases the cost of the product, it is important to determine whether a simply dried and ground herring milt product possesses similar benefits. Therefore, the current study was conducted to investigate the effect of herring milt dry powder (HMDP) on glucose control and the associated metabolic phenotypes and further to compare its efficacy with HMPH. Male C57BL/6J mice on a high-fat diet for 7 weeks were randomized based on body weight and blood glucose into three groups. One group continued on the high-fat diet and was used as the insulin-resistant/diabetic control and the other two groups were given the high-fat diet modified to have 70% of casein protein being replaced with the same amount of protein from HMDP or HMPH. A group of mice on a low-fat diet all the time was used as the normal control. The results demonstrated that mice on the high-fat diet increased weight gain and showed higher blood concentrations of glucose, insulin, and leptin, as well as impaired glucose tolerance and pancreatic β-cell function relative to those on the normal control diet. In comparison with the high-fat diet, the replacement of 70% dietary casein protein with the same amount of HMDP or HMPH protein decreased weight gain and significantly improved the aforementioned biomarkers, insulin sensitivity or resistance, and β-cell function. The HMDP and HMPH showed similar effects on every parameter except blood lipids where HMDP decreased total cholesterol and non-HDL-cholesterol levels while the effect of HMPH was not significant. The results demonstrate that substituting 70% of dietary casein protein with the equivalent amount of HMDP or HMPH protein protects against obesity and diabetes, and HMDP is also beneficial to cholesterol homeostasis.
Collapse
|
28
|
Gandoy-Fieiras N, Gonzalez-Juanatey JR, Eiras S. Myocardium Metabolism in Physiological and Pathophysiological States: Implications of Epicardial Adipose Tissue and Potential Therapeutic Targets. Int J Mol Sci 2020; 21:2641. [PMID: 32290181 PMCID: PMC7177518 DOI: 10.3390/ijms21072641] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 04/05/2020] [Accepted: 04/08/2020] [Indexed: 01/01/2023] Open
Abstract
The main energy substrate of adult cardiomyocytes for their contractility are the fatty acids. Its metabolism generates high ATP levels at the expense of high oxygen consumption in the mitochondria. Under low oxygen supply, they can get energy from other substrates, mainly glucose, lactate, ketone bodies, etc., but the mitochondrial dysfunction, in pathological conditions, reduces the oxidative metabolism. In consequence, fatty acids are stored into epicardial fat and its accumulation provokes inflammation, insulin resistance, and oxidative stress, which enhance the myocardium dysfunction. Some therapies focused on improvement the fatty acids entry into mitochondria have failed to demonstrate benefits on cardiovascular disorders. Oppositely, those therapies with effects on epicardial fat volume and inflammation might improve the oxidative metabolism of myocardium and might reduce the cardiovascular disease progression. This review aims at explain (a) the energy substrate adaptation of myocardium in physiological conditions, (b) the reduction of oxidative metabolism in pathological conditions and consequences on epicardial fat accumulation and insulin resistance, and (c) the reduction of cardiovascular outcomes after regulation by some therapies.
Collapse
Affiliation(s)
- Nerea Gandoy-Fieiras
- Translational Cardiology Group, Health Research Institute, 15782 Santiago de Compostela, Spain;
| | - Jose Ramon Gonzalez-Juanatey
- Cardiovascular Department, University Hospital of Santiago de Compostela, 15782 Santiago de Compostela, Spain;
- Cardiology Group, Health Research Institute, 17582 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Sonia Eiras
- Translational Cardiology Group, Health Research Institute, 15782 Santiago de Compostela, Spain;
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| |
Collapse
|
29
|
Arima N, Sasaki Y, Lee LH, Zhang H, Figueiredo JL, Mlynarchik AK, Qiao J, Yamada I, Higashi H, Ha AH, Halu A, Mizuno K, Singh SA, Yamazaki Y, Aikawa M. Multiorgan Systems Study Reveals Igfbp7 as a Suppressor of Gluconeogenesis after Gastric Bypass Surgery. J Proteome Res 2020; 19:129-143. [PMID: 31661273 DOI: 10.1021/acs.jproteome.9b00441] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Roux-en-Y gastric bypass (RYGB) surgery reduces weight in obese patients. A marked decrease in blood glucose levels occurs before weight loss; however, key molecules that improve the glycemic profile remain largely unknown. Using a murine RYGB surgery model, we performed multiorgan proteomics and bioinformatics to monitor the proteins and molecular pathways that change in this early glycemic response. Multiplexed proteomic kinetics data analysis revealed that the Roux limb, biliopancreatic limb, liver, and pancreas each exhibited unique temporal and molecular responses to the RYGB surgery. In addition, protein-protein network analysis indicated that the changes to the microbial environment in the intestine may play a crucial role in the beneficial effects of RYGB surgery. Furthermore, insulin-like growth factor binding protein 7 (Igfbp7) was identified as an early induced protein in the Roux limb. Known secretory properties of Igfbp7 prompted us to further investigate its role as a remote organ regulator of glucose metabolism. Igfbp7 overexpression decreased blood glucose levels in diet-induced obese mice and attenuated gluconeogenic gene expression in the liver. Secreted Igfbp7 appeared to mediate these beneficial effects. These results demonstrate that organs responded differentially to RYGB surgery and indicate that Igfbp7 may play an important role in improving blood glucose levels.
Collapse
Affiliation(s)
- Naoaki Arima
- Tokyo New Drug Research Laboratories , Kowa Company, Ltd. , Tokyo 189-0022 , Japan
| | - Yusuke Sasaki
- Tokyo New Drug Research Laboratories , Kowa Company, Ltd. , Tokyo 189-0022 , Japan
| | | | | | | | | | | | - Iwao Yamada
- Tokyo New Drug Research Laboratories , Kowa Company, Ltd. , Tokyo 189-0022 , Japan
| | - Hideyuki Higashi
- Tokyo New Drug Research Laboratories , Kowa Company, Ltd. , Tokyo 189-0022 , Japan
| | | | | | - Ken Mizuno
- Tokyo New Drug Research Laboratories , Kowa Company, Ltd. , Tokyo 189-0022 , Japan
| | | | - Yukiyoshi Yamazaki
- Tokyo New Drug Research Laboratories , Kowa Company, Ltd. , Tokyo 189-0022 , Japan
| | - Masanori Aikawa
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health , Moscow , Russian Federation , 119146
| |
Collapse
|
30
|
Garmes HM, Castillo AR. Insulin signaling in the whole spectrum of GH deficiency. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2019; 63:582-591. [PMID: 31939483 PMCID: PMC10522230 DOI: 10.20945/2359-3997000000188] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 10/12/2019] [Indexed: 11/23/2022]
Abstract
GH is one of the insulin counterregulatory hormones which acts in the opposite way to insulin, increasing the glucose production by the liver and kidneys and decreasing glucose uptake from peripheral tissues, thus being a hyperglycemic hormone. When in excess, as in acromegaly, it induces glucose intolerance and diabetes. As expected, patients with GH deficiency (GHD) have hypoglycemia, especially in early childhood, but as GH is also a lipolytic hormone, these patients are becoming obese with higher percentages of body fat. Although obesity in general is directly related to insulin resistance, in patients with GH secretion disorders this relationship may be altered. In acromegaly there is a decrease in fat mass with worsening insulin sensitivity and mice with isolated GHD are characterized by greater insulin sensitivity despite excess fat mass. In humans with GHD, body composition shows increased body fat and decreased free fat mass, but the results regarding insulin sensitivity are still controversial in these patients. These discrepant results regarding insulin sensitivity in patients with GHD suggest the existence of other variables influencing these results. In the present review, we will try to follow the path of the different researches conducted on this subject, both in animal and human models, with the goal of understanding the current knowledge of insulin sensitivity across the spectrum of GHD. Arch Endocrinol Metab. 2019;63(6):582-91.
Collapse
Affiliation(s)
- Heraldo Mendes Garmes
- Departamento de Clínica MédicaFaculdade de Ciências MédicasUniversidade Estadual de CampinasCampinasSPBrasil Divisão de Endocrinologia, Departamento de Clínica Médica, Faculdade de Ciências Médicas da Universidade Estadual de Campinas (FCM-Unicamp), Campinas, SP, Brasil
| | - Alejandro Rosell Castillo
- Departamento de Clínica MédicaFaculdade de Ciências MédicasUniversidade Estadual de CampinasCampinasSPBrasil Divisão de Endocrinologia, Departamento de Clínica Médica, Faculdade de Ciências Médicas da Universidade Estadual de Campinas (FCM-Unicamp), Campinas, SP, Brasil
| |
Collapse
|
31
|
Chhabra Y, Nelson CN, Plescher M, Barclay JL, Smith AG, Andrikopoulos S, Mangiafico S, Waxman DJ, Brooks AJ, Waters MJ. Loss of growth hormone-mediated signal transducer and activator of transcription 5 (STAT5) signaling in mice results in insulin sensitivity with obesity. FASEB J 2019; 33:6412-6430. [PMID: 30779881 PMCID: PMC6463913 DOI: 10.1096/fj.201802328r] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Growth hormone (GH) has an important function as an insulin antagonist with elevated insulin sensitivity evident in humans and mice lacking a functional GH receptor (GHR). We sought the molecular basis for this sensitivity by utilizing a panel of mice possessing specific deletions of GHR signaling pathways. Metabolic clamps and glucose homeostasis tests were undertaken in these obese adult C57BL/6 male mice, which indicated impaired hepatic gluconeogenesis. Insulin sensitivity and glucose disappearance rate were enhanced in muscle and adipose of mice lacking the ability to activate the signal transducer and activator of transcription (STAT)5 via the GHR (Ghr-391-/-) as for GHR-null (GHR-/-) mice. These changes were associated with a striking inhibition of hepatic glucose output associated with altered glycogen metabolism and elevated hepatic glycogen content during unfed state. The enhanced hepatic insulin sensitivity was associated with increased insulin receptor β and insulin receptor substrate 1 activation along with activated downstream protein kinase B signaling cascades. Although phosphoenolpyruvate carboxykinase (Pck)-1 expression was unchanged, its inhibitory acetylation was elevated because of decreased sirtuin-2 expression, thereby promoting loss of PCK1. Loss of STAT5 signaling to defined chromatin immunoprecipitation targets would further increase lipogenesis, supporting hepatosteatosis while lowering glucose output. Finally, up-regulation of IL-15 expression in muscle, with increased secretion of adiponectin and fibroblast growth factor 1 from adipose tissue, is expected to promote insulin sensitivity.-Chhabra, Y., Nelson, C. N., Plescher, M., Barclay, J. L., Smith, A. G., Andrikopoulos, S., Mangiafico, S., Waxman, D. J., Brooks, A. J., Waters, M. J. Loss of growth hormone-mediated signal transducer and activator of transcription 5 (STAT5) signaling in mice results in insulin sensitivity with obesity.
Collapse
Affiliation(s)
- Yash Chhabra
- University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia.,Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Caroline N Nelson
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Monika Plescher
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia.,Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Johanna L Barclay
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia.,Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Aaron G Smith
- School of Biomedical Sciences, Institute of Health and Biomedical Innovation, The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Sof Andrikopoulos
- Department of Medicine, The University of Melbourne, Victoria, Australia
| | | | - David J Waxman
- Department of Biology and Bioinformatics Program, Boston University, Boston, Massachusetts, USA
| | - Andrew J Brooks
- University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia.,Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Michael J Waters
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
32
|
Goldstein JM, Hale T, Foster SL, Tobet SA, Handa RJ. Sex differences in major depression and comorbidity of cardiometabolic disorders: impact of prenatal stress and immune exposures. Neuropsychopharmacology 2019; 44:59-70. [PMID: 30030541 PMCID: PMC6235859 DOI: 10.1038/s41386-018-0146-1] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 06/15/2018] [Accepted: 06/21/2018] [Indexed: 12/11/2022]
Abstract
Major depressive disorder topped ischemic heart disease as the number one cause of disability worldwide in 2012, and women have twice the risk of men. Further, the comorbidity of depression and cardiometabolic disorders will be one of the primary causes of disability worldwide by 2020, with women at twice the risk. Thus, understanding the sex-dependent comorbidities has public health consequences worldwide. We propose here that sex differences in MDD-cardiometabolic comorbidity originate, in part, from pathogenic processes initiated in fetal development that involve sex differences in shared pathophysiology between the brain, the vascular system, the CNS control of the heart and associated hormonal, immune, and metabolic physiology. Pathways implicate neurotrophic and angiogenic growth factors, gonadal hormone receptors, and neurotransmitters such as gamma amino butyric acid (GABA) on neuronal and vascular development of HPA axis regions, such as the paraventricular nucleus (PVN), in addition to blood pressure, in part through the renin-angiotensin system, and insulin and glucose metabolism. We show that the same prenatal exposures have consequences for sex differences across multiple organ systems that, in part, share common pathophysiology. Thus, we believe that applying a sex differences lens to understanding shared biologic substrates underlying these comorbidities will provide novel insights into the development of sex-dependent therapeutics. Further, taking a lifespan perspective beginning in fetal development provides the opportunity to target abnormalities early in the natural history of these disorders in a sex-dependent way.
Collapse
Affiliation(s)
- Jill M Goldstein
- Departments of Psychiatry and Obstetrics and Gynecology, Massachusetts General Hospital (MGH), Boston, MA, 02120, USA.
- Departments of Psychiatry and Medicine, Harvard Medical School, Boston, MA, USA.
| | - Taben Hale
- Department of Basic Medical Science, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, 85004, USA
| | - Simmie L Foster
- Department of Psychiatry, Harvard Medical School, at Massachusetts General Hospital, Boston, MA, USA
| | - Stuart A Tobet
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO, 80523, USA
| | - Robert J Handa
- Department of Basic Medical Science, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, 85004, USA
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA
| |
Collapse
|
33
|
Emamat H, Tangestani H, Bahadoran Z, Khalili-Moghadam S, Mirmiran P. The Associations of Dietary Acid Load with Insulin Resistance and Type 2 Diabetes: A Systematic Review of Existing Human Studies. Recent Pat Food Nutr Agric 2018; 10:27-33. [PMID: 30246649 DOI: 10.2174/2212798410666180924142222] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 08/15/2018] [Accepted: 09/15/2018] [Indexed: 11/22/2022]
Abstract
BACKGROUND Type 2 diabetes is a multifactorial disorder influenced by both genetic and environmental factors and is rising dramatically throughout the world. Recently, consideration to dietary acid-base load has been raised as a nutritional indicator that could have metabolic effects. OBJECTIVE The aim of this study was to systematically investigate the associations of dietary acid load indices with glucose/insulin homeostasis and type 2 diabetes in a patent based review. METHODS Systematic literature review was conducted using PubMed, Scopus and Google Scholar, from inception up to 10 January 2018. All human studies publications investigated the association of dietary acid load indices (PRAL or NEAP or Pro:K) and incidence of diabetes or glucose/insulin metabolism were included. RESULTS In all, 174 studies were included for the title and abstract screening. A total of 164 articles were excluded because they did not meet the inclusion criteria. Finally, 10 articles (five crosssectional studies and five cohort studies) in accordance with our inclusion criteria were chosen for further evaluations that were published between 2008 and 2017. CONCLUSION Although there are some evidence of an association between dietary acid load and type 2 diabetes, definitive declarations in this regard will be needed to intervention human studies modifying acid-base dietary intake.
Collapse
Affiliation(s)
- Hadi Emamat
- Student Research Committee, Department and Faculty of Nutrition Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hadith Tangestani
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Zahra Bahadoran
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sajjad Khalili-Moghadam
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parvin Mirmiran
- Department of Nutrition and Clinical Dietetics, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
34
|
Kineman RD, del Rio-Moreno M, Sarmento-Cabral A. 40 YEARS of IGF1: Understanding the tissue-specific roles of IGF1/IGF1R in regulating metabolism using the Cre/loxP system. J Mol Endocrinol 2018; 61:T187-T198. [PMID: 29743295 PMCID: PMC7721256 DOI: 10.1530/jme-18-0076] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 05/09/2018] [Indexed: 12/13/2022]
Abstract
It is clear that insulin-like growth factor-1 (IGF1) is important in supporting growth and regulating metabolism. The IGF1 found in the circulation is primarily produced by the liver hepatocytes, but healthy mature hepatocytes do not express appreciable levels of the IGF1 receptor (IGF1R). Therefore, the metabolic actions of IGF1 are thought to be mediated via extra-hepatocyte actions. Given the structural and functional homology between IGF1/IGF1R and insulin receptor (INSR) signaling, and the fact that IGF1, IGF1R and INSR are expressed in most tissues of the body, it is difficult to separate out the tissue-specific contributions of IGF1/IGF1R in maintaining whole body metabolic function. To circumvent this problem, over the last 20 years, investigators have taken advantage of the Cre/loxP system to manipulate IGF1/IGF1R in a tissue-dependent, and more recently, an age-dependent fashion. These studies have revealed that IGF1/IGF1R can alter extra-hepatocyte function to regulate hormonal inputs to the liver and/or alter tissue-specific carbohydrate and lipid metabolism to alter nutrient flux to liver, where these actions are not mutually exclusive, but serve to integrate the function of all tissues to support the metabolic needs of the organism.
Collapse
Affiliation(s)
- Rhonda D Kineman
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Illinois at Chicago,1819 W Polk St. M/C 646 Chicago, IL, 60612
- Research and Development Division, Jesse Brown VA Medical Center, Suite 6215, MP 191, 820 S Damen Ave. Chicago, IL 60612
- Corresponding author: Rhonda D Kineman, . University of Illinois at Chicago, Medicine, 1819 W. Polk St., MC 640, Chicago, IL, USA 60612
| | - Mercedes del Rio-Moreno
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Illinois at Chicago,1819 W Polk St. M/C 646 Chicago, IL, 60612
| | - André Sarmento-Cabral
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Illinois at Chicago,1819 W Polk St. M/C 646 Chicago, IL, 60612
| |
Collapse
|
35
|
Li J, Mao Z, Huang J, Xia J. PICK1 is essential for insulin production and the maintenance of glucose homeostasis. Mol Biol Cell 2018; 29:587-596. [PMID: 29298842 PMCID: PMC6004578 DOI: 10.1091/mbc.e17-03-0204] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 12/22/2017] [Accepted: 12/27/2017] [Indexed: 01/11/2023] Open
Abstract
Protein interacting with C-kinase 1 (PICK1) is a peripheral membrane protein that controls insulin granule formation, trafficking, and maturation in INS-1E cells. However, global Pick1-knockout mice showed only a subtle diabetes-like phenotype. This raises the possibility that compensatory effects from tissues other than pancreatic beta cells may obscure the effects of insulin deficiency. To explore the role of PICK1 in pancreatic islets, we generated mice harboring a conditional Pick1 allele in a C57BL/6J background. The conditional Pick1-knockout mice exhibited impaired glucose tolerance, profound insulin deficiency, and hyperglycemia. In vitro experiments showed that the ablation of Pick1 in pancreatic beta cells selectively decreased the initial rapid release of insulin and the total insulin levels in the islets. Importantly, the specific ablation of Pick1 induced elevated proinsulin levels in the circulation and in the islets, accompanied by a reduction in the proinsulin processing enzymes prohormone convertase 1/3 (PC1/3). The deletion of Pick1 triggered the specific elimination of chromogranin B in pancreatic beta cells, which is believed to control granule formation and release. Collectively, these data demonstrate the critical role of PICK1 in secretory granule biogenesis, proinsulin processing, and beta cell function. We conclude that the beta cell-specific deletion of Pick1 in mice led to hyperglycemia and eventually to diabetes.
Collapse
Affiliation(s)
- Jia Li
- Department of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, China
- College of Life Science, Shaanxi Normal University, Shaanxi 710119, China
| | - Zhuo Mao
- Department of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, China
- Center for Diabetes, Obesity and Metabolism, Department of Physiology, Shenzhen University Health Science Center, Shenzhen, Guangdong 518061, China
| | - Jiandong Huang
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- HKU-Shenzhen Institute of Research and Innovation, Shenzhen 518057, China
- Shenzhen Institute of Advanced Technologies, Shenzhen 518055, China
| | - Jun Xia
- Department of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, China
| |
Collapse
|
36
|
Abstract
Anabolic-androgenic steroids (AAS) and other hormones such as growth hormone (GH) and insulin-like growth factor-1 (IGF-1) have been shown to increase muscle mass in patients suffering from various diseases related to muscle atrophy. Despite known side-effects associated with supraphysiologic doses of such drugs, their anabolic effects have led to their widespread use and abuse by bodybuilders and athletes such as strength athletes seeking to improve performance and muscle mass. On the other hand, resistance training (RT) has also been shown to induce significant endogenous hormonal (testosterone (T), GH, IGF-1) elevations. Therefore, some bodybuilders employ RT protocols designed to elevate hormonal levels in order to maximize anabolic responses. In this article, we reviewed current RT protocol outcomes with and without performance enhancing drug usage. Acute RT-induced hormonal elevations seem not to be directly correlated with muscle growth. On the other hand, supplementation with AAS and other hormones might lead to supraphysiological muscle hypertrophy, especially when different compounds are combined.
Collapse
Affiliation(s)
- Julius Fink
- a Graduate School of Medicine, Department of Metabolism and Endocrinology , Juntendo University , Tokyo , Japan
| | | | - Koichi Nakazato
- c Graduate Schools of Health and Sport Science , Nippon Sport Science University , Tokyo , Japan
| |
Collapse
|
37
|
Unterman TG. Regulation of Hepatic Glucose Metabolism by FoxO Proteins, an Integrated Approach. Curr Top Dev Biol 2018; 127:119-147. [DOI: 10.1016/bs.ctdb.2017.10.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
38
|
Central IGF1 improves glucose tolerance and insulin sensitivity in mice. Nutr Diabetes 2017; 7:2. [PMID: 29259155 PMCID: PMC5865549 DOI: 10.1038/s41387-017-0002-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 07/26/2017] [Accepted: 08/01/2017] [Indexed: 02/05/2023] Open
Abstract
Insulin-like growth factor 1 (IGF1) is a key factor for tissue growth and fuel metabolism. The potential function of central IGF1 remains unclear. We previously observed that IGF1 expression is increased in the hypothalamus of obese mice lacking STAT5 in the central nervous system (CNS). In this study, we explored the potential metabolic function of central IGF1 by intracerebroventricular (ICV) injection of IGF1, over-expression of central IGF1 by administering an adeno-associated virus (AAV), and ICV injection of an anti-IGF1 antibody. Mice that over-expressed central IGF1 displayed increased appetite, improved glucose tolerance and insulin sensitivity, decreased Pomc levels in the hypothalamus, and increased UCP1 expression in brown fat tissue. This is the first study demonstrating that central IGF1 regulates several important metabolic functions.
Collapse
|
39
|
Bertucci JI, Blanco AM, Canosa LF, Unniappan S. Direct actions of macronutrient components on goldfish hepatopancreas in vitro to modulate the expression of ghr-I, ghr-II, igf-I and igf-II mRNAs. Gen Comp Endocrinol 2017; 250:1-8. [PMID: 28549738 DOI: 10.1016/j.ygcen.2017.05.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 05/02/2017] [Accepted: 05/22/2017] [Indexed: 12/18/2022]
Abstract
In mammals and fish, somatic growth and metabolism are coordinated by the GH-IGF axis, composed of growth hormone (GH), growth hormone receptors I and II (GHR-I and GHR-II), and the insulin-like growth factors I and II (IGF-I and IGF-II). In order to determine if dietary macronutrients regulate the hepatopancreatic expression of ghr-I, ghr-II, igf-I and igf-II independently of circulating GH, organ culture experiments were conducted. Briefly, goldfish hepatopancreas sections were incubated with different doses of glucose; L-tryptophan; oleic acid; linolenic acid (LNA); eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA). After two and four hours of treatment, the expression of ghr-I, ghr-II, igf-I and igf-II mRNAs was quantified. We found that glucose and L-tryptophan globally upregulate the mRNA expression of ghr-I; ghr-II; igf-I and igf-II. Duration of exposure, and unsaturation level of fatty acids differentially modulate ghr-I, ghr-II and igf-II mRNA expression. Additionally, we found that fatty acids increase the expression of igf-I depending on incubation time and fatty acid class. In conclusion, here we present evidence for GH-independent, direct effects exerted by dietary macronutrients on GHR and IGF in goldfish hepatopancreas.
Collapse
Affiliation(s)
- Juan Ignacio Bertucci
- Instituto de Investigaciones Biotecnológicas-Instituto Tecnológico Chascomús, Buenos Aires, Argentina
| | - Ayelén Melisa Blanco
- Departamento de Fisiología (Fisiología Animal II), Facultad de Biología, Universidad Complutense de Madrid, Madrid, Spain
| | - Luis Fabián Canosa
- Instituto de Investigaciones Biotecnológicas-Instituto Tecnológico Chascomús, Buenos Aires, Argentina.
| | - Suraj Unniappan
- Laboratory of Integrative Neuroendocrinology, Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada.
| |
Collapse
|
40
|
Insulin-like growth factor-1 signaling in cardiac aging. Biochim Biophys Acta Mol Basis Dis 2017; 1864:1931-1938. [PMID: 28847512 DOI: 10.1016/j.bbadis.2017.08.029] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 08/22/2017] [Accepted: 08/23/2017] [Indexed: 12/31/2022]
Abstract
Cardiovascular disease (CVD) is the leading cause of death in most developed countries. Aging is associated with enhanced risk of CVD. Insulin-like growth factor-1 (IGF-1) binds to its cognate receptor, IGF-1 receptor (IGF-1R), and exerts pleiotropic effects on cell growth, differentiation, development, and tissue repair. Importantly, IGF-1/IGF-1R signaling is implicated in cardiac aging and longevity. Cardiac aging is an intrinsic process that results in cardiac dysfunction, accompanied by molecular and cellular changes. In this review, we summarize the current state of knowledge regarding the link between the IGF-1/IGF-1R system and cardiac aging. The biological effects of IGF-1R and insulin receptor will be discussed and compared. Furthermore, we describe data regarding how deletion of IGF-1R in cardiomyocytes of aged knockout mice may delay the development of senescence-associated myocardial pathologies. This article is part of a Special issue entitled Cardiac adaptations to obesity, diabetes and insulin resistance, edited by Professors Jan F.C. Glatz, Jason R.B. Dyck and Christine Des Rosiers.
Collapse
|
41
|
Partial IGF-1 deficiency is sufficient to reduce heart contractibility, angiotensin II sensibility, and alter gene expression of structural and functional cardiac proteins. PLoS One 2017; 12:e0181760. [PMID: 28806738 PMCID: PMC5555709 DOI: 10.1371/journal.pone.0181760] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 07/06/2017] [Indexed: 01/15/2023] Open
Abstract
Circulating levels of IGF-1 may decrease under several circumstances like ageing, metabolic syndrome, and advanced cirrhosis. This reduction is associated with insulin resistance, dyslipidemia, progression to type 2 diabetes, and increased risk for cardiovascular diseases. However, underlying mechanisms between IGF-1 deficiency and cardiovascular disease remain elusive. The specific aim of the present work was to study whether the partial IGF-1 deficiency influences heart and/or coronary circulation, comparing vasoactive factors before and after of ischemia-reperfusion (I/R). In addition, histology of the heart was performed together with cardiac gene expression for proteins involved in structure and function (extracellular matrix, contractile proteins, active peptides); carried out using microarrays, followed by RT-qPCR confirmation of the three experimental groups. IGF-1 partial deficiency is associated to a reduction in contractility and angiotensin II sensitivity, interstitial fibrosis as well as altered expression pattern of genes involved in extracellular matrix proteins, calcium dynamics, and cardiac structure and function. Although this work is descriptive, it provides a clear insight of the impact that partial IGF-1 deficiency on the heart and establishes this experimental model as suitable for studying cardiac disease mechanisms and exploring therapeutic options for patients under IGF-1 deficiency conditions.
Collapse
|
42
|
Birzniece V, Barrett PHR, Ho KKY. Tamoxifen reduces hepatic VLDL production and GH secretion in women: a possible mechanism for steatosis development. Eur J Endocrinol 2017; 177:137-143. [PMID: 28500244 DOI: 10.1530/eje-17-0151] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 04/18/2017] [Accepted: 05/12/2017] [Indexed: 11/08/2022]
Abstract
CONTEXT Growth hormone (GH) stimulates hepatic synthesis of very-low-density lipoproteins (VLDL), whereas hepatic steatosis develops as a result of GH deficiency. Steatosis is also a complication of tamoxifen treatment, the cause of which is not known. As tamoxifen inhibits the secretion and action of GH, we hypothesize that it induces steatosis by inhibiting hepatic VLDL export. AIM To investigate whether tamoxifen reduces hepatic VLDL secretion. DESIGN Eight healthy, normolipidemic women (age: 64.4 ± 2.1 years) were studied in random sequence at baseline, after 2 weeks of tamoxifen (20 mg/day) and after 2 weeks of estradiol valerate (EV; 2 mg/day) treatments, separated by a 4-week washout period. The kinetics of apolipoprotein B (apoB), the structural protein of VLDL particles, were measured using a stable isotope 2H3-leucine turnover technique. VLDL-apoB fractional catabolic rate (FCR) was determined using a multicompartment model. VLDL-apoB secretion was estimated as the product of FCR and VLDL-apoB concentration. GH response to arginine stimulation, circulating levels of IGF-1, FFA, and TG, along with TG content in VLDL were measured. RESULTS Tamoxifen significantly (P < 0.05) reduced VLDL-apoB concentration and secretion by 27.3 ± 7.8% and 29.8 ± 10.2%, respectively. In contrast, EV did not significantly change VLDL-apoB concentration or secretion. Tamoxifen but not EV significantly reduced (P < 0.05) GH response to arginine stimulation. Both treatments significantly lowered (P < 0.05) circulating IGF-1. CONCLUSION Inhibition of VLDL secretion may contribute to the development of fatty liver during tamoxifen therapy. As GH stimulates VLDL secretion, the development of steatosis may arise secondarily from GH insufficiency induced by tamoxifen.
Collapse
Affiliation(s)
- Vita Birzniece
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- School of Medicine, Western Sydney University, Penrith, New South Wales, Australia
- School of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - P Hugh R Barrett
- School of Biomedical Sciences, Faculty of Health and Medical Sciences, and Faculty of Engineering and Mathematical Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Ken K Y Ho
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- Centres of Health Research, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| |
Collapse
|
43
|
Berryman DE, List EO. Growth Hormone's Effect on Adipose Tissue: Quality versus Quantity. Int J Mol Sci 2017; 18:ijms18081621. [PMID: 28933734 PMCID: PMC5578013 DOI: 10.3390/ijms18081621] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 07/10/2017] [Accepted: 07/17/2017] [Indexed: 02/07/2023] Open
Abstract
Obesity is an excessive accumulation or expansion of adipose tissue (AT) due to an increase in either the size and/or number of its characteristic cell type, the adipocyte. As one of the most significant public health problems of our time, obesity and its associated metabolic complications have demanded that attention be given to finding effective therapeutic options aimed at reducing adiposity or the metabolic dysfunction associated with its accumulation. Growth hormone (GH) has therapeutic potential due to its potent lipolytic effect and resultant ability to reduce AT mass while preserving lean body mass. However, AT and its resident adipocytes are significantly more dynamic and elaborate than once thought and require one not to use the reduction in absolute mass as a readout of efficacy alone. Paradoxically, therapies that reduce GH action may ultimately prove to be healthier, in part because GH also possesses potent anti-insulin activities along with concerns that GH may promote the growth of certain cancers. This review will briefly summarize some of the newer complexities of AT relevant to GH action and describe the current understanding of how GH influences this tissue using data from both humans and mice. We will conclude by considering the therapeutic use of GH or GH antagonists in obesity, as well as important gaps in knowledge regarding GH and AT.
Collapse
Affiliation(s)
- Darlene E Berryman
- The Diabetes Institute at Ohio University, 108 Konneker Research Labs, Ohio University, Athens, OH 45701, USA.
- Edison Biotechnology Institute, 218 Konneker Research Labs, Ohio University, Athens, OH 45701, USA.
| | - Edward O List
- The Diabetes Institute at Ohio University, 108 Konneker Research Labs, Ohio University, Athens, OH 45701, USA.
- Edison Biotechnology Institute, 218 Konneker Research Labs, Ohio University, Athens, OH 45701, USA.
| |
Collapse
|
44
|
Mukhi D, Nishad R, Menon RK, Pasupulati AK. Novel Actions of Growth Hormone in Podocytes: Implications for Diabetic Nephropathy. Front Med (Lausanne) 2017; 4:102. [PMID: 28748185 PMCID: PMC5506074 DOI: 10.3389/fmed.2017.00102] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Accepted: 06/26/2017] [Indexed: 02/05/2023] Open
Abstract
The kidney regulates water, electrolyte, and acid-base balance and thus maintains body homeostasis. The kidney's potential to ensure ultrafiltered and almost protein-free urine is compromised in various metabolic and hormonal disorders such as diabetes mellitus (DM). Diabetic nephropathy (DN) accounts for ~20-40% of mortality in DM. Proteinuria, a hallmark of renal glomerular diseases, indicates injury to the glomerular filtration barrier (GFB). The GFB is composed of glomerular endothelium, basement membrane, and podocytes. Podocytes are terminally differentiated epithelial cells with limited ability to replicate. Podocyte shape and number are both critical for the integrity and function of the GFB. Podocytes are vulnerable to various noxious stimuli prevalent in a diabetic milieu that could provoke podocytes to undergo changes to their unique architecture and function. Effacement of podocyte foot process is a typical morphological alteration associated with proteinuria. The dedifferentiation of podocytes from epithelial-to-mesenchymal phenotype and consequential loss results in proteinuria. Poorly controlled type 1 DM is associated with elevated levels of circulating growth hormone (GH), which is implicated in the pathophysiology of various diabetic complications including DN. Recent studies demonstrate that functional GH receptors are expressed in podocytes and that GH may exert detrimental effects on the podocyte. In this review, we summarize recent advances that shed light on actions of GH on the podocyte that could play a role in the pathogenesis of DN.
Collapse
Affiliation(s)
- Dhanunjay Mukhi
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Rajkishor Nishad
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Ram K. Menon
- Department of Pediatric Endocrinology and Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Anil Kumar Pasupulati
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, India
| |
Collapse
|
45
|
Abstract
PURPOSE OF REVIEW Acid-base homeostasis is impaired in chronic kidney disease (CKD) and may contribute to disease progression. Diabetes, a major cause of CKD worldwide, may exacerbate acidosis further due to differences in acid production and excretion. Here, we review the role of abnormal acid-base homeostasis in the pathogenesis and progression of diabetes and diabetic kidney disease. RECENT FINDINGS Acidosis and dietary acid loading may contribute to the development and worsening of insulin resistance and hypertension, thereby promoting diabetes and diabetic CKD. However, although metabolic acidosis associates with progression of CKD generally, the results in diabetic CKD are mixed. Data suggests that metabolic acid production in diabetes may be higher than would be predicted based on dietary intake alone, and new observational data suggests that this higher diet-independent acid production could potentially be protective. The role of acid-base homeostasis in diabetic CKD progression is complex and must consider differences in endogenous acid production and excretion in diabetes. Ongoing observational and interventional studies in this field should consider the unique physiology of diabetes.
Collapse
Affiliation(s)
- Pascale Khairallah
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Julia J Scialla
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC, USA.
- Department of Medicine, Durham Veterans Affairs Medical Center, Durham, NC, USA.
| |
Collapse
|
46
|
Liu Z, Cordoba-Chacon J, Kineman RD, Cronstein BN, Muzumdar R, Gong Z, Werner H, Yakar S. Growth Hormone Control of Hepatic Lipid Metabolism. Diabetes 2016; 65:3598-3609. [PMID: 27679560 PMCID: PMC5127251 DOI: 10.2337/db16-0649] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 09/20/2016] [Indexed: 12/15/2022]
Abstract
In humans, low levels of growth hormone (GH) and its mediator, IGF-1, associate with hepatic lipid accumulation. In mice, congenital liver-specific ablation of the GH receptor (GHR) results in reductions in circulating IGF-1 and hepatic steatosis, associated with systemic insulin resistance. Due to the intricate relationship between GH and IGF-1, the relative contribution of each hormone to the development of hepatic steatosis is unclear. Our goal was to dissect the mechanisms by which hepatic GH resistance leads to steatosis and overall insulin resistance, independent of IGF-1. We have generated a combined mouse model with liver-specific ablation of GHR in which we restored liver IGF-1 expression via the hepatic IGF-1 transgene. We found that liver GHR ablation leads to increases in lipid uptake, de novo lipogenesis, hyperinsulinemia, and hyperglycemia accompanied with severe insulin resistance and increased body adiposity and serum lipids. Restoration of IGF-1 improved overall insulin sensitivity and lipid profile in serum and reduced body adiposity, but was insufficient to protect against steatosis-induced hepatic inflammation or oxidative stress. We conclude that the impaired metabolism in states of GH resistance results from direct actions of GH on lipid uptake and de novo lipogenesis, whereas its actions on extrahepatic tissues are mediated by IGF-1.
Collapse
Affiliation(s)
- Zhongbo Liu
- Department of Basic Science & Craniofacial Biology, David B. Kriser Dental Center, NYU College of Dentistry, New York, NY
| | - Jose Cordoba-Chacon
- Research and Development, Jesse Brown VA Medical Center, Chicago, IL
- Department of Medicine, Section of Endocrinology, Diabetes and Metabolism, University of Illinois at Chicago, Chicago, IL
| | - Rhonda D Kineman
- Research and Development, Jesse Brown VA Medical Center, Chicago, IL
- Department of Medicine, Section of Endocrinology, Diabetes and Metabolism, University of Illinois at Chicago, Chicago, IL
| | | | - Radhika Muzumdar
- Division of Pediatric Endocrinology, Diabetes and Metabolism Consultation, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Zhenwei Gong
- Division of Pediatric Endocrinology, Diabetes and Metabolism Consultation, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Haim Werner
- Department of Human Molecular Genetics and Biochemistry, The Sackler School of Medicine, Tel Aviv University, Ramat Aviv, Israel
| | - Shoshana Yakar
- Department of Basic Science & Craniofacial Biology, David B. Kriser Dental Center, NYU College of Dentistry, New York, NY
| |
Collapse
|
47
|
Genetic variation in the insulin, insulin-like growth factor, growth hormone, and leptin pathways in relation to breast cancer in African-American women: the AMBER consortium. NPJ Breast Cancer 2016; 2. [PMID: 27942580 PMCID: PMC5142758 DOI: 10.1038/npjbcancer.2016.34] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The insulin/insulin-like growth factor (IGF) system and related pathways such as growth hormone, and leptin signaling have a key role in cancer development. It is unclear how germline variation in these pathways affects breast cancer risk. We conducted gene-based analyses of 184 genes in the insulin/IGF, growth hormone, and leptin pathways to identify genetic variation associated with risk of breast cancer overall, and for estrogen receptor (ER) subtypes. Tag single-nucleotide polymorphisms (SNPs) for each gene were selected and genotyped on a customized Illumina SNP array. Imputation was carried out using 1000 Genomes haplotypes. The analysis included 91,627 SNPs genotyped or imputed in 3,663 breast cancer cases, (1,983 ER-positive and 1,098 ER-negative) and 4,687 controls from the African American Breast Cancer Epidemiology and Risk consortium, a collaborative project of four large studies of breast cancer in African-American women (Carolina Breast Cancer Study, Black Women's Health Study, Women's Circle of Health Study, and Multiethnic Cohort). We used a multi-locus adaptive joint test to determine the association of each gene with overall breast cancer and ER subtypes. The most significant gene associations (P⩽0.01) were BAIAP2 and CALM2 for overall breast cancer; BAIAP2 and CSNK2A1 for ER+ breast cancer; and BRAF, BAD, and MAPK3 for ER− breast cancer. The association of BAD with ER− breast cancer was explained by a two-SNP risk model; all other associations were best explained by one-SNP risk models. In total, six genes and seven SNPs had suggestive associations with overall breast cancer or ER subtypes in African-American women.
Collapse
|
48
|
Svensson J, Sjögren K, Levin M, Borén J, Tivesten Å, Ohlsson C. Increased diet-induced fatty streak formation in female mice with deficiency of liver-derived insulin-like growth factor-I. Endocrine 2016; 52:550-60. [PMID: 26627099 PMCID: PMC4879167 DOI: 10.1007/s12020-015-0809-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 11/17/2015] [Indexed: 01/28/2023]
Abstract
The role of endocrine IGF-I for atherosclerosis is unclear. We determined the importance of circulating, liver-derived IGF-I for fatty streak formation in mice. Mice with adult, liver-specific IGF-I inactivation (LI-IGF-I(-/-) mice, serum IGF-I reduced by approximately 80 %) and control mice received an atherogenic (modified Paigen) diet between 6 and 12 months of age. At study end, Oil Red O staining of aortic root cryosections showed increased fatty streak area and lipid deposition in female but not in male LI-IGF-I(-/-) mice compared to controls. Mac-2 staining of aortic root and measurements of CD68 mRNA level in femoral artery revealed increased macrophage accumulation in proportion to the increased fatty streak area in female LI-IGF-I(-/-) mice. Moreover, female LI-IGF-I(-/-) mice displayed increased serum cholesterol and interleukin-6 as well as increased vascular cell-adhesion molecule 1 (VCAM1) mRNA levels in the femoral artery and elevated VCAM1 protein expression in the aortic root. Thus, increased diet-induced fatty streak formation in female LI-IGF-I(-/-) mice was associated with increased serum cholesterol and signs of systemic inflammation, endothelial activation, lipid deposition, and macrophage infiltration in the vascular wall.
Collapse
Affiliation(s)
- Johan Svensson
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 45, Göteborg, Sweden.
- Department of Internal Medicine, Sahlgrenska University Hospital, Gröna Stråket 8, 413 45, Göteborg, Sweden.
| | - Klara Sjögren
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 45, Göteborg, Sweden
| | - Malin Levin
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 45, Göteborg, Sweden
| | - Jan Borén
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 45, Göteborg, Sweden
| | - Åsa Tivesten
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 45, Göteborg, Sweden
| | - Claes Ohlsson
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 45, Göteborg, Sweden
| |
Collapse
|
49
|
Akter S, Kurotani K, Kashino I, Goto A, Mizoue T, Noda M, Sawada N, Tsugane S. High Dietary Acid Load Score Is Associated with Increased Risk of Type 2 Diabetes in Japanese Men: The Japan Public Health Center-based Prospective Study. J Nutr 2016; 146:1076-1083. [PMID: 27052540 DOI: 10.3945/jn.115.225177] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 03/01/2016] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Diet-induced metabolic acidosis has been linked to cardiometabolic abnormalities in Westerners, but the evidence on this issue is scarce in Asians. OBJECTIVE The present study prospectively examined the association between dietary acid load and type 2 diabetes (T2D) in Japanese adults. METHODS Study participants were 27,809 men and 36,851 women, aged 45-75 y, who completed a dietary questionnaire of the second survey of the Japan Public Health Center-based Prospective Study and had no previous history of T2D. Dietary intake was assessed by using a validated 147-item food-frequency questionnaire. Potential renal acid load (PRAL) and net endogenous acid production (NEAP) scores were derived from the nutrient intake. A multilevel logistic regression model was used to estimate ORs and 95% CIs of self-reported, physician-diagnosed T2D over 5 y, with adjustment for potential confounding variables. RESULTS A total of 1191 cases of newly diagnosed T2D were reported. PRAL score was positively associated with T2D in men; the multivariable-adjusted ORs (95% CIs) for the lowest through the highest quartiles of PRAL were 1.00, 1.09 (0.87, 1.36), 1.10 (0.88, 1.37), and 1.25 (1.01, 1.55) (P-trend = 0.047). Further adjustment for dietary intake strengthened the association. NEAP score was not associated with the risk of T2D (P-trend = 0.20). In stratified analyses, the positive association between PRAL and T2D was confined to younger men (age <50 y; P-trend = 0.046). There was no association between dietary acid load score and T2D in women. CONCLUSION A high dietary acid load score is associated with an increased risk of T2D in Japanese men.
Collapse
Affiliation(s)
- Shamima Akter
- Department of Epidemiology and Prevention, Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo, Japan;
| | - Kayo Kurotani
- Department of Epidemiology and Prevention, Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo, Japan
| | - Ikuko Kashino
- Department of Epidemiology and Prevention, Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo, Japan
| | - Atsushi Goto
- Department of Public Health, Tokyo Women's University, Tokyo, Japan; Department of Diabetes Research, National Center for Global Health and Medicine, Tokyo, Japan; and
| | - Tetsuya Mizoue
- Department of Epidemiology and Prevention, Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo, Japan
| | - Mitsushiko Noda
- Department of Public Health, Tokyo Women's University, Tokyo, Japan
| | - Norie Sawada
- Epidemiology and Prevention Group, Research Center for Cancer Prevention and Screening, National Cancer Center, Tokyo, Japan
| | - Shoichiro Tsugane
- Epidemiology and Prevention Group, Research Center for Cancer Prevention and Screening, National Cancer Center, Tokyo, Japan
| |
Collapse
|
50
|
Bartke A, List EO, Kopchick JJ. The somatotropic axis and aging: Benefits of endocrine defects. Growth Horm IGF Res 2016; 27:41-45. [PMID: 26925766 PMCID: PMC4792645 DOI: 10.1016/j.ghir.2016.02.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 01/27/2016] [Accepted: 02/12/2016] [Indexed: 12/15/2022]
Abstract
Reduced somatotropic [growth hormone (GH) and insulin-like growth factor-1 (IGF-1)] action has been associated with delayed and slower aging, reduced risk of frailty, reduced age-related disease and functional decline, and with remarkably extended longevity. Recent studies have added to the evidence that these relationships discovered in laboratory populations of mice apply to other mammalian species. However, the relationship of the somatotropic signaling to human aging is less striking, complex and controversial. In mice, targeted deletion of GH receptors (GHR) in the liver, muscle or adipose tissue affected multiple metabolic parameters but failed to reproduce the effects of global GHR deletion on longevity. Continued search for mechanisms of extended longevity in animals with GH deficiency or resistance focused attention on different pathways of mechanistic target of rapamycin (mTOR), energy metabolism, regulation of local IGF-1 levels and resistance to high-fat diet (HFD).
Collapse
Affiliation(s)
- Andrzej Bartke
- SIU School of Medicine, Department of Internal Medicine, 801 N. Rutledge, P.O. Box 19628, Springfield, IL 62794-9628, United States.
| | - Edward O List
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, United States; Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, United States
| | - John J Kopchick
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, United States; Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, United States
| |
Collapse
|