1
|
Guerra-Cantera S, Frago LM, Jiménez-Hernaiz M, Collado-Pérez R, Canelles S, Ros P, García-Piqueras J, Pérez-Nadador I, Barrios V, Argente J, Chowen JA. The metabolic effects of resumption of a high fat diet after weight loss are sex dependent in mice. Sci Rep 2023; 13:13227. [PMID: 37580448 PMCID: PMC10425431 DOI: 10.1038/s41598-023-40514-w] [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: 05/29/2023] [Accepted: 08/11/2023] [Indexed: 08/16/2023] Open
Abstract
Dietary restriction is a frequent strategy for weight loss, but adherence is difficult and returning to poor dietary habits can result in more weight gain than that previously lost. How weight loss due to unrestricted intake of a healthy diet affects the response to resumption of poor dietary habits is less studied. Moreover, whether this response differs between the sexes and if the insulin-like growth factor (IGF) system, sex dependent and involved in metabolic control, participates is unknown. Mice received rodent chow (6% Kcal from fat) or a high-fat diet (HFD, 62% Kcal from fat) for 4 months, chow for 3 months plus 1 month of HFD, or HFD for 2 months, chow for 1 month then HFD for 1 month. Males and females gained weight on HFD and lost weight when returned to chow at different rates (p < 0.001), but weight gain after resumption of HFD intake was not affected by previous weight loss in either sex. Glucose metabolism was more affected by HFD, as well as the re-exposure to HFD after weight loss, in males. This was associated with increases in hypothalamic mRNA levels of IGF2 (p < 0.01) and IGF binding protein (IGFBP) 2 (p < 0.05), factors involved in glucose metabolism, again only in males. Likewise, IGF2 increased IGFBP2 mRNA levels only in hypothalamic astrocytes from males (p < 0.05). In conclusion, the metabolic responses to dietary changes were less severe and more delayed in females and the IGF system might be involved in some of the sex specific observations.
Collapse
Affiliation(s)
- Santiago Guerra-Cantera
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Laura M Frago
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - María Jiménez-Hernaiz
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Roberto Collado-Pérez
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
| | - Sandra Canelles
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Purificación Ros
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
- Department of Endocrinology, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | - Jorge García-Piqueras
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
| | - Iris Pérez-Nadador
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
| | - Vicente Barrios
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Jesús Argente
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain.
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain.
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.
- IMDEA Food Institute, CEI UAM + CSIC, Madrid, Spain.
| | - Julie A Chowen
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain.
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.
- IMDEA Food Institute, CEI UAM + CSIC, Madrid, Spain.
| |
Collapse
|
2
|
Macvanin M, Gluvic Z, Radovanovic J, Essack M, Gao X, Isenovic ER. New insights on the cardiovascular effects of IGF-1. Front Endocrinol (Lausanne) 2023; 14:1142644. [PMID: 36843588 PMCID: PMC9947133 DOI: 10.3389/fendo.2023.1142644] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 01/26/2023] [Indexed: 02/11/2023] Open
Abstract
INTRODUCTION Cardiovascular (CV) disorders are steadily increasing, making them the world's most prevalent health issue. New research highlights the importance of insulin-like growth factor 1 (IGF-1) for maintaining CV health. METHODS We searched PubMed and MEDLINE for English and non-English articles with English abstracts published between 1957 (when the first report on IGF-1 identification was published) and 2022. The top search terms were: IGF-1, cardiovascular disease, IGF-1 receptors, IGF-1 and microRNAs, therapeutic interventions with IGF-1, IGF-1 and diabetes, IGF-1 and cardiovascular disease. The search retrieved original peer-reviewed articles, which were further analyzed, focusing on the role of IGF-1 in pathophysiological conditions. We specifically focused on including the most recent findings published in the past five years. RESULTS IGF-1, an anabolic growth factor, regulates cell division, proliferation, and survival. In addition to its well-known growth-promoting and metabolic effects, there is mounting evidence that IGF-1 plays a specialized role in the complex activities that underpin CV function. IGF-1 promotes cardiac development and improves cardiac output, stroke volume, contractility, and ejection fraction. Furthermore, IGF-1 mediates many growth hormones (GH) actions. IGF-1 stimulates contractility and tissue remodeling in humans to improve heart function after myocardial infarction. IGF-1 also improves the lipid profile, lowers insulin levels, increases insulin sensitivity, and promotes glucose metabolism. These findings point to the intriguing medicinal potential of IGF-1. Human studies associate low serum levels of free or total IGF-1 with an increased risk of CV and cerebrovascular illness. Extensive human trials are being conducted to investigate the therapeutic efficacy and outcomes of IGF-1-related therapy. DISCUSSION We anticipate the development of novel IGF-1-related therapy with minimal side effects. This review discusses recent findings on the role of IGF-1 in the cardiovascular (CVD) system, including both normal and pathological conditions. We also discuss progress in therapeutic interventions aimed at targeting the IGF axis and provide insights into the epigenetic regulation of IGF-1 mediated by microRNAs.
Collapse
Affiliation(s)
- Mirjana Macvanin
- Department of Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
- *Correspondence: Mirjana Macvanin,
| | - Zoran Gluvic
- Clinic for Internal Medicine, Department of Endocrinology and Diabetes, Zemun Clinical Hospital, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Jelena Radovanovic
- Department of Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Magbubah Essack
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
- Computer Science Program, Computer, Electrical and Mathematical Sciences and Engineering Division (CEMSE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Xin Gao
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
- Computer Science Program, Computer, Electrical and Mathematical Sciences and Engineering Division (CEMSE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Esma R. Isenovic
- Department of Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
3
|
Cao C, Duan P, Li W, Guo Y, Zhang J, Gui Y, Yuan S. Lack of miR-379/miR-544 Cluster Resists High-Fat Diet-Induced Obesity and Prevents Hepatic Triglyceride Accumulation in Mice. Front Cell Dev Biol 2021; 9:720900. [PMID: 34527673 PMCID: PMC8435714 DOI: 10.3389/fcell.2021.720900] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 08/10/2021] [Indexed: 12/21/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) affects obesity-associated metabolic syndrome, which exhibits hepatic steatosis, insulin insensitivity and glucose intolerance. Emerging evidence suggests that microRNAs (miRNAs) are essential for the metabolic homeostasis of liver tissues. Many hepatic miRNAs located in the miR-379/miR-544 cluster were significantly increased in leptin-receptor-deficient type 2 mice (db/db), a mouse model of diabetes. However, the function of the miR-379/miR-544 cluster in the process of hepatic steatosis remains unclear. Here, we report that the novel function of miR-379/miR-544 cluster in regulating obesity-mediated metabolic dysfunction. Genetical mutation of miR-379/miR-544 cluster in mice displayed resistance to high-fat diet (HFD)-induced obesity with moderate hepatic steatosis and hypertriglyceridemia. In vitro studies revealed that silencing of miR-379 in human hepatocellular carcinoma (HepG2) cells ameliorated palmitic acid-induced elevation of cellular triglycerides, and overexpression of miR-379 had the opposite effect. Moreover, Igf1r (Insulin-like growth factor 1 receptor) and Dlk1 (Delta-like homolog 1) were directly targeted by miR-379 and miR-329, respectively, and elevated in the livers of the miR-379/miR-544 cluster knockout mice fed on HFD. Further transcriptome analyses revealed that the hepatic gene expressions are dysregulated in miR-379/miR-544 knockout mice fed with HFD. Collectively, our findings identify the miR-379/miR-544 cluster as integral components of a regulatory circuit that functions under conditions of metabolic stress to control hepatic steatosis. Thus, this miRNA cluster provides potential targets for pharmacologic intervention in obesity and NAFLD.
Collapse
Affiliation(s)
- Congcong Cao
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen-Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Peng Duan
- Department of Obstetrics and Gynaecology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, China
| | - Wencun Li
- The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yang Guo
- College of Pharmacy, Hubei University of Medicine, Shiyan, China
| | - Jin Zhang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yaoting Gui
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen-Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Shuiqiao Yuan
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, China
| |
Collapse
|
4
|
Insulin mitigates apoptosis of porcine follicular granulosa cells by downregulating BimEL. Reprod Biol 2019; 19:293-298. [PMID: 31561987 DOI: 10.1016/j.repbio.2019.08.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 07/31/2019] [Accepted: 08/08/2019] [Indexed: 01/02/2023]
Abstract
Follicular growth or atresia is governed by the survival and apoptosis of granulosa cells. Increasing evidence shows that follicle growth is influenced by energy intake, which is positively related to insulin levels. However, the function of insulin in granulosa cell survival is poorly understood. This study focused on the effects of insulin on porcine medium follicle granulosa cell survival. In the present study, we showed that insulin markedly mitigated the apoptosis of porcine granulosa cells following serum starvation. Moreover, insulin activated the PI3K/Akt pathway to downregulate bim mRNA expression and simultaneously promoted the phosphorylation of BimEL through activating ERK 1/2, both of which reduced the level of BimEL. The results demonstrate that insulin protected the granulosa cells against apoptosis by reducing levels of the pro-apoptotic protein BimEL. However, the concentration of insulin (1 μg/ml) was relatively high. High levels of insulin partly combined with the IGF-1 receptor to play its roles in granulosa cells. This experiment provides new insight into the role of insulin in granulosa cells and sheds light on nutrition-reproduction interactions.
Collapse
|
5
|
Aguirre GA, De Ita JR, de la Garza RG, Castilla-Cortazar I. Insulin-like growth factor-1 deficiency and metabolic syndrome. J Transl Med 2016; 14:3. [PMID: 26733412 PMCID: PMC4702316 DOI: 10.1186/s12967-015-0762-z] [Citation(s) in RCA: 186] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 12/26/2015] [Indexed: 02/06/2023] Open
Abstract
Consistent evidence associates IGF-1 deficiency and metabolic syndrome. In this review, we will focus on the metabolic effects of IGF-1, the concept of metabolic syndrome and its clinical manifestations (impaired lipid profile, insulin resistance, increased glucose levels, obesity, and cardiovascular disease), discussing whether IGF-1 replacement therapy could be a beneficial strategy for these patients. The search plan was made in Medline for Pubmed with the following mesh terms: IGF-1 and "metabolism, carbohydrate, lipids, proteins, amino acids, metabolic syndrome, cardiovascular disease, diabetes" between the years 1963-2015. The search includes animal and human protocols. In this review we discuss the relevant actions of IGF-1 on metabolism and the implication of IGF-1 deficiency in the establishment of metabolic syndrome. Multiple studies (in vitro and in vivo) demonstrate the association between IGF-1 deficit and deregulated lipid metabolism, cardiovascular disease, diabetes, and an altered metabolic profile of diabetic patients. Based on the available data we propose IGF-1 as a key hormone in the pathophysiology of metabolic syndrome; due to its implications in the metabolism of carbohydrates and lipids. Previous data demonstrates how IGF-1 can be an effective option in the treatment of this worldwide increasing condition. It has to distinguished that the replacement therapy should be only undertaken to restore the physiological levels, never to exceed physiological ranges.
Collapse
Affiliation(s)
- G A Aguirre
- Escuela de Medicina, Tecnologico de Monterrey, Avenida Morones Prieto No. 3000 Pte. Col. Los Doctores, 64710, Monterrey, Nuevo León, Mexico.
| | - J Rodríguez De Ita
- Escuela de Medicina, Tecnologico de Monterrey, Avenida Morones Prieto No. 3000 Pte. Col. Los Doctores, 64710, Monterrey, Nuevo León, Mexico.
| | - R G de la Garza
- Escuela de Medicina, Tecnologico de Monterrey, Avenida Morones Prieto No. 3000 Pte. Col. Los Doctores, 64710, Monterrey, Nuevo León, Mexico.
| | - I Castilla-Cortazar
- Escuela de Medicina, Tecnologico de Monterrey, Avenida Morones Prieto No. 3000 Pte. Col. Los Doctores, 64710, Monterrey, Nuevo León, Mexico.
- Fundación de Investigación HM Hospitales, Madrid, Spain.
| |
Collapse
|
6
|
Hwang JY, Sim X, Wu Y, Liang J, Tabara Y, Hu C, Hara K, Tam CHT, Cai Q, Zhao Q, Jee S, Takeuchi F, Go MJ, Ong RTH, Ohkubo T, Kim YJ, Zhang R, Yamauchi T, So WY, Long J, Gu D, Lee NR, Kim S, Katsuya T, Oh JH, Liu J, Umemura S, Kim YJ, Jiang F, Maeda S, Chan JCN, Lu W, Hixson JE, Adair LS, Jung KJ, Nabika T, Bae JB, Lee MH, Seielstad M, Young TL, Teo YY, Kita Y, Takashima N, Osawa H, Lee SH, Shin MH, Shin DH, Choi BY, Shi J, Gao YT, Xiang YB, Zheng W, Kato N, Yoon M, He J, Shu XO, Ma RCW, Kadowaki T, Jia W, Miki T, Qi L, Tai ES, Mohlke KL, Han BG, Cho YS, Kim BJ. Genome-wide association meta-analysis identifies novel variants associated with fasting plasma glucose in East Asians. Diabetes 2015; 64:291-8. [PMID: 25187374 PMCID: PMC4274808 DOI: 10.2337/db14-0563] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Fasting plasma glucose (FPG) has been recognized as an important indicator for the overall glycemic state preceding the onset of metabolic diseases. So far, most indentified genome-wide association loci for FPG were derived from populations with European ancestry, with a few exceptions. To extend a thorough catalog for FPG loci, we conducted meta-analyses of 13 genome-wide association studies in up to 24,740 nondiabetic subjects with East Asian ancestry. Follow-up replication analyses in up to an additional 21,345 participants identified three new FPG loci reaching genome-wide significance in or near PDK1-RAPGEF4, KANK1, and IGF1R. Our results could provide additional insight into the genetic variation implicated in fasting glucose regulation.
Collapse
Affiliation(s)
- Joo-Yeon Hwang
- Center for Genome Science, National Institute of Health, Osong Health Technology Administration Complex, Chungcheongbuk-do, Republic of Korea
| | - Xueling Sim
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, MI Centre for Molecular Epidemiology, National University of Singapore, Singapore, Singapore
| | - Ying Wu
- Department of Genetics, University of North Carolina, Chapel Hill, NC
| | - Jun Liang
- Department of Nutrition, Harvard School of Public Health, Boston, MA
| | - Yasuharu Tabara
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Cheng Hu
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Kazuo Hara
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan Department of Integrated Molecular Science on Metabolic Diseases, 22nd Century Medical and Research Center, The University of Tokyo, Tokyo, Japan
| | - Claudia H T Tam
- Department of Medicine and Therapeutics, Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong
| | - Qiuyin Cai
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center; and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN
| | - Qi Zhao
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA
| | - Sunha Jee
- Institute for Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Korea
| | - Fumihiko Takeuchi
- Department of Gene Diagnostics and Therapeutics, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Min Jin Go
- Center for Genome Science, National Institute of Health, Osong Health Technology Administration Complex, Chungcheongbuk-do, Republic of Korea
| | - Rick Twee Hee Ong
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
| | - Takayoshi Ohkubo
- Department of Planning for Drug Development and Clinical Evaluation, Tohoku University Graduate School of Pharmaceutical Sciences, Sendai, Japan Department of Health Science, Shiga University of Medical Science, Shiga, Japan
| | - Young Jin Kim
- Center for Genome Science, National Institute of Health, Osong Health Technology Administration Complex, Chungcheongbuk-do, Republic of Korea
| | - Rong Zhang
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Toshimasa Yamauchi
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Wing Yee So
- Department of Medicine and Therapeutics, Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong
| | - Jirong Long
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center; and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN
| | - Dongfeng Gu
- State Key Laboratory of Cardiovascular Disease, Department of Epidemiology, National Center for Cardiovascular Diseases, Beijing, China Population Genetics, Fuwai Hospital, National Center of Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China Peking Union Medical College, Beijing, China
| | - Nanette R Lee
- Office of Population Studies Foundation, University of San Carlos, Cebu City, Philippines
| | - Soriul Kim
- Institute for Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Korea
| | - Tomohiro Katsuya
- Department of Clinical Gene Therapy, Osaka University Graduate School of Medicine, Suita, Japan
| | - Ji Hee Oh
- Center for Genome Science, National Institute of Health, Osong Health Technology Administration Complex, Chungcheongbuk-do, Republic of Korea
| | - Jianjun Liu
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | - Satoshi Umemura
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University School of Medicine, Yokohama, Japan
| | - Yeon-Jung Kim
- Center for Genome Science, National Institute of Health, Osong Health Technology Administration Complex, Chungcheongbuk-do, Republic of Korea
| | - Feng Jiang
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Shiro Maeda
- Laboratory for Endocrinology, Metabolism and Kidney Diseases, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Juliana C N Chan
- Department of Medicine and Therapeutics, Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong
| | - Wei Lu
- Shanghai Municipal Center for Disease Control & Prevention, Shanghai, China
| | - James E Hixson
- Human Genetics Center, The University of Texas School of Public Health, Houston, TX
| | - Linda S Adair
- Department of Nutrition, University of North Carolina, Chapel Hill, NC
| | - Keum Ji Jung
- Institute for Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Korea
| | - Toru Nabika
- Department of Functional Pathology, Shimane University School of Medicine, Izumo, Japan
| | - Jae-Bum Bae
- Center for Genome Science, National Institute of Health, Osong Health Technology Administration Complex, Chungcheongbuk-do, Republic of Korea
| | - Mi Hee Lee
- Center for Genome Science, National Institute of Health, Osong Health Technology Administration Complex, Chungcheongbuk-do, Republic of Korea
| | - Mark Seielstad
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA
| | - Terri L Young
- Center for Human Genetics, Duke University Medical Center, Durham, NC
| | - Yik Ying Teo
- Centre for Molecular Epidemiology, National University of Singapore, Singapore, Singapore Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore Department of Statistics and Applied Probability, National University of Singapore, Singapore, Singapore Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore, Singapore
| | - Yoshikuni Kita
- Department of Health Science, Shiga University of Medical Science, Shiga, Japan
| | - Naoyuki Takashima
- Department of Health Science, Shiga University of Medical Science, Shiga, Japan
| | - Haruhiko Osawa
- Department of Molecular and Genetic Medicine, Ehime University Graduate School of Medicine, Toon, Japan
| | - So-Hyun Lee
- Center for Genome Science, National Institute of Health, Osong Health Technology Administration Complex, Chungcheongbuk-do, Republic of Korea
| | - Min-Ho Shin
- Department of Preventive Medicine, Chonnam National University Medical School, Gwangju, South Korea
| | - Dong Hoon Shin
- Department of Occupational and Environmental Medicine, Keimyung University Dongsan Medical Center, Daegu, South Korea
| | - Bo Youl Choi
- Department of Preventive Medicine, College of Medicine, Hanyang University, Seoul, Korea
| | - Jiajun Shi
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center; and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN
| | - Yu-Tang Gao
- Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yong-Bing Xiang
- Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center; and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN
| | - Norihiro Kato
- Department of Gene Diagnostics and Therapeutics, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Miwuk Yoon
- Institute for Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Korea
| | - Jiang He
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA
| | - Xiao Ou Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center; and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN
| | - Ronald C W Ma
- Department of Medicine and Therapeutics, Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong
| | - Takashi Kadowaki
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Weiping Jia
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Tetsuro Miki
- Department of Geriatric Medicine, Ehime University Graduate School of Medicine, Toon, Japan
| | - Lu Qi
- Department of Nutrition, Harvard School of Public Health, Boston, MA
| | - E Shyong Tai
- Department of Gene Diagnostics and Therapeutics, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan Department of Medicine, National University of Singapore, Singapore, Singapore Duke-National University of Singapore Graduate Medical School, Singapore, Singapore
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, NC
| | - Bok-Ghee Han
- Center for Genome Science, National Institute of Health, Osong Health Technology Administration Complex, Chungcheongbuk-do, Republic of Korea
| | - Yoon Shin Cho
- Department of Biomedical Science, Hallym University, Chuncheon, Korea
| | - Bong-Jo Kim
- Center for Genome Science, National Institute of Health, Osong Health Technology Administration Complex, Chungcheongbuk-do, Republic of Korea
| |
Collapse
|
7
|
Abstract
Persistent nutrient excess results in a compensatory increase in the β-cell number in mammals. It is unknown whether this response occurs in nonmammalian vertebrates, including zebrafish, a model for genetics and chemical genetics. We investigated the response of zebrafish β-cells to nutrient excess and the underlying mechanisms by culturing transgenic zebrafish larvae in solutions of different nutrient composition. The number of β-cells rapidly increases after persistent, but not intermittent, exposure to glucose or a lipid-rich diet. The response to glucose, but not the lipid-rich diet, required mammalian target of rapamycin activity. In contrast, inhibition of insulin/IGF-1 signaling in β-cells blocked the response to the lipid-rich diet, but not to glucose. Lineage tracing and marker expression analyses indicated that the new β-cells were not from self-replication but arose through differentiation of postmitotic precursor cells. On the basis of transgenic markers, we identified two groups of newly formed β-cells: one with nkx2.2 promoter activity and the other with mnx1 promoter activity. Thus, nutrient excess in zebrafish induces a rapid increase in β-cells though differentiation of two subpopulations of postmitotic precursor cells. This occurs through different mechanisms depending on the nutrient type and likely involves paracrine signaling between the differentiated β-cells and the precursor cells.
Collapse
|
8
|
Abstract
Insulin-like growth factor 1 (IGF-1) is a pleiotropic polypeptide. Its expression is tightly regulated and it plays significant roles during early development, maturation, and adulthood. This article discusses the roles of IGF-1 in determination of body size, skeletal acquisition, muscle growth, carbohydrate metabolism, and longevity, as learned from mouse models.
Collapse
Affiliation(s)
- Shoshana Yakar
- Department of Basic Science and Craniofacial Biology, David B. Kriser Dental Center, New York University College of Dentistry, New York, NY 10010-4086, USA.
| | | |
Collapse
|
9
|
Clemmons DR. Metabolic actions of insulin-like growth factor-I in normal physiology and diabetes. Endocrinol Metab Clin North Am 2012; 41:425-43, vii-viii. [PMID: 22682639 PMCID: PMC3374394 DOI: 10.1016/j.ecl.2012.04.017] [Citation(s) in RCA: 193] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Insulin-like growth factor-I (IGF-I) is closely related to insulin but has distinct metabolic actions. IGF-I is an important stimulant of protein synthesis in muscle, but it also stimulates free fatty acid use. The administration of IGF-I to patients with extreme insulin resistance results in improvement in glycemic control, and IGF-I is associated with lowering glucose and enhancing insulin sensitivity in Type 1 and Type 2 diabetes. However, patients with diabetes are also sensitive to stimulation of side effects in response to IGF-I. IGF-I coordinately links growth hormone and insulin actions and has direct effects on intermediary metabolism.
Collapse
Affiliation(s)
- David R Clemmons
- Division of Endocrinology, Department of Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC 27599-7170, USA.
| |
Collapse
|
10
|
Sugita M, Sugita H, Kim M, Mao J, Yasuda Y, Habiro M, Shinozaki S, Yasuhara S, Shimizu N, Martyn JJ, Kaneki M. Inducible nitric oxide synthase deficiency ameliorates skeletal muscle insulin resistance but does not alter unexpected lower blood glucose levels after burn injury in C57BL/6 mice. Metabolism 2012; 61:127-36. [PMID: 21816442 PMCID: PMC3304504 DOI: 10.1016/j.metabol.2011.06.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2011] [Revised: 05/30/2011] [Accepted: 06/02/2011] [Indexed: 01/04/2023]
Abstract
Burn injury is associated with inflammatory responses and metabolic alterations including insulin resistance. Impaired insulin receptor substrate-1 (IRS-1)-mediated insulin signal transduction is a major component of insulin resistance in skeletal muscle following burn injury. To further investigate molecular mechanisms that underlie burn injury-induced insulin resistance, we study a role of inducible nitric oxide synthase (iNOS), a major mediator of inflammation, on burn-induced muscle insulin resistance in iNOS-deficient mice. Full-thickness third-degree burn injury comprising 12% of total body surface area was produced in wild-type and iNOS-deficient C57BL/6 mice. Insulin-stimulated activation (phosphorylation) of IR, IRS-1, and Akt was assessed by immunoblotting and immunoprecipitation. Insulin-stimulated glucose uptake by skeletal muscle was evaluated ex vivo. Burn injury caused induction of iNOS in skeletal muscle of wild-type mice. The increase of iNOS expression paralleled the increase of insulin resistance, as evidenced by decreased tyrosine phosphorylation of IR and IRS-1, IRS-1 expression, insulin-stimulated activation of phosphatidylinositol 3-kinase and Akt/PKB, and insulin-stimulated glucose uptake in mouse skeletal muscle. The absence of iNOS in genetically engineered mice significantly lessened burn injury-induced insulin resistance in skeletal muscle. In wild-type mice, insulin tolerance test revealed whole-body insulin resistance in burned mice compared with sham-burned controls. This effect was reversed by iNOS deficiency. Unexpectedly, however, blood glucose levels were depressed in both wild-type and iNOS-deficient mice after burn injury. Gene disruption of iNOS ameliorated the effect of burn on IRS-1-mediated insulin signaling in skeletal muscle of mice. These findings indicate that iNOS plays a significant role in burn injury-induced skeletal muscle insulin resistance.
Collapse
Affiliation(s)
- Michiko Sugita
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Shriners Hospitals for Children, Boston, MA 02114, USA
| | - Hiroki Sugita
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Shriners Hospitals for Children, Boston, MA 02114, USA
| | - Minhye Kim
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Ji Mao
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Shriners Hospitals for Children, Boston, MA 02114, USA
| | - Yoshikazu Yasuda
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Shriners Hospitals for Children, Boston, MA 02114, USA
| | - Mayu Habiro
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Shohei Shinozaki
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Shriners Hospitals for Children, Boston, MA 02114, USA
| | - Shingo Yasuhara
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Shriners Hospitals for Children, Boston, MA 02114, USA
| | - Nobuyuki Shimizu
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Shriners Hospitals for Children, Boston, MA 02114, USA
| | - J.A. Jeevendra Martyn
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Shriners Hospitals for Children, Boston, MA 02114, USA
| | - Masao Kaneki
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Shriners Hospitals for Children, Boston, MA 02114, USA
- Corresponding author. Masao Kaneki; Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, 149 Thirteenth Street, Charlestown, MA 02129, USA, Phone: +1(617) 726-8122; Fax: +1(617) 726-8134;
| |
Collapse
|
11
|
Maor G, Vasiliver-Shamis G, Hazan-Brill R, Wertheimer E, Karnieli E. GLUT4 in murine bone growth: from uptake and translocation to proliferation and differentiation. Am J Physiol Endocrinol Metab 2011; 300:E613-23. [PMID: 20923959 DOI: 10.1152/ajpendo.90484.2008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Skeletal growth, taking place in the cartilaginous growth plates of long bones, consumes high levels of glucose for both metabolic and anabolic purposes. We previously showed that Glut4 is present in growing bone and is decreased in diabetes. In the present study, we examined the hypothesis that in bone, GLUT4 gene expression and function are regulated via the IGF-I receptor (IGF-IR) and that Glut4 plays an important role in bone growth. Insulin and IGF-I actions on skeletal growth and glucose uptake were determined using mandibular condyle (MC) organ cultures and MC-derived primary cell cultures (MCDC). Chondrogenesis was determined by following proliferation and differentiation activities using immunohistochemical (IHC) analysis of proliferating cell nuclear antigen and type II collagen expression, respectively. Overall condylar growth was assessed morphometrically. GLUT4 mRNA and protein levels were determined using in situ hybridization and IHC, respectively. Glut4 translocation to the cell membrane was assessed using confocal microscopy analysis of GFP-Glut4 fusion-transfected cells and immunogold and electron microscopy on MC sections; glucose uptake was assayed by 2-deoxyglucose (2-DOG) uptake. Both IGF-I and insulin-stimulated glucose uptake in MCDC, with IGF-I being tenfold more potent than insulin. Blockage of IGF-IR abrogated both IGF-I- and insulin-induced chondrogenesis and glucose metabolism. IGF-I, but not insulin, induced Glut4 translocation to the plasma membrane. Additionally, insulin induced both GLUT4 and IGF-IR gene expression and improved condylar growth in insulin receptor knockout mice-derived MC. Moreover, silencing of GLUT4 gene in MCDC culture abolished both IGF-I-induced glucose uptake and chondrocytic proliferation and differentiation. In growing bone, the IGF-IR pathway stimulates Glut4 translocation and enhances glucose uptake. Moreover, intact Glut4 cellular levels and translocation machinery are essential for early skeletal growth.
Collapse
Affiliation(s)
- Gila Maor
- Department of Anatomy and Cell Biology, Rappaport Faculty of Medicine, Tel Aviv University, Israel
| | | | | | | | | |
Collapse
|
12
|
Smith TJ. Insulin-like growth factor-I regulation of immune function: a potential therapeutic target in autoimmune diseases? Pharmacol Rev 2010; 62:199-236. [PMID: 20392809 DOI: 10.1124/pr.109.002469] [Citation(s) in RCA: 185] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
This topically limited review explores the relationship between the immune system and insulin-like growth factors (IGF-I and IGF-II) and the proteins through which they act, including IGF-I receptor (IGF-IR) and the IGF-I binding proteins. The IGF/IGF-IR pathway plays important and diverse roles in tissue development and function. It regulates cell cycle progression, apoptosis, and the translation of proteins. Many of the consequences ascribed to IGF-IR activation result from its association with several accessory proteins that are either identical or closely related to those involved in insulin receptor signaling. Relatively recent awareness that IGF-I and IGF-IR regulate immune function has cast this pathway in an unexpected light; it may represent an important switch governing the quality and amplitude of immune responses. IGF-I/IGF-IR signaling may also participate in the pathogenesis of autoimmune diseases, although its relationship with these processes seems complex and relatively unexplored. On the one hand, IGF-I seems to protect experimental animals from developing insulin-deficient diabetes mellitus. In contrast, activating antibodies directed at IGF-IR have been detected in patients with Graves' disease, where the receptor is overexpressed by multiple cell types. The frequency of IGF-IR+ B and T cells is substantially increased in patients with that disease. Potential involvement of IGF-I and IGF-IR in the pathogenesis of autoimmune diseases suggests that this pathway might constitute an attractive therapeutic target. IGF-IR has been targeted in efforts directed toward drug development for cancer, employing both small-molecule and monoclonal antibody approaches. These have been generally well-tolerated. Recognizing the broader role of IGF-IR in regulating both normal and pathological immune responses may offer important opportunities for therapeutic intervention in several allied diseases that have proven particularly difficult to treat.
Collapse
Affiliation(s)
- Terry J Smith
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan Medical School, 1000 Wall Street, Ann Arbor, MI 48105, USA.
| |
Collapse
|
13
|
Boucher J, Tseng YH, Kahn CR. Insulin and insulin-like growth factor-1 receptors act as ligand-specific amplitude modulators of a common pathway regulating gene transcription. J Biol Chem 2010; 285:17235-45. [PMID: 20360006 DOI: 10.1074/jbc.m110.118620] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Insulin and insulin-like growth factor-1 (IGF-1) act on highly homologous receptors, yet in vivo elicit distinct effects on metabolism and growth. To investigate how the insulin and IGF-1 receptors exert specificity in their biological responses, we assessed their role in the regulation of gene expression using three experimental paradigms: 1) preadipocytes before and after differentiation into adipocytes that express both receptors, but at different ratios; 2) insulin receptor (IR) or IGF1R knock-out preadipocytes that only express the complimentary receptor; and 3) IR/IGF1R double knock-out (DKO) cells reconstituted with the IR, IGF1R, or both. In wild-type preadipocytes, which express predominantly IGF1R, microarray analysis revealed approximately 500 IGF-1 regulated genes (p < 0.05). The largest of these were confirmed by quantitative PCR, which also revealed that insulin produced a similar effect, but with a smaller magnitude of response. After differentiation, when IR levels increase and IGF1R decrease, insulin became the dominant regulator of each of these genes. Measurement of the 50 most highly regulated genes by quantitative PCR did not reveal a single gene regulated uniquely via the IR or IGF1R using cells expressing exclusively IGF-1 or insulin receptors. Insulin and IGF-1 dose responses from 1 to 100 nm in WT, IRKO, IGFRKO, and DKO cells re-expressing IR, IGF1R, or both showed that insulin and IGF-1 produced effects in proportion to the concentration of ligand and the specific receptor on which they act. Thus, IR and IGF1R act as identical portals to the regulation of gene expression, with differences between insulin and IGF-1 effects due to a modulation of the amplitude of the signal created by the specific ligand-receptor interaction.
Collapse
Affiliation(s)
- Jeremie Boucher
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Joslin Diabetes Center, Boston, Massachusetts 02215, USA
| | | | | |
Collapse
|
14
|
Genetic Engineering Technology and Pharmacological Evaluation of Chinese Medicine. Chin J Nat Med 2010. [DOI: 10.3724/sp.j.1009.2009.00476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
15
|
Palsgaard J, Brown AE, Jensen M, Borup R, Walker M, De Meyts P. Insulin-like growth factor I (IGF-I) is a more potent regulator of gene expression than insulin in primary human myoblasts and myotubes. Growth Horm IGF Res 2009; 19:168-178. [PMID: 19010705 DOI: 10.1016/j.ghir.2008.09.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2008] [Revised: 09/15/2008] [Accepted: 09/24/2008] [Indexed: 11/28/2022]
Abstract
Conventionally, insulin is believed to induce a metabolic response, and IGF-I a mitogenic/differentiation response in vivo. However, several studies indicate that the roles of insulin and IGF-I may not be that easy to separate. In this study, insulin and IGF-I specificity in terms of gene regulation was investigated in primary human skeletal muscle cells before and after differentiation. Cell cultures were treated with 100 nM insulin, IGF-I or nothing for 4h, and gene expression was subsequently determined using the Affymetrix microarray platform. Insulin and IGF-I receptor levels were determined by qRT-PCR and by radioligand binding assays. In primary myoblasts, insulin did not have any significant effect on gene expression, whereas IGF-I regulated 229 genes. In primary myotubes, insulin regulated 105 genes, whereas IGF-I regulated 697 genes. Additionally, 99 genes were found to be differentially regulated by insulin and IGF-I in a direct comparison. The majority of these genes were specifically regulated by IGF-I, 16 genes were regulated by both ligands, and no genes were regulated by only insulin. The microarray results correlated with low levels of insulin receptors compared to IGF-I receptors as determined by radioligand binding assays. In the myotubes, we did not identify any ligand specificity in terms of functional categories. The major difference between the two ligands was their respective potencies in gene regulation, which was higher for IGF-I than for insulin. This was true for genes involved in both mitogenic and metabolic regulations. The data suggest that IGF-I is a more important metabolic regulator in skeletal muscle than previously estimated.
Collapse
Affiliation(s)
- Jane Palsgaard
- Receptor Systems Biology Laboratory, Hagedorn Research Institute, Niels Steensens vej 6, 2820 Gentofte, Denmark.
| | | | | | | | | | | |
Collapse
|
16
|
Nojima K, Ikegami H, Fujisawa T, Ueda H, Babaya N, Itoi-Babaya M, Yamaji K, Shibata M, Ogihara T. Food hardness as environmental factor in development of type 2 diabetes. Diabetes Res Clin Pract 2006; 74:1-7. [PMID: 16730844 DOI: 10.1016/j.diabres.2006.03.031] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2005] [Revised: 01/13/2006] [Accepted: 03/14/2006] [Indexed: 11/22/2022]
Abstract
The effect of hardness of the diet as an environmental factor on the development of diabetes was investigated in a mouse model of type 2 diabetes. NSY and control C3H/He mice were fed several types of dietary chow from 4 weeks of age. Autoclaved CRF-1, whose major components are almost the same as those of the MF diet except for increased pellet hardness, resulted in a significant reduction in body weight in both NSY (p<0.05) and C3H (p<0.001) mice at 16 weeks of age. The prevalence of diabetes in NSY mice fed autoclaved CRF-1 was significantly lower than that in those fed MF at 36 weeks of age (p<0.05), which was associated with a significant decrease in body weight (p<0.0001). At 16 weeks of age, NSY mice fed with a hard diet (autoclaved CRF-1) showed a significantly lower body weight (32.1+/-0.3g) and blood glucose levels during ipGTT than those with fed a normal (gamma-irradiated CRF-1) (35.6+/-1.3g, p<0.05 and <0.01, respectively) or soft (powdered CRF-1) (p<0.05 and <0.05, respectively) diet. Switching from normal (gamma-irradiated) to hard (autoclaved) chow, even after the development diabetes at 36 weeks of age, markedly improved glucose intolerance after 4 weeks in NSY mice despite the small change in body weight. These results indicate the importance of food hardness as an environmental factor in the development of type 2 diabetes.
Collapse
Affiliation(s)
- Koji Nojima
- Department of Geriatric Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Affiliation(s)
- Liam J Murphy
- Department of Internal Medicine, Room 843 John Buhler Research Centre, University of Manitoba, 715 McDermot Avenue, Winnipeg, Manitoba, Canada R3E 3P4.
| |
Collapse
|
18
|
Ning Y, Schuller AGP, Bradshaw S, Rotwein P, Ludwig T, Frystyk J, Pintar JE. Diminished growth and enhanced glucose metabolism in triple knockout mice containing mutations of insulin-like growth factor binding protein-3, -4, and -5. Mol Endocrinol 2006; 20:2173-86. [PMID: 16675541 DOI: 10.1210/me.2005-0196] [Citation(s) in RCA: 127] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
IGF-I and IGF-II are essential regulators of mammalian growth, development and metabolism, whose actions are modified by six high-affinity IGF binding proteins (IGFBPs). New lines of knockout (KO) mice lacking either IGFBP-3, -4, or -5 had no apparent deficiencies in growth or metabolism beyond a modest growth impairment (approximately 85-90% of wild type) when IGFBP-4 was eliminated. To continue to address the roles of these proteins in whole animal physiology, we generated combinational IGFBP KO mice. Mice homozygous for targeted defects in IGFBP-3, -4, and -5 remain viable and at birth were the same size as IGFBP-4 KO mice. Unlike IGFBP-4 KO mice, however, the triple KO mice became significantly smaller by adulthood (78% wild type) and had significant reductions in fat pad accumulation (P < 0.05), circulating levels of total IGF-I (45% of wild type; P < 0.05) and IGF-I bioactivity (37% of wild type; P < 0.05). Metabolically, triple KO mice showed normal insulin tolerance, but a 37% expansion (P < 0.05) of beta-cell number and significantly increased insulin secretion after glucose challenge, which leads to enhanced glucose disposal. Finally, triple KO mice demonstrated a tissue-specific decline in activation of the Erk signaling pathway as well as weight of the quadriceps muscle. Taken together, these data provide direct evidence for combinatorial effects of IGFBP-3, -4, and -5 in both metabolism and at least some soft tissues and strongly suggest overlapping roles for IGFBP-3 and -5 in maintaining IGF-I-mediated postnatal growth in mice.
Collapse
Affiliation(s)
- Yun Ning
- Department of Neuroscience and Cell Biology, University of Medicine and Dentistry of New Jersey, Piscataway, New Jersey 08854, USA
| | | | | | | | | | | | | |
Collapse
|
19
|
Fernandez AM, LeRoith D. Skeletal Muscle. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2005; 567:117-47. [PMID: 16370138 DOI: 10.1007/0-387-26274-1_5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
20
|
Frystyk J. Free insulin-like growth factors -- measurements and relationships to growth hormone secretion and glucose homeostasis. Growth Horm IGF Res 2004; 14:337-375. [PMID: 15336229 DOI: 10.1016/j.ghir.2004.06.001] [Citation(s) in RCA: 241] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
IGF-I is a multipotent growth factor with important actions on normal tissue growth and regeneration. In addition, IGF-I has been suggested to have beneficial effects on glucose homeostasis due to its glucose lowering and insulin sensitizing actions. However, not all effects of IGF-I are considered to be favorable; thus, epidemiological studies suggest that IGF-I is also involved in the development of common cancers, atherosclerosis and type 2 diabetes. The biological actions of IGF-I are modulated by at least six IGF-binding proteins, which bind approximately 99% of the circulating IGF-I pool. So far, most in vivo studies have used serum or plasma total (extractable IGF-I) as an estimate of the bioactivity of IGF-I in vivo. However, within the last decade, validated assays for measurement of free IGF-I have been described. This review aims to discuss the current assays for free IGF-I and their advances in relation to the traditional measurement of total IGF-I. The literature overview will focus on the role of circulating free versus total IGF-I in the feedback regulation of GH release, and the possible involvement of the circulating IGF-system in glucose homeostasis.
Collapse
Affiliation(s)
- Jan Frystyk
- Medical Research Laboratories and Medical Department M, Aarhus University Hospital, Norrebrogade, Aarhus, Denmark.
| |
Collapse
|