51
|
Dommerholt MB, Dionne DA, Hutchinson DF, Kruit JK, Johnson JD. Metabolic effects of short-term caloric restriction in mice with reduced insulin gene dosage. J Endocrinol 2018; 237:59-71. [PMID: 29439088 DOI: 10.1530/joe-17-0505] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 02/08/2018] [Indexed: 11/08/2022]
Abstract
Caloric restriction (CR) is the only environmental intervention with robust evidence that it extends lifespan and delays the symptoms of aging, but its mechanisms are incompletely understood. Based on the prolonged longevity of knockout models, it was hypothesized that the insulin-IGF pathway could be a target for developing a CR mimic. This study aimed to test whether CR has additive effects on glucose homeostasis and beta-cell function in mice with reduced insulin gene dosage. To study models with a range of basal insulin levels, wild-type C57BL/6J and mice on an Ins2-/- background, were put on 8 weeks of 40% CR at various ages. Both male and female mice rapidly lost weight due to a reduced WAT mass. Glucose tolerance was improved and fasting glucose levels were reduced by CR in both wild type and 45- and 70-week-old Ins2-/- mice. The effects of CR and reduced insulin on glucose tolerance were non-additive in 20-week-old mice. Interestingly, mice on CR generally exhibited an inability to further depress blood glucose after insulin injection, pointing to possible alterations in insulin sensitivity. In conclusion, our results demonstrate that CR can cause weight loss in the context of reduced insulin production, but that CR-improved glucose homeostasis does not occur near the 'insulin floor' in young mice. Collectively, these data shed further light on the relationships between CR, insulin and glucose homeostasis.
Collapse
Affiliation(s)
- Marleen B Dommerholt
- Department of Cellular and Physiological SciencesUniversity of British Columbia, Vancouver, Canada
- Department of PediatricsUniversity Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Derek A Dionne
- Department of Cellular and Physiological SciencesUniversity of British Columbia, Vancouver, Canada
| | - Daria F Hutchinson
- Department of Cellular and Physiological SciencesUniversity of British Columbia, Vancouver, Canada
| | - Janine K Kruit
- Department of PediatricsUniversity Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - James D Johnson
- Department of Cellular and Physiological SciencesUniversity of British Columbia, Vancouver, Canada
| |
Collapse
|
52
|
Ma L, Wang R, Dong W, Zhao Z. Caloric restriction can improve learning and memory in C57/BL mice probably via regulation of the AMPK signaling pathway. Exp Gerontol 2018; 102:28-35. [DOI: 10.1016/j.exger.2017.11.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Revised: 10/24/2017] [Accepted: 11/22/2017] [Indexed: 10/18/2022]
|
53
|
Davy PMC, Allsopp RC, Donlon TA, Morris BJ, Willcox DC, Willcox BJ. FOXO3 and Exceptional Longevity: Insights From Hydra to Humans. Curr Top Dev Biol 2018; 127:193-212. [PMID: 29433738 DOI: 10.1016/bs.ctdb.2017.10.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Aging is a complex, multifactorial process with significant plasticity. While several biological pathways appear to influence aging, few genes have been identified that are both evolutionarily conserved and have a strong impact on aging and age-related phenotypes. The FoxO3 gene (FOXO3), and its homologs in model organisms, appears especially important, forming a key gene in the insulin/insulin-like growth factor-signaling pathway, and influencing life span across diverse species. We highlight some of the key findings that are associated with FoxO3 protein, its gene and homologs in relation to lifespan in different species, and the insights these findings might provide about the molecular, cellular, and physiological processes that modulate aging and longevity in humans.
Collapse
Affiliation(s)
- Philip M C Davy
- Institute for Biogenesis Research, University of Hawaii, Honolulu, HI, United States
| | - Richard C Allsopp
- Institute for Biogenesis Research, University of Hawaii, Honolulu, HI, United States
| | - Timothy A Donlon
- Honolulu Heart Program/Honolulu-Asia Aging Study, Kuakini Medical Center, Honolulu, HI, United States; Ohana Genetics, Honolulu, HI, United States
| | - Brian J Morris
- Honolulu Heart Program/Honolulu-Asia Aging Study, Kuakini Medical Center, Honolulu, HI, United States; John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, United States; School of Medical Sciences and Bosch Institute, University of Sydney, Sydney, NSW, Australia
| | - Donald Craig Willcox
- Honolulu Heart Program/Honolulu-Asia Aging Study, Kuakini Medical Center, Honolulu, HI, United States; John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, United States; Okinawa International University, Ginowan, Okinawa, Japan
| | - Bradley J Willcox
- Honolulu Heart Program/Honolulu-Asia Aging Study, Kuakini Medical Center, Honolulu, HI, United States; John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, United States.
| |
Collapse
|
54
|
Bettedi L, Foukas LC. Growth factor, energy and nutrient sensing signalling pathways in metabolic ageing. Biogerontology 2017; 18:913-929. [PMID: 28795262 PMCID: PMC5684302 DOI: 10.1007/s10522-017-9724-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 07/21/2017] [Indexed: 01/24/2023]
Abstract
The field of the biology of ageing has received increasing attention from a biomedical point of view over the past decades. The main reason has been the realisation that increases in human population life expectancy are accompanied by late onset diseases. Indeed, ageing is the most important risk factor for a number of neoplastic, neurodegenerative and metabolic pathologies. Advances in the knowledge of the genetics of ageing, mainly through research in model organisms, have implicated various cellular processes and the respective signalling pathways that regulate them in cellular and organismal ageing. Associated with ageing is a dysregulation of metabolic homeostasis usually manifested as age-related obesity, diminished insulin sensitivity and impaired glucose and lipid homeostasis. Metabolic deterioration contributes to the ageing phenotype and metabolic pathologies are thought to be one of the main factors limiting the potential for lifespan extension. Great efforts have been directed towards identifying pharmacological interventions with the potential to improve healthspan and a number of natural and synthetic compounds have shown promise in achieving beneficial metabolic effects.
Collapse
Affiliation(s)
- Lucia Bettedi
- Institute of Healthy Ageing and Department of Genetics, Evolution and Environment, University College London, London, UK
- Cell Biology and Neurobiology Branch, National Institutes of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Lazaros C Foukas
- Institute of Healthy Ageing and Department of Genetics, Evolution and Environment, University College London, London, UK.
| |
Collapse
|
55
|
Saccon TD, Moreira F, Cruz LA, Mondadori RG, Fang Y, Barros CC, Spinel L, Bartke A, Masternak MM, Schneider A. Ovarian aging and the activation of the primordial follicle reserve in the long-lived Ames dwarf and the short-lived bGH transgenic mice. Mol Cell Endocrinol 2017; 455:23-32. [PMID: 27771355 PMCID: PMC5397383 DOI: 10.1016/j.mce.2016.10.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 06/22/2016] [Accepted: 10/15/2016] [Indexed: 12/09/2022]
Abstract
The aim of this study was to evaluate the effect of growth hormone (GH) in the maintenance of the ovarian primordial follicle reserve. Ovaries from 16 mo old GH-deficient Ames Dwarf (df/df) and Normal (N/df) mice were used. A subgroup of df/df and N mice received GH or saline injections for six weeks starting at 14 mo of age. In addition, ovaries from 12 mo old mice overexpressing bovine GH (bGH) and controls were used. df/df mice had higher number of primordial and total follicles than N/df mice (p < 0.05), while GH treatment decreased follicle counts in both genotypes (p < 0.05). In addition, bGH mice had lower number of primordial and total follicles than the controls (p < 0.05). pFoxO3a levels were higher in mice treated with GH and in bGH mice (p < 0.05) when comparing with age match controls. These results indicate that increased circulating GH is associated with a reduced ovarian primordial follicle reserve and increased pFoxO3a content in oocytes.
Collapse
Affiliation(s)
- Tatiana D Saccon
- Faculdade de Nutrição, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Fabiana Moreira
- Campus Araquari, Instituto Federal Catarinense, Araquari, SC, Brazil
| | - Luis A Cruz
- Instituto de Biologia, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Rafael G Mondadori
- Instituto de Biologia, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Yimin Fang
- Departments of Internal Medicine and Physiology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Carlos C Barros
- Faculdade de Nutrição, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - L Spinel
- College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, USA
| | - A Bartke
- Departments of Internal Medicine and Physiology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Michal M Masternak
- College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, USA; Department of Head and Neck Surgery, The Greater Poland Cancer Centre, Poznan, Poland
| | - A Schneider
- Faculdade de Nutrição, Universidade Federal de Pelotas, Pelotas, RS, Brazil; College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, USA.
| |
Collapse
|
56
|
Tyml K, Swarbreck S, Pape C, Secor D, Koropatnick J, Feng Q, Veldhuizen RAW, Gill SE. Voluntary running exercise protects against sepsis-induced early inflammatory and pro-coagulant responses in aged mice. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2017; 21:210. [PMID: 28789683 PMCID: PMC5549433 DOI: 10.1186/s13054-017-1783-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 06/29/2017] [Indexed: 01/10/2023]
Abstract
Background Despite many animal studies and clinical trials, mortality in sepsis remains high. This may be due to the fact that most experimental studies of sepsis employ young animals, whereas the majority of septic patients are elderly (60 − 70 years). The objective of the present study was to examine the sepsis-induced inflammatory and pro-coagulant responses in aged mice. Since running exercise protects against a variety of diseases, we also examined the effect of voluntary running on septic responses in aged mice. Methods Male C57BL/6 mice were housed in our institute from 2–3 to 22 months (an age mimicking that of the elderly). Mice were prevented from becoming obese by food restriction (given 70–90% of ad libitum consumption amount). Between 20 and 22 months, a subgroup of mice ran voluntarily on wheels, alternating 1–3 days of running with 1–2 days of rest. At 22 months, mice were intraperitoneally injected with sterile saline (control) or 3.75 g/kg fecal slurry (septic). At 7 h post injection, we examined (1) neutrophil influx in the lung and liver by measuring myeloperoxidase and/or neutrophil elastase in the tissue homogenates by spectrophotometry, (2) interleukin 6 (IL6) and KC in the lung lavage by ELISA, (3) pulmonary surfactant function by measuring percentage of large aggregates, (4) capillary plugging (pro-coagulant response) in skeletal muscle by intravital microscopy, (5) endothelial nitric oxide synthase (eNOS) protein in skeletal muscle (eNOS-derived NO is putative inhibitor of capillary plugging) by immunoblotting, and (6) systemic blood platelet counts by hemocytometry. Results Sepsis caused high levels of pulmonary myeloperoxidase, elastase, IL6, KC, liver myeloperoxidase, and capillary plugging. Sepsis also caused low levels of surfactant function and platelet counts. Running exercise increased eNOS protein and attenuated the septic responses. Conclusions Voluntary running protects against exacerbated sepsis-induced inflammatory and pro-coagulant responses in aged mice. Protection against pro-coagulant responses may involve eNOS upregulation. The present discovery in aged mice calls for clinical investigation into potential beneficial effects of exercise on septic outcomes in the elderly. Electronic supplementary material The online version of this article (doi:10.1186/s13054-017-1783-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Karel Tyml
- Centre for Critical Illness Research, London, Ontario, Canada.,Department of Medical Biophysics, University of Western Ontario, London, Ontario, Canada
| | - Scott Swarbreck
- Centre for Critical Illness Research, London, Ontario, Canada
| | - Cynthia Pape
- Centre for Critical Illness Research, London, Ontario, Canada.,Department of Medicine, University of Western Ontario, London, Ontario, Canada
| | - Dan Secor
- Centre for Critical Illness Research, London, Ontario, Canada
| | - James Koropatnick
- Cancer Research Program, Lawson Health Research Institute, London, Ontario, Canada.,Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada.,Department of Oncology, University of Western Ontario, London, Ontario, Canada.,Department of Pathology, University of Western Ontario, London, Ontario, Canada
| | - Qingping Feng
- Centre for Critical Illness Research, London, Ontario, Canada.,Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| | - Ruud A W Veldhuizen
- Centre for Critical Illness Research, London, Ontario, Canada.,Division of Respirology, University of Western Ontario, London, Ontario, Canada.,Department of Medicine, University of Western Ontario, London, Ontario, Canada.,Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| | - Sean E Gill
- Centre for Critical Illness Research, London, Ontario, Canada. .,Division of Respirology, University of Western Ontario, London, Ontario, Canada. .,Department of Medicine, University of Western Ontario, London, Ontario, Canada. .,Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada.
| |
Collapse
|
57
|
Gesing A, Wiesenborn D, Do A, Menon V, Schneider A, Victoria B, Stout MB, Kopchick JJ, Bartke A, Masternak MM. A Long-lived Mouse Lacking Both Growth Hormone and Growth Hormone Receptor: A New Animal Model for Aging Studies. J Gerontol A Biol Sci Med Sci 2017; 72:1054-1061. [PMID: 27688483 PMCID: PMC5861925 DOI: 10.1093/gerona/glw193] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 09/13/2016] [Indexed: 11/14/2022] Open
Abstract
Disruption of the growth hormone (GH) signaling pathway promotes insulin sensitivity and is associated with both delayed aging and extended longevity. Two kinds of long-lived mice-Ames dwarfs (df/df) and GH receptor gene-disrupted knockouts (GHRKO) are characterized by a suppressed GH axis with a significant reduction of body size and decreased plasma insulin-like growth factor-1 (IGF-1) and insulin levels. Ames dwarf mice are deficient in GH, prolactin, and thyrotropin, whereas GHRKOs are GH resistant and are dwarf with decreased circulating IGF-1 and increased GH. Crossing Ames dwarfs and GHRKOs produced a new mouse line (df/KO), lacking both GH and GH receptor. These mice are characterized by improved glucose tolerance and increased adiponectin level, which could imply that these mice should be also characterized by additional life-span extension when comparing with GHRKOs and Ames dwarfs. Importantly, our longevity experiments showed that df/KO mice maintain extended longevity when comparing with N control mice; however, they do not live longer than GHRKO and Ames df/df mice. These important findings indicate that silencing GH signal is important to extend the life span; however, further decrease of body size in mice with already inhibited GH signal does not extend the life span regardless of improved some health-span markers.
Collapse
Affiliation(s)
- Adam Gesing
- College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando
- Department of Internal Medicine, Geriatrics Research, Southern Illinois University School of Medicine, Springfield
- Department of Oncological Endocrinology, Medical University of Lodz, Poland
| | - Denise Wiesenborn
- College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando
- Department of Medical Biochemistry and Molecular Biology, University of Saarland, Homburg, Germany
- Department of Biotechnology, University of Applied Sciences Kaiserslautern, Zweibrücken, Germany
| | - Andrew Do
- College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton
| | - Vinal Menon
- College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia
| | - Augusto Schneider
- College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando
- Faculdade de Nutrição, Universidade Federal de Pelotas, Rio Grande do Sul, Brazil
| | - Berta Victoria
- College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando
| | - Michael B Stout
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota
| | - John J Kopchick
- Edison Biotechnology Institute and Heritage College of Osteopathic Medicine, Ohio University, Athens
| | - Andrzej Bartke
- Department of Internal Medicine, Geriatrics Research, Southern Illinois University School of Medicine, Springfield
| | - Michal M Masternak
- College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando
- Department of Head and Neck Surgery, The Greater Poland Cancer Centre, Poznan, Poland
| |
Collapse
|
58
|
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
|
59
|
Hine C, Kim HJ, Zhu Y, Harputlugil E, Longchamp A, Matos MS, Ramadoss P, Bauerle K, Brace L, Asara JM, Ozaki CK, Cheng SY, Singha S, Ahn KH, Kimmelman A, Fisher FM, Pissios P, Withers DJ, Selman C, Wang R, Yen K, Longo VD, Cohen P, Bartke A, Kopchick JJ, Miller R, Hollenberg AN, Mitchell JR. Hypothalamic-Pituitary Axis Regulates Hydrogen Sulfide Production. Cell Metab 2017; 25:1320-1333.e5. [PMID: 28591635 PMCID: PMC5722247 DOI: 10.1016/j.cmet.2017.05.003] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 01/20/2017] [Accepted: 05/11/2017] [Indexed: 01/27/2023]
Abstract
Decreased growth hormone (GH) and thyroid hormone (TH) signaling are associated with longevity and metabolic fitness. The mechanisms underlying these benefits are poorly understood, but may overlap with those of dietary restriction (DR), which imparts similar benefits. Recently we discovered that hydrogen sulfide (H2S) is increased upon DR and plays an essential role in mediating DR benefits across evolutionary boundaries. Here we found increased hepatic H2S production in long-lived mouse strains of reduced GH and/or TH action, and in a cell-autonomous manner upon serum withdrawal in vitro. Negative regulation of hepatic H2S production by GH and TH was additive and occurred via distinct mechanisms, namely direct transcriptional repression of the H2S-producing enzyme cystathionine γ-lyase (CGL) by TH, and substrate-level control of H2S production by GH. Mice lacking CGL failed to downregulate systemic T4 metabolism and circulating IGF-1, revealing an essential role for H2S in the regulation of key longevity-associated hormones.
Collapse
Affiliation(s)
- Christopher Hine
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Hyo-Jeong Kim
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Yan Zhu
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Eylul Harputlugil
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Alban Longchamp
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Department of Surgery, Heart and Vascular Center Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Marina Souza Matos
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Preeti Ramadoss
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Kevin Bauerle
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Lear Brace
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - John M Asara
- Division of Signal Transduction, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - C Keith Ozaki
- Department of Surgery, Heart and Vascular Center Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Sheue-Yann Cheng
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Subhankar Singha
- Department of Chemistry, Center for Electro-Photo Behaviors in Advanced Molecular Systems, POSTECH, 77 Cheongam-Ro, Nam-Gu, Pohang 790-784, Republic of Korea
| | - Kyo Han Ahn
- Department of Chemistry, Center for Electro-Photo Behaviors in Advanced Molecular Systems, POSTECH, 77 Cheongam-Ro, Nam-Gu, Pohang 790-784, Republic of Korea
| | - Alec Kimmelman
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Ffolliott M Fisher
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Pavlos Pissios
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Dominic J Withers
- Medical Research Council Clinical Science Centre, Imperial College, London W12 0NN, UK
| | - Colin Selman
- Glasgow Ageing Research Network, Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Rui Wang
- Department of Biology, Lakehead University, Thunder Bay, ON P7B 5E1, Canada
| | - Kelvin Yen
- Department of Biological Sciences, Longevity Institute, School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Valter D Longo
- Department of Biological Sciences, Longevity Institute, School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Pinchas Cohen
- Department of Biological Sciences, Longevity Institute, School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Andrzej Bartke
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL 62794, USA
| | - John J Kopchick
- Edison Biotechnology Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Richard Miller
- Department of Pathology & Geriatrics Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Anthony N Hollenberg
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | - James R Mitchell
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA.
| |
Collapse
|
60
|
Fujii N, Narita T, Okita N, Kobayashi M, Furuta Y, Chujo Y, Sakai M, Yamada A, Takeda K, Konishi T, Sudo Y, Shimokawa I, Higami Y. Sterol regulatory element-binding protein-1c orchestrates metabolic remodeling of white adipose tissue by caloric restriction. Aging Cell 2017; 16:508-517. [PMID: 28256090 PMCID: PMC5418191 DOI: 10.1111/acel.12576] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/02/2017] [Indexed: 12/31/2022] Open
Abstract
Caloric restriction (CR) can delay onset of several age‐related pathophysiologies and extend lifespan in various species, including rodents. CR also induces metabolic remodeling involved in activation of lipid metabolism, enhancement of mitochondrial biogenesis, and reduction of oxidative stress in white adipose tissue (WAT). In studies using genetically modified mice with extended lifespans, WAT characteristics influenced mammalian lifespans. However, molecular mechanisms underlying CR‐associated metabolic remodeling of WAT remain unclear. Sterol regulatory element‐binding protein‐1c (Srebp‐1c), a master transcription factor of fatty acid (FA) biosynthesis, is responsible for the pathogenesis of fatty liver (steatosis). Our study showed that, under CR conditions, Srebp‐1c enhanced mitochondrial biogenesis via increased expression of peroxisome proliferator‐activated receptor gamma coactivator‐1α (Pgc‐1α) and upregulated expression of proteins involved in FA biosynthesis within WAT. However, via Srebp‐1c, most of these CR‐associated metabolic alterations were not observed in other tissues, including the liver. Moreover, our data indicated that Srebp‐1c may be an important factor both for CR‐associated suppression of oxidative stress, through increased synthesis of glutathione in WAT, and for the prolongevity action of CR. Our results strongly suggested that Srebp‐1c, the primary FA biosynthesis‐promoting transcriptional factor implicated in fatty liver disease, is also the food shortage‐responsive factor in WAT. This indicated that Srebp‐1c is a key regulator of metabolic remodeling leading to the beneficial effects of CR.
Collapse
Affiliation(s)
- Namiki Fujii
- Laboratory of Molecular Pathology and Metabolic Disease; Faculty of Pharmaceutical Sciences; Tokyo University of Science; 2641 Yamazaki Noda, Chiba 278-8510 Japan
| | - Takumi Narita
- Laboratory of Molecular Pathology and Metabolic Disease; Faculty of Pharmaceutical Sciences; Tokyo University of Science; 2641 Yamazaki Noda, Chiba 278-8510 Japan
- Translational Research Center, Research Institute of Science and Technology; Tokyo University of Science; 2641 Yamazaki Noda, Chiba 278-8510 Japan
| | - Naoyuki Okita
- Translational Research Center, Research Institute of Science and Technology; Tokyo University of Science; 2641 Yamazaki Noda, Chiba 278-8510 Japan
- Department of Internal Medicine Research; Sasaki Institute; Sasaki Foundation; 2-2 Kandasurugadai Chiyoda-ku, Tokyo 101-0062 Japan
| | - Masaki Kobayashi
- Laboratory of Molecular Pathology and Metabolic Disease; Faculty of Pharmaceutical Sciences; Tokyo University of Science; 2641 Yamazaki Noda, Chiba 278-8510 Japan
- Translational Research Center, Research Institute of Science and Technology; Tokyo University of Science; 2641 Yamazaki Noda, Chiba 278-8510 Japan
| | - Yurika Furuta
- Laboratory of Molecular Pathology and Metabolic Disease; Faculty of Pharmaceutical Sciences; Tokyo University of Science; 2641 Yamazaki Noda, Chiba 278-8510 Japan
| | - Yoshikazu Chujo
- Laboratory of Molecular Pathology and Metabolic Disease; Faculty of Pharmaceutical Sciences; Tokyo University of Science; 2641 Yamazaki Noda, Chiba 278-8510 Japan
| | - Masahiro Sakai
- Laboratory of Molecular Pathology and Metabolic Disease; Faculty of Pharmaceutical Sciences; Tokyo University of Science; 2641 Yamazaki Noda, Chiba 278-8510 Japan
| | - Atsushi Yamada
- Laboratory of Molecular Pathology and Metabolic Disease; Faculty of Pharmaceutical Sciences; Tokyo University of Science; 2641 Yamazaki Noda, Chiba 278-8510 Japan
| | - Kanae Takeda
- Laboratory of Molecular Pathology and Metabolic Disease; Faculty of Pharmaceutical Sciences; Tokyo University of Science; 2641 Yamazaki Noda, Chiba 278-8510 Japan
| | - Tomokazu Konishi
- Faculty of Bioresource Sciences; Akita Prefectural University; Shimoshinjo Nakano, Akita 010-0195 Japan
| | - Yuka Sudo
- Laboratory of Molecular Pathology and Metabolic Disease; Faculty of Pharmaceutical Sciences; Tokyo University of Science; 2641 Yamazaki Noda, Chiba 278-8510 Japan
- Translational Research Center, Research Institute of Science and Technology; Tokyo University of Science; 2641 Yamazaki Noda, Chiba 278-8510 Japan
| | - Isao Shimokawa
- Translational Research Center, Research Institute of Science and Technology; Tokyo University of Science; 2641 Yamazaki Noda, Chiba 278-8510 Japan
- Department of Pathology; Nagasaki University Graduate School of Biomedical Sciences; 1-12-4 Sakamoto Nagasaki 852-8523 Japan
| | - Yoshikazu Higami
- Laboratory of Molecular Pathology and Metabolic Disease; Faculty of Pharmaceutical Sciences; Tokyo University of Science; 2641 Yamazaki Noda, Chiba 278-8510 Japan
- Translational Research Center, Research Institute of Science and Technology; Tokyo University of Science; 2641 Yamazaki Noda, Chiba 278-8510 Japan
| |
Collapse
|
61
|
Differential effects of early-life nutrient restriction in long-lived GHR-KO and normal mice. GeroScience 2017; 39:347-356. [PMID: 28523599 DOI: 10.1007/s11357-017-9978-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 05/12/2017] [Indexed: 02/07/2023] Open
Abstract
There is increasing evidence that growth hormone (GH) and insulin-like growth factor 1 (IGF-1) signaling (collectively referred to as somatotropic signaling) during development has a profound influence on aging and longevity. Moreover, the absence of GH action was shown to modify responses of adult mice to calorie restriction (CR) and other antiaging interventions. It was therefore of interest to determine whether GH resistance in GH receptor knockout (GHR-KO) mice would modify the effects of mild pre-weaning CR imposed by increasing the number of pups in a litter (the so-called litter crowding). In addition to the expected impact on body weight, litter crowding affected glucose homeostasis, hepatic expression of IGF-1 and genes related to lipid metabolism, and expression of inflammatory markers in white adipose tissue, with some of these effects persisting until the age of 2 years. Litter crowding failed to further extend the remarkable longevity of GHR-KO mice and, instead, reduced late life survival of GHR-KO females, an effect opposite to the changes detected in normal animals. We conclude that the absence of GH actions alters the responses to pre-weaning CR and prevents this intervention from extending longevity.
Collapse
|
62
|
Hjortebjerg R, Berryman DE, Comisford R, Frank SJ, List EO, Bjerre M, Frystyk J, Kopchick JJ. Insulin, IGF-1, and GH Receptors Are Altered in an Adipose Tissue Depot-Specific Manner in Male Mice With Modified GH Action. Endocrinology 2017; 158:1406-1418. [PMID: 28323915 PMCID: PMC5460824 DOI: 10.1210/en.2017-00084] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 02/22/2017] [Indexed: 12/28/2022]
Abstract
Growth hormone (GH) is a determinant of glucose homeostasis and adipose tissue (AT) function. Using 7-month-old transgenic mice expressing the bovine growth hormone (bGH) gene and growth hormone receptor knockout (GHR-/-) mice, we examined whether changes in GH action affect glucose, insulin, and pyruvate tolerance and AT expression of proteins involved in the interrelated signaling pathways of GH, insulinlike growth factor 1 (IGF-1), and insulin. Furthermore, we searched for AT depot-specific differences in control mice. Glycated hemoglobin levels were reduced in bGH and GHR-/- mice, and bGH mice displayed impaired gluconeogenesis as judged by pyruvate tolerance testing. Serum IGF-1 was elevated by 90% in bGH mice, whereas IGF-1 and insulin were reduced by 97% and 61% in GHR-/- mice, respectively. Igf1 RNA was increased in subcutaneous, epididymal, retroperitoneal, and brown adipose tissue (BAT) depots in bGH mice (mean increase ± standard error of the mean in all five depots, 153% ± 27%) and decreased in all depots in GHR-/- mice (mean decrease, 62% ± 4%). IGF-1 receptor expression was decreased in all AT depots of bGH mice (mean decrease, 49% ± 6%) and increased in all AT depots of GHR-/- mice (mean increase, 94% ± 8%). Insulin receptor expression was reduced in retroperitoneal, mesenteric, and BAT depots in bGH mice (mean decrease in all depots, 56% ± 4%) and augmented in subcutaneous, retroperitoneal, mesenteric, and BAT depots in GHR-/- mice (mean increase: 51% ± 1%). Collectively, our findings indicate a role for GH in influencing hormone signaling in AT in a depot-dependent manner.
Collapse
Affiliation(s)
- Rikke Hjortebjerg
- Medical Research Laboratory, Department of Clinical Medicine, Faculty of Health, Aarhus University, 8000 Aarhus, Denmark
- Danish Diabetes Academy, 5000 Odense, Denmark
- Edison Biotechnology Institute, Ohio University, Athens, Ohio 45701
| | - Darlene E. Berryman
- Edison Biotechnology Institute, Ohio University, Athens, Ohio 45701
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio 45701
- The Diabetes Institute at Ohio University, Ohio University, Athens, Ohio 45701
| | - Ross Comisford
- Edison Biotechnology Institute, Ohio University, Athens, Ohio 45701
- The Diabetes Institute at Ohio University, Ohio University, Athens, Ohio 45701
| | - Stuart J. Frank
- Division of Endocrinology, Diabetes and Metabolism, University of Alabama at Birmingham, Birmingham, Alabama 35924
- Medical Service, Birmingham Veterans Affairs Medical Center, Birmingham, Alabama 35233
| | - Edward O. List
- Edison Biotechnology Institute, Ohio University, Athens, Ohio 45701
| | - Mette Bjerre
- Medical Research Laboratory, Department of Clinical Medicine, Faculty of Health, Aarhus University, 8000 Aarhus, Denmark
| | - Jan Frystyk
- Medical Research Laboratory, Department of Clinical Medicine, Faculty of Health, Aarhus University, 8000 Aarhus, Denmark
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, 8000 Aarhus, Denmark
| | - John J. Kopchick
- Edison Biotechnology Institute, Ohio University, Athens, Ohio 45701
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio 45701
| |
Collapse
|
63
|
Koopman JJE, van Heemst D, van Bodegom D, Bonkowski MS, Sun LY, Bartke A. Measuring aging rates of mice subjected to caloric restriction and genetic disruption of growth hormone signaling. Aging (Albany NY) 2017; 8:539-46. [PMID: 26959761 PMCID: PMC4833144 DOI: 10.18632/aging.100919] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Caloric restriction and genetic disruption of growth hormone signaling have been shown to counteract aging in mice. The effects of these interventions on aging are examined through age-dependent survival or through the increase in age-dependent mortality rates on a logarithmic scale fitted to the Gompertz model. However, these methods have limitations that impede a fully comprehensive disclosure of these effects. Here we examine the effects of these interventions on murine aging through the increase in age-dependent mortality rates on a linear scale without fitting them to a model like the Gompertz model. Whereas these interventions negligibly and non-consistently affected the aging rates when examined through the age-dependent mortality rates on a logarithmic scale, they caused the aging rates to increase at higher ages and to higher levels when examined through the age-dependent mortality rates on a linear scale. These results add to the debate whether these interventions postpone or slow aging and to the understanding of the mechanisms by which they affect aging. Since different methods yield different results, it is worthwhile to compare their results in future research to obtain further insights into the effects of dietary, genetic, and other interventions on the aging of mice and other species.
Collapse
Affiliation(s)
- Jacob J E Koopman
- Section of Gerontology and Geriatrics, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands.,Leyden Academy on Vitality and Ageing, Leiden, the Netherlands
| | - Diana van Heemst
- Section of Gerontology and Geriatrics, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands.,Leyden Academy on Vitality and Ageing, Leiden, the Netherlands
| | - David van Bodegom
- Section of Gerontology and Geriatrics, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands.,Leyden Academy on Vitality and Ageing, Leiden, the Netherlands
| | - Michael S Bonkowski
- Division of Geriatric Research, Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL 62794-9628, USA.,Paul F. Glenn Laboratory, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Liou Y Sun
- Division of Geriatric Research, Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL 62794-9628, USA.,Department of Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Andrzej Bartke
- Division of Geriatric Research, Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL 62794-9628, USA
| |
Collapse
|
64
|
Mathew R, Pal Bhadra M, Bhadra U. Insulin/insulin-like growth factor-1 signalling (IIS) based regulation of lifespan across species. Biogerontology 2017; 18:35-53. [DOI: 10.1007/s10522-016-9670-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 11/25/2016] [Indexed: 12/21/2022]
|
65
|
Bartke A, Sun L, Fang Y, Hill C. Growth hormone actions during development influence adult phenotype and longevity. Exp Gerontol 2016; 86:22-27. [PMID: 26752217 PMCID: PMC4930735 DOI: 10.1016/j.exger.2015.12.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 12/21/2015] [Accepted: 12/22/2015] [Indexed: 11/22/2022]
Abstract
There is considerable evidence that exposure to undernutrition, overnutrition, stress or endocrine disruptors during fetal development can increase the probability of obesity, hypertension, cardiovascular disease and other problems in adult life. In contrast to these findings, reducing early postnatal growth by altering maternal diet or number of pups in a litter can increase longevity. In hypopituitary Ames dwarf mice, which are remarkably long lived, a brief period of growth hormone therapy starting at 1 or 2weeks of age reduces longevity and normalizes ("rescues") multiple aging-related traits. Collectively, these findings indicate that nutritional and hormonal signals during development can have profound impact on the trajectory of aging. We suspect that altered "programming" of aging during development may represent one of the mechanisms of the Developmental Origins of Health and Disease (DOHaD) and the detrimental effects of "catch-up" growth.
Collapse
Affiliation(s)
- A Bartke
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL, United States.
| | - L Sun
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Y Fang
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - C Hill
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL, United States
| |
Collapse
|
66
|
Martyniak K, Masternak MM. Changes in adipose tissue cellular composition during obesity and aging as a cause of metabolic dysregulation. Exp Gerontol 2016; 94:59-63. [PMID: 27939445 DOI: 10.1016/j.exger.2016.12.007] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 12/02/2016] [Accepted: 12/07/2016] [Indexed: 12/17/2022]
Abstract
Adipose tissue represents complex endocrine organ containing several different cellular populations including adipocytes, pre-adipocytes, mesenchymal stem cells, macrophages and lymphocytes. It is well establishing that these populations are not static but alter during obesity and aging. Changes in cellular populations alter inflammatory status and other common metabolic complications arise, therefore adipose tissue cellular composition helps dictate its endocrine and regulatory function. During excessive weight gain in obese individuals and as we age there is shift towards increase populations of inflammatory macrophages with a decrease of regulatory T cell. This altered cellular composition promote chronic low grade inflammation negatively affecting mesenchymal stem cell progenitor self-renewal, which result in deterioration of adipogenesis and increased cellular stress in adipocytes. All these changes promote metabolic disorders including age- or obese-related insulin resistance leading to type 2 diabetes.
Collapse
Affiliation(s)
- Kari Martyniak
- College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL 32827, United States
| | - Michal M Masternak
- College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL 32827, United States; Department of Head and Neck Surgery, The Greater Poland Cancer Centre, Poznan, Poland.
| |
Collapse
|
67
|
Lin R, Zhang Y, Yan D, Liao X, Wang X, Fu Y, Cai W. Genetic Association Analysis of Common Variants in FOXO3 Related to Longevity in a Chinese Population. PLoS One 2016; 11:e0167918. [PMID: 27936216 PMCID: PMC5148017 DOI: 10.1371/journal.pone.0167918] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 11/22/2016] [Indexed: 12/14/2022] Open
Abstract
Recent studies suggested that forkhead box class O3 (FOXO3) functions as a key regulator for the insulin/insulin-like growth factor-1signaling pathway that influence aging and longevity. This study aimed to comprehensively elucidate the association of common genetic variants in FOXO3 with human longevity in a Chinese population. Eighteen single-nucleotide polymorphisms (SNPs) in FOXO3 were successfully genotyped in 616 unrelated long-lived individuals and 846 younger controls. No nominally significant effects were found. However, when stratifying by gender, four SNPs (rs10499051, rs7762395, rs4946933 and rs3800230) previously reported to be associated with longevity and one novel SNP (rs4945815) showed significant association with male longevity (P-values: 0.007–0.032), but all SNPs were not associated with female longevity. Correspondingly, males carrying the G-G-T-G haplotype of rs10499051, rs7762395, rs4945815 and rs3800230 tended to have longer lifespan than those carrying the most common haplotype A-G-C-T (odds ratio = 2.36, 95% confidence interval = 1.20–4.63, P = 0.013). However, none of the associated SNPs and haplotype remained significant after Bonferroni correction. In conclusion, our findings revealed that the FOXO3 variants we tested in our population of Chinese men and women were associated with longevity in men only. None of these associations passed Bonferroni correction. Bonferroni correction is very stringent for association studies. We therefore believe the effects of these nominally significant variants on human longevity will be confirmed by future studies.
Collapse
Affiliation(s)
- Rong Lin
- Department of Biology, Hainan Medical College, Haikou, Hainan, China
- * E-mail: (RL); (WC)
| | - Yunxia Zhang
- Department of Biochemistry and Molecular Biology, Hainan Medical College, Haikou, Hainan, China
| | - Dongjing Yan
- Department of Biochemistry and Molecular Biology, Hainan Medical College, Haikou, Hainan, China
| | - Xiaoping Liao
- Department of Neurology, the First Affiliated Hospital of Hainan Medical College, Haikou, Hainan, China
| | - Xianshou Wang
- Specialized Biotechnologies Lab, Hainan Medical College, Haikou, Hainan, China
| | - Yunxin Fu
- Division of Biostatistics and Human Genetics Center, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
- Laboratory for Conservation and Utilization of Bio-Resources, Yunnan University, Kunming, Yunnan, China
| | - Wangwei Cai
- Department of Biochemistry and Molecular Biology, Hainan Medical College, Haikou, Hainan, China
- * E-mail: (RL); (WC)
| |
Collapse
|
68
|
Stout MB, Swindell WR, Zhi X, Rohde K, List EO, Berryman DE, Kopchick JJ, Gesing A, Fang Y, Masternak MM. Transcriptome profiling reveals divergent expression shifts in brown and white adipose tissue from long-lived GHRKO mice. Oncotarget 2016; 6:26702-15. [PMID: 26436954 PMCID: PMC4694946 DOI: 10.18632/oncotarget.5760] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Accepted: 08/29/2015] [Indexed: 12/24/2022] Open
Abstract
Mice lacking the growth hormone receptor (GHRKO) exhibit improved lifespan and healthspan due to loss of growth hormone signaling. Both the distribution and activity of brown and white adipose tissue (BAT and WAT) are altered in GHRKO mice, but the contribution of each tissue to age-related phenotypes has remained unclear. We therefore used whole-genome microarrays to evaluate transcriptional differences in BAT and WAT depots between GHRKO and normal littermates at six months of age. Our findings reveal a unique BAT transcriptome as well as distinctive responses of BAT to Ghr ablation. BAT from GHRKO mice exhibited elevated expression of genes associated with mitochondria and metabolism, along with reduced expression of genes expressed by monocyte-derived cells (dendritic cells [DC] and macrophages). Largely the opposite was observed in WAT, with increased expression of DC-expressed genes and reduced expression of genes associated with metabolism, cellular respiration and the mitochondrial inner envelope. These findings demonstrate divergent response patterns of BAT and WAT to loss of GH signaling in GHRKO mice. These patterns suggest both BAT and WAT contribute in different ways to phenotypes in GHRKO mice, with Ghr ablation blunting inflammation in BAT as well as cellular metabolism and mitochondrial biogenesis in WAT.
Collapse
Affiliation(s)
- Michael B Stout
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | | | - Xu Zhi
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.,College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, USA
| | - Kyle Rohde
- College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, USA
| | - Edward O List
- Edison Biotechnology Institute and Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Darlene E Berryman
- Edison Biotechnology Institute and Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - John J Kopchick
- Edison Biotechnology Institute and Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Adam Gesing
- Department of Oncological Endocrinology, Medical University of Lodz, Lodz, Poland
| | - Yimin Fang
- Geriatrics Research Laboratory, Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Michal M Masternak
- College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, USA.,Department of Head and Neck Surgery, The Greater Poland Cancer Centre, Poznan, Poland
| |
Collapse
|
69
|
Schultz MB, Sinclair DA. When stem cells grow old: phenotypes and mechanisms of stem cell aging. Development 2016; 143:3-14. [PMID: 26732838 DOI: 10.1242/dev.130633] [Citation(s) in RCA: 214] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
All multicellular organisms undergo a decline in tissue and organ function as they age. An attractive theory is that a loss in stem cell number and/or activity over time causes this decline. In accordance with this theory, aging phenotypes have been described for stem cells of multiple tissues, including those of the hematopoietic system, intestine, muscle, brain, skin and germline. Here, we discuss recent advances in our understanding of why adult stem cells age and how this aging impacts diseases and lifespan. With this increased understanding, it is feasible to design and test interventions that delay stem cell aging and improve both health and lifespan.
Collapse
Affiliation(s)
- Michael B Schultz
- Paul F. Glenn Center for the Biological Mechanisms of Aging, Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - David A Sinclair
- Paul F. Glenn Center for the Biological Mechanisms of Aging, Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
70
|
Abstract
Sex differences in longevity can provide insights into novel mechanisms of aging, yet they have been little studied. Surprisingly, sex-specific longevity patterns are best known in wild animals. Evolutionary hypotheses accounting for longevity patterns in natural populations include differential vulnerability to environmental hazards, differential intensity of sexual selection, and distinct patterns of parental care. Mechanistic hypotheses focus on hormones, asymmetric inheritance of sex chromosomes and mitochondria. Virtually all intensively studied species show conditional sex differences in longevity. Humans are the only species in which one sex is known to have a ubiquitous survival advantage. Paradoxically, although women live longer, they suffer greater morbidity particularly late in life. This mortality-morbidity paradox may be a consequence of greater connective tissue responsiveness to sex hormones in women. Human females' longevity advantage may result from hormonal influences on inflammatory and immunological responses, or greater resistance to oxidative damage; current support for these mechanisms is weak.
Collapse
Affiliation(s)
- Steven N Austad
- Department of Biology and Nathan Shock Center of Excellence in the Basic Biology of Aging, University of Alabama at Birmingham, Birmingham, AL 35294-1170, USA.
| | - Kathleen E Fischer
- Department of Biology and Nathan Shock Center of Excellence in the Basic Biology of Aging, University of Alabama at Birmingham, Birmingham, AL 35294-1170, USA
| |
Collapse
|
71
|
Jara A, Liu X, Sim D, Benner CM, Duran-Ortiz S, Qian Y, List EO, Berryman DE, Kim JK, Kopchick JJ. Cardiac-Specific Disruption of GH Receptor Alters Glucose Homeostasis While Maintaining Normal Cardiac Performance in Adult Male Mice. Endocrinology 2016; 157:1929-41. [PMID: 27035649 PMCID: PMC4870885 DOI: 10.1210/en.2015-1686] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
GH is considered necessary for the proper development and maintenance of several tissues, including the heart. Studies conducted in both GH receptor null and bovine GH transgenic mice have demonstrated specific cardiac structural and functional changes. In each of these mouse lines, however, GH-induced signaling is altered systemically, being decreased in GH receptor null mice and increased in bovine GH transgenic mice. Therefore, to clarify the direct effects GH has on cardiac tissue, we developed a tamoxifen-inducible, cardiac-specific GHR disrupted (iC-GHRKO) mouse line. Cardiac GH receptor was disrupted in 4-month-old iC-GHRKO mice to avoid developmental effects due to perinatal GHR gene disruption. Surprisingly, iC-GHRKO mice showed no difference vs controls in baseline or postdobutamine stress test echocardiography measurements, nor did iC-GHRKO mice show differences in longitudinal systolic blood pressure measurements. Interestingly, iC-GHRKO mice had decreased fat mass and improved insulin sensitivity at 6.5 months of age. By 12.5 months of age, however, iC-GHRKO mice no longer had significant decreases in fat mass and had developed glucose intolerance and insulin resistance. Furthermore, investigation via immunoblot analysis demonstrated that iC-GHRKO mice had appreciably decreased insulin stimulated Akt phosphorylation, specifically in heart and liver, but not in epididymal white adipose tissue. These changes were accompanied by a decrease in circulating IGF-1 levels in 12.5-month-old iC-GHRKO mice. These data indicate that whereas the disruption of cardiomyocyte GH-induced signaling in adult mice does not affect cardiac function, it does play a role in systemic glucose homeostasis, in part through modulation of circulating IGF-1.
Collapse
Affiliation(s)
- Adam Jara
- Edison Biotechnology Institute (A.J., X.L., D.S., C.M.B., S.D.-O., Y.Q., E.O.L., D.E.B., J.J.K.), Departments of Biomedical Sciences (A.J., D.E.B., J.J.K.) and Specialty Medicine (E.O.L.), Heritage College of Osteopathic Medicine, Department of Biological Sciences (S.D.-O., J.J.K.), School of Applied Health Sciences and Wellness (X.L., C.M.B., D.E.B.), College of Health Sciences and Professions, Department of Social and Public Health (D.S.), Ohio University, Athens, Ohio 45701; and Program in Molecular Medicine (J.K.K.), University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Xingbo Liu
- Edison Biotechnology Institute (A.J., X.L., D.S., C.M.B., S.D.-O., Y.Q., E.O.L., D.E.B., J.J.K.), Departments of Biomedical Sciences (A.J., D.E.B., J.J.K.) and Specialty Medicine (E.O.L.), Heritage College of Osteopathic Medicine, Department of Biological Sciences (S.D.-O., J.J.K.), School of Applied Health Sciences and Wellness (X.L., C.M.B., D.E.B.), College of Health Sciences and Professions, Department of Social and Public Health (D.S.), Ohio University, Athens, Ohio 45701; and Program in Molecular Medicine (J.K.K.), University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Don Sim
- Edison Biotechnology Institute (A.J., X.L., D.S., C.M.B., S.D.-O., Y.Q., E.O.L., D.E.B., J.J.K.), Departments of Biomedical Sciences (A.J., D.E.B., J.J.K.) and Specialty Medicine (E.O.L.), Heritage College of Osteopathic Medicine, Department of Biological Sciences (S.D.-O., J.J.K.), School of Applied Health Sciences and Wellness (X.L., C.M.B., D.E.B.), College of Health Sciences and Professions, Department of Social and Public Health (D.S.), Ohio University, Athens, Ohio 45701; and Program in Molecular Medicine (J.K.K.), University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Chance M Benner
- Edison Biotechnology Institute (A.J., X.L., D.S., C.M.B., S.D.-O., Y.Q., E.O.L., D.E.B., J.J.K.), Departments of Biomedical Sciences (A.J., D.E.B., J.J.K.) and Specialty Medicine (E.O.L.), Heritage College of Osteopathic Medicine, Department of Biological Sciences (S.D.-O., J.J.K.), School of Applied Health Sciences and Wellness (X.L., C.M.B., D.E.B.), College of Health Sciences and Professions, Department of Social and Public Health (D.S.), Ohio University, Athens, Ohio 45701; and Program in Molecular Medicine (J.K.K.), University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Silvana Duran-Ortiz
- Edison Biotechnology Institute (A.J., X.L., D.S., C.M.B., S.D.-O., Y.Q., E.O.L., D.E.B., J.J.K.), Departments of Biomedical Sciences (A.J., D.E.B., J.J.K.) and Specialty Medicine (E.O.L.), Heritage College of Osteopathic Medicine, Department of Biological Sciences (S.D.-O., J.J.K.), School of Applied Health Sciences and Wellness (X.L., C.M.B., D.E.B.), College of Health Sciences and Professions, Department of Social and Public Health (D.S.), Ohio University, Athens, Ohio 45701; and Program in Molecular Medicine (J.K.K.), University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Yanrong Qian
- Edison Biotechnology Institute (A.J., X.L., D.S., C.M.B., S.D.-O., Y.Q., E.O.L., D.E.B., J.J.K.), Departments of Biomedical Sciences (A.J., D.E.B., J.J.K.) and Specialty Medicine (E.O.L.), Heritage College of Osteopathic Medicine, Department of Biological Sciences (S.D.-O., J.J.K.), School of Applied Health Sciences and Wellness (X.L., C.M.B., D.E.B.), College of Health Sciences and Professions, Department of Social and Public Health (D.S.), Ohio University, Athens, Ohio 45701; and Program in Molecular Medicine (J.K.K.), University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Edward O List
- Edison Biotechnology Institute (A.J., X.L., D.S., C.M.B., S.D.-O., Y.Q., E.O.L., D.E.B., J.J.K.), Departments of Biomedical Sciences (A.J., D.E.B., J.J.K.) and Specialty Medicine (E.O.L.), Heritage College of Osteopathic Medicine, Department of Biological Sciences (S.D.-O., J.J.K.), School of Applied Health Sciences and Wellness (X.L., C.M.B., D.E.B.), College of Health Sciences and Professions, Department of Social and Public Health (D.S.), Ohio University, Athens, Ohio 45701; and Program in Molecular Medicine (J.K.K.), University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Darlene E Berryman
- Edison Biotechnology Institute (A.J., X.L., D.S., C.M.B., S.D.-O., Y.Q., E.O.L., D.E.B., J.J.K.), Departments of Biomedical Sciences (A.J., D.E.B., J.J.K.) and Specialty Medicine (E.O.L.), Heritage College of Osteopathic Medicine, Department of Biological Sciences (S.D.-O., J.J.K.), School of Applied Health Sciences and Wellness (X.L., C.M.B., D.E.B.), College of Health Sciences and Professions, Department of Social and Public Health (D.S.), Ohio University, Athens, Ohio 45701; and Program in Molecular Medicine (J.K.K.), University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Jason K Kim
- Edison Biotechnology Institute (A.J., X.L., D.S., C.M.B., S.D.-O., Y.Q., E.O.L., D.E.B., J.J.K.), Departments of Biomedical Sciences (A.J., D.E.B., J.J.K.) and Specialty Medicine (E.O.L.), Heritage College of Osteopathic Medicine, Department of Biological Sciences (S.D.-O., J.J.K.), School of Applied Health Sciences and Wellness (X.L., C.M.B., D.E.B.), College of Health Sciences and Professions, Department of Social and Public Health (D.S.), Ohio University, Athens, Ohio 45701; and Program in Molecular Medicine (J.K.K.), University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - John J Kopchick
- Edison Biotechnology Institute (A.J., X.L., D.S., C.M.B., S.D.-O., Y.Q., E.O.L., D.E.B., J.J.K.), Departments of Biomedical Sciences (A.J., D.E.B., J.J.K.) and Specialty Medicine (E.O.L.), Heritage College of Osteopathic Medicine, Department of Biological Sciences (S.D.-O., J.J.K.), School of Applied Health Sciences and Wellness (X.L., C.M.B., D.E.B.), College of Health Sciences and Professions, Department of Social and Public Health (D.S.), Ohio University, Athens, Ohio 45701; and Program in Molecular Medicine (J.K.K.), University of Massachusetts Medical School, Worcester, Massachusetts 01605
| |
Collapse
|
72
|
Abstract
Caloric restriction is the most effective and reproducible dietary intervention known to regulate aging and increase the healthy lifespan in various model organisms, ranging from the unicellular yeast to worms, flies, rodents, and primates. However, caloric restriction, which in most cases entails a 20–40% reduction of food consumption relative to normal intake, is a severe intervention that results in both beneficial and detrimental effects. Specific types of chronic, intermittent, or periodic dietary restrictions without chronic caloric restriction have instead the potential to provide a significant healthspan increase while minimizing adverse effects. Improved periodic or targeted dietary restriction regimens that uncouple the challenge of food deprivation from the beneficial effects will allow a safe intervention feasible for a major portion of the population. Here we focus on healthspan interventions that are not chronic or do not require calorie restriction.
Collapse
Affiliation(s)
- Changhan Lee
- Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Valter Longo
- Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA; IFOM - FIRC Institute of Molecular Oncology Foundation, Milan, Italy
| |
Collapse
|
73
|
Mitchell SJ, Scheibye-Knudsen M, Longo DL, de Cabo R. Animal models of aging research: implications for human aging and age-related diseases. Annu Rev Anim Biosci 2016; 3:283-303. [PMID: 25689319 DOI: 10.1146/annurev-animal-022114-110829] [Citation(s) in RCA: 218] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Aging is characterized by an increasing morbidity and functional decline that eventually results in the death of an organism. Aging is the largest risk factor for numerous human diseases, and understanding the aging process may thereby facilitate the development of new treatments for age-associated diseases. The use of humans in aging research is complicated by many factors, including ethical issues; environmental and social factors; and perhaps most importantly, their long natural life span. Although cellular models of human disease provide valuable mechanistic information, they are limited in that they may not replicate the in vivo biology. Almost all organisms age, and thus animal models can be useful for studying aging. Herein, we review some of the major models currently used in aging research and discuss their benefits and pitfalls, including interventions known to extend life span and health span. Finally, we conclude by discussing the future of animal models in aging research.
Collapse
|
74
|
Bitto A, Wang AM, Bennett CF, Kaeberlein M. Biochemical Genetic Pathways that Modulate Aging in Multiple Species. Cold Spring Harb Perspect Med 2015; 5:5/11/a025114. [PMID: 26525455 DOI: 10.1101/cshperspect.a025114] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The mechanisms underlying biological aging have been extensively studied in the past 20 years with the avail of mainly four model organisms: the budding yeast Saccharomyces cerevisiae, the nematode Caenorhabditis elegans, the fruitfly Drosophila melanogaster, and the domestic mouse Mus musculus. Extensive research in these four model organisms has identified a few conserved genetic pathways that affect longevity as well as metabolism and development. Here, we review how the mechanistic target of rapamycin (mTOR), sirtuins, adenosine monophosphate-activated protein kinase (AMPK), growth hormone/insulin-like growth factor 1 (IGF-1), and mitochondrial stress-signaling pathways influence aging and life span in the aforementioned models and their possible implications for delaying aging in humans. We also draw some connections between these biochemical pathways and comment on what new developments aging research will likely bring in the near future.
Collapse
Affiliation(s)
- Alessandro Bitto
- Department of Pathology, University of Washington, Seattle, Washington 98195
| | - Adrienne M Wang
- Department of Pathology, University of Washington, Seattle, Washington 98195
| | | | - Matt Kaeberlein
- Department of Pathology, University of Washington, Seattle, Washington 98195
| |
Collapse
|
75
|
Abstract
The somatotropic signaling pathway has been implicated in aging and longevity studies in mice and other species. The physiology and lifespans of a variety of mutant mice, both spontaneous and genetically engineered, have contributed to our current understanding of the role of growth hormone and insulin-like growth factor I on aging-related processes. Several other mice discovered to live longer than their wild-type control counterparts also exhibit differences in growth factor levels; however, the complex nature of the phenotypic changes in these animals may also impact lifespan. The somatotropic axis impacts several pathways that dictate insulin sensitivity, nutrient sensing, mitochondrial function, and stress resistance as well as others that are thought to be involved in lifespan regulation.
Collapse
Affiliation(s)
- H M Brown-Borg
- Department of Biomedical Sciences, University of North Dakota, School of Medicine and Health Sciences, Grand Forks, North Dakota
| |
Collapse
|
76
|
Huang CW, Wang HD, Bai H, Wu MS, Yen JH, Tatar M, Fu TF, Wang PY. Tequila Regulates Insulin-Like Signaling and Extends Life Span in Drosophila melanogaster. J Gerontol A Biol Sci Med Sci 2015; 70:1461-9. [PMID: 26265729 DOI: 10.1093/gerona/glv094] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 05/30/2015] [Indexed: 11/13/2022] Open
Abstract
The aging process is a universal phenomenon shared by all living organisms. The identification of longevity genes is important in that the study of these genes is likely to yield significant insights into human senescence. In this study, we have identified Tequila as a novel candidate gene involved in the regulation of longevity in Drosophila melanogaster. We have found that a hypomorphic mutation of Tequila (Teq(f01792)), as well as cell-specific downregulation of Tequila in insulin-producing neurons of the fly, significantly extends life span. Tequila deficiency-induced life-span extension is likely to be associated with reduced insulin-like signaling, because Tequila mutant flies display several common phenotypes of insulin dysregulation, including reduced circulating Drosophila insulin-like peptide 2 (Dilp2), reduced Akt phosphorylation, reduced body size, and altered glucose homeostasis. These observations suggest that Tequila may confer life-span extension by acting as a modulator of Drosophila insulin-like signaling.
Collapse
Affiliation(s)
- Cheng-Wen Huang
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, Taiwan. Institute of Biotechnology
| | - Horng-Dar Wang
- Institute of Biotechnology, Institute of Systems Neuroscience, and Department of Life Science, National Tsing Hua University, HsinChu, Taiwan
| | - Hua Bai
- Department of Ecology and Evolutionary Biology, Brown University, Providence, Rhode Island
| | - Ming-Shiang Wu
- Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University, Taipei, Taiwan
| | - Jui-Hung Yen
- Department of Microbiology and Immunology, Indiana University School of Medicine, Fort Wayne, Indiana
| | - Marc Tatar
- Department of Ecology and Evolutionary Biology, Brown University, Providence, Rhode Island
| | - Tsai-Feng Fu
- Department of Applied Chemistry, National Chinan University, Nantou, Taiwan
| | - Pei-Yu Wang
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
77
|
Castillo-Quan JI, Kinghorn KJ, Bjedov I. Genetics and pharmacology of longevity: the road to therapeutics for healthy aging. ADVANCES IN GENETICS 2015; 90:1-101. [PMID: 26296933 DOI: 10.1016/bs.adgen.2015.06.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Aging can be defined as the progressive decline in tissue and organismal function and the ability to respond to stress that occurs in association with homeostatic failure and the accumulation of molecular damage. Aging is the biggest risk factor for human disease and results in a wide range of aging pathologies. Although we do not completely understand the underlying molecular basis that drives the aging process, we have gained exceptional insights into the plasticity of life span and healthspan from the use of model organisms such as the worm Caenorhabditis elegans and the fruit fly Drosophila melanogaster. Single-gene mutations in key cellular pathways that regulate environmental sensing, and the response to stress, have been identified that prolong life span across evolution from yeast to mammals. These genetic manipulations also correlate with a delay in the onset of tissue and organismal dysfunction. While the molecular genetics of aging will remain a prosperous and attractive area of research in biogerontology, we are moving towards an era defined by the search for therapeutic drugs that promote healthy aging. Translational biogerontology will require incorporation of both therapeutic and pharmacological concepts. The use of model organisms will remain central to the quest for drug discovery, but as we uncover molecular processes regulated by repurposed drugs and polypharmacy, studies of pharmacodynamics and pharmacokinetics, drug-drug interactions, drug toxicity, and therapeutic index will slowly become more prevalent in aging research. As we move from genetics to pharmacology and therapeutics, studies will not only require demonstration of life span extension and an underlying molecular mechanism, but also the translational relevance for human health and disease prevention.
Collapse
Affiliation(s)
- Jorge Iván Castillo-Quan
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, UK; Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, London, UK
| | - Kerri J Kinghorn
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, UK; Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, London, UK
| | - Ivana Bjedov
- Cancer Institute, University College London, London, UK
| |
Collapse
|
78
|
Sell C. Minireview: The Complexities of IGF/Insulin Signaling in Aging: Why Flies and Worms Are Not Humans. Mol Endocrinol 2015; 29:1107-13. [PMID: 26102060 DOI: 10.1210/me.2015-1074] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
A remarkable plasticity in life span has been uncovered in recent years, offering hope that the basic mechanisms of aging and interventions that delay aging may be identified in the coming decades. Life span extension has been achieved by genetic manipulation in multiple organisms including Sarcomyces cervisae, Caenorhabditis elegans, and Drosophila melanogaster, resulting in more than a doubling of life span in some cases. Typically, a reduction in function has been the most effective approach to extending life span, and a reduction in the insulin/IGF-1 signaling pathway appears to provide the most robust increase in life span. This highly conserved pathway integrates growth/survival signals with nutrient status. In mammals, it comprises part of the neuroendocrine axis, a critical regulator of growth and development. Reduced functionality of the neuroendocrine axis itself promotes life span extension in mammals; however, reduced activity of the IGF-1 signaling pathway specifically leads to less robust increases in life span. This review examines the differences in the insulin/IGF-1 axis between invertebrate and mammalian systems and discusses implications of these differences in terms of life span modulation.
Collapse
Affiliation(s)
- Christian Sell
- Drexel University College of Medicine, Philadelphia, Pennsylvania 19102
| |
Collapse
|
79
|
Ruetenik A, Barrientos A. Dietary restriction, mitochondrial function and aging: from yeast to humans. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2015; 1847:1434-47. [PMID: 25979234 DOI: 10.1016/j.bbabio.2015.05.005] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 04/30/2015] [Accepted: 05/05/2015] [Indexed: 12/20/2022]
Abstract
Dietary restriction (DR) attenuates many detrimental effects of aging and consequently promotes health and increases longevity across organisms. While over the last 15 years extensive research has been devoted towards understanding the biology of aging, the precise mechanistic aspects of DR are yet to be settled. Abundant experimental evidence indicates that the DR effect on stimulating health impinges several metabolic and stress-resistance pathways. Downstream effects of these pathways include a reduction in cellular damage induced by oxidative stress, enhanced efficiency of mitochondrial functions and maintenance of mitochondrial dynamics and quality control, thereby attenuating age-related declines in mitochondrial function. However, the literature also accumulates conflicting evidence regarding how DR ameliorates mitochondrial performance and whether that is enough to slow age-dependent cellular and organismal deterioration. Here, we will summarize the current knowledge about how and to which extent the influence of different DR regimes on mitochondrial biogenesis and function contribute to postpone the detrimental effects of aging on health-span and lifespan. This article is part of a Special Issue entitled: Mitochondrial Dysfunction in Aging.
Collapse
Affiliation(s)
| | - Antoni Barrientos
- Neuroscience Graduate Program; Department of Neurology, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
80
|
de Cavanagh EMV, Inserra F, Ferder L. Angiotensin II blockade: how its molecular targets may signal to mitochondria and slow aging. Coincidences with calorie restriction and mTOR inhibition. Am J Physiol Heart Circ Physiol 2015; 309:H15-44. [PMID: 25934099 DOI: 10.1152/ajpheart.00459.2014] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 04/30/2015] [Indexed: 02/07/2023]
Abstract
Caloric restriction (CR), renin angiotensin system blockade (RAS-bl), and rapamycin-mediated mechanistic target of rapamycin (mTOR) inhibition increase survival and retard aging across species. Previously, we have summarized CR and RAS-bl's converging effects, and the mitochondrial function changes associated with their physiological benefits. mTOR inhibition and enhanced sirtuin and KLOTHO signaling contribute to the benefits of CR in aging. mTORC1/mTORC2 complexes contribute to cell growth and metabolic regulation. Prolonged mTORC1 activation may lead to age-related disease progression; thus, rapamycin-mediated mTOR inhibition and CR may extend lifespan and retard aging through mTORC1 interference. Sirtuins by deacetylating histone and transcription-related proteins modulate signaling and survival pathways and mitochondrial functioning. CR regulates several mammalian sirtuins favoring their role in aging regulation. KLOTHO/fibroblast growth factor 23 (FGF23) contribute to control Ca(2+), phosphate, and vitamin D metabolism, and their dysregulation may participate in age-related disease. Here we review how mTOR inhibition extends lifespan, how KLOTHO functions as an aging suppressor, how sirtuins mediate longevity, how vitamin D loss may contribute to age-related disease, and how they relate to mitochondrial function. Also, we discuss how RAS-bl downregulates mTOR and upregulates KLOTHO, sirtuin, and vitamin D receptor expression, suggesting that at least some of RAS-bl benefits in aging are mediated through the modulation of mTOR, KLOTHO, and sirtuin expression and vitamin D signaling, paralleling CR actions in age retardation. Concluding, the available evidence endorses the idea that RAS-bl is among the interventions that may turn out to provide relief to the spreading issue of age-associated chronic disease.
Collapse
Affiliation(s)
- Elena M V de Cavanagh
- Center of Hypertension, Cardiology Department, Austral University Hospital, Derqui, Argentina; School of Biomedical Sciences, Austral University, Buenos Aires, Argentina; and
| | - Felipe Inserra
- Center of Hypertension, Cardiology Department, Austral University Hospital, Derqui, Argentina; School of Biomedical Sciences, Austral University, Buenos Aires, Argentina; and
| | - León Ferder
- Department of Physiology and Pharmacology, Ponce School of Medicine, Ponce, Puerto Rico
| |
Collapse
|
81
|
Jain S, Singh SN. Effect of L-carnitine Supplementation on Nutritional Status and Physical Performance Under Calorie Restriction. Indian J Clin Biochem 2015; 30:187-93. [PMID: 25883427 DOI: 10.1007/s12291-014-0437-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 04/15/2014] [Indexed: 12/24/2022]
Abstract
L-carnitine is popular as a potential ergogenic aid because of its role in the conversion of fat into energy. The present study was undertaken to investigate the effect of short term supplementation of L-carnitine on metabolic markers and physical efficiency tests under short term calorie restriction. Male albino rats were divided into four groups (n = 12 in each)-control, calorie restricted (CR for 5 days, 25 % of basal food intake), L-carnitine supplemented (CAR, given orally for 5 days at a dose of 100 mg/kg), CR with L-carnitine supplementation (CR + CAR). Food intake and body weight of the rats were measured along with biochemical variables like blood glucose, tissue glycogen, plasma and muscle protein and enzymatic activities of CPT-1 (carnitine palmitoyl transferase-1) and AMP kinase. Results demonstrated that L-carnitine caused marked increase in muscle glycogen, plasma protein, CPT-1 activity and swim time of rats (P < 0.05) on short term supplementation. In addition to the substantive effects caused by CR alone, L-carnitine under CR significantly affected muscle glycogen, plasma protein, CPT-1 activity and AMP kinase (P < 0.05). Short term CR along with L-carnitine also resulted in increased swim time of rats than control, CR and L-carnitine treated rats (P < 0.05). The present study was an attempt towards developing an approach for better adherence to dietary restriction regimen, with the use of L-carnitine.
Collapse
Affiliation(s)
- Swati Jain
- Nutrition Division, Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur, Delhi, 110054 India
| | - Som Nath Singh
- Nutrition Division, Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur, Delhi, 110054 India
| |
Collapse
|
82
|
Dominick G, Berryman DE, List EO, Kopchick JJ, Li X, Miller RA, Garcia GG. Regulation of mTOR activity in Snell dwarf and GH receptor gene-disrupted mice. Endocrinology 2015; 156:565-75. [PMID: 25456069 PMCID: PMC4298324 DOI: 10.1210/en.2014-1690] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The involvement of mammalian target of rapamycin (mTOR) in lifespan control in invertebrates, calorie-restricted rodents, and extension of mouse lifespan by rapamycin have prompted speculation that diminished mTOR function may contribute to mammalian longevity in several settings. We show here that mTOR complex-1 (mTORC1) activity is indeed lower in liver, muscle, heart, and kidney tissue of Snell dwarf and global GH receptor (GHR) gene-disrupted mice (GHR-/-), consistent with previous studies. Surprisingly, activity of mTORC2 is higher in fasted Snell and GHR-/- than in littermate controls in all 4 tissues tested. Resupply of food enhanced mTORC1 activity in both controls and long-lived mutant mice but diminished mTORC2 activity only in the long-lived mice. Mice in which GHR has been disrupted only in the liver do not show extended lifespan and also fail to show the decline in mTORC1 and increase in mTORC2 seen in mice with global loss of GHR. The data suggest that the antiaging effects in the Snell dwarf and GHR-/- mice are accompanied by both a decline in mTORC1 in multiple organs and an increase in fasting levels of mTORC2. Neither the lifespan nor mTOR effects appear to be mediated by direct GH effects on liver or by the decline in plasma IGF-I, a shared trait in both global and liver-specific GHR-/- mice. Our data suggest that a more complex pattern of hormonal effects and intertissue interactions may be responsible for regulating both lifespan and mTORC2 function in these mouse models of delayed aging.
Collapse
Affiliation(s)
- Graham Dominick
- Department of Molecular, Cellular, and Developmental Biology (G.D.), University of Michigan College of Literature, Science and the Arts, Ann Arbor, Michigan 48109; Edison Biotechnology Institute (D.E.B., E.O.L., J.J.K.), Ohio University, Athens, Ohio 45701; Department of Pathology (X.L., R.A.M., G.G.G.), University of Michigan School of Medicine Ann Arbor, Michigan 48109; and University of Michigan Geriatrics Center (R.A.M.), Ann Arbor, Michigan 48109
| | | | | | | | | | | | | |
Collapse
|
83
|
Lamming DW. Diminished mTOR signaling: a common mode of action for endocrine longevity factors. SPRINGERPLUS 2014; 3:735. [PMID: 25674466 PMCID: PMC4320218 DOI: 10.1186/2193-1801-3-735] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 12/07/2014] [Indexed: 12/12/2022]
Abstract
Since the initial observation that a calorie-restricted (CR) diet can extend rodent lifespan, many genetic and pharmaceutical interventions that also extend lifespan in mammals have been discovered. The mechanism by which CR and these other interventions extend lifespan is the subject of significant debate and research. One proposed mechanism is that CR promotes longevity by increasing insulin sensitivity, but recent findings that dissociate longevity and insulin sensitivity cast doubt on this hypothesis. These findings can be reconciled if longevity is promoted not via increased insulin sensitivity, but instead via decreased PI3K/Akt/mTOR pathway signaling. This review presents a unifying hypothesis that explains the lifespan-extending effects of a variety of genetic mutations and pharmaceutical interventions and points towards new molecular pathways which may also be leveraged to promote healthy aging.
Collapse
Affiliation(s)
- Dudley W Lamming
- Division of Endocrinology, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin USA ; William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin USA
| |
Collapse
|
84
|
Arum O, Boparai RK, Saleh JK, Wang F, Dirks AL, Turner JG, Kopchick JJ, Liu J, Khardori RK, Bartke A. Specific suppression of insulin sensitivity in growth hormone receptor gene-disrupted (GHR-KO) mice attenuates phenotypic features of slow aging. Aging Cell 2014; 13:981-1000. [PMID: 25244225 PMCID: PMC4326932 DOI: 10.1111/acel.12262] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2014] [Indexed: 12/20/2022] Open
Abstract
In addition to their extended lifespans, slow-aging growth hormone receptor/binding protein gene-disrupted (knockout) (GHR-KO) mice are hypoinsulinemic and highly sensitive to the action of insulin. It has been proposed that this insulin sensitivity is important for their longevity and increased healthspan. We tested whether this insulin sensitivity of the GHR-KO mouse is necessary for its retarded aging by abrogating that sensitivity with a transgenic alteration that improves development and secretory function of pancreatic β-cells by expressing Igf-1 under the rat insulin promoter 1 (RIP::IGF-1). The RIP::IGF-1 transgene increased circulating insulin content in GHR-KO mice, and thusly fully normalized their insulin sensitivity, without affecting the proliferation of any non-β-cell cell types. Multiple (nonsurvivorship) longevity-associated physiological and endocrinological characteristics of these mice (namely beneficial blood glucose regulatory control, altered metabolism, and preservation of memory capabilities) were partially or completely normalized, thus supporting the causal role of insulin sensitivity for the decelerated senescence of GHR-KO mice. We conclude that a delayed onset and/or decreased pace of aging can be hormonally regulated.
Collapse
Affiliation(s)
- Oge Arum
- Department of Internal Medicine Southern Illinois University‐School of Medicine Springfield IL 62794USA
| | - Ravneet K. Boparai
- Department of Internal Medicine Southern Illinois University‐School of Medicine Springfield IL 62794USA
| | - Jamal K. Saleh
- Department of Internal Medicine Southern Illinois University‐School of Medicine Springfield IL 62794USA
| | - Feiya Wang
- Department of Internal Medicine Southern Illinois University‐School of Medicine Springfield IL 62794USA
| | - Angela L. Dirks
- Department of Internal Medicine Southern Illinois University‐School of Medicine Springfield IL 62794USA
| | - Jeremy G. Turner
- Division of ENT‐Otolaryngology Department of Surgery Southern Illinois University‐School of Medicine Springfield IL 62794USA
| | - John J. Kopchick
- Edison Biotechnology Institute and Department of Biomedical Sciences Heritage College of Osteopathic Medicine Ohio University Athens OH 45701USA
| | - Jun‐Li Liu
- Fraser Laboratories for Diabetes Research Department of Medicine McGill University Health Centre 687 Pine Avenue West Montreal QC H3A 1A1 Canada
| | - Romesh K. Khardori
- Division of Endocrinology & Metabolism Department of Internal Medicine Eastern Virginia Medical School 700 West Olney Road Norfolk VA 23507 USA
| | - Andrzej Bartke
- Department of Internal Medicine Southern Illinois University‐School of Medicine Springfield IL 62794USA
| |
Collapse
|
85
|
Liu Y, Berry PA, Zhang Y, Jiang J, Lobie PE, Paulmurugan R, Langenheim JF, Chen WY, Zinn KR, Frank SJ. Dynamic analysis of GH receptor conformational changes by split luciferase complementation. Mol Endocrinol 2014; 28:1807-1819. [PMID: 25188449 PMCID: PMC4213363 DOI: 10.1210/me.2014-1153] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 08/28/2014] [Indexed: 11/19/2022] Open
Abstract
The transmembrane GH receptor (GHR) exists at least in part as a preformed homodimer on the cell surface. Structural and biochemical studies suggest that GH binds GHR in a 1:2 stoichiometry to effect acute GHR conformational changes that trigger the activation of the receptor-associated tyrosine kinase, Janus kinase 2 (JAK2), and downstream signaling. Despite information about GHR-GHR association derived from elegant fluorescence resonance energy transfer/bioluminescence resonance energy transfer studies, an assessment of the dynamics of GH-induced GHR conformational changes has been lacking. To this end, we used a split luciferase complementation assay that allowed detection in living cells of specific ligand-independent GHR-GHR interaction. Furthermore, GH treatment acutely augmented complementation of enzyme activity between GHRs fused, respectively, to N- and C-terminal fragments of firefly luciferase. Analysis of the temporal pattern of GH-induced complementation changes, pharmacological manipulation, genetic alteration of JAK2 levels, and truncation of the GHR intracellular domain (ICD) tail suggested that GH acutely enhances proximity of the GHR homodimer partners independent of the presence of JAK2, phosphorylation of GHR-luciferase chimeras, or an intact ICD. However, subsequent reduction of complementation requires JAK2 kinase activity and the ICD tail. This conclusion is in contrast to existing models of the GHR activation process.
Collapse
Affiliation(s)
- Ying Liu
- Department of Medicine (Y.L., P.A.B., Y.Z., J.J., S.J.F.), Division of Endocrinology, Diabetes, and Metabolism, and Departments of Radiology (K.R.Z.), and Cell, Developmental, and Integrative Biology (S.J.F.), University of Alabama at Birmingham, Birmingham, Alabama 35294; Cancer Science Institute of Singapore and Department of Pharmacology (P.E.L.), National University of Singapore, Singapore 119077; Department of Radiology (R.P.), Stanford University School of Medicine, Palo Alto, California 94304; Department of Biological Sciences (J.F.L., W.Y.C.), Clemson University, Clemson, South Carolina 29634; and Endocrinology Section (S.J.F.), Medical Service, Veterans Affairs Medical Center, Birmingham, Alabama 35233
| | | | | | | | | | | | | | | | | | | |
Collapse
|
86
|
Bozhkov AI, Nikitchenko YV. Thermogenesis and longevity in mammals. Thyroxin model of accelerated aging. Exp Gerontol 2014; 60:173-82. [PMID: 25446982 DOI: 10.1016/j.exger.2014.10.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 10/29/2014] [Accepted: 10/31/2014] [Indexed: 01/18/2023]
Abstract
Development of experimental models of life span regulation is an important goal of modern gerontology. We proposed a thyroxin model of accelerated aging. Male Wistar rats at the age of 17 months received thyroxin in drinking water at a concentration of 6 mg/L for 2 months as a model of induced hyperthyroidism (IH). Administration of thyroxin resulted in a decrease in life span and a 2°C increase in body temperature that was accompanied by a 2 fold increase in thyroxin level and a 40% increase in triiodothyronine in blood serum. Induced hyperthyroidism can be used as a model of accelerated aging. We also found that thyroxin administration acts as uncoupler of oxidative phosphorylation as treatment was accompanied by an increase in the generation of superoxide radicals by 50%. Antioxidant enzyme activity remained unchanged (glutathione peroxidase, glutathione reductase mitochondrial) or was reduced (glutathione-S-transferase by 1.7 times) as compared with the control. The activity of glucose-6-transferase was increased by 2.8 times as compared with control, and malate dehydrogenase activity in liver increased by 6.8 times. Induced hyperthyroidism in rats resulted in distinct epigenotype which was accompanied by a decrease in life span.
Collapse
Affiliation(s)
- A I Bozhkov
- Research Institute of Biology, V.N. Karazin Kharkov National University, 61022 Kharkov, Ukraine.
| | - Yu V Nikitchenko
- Research Institute of Biology, V.N. Karazin Kharkov National University, 61022 Kharkov, Ukraine
| |
Collapse
|
87
|
Arum O, Saleh J, Boparai R, Turner J, Kopchick J, Khardori R, Bartke A. Interaction of growth hormone receptor/binding protein gene disruption and caloric restriction for insulin sensitivity and attenuated aging. F1000Res 2014; 3:256. [PMID: 25789159 PMCID: PMC4358413 DOI: 10.12688/f1000research.5378.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/27/2014] [Indexed: 11/20/2022] Open
Abstract
The correlation of physiological sensitivity to insulin ( vis-à-vis glycemic regulation) and longevity is extensively established, creating a justifiable gerontological interest on whether insulin sensitivity is causative, or even predictive, of some or all phenotypes of slowed senescence (including longevity). The growth hormone receptor/ binding protein gene-disrupted (GHR-KO) mouse is the most extensively investigated insulin-sensitive, attenuated aging model. It was reported that, in a manner divergent from similar mutants, GHR-KO mice fail to respond to caloric restriction (CR) by altering their insulin sensitivity. We hypothesized that maximized insulin responsiveness is what causes GHR-KO mice to exhibit a suppressed survivorship response to dietary (including caloric) restriction; and attempted to refute this hypothesis by assessing the effects of CR on GHR-KO mice for varied slow-aging-associated phenotypes. In contrast to previous reports, we found GHR-KO mice on CR to be less responsive than their ad libitum (A.L.) counterparts to the hypoglycemia-inducing effects of insulin. Further, CR had negligible effects on the metabolism or cognition of GHR-KO mice. Therefore, our data suggest that the effects of CR on the insulin sensitivity of GHR-KO mice do not concur with the effects of CR on the aging of GHR-KO mice.
Collapse
Affiliation(s)
- Oge Arum
- Department of Internal Medicine, Southern Illinois University-School of Medicine, Springfield, IL, 62794, USA
| | - Jamal Saleh
- Department of Internal Medicine, Southern Illinois University-School of Medicine, Springfield, IL, 62794, USA
| | - Ravneet Boparai
- Department of Internal Medicine, Southern Illinois University-School of Medicine, Springfield, IL, 62794, USA
| | - Jeremy Turner
- Department of Surgery, Division of ENT-Otolaryngology, Southern Illinois University-School of Medicine, Springfield, IL, 62794, USA
| | - John Kopchick
- Edison Biotechnology Institute and Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, 45701, USA
| | - Romesh Khardori
- Department of Internal Medicine, Division of Endocrinology & Metabolism, Eastern Virginia Medical School, Norfolk, VA, 23507, USA
| | - Andrzej Bartke
- Department of Internal Medicine, Southern Illinois University-School of Medicine, Springfield, IL, 62794, USA
| |
Collapse
|
88
|
Growth hormone abolishes beneficial effects of calorie restriction in long-lived Ames dwarf mice. Exp Gerontol 2014; 58:219-229. [PMID: 25152388 DOI: 10.1016/j.exger.2014.08.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 08/16/2014] [Accepted: 08/20/2014] [Indexed: 11/23/2022]
Abstract
Disruption of the growth hormone (GH) axis promotes longevity and delays aging. In contrast, GH over-expression may lead to accelerated aging and shorter life. Calorie restriction (CR) improves insulin sensitivity and may extend lifespan. Long-lived Ames dwarf (df/df) mice have additional extension of longevity when subjected to 30% CR. The aim of the study was to assess effects of CR or GH replacement therapy separately and as a combined (CR+GH) treatment in GH-deficient df/df and normal mice, on selected metabolic parameters (e.g., insulin, glucose, cholesterol), insulin signaling components (e.g., insulin receptor [IR] β-subunit, phosphorylated form of IR [IR pY1158], protein kinase C ζ/λ [p-PKCζ/λ] and mTOR [p-mTOR]), transcription factor p-CREB, and components of the mitogen-activated protein kinase (MAPK) signaling (p-ERK1/2, p-p38), responsible for cell proliferation, differentiation and survival. CR decreased plasma levels of insulin, glucose, cholesterol and leptin, and increased hepatic IR β-subunit and IR pY1158 levels as well as IR, IRS-1 and GLUT-2 gene expression compared to ad libitum feeding, showing a significant beneficial diet intervention effect. Moreover, hepatic protein levels of p-PKCζ/λ, p-mTOR and p-p38 decreased, and p-CREB increased in CR mice. On the contrary, GH increased levels of glucose, cholesterol and leptin in plasma, and p-mTOR or p-p38 in livers, and decreased plasma adiponectin and hepatic IR β-subunit compared to saline treatment. There were no GH effects on adiponectin in N mice. Moreover, GH replacement therapy did not affect IR, IRS-1 and GLUT-2 gene expression. GH treatment abolishes the beneficial effects of CR; it may suggest an important role of GH-IGF1 axis in mediating the CR action. Suppressed somatotrophic signaling seems to predominate over GH replacement therapy in the context of the examined parameters and signaling pathways.
Collapse
|
89
|
Park CH, Kim DH, Park MH, Kim MK, Kim ND, Kim CM, Tanaka T, Yokozawa T, Chung HY, Moon HR. Chinese Prescription Kangen-karyu and Salviae Miltiorrhizae Radix Improve Age-Related Oxidative Stress and Inflammatory Response through the PI3K/Akt or MAPK Pathways. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2014; 42:987-1005. [DOI: 10.1142/s0192415x14500621] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
This study examined whether Kangen-karyu and its crude drug, Salviae Miltiorrhizae Radix, have a reno-protective effect on the age-related oxidative stress and inflammatory response through the phosphoinositide 3-kinase (PI3K)/Akt or mitogen-activated protein kinase (MAPK) pathways in aged rats. Kangen-karyu or Salviae Miltiorrhizae Radix (20 mg/kg body weight/day) was administered orally to old groups of rats for 16 days, and their effects were compared with the vehicle-treated old and young rats. The administration of Kangen-karyu caused a slight decrease in the serum glucose level and a significant decrease in the serum insulin level in the old rats. The increased levels of serum renal functional parameter (urea-nitrogen) and oxidative parameter were significantly reduced by both Kangen-karyu and Salviae Miltiorrhizae Radix. The old rats exhibited a dysregulation of the protein expression related to insulin resistance, oxidative stress, and inflammation in the kidneys, but Kangen-karyu administration significantly reduced the expression of the inflammatory proteins through the PI3K/Akt pathway. On the other hand, the Salviae Miltiorrhizae Radix-treated old rats showed a decrease in the inflammatory cytokines through the MAPK pathway. These results provide important evidence that Kangen-karyu and Salviae Miltiorrhizae Radix have a pleiotropic effect on the PI3K/Akt and MAPK pathways, showing renoprotective effects against the development of inflammation in old rats. This study provides scientific evidence that Kangen-karyu and Salviae Miltiorrhizae Radix improve the inflammatory responses via the PI3K/Akt or MAPK pathways in the kidney of old rats.
Collapse
Affiliation(s)
- Chan Hum Park
- Molecular Inflammation Research Center for Aging Intervention (MRCA), Pusan National University, Geumjeong-gu, Busan 609-735, Korea
| | - Dae Hyun Kim
- Molecular Inflammation Research Center for Aging Intervention (MRCA), Pusan National University, Geumjeong-gu, Busan 609-735, Korea
| | - Min Hi Park
- Molecular Inflammation Research Center for Aging Intervention (MRCA), Pusan National University, Geumjeong-gu, Busan 609-735, Korea
| | - Mi Kyung Kim
- Research Center for Anti-Aging Technology Development, Pusan National University, Geumjeong-gu, Busan 609-735, Korea
| | - Nam Deuk Kim
- Molecular Inflammation Research Center for Aging Intervention (MRCA), Pusan National University, Geumjeong-gu, Busan 609-735, Korea
- Research Center for Anti-Aging Technology Development, Pusan National University, Geumjeong-gu, Busan 609-735, Korea
| | - Cheol Min Kim
- Research Center for Anti-Aging Technology Development, Pusan National University, Geumjeong-gu, Busan 609-735, Korea
| | - Takashi Tanaka
- Graduate School of Biomedical Sciences, Nagasaki University, 1-14 Bunkyo-cho, Nagasaki 852-8521, Japan
| | - Takako Yokozawa
- Molecular Inflammation Research Center for Aging Intervention (MRCA), Pusan National University, Geumjeong-gu, Busan 609-735, Korea
- Graduate School of Science and Engineering for Research, University of Toyama, 3190 Gofuku, Toyama 930-8555, Japan
| | - Hae Young Chung
- Molecular Inflammation Research Center for Aging Intervention (MRCA), Pusan National University, Geumjeong-gu, Busan 609-735, Korea
| | - Hyung Ryong Moon
- Molecular Inflammation Research Center for Aging Intervention (MRCA), Pusan National University, Geumjeong-gu, Busan 609-735, Korea
| |
Collapse
|
90
|
Arum O, Saleh JK, Boparai RK, Kopchick JJ, Khardori RK, Bartke A. Preservation of blood glucose homeostasis in slow-senescing somatotrophism-deficient mice subjected to intermittent fasting begun at middle or old age. AGE (DORDRECHT, NETHERLANDS) 2014; 36:9651. [PMID: 24789008 PMCID: PMC4082609 DOI: 10.1007/s11357-014-9651-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 03/26/2014] [Indexed: 05/19/2023]
Abstract
Poor blood glucose homeostatic regulation is common, consequential, and costly for older and elderly populations, resulting in pleiotrophically adverse clinical outcomes. Somatotrophic signaling deficiency and dietary restriction have each been shown to delay the rate of senescence, resulting in salubrious phenotypes such as increased survivorship. Using two growth hormone (GH) signaling-related, slow-aging mouse mutants we tested, via longitudinal analyses, whether genetic perturbations that increase survivorship also improve blood glucose homeostatic regulation in senescing mammals. Furthermore, we institute a dietary restriction paradigm that also decelerates aging, an intermittent fasting (IF) feeding schedule, as either a short-term or a sustained intervention beginning at either middle or old age, and assess its effects on blood glucose control. We find that either of the two genetic alterations in GH signaling ameliorates fasting hyperglycemia; additionally, both longevity-inducing somatotrophic mutations improve insulin sensitivity into old age. Strikingly, we observe major and broad improvements in blood glucose homeostatic control by IF: IF improves ad libitum-fed hyperglycemia, glucose tolerance, and insulin sensitivity, and reduces hepatic gluconeogenesis, in aging mutant and normal mice. These results on correction of aging-resultant blood glucose dysregulation have potentially important clinical and public health implications for our ever-graying global population, and are consistent with the Longevity Dividend concept.
Collapse
Affiliation(s)
- Oge Arum
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL, 62794, USA,
| | | | | | | | | | | |
Collapse
|
91
|
Sadagurski M, Landeryou T, Blandino-Rosano M, Cady G, Elghazi L, Meister D, See L, Bartke A, Bernal-Mizrachi E, Miller RA. Long-lived crowded-litter mice exhibit lasting effects on insulin sensitivity and energy homeostasis. Am J Physiol Endocrinol Metab 2014; 306:E1305-14. [PMID: 24735888 PMCID: PMC4042097 DOI: 10.1152/ajpendo.00031.2014] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 04/14/2014] [Indexed: 12/25/2022]
Abstract
The action of nutrients on early postnatal growth can influence mammalian aging and longevity. Recent work has demonstrated that limiting nutrient availability in the first 3 wk of life [by increasing the number of pups in the crowded-litter (CL) model] leads to extension of mean and maximal lifespan in genetically normal mice. In this study, we aimed to characterize the impact of early-life nutrient intervention on glucose metabolism and energy homeostasis in CL mice. In our study, we used mice from litters supplemented to 12 or 15 pups and compared those to control litters limited to eight pups. At weaning and then throughout adult life, CL mice are significantly leaner and consume more oxygen relative to control mice. At 6 mo of age, CL mice had low fasting leptin concentrations, and low-dose leptin injections reduced body weight and food intake more in CL female mice than in controls. At 22 mo, CL female mice also have smaller adipocytes compared with controls. Glucose and insulin tolerance tests show an increase in insulin sensitivity in 6 mo old CL male mice, and females become more insulin sensitive later in life. Furthermore, β-cell mass was significantly reduced in the CL male mice and was associated with reduction in β-cell proliferation rate in these mice. Together, these data show that early-life nutrient intervention has a significant lifelong effect on metabolic characteristics that may contribute to the increased lifespan of CL mice.
Collapse
Affiliation(s)
- Marianna Sadagurski
- Department of Internal Medicine, Division of Geriatric and Palliative Medicine, University of Michigan, Ann Arbor, Michigan;
| | - Taylor Landeryou
- Department of Pathology and Geriatrics Center, University of Michigan, Ann Arbor, Michigan
| | - Manuel Blandino-Rosano
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, Michigan
| | - Gillian Cady
- Department of Pathology and Geriatrics Center, University of Michigan, Ann Arbor, Michigan
| | - Lynda Elghazi
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, Michigan
| | - Daniel Meister
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, Michigan
| | - Lauren See
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, Michigan
| | - Andrzej Bartke
- Department of Internal Medicine-Geriatrics Research, Southern Illinois University School of Medicine, Springfield, Illinois; and
| | - Ernesto Bernal-Mizrachi
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, Michigan; Endocrinology Section, Medical Service, Veterans Affairs Medical Center, Ann Arbor, Michigan
| | - Richard A Miller
- Department of Pathology and Geriatrics Center, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
92
|
Development of a murine model of early sepsis in diet-induced obesity. BIOMED RESEARCH INTERNATIONAL 2014; 2014:719853. [PMID: 24967393 PMCID: PMC4054609 DOI: 10.1155/2014/719853] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 02/25/2014] [Indexed: 12/15/2022]
Abstract
Sepsis, a global health issue, is the most common cause of mortality in the intensive care unit. The aim of this study was to develop a new model of sepsis that investigates the impact of prolonged western diet (WD) induced obesity on the response to early sepsis. Male C57BL/6 mice were fed either a high fat WD or normal chow diet (NCD) for 6, 15, or 27 weeks. Septic obese mice at 15 and 27 weeks had significantly lower levels of lung myeloperoxidase (26.3 ± 3.80 U/mg tissue) compared to age matched ad lib (44.1 ± 2.86 U/mg tissue) and diet restricted (63.2 ± 5.60 U/mg tissue) controls. Low levels of lung inflammation were not associated with changes in hepatic cytokines and oxidative stress levels. Obese mice had significantly (P < 0.0001) larger livers compared to controls. Histological examination of the livers demonstrated that WD fed mice had increased inflammation with pronounced fat infiltration, steatosis, and hepatocyte ballooning. Using this model of prolonged exposure to high fat diet we have data that agree with recent clinical observations suggesting obese individuals are protected from sepsis-induced lung injury. This model will allow us to investigate the links between damage to the hepatic microcirculation, immune response, and lung injury.
Collapse
|
93
|
Lin WS, Chen JY, Wang JC, Chen LY, Lin CH, Hsieh TR, Wang MF, Fu TF, Wang PY. The anti-aging effects of Ludwigia octovalvis on Drosophila melanogaster and SAMP8 mice. AGE (DORDRECHT, NETHERLANDS) 2014; 36:689-703. [PMID: 24338263 PMCID: PMC4039272 DOI: 10.1007/s11357-013-9606-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Accepted: 12/02/2013] [Indexed: 05/29/2023]
Abstract
We investigated the anti-aging effects of Ludwigia octovalvis (Jacq.) P. H. Raven (Onagraceae), an extract of which is widely consumed as a healthful drink in a number of countries. Using the fruit fly, Drosophila melanogaster, as a model organism, we demonstrated that L. octovalvis extract (LOE) significantly extended fly lifespan on a high, but not a low, calorie diet, indicating that LOE may regulate lifespan through a dietary restriction (DR)-related pathway. LOE also attenuated age-related cognitive decline in both flies and in the senescence-accelerated-prone 8 (SAMP8) mouse, without causing any discernable negative trade-offs, including water intake, food intake, fecundity, or spontaneous motor activity. LOE contained high levels of polyphenols and flavonoids, which possess strong DPPH radical scavenging activity, and was shown to attenuate paraquat-induced oxidative damage and lethality in flies. Gas chromatography-mass spectrometry (GC-MS) analyses identified 17 known molecules, of which β-sitosterol and squalene were the two most abundant. We further demonstrated that β-sitosterol was capable of extending lifespan, likely through activating AMP-activated protein kinase (AMPK) in the fat body of adult flies. Taken together, our data suggest that LOE is a potent anti-aging intervention with potential for treating age-related disorders.
Collapse
Affiliation(s)
- Wei-Sheng Lin
- />Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, No. 1, Sec. 1, Renai Rd., Taipei, 10051 Taiwan
| | - Jun-Yi Chen
- />Institute of Neuroscience, National Chengchi University, Taipei, Taiwan
| | - Jo-Chiao Wang
- />Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, No. 1, Sec. 1, Renai Rd., Taipei, 10051 Taiwan
| | - Liang-Yu Chen
- />Department of Biotechnology, Mingchuan University, Taoyuan, Taiwan
| | - Che-Hao Lin
- />Hsiehyu Biotech Company Ltd., Taipei, Taiwan
| | | | - Ming-Fu Wang
- />Department of Food and Nutrition, Providence University, Taichung, Taiwan
| | - Tsai-Feng Fu
- />Department of Applied Chemistry, National Chinan University, Nantou, Taiwan
| | - Pei-Yu Wang
- />Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, No. 1, Sec. 1, Renai Rd., Taipei, 10051 Taiwan
| |
Collapse
|
94
|
Long YC, Tan TMC, Takao I, Tang BL. The biochemistry and cell biology of aging: metabolic regulation through mitochondrial signaling. Am J Physiol Endocrinol Metab 2014; 306:E581-91. [PMID: 24452454 DOI: 10.1152/ajpendo.00665.2013] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cellular and organ metabolism affects organismal lifespan. Aging is characterized by increased risks for metabolic disorders, with age-associated degenerative diseases exhibiting varying degrees of mitochondrial dysfunction. The traditional view of the role of mitochondria generated reactive oxygen species (ROS) in cellular aging, assumed to be causative and simply detrimental for a long time now, is in need of reassessment. While there is little doubt that high levels of ROS are detrimental, mounting evidence points toward a lifespan extension effect exerted by mild to moderate ROS elevation. Dietary caloric restriction, inhibition of insulin-like growth factor-I signaling, and inhibition of the nutrient-sensing mechanistic target of rapamycin are robust longevity-promoting interventions. All of these appear to elicit mitochondrial retrograde signaling processes (defined as signaling from the mitochondria to the rest of the cell, for example, the mitochondrial unfolded protein response, or UPR(mt)). The effects of mitochondrial retrograde signaling may even spread to other cells/tissues in a noncell autonomous manner by yet unidentified signaling mediators. Multiple recent publications support the notion that an evolutionarily conserved, mitochondria-initiated signaling is central to the genetic and epigenetic regulation of cellular aging and organismal lifespan.
Collapse
Affiliation(s)
- Yun Chau Long
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University Health System, Singapore; and
| | | | | | | |
Collapse
|
95
|
Arum O, Rickman DJ, Kopchick JJ, Bartke A. The slow-aging growth hormone receptor/binding protein gene-disrupted (GHR-KO) mouse is protected from aging-resultant neuromusculoskeletal frailty. AGE (DORDRECHT, NETHERLANDS) 2014; 36:117-27. [PMID: 23824747 PMCID: PMC3889906 DOI: 10.1007/s11357-013-9551-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Accepted: 06/03/2013] [Indexed: 05/19/2023]
Abstract
Neuromusculoskeletal (physical) frailty is an aging-attributable biomedical issue of extremely high import, from both public health and individual perspectives. Yet, it is rarely studied in nonhuman research subjects and very rarely studied in animals with extended longevity. In an effort to address this relatively neglected area, we have conducted a longitudinal investigation of the neuromusculoskeletal healthspan in mice with two senescence-slowing interventions: growth hormone (GH) resistance, produced by GH receptor "knockout" (GHR-KO), and caloric restriction (CR). We report marked improvements in the retention of strength, balance, and motor coordination by the longevity-conferring GHR/BP gene disruption, CR regimen, or a combination of the two. Specifically, GHR-KO mice exhibit superior grip strength, balance, and motor coordination at middle age, and CR mice display superior grip strength at middle age. The advantageous effects established by middle-age are more pronounced in old-age, and these robust alterations are, generally, not gender-specific. Thus, we show that genetic and/or dietary interventions that engender longevity are also beneficial for the retention of neuromusculoskeletal health and functionality. The translational knowledge to be gained from subsequent molecular or histological investigations of these models of preserved functionality and decelerated senescence is potentially relevant to the efforts to reduce the specter of fear, falls, fracture, and frailty in the elderly.
Collapse
Affiliation(s)
- Oge Arum
- />Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL 62794 USA
- />631 N. 6th St., 2B, Springfield, IL 62702 USA
| | - Dustin J. Rickman
- />Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL 62794 USA
| | - John J. Kopchick
- />Edison Biotechnology Institute and Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701 USA
| | - Andrzej Bartke
- />Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL 62794 USA
| |
Collapse
|
96
|
Xu J, Gontier G, Chaker Z, Lacube P, Dupont J, Holzenberger M. Longevity effect of IGF-1R(+/-) mutation depends on genetic background-specific receptor activation. Aging Cell 2014; 13:19-28. [PMID: 23898955 PMCID: PMC4326867 DOI: 10.1111/acel.12145] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/21/2013] [Indexed: 12/22/2022] Open
Abstract
Growth hormone (GH) and insulin-like growth factor (IGF) signaling regulates lifespan in mice. The modulating effects of genetic background gained much attention because it was shown that life-prolonging effects in Snell dwarf and GH receptor knockout vary between mouse strains. We previously reported that heterozygous IGF-1R inactivation (IGF-1R(+/-) ) extends lifespan in female mice on 129/SvPas background, but it remained unclear whether this mutation produces a similar effect in other genetic backgrounds and which molecules possibly modify this effect. Here, we measured the life-prolonging effect of IGF-1R(+/-) mutation in C57BL/6J background and investigated the role of insulin/IGF signaling molecules in strain-dependent differences. We found significant lifespan extension in female IGF-1R(+/-) mutants on C57BL/6J background, but the effect was smaller than in 129/SvPas, suggesting strain-specific penetrance of longevity phenotypes. Comparing GH/IGF pathways between wild-type 129/SvPas and C57BL/6J mice, we found that circulating IGF-I and activation of IGF-1R, IRS-1, and IRS-2 were markedly elevated in 129/SvPas, while activation of IGF pathways was constitutively low in spontaneously long-lived C57BL/6J mice. Importantly, we demonstrated that loss of one IGF-1R allele diminished the level of activated IGF-1R and IRS more profoundly and triggered stronger endocrine feedback in 129/SvPas background than in C57BL/6J. We also revealed that acute oxidative stress entails robust IGF-1R pathway activation, which could account for the fact that IGF-1R(+/-) stress resistance phenotypes are fully penetrant in both backgrounds. Together, these results provide a possible explanation why IGF-1R(+/-) was less efficient in extending lifespan in C57BL/6J compared with 129/SvPas.
Collapse
Affiliation(s)
- Jie Xu
- INSERM; Hôpital Saint-Antoine; Paris 75012 France
- Université Pierre et Marie Curie; UPMC; Paris 75005 France
| | - Géraldine Gontier
- INSERM; Hôpital Saint-Antoine; Paris 75012 France
- Université Pierre et Marie Curie; UPMC; Paris 75005 France
| | - Zayna Chaker
- INSERM; Hôpital Saint-Antoine; Paris 75012 France
- Université Pierre et Marie Curie; UPMC; Paris 75005 France
- Faculté de Médecine; Université Paris Descartes; Paris 75006 France
| | - Philippe Lacube
- INSERM; Hôpital Saint-Antoine; Paris 75012 France
- Université Pierre et Marie Curie; UPMC; Paris 75005 France
| | - Joëlle Dupont
- INRA UMR7247; Nouzilly 37380 France
- CNRS UMR6175; Nouzilly 37380 France
- Université François Rabelais; Tours 37041 France
| | - Martin Holzenberger
- INSERM; Hôpital Saint-Antoine; Paris 75012 France
- Université Pierre et Marie Curie; UPMC; Paris 75005 France
| |
Collapse
|
97
|
|
98
|
Sun LY, Spong A, Swindell WR, Fang Y, Hill C, Huber JA, Boehm JD, Westbrook R, Salvatori R, Bartke A. Growth hormone-releasing hormone disruption extends lifespan and regulates response to caloric restriction in mice. eLife 2013; 2:e01098. [PMID: 24175087 PMCID: PMC3810783 DOI: 10.7554/elife.01098] [Citation(s) in RCA: 113] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Accepted: 09/25/2013] [Indexed: 12/18/2022] Open
Abstract
We examine the impact of targeted disruption of growth hormone-releasing hormone (GHRH) in mice on longevity and the putative mechanisms of delayed aging. GHRH knockout mice are remarkably long-lived, exhibiting major shifts in the expression of genes related to xenobiotic detoxification, stress resistance, and insulin signaling. These mutant mice also have increased adiponectin levels and alterations in glucose homeostasis consistent with the removal of the counter-insulin effects of growth hormone. While these effects overlap with those of caloric restriction, we show that the effects of caloric restriction (CR) and the GHRH mutation are additive, with lifespan of GHRH-KO mutants further increased by CR. We conclude that GHRH-KO mice feature perturbations in a network of signaling pathways related to stress resistance, metabolic control and inflammation, and therefore provide a new model that can be used to explore links between GHRH repression, downregulation of the somatotropic axis, and extended longevity. DOI:http://dx.doi.org/10.7554/eLife.01098.001 There is increasing evidence that the hormonal systems involved in growth, the metabolism of glucose, and the processes that balance energy intake and expenditure might also be involved in the aging process. In rodents, mutations in genes involved in these hormone-signaling pathways can substantially increase lifespan, as can a diet that is low in calories but which avoids malnutrition. As well as living longer, such mice also show reductions in age-related conditions such as diabetes, memory loss and cancer. Many of these effects appear to involve the actions of growth hormone. Mice with mutations that disrupt the development of the pituitary gland, which produces growth hormone, show increased longevity, as do mice that lack the receptor for growth hormone. However, these animals also show changes in a number of other hormones, making it difficult to be sure that the reduction in growth hormone signaling is responsible for their increased lifespan. Now, Sun et al. have studied mutant mice that lack a gene called GHRH, which promotes the release of growth hormone. These mice, which have normal levels of all other pituitary hormones, lived for up to 50% longer than their wild-type littermates. They were more active than normal mice and had more body fat, and showed greatly increased sensitivity to insulin. Some of the changes in these mutant mice resembled those seen in animals with a restricted calorie intake, suggesting that the same mechanisms may be implicated in both. However, Sun et al. found that caloric restriction further increased the lifespans of their GHRH knockout mice, indicating that at least some of the effects of caloric restriction are independent of disrupted growth hormone signaling. The results of this study are an important step forward for understanding how growth hormone signaling and caloric restriction regulate aging, both individually and in combination. The GHRH knockout mice are likely to become an important model system for studying these processes and for understanding the complex interactions between diet and hormonal pathways. DOI:http://dx.doi.org/10.7554/eLife.01098.002
Collapse
Affiliation(s)
- Liou Y Sun
- Department of Internal Medicine , Southern Illinois University School of Medicine , Springfield , United States
| | | | | | | | | | | | | | | | | | | |
Collapse
|
99
|
Li X, Bartke A, Berryman DE, Funk K, Kopchick JJ, List EO, Sun L, Miller RA. Direct and indirect effects of growth hormone receptor ablation on liver expression of xenobiotic metabolizing genes. Am J Physiol Endocrinol Metab 2013; 305:E942-50. [PMID: 23941873 PMCID: PMC3798695 DOI: 10.1152/ajpendo.00304.2013] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Detoxification of ingested xenobiotic chemicals, and of potentially toxic endogenous metabolites, is carried out largely through a series of enzymes synthesized in the liver, sometimes called "xenobiotic metabolizing enzymes" (XME). Expression of these XME is sexually dimorphic in rodents and humans, with many of the XME expressed at higher levels in females. This expression pattern is thought to be regulated, in part, by the sex differences in circadian growth hormone (GH) pulsatility. We have evaluated mRNA, in the liver, for 52 XME genes in male and female mice of four mutant stocks, with diminished levels of GH receptor (GHR) either globally (GKO), or in liver (LKO), fat (FKO), or muscle (MKO) tissue specifically. The data show complex, sex-specific changes. For some XME, the expression pattern is consistent with direct control of hepatic mRNA by GHR in the liver. In contrast, other XME show evidence for indirect pathways in which hepatic XME expression is altered by GH signals in fat or skeletal muscle. The effects of GHR-null mutations on glucose control, responses to dietary interventions, steroid metabolism, detoxification pathways, and lifespan may depend on a mixture of direct hepatic effects and cross talk between different GH-responsive tissues.
Collapse
Affiliation(s)
- Xinna Li
- 1Department of Pathology, and Geriatrics Center, University of Michigan School of Medicine, Ann Arbor, Michigan;
| | - Andrzej Bartke
- 2Department of Internal Medicine, Geriatrics Research, Southern Illinois University School of Medicine, Springfield, Illinois;
| | - Darlene E. Berryman
- 3Edison Biotechnology Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio;
- 4School of Applied Health Sciences and Wellness, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio;
- 5Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio; and
| | - Kevin Funk
- 3Edison Biotechnology Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio;
| | - John J. Kopchick
- 3Edison Biotechnology Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio;
- 5Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio; and
| | - Edward O. List
- 3Edison Biotechnology Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio;
- 6Department of Specialty Medicine, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio
| | - Liou Sun
- 2Department of Internal Medicine, Geriatrics Research, Southern Illinois University School of Medicine, Springfield, Illinois;
| | - Richard A. Miller
- 1Department of Pathology, and Geriatrics Center, University of Michigan School of Medicine, Ann Arbor, Michigan;
| |
Collapse
|
100
|
Słuczanowska-Głąbowska S, Laszczyńska M, Piotrowska K, Głąbowski W, Rumianowski B, Masternak M, Arum O, Kucia M, Kopchick JJ, Bartke A, Ratajczak MZ. The effect of calorie restriction on the presence of apoptotic ovarian cells in normal wild type mice and low-plasma-IGF-1 Laron dwarf mice. J Ovarian Res 2013; 6:67. [PMID: 24063422 PMCID: PMC3849152 DOI: 10.1186/1757-2215-6-67] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2013] [Accepted: 09/20/2013] [Indexed: 11/24/2022] Open
Abstract
Background It is known that caloric restriction extends lifespan and can minimize age-related dysfunction of the reproductive system. We became interested in how caloric restriction influences apoptosis, which is a crucial process that maintains ovarian cell homeostasis. Methods We examined ovarian cells in: 2.5-year-old wild type mice on caloric restriction (CR) or fed ad libitum (AL) and Laron dwarf mice (GHR-KO) at the same ages on CR or fed AL. Apoptosis was assessed by histochemical analysis on paraffin sections of ovarian tissue. Results Morphological and histochemical analysis revealed that CR improved reproductive potential in 2.5-year-old WT littermates and GHR-KO female mice, as indicated by the increased number of ovarian follicles. The level of apoptosis in ovarian tissue was higher in WT mice on a CR diet compared with WT mice on the AL diet. In GHR-KO mice, the level of apoptosis in ovaries was similar for mice on CR and on AL diets and bigger than in WT mice on CR. Conclusions Morphological and histochemical analysis revealed a younger biological age of the ovaries in 2-year-old WT littermates and GHR-KO female mice on CR compared with animals fed AL.
Collapse
|