1
|
Conti B, de Cabo R. Promoting health and survival through lowered body temperature. NATURE AGING 2025; 5:740-749. [PMID: 40205073 DOI: 10.1038/s43587-025-00850-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 03/13/2025] [Indexed: 04/11/2025]
Abstract
Core body temperature (Tb) is a long-established determinant of longevity across species. In this Perspective, we first summarize evidence demonstrating that reducing Tb increases lifespan and that lowered Tb contributes to the antiaging effects of calorie restriction. Next, we discuss recent data that diverge from prior hypotheses on the mechanisms by which Tb affects longevity, suggesting these are limited neither to the thermodynamics of nonenzymatic chemical reactions, nor reduced formation of mitochondrial reactive oxygen species nor lowered metabolic rate. Instead, recent findings in invertebrates show that cold promotes longevity via specific pathways including nutrient sensing and proteostasis, as well as modulating the thermodynamics of proteins and nucleic acids by changing their structure and function, for example, affecting temperature-sensitive ion channels, long-lived temperature-sensitive dauer mutations, base-pair stability and stem-loop RNA structures. Temperature affects the epigenetic signature and inflammation, and lowering Tb can also induce RNA-binding cold shock proteins, activate cold-sensitive kinases and differential splicing to potentially reshape the cellular environment. Finally, we reflect on important future work and the translational potential of temperature management and temperature mimetics.
Collapse
Affiliation(s)
- Bruno Conti
- San Diego Biomedical Research Institute, San Diego, CA, USA.
| | - Rafael de Cabo
- National Institute on Aging, National Institutes of Health, Baltimore, MD, USA.
| |
Collapse
|
2
|
Wibisono S, Wibisono P, Chen CH, Sun J, Liu Y. The Caenorhabditis elegans neuronal GPCR OCTR-1 modulates longevity responses to both warm and cold temperatures. iScience 2025; 28:112279. [PMID: 40264795 PMCID: PMC12013480 DOI: 10.1016/j.isci.2025.112279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 01/27/2025] [Accepted: 03/19/2025] [Indexed: 04/24/2025] Open
Abstract
Many animal species live longer in cold climates than in warm climates, which was traditionally explained using the rate of living theory, i.e., higher temperatures increase chemical reaction rates, thus speeding up the aging process. However, recent studies have identified specific molecules and cells that are involved in longevity responses to temperature, indicating that such responses are not simply thermodynamic but are regulated processes. Here, we report that Caenorhabditis elegans lacking the neuronal G protein-coupled receptor OCTR-1 have extended lifespans at a warm temperature but shortened lifespans at a cold temperature, demonstrating that OCTR-1 modulates temperature-induced longevity responses. These responses are regulated by the OCTR-1-expressing, chemosensory ASH neurons. Furthermore, the OCTR-1 pathway controls such responses to warm and cold temperatures by regulating the expressions of immune response genes and the intestinal transcriptional factor ELT-2, respectively. Overall, our study provides cellular and molecular insights into the relationship between temperature and longevity.
Collapse
Affiliation(s)
- Shawndra Wibisono
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, USA
| | - Phillip Wibisono
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, USA
| | - Chia-Hui Chen
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, USA
| | - Jingru Sun
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, USA
| | - Yiyong Liu
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, USA
- Genomics Core, Washington State University, Spokane, WA, USA
| |
Collapse
|
3
|
Wang A, Speakman JR. Potential downsides of calorie restriction. Nat Rev Endocrinol 2025:10.1038/s41574-025-01111-1. [PMID: 40247012 DOI: 10.1038/s41574-025-01111-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/26/2025] [Indexed: 04/19/2025]
Abstract
Although the potential benefits of calorie restriction on human lifespan remain uncertain, it is currently one of the most extensively researched non-genetic approaches to extending both lifespan and healthspan in animals. Calorie restriction offers numerous health benefits, including a reduced incidence of age-related diseases. However, calorie restriction also produces a range of negative effects, which are not fully documented and require further investigation, particularly in humans. As the viability of calorie restriction in humans will depend on the balance of benefits and detrimental effects, it is crucial to understand the nature of these negative effects and what drives them. In this Review, we summarize the effects of calorie restriction on wound healing, hunger, cold sensitivity, bone health, brain size, cognition, reproductive performance and infection, primarily based on studies of rodents with some data from other species and from humans. Overall, the detrimental effects of calorie restriction seem to stem directly from prioritization of vital functions and downregulation or suppression of energy-demanding processes, which helps preserve survival but can also lead to impaired physiological performance and increased vulnerability to stressors. The exact mechanisms underlying these effects remain unclear. Whether it might be possible to engage in calorie restriction but avoid these negative effects remains uncertain.
Collapse
Affiliation(s)
- Anyongqi Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- Beijing Life Science Academy, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - John Roger Speakman
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.
- Shenzhen Key Laboratory of Metabolic Health, Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
- Faculty of Pharmaceutical Sciences, Shenzhen University of Advanced Technology, Shenzhen, China.
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, UK.
- Institute of Health Sciences, China Medical University, Shenyang, China.
| |
Collapse
|
4
|
Shashikadze B, Franzmeier S, Hofmann I, Kraetzl M, Flenkenthaler F, Blutke A, Fröhlich T, Wolf E, Hinrichs A. Structural and proteomic repercussions of growth hormone receptor deficiency on the pituitary gland: Lessons from a translational pig model. J Neuroendocrinol 2024; 36:e13277. [PMID: 37160285 DOI: 10.1111/jne.13277] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 04/03/2023] [Accepted: 04/04/2023] [Indexed: 05/11/2023]
Abstract
Growth hormone receptor deficiency (GHRD) results in low serum insulin-like growth factor 1 (IGF1) and high, but non-functional serum growth hormone (GH) levels in human Laron syndrome (LS) patients and animal models. This study investigated the quantitative histomorphological and molecular alterations associated with GHRD. Pituitary glands from 6 months old growth hormone receptor deficient (GHR-KO) and control pigs were analyzed using a quantitative histomorphological approach in paraffin (9 GHR-KO [5 males, 4 females] vs. 11 controls [5 males, 6 females]), ultrathin sections tissue sections (3 male GHR-KO vs. 3 male controls) and label-free proteomics (4 GHR-KO vs. 4 control pigs [2 per sex]). GHR-KO pigs displayed reduced body weights (60% reduction in comparison to controls; p < .0001) and decreased pituitary volumes (54% reduction in comparison to controls; p < .0001). The volume proportion of the adenohypophysis did not differ in GHR-KO and control pituitaries (65% vs. 71%; p = .0506) and GHR-KO adenohypophyses displayed a reduced absolute volume but an unaltered volume density of somatotrophs in comparison to controls (21% vs. 18%; p = .3164). In GHR-KO pigs, somatotroph cells displayed a significantly reduced volume density of granules (23.5%) as compared to controls (67.7%; p < .0001). Holistic proteome analysis of adenohypophysis samples identified 4660 proteins, of which 592 were differentially abundant between the GHR-KO and control groups. In GHR-KO samples, the abundance of somatotropin precursor was decreased, whereas increased abundances of proteins involved in protein production, transport and endoplasmic reticulum (ER) stress were revealed. Increased protein production and secretion as well as significantly reduced proportion of GH-storing granules in somatotroph cells of the adenohypophysis without an increase in volume density of somatotroph cells in the adenohypophysis could explain elevated serum GH levels in GHR-KO pigs.
Collapse
Affiliation(s)
- Bachuki Shashikadze
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Sophie Franzmeier
- Institute of Veterinary Pathology at the Center for Clinical Veterinary Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Isabel Hofmann
- Institute of Veterinary Pathology at the Center for Clinical Veterinary Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Martin Kraetzl
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, Munich, Germany
- Center for Innovative Medical Models (CiMM), Ludwig-Maximilians-Universität München, Oberschleissheim, Germany
| | - Florian Flenkenthaler
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Andreas Blutke
- Institute of Veterinary Pathology at the Center for Clinical Veterinary Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Thomas Fröhlich
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Eckhard Wolf
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, Ludwig-Maximilians-Universität München, Munich, Germany
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, Munich, Germany
- Center for Innovative Medical Models (CiMM), Ludwig-Maximilians-Universität München, Oberschleissheim, Germany
| | - Arne Hinrichs
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, Munich, Germany
- Center for Innovative Medical Models (CiMM), Ludwig-Maximilians-Universität München, Oberschleissheim, Germany
| |
Collapse
|
5
|
Austad SN, Smith JR, Hoffman JM. Amino acid restriction, aging, and longevity: an update. FRONTIERS IN AGING 2024; 5:1393216. [PMID: 38757144 PMCID: PMC11096585 DOI: 10.3389/fragi.2024.1393216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 04/18/2024] [Indexed: 05/18/2024]
Abstract
Various so-called dietary restriction paradigms have shown promise for extending health and life. All such paradigms rely on ad libitum (hereafter ad lib) feeding, something virtually never employed in animals whose long-term health we value, either as a control or, except for food restriction itself, for both control and treatment arms of the experiment. Even though the mechanism(s) remain only vaguely understood, compared to ad lib-fed animals a host of dietary manipulations, including calorie restriction, low protein, methionine, branched-chain amino acids, and even low isoleucine have demonstrable health benefits in laboratory species in a standard laboratory environment. The remaining challenge is to determine whether these health benefits remain in more realistic environments and how they interact with other health enhancing treatments such as exercise or emerging geroprotective drugs. Here we review the current state of the field of amino acid restriction on longevity of animal models and evaluate its translational potential.
Collapse
Affiliation(s)
- S. N. Austad
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - J. R. Smith
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - J. M. Hoffman
- Department of Biological Sciences, Augusta University, Augusta, GA, United States
| |
Collapse
|
6
|
List EO, Duran-Ortiz S, Kulkarni P, Davis E, Mora-Criollo P, Berryman DE, Kopchick JJ. Growth hormone receptor gene disruption. VITAMINS AND HORMONES 2023; 123:109-149. [PMID: 37717983 PMCID: PMC11462719 DOI: 10.1016/bs.vh.2022.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2023]
Abstract
Much of our understanding of growth hormone's (GH)'s numerous activities stems from studies utilizing GH receptor (GHR) knockout mice. More recently, the role of GH action has been examined by creating mice with tissue-specific or temporal GHR disruption. To date, 37 distinct GHR knockout mouse lines have been created. Targeted tissues include fat, liver, muscle, heart, bone, brain, macrophage, intestine, hematopoietic stem cells, pancreatic β cells, and inducible multi-tissue "global" disruption at various ages. In this chapter, a summary of each mouse line is provided with background information on the generation of the mouse line as well as important physiological outcomes resulting from GHR gene disruption. Collectively, these mouse lines provide unique insights into GH action and have resulted in the development of new hypotheses about the functions ascribed to GH action in particular tissues.
Collapse
Affiliation(s)
- Edward O List
- The Edison Biotechnology Institute, and the Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, United States
| | - Silvana Duran-Ortiz
- The Edison Biotechnology Institute, and the Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, United States
| | - Prateek Kulkarni
- The Edison Biotechnology Institute, and the Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, United States
| | - Emily Davis
- The Edison Biotechnology Institute, and the Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, United States
| | - Patricia Mora-Criollo
- The Edison Biotechnology Institute, and the Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, United States
| | - Darlene E Berryman
- The Edison Biotechnology Institute, and the Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, United States
| | - John J Kopchick
- The Edison Biotechnology Institute, and the Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, United States.
| |
Collapse
|
7
|
Growth hormone receptor (GHR) in AgRP neurons regulates thermogenesis in a sex-specific manner. GeroScience 2023:10.1007/s11357-023-00726-4. [PMID: 36633824 PMCID: PMC10400518 DOI: 10.1007/s11357-023-00726-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 01/03/2023] [Indexed: 01/13/2023] Open
Abstract
Evidence for hypothalamic regulation of energy homeostasis and thermoregulation in brown adipose tissue (BAT) during aging has been well recognized, yet the central molecular mediators involved in this process are poorly understood. The arcuate hypothalamus, orexigenic agouti-related peptide (AgRP) neurons control nutrient intake, energy homeostasis, and BAT thermogenesis. To determine the roles of growth hormone receptor (GHR) signaling in the AgRP neurons, we used mice with the AgRP-specific GHR deletion (AgRPΔGHR). We found that female AgRPΔGHR mice were resistant to temperature adaptation, and their body core temperature remained significantly lower when held at 10 °C, 22 °C, or 30 °C, compared to control mice. Low body core temperature in female AgRPΔGHR mice has been associated with significant reductions in Ucp1 and Pgc1α expression in the BAT. Further, neuronal activity in AgRP in response to cold exposure was blunted in AgRPΔGHR female mice, while the number of Fos+ AgRP neurons was increased in female controls exposed to cold. Global transcriptome from BAT identified increased the expression of genes related to immune responses and chemokine activity and decreased the expression of genes involved in triglyceride synthesis and metabolic pathways in AgRPΔGHR female mice. Importantly, these were the same genes that are downregulated by thermoneutrality in control mice but not in the AgRPΔGHR animals. Collectively, these data demonstrate a novel sex-specific role for GHR signaling in AgRP neurons in thermal regulation, which might be particularly relevant during aging.
Collapse
|
8
|
Qian Y, Berryman DE, Basu R, List EO, Okada S, Young JA, Jensen EA, Bell SRC, Kulkarni P, Duran-Ortiz S, Mora-Criollo P, Mathes SC, Brittain AL, Buchman M, Davis E, Funk KR, Bogart J, Ibarra D, Mendez-Gibson I, Slyby J, Terry J, Kopchick JJ. Mice with gene alterations in the GH and IGF family. Pituitary 2022; 25:1-51. [PMID: 34797529 PMCID: PMC8603657 DOI: 10.1007/s11102-021-01191-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/21/2021] [Indexed: 01/04/2023]
Abstract
Much of our understanding of GH's action stems from animal models and the generation and characterization of genetically altered or modified mice. Manipulation of genes in the GH/IGF1 family in animals started in 1982 when the first GH transgenic mice were produced. Since then, multiple laboratories have altered mouse DNA to globally disrupt Gh, Ghr, and other genes upstream or downstream of GH or its receptor. The ability to stay current with the various genetically manipulated mouse lines within the realm of GH/IGF1 research has been daunting. As such, this review attempts to consolidate and summarize the literature related to the initial characterization of many of the known gene-manipulated mice relating to the actions of GH, PRL and IGF1. We have organized the mouse lines by modifications made to constituents of the GH/IGF1 family either upstream or downstream of GHR or to the GHR itself. Available data on the effect of altered gene expression on growth, GH/IGF1 levels, body composition, reproduction, diabetes, metabolism, cancer, and aging are summarized. For the ease of finding this information, key words are highlighted in bold throughout the main text for each mouse line and this information is summarized in Tables 1, 2, 3 and 4. Most importantly, the collective data derived from and reported for these mice have enhanced our understanding of GH action.
Collapse
Affiliation(s)
- Yanrong Qian
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
| | - Darlene E Berryman
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Reetobrata Basu
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
| | - Edward O List
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
| | - Shigeru Okada
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- Department of Pediatrics, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Jonathan A Young
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Elizabeth A Jensen
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
- Translational Biomedical Sciences Doctoral Program, Ohio University, Athens, OH, USA
| | - Stephen R C Bell
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Prateek Kulkarni
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- Department of Biological Sciences, College of Arts and Sciences, Ohio University, Athens, OH, USA
- Molecular and Cellular Biology Program, Ohio University, Athens, OH, USA
| | | | - Patricia Mora-Criollo
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- Translational Biomedical Sciences Doctoral Program, Ohio University, Athens, OH, USA
| | - Samuel C Mathes
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
| | - Alison L Brittain
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
- Molecular and Cellular Biology Program, Ohio University, Athens, OH, USA
| | - Mat Buchman
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Emily Davis
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- Department of Biological Sciences, College of Arts and Sciences, Ohio University, Athens, OH, USA
- Molecular and Cellular Biology Program, Ohio University, Athens, OH, USA
| | - Kevin R Funk
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- Department of Biological Sciences, College of Arts and Sciences, Ohio University, Athens, OH, USA
- Molecular and Cellular Biology Program, Ohio University, Athens, OH, USA
| | - Jolie Bogart
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- Department of Biological Sciences, College of Arts and Sciences, Ohio University, Athens, OH, USA
| | - Diego Ibarra
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- Department of Chemistry and Biochemistry, College of Arts and Sciences, Ohio University, Athens, OH, USA
| | - Isaac Mendez-Gibson
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- College of Health Sciences and Professions, Ohio University, Athens, OH, USA
| | - Julie Slyby
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- Department of Biological Sciences, College of Arts and Sciences, Ohio University, Athens, OH, USA
| | - Joseph Terry
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- Department of Biological Sciences, College of Arts and Sciences, Ohio University, Athens, OH, USA
| | - John J Kopchick
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA.
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA.
| |
Collapse
|
9
|
Morgan SA, Berryman DE, List EO, Lavery GG, Stewart PM, Kopchick JJ. Regulation of 11β-HSD1 by GH/IGF-1 in key metabolic tissues may contribute to metabolic disease in GH deficient patients. Growth Horm IGF Res 2022; 62:101440. [PMID: 34814007 DOI: 10.1016/j.ghir.2021.101440] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 11/01/2021] [Accepted: 11/14/2021] [Indexed: 11/19/2022]
Abstract
Patients with growth hormone deficiency (GHD) have many clinical features in common with Cushing's syndrome (glucocorticoid excess) - notably visceral obesity, insulin resistance, muscle myopathy and increased vascular mortality. Within key metabolic tissues, 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) converts cortisone to the active glucocorticoid, cortisol (11-dehydrocorticosterone and corticosterone in rodents respectively), and thus amplifies local glucocorticoid action. We hypothesize that 11β-HSD1 expression is negatively regulated by growth hormone (GH), and that GHD patients have elevated 11β-HSD1 within key metabolic tissues (leading to increased intracellular cortisol generation) which contributes to the clinical features of this disease. To identify the impact of GH excess/resistance on 11β-HSD1 in vivo, we measured mRNA expression in key metabolic tissues of giant mice expressing the bovine GH (bGH) gene, dwarf mice with a disrupted GH receptor (GHRKO) gene and mice expressing a gene encoding a GH receptor antagonist (GHA). Additionally, we assessed urine steroid markers of 11β-HSD1 activity in both GHRKO and bGH animals. 11β-HSD1 expression was decreased in gastrocnemius muscle (0.43-fold, p < 0.05), subcutaneous adipose (0.53-fold, p < 0.05) and epididymal adipose tissue (0.40-fold, p < 0.05), but not liver, in bGH mice compared to WT controls. This was paralleled by an increased percentage of 11-DHC (inactive glucocorticoid) present in the urine of bGH mice compared to WT controls (2.5-fold, p < 0.01) - consistent with decreased systemic 11β-HSD1 activity. By contrast, expression of 11β-HSD1 was increased in the liver of GHRKO (2.7-fold, p < 0.05) and GHA mice (2.0-fold, p < 0.05) compared to WT controls, but not gastrocnemius muscle, subcutaneous adipose tissue or epididymal adipose tissue. In summary, we have demonstrated a negative relationship between GH action and 11β-HSD1 expression which appears to be tissue specific. These data provide evidence that increased intracellular cortisol production within key tissues may contribute to metabolic disease in GHD patients.
Collapse
Affiliation(s)
- Stuart A Morgan
- Institute of Metabolism & Systems Research, College of Medical and Dental Sciences, Institute of Biomedical Research, University of Birmingham, Birmingham B15 2TT, UK.
| | - Darlene E Berryman
- Edison Biotechnology Institute, Ohio University/The Ridges, 1 Water Tower Drive, Building #25, Athens, OH 45701, USA
| | - Edward O List
- Edison Biotechnology Institute, Ohio University/The Ridges, 1 Water Tower Drive, Building #25, Athens, OH 45701, USA
| | - Gareth G Lavery
- Institute of Metabolism & Systems Research, College of Medical and Dental Sciences, Institute of Biomedical Research, University of Birmingham, Birmingham B15 2TT, UK
| | - Paul M Stewart
- Institute of Metabolism & Systems Research, College of Medical and Dental Sciences, Institute of Biomedical Research, University of Birmingham, Birmingham B15 2TT, UK; Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - John J Kopchick
- Edison Biotechnology Institute, Ohio University/The Ridges, 1 Water Tower Drive, Building #25, Athens, OH 45701, USA
| |
Collapse
|
10
|
Matzkin ME, Calandra RS, Rossi SP, Bartke A, Frungieri MB. Hallmarks of Testicular Aging: The Challenge of Anti-Inflammatory and Antioxidant Therapies Using Natural and/or Pharmacological Compounds to Improve the Physiopathological Status of the Aged Male Gonad. Cells 2021; 10:cells10113114. [PMID: 34831334 PMCID: PMC8619877 DOI: 10.3390/cells10113114] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/06/2021] [Accepted: 11/08/2021] [Indexed: 12/21/2022] Open
Abstract
The evolutionary theory of aging supports a trade-off relationship between reproduction and aging. Aging of the male reproductive system primarily affects the testes, leading to a decrease in the levels of sexual hormones, alterations in sperm quality and production, and a decline in fertility that does not necessarily involve a complete cessation of spermatogenesis. Inflammation, oxidation, and apoptosis are events considered as predictors of pathogenesis and the development of age-related diseases that are frequently observed in aged testes. Although the molecular mechanisms are still poorly understood, accumulating evidence points toward pro-inflammatory molecules and reactive oxygen species as primary contributing factors for testicular aging. However, the real impact of aging-related testicular alterations on fertility, reproductive health, and life span is far from being fully revealed. This work discusses the current knowledge on the impact of aging in the testis, particularly of aging-related dysregulated inflammation and oxidative damage on the functioning of its different cell populations. More interestingly, this review covers the potential benefits of anti-aging interventions and therapies using either pharmacological compounds (such as non-selective non-steroidal anti-inflammatory medication) or more natural alternatives (such as various nutraceuticals or even probiotics) that exhibit anti-inflammatory, antioxidant, and anti-apoptotic properties. Some of these are currently being investigated or are already in clinical use to delay or prevent testicular aging.
Collapse
Affiliation(s)
- María Eugenia Matzkin
- Instituto de Biología y Medicina Experimental, CONICET, Ciudad de Buenos Aires C1428ADN, Argentina; (R.S.C.); (S.P.R.); (M.B.F.)
- Departamento de Bioquímica Humana, Cátedra I, Facultad de Medicina, Universidad de Buenos Aires, Ciudad de Buenos Aires C1121ABG, Argentina
- Correspondence: ; Tel.: +54-114783-2869 (ext. 1209)
| | - Ricardo Saúl Calandra
- Instituto de Biología y Medicina Experimental, CONICET, Ciudad de Buenos Aires C1428ADN, Argentina; (R.S.C.); (S.P.R.); (M.B.F.)
| | - Soledad Paola Rossi
- Instituto de Biología y Medicina Experimental, CONICET, Ciudad de Buenos Aires C1428ADN, Argentina; (R.S.C.); (S.P.R.); (M.B.F.)
- Departamento de Bioquímica Humana, Cátedra I, Facultad de Medicina, Universidad de Buenos Aires, Ciudad de Buenos Aires C1121ABG, Argentina
| | - Andrzej Bartke
- Geriatrics Research, Department of Internal Medicine, School of Medicine, Southern Illinois University, Springfield, IL 62794, USA;
| | - Mónica Beatriz Frungieri
- Instituto de Biología y Medicina Experimental, CONICET, Ciudad de Buenos Aires C1428ADN, Argentina; (R.S.C.); (S.P.R.); (M.B.F.)
- Cátedra de Química, Ciclo Básico Común, Universidad de Buenos Aires, Ciudad de Buenos Aires C1405CAE, Argentina
| |
Collapse
|
11
|
Schneider A, Victoria B, Schiavon Cousen MI, Fang Y, McFadden S, Darcy J, Gesing A, Hascup ER, Hascup KN, Bartke A, Masternak MM. Growth hormone signaling shapes the impact of environmental temperature on transcriptomic profile of different adipose tissue depots in male mice. J Gerontol A Biol Sci Med Sci 2021; 77:941-946. [PMID: 34614153 PMCID: PMC9071461 DOI: 10.1093/gerona/glab291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Indexed: 11/14/2022] Open
Abstract
Growth hormone receptor knockout (GHRKO) mice are smaller, long living and have an increased metabolic rate compared with normal (N) littermates. However, it is known that thermoneutral conditions (30°-32°C) elicit metabolic adaptations in mice, increasing the metabolic rate. Therefore, we hypothesized that environmental temperature would affect the expression profile of different adipose tissue depots in GHRKO mice. For this, N (n=12) and GHRKO (n=11) male mice were maintained at 23°C or 30°C from weaning until 11 months of age. RNA sequencing from adipose tissue depots (epididymal - eWAT, perirenal - pWAT, subcutaneous - sWAT and brown fat - BAT) was performed. Thermoneutrality increased body weight gain in GHRKO but not N mice. Only a few genes were commonly regulated by temperature in N and GHRKO mice. The BAT was the most responsive to changes in temperature in both N and GHRKO mice. BAT Ucp1 and Ucp3 expression were decreased to a similar extent in both N and GHRKO mice under thermoneutrality. In contrast, eWAT was mostly unresponsive to changes in temperature. The response to thermoneutrality in GHRKO mice was most divergent from N mice in sWAT. Relative weight of sWAT was almost four times greater in GHRKO mice. Very few genes were regulated in N mice sWAT when compared to GHRKO mice. This suggest that this WAT depot has a central role in the adaptation of GHRKO mice to changes in temperature.
Collapse
Affiliation(s)
- Augusto Schneider
- Faculdade de Nutrição, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Berta Victoria
- College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL
| | | | - Yimin Fang
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer's Research and Treatment, Depts of Neurology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Samuel McFadden
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer's Research and Treatment, Depts of Neurology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Justin Darcy
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Adam Gesing
- Department of Endocrinology of Ageing, Medical University of Lodz, Poland
| | - Erin R Hascup
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer's Research and Treatment, Depts of Neurology, Southern Illinois University School of Medicine, Springfield, IL, USA.,Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Kevin N Hascup
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer's Research and Treatment, Depts of Neurology, Southern Illinois University School of Medicine, Springfield, IL, USA.,Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA.,Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Andrzej Bartke
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer's Research and Treatment, Depts of Neurology, Southern Illinois University School of Medicine, Springfield, IL, USA.,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.,Department of Head and Neck Surgery, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
12
|
Frungieri MB, Calandra RS, Bartke A, Matzkin ME. Male and female gonadal ageing: its impact on health span and life span. Mech Ageing Dev 2021; 197:111519. [PMID: 34139215 DOI: 10.1016/j.mad.2021.111519] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 06/03/2021] [Accepted: 06/08/2021] [Indexed: 02/07/2023]
Abstract
Ageing is linked to changes in the hypothalamic-pituitary-gonadal axis and a progressive decline in gonadal function. While women become infertile when they enter menopause, fertility decline in ageing men does not necessarily involve a complete cessation of spermatogenesis. Gonadal dysfunction in elderly people is characterized by morphological, endocrine and metabolic alterations affecting the reproductive function and quality of life. With advancing age, sexuality turns into a critical emotional and physical factor actually defining the number of years that ageing people live a healthy life. Gonadal ageing correlates with comorbidities and an increased risk of age-related diseases including diabetes, kidney problems, cardiovascular failures and cancer. This article briefly summarizes the current state of knowledge on ovarian and testicular senescence, explores the experimental models used in the study of gonadal ageing, and describes the local pro-inflammatory, oxidative and apoptotic events and the associated signalling pathways that take place in the gonads while people get older. Overall, literature reports that ageing exacerbates a mutual crosstalk among oxidative stress, apoptosis and the inflammatory response in the gonads leading to detrimental effects on fertility. Data also highlight the clinical implications of novel therapeutic interventions using antioxidant, anti-apoptotic and anti-inflammatory drugs on health span and life span.
Collapse
Affiliation(s)
- Mónica B Frungieri
- Instituto de Biología y Medicina Experimental, CONICET, Ciudad de Buenos Aires, C1428ADN, Argentina; Cátedra de Química, Ciclo Básico Común, Ciudad de Buenos Aires, C1405CAE, Argentina.
| | - Ricardo S Calandra
- Instituto de Biología y Medicina Experimental, CONICET, Ciudad de Buenos Aires, C1428ADN, Argentina
| | - Andrzej Bartke
- Division of Geriatrics Research, Department of Internal Medicine, Southern Illinois University, School of Medicine, Springfield, IL 62702, USA
| | - María E Matzkin
- Instituto de Biología y Medicina Experimental, CONICET, Ciudad de Buenos Aires, C1428ADN, Argentina; Cátedra de Bioquímica Humana, Facultad de Medicina, Universidad de Buenos Aires, Ciudad de Buenos Aires, C1121ABG, Argentina
| |
Collapse
|
13
|
Fuentealba M, Fabian DK, Dönertaş HM, Thornton JM, Partridge L. Transcriptomic profiling of long- and short-lived mutant mice implicates mitochondrial metabolism in ageing and shows signatures of normal ageing in progeroid mice. Mech Ageing Dev 2021; 194:111437. [PMID: 33454277 PMCID: PMC7895802 DOI: 10.1016/j.mad.2021.111437] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 12/09/2020] [Accepted: 01/11/2021] [Indexed: 12/21/2022]
Abstract
Genetically modified mouse models of ageing are the living proof that lifespan and healthspan can be lengthened or shortened, and provide a powerful context in which to unravel the molecular mechanisms at work. In this study, we analysed and compared gene expression data from 10 long-lived and 8 short-lived mouse models of ageing. Transcriptome-wide correlation analysis revealed that mutations with equivalent effects on lifespan induce more similar transcriptomic changes, especially if they target the same pathway. Using functional enrichment analysis, we identified 58 gene sets with consistent changes in long- and short-lived mice, 55 of which were up-regulated in long-lived mice and down-regulated in short-lived mice. Half of these sets represented genes involved in energy and lipid metabolism, among which Ppargc1a, Mif, Aldh5a1 and Idh1 were frequently observed. Based on the gene sets with consistent changes, and also the whole transcriptome, the gene expression changes during normal ageing resembled the transcriptome of short-lived models, suggesting that accelerated ageing models reproduce partially the molecular changes of ageing. Finally, we identified new genetic interventions that may ameliorate ageing, by comparing the transcriptomes of 51 mouse mutants not previously associated with ageing to expression signatures of long- and short-lived mice and ageing-related changes.
Collapse
Affiliation(s)
- Matias Fuentealba
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, London, WC1E 6BT, UK; European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, CB10 1SD, UK
| | - Daniel K Fabian
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, London, WC1E 6BT, UK; European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, CB10 1SD, UK
| | - Handan Melike Dönertaş
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, CB10 1SD, UK
| | - Janet M Thornton
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, CB10 1SD, UK
| | - Linda Partridge
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, London, WC1E 6BT, UK; Max Planck Institute for Biology of Ageing, Cologne, Germany.
| |
Collapse
|
14
|
Endicott SJ, Boynton DN, Beckmann LJ, Miller RA. Long-lived mice with reduced growth hormone signaling have a constitutive upregulation of hepatic chaperone-mediated autophagy. Autophagy 2021; 17:612-625. [PMID: 32013718 PMCID: PMC8032237 DOI: 10.1080/15548627.2020.1725378] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 01/26/2020] [Accepted: 01/29/2020] [Indexed: 02/01/2023] Open
Abstract
Chaperone-mediated autophagy (CMA) is the most selective form of lysosomal proteolysis. CMA modulates proteomic organization through selective protein degradation, with targets including metabolic enzymes, cell growth regulators, and neurodegeneration-related proteins. CMA activity is low in ad libitum-fed rodents but is increased by prolonged fasting. AKT negatively regulates CMA at the lysosomal membrane by phosphorylating and inhibiting the CMA regulator GFAP. We have previously reported that long-lived Pou1f1/Pit1 mutant (Snell) mice and ghr (growth hormone receptor) knockout mice (ghr KO) have lower AKT activity when fed compared to littermate controls, suggesting the hypothesis that these mice have increased baseline CMA activity. Here, we report that liver lysosomes from fed Snell dwarf mice and ghr KO mice have decreased GFAP phosphorylation and increased CMA substrate uptake activity. Liver lysosomes isolated from fed Snell dwarf mice and ghr KO mice injected with the protease inhibitor leupeptin had increased accumulation of endogenous CMA substrates, compared to littermate controls, suggesting an increase in CMA in vivo. Mice with liver-specific ablation of GH (growth hormone) signaling did not have increased liver CMA, suggesting that a signaling effect resulting from a loss of growth hormone in another tissue causes enhanced CMA in Snell dwarf and ghr KO mice. Finally, we find Snell dwarf mice have decreased protein levels (in liver and kidney) of CIP2A, a well-characterized CMA target protein, without an associated change in Cip2a mRNA. Collectively, these data suggest that CMA is enhanced downstream of an endocrine change resulting from whole-body ablation of GH signaling.Abbreviations: CMA: chaperone-mediated autophagy; GH: growth hormone; ghr KO: growth hormone receptor knockout; LAMP2A: splice variant 1 of Lamp2 transcript; LC3-I: non-lipidated MAP1LC3; LC3-II: lipidated MAP1LC3; Li-ghr KO: liver-specific ghr knockout; MA: macroautophagy; MTORC1: mechanistic target of rapamycin kinase complex 1; MTORC2: mechanistic target of rapamycin kinase complex 2; PBS: phosphate-buffered saline.
Collapse
Affiliation(s)
- S. Joseph Endicott
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Dennis N. Boynton
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI, USA
| | - Logan J. Beckmann
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI, USA
| | - Richard A. Miller
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
- Institute of Gerontology, University of Michigan Geriatrics Center, Ann Arbor, MI, USA
| |
Collapse
|
15
|
Iwase H, Ball S, Adams K, Eyestone W, Walters A, Cooper DKC. Growth hormone receptor knockout: Relevance to xenotransplantation. Xenotransplantation 2020; 28:e12652. [PMID: 33058285 DOI: 10.1111/xen.12652] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 09/03/2020] [Accepted: 09/15/2020] [Indexed: 02/06/2023]
Abstract
Xenotransplantation research has made considerable progress in recent years, largely through the increasing availability of pigs with multiple genetic modifications, effective immunosuppressive therapy, and anti-inflammatory therapy to protect pig tissues from the primate immune and inflammatory responses and correct molecular incompatibilities. Further study is required regarding identification and investigation of physiological incompatibilities. Although the exact cause remains uncertain, we and others have observed relatively rapid growth of kidney xenografts after transplantation into nonhuman primates (NHPs). There has also been some evidence of growth, or at least ventricular hypertrophy, of the pig heart after orthotopic transplantation into NHPs. Rapid growth could be problematic, particularly with regard to the heart within the relatively restricted confines of the chest. It has been suggested that the problem of rapid growth of the pig organ after transplantation could be resolved by growth hormone receptor (GHR) gene knockout in the pig. The GHR, although most well-known for regulating growth, has many other biological functions, including regulating metabolism and controlling physiological processes. Genetically modified GHRKO pigs have recently become available. We provide data on their growth compared to comparable pigs that do not include GHRKO, and we have reviewed the literature regarding the effect of GHRKO, and its relevance to xenotransplantation.
Collapse
Affiliation(s)
- Hayato Iwase
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | | | | | - David K C Cooper
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
16
|
List EO, Duran-Ortiz S, Kopchick JJ. Effects of tissue-specific GH receptor knockouts in mice. Mol Cell Endocrinol 2020; 515:110919. [PMID: 32592744 DOI: 10.1016/j.mce.2020.110919] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 06/15/2020] [Accepted: 06/17/2020] [Indexed: 11/18/2022]
Abstract
Growth hormone (GH) is pituitary derived hormone which acts on most tissues of the body either directly or indirectly and affects many metabolic processes throughout life. Genetically engineered mouse lines have become vital tools for uncovering the various in vivo activities of a GH. A particularly useful mouse line has been the GH receptor (GHR) gene disrupted or knockout (KO) mouse which has been used world-wide in many studies. Recent advances in biotechnology have allowed the development of tissue-specific knockout mouse lines which allows for more direct enquiries on the activities of a given protein in specific tissues or cell types. Accordingly, twenty-two novel tissue-specific GHRKO mouse lines have been developed in the last eleven years. In this paper we provide a detailed list and review the phenotypic changes that occur in each of these tissue-specific GHRKO mouse lines.
Collapse
Affiliation(s)
- Edward O List
- Edison Biotechnology Institute, Ohio University, Athens, OH, 45701, USA; Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, 45701, USA
| | - Silvana Duran-Ortiz
- Edison Biotechnology Institute, Ohio University, Athens, OH, 45701, USA; Molecular and Cellular Biology Program, Department of Biomedical Sciences, Ohio University, Athens, OH, 45701, USA
| | - John J Kopchick
- Edison Biotechnology Institute, Ohio University, Athens, OH, 45701, USA; Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, 45701, USA; Molecular and Cellular Biology Program, Department of Biomedical Sciences, Ohio University, Athens, OH, 45701, USA.
| |
Collapse
|
17
|
Li X, Frazier JA, Spahiu E, McPherson M, Miller RA. Muscle-dependent regulation of adipose tissue function in long-lived growth hormone-mutant mice. Aging (Albany NY) 2020; 12:8766-8789. [PMID: 32464603 PMCID: PMC7288969 DOI: 10.18632/aging.103380] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 05/14/2020] [Indexed: 01/24/2023]
Abstract
Altered adipose tissue may contribute to the longevity of Snell dwarf and growth hormone receptor (GHR) knock-out mice. We report here that white (WAT) and brown (BAT) fat have elevated UCP1 in both kinds of mice, and that adipocytes in WAT depots turn beige/brown. These imply increased thermogenesis and are expected to lead to improved glucose control. Both kinds of long-lived mice show lower levels of inflammatory M1 macrophages and higher levels of anti-inflammatory M2 macrophages in BAT and WAT, with correspondingly lower levels of TNFα, IL-6, and MCP1. Experiments with mice with tissue-specific disruption of GHR showed that these adipocyte and macrophage changes were not due to hepatic IGF1 production nor to direct GH effects on adipocytes, but instead reflect GH effects on muscle. Muscles deprived of GH signals, either globally (GKO) or in muscle only (MKO), produce higher levels of circulating irisin and its precursor FNDC5. The data thus suggest that the changes in adipose tissue differentiation and inflammatory status seen in long-lived mutant mice reflect interruption of GH-dependent irisin inhibition, with consequential effects on metabolism and thermogenesis.
Collapse
Affiliation(s)
- Xinna Li
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, Michigan 48109, USA
| | - Jacquelyn A. Frazier
- College of Literature, Sciences, and The Arts, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Edward Spahiu
- College of Literature, Sciences, and The Arts, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Madaline McPherson
- College of Literature, Sciences, and The Arts, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Richard A. Miller
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, Michigan 48109, USA,University of Michigan Geriatrics Center, Ann Arbor, Michigan 48109, USA
| |
Collapse
|
18
|
Icyuz M, Fitch M, Zhang F, Challa A, Sun LY. Physiological and metabolic features of mice with CRISPR/Cas9-mediated loss-of-function in growth hormone-releasing hormone. Aging (Albany NY) 2020; 12:9761-9780. [PMID: 32422607 PMCID: PMC7288930 DOI: 10.18632/aging.103242] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 04/20/2020] [Indexed: 12/12/2022]
Abstract
Our previous study demonstrated that the loss of growth hormone releasing hormone (GHRH) results in increased lifespan and improved metabolic homeostasis in the mouse model generated by classical embryonic stem cell-based gene-targeting method. In this study, we targeted the GHRH gene using the CRISPR/Cas9 technology to avoid passenger alleles/mutations and performed in-depth physiological and metabolic characterization. In agreement with our previous observations, male and female GHRH-/- mice have significantly reduced body weight and enhanced insulin sensitivity when compared to wild type littermates. Dual-energy X-ray absorptiometry showed that there were significant decreases in lean mass, bone mineral content and density, and a dramatic increase in fat mass of GHRH-/- mice when compared to wild type littermates. Indirect calorimetry measurements showed dramatic reductions in oxygen consumption, carbon dioxide production and energy expenditure in GHRH-/- mice compared to wild type mice in both light and dark cycles. Respiratory exchange ratio was significantly lower in GHRH-/- mice during the light cycle, but not during the dark cycle, indicating a circadian related metabolic shift towards fat utilization in the growth hormone deficient mice. The novel CRISPR/Cas9 GHRH-/- mice are exhibiting the consistent and unique physiological and metabolic characteristics, which might mediate the longevity effects of growth hormone deficiency in mice.
Collapse
Affiliation(s)
- Mert Icyuz
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Michael Fitch
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Fang Zhang
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Anil Challa
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Liou Y. Sun
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| |
Collapse
|
19
|
Fang Y, McFadden S, Darcy J, Hascup ER, Hascup KN, Bartke A. Lifespan of long-lived growth hormone receptor knockout mice was not normalized by housing at 30°C since weaning. Aging Cell 2020; 19:e13123. [PMID: 32110850 PMCID: PMC7253058 DOI: 10.1111/acel.13123] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 01/22/2020] [Accepted: 01/30/2020] [Indexed: 12/18/2022] Open
Abstract
Growth hormone receptor knockout (GHRKO) mice are remarkably long-lived and have improved glucose homeostasis along with altered energy metabolism which manifests through decreased respiratory quotient (RQ) and increased oxygen consumption (VO2 ). Short-term exposure of these animals to increased environmental temperature (eT) at 30°C can normalize their VO2 and RQ. We hypothesized that increased heat loss in the diminutive GHRKO mice housed at 23°C and the consequent metabolic adjustments to meet the increased energy demand for thermogenesis may promote extension of longevity, and preventing these adjustments by chronic exposure to increased eT will reduce or eliminate their longevity advantage. To test these hypotheses, GHRKO mice were housed at increased eT (30°C) since weaning. Here, we report that contrasting with the effects of short-term exposure of adult GHRKO mice to 30°C, transferring juvenile GHRKO mice to chronic housing at 30°C did not normalize the examined parameters of energy metabolism and glucose homeostasis. Moreover, despite decreased expression levels of thermogenic genes in brown adipose tissue (BAT) and elevated core body temperature, the lifespan of male GHRKO mice was not reduced, while the lifespan of female GHRKO mice was increased, along with improved glucose homeostasis. The results indicate that GHRKO mice have intrinsic features that help maintain their delayed, healthy aging, and extended longevity at both 23°C and 30°C.
Collapse
Affiliation(s)
- Yimin Fang
- Department of NeurologySouthern Illinois University School of MedicineSpringfieldILUSA
| | - Samuel McFadden
- Department of NeurologySouthern Illinois University School of MedicineSpringfieldILUSA
| | - Justin Darcy
- Department of Internal MedicineSouthern Illinois University School of MedicineSpringfieldILUSA
- Present address:
Section on Integrative Physiology and MetabolismJoslin Diabetes CenterHarvard Medical SchoolBostonMAUSA
| | - Erin R. Hascup
- Department of NeurologySouthern Illinois University School of MedicineSpringfieldILUSA
- Department of PharmacologySouthern Illinois University School of MedicineSpringfieldILUSA
| | - Kevin N. Hascup
- Department of NeurologySouthern Illinois University School of MedicineSpringfieldILUSA
- Department of PharmacologySouthern Illinois University School of MedicineSpringfieldILUSA
- Department of Molecular Biology, Microbiology and BiochemistrySouthern Illinois University School of MedicineSpringfieldILUSA
| | - Andrzej Bartke
- Department of Internal MedicineSouthern Illinois University School of MedicineSpringfieldILUSA
| |
Collapse
|
20
|
Buffenstein R, Lewis KN, Gibney PA, Narayan V, Grimes KM, Smith M, Lin TD, Brown-Borg HM. Probing Pedomorphy and Prolonged Lifespan in Naked Mole-Rats and Dwarf Mice. Physiology (Bethesda) 2020; 35:96-111. [DOI: 10.1152/physiol.00032.2019] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Pedomorphy, maintenance of juvenile traits throughout life, is most pronounced in extraordinarily long-lived naked mole-rats. Many of these traits (e.g., slow growth rates, low hormone levels, and delayed sexual maturity) are shared with spontaneously mutated, long-lived dwarf mice. Although some youthful traits likely evolved as adaptations to subterranean habitats (e.g., thermolability), the nature of these intrinsic pedomorphic features may also contribute to their prolonged youthfulness, longevity, and healthspan.
Collapse
Affiliation(s)
| | | | - Patrick A. Gibney
- Calico Life Sciences LLC, South San Francisco, California
- Department of Food Science, College of Agriculture and Life Sciences, Stocking Hall, Cornell University, Ithaca, New York
| | - Vikram Narayan
- Calico Life Sciences LLC, South San Francisco, California
| | - Kelly M. Grimes
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Megan Smith
- Calico Life Sciences LLC, South San Francisco, California
| | - Tzuhua D. Lin
- Calico Life Sciences LLC, South San Francisco, California
| | - Holly M. Brown-Borg
- Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota
| |
Collapse
|
21
|
Liu Z, Solesio ME, Schaffler MB, Frikha-Benayed D, Rosen CJ, Werner H, Kopchick JJ, Pavlov EV, Abramov AY, Yakar S. Mitochondrial Function Is Compromised in Cortical Bone Osteocytes of Long-Lived Growth Hormone Receptor Null Mice. J Bone Miner Res 2019; 34:106-122. [PMID: 30216544 PMCID: PMC7080402 DOI: 10.1002/jbmr.3573] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 08/03/2018] [Accepted: 08/08/2018] [Indexed: 12/12/2022]
Abstract
Despite increased longevity and resistance to multiple stressors, growth hormone receptor null (GHRKO) mice exhibit severe skeletal impairment. The role of GHR in maintaining osteocyte mitochondrial function is unknown. We found that GHR ablation was detrimental to osteocyte mitochondrial function. In vivo multiphoton microscopy revealed significant reductions of >10% in mitochondrial membrane potential (MMP) in GHRKO osteocytes and reduced mitochondrial volumetric density. Reductions in MMP were accompanied by reductions in glucose transporter-1 levels, steady state ATP, NADH redox index, oxygen consumption rate, and mitochondrial reserve capacity in GHRKO osteocytes. Glycolytic capacity did not differ between control and GHRKO males' osteocytes. However, osteocytes from aged female GHRKO mice exhibited reductions in glycolytic parameters, indicating impairments in glucose metabolism, which may be sex dependent. GHRKO osteocytes exhibited increased levels of cytoplasmic reactive oxygen species (ROS) (both basal and in response to high glucose), insulin-like growth factor-1 (IGF-1), and insulin. Mitochondrial ROS levels were increased and correlated with reduced glutathione in GHRKO osteocytes. Overall, the compromised osteocyte mitochondrial function and responses to metabolic insults strongly correlated with skeletal impairments, suggesting that despite increased life span of the GHRKO mice, skeletal health span is decreased. © 2018 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Zhongbo Liu
- David B. Kriser Dental Center, Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, USA
| | - Maria E Solesio
- David B. Kriser Dental Center, Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, USA
| | - Mitchell B Schaffler
- Department of Biomedical Engineering, City College of New York, New York, NY, USA
| | - Dorra Frikha-Benayed
- Department of Biomedical Engineering, City College of New York, New York, NY, USA
| | | | - Haim Werner
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - John J Kopchick
- Edison Biotechnology Institute and Department of Biomedical Sciences, Ohio University, Athens, OH, USA
| | - Evgeny V Pavlov
- David B. Kriser Dental Center, Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, USA
| | - Andrey Y Abramov
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, UK
| | - Shoshana Yakar
- David B. Kriser Dental Center, Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, USA
| |
Collapse
|
22
|
Grant AD, Wilsterman K, Smarr BL, Kriegsfeld LJ. Evidence for a Coupled Oscillator Model of Endocrine Ultradian Rhythms. J Biol Rhythms 2018; 33:475-496. [PMID: 30132387 DOI: 10.1177/0748730418791423] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Whereas long-period temporal structures in endocrine dynamics have been well studied, endocrine rhythms on the scale of hours are relatively unexplored. The study of these ultradian rhythms (URs) has remained nascent, in part, because a theoretical framework unifying ultradian patterns across systems has not been established. The present overview proposes a conceptual coupled oscillator network model of URs in which oscillating hormonal outputs, or nodes, are connected by edges representing the strength of node-node coupling. We propose that variable-strength coupling exists both within and across classic hormonal axes. Because coupled oscillators synchronize, such a model implies that changes across hormonal systems could be inferred by surveying accessible nodes in the network. This implication would at once simplify the study of URs and open new avenues of exploration into conditions affecting coupling. In support of this proposed framework, we review mammalian evidence for (1) URs of the gut-brain axis and the hypothalamo-pituitary-thyroid, -adrenal, and -gonadal axes, (2) UR coupling within and across these axes; and (3) the relation of these URs to body temperature. URs across these systems exhibit behavior broadly consistent with a coupled oscillator network, maintaining both consistent URs and coupling within and across axes. This model may aid the exploration of mammalian physiology at high temporal resolution and improve the understanding of endocrine system dynamics within individuals.
Collapse
Affiliation(s)
- Azure D Grant
- The Helen Wills Neuroscience Institute, University of California, Berkeley, California
| | - Kathryn Wilsterman
- Department of Integrative Biology, University of California, Berkeley, California
| | - Benjamin L Smarr
- Department of Psychology, University of California, Berkeley, California
| | - Lance J Kriegsfeld
- The Helen Wills Neuroscience Institute, University of California, Berkeley, California.,Department of Psychology, University of California, Berkeley, California
| |
Collapse
|
23
|
Sparkman AM, Clark AD, Brummett LJ, Chism KR, Combrink LL, Kabey NM, Schwartz TS. Convergence in reduced body size, head size, and blood glucose in three island reptiles. Ecol Evol 2018; 8:6169-6182. [PMID: 29988440 PMCID: PMC6024148 DOI: 10.1002/ece3.4171] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 04/17/2018] [Accepted: 04/23/2018] [Indexed: 12/12/2022] Open
Abstract
Many oceanic islands harbor diverse species that differ markedly from their mainland relatives with respect to morphology, behavior, and physiology. A particularly common morphological change exhibited by a wide range of species on islands worldwide involves either a reduction in body size, termed island dwarfism, or an increase in body size, termed island gigantism. While numerous instances of dwarfism and gigantism have been well documented, documentation of other morphological changes on islands remains limited. Furthermore, we lack a basic understanding of the physiological mechanisms that underlie these changes, and whether they are convergent. A major hypothesis for the repeated evolution of dwarfism posits selection for smaller, more efficient body sizes in the context of low resource availability. Under this hypothesis, we would expect the physiological mechanisms known to be downregulated in model organisms exhibiting small body sizes due to dietary restriction or artificial selection would also be downregulated in wild species exhibiting dwarfism on islands. We measured body size, relative head size, and circulating blood glucose in three species of reptiles-two snakes and one lizard-in the California Channel Islands relative to mainland populations. Collating data from 6 years of study, we found that relative to mainland population the island populations had smaller body size (i.e., island dwarfism), smaller head sizes relative to body size, and lower levels of blood glucose, although with some variation by sex and year. These findings suggest that the island populations of these three species have independently evolved convergent physiological changes (lower glucose set point) corresponding to convergent changes in morphology that are consistent with a scenario of reduced resource availability and/or changes in prey size on the islands. This provides a powerful system to further investigate ecological, physiological, and genetic variables to elucidate the mechanisms underlying convergent changes in life history on islands.
Collapse
Affiliation(s)
| | - Amanda D. Clark
- Department of Biological SciencesAuburn UniversityAuburnAlabama
| | | | | | | | - Nicole M. Kabey
- Department of BiologyWestmont CollegeSanta BarbaraCalifornia
| | | |
Collapse
|
24
|
Basu R, Qian Y, Kopchick JJ. MECHANISMS IN ENDOCRINOLOGY: Lessons from growth hormone receptor gene-disrupted mice: are there benefits of endocrine defects? Eur J Endocrinol 2018; 178:R155-R181. [PMID: 29459441 DOI: 10.1530/eje-18-0018] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 02/19/2018] [Indexed: 12/12/2022]
Abstract
Growth hormone (GH) is produced primarily by anterior pituitary somatotroph cells. Numerous acute human (h) GH treatment and long-term follow-up studies and extensive use of animal models of GH action have shaped the body of GH research over the past 70 years. Work on the GH receptor (R)-knockout (GHRKO) mice and results of studies on GH-resistant Laron Syndrome (LS) patients have helped define many physiological actions of GH including those dealing with metabolism, obesity, cancer, diabetes, cognition and aging/longevity. In this review, we have discussed several issues dealing with these biological effects of GH and attempt to answer the question of whether decreased GH action may be beneficial.
Collapse
Affiliation(s)
- Reetobrata Basu
- Edison Biotechnology Institute, Ohio University, Athens, Ohio, USA
| | - Yanrong Qian
- Edison Biotechnology Institute, Ohio University, Athens, Ohio, USA
| | - John J Kopchick
- Edison Biotechnology Institute, Ohio University, Athens, Ohio, USA
- Ohio University Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA
| |
Collapse
|
25
|
Paul BD, Snyder SH. Gasotransmitter hydrogen sulfide signaling in neuronal health and disease. Biochem Pharmacol 2018; 149:101-109. [PMID: 29203369 PMCID: PMC5868969 DOI: 10.1016/j.bcp.2017.11.019] [Citation(s) in RCA: 167] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 11/29/2017] [Indexed: 01/17/2023]
Abstract
Hydrogen sulfide is a gaseous signaling molecule or gasotransmitter which plays important roles in a wide spectrum of physiologic processes in the brain and peripheral tissues. Unlike nitric oxide and carbon monoxide, the other major gasotransmitters, research on hydrogen sulfide is still in its infancy. One of the modes by which hydrogen sulfide signals is via a posttranslational modification termed sulfhydration/persulfidation, which occurs on reactive cysteine residues on target proteins, where the reactive SH group is converted to an SSH group. Sulfhydration is a substantially prevalent modification, which modulates the structure or function of proteins being modified. Thus, precise control of endogenous hydrogen sulfide production and metabolism is critical for maintenance of optimal cellular function, with excess generation and paucity, both contributing to pathology. Dysregulation of the reverse transsulfuration pathway which generates hydrogen sulfide occurs in several neurodegenerative diseases such as Parkinson's disease, Huntington's disease and Alzheimer's disease. Accordingly, treatment with donors of hydrogen sulfide or stimulation of the reverse transsulfuration have proved beneficial in several neurodegenerative states. In this review we focus on hydrogen sulfide mediated neuronal signaling processes that contribute to neuroprotection.
Collapse
Affiliation(s)
- Bindu D Paul
- The Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Solomon H Snyder
- The Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Psychiatry, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
26
|
Insulin-like growth factor 1 receptor regulates hypothermia during calorie restriction. Proc Natl Acad Sci U S A 2017; 114:9731-9736. [PMID: 28827363 DOI: 10.1073/pnas.1617876114] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
When food resources are scarce, endothermic animals can lower core body temperature (Tb). This phenomenon is believed to be part of an adaptive mechanism that may have evolved to conserve energy until more food becomes available. Here, we found in the mouse that the insulin-like growth factor 1 receptor (IGF-1R) controls this response in the central nervous system. Pharmacological or genetic inhibition of IGF-1R enhanced the reduction of temperature and of energy expenditure during calorie restriction. Full blockade of IGF-1R affected female and male mice similarly. In contrast, genetic IGF-1R dosage was effective only in females, where it also induced transient and estrus-specific hypothermia in animals fed ad libitum. These effects were regulated in the brain, as only central, not peripheral, pharmacological activation of IGF-1R prevented hypothermia during calorie restriction. Targeted IGF-1R knockout selectively in forebrain neurons revealed that IGF signaling also modulates calorie restriction-dependent Tb regulation in regions rostral of the canonical hypothalamic nuclei involved in controlling body temperature. In aggregate, these data identify central IGF-1R as a mediator of the integration of nutrient and temperature homeostasis. They also show that calorie restriction, IGF-1R signaling, and body temperature, three of the main regulators of metabolism, aging, and longevity, are components of the same pathway.
Collapse
|
27
|
Hascup KN, Lynn MK, Fitzgerald PJ, Randall S, Kopchick JJ, Boger HA, Bartke A, Hascup ER. Enhanced Cognition and Hypoglutamatergic Signaling in a Growth Hormone Receptor Knockout Mouse Model of Successful Aging. J Gerontol A Biol Sci Med Sci 2017; 72:329-337. [PMID: 27208894 DOI: 10.1093/gerona/glw088] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 04/25/2016] [Indexed: 01/19/2023] Open
Abstract
Growth hormone receptor knockout (GHR-KO) mice are long lived with improved health span, making this an excellent model system for understanding biochemical mechanisms important to cognitive reserve. The purpose of the present study was to elucidate differences in cognition and glutamatergic dynamics between aged (20- to 24-month-old) GHR-KO and littermate controls. Glutamate plays a critical role in hippocampal learning and memory and is implicated in several neurodegenerative disorders, including Alzheimer's disease. Spatial learning and memory were assessed using the Morris water maze (MWM), whereas independent dentate gyrus (DG), CA3, and CA1 basal glutamate, release, and uptake measurements were conducted in isoflurane anesthetized mice utilizing an enzyme-based microelectrode array (MEA) coupled with constant potential amperometry. These MEAs have high temporal and low spatial resolution while causing minimal damage to the surrounding parenchyma. Littermate controls performed worse on the memory portion of the MWM behavioral task and had elevated DG, CA3, and CA1 basal glutamate and stimulus-evoked release compared with age-matched GHR-KO mice. CA3 basal glutamate negatively correlated with MWM performance. These results support glutamatergic regulation in learning and memory and may have implications for therapeutic targets to delay the onset of, or reduce cognitive decline, in Alzheimer's disease.
Collapse
Affiliation(s)
- Kevin N Hascup
- Department of Neurology, Center for Alzheimer's Disease and Related Disorders, Southern Illinois University School of Medicine, Springfield
| | - Mary K Lynn
- Department of Neuroscience, Medical University of South Carolina, Charleston
| | - Patrick J Fitzgerald
- Department of Neurology, Center for Alzheimer's Disease and Related Disorders, Southern Illinois University School of Medicine, Springfield
| | - Shari Randall
- Department of Neurology, Center for Alzheimer's Disease and Related Disorders, Southern Illinois University School of Medicine, Springfield
| | - John J Kopchick
- Edison Biotechnology Institute, Department of Biomedical Sciences, Ohio University, Athens
| | - Heather A Boger
- Department of Neuroscience, Medical University of South Carolina, Charleston.,Center on Aging, Medical University of South Carolina, Charleston
| | | | - Erin R Hascup
- Department of Neurology, Center for Alzheimer's Disease and Related Disorders, Southern Illinois University School of Medicine, Springfield.,Department of Pharmacology, Southern Illinois University School of Medicine, Springfield
| |
Collapse
|
28
|
TSH-independent release of thyroid hormones through cold exposure in aging rats. Oncotarget 2017; 8:89431-89438. [PMID: 29163760 PMCID: PMC5685681 DOI: 10.18632/oncotarget.19851] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 07/26/2017] [Indexed: 01/27/2023] Open
Abstract
Thyroid function decreases and cold exposure response becomes impaired with increasing age. We investigated the age-related changes in thyroid structure and function and cold-induced changes in the thyroid activity of aging rats. Thirty-two male Sprague–Dawley rats were randomly divided into four groups (8 rats per group): young (7 months) and old (22 months) groups exposed to room temperature and cold stress. The active follicle ratio and serum free T3, T4 and TSH, and TSH receptor (TSHR) concentrations in the thyroid tissues of the rats from each group were compared. At room temperature, old rats had significantly lower active follicle ratio and free T3 and T4 concentrations than young rats. Furthermore, old rats displayed higher TSH level than young. Exposure to cold temperature led to significantly increased active colloid ratio and free T3 and T4 concentrations among old rats, but no significant differences were found among young rats. Additionally, no significant changes in the TSH and TSHR levels were observed after cold exposure in both young and old rats. Old rats have lower thyroid function than young rats under normal temperature. Aging rats are more susceptible to cold stress than young rats, and cold-induced thyroid activation occurs independently of TSH. We investigated the age-related changes in the thyroid structure and function and cold-induced changes in the thyroid activity of aging rats. Aging rats have structurally less active thyroid follicles and functionally lower thyroid hormone levels than young rats. Furthermore, old rats are more susceptible to cold stress than young rats, and cold-induced thyroid activation occurs independently of TSH.
Collapse
|
29
|
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
|
30
|
Abstract
Reduced insulin-like growth factor 1/insulin signaling (IIS) has been linked to extended longevity in species ranging from yeast to mammals. In mammals, this is exemplified in Ames dwarf (Prop1df/df) mice, which have a 40%-60% increase in longevity (males and females, respectively) due to their recessive Prop1 loss-of-function mutation that results in lack of growth hormone (GH), thyroid-stimulating hormone and prolactin. Our laboratory has previously shown that Ames dwarf mice have functionally unique white adipose tissue (WAT) that improves, rather than impairs, insulin sensitivity. Because GH and thyroid hormone are integral to adipose tissue development and function, we hypothesized that brown adipose tissue (BAT) in Ames dwarf mice may also be functionally unique and/or enhanced. Here, we elaborate on our recent findings, which demonstrate that BAT is functionally enhanced in Ames dwarf mice, and suggest that BAT removal in these mice results in utilization of WAT depots as an energy source. We also discuss how our findings compare to those in other long-lived dwarf mice with altered IIS, which unlike Ames dwarf mice, are essentially euthyroid. Lastly, we provide some insights into the implications of these findings and discuss some of the necessary future work in this area.
Collapse
Affiliation(s)
- Justin Darcy
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, Illinois, USA
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, Illinois, USA
| | - Andrzej Bartke
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, Illinois, USA
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, Illinois, USA
| |
Collapse
|
31
|
Brewer RA, Gibbs VK, Smith DL. Targeting glucose metabolism for healthy aging. NUTRITION AND HEALTHY AGING 2016; 4:31-46. [PMID: 28035340 PMCID: PMC5166514 DOI: 10.3233/nha-160007] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Advancing age is the greatest single risk factor for numerous chronic diseases. Thus, the ability to target the aging process can facilitate improved healthspan and potentially lifespan. Lack of adequate glucoregulatory control remains a recurrent theme accompanying aging and chronic disease, while numerous longevity interventions result in maintenance of glucoregulatory control. In this review, we propose targeting glucose metabolism to enhance regulatory control as a means to ameliorate the aging process. We highlight that calorie restriction improves glucoregulatory control and extends both lifespan and healthspan in model organisms, but we also indicate more practical interventions (i.e., calorie restriction mimetics) are desirable for clinical application in humans. Of the calorie restriction mimetics being investigated, we focus on the type 2 diabetes drug acarbose, an α-glucosidase inhibitor that when taken with a meal, results in reduced enzymatic degradation and absorption of glucose from complex carbohydrates. We discuss alternatives to acarbose that yield similar physiologic effects and describe dietary sources (e.g., sweet potatoes, legumes, and berries) of bioactive compounds with α-glucosidase inhibitory activity. We indicate future research should include exploration of how non-caloric compounds like α-glucosidase inhibitors modify macronutrient metabolism prior to disease onset, which may guide nutritional/lifestyle interventions to support health and reduce age-related disease risk.
Collapse
Affiliation(s)
- Rachel A. Brewer
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Victoria K. Gibbs
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
- Nutrition Obesity Research Center, Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
- Comprehensive Center for Healthy Aging, University of Alabama at Birmingham, Birmingham, AL, USA
- Nathan Shock Center of Excellence in the Biology of Aging, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Daniel L. Smith
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
- Nutrition Obesity Research Center, Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
- Comprehensive Center for Healthy Aging, University of Alabama at Birmingham, Birmingham, AL, USA
- Nathan Shock Center of Excellence in the Biology of Aging, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
32
|
Darcy J, Fang Y, Hill CM, McFadden S, Sun LY, Bartke A. Original Research: Metabolic alterations from early life thyroxine replacement therapy in male Ames dwarf mice are transient. Exp Biol Med (Maywood) 2016; 241:1764-71. [PMID: 27190246 DOI: 10.1177/1535370216650292] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 04/25/2016] [Indexed: 01/03/2023] Open
Abstract
Ames dwarf mice are exceptionally long-lived due to a Prop1 loss of function mutation resulting in deficiency of growth hormone, thyroid-stimulating hormone and prolactin. Deficiency in thyroid-stimulating hormone and growth hormone leads to greatly reduced levels of circulating thyroid hormones and insulin-like growth factor 1, as well as a reduction in insulin secretion. Early life growth hormone replacement therapy in Ames dwarf mice significantly shortens their longevity, while early life thyroxine (T4) replacement therapy does not. Possible mechanisms by which early life growth hormone replacement therapy shortens longevity include deleterious effects on glucose homeostasis and energy metabolism, which are long lasting. A mechanism explaining why early life T4 replacement therapy does not shorten longevity remains elusive. Here, we look for a possible explanation as to why early life T4 replacement therapy does not impact longevity of Ames dwarf mice. We found that early life T4 replacement therapy increased body weight and advanced the age of sexual maturation. We also find that early life T4 replacement therapy does not impact glucose tolerance or insulin sensitivity, and any deleterious effects on oxygen consumption, respiratory quotient and heat production are transient. Lastly, we find that early life T4 replacement therapy has long-lasting effects on bone mineral density and bone mineral content. We suggest that the transient effects on energy metabolism and lack of effects on glucose homeostasis are the reasons why there is no shortening of longevity after early life T4 replacement therapy in Ames dwarf mice.
Collapse
Affiliation(s)
- Justin Darcy
- Department of Internal Medicine, Geriatrics Research, Southern Illinois University School of Medicine, Springfield, IL 62794, USA Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL 62794, USA
| | - Yimin Fang
- Department of Internal Medicine, Geriatrics Research, Southern Illinois University School of Medicine, Springfield, IL 62794, USA
| | - Cristal M Hill
- Department of Internal Medicine, Geriatrics Research, Southern Illinois University School of Medicine, Springfield, IL 62794, USA Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL 62794, USA
| | - Sam McFadden
- Department of Internal Medicine, Geriatrics Research, Southern Illinois University School of Medicine, Springfield, IL 62794, USA
| | - Liou Y Sun
- Department of Internal Medicine, Geriatrics Research, Southern Illinois University School of Medicine, Springfield, IL 62794, USA
| | - Andrzej Bartke
- Department of Internal Medicine, Geriatrics Research, Southern Illinois University School of Medicine, Springfield, IL 62794, USA
| |
Collapse
|
33
|
McIsaac RS, Lewis KN, Gibney PA, Buffenstein R. From yeast to human: exploring the comparative biology of methionine restriction in extending eukaryotic life span. Ann N Y Acad Sci 2016; 1363:155-70. [DOI: 10.1111/nyas.13032] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 01/25/2016] [Accepted: 01/27/2016] [Indexed: 12/19/2022]
|
34
|
Growth Hormone Influence on the Morphology and Size of the Mouse Meibomian Gland. J Ophthalmol 2016; 2016:5728071. [PMID: 26981277 PMCID: PMC4769763 DOI: 10.1155/2016/5728071] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 01/11/2016] [Accepted: 01/19/2016] [Indexed: 12/20/2022] Open
Abstract
Purpose. We hypothesize that growth hormone (GH) plays a significant role in the regulation of the meibomian gland. To test our hypothesis, we examined the influence of GH on mouse meibomian gland structure. Methods. We studied four groups of mice, including (1) bovine (b) GH transgenic mice with excess GH; (2) GH receptor (R) antagonist (A) transgenic mice with decreased GH; (3) GHR knockout (−/−) mice with no GH activity; and (4) wild type (WT) control mice. After mouse sacrifice, eyelids were processed for morphological and image analyses. Results. Our results show striking structural changes in the GH-deficient animals. Many of the GHR−/− and GHA meibomian glands featured hyperkeratinized and thickened ducts, acini inserting into duct walls, and poorly differentiated acini. In contrast, the morphology of WT and bGH meibomian glands appeared similar. The sizes of meibomian glands of bGH mice were significantly larger and those of GHA and GHR−/− mice were significantly smaller than glands of WT mice. Conclusions. Our findings support our hypothesis that the GH/IGF-1 axis plays a significant role in the control of the meibomian gland. In addition, our data show that GH modulates the morphology and size of this tissue.
Collapse
|
35
|
Basso A, Del Bello G, Piacenza F, Giacconi R, Costarelli L, Malavolta M. Circadian rhythms of body temperature and locomotor activity in aging BALB/c mice: early and late life span predictors. Biogerontology 2016; 17:703-14. [PMID: 26820297 DOI: 10.1007/s10522-016-9635-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 01/20/2016] [Indexed: 01/16/2023]
Abstract
Impairment of one or more parameters of circadian rhythms (CR) of body temperature (BT) and locomotor activity (LMA) are considered among the hallmarks of mammalian aging. These alterations are frequently used as markers for imminent death in laboratory mice. However, there are still contradictory data for particular strains and it is also uncertain which changes might predict senescence changes later in life, including the force of mortality. In the present paper we use telemetry to study LMA and CR of BT during aging of BALB/c mice. At our knowledge this is the first time that CR of BT and LMA are investigated in this strain in a range of age covering the whole lifespan, from young adult up to very old age. CR of BT was analyzed with a cosine model using a cross sectional approach and follow-up measurements. The results show that BT, LMA, amplitude, goodness-of-fit (GoF) to circadian cycle of temperature decrease with different shapes during chronological age. Moreover, we found that the % change of amplitude and BT in early life (5-19 months) can predict the remaining lifespan of the mice. Later in life (22-32 months), best predictors are single measurements of LMA and GoF. The results of this study also offer potential measures to rapidly identifying freely unrestrained mice with the worst longitudinal outcome and against which existing or novel biomarkers and treatments may be assessed.
Collapse
Affiliation(s)
- Andrea Basso
- Nutrition and Aging Centre, Scientific and Technological Pole - INRCA - National Institute of Health and Sciences on Ageing, Via Birarelli 8, 60121, Ancona, Italy
| | - Giovanna Del Bello
- Nutrition and Aging Centre, Scientific and Technological Pole - INRCA - National Institute of Health and Sciences on Ageing, Via Birarelli 8, 60121, Ancona, Italy
| | - Francesco Piacenza
- Nutrition and Aging Centre, Scientific and Technological Pole - INRCA - National Institute of Health and Sciences on Ageing, Via Birarelli 8, 60121, Ancona, Italy
| | - Robertina Giacconi
- Nutrition and Aging Centre, Scientific and Technological Pole - INRCA - National Institute of Health and Sciences on Ageing, Via Birarelli 8, 60121, Ancona, Italy
| | - Laura Costarelli
- Nutrition and Aging Centre, Scientific and Technological Pole - INRCA - National Institute of Health and Sciences on Ageing, Via Birarelli 8, 60121, Ancona, Italy
| | - Marco Malavolta
- Nutrition and Aging Centre, Scientific and Technological Pole - INRCA - National Institute of Health and Sciences on Ageing, Via Birarelli 8, 60121, Ancona, Italy.
| |
Collapse
|
36
|
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
|
37
|
Comisford R, Lubbers ER, Householder LA, Suer O, Tchkonia T, Kirkland JL, List EO, Kopchick JJ, Berryman DE. Growth Hormone Receptor Antagonist Transgenic Mice Have Increased Subcutaneous Adipose Tissue Mass, Altered Glucose Homeostasis and No Change in White Adipose Tissue Cellular Senescence. Gerontology 2015; 62:163-72. [PMID: 26372907 DOI: 10.1159/000439050] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 07/29/2015] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Growth hormone (GH)-resistant/deficient mice experience improved glucose homeostasis and substantially increased lifespan. Recent evidence suggests that long-lived GH-resistant/deficient mice are protected from white adipose tissue (WAT) dysfunction, including WAT cellular senescence, impaired adipogenesis and loss of subcutaneous WAT in old age. This preservation of WAT function has been suggested to be a potential mechanism for the extended lifespan of these mice. OBJECTIVE The objective of this study was to examine WAT senescence, WAT distribution and glucose homeostasis in dwarf GH receptor antagonist (GHA) transgenic mice, a unique mouse strain having decreased GH action but normal longevity. METHODS 18-month-old female GHA mice and wild-type (WT) littermate controls were used. Prior to dissection, body composition, fasting blood glucose as well as glucose and insulin tolerance tests were performed. WAT distribution was determined by weighing four distinct WAT depots at the time of dissection. Cellular senescence in four WAT depots was assessed using senescence-associated β-galactosidase staining to quantify the senescent cell burden, and real-time qPCR to quantify gene expression of senescence markers p16 and IL-6. RESULTS GHA mice had a 22% reduction in total body weight, a 33% reduction in lean mass and a 10% increase in body fat percentage compared to WT controls. GHA mice had normal fasting blood glucose and improved insulin sensitivity; however, they exhibited impaired glucose tolerance. Moreover, GHA mice displayed enhanced lipid storage in the inguinal subcutaneous WAT depot (p < 0.05) and a 1.7-fold increase in extra-/intraperitoneal WAT ratio compared to controls (p < 0.05). Measurements of WAT cellular senescence showed no difference between GHA mice and WT controls. CONCLUSIONS Similar to other mice with decreased GH action, female GHA mice display reduced age-related lipid redistribution and improved insulin sensitivity, but no change in cellular senescence. The similar abundance of WAT senescent cells in GHA and control mice suggests that any protection against generation of senescent cells afforded by decreased GH action, low insulin-like growth factor 1 and/or improved insulin sensitivity in the GHA mice may be offset by their severe adiposity, since obesity is known to increase senescence.
Collapse
Affiliation(s)
- Ross Comisford
- Edison Biotechnology Institute, Ohio University, Athens, Ohio, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Li M, Reynolds CM, Gray C, Vickers MH. Preweaning GH Treatment Normalizes Body Growth Trajectory and Reverses Metabolic Dysregulation in Adult Offspring After Maternal Undernutrition. Endocrinology 2015; 156:3228-38. [PMID: 25993526 DOI: 10.1210/en.2015-1041] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Maternal undernutrition (UN) results in growth disorders and metabolic dysfunction in offspring. Although dysregulation of the GH-IGF axis in offspring is a known consequence of maternal UN, little is known about the efficacy of GH treatment during the period of developmental plasticity on later growth and metabolic outcomes. The present study investigated the effect of preweaning GH treatment on growth, glucose metabolism, and the GH-IGF axis in adult male and female offspring after maternal UN. Female Sprague Dawley rats were fed either a chow diet ad libitum (control [CON]) or 50% of ad libitum (UN) throughout pregnancy. From postnatal day 3, CON and UN pups received either saline (CON-S and UN-S) or GH (2.5 μg/g·d CON-GH and UN-GH) daily throughout lactation. At weaning, male and female offspring were randomly selected from each litter and fed a standard chow diet for the remainder of the study. Preweaning GH treatment normalized maternal UN-induced alterations in postweaning growth trajectory and concomitant adiposity in offspring. Plasma leptin concentrations were increased in UN-S offspring and normalized in the UN-GH group. Hepatic GH receptor expression was significantly elevated in UN-S offspring and normalized with GH treatment. Hepatic IGF binding protein-2 gene expression and plasma IGF-1 to IGF binding protein-3 ratio was reduced in UN-S offspring and elevated with GH treatment. GH treatment during a critical developmental window prevented maternal UN-induced changes in postnatal growth patterns and related adiposity, suggesting that manipulation of the GH-IGF-1 axis in early development may represent a promising avenue to prevent adverse developmental programming effects in adulthood.
Collapse
Affiliation(s)
- Minglan Li
- Liggins Institute and Gravida: National Centre for Growth and Development, University of Auckland, Auckland 1142, New Zealand
| | - Clare M Reynolds
- Liggins Institute and Gravida: National Centre for Growth and Development, University of Auckland, Auckland 1142, New Zealand
| | - Clint Gray
- Liggins Institute and Gravida: National Centre for Growth and Development, University of Auckland, Auckland 1142, New Zealand
| | - Mark H Vickers
- Liggins Institute and Gravida: National Centre for Growth and Development, University of Auckland, Auckland 1142, New Zealand
| |
Collapse
|
39
|
Keil G, Cummings E, de Magalhães JP. Being cool: how body temperature influences ageing and longevity. Biogerontology 2015; 16:383-97. [PMID: 25832892 PMCID: PMC4486781 DOI: 10.1007/s10522-015-9571-2] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 03/24/2015] [Indexed: 12/12/2022]
Abstract
Temperature is a basic and essential property of any physical system, including living systems. Even modest variations in temperature can have profound effects on organisms, and it has long been thought that as metabolism increases at higher temperatures so should rates of ageing. Here, we review the literature on how temperature affects longevity, ageing and life history traits. From poikilotherms to homeotherms, there is a clear trend for lower temperature being associated with longer lifespans both in wild populations and in laboratory conditions. Many life-extending manipulations in rodents, such as caloric restriction, also decrease core body temperature. Nonetheless, an inverse relationship between temperature and lifespan can be obscured or reversed, especially when the range of body temperatures is small as in homeotherms. An example is observed in humans: women appear to have a slightly higher body temperature and yet live longer than men. The mechanisms involved in the relationship between temperature and longevity also appear to be less direct than once thought with neuroendocrine processes possibly mediating complex physiological responses to temperature changes. Lastly, we discuss species differences in longevity in mammals and how this relates to body temperature and argue that the low temperature of the long-lived naked mole-rat possibly contributes to its exceptional longevity.
Collapse
Affiliation(s)
- Gerald Keil
- Integrative Genomics of Ageing Group, Institute of Integrative Biology, University of Liverpool, Liverpool, L69 7ZB UK
| | - Elizabeth Cummings
- Integrative Genomics of Ageing Group, Institute of Integrative Biology, University of Liverpool, Liverpool, L69 7ZB UK
| | - João Pedro de Magalhães
- Integrative Genomics of Ageing Group, Institute of Integrative Biology, University of Liverpool, Liverpool, L69 7ZB UK
| |
Collapse
|
40
|
Thermosensation and longevity. J Comp Physiol A Neuroethol Sens Neural Behav Physiol 2015; 201:857-67. [PMID: 26101089 DOI: 10.1007/s00359-015-1021-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2014] [Revised: 06/06/2015] [Accepted: 06/08/2015] [Indexed: 12/25/2022]
Abstract
Temperature has profound effects on behavior and aging in both poikilotherms and homeotherms. To thrive under the ever fluctuating environmental temperatures, animals have evolved sophisticated mechanisms to sense and adapt to temperature changes. Animals sense temperature through various molecular thermosensors, such as thermosensitive transient receptor potential (TRP) channels expressed in neurons, keratinocytes, and intestine. These evolutionarily conserved thermosensitive TRP channels feature distinct activation thresholds, thereby covering a wide spectrum of ambient temperature. Temperature changes trigger complex thermosensory behaviors. Due to the simplicity of the nervous system in model organisms such as Caenorhabditis elegans and Drosophila, the mechanisms of thermosensory behaviors in these species have been extensively studied at the circuit and molecular levels. While much is known about temperature regulation of behavior, it remains largely unclear how temperature affects aging. Recent studies in C. elegans demonstrate that temperature modulation of longevity is not simply a passive thermodynamic phenomenon as suggested by the rate-of-living theory, but rather a process that is actively regulated by genes, including those encoding thermosensitive TRP channels. In this review, we discuss our current understanding of thermosensation and its role in aging.
Collapse
|
41
|
Arum O, Dawson JA, Smith DL, Kopchick JJ, Allison DB, Bartke A. Do altered energy metabolism or spontaneous locomotion 'mediate' decelerated senescence? Aging Cell 2015; 14:483-90. [PMID: 25720347 PMCID: PMC4406677 DOI: 10.1111/acel.12318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2015] [Indexed: 12/01/2022] Open
Abstract
That one or multiple measures of metabolic rate may be robustly associated with, or possibly even causative of, the progression of aging-resultant phenotypes such as lifespan is a long-standing, well-known mechanistic hypothesis. To broach this hypothesis, we assessed metabolic function and spontaneous locomotion in two genetic and one dietary mouse models for retarded aging, and subjected the data to mediation analyses to determine whether any metabolic or locomotor trait could be identified as a mediator of the effect of any of the interventions on senescence. We do not test the hypothesis of causality (which would require some experiments), but instead test whether the correlation structure of certain variables is consistent with one possible pathway model in which a proposed mediating variable has a causal role. Results for metabolic measures, including oxygen consumption and respiratory quotient, failed to support this hypothesis; similar negative results were obtained for three behavioral motion metrics. Therefore, our mediation analyses did not find support that any of these correlates of decelerated senescence was a substantial mediator of the effect of either of these genetic alterations (with or without caloric restriction) on longevity. Further studies are needed to relate the examined phenotypic characteristics to mechanisms of aging and control of longevity.
Collapse
Affiliation(s)
- Oge Arum
- Department of Internal Medicine Southern Illinois University‐School of Medicine Springfield IL 62794 USA
| | - John Alexander Dawson
- Section on Statistical Genetics Department of Biostatistics School of Public Health University of Alabama at Birmingham 1665 University Blvd RPHB 140J Birmingham AL 35294‐0022 USA
| | - Daniel Larry Smith
- Nutrition Obesity Research Center Department of Nutrition Sciences University of Alabama at Birmingham Birmingham AL 35294‐0022 USA
| | - John J. Kopchick
- Department of Biomedical Sciences Edison Biotechnology Institute Heritage College of Osteopathic Medicine Ohio University Athens OH 45701 USA
| | - David B. Allison
- Section on Statistical Genetics Department of Biostatistics School of Public Health University of Alabama at Birmingham 1665 University Blvd RPHB 140J Birmingham AL 35294‐0022 USA
- Nutrition Obesity Research Center Department of Nutrition Sciences University of Alabama at Birmingham Birmingham AL 35294‐0022 USA
| | - Andrzej Bartke
- Department of Internal Medicine Southern Illinois University‐School of Medicine Springfield IL 62794 USA
| |
Collapse
|
42
|
Kopchick JJ, List EO, Kelder B, Gosney ES, Berryman DE. Evaluation of growth hormone (GH) action in mice: discovery of GH receptor antagonists and clinical indications. Mol Cell Endocrinol 2014; 386:34-45. [PMID: 24035867 PMCID: PMC3943600 DOI: 10.1016/j.mce.2013.09.004] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Revised: 08/29/2013] [Accepted: 09/03/2013] [Indexed: 11/28/2022]
Abstract
The discovery of a growth hormone receptor antagonist (GHA) was initially established via expression of mutated GH genes in transgenic mice. Following this discovery, development of the compound resulted in a drug termed pegvisomant, which has been approved for use in patients with acromegaly. Pegvisomant treatment in a dose dependent manner results in normalization of IGF-1 levels in most patients. Thus, it is a very efficacious and safe drug. Since the GH/IGF-1 axis has been implicated in the progression of several types of cancers, many have suggested the use of pegvisomant as an anti-cancer therapeutic. In this manuscript, we will review the use of mouse strains that possess elevated or depressed levels of GH action for unraveling many of GH actions. Additionally, we will describe experiments in which the GHA was discovered, review results of pegvisomant's preclinical and clinical trials, and provide data suggesting pegvisomant's therapeutic value in selected types of cancer.
Collapse
Affiliation(s)
- John J Kopchick
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, United States; Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, United States.
| | - Edward O List
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, United States; Department of Specialty Medicine, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, United States
| | - Bruce Kelder
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, United States; Department of Pediatrics, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, United States
| | - Elahu S Gosney
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, United States
| | - Darlene E Berryman
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, United States; Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, United States; School of Applied Health Sciences and Wellness, Ohio University, Athens, OH 45701, United States
| |
Collapse
|
43
|
Vasiliadis I, Kolovou G, Kolovou V, Giannakopoulou V, Boutsikou M, Katsiki N, Papadopoulou E, Mavrogeni S, Sorontila K, Pantos C, Cokkinos DV. Gene polymorphisms and thyroid function in patients with heart failure. Endocrine 2014; 45:46-54. [PMID: 23543433 DOI: 10.1007/s12020-013-9926-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2012] [Accepted: 03/12/2013] [Indexed: 01/08/2023]
Abstract
We evaluated nuclear factor kappa B {NFkB, rs28362491 [-94ins/delATTG (W/D)]} and angiotensin converting enzyme {ACE; rs1799752 [Ins(I)/Del(D)]} gene polymorphisms and their correlation with thyroid function in patients with heart failure (HF). Peak oxygen uptake (VO(2)) was evaluated (by Weber classification) during a symptom-limited cardiopulmonary exercise test in 194 patients. Thyroid-stimulating hormone, triiodothyronine (T3), thyroxine (T4), and free (F) T3 and FT4 were also measured. According to their cardiovascular (CV) capacity, patients were subdivided into four groups: group A included patients with peak VO(2) >20 ml/kg/min, group B 16-20 ml/kg/min, group C 10-16 ml/kg/min, and group D 6-10 ml/kg/min. Patients were also genotyped for NFkB and ACE genetic variants. T3 was increased and FT3 was decreased for every raise in Weber's classification (p = 0.007 and p = 0.012, respectively). Del carriers had elevated FT3 levels compared with Ins carriers (p = 0.021). Patients with II genotype had elevated T4 levels compared with ID genotype (p = 0.044). Both T4 and FT4 were decreased in D allele carriers (p = 0.007 and p = 0.045, respectively). Thyroid hormones correlated with CV capacity. Associations between the NFkB and ACE gene polymorphisms and thyroid hormones levels were also observed. Further larger studies are required to clarify genes contribution in HF.
Collapse
Affiliation(s)
- Ioannis Vasiliadis
- Cardiology Department, Onassis Cardiac Surgery Center Athens, 356 Sygrou Ave, 176 74, Athens, Greece
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Rizza W, Veronese N, Fontana L. What are the roles of calorie restriction and diet quality in promoting healthy longevity? Ageing Res Rev 2014; 13:38-45. [PMID: 24291541 DOI: 10.1016/j.arr.2013.11.002] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 11/12/2013] [Accepted: 11/19/2013] [Indexed: 12/17/2022]
Abstract
Epidemiological and experimental data indicate that diet plays a central role in the pathogenesis of many age-associated chronic diseases, and in the biology of aging itself. Data from several animal studies suggest that the degree and time of calorie restriction (CR) onset, the timing of food intake as well as diet composition, play major roles in promoting health and longevity, breaking the old dogma that only calorie intake is important in extending healthy lifespan. Data from human studies indicate that long-term CR with adequate intake of nutrients results in several metabolic adaptations that reduce the risk of developing type 2 diabetes, hypertension, cardiovascular disease and cancer. Moreover, CR opposes the expected age-associated alterations in myocardial stiffness, autonomic function, and gene expression in the human skeletal muscle. However, it is possible that some of the beneficial effects on metabolic health are not entirely due to CR, but to the high quality diets consumed by the CR practitioners, as suggested by data collected in individuals consuming strict vegan diets. More studies are needed to understand the interactions among single nutrient modifications (e.g. protein/aminoacid, fatty acids, vitamins, phytochemicals, and minerals), the degree of CR and the frequency of food consumption in modulating anti-aging metabolic and molecular pathways, and in the prevention of age-associated diseases.
Collapse
|
45
|
Bartke A, Sun LY, Longo V. Somatotropic signaling: trade-offs between growth, reproductive development, and longevity. Physiol Rev 2013; 93:571-98. [PMID: 23589828 PMCID: PMC3768106 DOI: 10.1152/physrev.00006.2012] [Citation(s) in RCA: 227] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Growth hormone (GH) is a key determinant of postnatal growth and plays an important role in the control of metabolism and body composition. Surprisingly, deficiency in GH signaling delays aging and remarkably extends longevity in laboratory mice. In GH-deficient and GH-resistant animals, the "healthspan" is also extended with delays in cognitive decline and in the onset of age-related disease. The role of hormones homologous to insulin-like growth factor (IGF, an important mediator of GH actions) in the control of aging and lifespan is evolutionarily conserved from worms to mammals with some homologies extending to unicellular yeast. The combination of reduced GH, IGF-I, and insulin signaling likely contributes to extended longevity in GH or GH receptor-deficient organisms. Diminutive body size and reduced fecundity of GH-deficient and GH-resistant mice can be viewed as trade-offs for extended longevity. Mechanisms responsible for delayed aging of GH-related mutants include enhanced stress resistance and xenobiotic metabolism, reduced inflammation, improved insulin signaling, and various metabolic adjustments. Pathological excess of GH reduces life expectancy in men as well as in mice, and GH resistance or deficiency provides protection from major age-related diseases, including diabetes and cancer, in both species. However, there is yet no evidence of increased longevity in GH-resistant or GH-deficient humans, possibly due to non-age-related deaths. Results obtained in GH-related mutant mice provide striking examples of mutations of a single gene delaying aging, reducing age-related disease, and extending lifespan in a mammal and providing novel experimental systems for the study of mechanisms of aging.
Collapse
Affiliation(s)
- Andrzej Bartke
- Southern Illinois University School of Medicine, Department of Internal Medicine, Geriatric Research, Springfield, Illinois 62703, USA.
| | | | | |
Collapse
|
46
|
Giani JF, Miquet JG, Muñoz MC, Burghi V, Toblli JE, Masternak MM, Kopchick JJ, Bartke A, Turyn D, Dominici FP. Upregulation of the angiotensin-converting enzyme 2/angiotensin-(1-7)/Mas receptor axis in the heart and the kidney of growth hormone receptor knock-out mice. Growth Horm IGF Res 2012; 22:224-233. [PMID: 22947377 PMCID: PMC3698955 DOI: 10.1016/j.ghir.2012.08.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2012] [Revised: 08/06/2012] [Accepted: 08/07/2012] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Growth hormone (GH) resistance leads to enhanced insulin sensitivity, decreased systolic blood pressure and increased lifespan. The aim of this study was to determine if there is a shift in the balance of the renin-angiotensin system (RAS) towards the ACE2/Ang-(1-7)/Mas receptor axis in the heart and the kidney of a model of GH resistance and retarded aging, the GH receptor knockout (GHR-/-) mouse. DESIGN RAS components were evaluated in the heart and the kidney of GHR-/- and control mice by immunohistochemistry and Western blotting (n=12 for both groups). RESULTS The immunostaining of Ang-(1-7) was increased in both the heart and the kidney of GHR-/- mice. These changes were concomitant with an increased immunostaining of the Mas receptor and ACE2 in both tissues. The immunostaining of AT1 receptor was reduced in heart and kidney of GHR-/- mice while that of AT2 receptor was increased in the heart and unaltered in the kidney. Ang II, ACE and angiotensinogen levels remained unaltered in the heart and the kidney of GH resistant mice. These results were confirmed by Western blotting and correlated with a significant increase in the abundance of the endothelial nitric oxide synthase in both tissues. CONCLUSIONS The shift within the RAS towards an exacerbation of the ACE2/Ang-(1-7)/Mas receptor axis observed in GHR-/- mice could be related to a protective role in cardiac and renal function; and thus, possibly contribute to the decreased incidence of cardiovascular diseases displayed by this animal model of longevity.
Collapse
Affiliation(s)
- Jorge F Giani
- Instituto de Química y Fisicoquímica Biológica (IQUIFIB), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Ding J, Sackmann-Sala L, Kopchick JJ. Mouse models of growth hormone action and aging: a proteomic perspective. Proteomics 2012; 13:674-85. [PMID: 23019135 DOI: 10.1002/pmic.201200271] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Revised: 07/19/2012] [Accepted: 07/27/2012] [Indexed: 12/12/2022]
Abstract
Growth hormone (GH) is a protein secreted by the anterior pituitary and circulates throughout the body to exert important actions on growth and metabolism. GH stimulates the secretion of insulin-like growth factor-I (IGF-I) that mediates some of the growth promoting actions of GH. The GH/IGF-I axis has recently been recognized as important in terms of longevity in organisms ranging from Caenorhabditis elegans to mice. For example, GH transgenic mice possess short lifespans while GH receptor null (GHR-/-) mice have extended longevity. Thus, the actions of GH (or IGF-I) or lack thereof impact the aging process. In this review, we summarize the proteomic analyses of plasma and white adipose tissue in these two mouse models of GH action, i.e. GH transgenic and GHR-/- mice. At the protein level, we wanted to establish novel plasma biomarkers of GH action as a function of age and to determine differences in adipose tissue depots. We have shown that these proteomic approaches have not only confirmed several known physiological actions of GH, but also resulted in novel protein biomarkers and targets that may be indicative of the aging process and/or new functions of GH. These results may generate new directions for GH and/or aging research.
Collapse
Affiliation(s)
- Juan Ding
- Department of Ophthalmology, Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | | | | |
Collapse
|
48
|
Jeong DE, Artan M, Seo K, Lee SJ. Regulation of lifespan by chemosensory and thermosensory systems: findings in invertebrates and their implications in mammalian aging. Front Genet 2012; 3:218. [PMID: 23087711 PMCID: PMC3475297 DOI: 10.3389/fgene.2012.00218] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Accepted: 10/01/2012] [Indexed: 12/30/2022] Open
Abstract
Many environmental factors that dynamically change in nature influence various aspects of animal physiology. Animals are equipped with sensory neuronal systems that help them properly sense and respond to environmental factors. Several studies have shown that chemosensory and thermosensory neurons affect the lifespan of invertebrate model animals, including Caenorhabditis elegans and Drosophila melanogaster. Although the mechanisms by which these sensory systems modulate lifespan are incompletely understood, hormonal signaling pathways have been implicated in sensory system-mediated lifespan regulation. In this review, we describe findings regarding how sensory nervous system components elicit physiological changes to regulate lifespan in invertebrate models, and discuss their implications in mammalian aging.
Collapse
Affiliation(s)
- Dae-Eun Jeong
- Division of Molecular and Life Science, Pohang University of Science and Technology Pohang, South Korea
| | | | | | | |
Collapse
|
49
|
Gesing A, Bartke A, Masternak MM, Lewiński A, Karbownik-Lewińska M. Decreased thyroid follicle size in dwarf mice may suggest the role of growth hormone signaling in thyroid growth regulation. Thyroid Res 2012; 5:7. [PMID: 22897932 PMCID: PMC3464137 DOI: 10.1186/1756-6614-5-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Accepted: 08/08/2012] [Indexed: 12/31/2022] Open
Abstract
Background Altered somatotrophic signaling is among the most important potential mechanisms of extended longevity. Ames dwarf (df/df) mice are homozygous for mutation at the Prop-1 gene, leading to a lack of growth hormone (GH), prolactin and thyroid stimulating hormone (TSH). Mice homozygous for targeted disruption of the growth hormone receptor/growth hormone binding protein gene are known as GH receptor knockout (GHRKO) mice or “Laron dwarf”. Both, df/df and GHRKO mice, are characterized by reduced body size, low plasma insulin and insulin-like growth factor-I (IGF-I), remarkably extended longevity, and severe (in df/df mice) or mild (in GHRKO mice) thyroid hypofunction. Recently, by crossing df/df and GHRKO mice, double-mutant Ames dwarf/GHRKO (df/KO) mice were created. Interestingly, these mice are smaller than Ames dwarfs or GHRKOs, and also have reduced insulin and IGF-I levels. The aim of the study was to investigate if and to what extent certain thyroid morphological parameters, such as inner follicular surface area, inner follicular perimeter, as well as the follicular epithelium thickness are changed in the examined dwarf mice. Methods This quantification was performed in thyroids collected from df/df, GHRKO and df/KO female mice, at approximately 5–6 months of age. We used a computerized plotting programme that combines a live microscopic image of the slide with an operator-generated overlay. Results Inner follicular surface area and inner follicular perimeter were decreased in all examined kinds of dwarf mice as compared to normal animals. Furthermore, decreases in these two parameters were more pronounced in df/df and df/KO than in GHRKO mice. Concerning the follicular epithelium thickness, only a tendency towards decrease of this parameter was found in all three kinds of dwarf mice. Conclusions Parameters characterizing thyroid follicle size are decreased in all three examined models of dwarf mice, which may explain decreased thyroid hormone levels in both basal mutants (Ames dwarfs and GHRKOs). df/df mutation seems to predominate over GHRKO genetic intervention concerning their effects on thyroid growth. Beside TSH, also GH signaling seems to constitute a crucial element in the regulation of thyroid growth and, possibly, function.
Collapse
Affiliation(s)
- Adam Gesing
- Department of Oncological Endocrinology, Chair of Endocrinology and Metabolic Diseases, Medical University of Lodz, Lodz, Poland.
| | | | | | | | | |
Collapse
|
50
|
Brown-Borg HM, Bartke A. GH and IGF1: roles in energy metabolism of long-living GH mutant mice. J Gerontol A Biol Sci Med Sci 2012; 67:652-60. [PMID: 22466316 PMCID: PMC3348496 DOI: 10.1093/gerona/gls086] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Of the multiple theories to explain exceptional longevity, the most robust of these has centered on the reduction of three anabolic protein hormones, growth hormone (GH), insulin-like growth factor, and insulin. GH mutant mice live 50% longer and exhibit significant differences in several aspects of energy metabolism as compared with wild-type mice. Mitochondrial metabolism is upregulated in the absence of GH, whereas in GH transgenic mice and dwarf mice treated with GH, multiple aspects of these pathways are suppressed. Core body temperature is markedly lower in dwarf mice, yet whole-body metabolism, as measured by indirect calorimetry, is surprisingly higher in Ames dwarf and Ghr-/- mice compared with normal controls. Elevated adiponectin, a key antiinflammatory cytokine, is also very likely to contribute to longevity in these mice. Thus, several important components related to energy metabolism are altered in GH mutant mice, and these differences are likely critical in aging processes and life-span extension.
Collapse
Affiliation(s)
- Holly M Brown-Borg
- Department of Pharmacology, Physiology & Therapeutics, School of Medicine & Health Sciences, University of North Dakota, 501 North Columbia Road, Grand Forks, ND 58202-9037, USA.
| | | |
Collapse
|