551
|
Pilling LC, Joehanes R, Kacprowski T, Peters M, Jansen R, Karasik D, Kiel DP, Harries LW, Teumer A, Powell J, Levy D, Lin H, Lunetta K, Munson P, Bandinelli S, Henley W, Hernandez D, Singleton A, Tanaka T, van Grootheest G, Hofman A, Uitterlinden AG, Biffar R, Gläser S, Homuth G, Malsch C, Völker U, Penninx B, van Meurs JBJ, Ferrucci L, Kocher T, Murabito J, Melzer D. Gene transcripts associated with muscle strength: a CHARGE meta-analysis of 7,781 persons. Physiol Genomics 2016; 48:1-11. [PMID: 26487704 PMCID: PMC4757025 DOI: 10.1152/physiolgenomics.00054.2015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 10/08/2015] [Indexed: 12/22/2022] Open
Abstract
Lower muscle strength in midlife predicts disability and mortality in later life. Blood-borne factors, including growth differentiation factor 11 (GDF11), have been linked to muscle regeneration in animal models. We aimed to identify gene transcripts associated with muscle strength in adults. Meta-analysis of whole blood gene expression (overall 17,534 unique genes measured by microarray) and hand-grip strength in four independent cohorts (n = 7,781, ages: 20-104 yr, weighted mean = 56), adjusted for age, sex, height, weight, and leukocyte subtypes. Separate analyses were performed in subsets (older/younger than 60, men/women). Expression levels of 221 genes were associated with strength after adjustment for cofactors and for multiple statistical testing, including ALAS2 (rate-limiting enzyme in heme synthesis), PRF1 (perforin, a cytotoxic protein associated with inflammation), IGF1R, and IGF2BP2 (both insulin like growth factor related). We identified statistical enrichment for hemoglobin biosynthesis, innate immune activation, and the stress response. Ten genes were associated only in younger individuals, four in men only and one in women only. For example, PIK3R2 (a negative regulator of PI3K/AKT growth pathway) was negatively associated with muscle strength in younger (<60 yr) individuals but not older (≥ 60 yr). We also show that 115 genes (52%) have not previously been linked to muscle in NCBI PubMed abstracts. This first large-scale transcriptome study of muscle strength in human adults confirmed associations with known pathways and provides new evidence for over half of the genes identified. There may be age- and sex-specific gene expression signatures in blood for muscle strength.
Collapse
Affiliation(s)
- L C Pilling
- Epidemiology and Public Health Group, Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, United Kingdom
| | - R Joehanes
- The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, Massachusetts; Population Studies Branch, National Heart, Lung, and Blood Institute, Bethesda, Maryland
| | - T Kacprowski
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine and Ernst Moritz Arndt University Greifswald, Greifswald, Germany
| | - M Peters
- Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands; The Netherlands Genomics Initiative-sponsored Netherlands Consortium for Healthy Aging (NGI-NCHA), Leiden/Rotterdam, the Netherlands
| | - R Jansen
- Department of Psychiatry, VU University Medical Center, Neuroscience Campus Amsterdam, Amsterdam, the Netherlands
| | - D Karasik
- Hebrew SeniorLife Institute for Aging Research, Boston, Massachusetts
| | - D P Kiel
- Hebrew SeniorLife Institute for Aging Research, Boston, Massachusetts
| | - L W Harries
- RNA mechanisms of complex diseases group, Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, United Kingdom
| | - A Teumer
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine and Ernst Moritz Arndt University Greifswald, Greifswald, Germany
| | - J Powell
- Centre for Neurogenetics and Statistical Genomics, Queensland Brain Institute, University of Queensland, St. Lucia, Brisbane, Australia
| | - D Levy
- The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, Massachusetts; Population Studies Branch, National Heart, Lung, and Blood Institute, Bethesda, Maryland
| | - H Lin
- The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, Massachusetts; Section of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - K Lunetta
- The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, Massachusetts; Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - P Munson
- The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, Massachusetts; The Mathematical and Statistical Computing Laboratory, Center for Information Technology, National Institutes of Health, Bethesda, Maryland
| | - S Bandinelli
- Geriatric Unit, Azienda Sanitaria di Firenze, Florence, Italy
| | - W Henley
- Institute for Health Services Research, University of Exeter Medical School, Exeter, United Kingdom
| | - D Hernandez
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland
| | - A Singleton
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland
| | - T Tanaka
- Clinical Research Branch, National Institute on Aging, Baltimore, Maryland
| | - G van Grootheest
- Department of Psychiatry, VU University Medical Center, Neuroscience Campus Amsterdam, Amsterdam, the Netherlands
| | - A Hofman
- The Netherlands Genomics Initiative-sponsored Netherlands Consortium for Healthy Aging (NGI-NCHA), Leiden/Rotterdam, the Netherlands; Department of Epidemiology, Erasmus Medical Center Rotterdam, the Netherlands
| | - A G Uitterlinden
- Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands; The Netherlands Genomics Initiative-sponsored Netherlands Consortium for Healthy Aging (NGI-NCHA), Leiden/Rotterdam, the Netherlands; Department of Epidemiology, Erasmus Medical Center Rotterdam, the Netherlands
| | - R Biffar
- Department of Prosthetic Dentistry, Gerostomatology and Dental Materials, University Medicine Greifswald, Greifswald, Germany
| | - S Gläser
- Department of Internal Medicine B - Cardiology, Intensive Care, Pulmonary Medicine and Infectious Diseases, University of Greifswald, Greifswald, Germany
| | - G Homuth
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine and Ernst Moritz Arndt University Greifswald, Greifswald, Germany
| | - C Malsch
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine and Ernst Moritz Arndt University Greifswald, Greifswald, Germany
| | - U Völker
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine and Ernst Moritz Arndt University Greifswald, Greifswald, Germany
| | - B Penninx
- Department of Psychiatry, VU University Medical Center, Neuroscience Campus Amsterdam, Amsterdam, the Netherlands
| | - J B J van Meurs
- Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands; The Netherlands Genomics Initiative-sponsored Netherlands Consortium for Healthy Aging (NGI-NCHA), Leiden/Rotterdam, the Netherlands
| | - L Ferrucci
- Clinical Research Branch, National Institute on Aging, Baltimore, Maryland
| | - T Kocher
- Unit of Periodontology, Department of Restorative Dentistry, Periodontology and Endodontology, University Medicine Greifswald, Greifswald, Germany; and
| | - J Murabito
- The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, Massachusetts; General Internal Medicine Section, Boston University, Boston, Massachusetts
| | - D Melzer
- Epidemiology and Public Health Group, Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, United Kingdom;
| |
Collapse
|
552
|
Swahari V, Nakamura A. Speeding up the clock: The past, present and future of progeria. Dev Growth Differ 2015; 58:116-30. [DOI: 10.1111/dgd.12251] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 10/15/2015] [Accepted: 10/15/2015] [Indexed: 12/18/2022]
Affiliation(s)
- Vijay Swahari
- Neuroscience Center; University of North Carolina; Chapel Hill North Carolina USA
| | - Ayumi Nakamura
- Neuroscience Center; University of North Carolina; Chapel Hill North Carolina USA
| |
Collapse
|
553
|
Dhawan J, Laxman S. Decoding the stem cell quiescence cycle--lessons from yeast for regenerative biology. J Cell Sci 2015; 128:4467-74. [PMID: 26672015 PMCID: PMC5695657 DOI: 10.1242/jcs.177758] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
In the past decade, major advances have occurred in the understanding of mammalian stem cell biology, but roadblocks (including gaps in our fundamental understanding) remain in translating this knowledge to regenerative medicine. Interestingly, a close analysis of the Saccharomyces cerevisiae literature leads to an appreciation of how much yeast biology has contributed to the conceptual framework underpinning our understanding of stem cell behavior, to the point where such insights have been internalized into the realm of the known. This Opinion article focuses on one such example, the quiescent adult mammalian stem cell, and examines concepts underlying our understanding of quiescence that can be attributed to studies in yeast. We discuss the metabolic, signaling and gene regulatory events that control entry and exit into quiescence in yeast. These processes and events retain remarkable conservation and conceptual parallels in mammalian systems, and collectively suggest a regulated program beyond the cessation of cell division. We argue that studies in yeast will continue to not only reveal fundamental concepts in quiescence, but also leaven progress in regenerative medicine.
Collapse
Affiliation(s)
- Jyotsna Dhawan
- Institute for Stem Cell Biology and Regenerative Medicine, Bangalore, India CSIR Center for Cellular and Molecular Biology, Hyderabad, India
| | - Sunil Laxman
- Institute for Stem Cell Biology and Regenerative Medicine, Bangalore, India
| |
Collapse
|
554
|
|
555
|
Rochette L, Zeller M, Cottin Y, Vergely C. Growth and differentiation factor 11 (GDF11): Functions in the regulation of erythropoiesis and cardiac regeneration. Pharmacol Ther 2015; 156:26-33. [DOI: 10.1016/j.pharmthera.2015.10.006] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
556
|
Dey D, Goldhamer DJ, Yu PB. Contributions of muscle-resident progenitor cells to homeostasis and disease. CURRENT MOLECULAR BIOLOGY REPORTS 2015; 1:175-188. [PMID: 29075589 PMCID: PMC5654566 DOI: 10.1007/s40610-015-0025-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Adult skeletal muscle maintains a homeostatic state with modest levels of cellular turnover, unlike the skin or blood. However, the muscle is highly sensitive to tissue injury, which unleashes a cascade of regenerative and inflammatory processes. Muscle regeneration involves cross-talk between numerous cytokine signaling axes, and the coordinated activity of multiple muscle-resident and circulating progenitor populations. Satellite cells, closely associated with myofibers, are established as the canonical muscle stem cell, with self-renewal and myofiber-regenerating capacity. However, a heterogeneous group of mesenchymal progenitor cells residing within the muscle interstitium are also highly responsive to muscle injury and exhibit varying degrees of regenerative potential. These cells interact with satellite cells via direct and indirect mechanisms to regulate regeneration or repair. We describe the known phylogenetic and functional relationships of the multiple progenitor populations residing within skeletal muscle, their putative roles in the coordination of injury repair, and their possible contributions to health and disease.
Collapse
Affiliation(s)
- Devaveena Dey
- Department of Medicine, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Boston, MA 02115-6119, USA
| | - David J. Goldhamer
- Department of Molecular and Cell Biology, University of Connecticut Stem Cell Institute, University of Connecticut, Storrs, CT 06269-3125, USA
| | - Paul B. Yu
- Department of Medicine, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Boston, MA 02115-6119, USA
| |
Collapse
|
557
|
DeCarolis NA, Kirby ED, Wyss-Coray T, Palmer TD. The Role of the Microenvironmental Niche in Declining Stem-Cell Functions Associated with Biological Aging. Cold Spring Harb Perspect Med 2015; 5:5/12/a025874. [PMID: 26627453 DOI: 10.1101/cshperspect.a025874] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Aging is strongly correlated with decreases in neurogenesis, the process by which neural stem and progenitor cells proliferate and differentiate into new neurons. In addition to stem-cell-intrinsic factors that change within the aging stem-cell pool, recent evidence emphasizes new roles for systemic and microenvironmental factors in modulating the neurogenic niche. This article focuses on new insights gained through the use of heterochronic parabiosis models, in which an old mouse and a young circulatory system are joined. By studying the brains of both young and old mice, researchers are beginning to uncover circulating proneurogenic "youthful" factors and "aging" factors that decrease stem-cell activity and neurogenesis. Ultimately, the identification of factors that influence stem-cell aging may lead to strategies that slow or even reverse age-related decreases in neural-stem-cell (NSC) function and neurogenesis.
Collapse
Affiliation(s)
- Nathan A DeCarolis
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California 94305
| | - Elizabeth D Kirby
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California 94305
| | - Tony Wyss-Coray
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California 94305 Center for Tissue Regeneration, Repair, and Restoration, Veterans Administration, Palo Alto Health Care Systems, Palo Alto, California 94304
| | - Theo D Palmer
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California 94305 Department of Neurosurgery, Stanford University School of Medicine, Stanford, California 94305
| |
Collapse
|
558
|
Yu H, Fujii NL, Toyoda T, An D, Farese RV, Leitges M, Hirshman MF, Mul JD, Goodyear LJ. Contraction stimulates muscle glucose uptake independent of atypical PKC. Physiol Rep 2015; 3:3/11/e12565. [PMID: 26564060 PMCID: PMC4673624 DOI: 10.14814/phy2.12565] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Exercise increases skeletal muscle glucose uptake, but the underlying mechanisms are only partially understood. The atypical protein kinase C (PKC) isoforms λ and ζ (PKC‐λ/ζ) have been shown to be necessary for insulin‐, AICAR‐, and metformin‐stimulated glucose uptake in skeletal muscle, but not for treadmill exercise‐stimulated muscle glucose uptake. To investigate if PKC‐λ/ζ activity is required for contraction‐stimulated muscle glucose uptake, we used mice with tibialis anterior muscle‐specific overexpression of an empty vector (WT), wild‐type PKC‐ζ (PKC‐ζWT), or an enzymatically inactive T410A‐PKC‐ζ mutant (PKC‐ζT410A). We also studied skeletal muscle‐specific PKC‐λ knockout (MλKO) mice. Basal glucose uptake was similar between WT, PKC‐ζWT, and PKC‐ζT410A tibialis anterior muscles. In contrast, in situ contraction‐stimulated glucose uptake was increased in PKC‐ζT410A tibialis anterior muscles compared to WT or PKC‐ζWT tibialis anterior muscles. Furthermore, in vitro contraction‐stimulated glucose uptake was greater in soleus muscles of MλKO mice than WT controls. Thus, loss of PKC‐λ/ζ activity increases contraction‐stimulated muscle glucose uptake. These data clearly demonstrate that PKC‐λ/ζ activity is not necessary for contraction‐stimulated glucose uptake.
Collapse
Affiliation(s)
- Haiyan Yu
- Harvard Medical School, Joslin Diabetes Center, Boston, Massachusetts
| | - Nobuharu L Fujii
- Harvard Medical School, Joslin Diabetes Center, Boston, Massachusetts
| | - Taro Toyoda
- Harvard Medical School, Joslin Diabetes Center, Boston, Massachusetts
| | - Ding An
- Harvard Medical School, Joslin Diabetes Center, Boston, Massachusetts
| | | | - Michael Leitges
- The Biotechnology Center of Oslo, University of Oslo, Blindern, Oslo, Norway
| | | | - Joram D Mul
- Harvard Medical School, Joslin Diabetes Center, Boston, Massachusetts
| | - Laurie J Goodyear
- Harvard Medical School, Joslin Diabetes Center, Boston, Massachusetts
| |
Collapse
|
559
|
Blau HM, Cosgrove BD, Ho ATV. The central role of muscle stem cells in regenerative failure with aging. Nat Med 2015; 21:854-62. [PMID: 26248268 DOI: 10.1038/nm.3918] [Citation(s) in RCA: 311] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 07/10/2015] [Indexed: 02/07/2023]
Abstract
Skeletal muscle mass, function, and repair capacity all progressively decline with aging, restricting mobility, voluntary function, and quality of life. Skeletal muscle repair is facilitated by a population of dedicated muscle stem cells (MuSCs), also known as satellite cells, that reside in anatomically defined niches within muscle tissues. In adult tissues, MuSCs are retained in a quiescent state until they are primed to regenerate damaged muscle through cycles of self-renewal divisions. With aging, muscle tissue homeostasis is progressively disrupted and the ability of MuSCs to repair injured muscle markedly declines. Until recently, this decline has been largely attributed to extrinsic age-related alterations in the microenvironment to which MuSCs are exposed. However, as highlighted in this Perspective, recent reports show that MuSCs also progressively undergo cell-intrinsic alterations that profoundly affect stem cell regenerative function with aging. A more comprehensive understanding of the interplay of stem cell-intrinsic and extrinsic factors will set the stage for improving cell therapies capable of restoring tissue homeostasis and enhancing muscle repair in the aged.
Collapse
Affiliation(s)
- Helen M Blau
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Benjamin D Cosgrove
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| | - Andrew T V Ho
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
560
|
Rodgers BD, Eldridge JA. Reduced Circulating GDF11 Is Unlikely Responsible for Age-Dependent Changes in Mouse Heart, Muscle, and Brain. Endocrinology 2015; 156:3885-8. [PMID: 26372181 DOI: 10.1210/en.2015-1628] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Recent high-profile studies report conflicting data on the age-related change in circulating growth/differentiation factor 11 (GDF11) and myostatin as well as the former's influence on muscle regeneration. Both ligands bind and activate ActRIIB receptors with similar affinities and should therefore have similar actions, yet these studies suggest that GDF11 activates muscle regeneration whereas myostatin is well known to inhibit it. They also suggest that circulating GDF11 levels, but not those of myostatin, decline with age. We performed a careful assessment of the ELISA used to quantify circulating myostatin in these studies and determined that assay reagents significantly cross react with each protein, each of which is highly homologous. Circulating myostatin levels decreased with age and estimates of GDF11 levels using myostatin null mice indicate that they were almost 500 times lower than those for myostatin. This suggests that circulating GDF11 has little physiological relevance as it could not outcompete myostatin for ActRIIB binding sites. Together, these results further suggest that the previously reported aging muscle, heart, and brain phenotypes attributed to reduced circulating GDF11 should be reconsidered.
Collapse
Affiliation(s)
- Buel D Rodgers
- Department of Animal Sciences, Washington Center for Muscle Biology, Washington State University, Pullman, Washington 99164
| | - Jennifer A Eldridge
- Department of Animal Sciences, Washington Center for Muscle Biology, Washington State University, Pullman, Washington 99164
| |
Collapse
|
561
|
Londhe P, Guttridge DC. Inflammation induced loss of skeletal muscle. Bone 2015; 80:131-142. [PMID: 26453502 PMCID: PMC4600538 DOI: 10.1016/j.bone.2015.03.015] [Citation(s) in RCA: 204] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Revised: 03/03/2015] [Accepted: 03/18/2015] [Indexed: 12/21/2022]
Abstract
Inflammation is an important contributor to the pathology of diseases implicated in skeletal muscle dysfunction. A number of diseases and disorders including inflammatory myopathies and Chronic Obstructive Pulmonary Disorder (COPD) are characterized by chronic inflammation or elevation of the inflammatory mediators. While these disease states exhibit different pathologies, all have in common the loss of skeletal muscle mass and a deregulated skeletal muscle physiology. Pro-inflammatory cytokines are key contributors to chronic inflammation found in many of these diseases. This section of the review focuses on some of the known inflammatory disorders like COPD, Rheumatoid Arthritis (RA) and inflammatory myopathies that display skeletal muscle atrophy and also provides the reader an overview of the mediators of inflammation, their signaling pathways, and mechanisms of action. This article is part of a Special Issue entitled "Muscle Bone Interactions".
Collapse
Affiliation(s)
- Priya Londhe
- Department of Molecular Virology, Immunology, and Medical Genetics, Human Cancer Genetics Program, Arthur G. James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Denis C Guttridge
- Department of Molecular Virology, Immunology, and Medical Genetics, Human Cancer Genetics Program, Arthur G. James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
562
|
Amaral MD, Balch WE. Hallmarks of therapeutic management of the cystic fibrosis functional landscape. J Cyst Fibros 2015; 14:687-99. [PMID: 26526359 PMCID: PMC4644672 DOI: 10.1016/j.jcf.2015.09.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 09/16/2015] [Accepted: 09/16/2015] [Indexed: 01/29/2023]
Abstract
The cystic fibrosis (CF) transmembrane conductance regulator (CFTR) protein does not operate in isolation, rather in a dynamic network of interacting components that impact its synthesis, folding, stability, intracellular location and function, referred to herein as the 'CFTR Functional Landscape (CFFL)'. For the prominent F508del mutation, many of these interactors are deeply connected to a protein fold management system, the proteostasis network (PN). However, CF encompasses an additional 2000 CFTR variants distributed along its entire coding sequence (referred to as CFTR2), and each variant contributes a differential liability to PN management of CFTR and to a protein 'social network' (SN) that directs the probability of the (patho)physiologic events that impact ion transport in each cell, tissue and patient in health and disease. Recognition of the importance of the PN and SN in driving the unique patient CFFL leading to disease highlights the importance of precision medicine in therapeutic management of disease progression. We take the view herein that it is not CFTR, rather the PN/SN, and their impact on the CFFL, that are the key physiologic forces driving onset and clinical progression of CF. We posit that a deep understanding of each patients PN/SN gained by merging genomic, proteomic (mass spectrometry (MS)), and high-content microscopy (HCM) technologies in the context of novel network learning algorithms will lead to a paradigm shift in CF clinical management. This should allow for generation of new classes of patient specific PN/SN directed therapeutics for personalized management of the CFFL in the clinic.
Collapse
Affiliation(s)
- Margarida D Amaral
- University of Lisboa, Faculty of Sciences, BioISI - Biosystems & Integrative Sciences Institute, Lisboa, Portugal.
| | - William E Balch
- Department of Chemical Physiology, Department of Cell and Molecular Biology, The Skaggs Institute of Chemical Biology, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
563
|
Deshmukh AS, Cox J, Jensen LJ, Meissner F, Mann M. Secretome Analysis of Lipid-Induced Insulin Resistance in Skeletal Muscle Cells by a Combined Experimental and Bioinformatics Workflow. J Proteome Res 2015; 14:4885-95. [PMID: 26457550 DOI: 10.1021/acs.jproteome.5b00720] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Skeletal muscle has emerged as an important secretory organ that produces so-called myokines, regulating energy metabolism via autocrine, paracrine, and endocrine actions; however, the nature and extent of the muscle secretome has not been fully elucidated. Mass spectrometry (MS)-based proteomics, in principle, allows an unbiased and comprehensive analysis of cellular secretomes; however, the distinction of bona fide secreted proteins from proteins released upon lysis of a small fraction of dying cells remains challenging. Here we applied highly sensitive MS and streamlined bioinformatics to analyze the secretome of lipid-induced insulin-resistant skeletal muscle cells. Our workflow identified 1073 putative secreted proteins including 32 growth factors, 25 cytokines, and 29 metalloproteinases. In addition to previously reported proteins, we report hundreds of novel ones. Intriguingly, ∼40% of the secreted proteins were regulated under insulin-resistant conditions, including a protein family with signal peptide and EGF-like domain structure that had not yet been associated with insulin resistance. Finally, we report that secretion of IGF and IGF-binding proteins was down-regulated under insulin-resistant conditions. Our study demonstrates an efficient combined experimental and bioinformatics workflow to identify putative secreted proteins from insulin-resistant skeletal muscle cells, which could easily be adapted to other cellular models.
Collapse
Affiliation(s)
- Atul S Deshmukh
- Department of Proteomics and Signal Transduction, Max-Planck-Institute of Biochemistry , Am Klopferspitz 18, D-82152 Martinsried, Germany.,The Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen , Blegdamsvej 3B, Building 6.1, DK-2200 Copenhagen, Denmark
| | - Juergen Cox
- Department of Proteomics and Signal Transduction, Max-Planck-Institute of Biochemistry , Am Klopferspitz 18, D-82152 Martinsried, Germany
| | - Lars Juhl Jensen
- The Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen , Blegdamsvej 3B, Building 6.1, DK-2200 Copenhagen, Denmark
| | - Felix Meissner
- Department of Proteomics and Signal Transduction, Max-Planck-Institute of Biochemistry , Am Klopferspitz 18, D-82152 Martinsried, Germany
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max-Planck-Institute of Biochemistry , Am Klopferspitz 18, D-82152 Martinsried, Germany.,The Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen , Blegdamsvej 3B, Building 6.1, DK-2200 Copenhagen, Denmark
| |
Collapse
|
564
|
Young JC, Wakitani S, Loveland KL. TGF-β superfamily signaling in testis formation and early male germline development. Semin Cell Dev Biol 2015; 45:94-103. [PMID: 26500180 DOI: 10.1016/j.semcdb.2015.10.029] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 10/16/2015] [Indexed: 12/11/2022]
Abstract
The TGF-β ligand superfamily contains at least 40 members, many of which are produced and act within the mammalian testis to facilitate formation of sperm. Their progressive expression at key stages and in specific cell types determines the fertility of adult males, influencing testis development and controlling germline differentiation. BMPs are essential for the interactive instructions between multiple cell types in the early embryo that drive initial specification of gamete precursors. In the nascent foetal testis, several ligands including Nodal, TGF-βs, Activins and BMPs, serve as key masculinizing switches by regulating male germline pluripotency, somatic and germline proliferation, and testicular vascularization and architecture. In postnatal life, local production of these factors determine adult testis size by regulating Sertoli cell multiplication and differentiation, in addition to specifying germline differentiation and multiplication. Because TGF-β superfamily signaling is integral to testis formation, it affects processes that underlie testicular pathologies, including testicular cancer, and its potential to contribute to subfertility is beginning to be understood.
Collapse
Affiliation(s)
- Julia C Young
- Hudson Institute of Medical Research, Clayton, Victoria, Australia; Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Shoichi Wakitani
- Hudson Institute of Medical Research, Clayton, Victoria, Australia; Laboratory of Veterinary Biochemistry and Molecular Biology, University of Miyazaki, Japan
| | - Kate L Loveland
- Hudson Institute of Medical Research, Clayton, Victoria, Australia; School of Clinical Sciences, Monash University, Clayton, Victoria, Australia; Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
565
|
Snijders T, Nederveen JP, McKay BR, Joanisse S, Verdijk LB, van Loon LJC, Parise G. Satellite cells in human skeletal muscle plasticity. Front Physiol 2015; 6:283. [PMID: 26557092 PMCID: PMC4617172 DOI: 10.3389/fphys.2015.00283] [Citation(s) in RCA: 229] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 09/23/2015] [Indexed: 01/06/2023] Open
Abstract
Skeletal muscle satellite cells are considered to play a crucial role in muscle fiber maintenance, repair and remodeling. Our knowledge of the role of satellite cells in muscle fiber adaptation has traditionally relied on in vitro cell and in vivo animal models. Over the past decade, a genuine effort has been made to translate these results to humans under physiological conditions. Findings from in vivo human studies suggest that satellite cells play a key role in skeletal muscle fiber repair/remodeling in response to exercise. Mounting evidence indicates that aging has a profound impact on the regulation of satellite cells in human skeletal muscle. Yet, the precise role of satellite cells in the development of muscle fiber atrophy with age remains unresolved. This review seeks to integrate recent results from in vivo human studies on satellite cell function in muscle fiber repair/remodeling in the wider context of satellite cell biology whose literature is largely based on animal and cell models.
Collapse
Affiliation(s)
- Tim Snijders
- Department of Kinesiology and Medical Physics and Applied Radiation Sciences, McMaster University Hamilton, ON, Canada ; Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Maastricht, Netherlands
| | - Joshua P Nederveen
- Department of Kinesiology and Medical Physics and Applied Radiation Sciences, McMaster University Hamilton, ON, Canada
| | - Bryon R McKay
- Department of Kinesiology and Medical Physics and Applied Radiation Sciences, McMaster University Hamilton, ON, Canada
| | - Sophie Joanisse
- Department of Kinesiology and Medical Physics and Applied Radiation Sciences, McMaster University Hamilton, ON, Canada
| | - Lex B Verdijk
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Maastricht, Netherlands
| | - Luc J C van Loon
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Maastricht, Netherlands
| | - Gianni Parise
- Department of Kinesiology and Medical Physics and Applied Radiation Sciences, McMaster University Hamilton, ON, Canada
| |
Collapse
|
566
|
Yang J, Huang T, Petralia F, Long Q, Zhang B, Argmann C, Zhao Y, Mobbs CV, Schadt EE, Zhu J, Tu Z. Synchronized age-related gene expression changes across multiple tissues in human and the link to complex diseases. Sci Rep 2015; 5:15145. [PMID: 26477495 PMCID: PMC4609956 DOI: 10.1038/srep15145] [Citation(s) in RCA: 144] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 09/21/2015] [Indexed: 01/06/2023] Open
Abstract
Aging is one of the most important biological processes and is a known risk factor for many age-related diseases in human. Studying age-related transcriptomic changes in tissues across the whole body can provide valuable information for a holistic understanding of this fundamental process. In this work, we catalogue age-related gene expression changes in nine tissues from nearly two hundred individuals collected by the Genotype-Tissue Expression (GTEx) project. In general, we find the aging gene expression signatures are very tissue specific. However, enrichment for some well-known aging components such as mitochondria biology is observed in many tissues. Different levels of cross-tissue synchronization of age-related gene expression changes are observed, and some essential tissues (e.g., heart and lung) show much stronger "co-aging" than other tissues based on a principal component analysis. The aging gene signatures and complex disease genes show a complex overlapping pattern and only in some cases, we see that they are significantly overlapped in the tissues affected by the corresponding diseases. In summary, our analyses provide novel insights to the co-regulation of age-related gene expression in multiple tissues; it also presents a tissue-specific view of the link between aging and age-related diseases.
Collapse
Affiliation(s)
- Jialiang Yang
- Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, NY, 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY, 10029, USA
| | - Tao Huang
- Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, NY, 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY, 10029, USA
| | - Francesca Petralia
- Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, NY, 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY, 10029, USA
| | - Quan Long
- Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, NY, 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY, 10029, USA
| | - Bin Zhang
- Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, NY, 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY, 10029, USA
| | - Carmen Argmann
- Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, NY, 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY, 10029, USA
| | - Yong Zhao
- Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, NY, 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY, 10029, USA
| | - Charles V. Mobbs
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, NY, 10029, USA
- Department of Geriatrics and Palliative Medicine, Icahn School of Medicine at Mount Sinai, NY, 10029, USA
- Department of Medicine, Endocrinology, Diabetes and Bone Disease, Icahn School of Medicine at Mount Sinai, NY, 10029, USA
| | - Eric E. Schadt
- Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, NY, 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY, 10029, USA
| | - Jun Zhu
- Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, NY, 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY, 10029, USA
| | - Zhidong Tu
- Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, NY, 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY, 10029, USA
| |
Collapse
|
567
|
Latres E, Pangilinan J, Miloscio L, Bauerlein R, Na E, Potocky TB, Huang Y, Eckersdorff M, Rafique A, Mastaitis J, Lin C, Murphy AJ, Yancopoulos GD, Gromada J, Stitt T. Myostatin blockade with a fully human monoclonal antibody induces muscle hypertrophy and reverses muscle atrophy in young and aged mice. Skelet Muscle 2015; 5:34. [PMID: 26457176 PMCID: PMC4600334 DOI: 10.1186/s13395-015-0060-8] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 09/29/2015] [Indexed: 01/08/2023] Open
Abstract
Background Loss of skeletal muscle mass and function in humans is associated with significant morbidity and mortality. The role of myostatin as a key negative regulator of skeletal muscle mass and function has supported the concept that inactivation of myostatin could be a useful approach for treating muscle wasting diseases. Methods We generated a myostatin monoclonal blocking antibody (REGN1033) and characterized its effects in vitro using surface plasmon resonance biacore and cell-based Smad2/3 signaling assays. REGN1033 was tested in mice for the ability to induce skeletal muscle hypertrophy and prevent atrophy induced by immobilization, hindlimb suspension, or dexamethasone. The effect of REGN1033 on exercise training was tested in aged mice. Messenger RNA sequencing, immunohistochemistry, and ex vivo force measurements were performed on skeletal muscle samples from REGN1033-treated mice. Results The human monoclonal antibody REGN1033 is a specific and potent myostatin antagonist. Chronic treatment of mice with REGN1033 increased muscle fiber size, muscle mass, and force production. REGN1033 prevented the loss of muscle mass induced by immobilization, glucocorticoid treatment, or hindlimb unweighting and increased the gain of muscle mass during recovery from pre-existing atrophy. In aged mice, REGN1033 increased muscle mass and strength and improved physical performance during treadmill exercise. Conclusions We show that specific myostatin antagonism with the human antibody REGN1033 enhanced muscle mass and function in young and aged mice and had beneficial effects in models of skeletal muscle atrophy.
Collapse
Affiliation(s)
- Esther Latres
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591 USA
| | - Jeffrey Pangilinan
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591 USA
| | - Lawrence Miloscio
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591 USA
| | - Roy Bauerlein
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591 USA
| | - Erqian Na
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591 USA
| | - Terra B Potocky
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591 USA
| | - Ying Huang
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591 USA
| | - Mark Eckersdorff
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591 USA
| | - Ashique Rafique
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591 USA
| | - Jason Mastaitis
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591 USA
| | - Calvin Lin
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591 USA
| | - Andrew J Murphy
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591 USA
| | - George D Yancopoulos
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591 USA
| | - Jesper Gromada
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591 USA
| | - Trevor Stitt
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591 USA
| |
Collapse
|
568
|
Abstract
Sarcopenia and cachexia are muscle wasting syndromes associated with aging and with many chronic diseases, such as congestive heart failure (CHF), diabetes, cancer, chronic obstructive pulmonary disease and chronic kidney disease (CKD). While mechanisms are complex, these conditions are often accompanied by elevated angiotensin II (Ang II). Patients with advanced CHF or CKD often have increased Ang II levels and cachexia, and angiotensin-converting enzyme inhibitor treatment improves weight loss. It was found that Ang II infusion in rodents leads to skeletal muscle wasting. Ang II increases cytokines and circulating hormones, such as tumor necrosis factor-α, interleukin-6, serum amyloid-A and glucocorticoids, which regulate muscle protein synthesis and degradation. Ang II-induced muscle wasting is caused by alterations in insulin-like growth factor-1 signaling, enhanced muscle protein breakdown via the ubiquitin-proteasome system and decreased appetite resulting from the downregulation of hypothalamic orexigenic neuropeptides, such as Npy and orexin. Ang II also inhibits 5' adenosine monophosphate-activated protein kinase activity and disrupts normal energy balance via the activation of 5' adenosine monophosphate-activated protein kinase phosphatase PP2Cα. Furthermore, Ang II inhibits skeletal muscle stem (satellite) cell proliferation, leading to lowered muscle regenerative capacity. Distinct satellite cell angiotensin receptor subtypes have different effects on different stages of differentiation and are critical for the regulation of muscle regeneration. These data suggest that the renin-angiotensin system plays a critical role in mechanisms underlying cachexia in chronic disease states, and it is a promising target for the treatment of muscle atrophy in patients with diseases such as CHF and CKD.
Collapse
Affiliation(s)
- Tadashi Yoshida
- Department of Medicine, University of Missouri-Columbia, Columbia, MO
| | | |
Collapse
|
569
|
Bickford PC, Kaneko Y, Grimmig B, Pappas C, Small B, Sanberg CD, Sanberg PR, Tan J, Douglas Shytle R. Nutraceutical intervention reverses the negative effects of blood from aged rats on stem cells. AGE (DORDRECHT, NETHERLANDS) 2015; 37:103. [PMID: 26410618 PMCID: PMC5005857 DOI: 10.1007/s11357-015-9840-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 09/15/2015] [Indexed: 06/05/2023]
Abstract
Aging is associated with a decline in function in many of the stem cell niches of the body. An emerging body of literature suggests that one of the reasons for this decline in function is due to cell non-autonomous influences on the niche from the body. For example, studies using the technique of parabiosis have demonstrated a negative influence of blood from aged mice on muscle satellite cells and neurogenesis in young mice. We examined if we could reverse this effect of aged serum on stem cell proliferation by treating aged rats with NT-020, a dietary supplement containing blueberry, green tea, vitamin D3, and carnosine that has been shown to increase neurogenesis in aged rats. Young and aged rats were administered either control NIH-31 diet or one supplemented with NT-020 for 28 days, and serum was collected upon euthanasia. The serum was used in cultures of both rat hippocampal neural progenitor cells (NPCs) and rat bone marrow-derived mesenchymal stem cells (MSCs). Serum from aged rats significantly reduced cell proliferation as measured by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and 5-bromo-2'-deoxyuridine (BrdU) assays in both NPCs and MSCs. Serum from aged rats treated with NT-020 was not different from serum from young rats. Therefore, NT-020 rescued the effect of serum from aged rats to reduce stem cell proliferation.
Collapse
Affiliation(s)
- Paula C Bickford
- Research Service, James A Haley Veterans Affairs Hospital|, Tampa, FL, 33620, USA.
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, MDC-78, USF Morsani College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL, 33620, USA.
| | - Yuji Kaneko
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, MDC-78, USF Morsani College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL, 33620, USA
| | - Bethany Grimmig
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, MDC-78, USF Morsani College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL, 33620, USA
- Department of Molecular Pharmacology and Physiology, USF Morsani College of Medicine, Tampa, FL, 33620, USA
| | | | - Brent Small
- School of Aging Studies, USF, Tampa, FL, 33620, USA
| | | | - Paul R Sanberg
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, MDC-78, USF Morsani College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL, 33620, USA
| | - Jun Tan
- Department of Psychiatry, USF Morsani College of Medicine, Tampa, FL, 33620, USA
| | - R Douglas Shytle
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, MDC-78, USF Morsani College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL, 33620, USA
| |
Collapse
|
570
|
Zhang R, Chen HZ, Liu DP. The Four Layers of Aging. Cell Syst 2015; 1:180-6. [PMID: 27135911 DOI: 10.1016/j.cels.2015.09.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 07/06/2015] [Accepted: 09/04/2015] [Indexed: 02/06/2023]
Abstract
Instead of considering aging in terms of discrete hallmarks, we suggest that it operates in four layers, each at a different biological scale. Malfunctions within each layer-and connections between them-produce the aged phenotype and its associated susceptibility to disease.
Collapse
Affiliation(s)
- Ran Zhang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, P.R. China
| | - Hou-Zao Chen
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, P.R. China.
| | - De-Pei Liu
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, P.R. China.
| |
Collapse
|
571
|
Abstract
Many organisms have developed a robust ability to adapt and survive in the face of environmental perturbations that threaten the integrity of their genome, proteome, or metabolome. Studies in multiple model organisms have shown that, in general, when exposed to stress, cells activate a complex prosurvival signaling network that includes immune and DNA damage response genes, chaperones, antioxidant enzymes, structural proteins, metabolic enzymes, and noncoding RNAs. The manner of activation runs the gamut from transcriptional induction of genes to increased stability of transcripts to posttranslational modification of important biosynthetic proteins within the stressed tissue. Superimposed on these largely autonomous effects are nonautonomous responses in which the stressed tissue secretes peptides and other factors that stimulate tissues in different organs to embark on processes that ultimately help the organism as a whole cope with stress. This review focuses on the mechanisms by which tissues in one organ adapt to environmental challenges by regulating stress responses in tissues of different organs.
Collapse
Affiliation(s)
- Edward Owusu-Ansah
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, NY 10032;
| | | |
Collapse
|
572
|
Rivella S. β-thalassemias: paradigmatic diseases for scientific discoveries and development of innovative therapies. Haematologica 2015; 100:418-30. [PMID: 25828088 DOI: 10.3324/haematol.2014.114827] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
β-thalassemias are monogenic disorders characterized by defective synthesis of the β-globin chain, one of the major components of adult hemoglobin. A large number of mutations in the β-globin gene or its regulatory elements have been associated with β-thalassemias. Due to the complexity of the regulation of the β-globin gene and the role of red cells in many physiological processes, patients can manifest a large spectrum of phenotypes, and clinical requirements vary from patient to patient. It is important to consider the major differences in the light of potential novel therapeutics. This review summarizes the main discoveries and mechanisms associated with the synthesis of β-globin and abnormal erythropoiesis, as well as current and novel therapies.
Collapse
Affiliation(s)
- Stefano Rivella
- Department of Pediatrics Hematology-Oncology Department of Cell and Developmental Biology Weill Cornell Medical College New York, NY, USA
| |
Collapse
|
573
|
Smith SC, Zhang X, Zhang X, Gross P, Starosta T, Mohsin S, Franti M, Gupta P, Hayes D, Myzithras M, Kahn J, Tanner J, Weldon SM, Khalil A, Guo X, Sabri A, Chen X, MacDonnell S, Houser SR. GDF11 does not rescue aging-related pathological hypertrophy. Circ Res 2015; 117:926-32. [PMID: 26383970 DOI: 10.1161/circresaha.115.307527] [Citation(s) in RCA: 159] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 09/17/2015] [Indexed: 11/16/2022]
Abstract
RATIONALE Growth differentiation factor 11 (GDF11) is a member of the transforming growth factor-β super family of secreted factors. A recent study showed that reduced GDF11 blood levels with aging was associated with pathological cardiac hypertrophy (PCH) and restoring GDF11 to normal levels in old mice rescued PCH. OBJECTIVE To determine whether and by what mechanism GDF11 rescues aging dependent PCH. METHODS AND RESULTS Twenty-four-month-old C57BL/6 mice were given a daily injection of either recombinant (r) GDF11 at 0.1 mg/kg or vehicle for 28 days. rGDF11 bioactivity was confirmed in vitro. After treatment, rGDF11 levels were significantly increased, but there was no significant effect on either heart weight or body weight. Heart weight/body weight ratios of old mice were not different from 8- or 12-week-old animals, and the PCH marker atrial natriuretic peptide was not different in young versus old mice. Ejection fraction, internal ventricular dimension, and septal wall thickness were not significantly different between rGDF11 and vehicle-treated animals at baseline and remained unchanged at 1, 2, and 4 weeks of treatment. There was no difference in myocyte cross-sectional area rGDF11 versus vehicle-treated old animals. In vitro studies using phenylephrine-treated neonatal rat ventricular myocytes, to explore the putative antihypertrophic effects of GDF11, showed that GDF11 did not reduce neonatal rat ventricular myocytes hypertrophy, but instead induced hypertrophy. CONCLUSIONS Our studies show that there is no age-related PCH in disease-free 24-month-old C57BL/6 mice and that restoring GDF11 in old mice has no effect on cardiac structure or function.
Collapse
Affiliation(s)
- Shavonn C Smith
- From the Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA (S.C.S., Xiaoxiao Zhang, Xiaoying Zhang, P.G., T.S., S Mohsin, X.G., A.S., X.C., S.R.H.); and Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT (M.F., P.G., D.H., M.M., J.K., J.T., S.M.W., A.K., S. MacDonnell)
| | - Xiaoxiao Zhang
- From the Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA (S.C.S., Xiaoxiao Zhang, Xiaoying Zhang, P.G., T.S., S Mohsin, X.G., A.S., X.C., S.R.H.); and Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT (M.F., P.G., D.H., M.M., J.K., J.T., S.M.W., A.K., S. MacDonnell)
| | - Xiaoying Zhang
- From the Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA (S.C.S., Xiaoxiao Zhang, Xiaoying Zhang, P.G., T.S., S Mohsin, X.G., A.S., X.C., S.R.H.); and Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT (M.F., P.G., D.H., M.M., J.K., J.T., S.M.W., A.K., S. MacDonnell)
| | - Polina Gross
- From the Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA (S.C.S., Xiaoxiao Zhang, Xiaoying Zhang, P.G., T.S., S Mohsin, X.G., A.S., X.C., S.R.H.); and Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT (M.F., P.G., D.H., M.M., J.K., J.T., S.M.W., A.K., S. MacDonnell)
| | - Timothy Starosta
- From the Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA (S.C.S., Xiaoxiao Zhang, Xiaoying Zhang, P.G., T.S., S Mohsin, X.G., A.S., X.C., S.R.H.); and Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT (M.F., P.G., D.H., M.M., J.K., J.T., S.M.W., A.K., S. MacDonnell)
| | - Sadia Mohsin
- From the Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA (S.C.S., Xiaoxiao Zhang, Xiaoying Zhang, P.G., T.S., S Mohsin, X.G., A.S., X.C., S.R.H.); and Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT (M.F., P.G., D.H., M.M., J.K., J.T., S.M.W., A.K., S. MacDonnell)
| | - Michael Franti
- From the Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA (S.C.S., Xiaoxiao Zhang, Xiaoying Zhang, P.G., T.S., S Mohsin, X.G., A.S., X.C., S.R.H.); and Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT (M.F., P.G., D.H., M.M., J.K., J.T., S.M.W., A.K., S. MacDonnell)
| | - Priyanka Gupta
- From the Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA (S.C.S., Xiaoxiao Zhang, Xiaoying Zhang, P.G., T.S., S Mohsin, X.G., A.S., X.C., S.R.H.); and Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT (M.F., P.G., D.H., M.M., J.K., J.T., S.M.W., A.K., S. MacDonnell)
| | - David Hayes
- From the Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA (S.C.S., Xiaoxiao Zhang, Xiaoying Zhang, P.G., T.S., S Mohsin, X.G., A.S., X.C., S.R.H.); and Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT (M.F., P.G., D.H., M.M., J.K., J.T., S.M.W., A.K., S. MacDonnell)
| | - Maria Myzithras
- From the Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA (S.C.S., Xiaoxiao Zhang, Xiaoying Zhang, P.G., T.S., S Mohsin, X.G., A.S., X.C., S.R.H.); and Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT (M.F., P.G., D.H., M.M., J.K., J.T., S.M.W., A.K., S. MacDonnell)
| | - Julius Kahn
- From the Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA (S.C.S., Xiaoxiao Zhang, Xiaoying Zhang, P.G., T.S., S Mohsin, X.G., A.S., X.C., S.R.H.); and Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT (M.F., P.G., D.H., M.M., J.K., J.T., S.M.W., A.K., S. MacDonnell)
| | - James Tanner
- From the Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA (S.C.S., Xiaoxiao Zhang, Xiaoying Zhang, P.G., T.S., S Mohsin, X.G., A.S., X.C., S.R.H.); and Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT (M.F., P.G., D.H., M.M., J.K., J.T., S.M.W., A.K., S. MacDonnell)
| | - Steven M Weldon
- From the Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA (S.C.S., Xiaoxiao Zhang, Xiaoying Zhang, P.G., T.S., S Mohsin, X.G., A.S., X.C., S.R.H.); and Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT (M.F., P.G., D.H., M.M., J.K., J.T., S.M.W., A.K., S. MacDonnell)
| | - Ashraf Khalil
- From the Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA (S.C.S., Xiaoxiao Zhang, Xiaoying Zhang, P.G., T.S., S Mohsin, X.G., A.S., X.C., S.R.H.); and Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT (M.F., P.G., D.H., M.M., J.K., J.T., S.M.W., A.K., S. MacDonnell)
| | - Xinji Guo
- From the Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA (S.C.S., Xiaoxiao Zhang, Xiaoying Zhang, P.G., T.S., S Mohsin, X.G., A.S., X.C., S.R.H.); and Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT (M.F., P.G., D.H., M.M., J.K., J.T., S.M.W., A.K., S. MacDonnell)
| | - Abdelkarim Sabri
- From the Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA (S.C.S., Xiaoxiao Zhang, Xiaoying Zhang, P.G., T.S., S Mohsin, X.G., A.S., X.C., S.R.H.); and Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT (M.F., P.G., D.H., M.M., J.K., J.T., S.M.W., A.K., S. MacDonnell)
| | - Xiongwen Chen
- From the Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA (S.C.S., Xiaoxiao Zhang, Xiaoying Zhang, P.G., T.S., S Mohsin, X.G., A.S., X.C., S.R.H.); and Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT (M.F., P.G., D.H., M.M., J.K., J.T., S.M.W., A.K., S. MacDonnell)
| | - Scott MacDonnell
- From the Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA (S.C.S., Xiaoxiao Zhang, Xiaoying Zhang, P.G., T.S., S Mohsin, X.G., A.S., X.C., S.R.H.); and Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT (M.F., P.G., D.H., M.M., J.K., J.T., S.M.W., A.K., S. MacDonnell)
| | - Steven R Houser
- From the Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA (S.C.S., Xiaoxiao Zhang, Xiaoying Zhang, P.G., T.S., S Mohsin, X.G., A.S., X.C., S.R.H.); and Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT (M.F., P.G., D.H., M.M., J.K., J.T., S.M.W., A.K., S. MacDonnell).
| |
Collapse
|
574
|
Sousa-Victor P, Perdiguero E, Muñoz-Cánoves P. Geroconversion of aged muscle stem cells under regenerative pressure. Cell Cycle 2015; 13:3183-90. [PMID: 25485497 DOI: 10.4161/15384101.2014.965072] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Regeneration of skeletal muscle relies on a population of quiescent stem cells (satellite cells) and is impaired in very old (geriatric) individuals undergoing sarcopenia. Stem cell function is essential for organismal homeostasis, providing a renewable source of cells to repair damaged tissues. In adult organisms, age-dependent loss-of-function of tissue-specific stem cells is causally related with a decline in regenerative potential. Although environmental manipulations have shown good promise in the reversal of these conditions, recently we demonstrated that muscle stem cell aging is, in fact, a progressive process that results in persistent and irreversible changes in stem cell intrinsic properties. Global gene expression analyses uncovered an induction of p16(INK4a) in satellite cells of physiologically aged geriatric and progeric mice that inhibits satellite cell-dependent muscle regeneration. Aged satellite cells lose the repression of the INK4a locus, which switches stem cell reversible quiescence into a pre-senescent state; upon regenerative or proliferative pressure, these cells undergo accelerated senescence (geroconversion), through Rb-mediated repression of E2F target genes. p16(INK4a) silencing rejuvenated satellite cells, restoring regeneration in geriatric and progeric muscles. Thus, p16(INK4a)/Rb-driven stem cell senescence is causally implicated in the intrinsic defective regeneration of sarcopenic muscle. Here we discuss on how cellular senescence may be a common mechanism of stem cell aging at the organism level and show that induction of p16(INK4a) in young muscle stem cells through deletion of the Polycomb complex protein Bmi1 recapitulates the geriatric phenotype.
Collapse
|
575
|
Jurberg AD, Vasconcelos-Fontes L, Cotta-de-Almeida V. A Tale from TGF-β Superfamily for Thymus Ontogeny and Function. Front Immunol 2015; 6:442. [PMID: 26441956 PMCID: PMC4564722 DOI: 10.3389/fimmu.2015.00442] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Accepted: 08/14/2015] [Indexed: 12/16/2022] Open
Abstract
Multiple signaling pathways control every aspect of cell behavior, organ formation, and tissue homeostasis throughout the lifespan of any individual. This review takes an ontogenetic view focused on the large superfamily of TGF-β/bone morphogenetic protein ligands to address thymus morphogenesis and function in T cell differentiation. Recent findings on a role of GDF11 for reversing aging-related phenotypes are also discussed.
Collapse
Affiliation(s)
- Arnon Dias Jurberg
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (Fiocruz) , Rio de Janeiro , Brazil ; Graduate Program in Cell and Developmental Biology, Institute of Biomedical Sciences, Federal University of Rio de Janeiro , Rio de Janeiro , Brazil
| | - Larissa Vasconcelos-Fontes
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (Fiocruz) , Rio de Janeiro , Brazil
| | - Vinícius Cotta-de-Almeida
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (Fiocruz) , Rio de Janeiro , Brazil
| |
Collapse
|
576
|
Cottin Y, Rochette L. A member of the TGF-β superfamily, GDF11: functions in the cardiac regeneration, perhaps an "elixir of youth?". Arch Cardiovasc Dis 2015; 108:409-11. [PMID: 26343500 DOI: 10.1016/j.acvd.2015.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 08/10/2015] [Indexed: 11/24/2022]
Affiliation(s)
- Yves Cottin
- Laboratoire de physiopathologie et pharmacologies cardio-métaboliques (LPPCM), 21000 Dijon, France; Service de cardiologie, CHU de Dijon, Inserm UMR866, facultés des sciences de santé, université de Bourgogne, 21000 Dijon, France
| | - Luc Rochette
- Laboratoire de physiopathologie et pharmacologies cardio-métaboliques (LPPCM), 21000 Dijon, France.
| |
Collapse
|
577
|
Tower J. Programmed cell death in aging. Ageing Res Rev 2015; 23:90-100. [PMID: 25862945 DOI: 10.1016/j.arr.2015.04.002] [Citation(s) in RCA: 287] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 03/15/2015] [Accepted: 04/01/2015] [Indexed: 02/08/2023]
Abstract
Programmed cell death (PCD) pathways, including apoptosis and regulated necrosis, are required for normal cell turnover and tissue homeostasis. Mis-regulation of PCD is increasingly implicated in aging and aging-related disease. During aging the cell turnover rate declines for several highly-mitotic tissues. Aging-associated disruptions in systemic and inter-cell signaling combined with cell-autonomous damage and mitochondrial malfunction result in increased PCD in some cell types, and decreased PCD in other cell types. Increased PCD during aging is implicated in immune system decline, skeletal muscle wasting (sarcopenia), loss of cells in the heart, and neurodegenerative disease. In contrast, cancer cells and senescent cells are resistant to PCD, enabling them to increase in abundance during aging. PCD pathways limit life span in fungi, but whether PCD pathways normally limit adult metazoan life span is not yet clear. PCD is regulated by a balance of negative and positive factors, including the mitochondria, which are particularly subject to aging-associated malfunction.
Collapse
|
578
|
Müller L, Pawelec G. As we age: Does slippage of quality control in the immune system lead to collateral damage? Ageing Res Rev 2015; 23:116-23. [PMID: 25676139 DOI: 10.1016/j.arr.2015.01.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 01/16/2015] [Accepted: 01/26/2015] [Indexed: 12/22/2022]
Abstract
The vertebrate adaptive immune system is remarkable for its possession of a very broad range of antigen receptors imbuing the system with exquisite specificity, in addition to the phagocytic and inflammatory cells of the innate system shared with invertebrates. This system requires strict control both at the level of the generation the cells carrying these receptors and at the level of their activation and effector function mediation in order to avoid autoimmunity and mitigate immune pathology. Thus, quality control checkpoints are built into the system at multiple nodes in the response, relying on clonal selection and regulatory networks to maximize pathogen-directed effects and minimize collateral tissue damage. However, these checkpoints are compromised with age, resulting in poorer immune control manifesting as tissue-damaging autoimmune and inflammatory phenomena which can cause widespread systemic disease, paradoxically compounding the problems associated with increased susceptibility to infectious disease and possibly cancer in the elderly. Better understanding the reasons for slippage of immune control will pave the way for developing rational strategies for interventions to maintain appropriate immunity while reducing immunopathology.
Collapse
|
579
|
Hommerding CJ, Childs BG, Baker DJ. The Role of Stem Cell Genomic Instability in Aging. CURRENT STEM CELL REPORTS 2015. [DOI: 10.1007/s40778-015-0020-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
580
|
Mendelsohn AR, Larrick JW. Systemic factors mediate reversible age-associated brain dysfunction. Rejuvenation Res 2015; 17:525-8. [PMID: 25400086 DOI: 10.1089/rej.2014.1643] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Brain function declines in aging mammals. Recent work has identified dysregulation of key blood-borne factors whose altered expression during aging diminishes brain function in mice. Increased C-C motif chemokine 11 (CCL11) expression with aging is detrimental to brain function. On the other hand, plasma levels of the trophic factor growth/differentiation factor 11 (GDF11) decrease with aging. Restoration of youthful levels of GDF11 by injection partially restores brain function and neurogenesis by improving endothelial cell function and vasculature. Moreover, GDF11 has a rejuvenative effect on cardiac and skeletal muscle. Decreased type II interferon (IFN-II) and increased type I interferon (IFN-I) signaling during aging at the choroid plexus (CP), which constitutes the brain-cerebrospinal fluid barrier (B-CSF-B), negatively effects brain function. Blood from young mice contains factors that restore IFN-II levels. IFN-II is required for maintenance of the CP, and low IFN-II levels are associated with decreased cognitive abilities. IFN-I levels appear to drive increased CCL11 expression through the CSF. Blood from young animals does not restore IFN-I levels. However, injecting anti-interferon-α/β receptor (IFNAR) antibodies into the CSF inhibits downstream IFN-I gene and protein expression and decreases expression of CCL11, partially restoring neurogenesis and cognitive function. These results suggest that IFN-I plays a critical role in increasing CCL11 during aging of the brain. An emerging theme is that aging-associated loss of function in mammals may involve a set of defined, potentially reversible changes in many tissues and organs, including the brain, permitting development of potential rejuvenative therapies.
Collapse
Affiliation(s)
- Andrew R Mendelsohn
- Panorama Research Institute and Regenerative Sciences Institute , Sunnyvale, California
| | | |
Collapse
|
581
|
Lee KY, Singh MK, Ussar S, Wetzel P, Hirshman MF, Goodyear LJ, Kispert A, Kahn CR. Tbx15 controls skeletal muscle fibre-type determination and muscle metabolism. Nat Commun 2015; 6:8054. [PMID: 26299309 PMCID: PMC4552045 DOI: 10.1038/ncomms9054] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 07/13/2015] [Indexed: 01/15/2023] Open
Abstract
Skeletal muscle is composed of both slow-twitch oxidative myofibers and fast-twitch glycolytic myofibers that differentially impact muscle metabolism, function and eventually whole-body physiology. Here we show that the mesodermal transcription factor T-box 15 (Tbx15) is highly and specifically expressed in glycolytic myofibers. Ablation of Tbx15 in vivo leads to a decrease in muscle size due to a decrease in the number of glycolytic fibres, associated with a small increase in the number of oxidative fibres. This shift in fibre composition results in muscles with slower myofiber contraction and relaxation, and also decreases whole-body oxygen consumption, reduces spontaneous activity, increases adiposity and glucose intolerance. Mechanistically, ablation of Tbx15 leads to activation of AMPK signalling and a decrease in Igf2 expression. Thus, Tbx15 is one of a limited number of transcription factors to be identified with a critical role in regulating glycolytic fibre identity and muscle metabolism. The transcriptional regulator Tbx15 has a role in organ development. Here Lee et al. show that Tbx15 influences fibre-type determination in murine skeletal muscles, explaining local and systemic metabolic derangements in heterozygous Tbx15 knockout mice.
Collapse
Affiliation(s)
- Kevin Y Lee
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, 1 Joslin Plaza, Boston, Massachusetts 02215, USA
| | - Manvendra K Singh
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany.,Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-NUS Graduate Medical School Singapore, National Heart Centre Singapore, 8 College Road, Singapore 169857, Singapore
| | - Siegfried Ussar
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, 1 Joslin Plaza, Boston, Massachusetts 02215, USA.,Institute for Diabetes and Obesity, Helmholtz Center, Parkring, 1385748 Munich/Garching, Germany
| | - Petra Wetzel
- Zentrum Physiologie, Medizinische Hochschule Hannover, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany
| | - Michael F Hirshman
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, 1 Joslin Plaza, Boston, Massachusetts 02215, USA
| | - Laurie J Goodyear
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, 1 Joslin Plaza, Boston, Massachusetts 02215, USA
| | - Andreas Kispert
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany
| | - C Ronald Kahn
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, 1 Joslin Plaza, Boston, Massachusetts 02215, USA
| |
Collapse
|
582
|
Affiliation(s)
- Vishal K Patel
- Medical School, Albert Einstein College of Medicine Bronx, NY 10461, USA
| | | |
Collapse
|
583
|
Olson KA, Beatty AL, Heidecker B, Regan MC, Brody EN, Foreman T, Kato S, Mehler RE, Singer BS, Hveem K, Dalen H, Sterling DG, Lawn RM, Schiller NB, Williams SA, Whooley MA, Ganz P. Association of growth differentiation factor 11/8, putative anti-ageing factor, with cardiovascular outcomes and overall mortality in humans: analysis of the Heart and Soul and HUNT3 cohorts. Eur Heart J 2015; 36:3426-34. [PMID: 26294790 DOI: 10.1093/eurheartj/ehv385] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 07/21/2015] [Indexed: 01/19/2023] Open
Abstract
AIMS Growth differentiation factor 11 and/or its homologue growth differentiation factor 8 (GDF11/8) reverses age-related cardiac hypertrophy and vascular ageing in mice. We investigated whether GDF11/8 associates with cardiovascular outcomes, left ventricular hypertrophy (LVH), or age in humans. METHODS AND RESULTS We measured plasma GDF11/8 levels in 928 participants with stable ischaemic heart disease in the Heart and Soul study. We adjudicated heart failure hospitalization, stroke, myocardial infarction, death, and their composite endpoint. Left ventricular hypertrophy was evaluated by echocardiography. We used multivariable Cox proportional hazards models to compare rates of cardiovascular events and death across GDF11/8 quartiles and logistic regression models to evaluate the association between GDF11/8 and LVH. Four hundred and fifty participants (48.5%) experienced a cardiovascular event or death during 8.9 years of follow-up. The adjusted risk of the composite endpoint was lower in the highest compared with the lowest GDF11/8 quartile [hazard ratio (HR), 0.45; 95% confidence interval (CI), 0.33-0.60; P < 0.001]. We replicated this relationship of GDF11/8 to adverse events in 971 participants in the HUNT3 cohort (adjusted HR, 0.34; 95% CI, 0.23-0.51; P < 0.001). Left ventricular hypertrophy was present in 368 participants (39.7%) at baseline. Participants in the highest quartile of GDF11/8 were less likely to have LVH than those in the lowest quartile (adjusted OR, 0.55; 95% CI, 0.35-0.86; P = 0.009). GDF11/8 levels were lower in older individuals (P < 0.001). CONCLUSION In patients with stable ischaemic heart disease, higher GDF11/8 levels are associated with lower risk of cardiovascular events and death. Our findings suggest that GDF11/8 has similar cardioprotective properties in humans to those demonstrated in mice.
Collapse
Affiliation(s)
- Kristoff A Olson
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Alexis L Beatty
- Department of Medicine, University of California, San Francisco, CA, USA Cardiology Section, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA Department of Medicine, University of Washington, Seattle, WA, USA
| | - Bettina Heidecker
- Department of Medicine, University of California, San Francisco, CA, USA
| | | | | | | | | | | | | | - Kristian Hveem
- Lifandis AS, Norway HUNT Research Center, Department of Public Health, NTNU, Trondheim, Norway
| | - Havard Dalen
- Department of Medicine, Levanger Hospital, Nord-Trøndelag Health Trust, Levanger, Norway MI Lab and Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Norway
| | | | | | - Nelson B Schiller
- Department of Medicine, University of California, San Francisco, CA, USA
| | | | - Mary A Whooley
- Department of Medicine, University of California, San Francisco, CA, USA Veterans Affairs Medical Center, San Francisco, CA, USA Department of Epidemiology and Biostatistics, University of California, San Francisco, USA
| | - Peter Ganz
- Department of Medicine, University of California, San Francisco, CA, USA Division of Cardiology, San Francisco General Hospital, San Francisco, CA, USA
| |
Collapse
|
584
|
|
585
|
Rachul CM, Percec I, Caulfield T. The Fountain of Stem Cell-Based Youth? Online Portrayals of Anti-Aging Stem Cell Technologies. Aesthet Surg J 2015; 35:730-6. [PMID: 25922365 DOI: 10.1093/asj/sju111] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/24/2014] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The hype surrounding stem cell science has created a market opportunity for the cosmetic industry. Cosmetic and anti-aging products and treatments that make claims regarding stem cell technology are increasingly popular, despite a lack of evidence for safety and efficacy of such products. OBJECTIVES This study explores how stem cell-based products and services are portrayed to the public through online sources, in order to gain insight into the key messages available to consumers. METHODS A content analysis of 100 web pages was conducted to examine the portrayals of stem cell-based cosmetic and anti-aging products and treatments. A qualitative discourse analysis of one web page further examined how language contributes to the portrayals of these products and treatments to public audiences. RESULTS The majority of web pages portrayed stem cell-based products as ready for public use. Very few web pages substantiated claims with scientific evidence, and even fewer mentioned any risks or limitations associated with stem cell science. The discourse analysis revealed that the framing and use of metaphor obscures the certainty of the efficacy of and length of time for stem cell-based anti-aging technology to be publicly available. CONCLUSIONS This study highlights the need to educate patients and the public on the current limits of stem cell applications in this context. In addition, generating scientific evidence for stem cell-based anti-aging and aesthetic applications is needed for optimizing benefits and minimizing adverse effects for the public. Having more evidence on efficacy and risks will help to protect patients who are eagerly seeking out these treatments.
Collapse
Affiliation(s)
- Christen M Rachul
- Ms Rachul is a PhD Candidate in Applied Linguistics and Language Studies, School of Linguistics and Language Studies, Carleton University, Ottawa, ON, Canada. Dr Percec is an Assistant Professor, Division of Plastic Surgery, Epigenetics Program, University of Pennsylvania, Philadelphia, PA. Professor Caulfield is the Canada Research Chair in Health Law and Policy; Trudeau Fellow and Professor, Faculty of Law and School of Public Health; and Research Director, Health Law Institute, University of Alberta, Edmonton, AB, Canada
| | - Ivona Percec
- Ms Rachul is a PhD Candidate in Applied Linguistics and Language Studies, School of Linguistics and Language Studies, Carleton University, Ottawa, ON, Canada. Dr Percec is an Assistant Professor, Division of Plastic Surgery, Epigenetics Program, University of Pennsylvania, Philadelphia, PA. Professor Caulfield is the Canada Research Chair in Health Law and Policy; Trudeau Fellow and Professor, Faculty of Law and School of Public Health; and Research Director, Health Law Institute, University of Alberta, Edmonton, AB, Canada
| | - Timothy Caulfield
- Ms Rachul is a PhD Candidate in Applied Linguistics and Language Studies, School of Linguistics and Language Studies, Carleton University, Ottawa, ON, Canada. Dr Percec is an Assistant Professor, Division of Plastic Surgery, Epigenetics Program, University of Pennsylvania, Philadelphia, PA. Professor Caulfield is the Canada Research Chair in Health Law and Policy; Trudeau Fellow and Professor, Faculty of Law and School of Public Health; and Research Director, Health Law Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
586
|
Crane JD, MacNeil LG, Lally JS, Ford RJ, Bujak AL, Brar IK, Kemp BE, Raha S, Steinberg GR, Tarnopolsky MA. Exercise-stimulated interleukin-15 is controlled by AMPK and regulates skin metabolism and aging. Aging Cell 2015; 14:625-34. [PMID: 25902870 PMCID: PMC4531076 DOI: 10.1111/acel.12341] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/14/2015] [Indexed: 12/28/2022] Open
Abstract
Aging is commonly associated with a structural deterioration of skin that compromises its barrier function, healing, and susceptibility to disease. Several lines of evidence show that these changes are driven largely by impaired tissue mitochondrial metabolism. While exercise is associated with numerous health benefits, there is no evidence that it affects skin tissue or that endocrine muscle-to-skin signaling occurs. We demonstrate that endurance exercise attenuates age-associated changes to skin in humans and mice and identify exercise-induced IL-15 as a novel regulator of mitochondrial function in aging skin. We show that exercise controls IL-15 expression in part through skeletal muscle AMP-activated protein kinase (AMPK), a central regulator of metabolism, and that the elimination of muscle AMPK causes a deterioration of skin structure. Finally, we establish that daily IL-15 therapy mimics some of the anti-aging effects of exercise on muscle and skin in mice. Thus, we elucidate a mechanism by which exercise confers health benefits to skin and suggest that low-dose IL-15 therapy may prove to be a beneficial strategy to attenuate skin aging.
Collapse
Affiliation(s)
- Justin D Crane
- Department of Kinesiology, McMaster UniversityHamilton, Ontario, Canada
- Department of Pediatrics, McMaster UniversityHamilton, Ontario, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster UniversityHamilton, Ontario, Canada
| | - Lauren G MacNeil
- Department of Pediatrics, McMaster UniversityHamilton, Ontario, Canada
| | - James S Lally
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster UniversityHamilton, Ontario, Canada
| | - Rebecca J Ford
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster UniversityHamilton, Ontario, Canada
| | - Adam L Bujak
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster UniversityHamilton, Ontario, Canada
| | - Ikdip K Brar
- Department of Pediatrics, McMaster UniversityHamilton, Ontario, Canada
| | - Bruce E Kemp
- Department of Medicine, St. Vincent’s Institute of Medical Research, University of MelbourneFitzroy, Vic., Australia
| | - Sandeep Raha
- Department of Pediatrics, McMaster UniversityHamilton, Ontario, Canada
| | - Gregory R Steinberg
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster UniversityHamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster UniversityHamilton, Ontario, Canada
| | | |
Collapse
|
587
|
Fadini GP, Menegazzo L, Bonora BM, Mazzucato M, Persano S, Vigili de Kreutzenberg S, Avogaro A. Effects of Age, Diabetes, and Vascular Disease on Growth Differentiation Factor 11: First-in-Human Study. Diabetes Care 2015; 38:e118-9. [PMID: 26116718 DOI: 10.2337/dc15-0868] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 05/01/2015] [Indexed: 02/03/2023]
Affiliation(s)
- Gian Paolo Fadini
- Department of Medicine, University of Padova, Padova, Italy Venetian Institute of Molecular Medicine, Padova, Italy
| | - Lisa Menegazzo
- Department of Medicine, University of Padova, Padova, Italy Venetian Institute of Molecular Medicine, Padova, Italy
| | | | | | - Sara Persano
- Department of Medicine, University of Padova, Padova, Italy
| | | | - Angelo Avogaro
- Department of Medicine, University of Padova, Padova, Italy Venetian Institute of Molecular Medicine, Padova, Italy
| |
Collapse
|
588
|
Farup J, Madaro L, Puri PL, Mikkelsen UR. Interactions between muscle stem cells, mesenchymal-derived cells and immune cells in muscle homeostasis, regeneration and disease. Cell Death Dis 2015. [PMID: 26203859 PMCID: PMC4650743 DOI: 10.1038/cddis.2015.198] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Recent evidence has revealed the importance of reciprocal functional interactions between different types of mononuclear cells in coordinating the repair of injured muscles. In particular, signals released from the inflammatory infiltrate and from mesenchymal interstitial cells (also known as fibro-adipogenic progenitors (FAPs)) appear to instruct muscle stem cells (satellite cells) to break quiescence, proliferate and differentiate. Interestingly, conditions that compromise the functional integrity of this network can bias muscle repair toward pathological outcomes that are typically observed in chronic muscular disorders, that is, fibrotic and fatty muscle degeneration as well as myofiber atrophy. In this review, we will summarize the current knowledge on the regulation of this network in physiological and pathological conditions, and anticipate the potential contribution of its cellular components to relatively unexplored conditions, such as aging and physical exercise.
Collapse
Affiliation(s)
- J Farup
- Section for Sports Science, Institute of Public Health, Aarhus University, Aarhus, Denmark
| | - L Madaro
- 1] Sanford-Burnham Medical Research Institute, Sanford Children's Health Research Center, La Jolla, CA, USA [2] IRCCS Fondazione Santa Lucia, Rome, Italy
| | - P L Puri
- 1] Sanford-Burnham Medical Research Institute, Sanford Children's Health Research Center, La Jolla, CA, USA [2] IRCCS Fondazione Santa Lucia, Rome, Italy
| | - U R Mikkelsen
- 1] Section for Sports Science, Institute of Public Health, Aarhus University, Aarhus, Denmark [2] Institute of Sports Medicine, Department of Orthopaedic Surgery M, Bispebjerg Hospital and Center for Healthy Aging, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
589
|
Mulloy B, Rider CC. The Bone Morphogenetic Proteins and Their Antagonists. VITAMINS AND HORMONES 2015; 99:63-90. [PMID: 26279373 DOI: 10.1016/bs.vh.2015.06.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The bone morphogenetic proteins (BMPs) and the growth and differentiation factors comprise a single family of some 20 homologous, dimeric cytokines which share the cystine-knot domain typical of the TGF-β superfamily. They control the differentiation and activity of a range of cell types, including many outside bone and cartilage. They serve as developmental morphogens, but are also important in chronic pathologies, including tissue fibrosis and cancer. One mechanism for enabling tight spatiotemporal control of their activities is through a number of antagonist proteins, including Noggin, Follistatin, Chordin, Twisted gastrulation (TSG), and the seven members of the Cerberus and Dan family. These antagonists are secreted proteins that bind selectively to particular BMPs with high affinity, thereby blocking receptor engagement and signaling. Most of these antagonists also possess a TGF-β cystine-knot domain. Here, we discuss current knowledge and understanding of the structures and activities of the BMPs and their antagonists, with a particular focus on the latter proteins. Recent advances in structural biology of BMP antagonists have begun the process of elucidating the molecular basis of their activity, displaying a surprising variety between the modes of action of these closely related proteins. We also discuss the interactions of the antagonists with the glycosaminoglycan heparan sulfate, which is found ubiquitously on cell surfaces and in the extracellular matrix.
Collapse
Affiliation(s)
- Barbara Mulloy
- Centre for Biomedical Sciences, School of Biological Sciences, Royal Holloway, University of London, Egham, Surrey, United Kingdom
| | - Chris C Rider
- Centre for Biomedical Sciences, School of Biological Sciences, Royal Holloway, University of London, Egham, Surrey, United Kingdom.
| |
Collapse
|
590
|
GDF11 Increases with Age and Inhibits Skeletal Muscle Regeneration. Cell Metab 2015; 22:164-74. [PMID: 26001423 PMCID: PMC4497834 DOI: 10.1016/j.cmet.2015.05.010] [Citation(s) in RCA: 423] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 04/23/2015] [Accepted: 05/06/2015] [Indexed: 12/15/2022]
Abstract
Age-related frailty may be due to decreased skeletal muscle regeneration. The role of TGF-β molecules myostatin and GDF11 in regeneration is unclear. Recent studies showed an age-related decrease in GDF11 and that GDF11 treatment improves muscle regeneration, which were contrary to prior studies. We now show that these recent claims are not reproducible and the reagents previously used to detect GDF11 are not GDF11 specific. We develop a GDF11-specific immunoassay and show a trend toward increased GDF11 levels in sera of aged rats and humans. GDF11 mRNA increases in rat muscle with age. Mechanistically, GDF11 and myostatin both induce SMAD2/3 phosphorylation, inhibit myoblast differentiation, and regulate identical downstream signaling. GDF11 significantly inhibited muscle regeneration and decreased satellite cell expansion in mice. Given early data in humans showing a trend for an age-related increase, GDF11 could be a target for pharmacologic blockade to treat age-related sarcopenia.
Collapse
|
591
|
Abstract
Identifying candidates that rejuvenate aged muscle stem cells is an important strategy toward developing therapies to treat age-related diseases. In this issue, Egerman et al. (2015) re-investigate the activity of GDF11 in myogenesis, recently suggested as an anti-aging agent, and instead find a potent inhibitory effect on skeletal muscle regeneration.
Collapse
Affiliation(s)
- Caroline E Brun
- Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, Regenerative Medicine Program, 501 Smyth Road, Ottawa, ON K1H 8L6, Canada; Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 501 Smyth Road, Ottawa, ON K1H 8L6, Canada
| | - Michael A Rudnicki
- Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, Regenerative Medicine Program, 501 Smyth Road, Ottawa, ON K1H 8L6, Canada; Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 501 Smyth Road, Ottawa, ON K1H 8L6, Canada.
| |
Collapse
|
592
|
A Periodic Diet that Mimics Fasting Promotes Multi-System Regeneration, Enhanced Cognitive Performance, and Healthspan. Cell Metab 2015; 22:86-99. [PMID: 26094889 PMCID: PMC4509734 DOI: 10.1016/j.cmet.2015.05.012] [Citation(s) in RCA: 603] [Impact Index Per Article: 60.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 04/02/2015] [Accepted: 05/08/2015] [Indexed: 12/14/2022]
Abstract
Prolonged fasting (PF) promotes stress resistance, but its effects on longevity are poorly understood. We show that alternating PF and nutrient-rich medium extended yeast lifespan independently of established pro-longevity genes. In mice, 4 days of a diet that mimics fasting (FMD), developed to minimize the burden of PF, decreased the size of multiple organs/systems, an effect followed upon re-feeding by an elevated number of progenitor and stem cells and regeneration. Bi-monthly FMD cycles started at middle age extended longevity, lowered visceral fat, reduced cancer incidence and skin lesions, rejuvenated the immune system, and retarded bone mineral density loss. In old mice, FMD cycles promoted hippocampal neurogenesis, lowered IGF-1 levels and PKA activity, elevated NeuroD1, and improved cognitive performance. In a pilot clinical trial, three FMD cycles decreased risk factors/biomarkers for aging, diabetes, cardiovascular disease, and cancer without major adverse effects, providing support for the use of FMDs to promote healthspan.
Collapse
|
593
|
Rea IM, Dellet M, Mills KI. Living long and ageing well: is epigenomics the missing link between nature and nurture? Biogerontology 2015; 17:33-54. [PMID: 26133292 DOI: 10.1007/s10522-015-9589-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Accepted: 06/22/2015] [Indexed: 12/12/2022]
Abstract
Human longevity is a complex trait and increasingly we understand that both genes and lifestyle interact in the longevity phenotype. Non-genetic factors, including diet, physical activity, health habits, and psychosocial factors contribute approximately 50% of the variability in human lifespan with another 25% explained by genetic differences. Family clusters of nonagenarian and centenarian siblings, who show both exceptional age-span and health-span, are likely to have inherited facilitatory gene groups, but also have nine decades of life experiences and behaviours which have interacted with their genetic profiles. Identification of their shared genes is just one small step in the link from genes to their physical and psychological profiles. Behavioural genomics is beginning to demonstrate links to biological mechanisms through regulation of gene expression, which directs the proteome and influences the personal phenotype. Epigenetics has been considered the missing link between nature and nurture. Although there is much that remains to be discovered, this article will discuss some of genetic and environmental factors which appear important in good quality longevity and link known epigenetic mechanisms to themes identified by nonagenarians themselves related to their longevity. Here we suggest that exceptional 90-year old siblings have adopted a range of behaviours and life-styles which have contributed to their ageing-well-phenotype and which link with important public health messages.
Collapse
Affiliation(s)
- Irene Maeve Rea
- School of Medicine, Dentistry and Biomedical Science, Queens University Belfast, Belfast, Northern Ireland, UK. .,School of Biomedical Sciences, University of Ulster, Coleraine, Northern Ireland, UK.
| | - Margaret Dellet
- School of Medicine, Dentistry and Biomedical Science, Queens University Belfast, Belfast, Northern Ireland, UK.,Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queens University Belfast , Belfast, Northern Ireland, UK
| | - Ken I Mills
- School of Medicine, Dentistry and Biomedical Science, Queens University Belfast, Belfast, Northern Ireland, UK.,Centre for Cancer Research and Cell Biology, School of Medicine, Dentistry and Biomedical Science, Queens University Belfast, Belfast, Northern Ireland, UK
| | | |
Collapse
|
594
|
Liu H, Xia X, Li B. Mesenchymal stem cell aging: Mechanisms and influences on skeletal and non-skeletal tissues. Exp Biol Med (Maywood) 2015; 240:1099-106. [PMID: 26088863 DOI: 10.1177/1535370215591828] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The aging population and the incidence of aging-related diseases such as osteoporosis are on the rise. Aging at the tissue and organ levels usually involves tissue stem cells. Human and animal model studies indicate that aging affects two aspects of mesenchymal stem cell (MSC): a decrease in the bone marrow MSC pool and biased differentiation into adipocyte at the cost of osteoblast, which underlie the etiology of osteoporosis. Aging of MSC cells is also detrimental to some non-skeletal tissues, in particular the hematopoietic system, where MSCs serve as a niche component. In addition, aging compromises the therapeutic potentials of MSC cells, including cells isolated from aged individuals or cells cultured for many passages. Here we discuss the recent progress on our understanding of MSC aging, with a focus on the effects of MSC aging on bone remodeling and hematopoiesis and the mechanisms of MSC aging.
Collapse
Affiliation(s)
- Huijuan Liu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xuechun Xia
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Baojie Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
595
|
Abstract
Stem cell decline is an important cellular driver of aging-associated pathophysiology in multiple tissues. Epigenetic regulation is central to establishing and maintaining stem cell function, and emerging evidence indicates that epigenetic dysregulation contributes to the altered potential of stem cells during aging. Unlike terminally differentiated cells, the impact of epigenetic dysregulation in stem cells is propagated beyond self; alterations can be heritably transmitted to differentiated progeny, in addition to being perpetuated and amplified within the stem cell pool through self-renewal divisions. This Review focuses on recent studies examining epigenetic regulation of tissue-specific stem cells in homeostasis, aging, and aging-related disease.
Collapse
Affiliation(s)
- Isabel Beerman
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; Program in Cellular and Molecular Medicine, Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02116, USA
| | - Derrick J Rossi
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; Program in Cellular and Molecular Medicine, Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02116, USA.
| |
Collapse
|
596
|
Sousa-Victor P, García-Prat L, Serrano AL, Perdiguero E, Muñoz-Cánoves P. Muscle stem cell aging: regulation and rejuvenation. Trends Endocrinol Metab 2015; 26:287-96. [PMID: 25869211 DOI: 10.1016/j.tem.2015.03.006] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 03/18/2015] [Accepted: 03/18/2015] [Indexed: 01/17/2023]
Abstract
Aging is characterized by a progressive decline of physiological integrity leading to the loss of tissue function and vulnerability to disease, but its causes remain poorly understood. Skeletal muscle has an outstanding regenerative capacity that relies on its resident stem cells (satellite cells). This capacity declines with aging, and recent discoveries have redefined our view of why this occurs. Here, we discuss how an interconnection of extrinsic changes in the systemic and local environment and cell-intrinsic mechanisms might provoke failure of normal muscle stem cell functions with aging. We focus particularly on the emergent biology of rejuvenation of old satellite cells, including cells of geriatric age, by restoring traits of youthfulness, with the final goal of improving human health during aging.
Collapse
Affiliation(s)
- Pedro Sousa-Victor
- Buck Institute for Research on Aging, Novato, CA, USA; Cell Biology Group, Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative diseases (CIBERNED), Barcelona, Spain
| | - Laura García-Prat
- Cell Biology Group, Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative diseases (CIBERNED), Barcelona, Spain
| | - Antonio L Serrano
- Cell Biology Group, Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative diseases (CIBERNED), Barcelona, Spain
| | - Eusebio Perdiguero
- Cell Biology Group, Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative diseases (CIBERNED), Barcelona, Spain
| | - Pura Muñoz-Cánoves
- Cell Biology Group, Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative diseases (CIBERNED), Barcelona, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain.
| |
Collapse
|
597
|
Cell-autonomous decrease in proliferative competitiveness of the aged hepatocyte. J Hepatol 2015; 62:1341-8. [PMID: 25617502 DOI: 10.1016/j.jhep.2015.01.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 12/03/2014] [Accepted: 01/04/2015] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS The regenerative potential of the liver declines with age, this might be dependent on a decrease in the intensity of the stimulus and/or an increased refractoriness of the target. In the present study, we compared the in vivo growth capacity of young and old hepatocytes transplanted into the same host. METHODS We utilized the retrorsine (RS)-based model for liver repopulation, which provides a specific and effective stimulus for transplanted hepatocytes. Rats of the dipeptidyl-peptidase type IV (DPP-IV)-deficient strain were given RS and were injected with a mix of hepatocytes isolated from either a 2-month old or an 18-month old donor. To follow the fate of transplanted cells, they were each identified through a specific tag: young hepatocytes expressed the green fluorescent protein (GFP(+)), while those from old donors were DPP-IV-positive. RESULTS At 1 month post-transplantation, DPP-IV-positive clusters (derived from old donor) were consistently smaller than those GFP(+) (young donor); the cross sectional area of clusters was decreased by 50%, while the mean volume was reduced to 1/3. Furthermore, when 2/3 partial hepatectomy (PH) was performed, the S-phase response of old hepatocyte-derived clusters was only 30-40% compared to that observed in cluster originating from young hepatocytes. No markers of cell senescence were expressed in clusters of transplanted hepatocytes. CONCLUSIONS This is the first direct evidence in vivo that hepatocytes in the aged liver express a cell-autonomous decline in their replicative capacity and in their regenerative response to PH compared to those from a young animal.
Collapse
|
598
|
Prognostic Significance and Determinants of the 6-Min Walk Test in Patients With Heart Failure and Preserved Ejection Fraction. JACC-HEART FAILURE 2015; 3:459-466. [DOI: 10.1016/j.jchf.2015.01.010] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Revised: 12/15/2014] [Accepted: 01/09/2015] [Indexed: 01/09/2023]
|
599
|
Ikemoto-Uezumi M, Uezumi A, Tsuchida K, Fukada SI, Yamamoto H, Yamamoto N, Shiomi K, Hashimoto N. Pro-Insulin-Like Growth Factor-II Ameliorates Age-Related Inefficient Regenerative Response by Orchestrating Self-Reinforcement Mechanism of Muscle Regeneration. Stem Cells 2015; 33:2456-68. [PMID: 25917344 DOI: 10.1002/stem.2045] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2014] [Revised: 03/03/2015] [Accepted: 03/28/2015] [Indexed: 12/14/2022]
Abstract
Sarcopenia, age-related muscle weakness, increases the frequency of falls and fractures in elderly people, which can trigger severe muscle injury. Rapid and successful recovery from muscle injury is essential not to cause further frailty and loss of independence. In fact, we showed insufficient muscle regeneration in aged mice. Although the number of satellite cells, muscle stem cells, decreases with age, the remaining satellite cells maintain the myogenic capacity equivalent to young mice. Transplantation of young green fluorescent protein (GFP)-Tg mice-derived satellite cells into young and aged mice revealed that age-related deterioration of the muscle environment contributes to the decline in regenerative capacity of satellite cells. Thus, extrinsic changes rather than intrinsic changes in satellite cells appear to be a major determinant of inefficient muscle regeneration with age. Comprehensive protein expression analysis identified a decrease in insulin-like growth factor-II (IGF-II) level in regenerating muscle of aged mice. We found that pro- and big-IGF-II but not mature IGF-II specifically express during muscle regeneration and the expressions are not only delayed but also decreased in absolute quantity with age. Supplementation of pro-IGF-II in aged mice ameliorated the inefficient regenerative response by promoting proliferation of satellite cells, angiogenesis, and suppressing adipogenic differentiation of platelet derived growth factor receptor (PDGFR)α(+) mesenchymal progenitors. We further revealed that pro-IGF-II but not mature IGF-II specifically inhibits the pathological adipogenesis of PDGFRα(+) cells. Together, these results uncovered a distinctive pro-IGF-II-mediated self-reinforcement mechanism of muscle regeneration and suggest that supplementation of pro-IGF-II could be one of the most effective therapeutic approaches for muscle injury in elderly people.
Collapse
Affiliation(s)
- Madoka Ikemoto-Uezumi
- Department of Regenerative Medicine, Research Institute, National Center for Geriatrics and Gerontology, Aichi, Japan
| | - Akiyoshi Uezumi
- Division for Therapies against Intractable Diseases, Institute for Comprehensive Medical Science, Fujita Health University, Aichi, Japan
| | - Kunihiro Tsuchida
- Division for Therapies against Intractable Diseases, Institute for Comprehensive Medical Science, Fujita Health University, Aichi, Japan
| | - So-ichiro Fukada
- Laboratory of Molecular and Cellular Physiology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Hiroshi Yamamoto
- Faculty of Pharmaceutical Sciences, Kobe Gakuin University, Kobe, Japan
| | - Naoki Yamamoto
- Laboratory of Molecular Biology and Histochemistry, Fujita Health University Joint Research Laboratory, Aichi, Japan
| | - Kosuke Shiomi
- Department of Regenerative Medicine, Research Institute, National Center for Geriatrics and Gerontology, Aichi, Japan
| | - Naohiro Hashimoto
- Department of Regenerative Medicine, Research Institute, National Center for Geriatrics and Gerontology, Aichi, Japan
| |
Collapse
|
600
|
|