51
|
Abstract
DNA damage response (DDR) and DNA repair pathways determine neoplastic cell transformation and therapeutic responses, as well as the aging process. Altered DDR functioning results in accumulation of unrepaired DNA damage, increased frequency of tumorigenic mutations, and premature aging. Recent evidence suggests that polypeptide hormones play a role in modulating DDR and DNA damage repair, while DNA damage accumulation may also affect hormonal status. We review the available reports elucidating involvement of insulin-like growth factor 1 (IGF1), growth hormone (GH), α-melanocyte stimulating hormone (αMSH), and gonadotropin-releasing hormone (GnRH)/gonadotropins in DDR and DNA repair as well as the current understanding of pathways enabling these actions. We discuss effects of DNA damage pathway mutations, including Fanconi anemia, on endocrine function and consider mechanisms underlying these phenotypes. (Endocrine Reviews 41: 1 - 19, 2020).
Collapse
Affiliation(s)
- Vera Chesnokova
- Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Shlomo Melmed
- Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| |
Collapse
|
52
|
Corrado A, Cici D, Rotondo C, Maruotti N, Cantatore FP. Molecular Basis of Bone Aging. Int J Mol Sci 2020; 21:ijms21103679. [PMID: 32456199 PMCID: PMC7279376 DOI: 10.3390/ijms21103679] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/18/2020] [Accepted: 05/21/2020] [Indexed: 12/16/2022] Open
Abstract
A decline in bone mass leading to an increased fracture risk is a common feature of age-related bone changes. The mechanisms underlying bone senescence are very complex and implicate systemic and local factors and are the result of the combination of several changes occurring at the cellular, tissue and structural levels; they include alterations of bone cell differentiation and activity, oxidative stress, genetic damage and the altered responses of bone cells to various biological signals and to mechanical loading. The molecular mechanisms responsible for these changes remain greatly unclear and many data derived from in vitro or animal studies appear to be conflicting and heterogeneous, probably due to the different experimental approaches; nevertheless, understanding the main physio-pathological processes that cause bone senescence is essential for the development of new potential therapeutic options for treating age-related bone loss. This article reviews the current knowledge concerning the molecular mechanisms underlying the pathogenesis of age-related bone changes.
Collapse
|
53
|
Pao PC, Patnaik D, Watson LA, Gao F, Pan L, Wang J, Adaikkan C, Penney J, Cam HP, Huang WC, Pantano L, Lee A, Nott A, Phan TX, Gjoneska E, Elmsaouri S, Haggarty SJ, Tsai LH. HDAC1 modulates OGG1-initiated oxidative DNA damage repair in the aging brain and Alzheimer's disease. Nat Commun 2020; 11:2484. [PMID: 32424276 PMCID: PMC7235043 DOI: 10.1038/s41467-020-16361-y] [Citation(s) in RCA: 139] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 04/28/2020] [Indexed: 02/08/2023] Open
Abstract
DNA damage contributes to brain aging and neurodegenerative diseases. However, the factors stimulating DNA repair to stave off functional decline remain obscure. We show that HDAC1 modulates OGG1-initated 8-oxoguanine (8-oxoG) repair in the brain. HDAC1-deficient mice display age-associated DNA damage accumulation and cognitive impairment. HDAC1 stimulates OGG1, a DNA glycosylase known to remove 8-oxoG lesions that are associated with transcriptional repression. HDAC1 deficiency causes impaired OGG1 activity, 8-oxoG accumulation at the promoters of genes critical for brain function, and transcriptional repression. Moreover, we observe elevated 8-oxoG along with reduced HDAC1 activity and downregulation of a similar gene set in the 5XFAD mouse model of Alzheimer's disease. Notably, pharmacological activation of HDAC1 alleviates the deleterious effects of 8-oxoG in aged wild-type and 5XFAD mice. Our work uncovers important roles for HDAC1 in 8-oxoG repair and highlights the therapeutic potential of HDAC1 activation to counter functional decline in brain aging and neurodegeneration.
Collapse
Affiliation(s)
- Ping-Chieh Pao
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Debasis Patnaik
- Chemical Neurobiology Laboratory, Departments of Neurology and Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - L Ashley Watson
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Fan Gao
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Caltech Bioinformatics Resource Center at Beckman Institute of Caltech, Pasadena, CA, 91225, USA
| | - Ling Pan
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Jun Wang
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Chinnakkaruppan Adaikkan
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Jay Penney
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Hugh P Cam
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Wen-Chin Huang
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Lorena Pantano
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Audrey Lee
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Alexi Nott
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Trongha X Phan
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Elizabeta Gjoneska
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, 27709, USA
| | - Sara Elmsaouri
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Stephen J Haggarty
- Chemical Neurobiology Laboratory, Departments of Neurology and Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Li-Huei Tsai
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
| |
Collapse
|
54
|
Wang T, He H, Liu S, Jia C, Fan Z, Zhong C, Yu J, Liu H, He C. Autophagy: A Promising Target for Age-related Osteoporosis. Curr Drug Targets 2020; 20:354-365. [PMID: 29943700 DOI: 10.2174/1389450119666180626120852] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Revised: 06/07/2018] [Accepted: 06/18/2018] [Indexed: 02/08/2023]
Abstract
Autophagy is a process the primary role of which is to clear up damaged cellular components such as long-lived proteins and organelles, thus participating in the conservation of different cells. Osteoporosis associated with aging is characterized by consistent changes in bone metabolism with suppression of bone formation as well as increased bone resorption. In advanced age, not only bone mass but also bone strength decrease in both sexes, resulting in an increased incidence of fractures. Clinical and animal experiments reveal that age-related bone loss is associated with many factors such as accumulation of autophagy, increased levels of reactive oxygen species, sex hormone deficiency, and high levels of endogenous glucocorticoids. Available basic and clinical studies indicate that age-associated factors can regulate autophagy. Those factors play important roles in bone remodeling and contribute to decreased bone mass and bone strength with aging. In this review, we summarize the mechanisms involved in bone metabolism related to aging and autophagy, supplying a theory for therapeutic targets to rescue bone mass and bone strength in older people.
Collapse
Affiliation(s)
- Tiantian Wang
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Hongchen He
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Shaxin Liu
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Chengsen Jia
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Ziyan Fan
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Can Zhong
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Jiadan Yu
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Honghong Liu
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Chengqi He
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
55
|
Kandlur A, Satyamoorthy K, Gangadharan G. Oxidative Stress in Cognitive and Epigenetic Aging: A Retrospective Glance. Front Mol Neurosci 2020; 13:41. [PMID: 32256315 PMCID: PMC7093495 DOI: 10.3389/fnmol.2020.00041] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 03/02/2020] [Indexed: 12/17/2022] Open
Abstract
Brain aging is the critical and common factor among several neurodegenerative disorders and dementia. Cellular, biochemical and molecular studies have shown intimate links between oxidative stress and cognitive dysfunction during aging and age-associated neuronal diseases. Brain aging is accompanied by oxidative damage of nuclear as well as mitochondrial DNA, and diminished repair. Recent studies have reported epigenetic alterations during aging of the brain which involves reactive oxygen species (ROS) that regulates various systems through distinct mechanisms. However, there are studies which depict differing roles of reactive oxidant species as a major factor during aging. In this review, we describe the evidence to show how oxidative stress is intricately linked to age-associated cognitive decline. The review will primarily focus on implications of age-associated oxidative damage on learning and memory, and the cellular events, with special emphasis on associated epigenetic machinery. A comprehensive understanding of these mechanisms may provide a perspective on the development of potential therapeutic targets within the oxidative system.
Collapse
Affiliation(s)
| | | | - Gireesh Gangadharan
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India
| |
Collapse
|
56
|
Parker GA, Kohn N, Spirina A, McMillen A, Huang W, Mackay TFC. Genetic Basis of Increased Lifespan and Postponed Senescence in Drosophila melanogaster. G3 (BETHESDA, MD.) 2020; 10:1087-1098. [PMID: 31969430 PMCID: PMC7056975 DOI: 10.1534/g3.120.401041] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 01/21/2020] [Indexed: 01/14/2023]
Abstract
Limited lifespan and senescence are near-universal phenomena. These quantitative traits exhibit variation in natural populations due to the segregation of many interacting loci and from environmental effects. Due to the complexity of the genetic control of lifespan and senescence, our understanding of the genetic basis of variation in these traits is incomplete. Here, we analyzed the pattern of genetic divergence between long-lived (O) Drosophila melanogaster lines selected for postponed reproductive senescence and unselected control (B) lines. We quantified the productivity of the O and B lines and found that reproductive senescence is maternally controlled. We therefore chose 57 candidate genes that are expressed in ovaries, 49 of which have human orthologs, and assessed the effects of RNA interference in ovaries and accessary glands on lifespan and reproduction. All but one candidate gene affected at least one life history trait in one sex or productivity week. In addition, 23 genes had antagonistic pleiotropic effects on lifespan and productivity. Identifying evolutionarily conserved genes affecting increased lifespan and delayed reproductive senescence is the first step toward understanding the evolutionary forces that maintain segregating variation at these loci in nature and may provide potential targets for therapeutic intervention to delay senescence while increasing lifespan.
Collapse
Affiliation(s)
- Grace A Parker
- Department of Biological Sciences
- Program in Genetics
- W. M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, North Carolina, 27695 and
| | | | | | | | - Wen Huang
- Department of Animal Science, Michigan State University, East Lansing, Michigan, 48824
| | - Trudy F C Mackay
- Department of Biological Sciences,
- Program in Genetics
- W. M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, North Carolina, 27695 and
| |
Collapse
|
57
|
Koelé MC, Lems WF, Willems HC. The Clinical Relevance of Hyperkyphosis: A Narrative Review. Front Endocrinol (Lausanne) 2020; 11:5. [PMID: 32038498 PMCID: PMC6993454 DOI: 10.3389/fendo.2020.00005] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 01/07/2020] [Indexed: 12/12/2022] Open
Abstract
The kyphosis angle of the thoracic spine tends to increase with aging. Hyperkyphosis is a kyphosis angle, exceeding the normal range. This narrative literature review aims to provide an overview of the current literature concerning kyphosis measurement methods, the etiology and adverse health effects of hyperkyphosis. As of yet, a well-defined threshold for hyperkyphosis is lacking. To attain more generalizability and to be able to compare study results in older adults, we propose to define age-related hyperkyphosis as a Cobb angle of 50° or more in standing position. Hyperkyphosis may be a potentially modifiable risk factor for adverse health outcomes, like fall risk and fractures. Additionally, hyperkyphosis may indicate the presence of osteoporosis, which is treatable. Prospective and intervention studies, using a Cobb angle of 50° as a clear and uniform definition of hyperkyphosis, are warranted to investigate the clinical relevance of hyperkyphosis.
Collapse
Affiliation(s)
- M C Koelé
- Division of Geriatrics, Department of Internal Medicine, Academic Medical Centre Amsterdam, Amsterdam Public Health Research Institute, Amsterdam UMC, Amsterdam, Netherlands
| | - W F Lems
- Department of Rheumatology, Amsterdam Movement Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - H C Willems
- Division of Geriatrics, Department of Internal Medicine, Academic Medical Centre Amsterdam, Amsterdam Public Health Research Institute, Amsterdam UMC, Amsterdam, Netherlands
| |
Collapse
|
58
|
Apostolou Z, Chatzinikolaou G, Stratigi K, Garinis GA. Nucleotide Excision Repair and Transcription-Associated Genome Instability. Bioessays 2019; 41:e1800201. [PMID: 30919497 DOI: 10.1002/bies.201800201] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 12/10/2018] [Indexed: 12/12/2022]
Abstract
Transcription is a potential threat to genome integrity, and transcription-associated DNA damage must be repaired for proper messenger RNA (mRNA) synthesis and for cells to transmit their genome intact into progeny. For a wide range of structurally diverse DNA lesions, cells employ the highly conserved nucleotide excision repair (NER) pathway to restore their genome back to its native form. Recent evidence suggests that NER factors function, in addition to the canonical DNA repair mechanism, in processes that facilitate mRNA synthesis or shape the 3D chromatin architecture. Here, these findings are critically discussed and a working model that explains the puzzling clinical heterogeneity of NER syndromes highlighting the relevance of physiological, transcription-associated DNA damage to mammalian development and disease is proposed.
Collapse
Affiliation(s)
- Zivkos Apostolou
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Nikolaou Plastira 100, Heraklion 70013, Crete, Greece.,Department of Biology, University of Crete, Vassilika Vouton, Heraklion GR71409, Crete, Greece
| | - Georgia Chatzinikolaou
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Nikolaou Plastira 100, Heraklion 70013, Crete, Greece
| | - Kalliopi Stratigi
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Nikolaou Plastira 100, Heraklion 70013, Crete, Greece
| | - George A Garinis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Nikolaou Plastira 100, Heraklion 70013, Crete, Greece.,Department of Biology, University of Crete, Vassilika Vouton, Heraklion GR71409, Crete, Greece
| |
Collapse
|
59
|
Abstract
Ageing appears to be a nearly universal feature of life, ranging from unicellular microorganisms to humans. Longevity depends on the maintenance of cellular functionality, and an organism's ability to respond to stress has been linked to functional maintenance and longevity. Stress response pathways might indeed become therapeutic targets of therapies aimed at extending the healthy lifespan. Various progeroid syndromes have been linked to genome instability, indicating an important causal role of DNA damage accumulation in the ageing process and the development of age-related pathologies. Recently, non-cell-autonomous mechanisms including the systemic consequences of cellular senescence have been implicated in regulating organismal ageing. We discuss here the role of cellular and systemic mechanisms of ageing and their role in ageing-associated diseases.
Collapse
Affiliation(s)
- Paulo F L da Silva
- Institute for Genome Stability in Ageing and Disease, Medical Faculty, University of Cologne, Joseph-Stelzmann-Strasse 26, 50931 Cologne, Germany.,Cologne Excellence Cluster for Cellular Stress Responses in Ageing-Associated Diseases (CECAD), Center for Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Strasse 26, 50931 Cologne, Germany
| | - Björn Schumacher
- Institute for Genome Stability in Ageing and Disease, Medical Faculty, University of Cologne, Joseph-Stelzmann-Strasse 26, 50931 Cologne, Germany.,Cologne Excellence Cluster for Cellular Stress Responses in Ageing-Associated Diseases (CECAD), Center for Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Strasse 26, 50931 Cologne, Germany
| |
Collapse
|
60
|
Schmeer C, Kretz A, Wengerodt D, Stojiljkovic M, Witte OW. Dissecting Aging and Senescence-Current Concepts and Open Lessons. Cells 2019; 8:cells8111446. [PMID: 31731770 PMCID: PMC6912776 DOI: 10.3390/cells8111446] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 11/12/2019] [Accepted: 11/13/2019] [Indexed: 01/10/2023] Open
Abstract
In contrast to the programmed nature of development, it is still a matter of debate whether aging is an adaptive and regulated process, or merely a consequence arising from a stochastic accumulation of harmful events that culminate in a global state of reduced fitness, risk for disease acquisition, and death. Similarly unanswered are the questions of whether aging is reversible and can be turned into rejuvenation as well as how aging is distinguishable from and influenced by cellular senescence. With the discovery of beneficial aspects of cellular senescence and evidence of senescence being not limited to replicative cellular states, a redefinition of our comprehension of aging and senescence appears scientifically overdue. Here, we provide a factor-based comparison of current knowledge on aging and senescence, which we converge on four suggested concepts, thereby implementing the newly emerging cellular and molecular aspects of geroconversion and amitosenescence, and the signatures of a genetic state termed genosenium. We also address the possibility of an aging-associated secretory phenotype in analogy to the well-characterized senescence-associated secretory phenotype and delineate the impact of epigenetic regulation in aging and senescence. Future advances will elucidate the biological and molecular fingerprints intrinsic to either process.
Collapse
Affiliation(s)
- Christian Schmeer
- Hans-Berger Department of Neurology, Jena University Hospital, 07747 Jena, Thuringia, Germany; (A.K.); (D.W.); (M.S.); (O.W.W.)
- Jena Center for Healthy Ageing, Jena University Hospital, 07747 Jena, Thuringia, Germany
- Correspondence:
| | - Alexandra Kretz
- Hans-Berger Department of Neurology, Jena University Hospital, 07747 Jena, Thuringia, Germany; (A.K.); (D.W.); (M.S.); (O.W.W.)
- Jena Center for Healthy Ageing, Jena University Hospital, 07747 Jena, Thuringia, Germany
| | - Diane Wengerodt
- Hans-Berger Department of Neurology, Jena University Hospital, 07747 Jena, Thuringia, Germany; (A.K.); (D.W.); (M.S.); (O.W.W.)
| | - Milan Stojiljkovic
- Hans-Berger Department of Neurology, Jena University Hospital, 07747 Jena, Thuringia, Germany; (A.K.); (D.W.); (M.S.); (O.W.W.)
- Jena Center for Healthy Ageing, Jena University Hospital, 07747 Jena, Thuringia, Germany
| | - Otto W. Witte
- Hans-Berger Department of Neurology, Jena University Hospital, 07747 Jena, Thuringia, Germany; (A.K.); (D.W.); (M.S.); (O.W.W.)
- Jena Center for Healthy Ageing, Jena University Hospital, 07747 Jena, Thuringia, Germany
| |
Collapse
|
61
|
Nagarajan P, Agudelo Garcia PA, Iyer CC, Popova LV, Arnold WD, Parthun MR. Early-onset aging and mitochondrial defects associated with loss of histone acetyltransferase 1 (Hat1). Aging Cell 2019; 18:e12992. [PMID: 31290578 PMCID: PMC6718594 DOI: 10.1111/acel.12992] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 05/07/2019] [Accepted: 05/27/2019] [Indexed: 12/18/2022] Open
Abstract
Histone acetyltransferase 1 (Hat1) is responsible for the acetylation of newly synthesized histone H4 on lysines 5 and 12 during the process of chromatin assembly. To understand the broader biological role of Hat1, we have generated a conditional mouse knockout model of this enzyme. We previously reported that Hat1 is required for viability and important for mammalian development and genome stability. In this study, we show that haploinsufficiency of Hat1 results in a significant decrease in lifespan. Defects observed in Hat1+/− mice are consistent with an early‐onset aging phenotype. These include lordokyphosis (hunchback), muscle atrophy, minor growth retardation, reduced subcutaneous fat, cancer, and paralysis. In addition, the expression of Hat1 is linked to the normal aging process as Hat1 mRNA and protein becomes undetectable in many tissues in old mice. At the cellular level, fibroblasts from Hat1 haploinsufficient embryos undergo early senescence and accumulate high levels of p21. Hat1+/− mouse embryonic fibroblasts (MEFs) display modest increases in endogenous DNA damage but have significantly higher levels of reactive oxygen species (ROS). Consistently, further studies show that Hat1−/− MEFs exhibit mitochondrial defects suggesting a critical role for Hat1 in mitochondrial function. Taken together, these data show that loss of Hat1 induces multiple hallmarks of early‐onset aging.
Collapse
Affiliation(s)
- Prabakaran Nagarajan
- Department of Biological Chemistry and Pharmacology, The Ohio State University Columbus Ohio
| | - Paula A. Agudelo Garcia
- Department of Biological Chemistry and Pharmacology, The Ohio State University Columbus Ohio
| | - Chitra C. Iyer
- Department of Neurology The Ohio State University Columbus Ohio
| | - Liudmila V. Popova
- Department of Biological Chemistry and Pharmacology, The Ohio State University Columbus Ohio
| | | | - Mark R. Parthun
- Department of Biological Chemistry and Pharmacology, The Ohio State University Columbus Ohio
| |
Collapse
|
62
|
Folgueras AR, Freitas-Rodríguez S, Velasco G, López-Otín C. Mouse Models to Disentangle the Hallmarks of Human Aging. Circ Res 2019; 123:905-924. [PMID: 30355076 DOI: 10.1161/circresaha.118.312204] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Model organisms have provided fundamental evidence that aging can be delayed and longevity extended. These findings gave rise to a new era in aging research aimed at elucidating the pathways and networks controlling this complex biological process. The identification of 9 hallmarks of aging has established a framework to evaluate the relative contribution of each hallmark and the interconnections among them. In this review, we revisit these hallmarks with the information obtained exclusively through the generation of genetically modified mouse models that have a significant impact on the aging process. We discuss within each hallmark those interventions that accelerate aging or that have been successful at increasing lifespan, with the final goal of identifying the most promising antiaging avenues based on the current knowledge provided by in vivo models.
Collapse
Affiliation(s)
- Alicia R Folgueras
- From the Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología del Principado de Asturias, Universidad de Oviedo, Spain
| | - Sandra Freitas-Rodríguez
- From the Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología del Principado de Asturias, Universidad de Oviedo, Spain
| | - Gloria Velasco
- From the Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología del Principado de Asturias, Universidad de Oviedo, Spain
| | - Carlos López-Otín
- From the Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología del Principado de Asturias, Universidad de Oviedo, Spain
| |
Collapse
|
63
|
Lans H, Hoeijmakers JHJ, Vermeulen W, Marteijn JA. The DNA damage response to transcription stress. Nat Rev Mol Cell Biol 2019; 20:766-784. [DOI: 10.1038/s41580-019-0169-4] [Citation(s) in RCA: 217] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/14/2019] [Indexed: 12/30/2022]
|
64
|
Hambright WS, Niedernhofer LJ, Huard J, Robbins PD. Murine models of accelerated aging and musculoskeletal disease. Bone 2019; 125:122-127. [PMID: 30844492 DOI: 10.1016/j.bone.2019.03.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Accepted: 03/03/2019] [Indexed: 12/13/2022]
Abstract
The primary risk factor for most musculoskeletal diseases, including osteoarthritis, osteoporosis and sarcopenia, is aging. To treat the diverse types of musculoskeletal diseases and pathologies, targeting their root cause, the aging process itself, has the potential to slow or prevent multiple age-related musculoskeletal conditions simultaneously. However, the development of approaches to delay onset of age related diseases, including musculoskeletal pathologies, has been slowed by the relatively long lifespan of rodent models of aging. Thus, to expedite the development of therapeutic approaches for age-related musculoskeletal disease, the implementation of mouse models of accelerated musculoskeletal aging are of great utility. Currently there are multiple genetically diverse mouse models that mirror certain aspects of normal human and mouse aging. Here, we provide a review of some of the most relevant murine models of accelerated aging that mimic many aspects of natural musculoskeletal aging, highlighting their relative strengths and weaknesses. Importantly, these murine models of accelerated aging recapitulate phenotypes of musculoskeletal age-related decline observed in humans.
Collapse
Affiliation(s)
- William S Hambright
- Department of Orthopaedic Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States of America
| | - Laura J Niedernhofer
- Institute on the Biology of Aging and Metabolism, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, United States of America; Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, United States of America
| | - Johnny Huard
- Department of Orthopaedic Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States of America; Steadman Philippon Research Institute, Vail, CO, United States of America.
| | - Paul D Robbins
- Institute on the Biology of Aging and Metabolism, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, United States of America; Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, United States of America.
| |
Collapse
|
65
|
Zhou Z, Wang L, Ge F, Gong P, Wang H, Wang F, Chen L, Liu L. Pold3 is required for genomic stability and telomere integrity in embryonic stem cells and meiosis. Nucleic Acids Res 2019; 46:3468-3486. [PMID: 29447390 PMCID: PMC6283425 DOI: 10.1093/nar/gky098] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Accepted: 02/01/2018] [Indexed: 12/29/2022] Open
Abstract
Embryonic stem cells (ESCs) and meiosis are featured by relatively higher frequent homologous recombination associated with DNA double strand breaks (DSB) repair. Here, we show that Pold3 plays important roles in DSB repair, telomere maintenance and genomic stability of both ESCs and spermatocytes in mice. By attempting to generate Pold3 deficient mice using CRISPR/Cas9 or transcription activator-like effector nucleases, we show that complete loss of Pold3 (Pold3−/−) resulted in early embryonic lethality at E6.5. Rapid DNA damage response and massive apoptosis occurred in both outgrowths of Pold3-null (Pold3−/−) blastocysts and Pold3 inducible knockout (iKO) ESCs. While Pold3−/− ESCs were not achievable, Pold3 iKO led to increased DNA damage response, telomere loss and chromosome breaks accompanied by extended S phase. Meanwhile, loss of Pold3 resulted in replicative stress, micronucleation and aneuploidy. Also, DNA repair was impaired in Pold3+/− or Pold3 knockdown ESCs. Moreover, Pold3 mediates DNA replication and repair by regulating 53BP1, RIF1, ATR and ATM pathways. Furthermore, spermatocytes of Pold3 haploinsufficient (Pold3+/−) mice with increasing age displayed impaired DSB repair, telomere shortening and loss, and chromosome breaks, like Pold3 iKO ESCs. These data suggest that Pold3 maintains telomere integrity and genomic stability of both ESCs and meiosis by suppressing replicative stress.
Collapse
Affiliation(s)
- Zhongcheng Zhou
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China.,Department of Cell Biology and Genetics, The Key Laboratory of Bioactive Materials Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Lingling Wang
- Department of Cell Biology and Genetics, The Key Laboratory of Bioactive Materials Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Feixiang Ge
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China.,Department of Cell Biology and Genetics, The Key Laboratory of Bioactive Materials Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Peng Gong
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China.,Department of Cell Biology and Genetics, The Key Laboratory of Bioactive Materials Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Hua Wang
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China.,Department of Cell Biology and Genetics, The Key Laboratory of Bioactive Materials Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Feng Wang
- Department of Genetics, School of basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Lingyi Chen
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China.,Department of Cell Biology and Genetics, The Key Laboratory of Bioactive Materials Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Lin Liu
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China.,Department of Cell Biology and Genetics, The Key Laboratory of Bioactive Materials Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| |
Collapse
|
66
|
Oakley PA, Ehsani NN, Harrison DE. The Scoliosis Quandary: Are Radiation Exposures From Repeated X-Rays Harmful? Dose Response 2019; 17:1559325819852810. [PMID: 31217755 PMCID: PMC6560808 DOI: 10.1177/1559325819852810] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 04/10/2019] [Accepted: 04/23/2019] [Indexed: 12/12/2022] Open
Abstract
X-rays have been the gold standard for diagnosis, evaluation, and management of spinal scoliosis for decades as other assessment methods are indirect, too expensive, or not practical in practice. The average scoliosis patient will receive 10 to 25 spinal X-rays over several years equating to a maximum estimated dose of 10 to 25 mGy. Some patients, those getting diagnosed at a younger age and receiving early and ongoing treatments, may receive up to 40 to 50 X-rays, approaching at most 50 mGy. There are concerns that repeated radiographs given to patients are carcinogenic. Some studies have used the linear no-threshold model to derive cancer-risk estimates; however, it is invalid for low-dose irradiation (ie, X-rays); these estimates are untrue. Other studies have calculated cancer-risk ratios from long-term health data of historic scoliosis cohorts. Since data indicate reduced cancer rates in a cohort receiving a total radiation dose between 50 and 300 mGy, it is unlikely that scoliosis patients would get cancer from repeated X-rays. Moreover, since the threshold for leukemia is about 1100 mGy, scoliosis patients will not likely develop cancers from spinal X-rays. Scoliosis patients likely have long-term health consequences, including cancers, from the actual disease entity itself and not from protracted X-ray radiation exposures that are essential and indeed safe.
Collapse
|
67
|
Niedernhofer LJ, Gurkar AU, Wang Y, Vijg J, Hoeijmakers JHJ, Robbins PD. Nuclear Genomic Instability and Aging. Annu Rev Biochem 2019; 87:295-322. [PMID: 29925262 DOI: 10.1146/annurev-biochem-062917-012239] [Citation(s) in RCA: 169] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The nuclear genome decays as organisms age. Numerous studies demonstrate that the burden of several classes of DNA lesions is greater in older mammals than in young mammals. More challenging is proving this is a cause rather than a consequence of aging. The DNA damage theory of aging, which argues that genomic instability plays a causal role in aging, has recently gained momentum. Support for this theory stems partly from progeroid syndromes in which inherited defects in DNA repair increase the burden of DNA damage leading to accelerated aging of one or more organs. Additionally, growing evidence shows that DNA damage accrual triggers cellular senescence and metabolic changes that promote a decline in tissue function and increased susceptibility to age-related diseases. Here, we examine multiple lines of evidence correlating nuclear DNA damage with aging. We then consider how, mechanistically, nuclear genotoxic stress could promote aging. We conclude that the evidence, in toto, supports a role for DNA damage as a nidus of aging.
Collapse
Affiliation(s)
- Laura J Niedernhofer
- Department of Molecular Medicine and the Center on Aging, The Scripps Research Institute Florida, Jupiter, Florida 33458, USA;
| | - Aditi U Gurkar
- Department of Molecular Medicine and the Center on Aging, The Scripps Research Institute Florida, Jupiter, Florida 33458, USA; .,Department of Medicine, Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
| | - Yinsheng Wang
- Department of Chemistry, University of California, Riverside, California 92521, USA
| | - Jan Vijg
- Department of Genetics, Albert Einstein College of Medicine, Michael F. Price Center, Bronx, New York 10461, USA
| | - Jan H J Hoeijmakers
- Department of Molecular Genetics, Erasmus University Medical Center, 3015 CE Rotterdam, The Netherlands
| | - Paul D Robbins
- Department of Molecular Medicine and the Center on Aging, The Scripps Research Institute Florida, Jupiter, Florida 33458, USA;
| |
Collapse
|
68
|
He J, Zhang A, Song Z, Guo S, Chen Y, Liu Z, Zhang J, Xu X, Liu J, Chu L. The resistant effect of SIRT1 in oxidative stress-induced senescence of rat nucleus pulposus cell is regulated by Akt-FoxO1 pathway. Biosci Rep 2019; 39:BSR20190112. [PMID: 30967498 PMCID: PMC6509061 DOI: 10.1042/bsr20190112] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 04/07/2019] [Accepted: 04/07/2019] [Indexed: 01/03/2023] Open
Abstract
Objective: The senescence of nucleus pulposus (NP) cells induced by oxidative stress is one of the important causes of intervertebral disc degeneration (IDD). Herein, we investigated the role and action mechanism of silent information regulator 1 (SIRT1) in oxidative stress-induced senescence of rat NP cell.Methods: Premature senescence of rat NP cells was induced by sublethal concentration of hydrogen peroxide (H2O2) (100 μM). SIRT1 was activated with SRT1720 (5 μM) to explore its effect on NP cells senescence. FoxO1 and Akt were inhibited by AS1842856 (0.2 μM) and MK-2206 (5 μM), respectively, to explore the role of Akt-FoxO1-SIRT1 axis in rat NP cells. Pretreatment with the resveratrol (20 μM), a common antioxidant and indirect activator of SIRT1, was done to investigate its role in senescent rat NP cells.Results: The mRNA and protein levels of SIRT1 were decreased in H2O2-induced senescent rat NP cells, and that specific activation of SIRT1 suppresses senescence. And the Akt-FoxO1 pathway, as the upstream of SIRT1, might be involved in the regulation of H2O2-induced senescence of rat NP cells by affecting the expression of SIRT1. In addition, the resveratrol played an anti-senescence role in rat NP cells, which might affect the Akt-FoxO1-SIRT1 axis.Conclusion: SIRT1 ameliorated oxidative stress-induced senescence of rat NP cell which was regulated by Akt-FoxO1 pathway, and resveratrol exerted anti-senescence effects by affecting this signaling axis.
Collapse
Affiliation(s)
- Junsheng He
- Department of Spinal surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Ailiang Zhang
- Department of Spinal surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Zhiwen Song
- Department of Spinal surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Shiwu Guo
- Department of Spinal surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Yuwei Chen
- Department of Spinal surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Zhiyuan Liu
- Department of Spinal surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
- Department of Orthopedics, The Affiliated Wujin Hospital of Jiangsu University, Changzhou 213003, China
| | - Jinlong Zhang
- Department of Spinal surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Xu Xu
- Department of Spinal surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Jinbo Liu
- Department of Spinal surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Lei Chu
- Department of Orthopedics, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| |
Collapse
|
69
|
Liang ST, Audira G, Juniardi S, Chen JR, Lai YH, Du ZC, Lin DS, Hsiao CD. Zebrafish Carrying pycr1 Gene Deficiency Display Aging and Multiple Behavioral Abnormalities. Cells 2019; 8:cells8050453. [PMID: 31091804 PMCID: PMC6562453 DOI: 10.3390/cells8050453] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 04/28/2019] [Accepted: 05/09/2019] [Indexed: 12/22/2022] Open
Abstract
Aging is a natural process that internal gene control and external stimuli mediate. Clinical data pointed out that homozygotic or heterozygotic mutation in the pyrroline-5-carboxylate reductase 1 (PYCR1) gene in humans caused cutis laxa (ARCL) disease, with progeroid appearance, lax and wrinkled skin, joint laxity, osteopenia, and mental retardation phenotypes. In this study, we aimed to generate pycr1 knockout (KO) zebrafish and carried out biochemical characterizations and behavior analyses. Marked apoptosis and senescence were detected in pycr1 KO zebrafish, which started from embryos/larvae stage. Biochemical assays showed that adult pycr1 KO fish have significantly reduced proline and extracellular matrix contents, lowered energy, and diminished superoxide dismutase (SOD) and telomerase activity when compared to the wild type fish, which suggested the pycr1 KO fish may have dysfunction in mitochondria. The pycr1 KO fish were viable; however, displayed progeria-like phenotype from the 4 months old and reach 50% mortality around six months old. In adult stage, we found that pycr1 KO fish showed reduced locomotion activity, aggression, predator avoidance, social interaction interest, as well as dysregulated color preference and circadian rhythm. In summary, we have identified multiple behavioral alterations in a novel fish model for aging with pycr1 gene loss-of-function by behavioral tests. This animal model may not only provide a unique vertebrate model to screen potential anti-aging drugs in the future, but also be an excellent in vivo model towards a better understanding of the corresponding behavioral alterations that accompany aging.
Collapse
Affiliation(s)
- Sung-Tzu Liang
- Department of Bioscience Technology, Chung Yuan Christian University, Chung-Li 32023, Taiwan.
| | - Gilbert Audira
- Department of Bioscience Technology, Chung Yuan Christian University, Chung-Li 32023, Taiwan.
- Department of Chemistry, Chung Yuan Christian University, Chung-Li 32023, Taiwan.
| | - Stevhen Juniardi
- Department of Bioscience Technology, Chung Yuan Christian University, Chung-Li 32023, Taiwan.
| | - Jung-Ren Chen
- Department of Biological Science & Technology, College of Medicine, I-Shou University, Kaohsiung 84001, Taiwan.
| | - Yu-Heng Lai
- Department of Chemistry, Chinese Culture University, Taipei 11114, Taiwan.
| | - Zheng-Cai Du
- Guangxi Scientific Experimental Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning 530200, China.
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning 530200, China.
| | - Dar-Shong Lin
- Department of Pediatrics, Mackay Memorial Hospital, Taipei 252, Taiwan.
- Department of Medical Research, Mackay Memorial Hospital, Taipei 252, Taiwan.
- Department of Medicine, Mackay Medical College, New Taipei 252, Taiwan.
| | - Chung-Der Hsiao
- Department of Bioscience Technology, Chung Yuan Christian University, Chung-Li 32023, Taiwan.
- Department of Chemistry, Chung Yuan Christian University, Chung-Li 32023, Taiwan.
- Center for Biomedical Technology, Chung Yuan Christian University, Chung-Li 32023, Taiwan.
- Center for Nanotechnology, Chung Yuan Christian University, Chung-Li 32023, Taiwan.
| |
Collapse
|
70
|
Baxter LL, Watkins-Chow DE, Pavan WJ, Loftus SK. A curated gene list for expanding the horizons of pigmentation biology. Pigment Cell Melanoma Res 2019; 32:348-358. [PMID: 30339321 PMCID: PMC10413850 DOI: 10.1111/pcmr.12743] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 09/01/2018] [Accepted: 09/29/2018] [Indexed: 12/27/2022]
Abstract
Over the past century, studies of human pigmentary disorders along with mouse and zebrafish models have shed light on the many cellular functions associated with visible pigment phenotypes. This has led to numerous genes annotated with the ontology term "pigmentation" in independent human, mouse, and zebrafish databases. Comparisons among these datasets revealed that each is individually incomplete in documenting all genes involved in integument-based pigmentation phenotypes. Additionally, each database contained inherent species-specific biases in data annotation, and the term "pigmentation" did not solely reflect integument pigmentation phenotypes. This review presents a comprehensive, cross-species list of 650 genes involved in pigmentation phenotypes that was compiled with extensive manual curation of genes annotated in OMIM, MGI, ZFIN, and GO. The resulting cross-species list of genes both intrinsic and extrinsic to integument pigment cells provides a valuable tool that can be used to expand our knowledge of complex, pigmentation-associated pathways.
Collapse
Affiliation(s)
- Laura L Baxter
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - Dawn E Watkins-Chow
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - William J Pavan
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - Stacie K Loftus
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
71
|
Abstract
Mutation provides the ultimate source of all new alleles in populations, including variants that cause disease and fuel adaptation. Recent whole genome sequencing studies have uncovered variation in the mutation rate among individuals and differences in the relative frequency of specific nucleotide changes (the mutation spectrum) between populations. Although parental age is a major driver of differences in overall mutation rate among individuals, the causes of variation in the mutation spectrum remain less well understood. Here, I use high-quality whole genome sequences from 29 inbred laboratory mouse strains to explore the root causes of strain variation in the mutation spectrum. My analysis leverages the unique, mosaic patterns of genetic relatedness among inbred mouse strains to identify strain private variants residing on haplotypes shared between multiple strains due to their recent descent from a common ancestor. I show that these strain-private alleles are strongly enriched for recent de novo mutations and lack signals of widespread purifying selection, suggesting their faithful recapitulation of the spontaneous mutation landscape in single strains. The spectrum of strain-private variants varies significantly among inbred mouse strains reared under standardized laboratory conditions. This variation is not solely explained by strain differences in age at reproduction, raising the possibility that segregating genetic differences affect the constellation of new mutations that arise in a given strain. Collectively, these findings imply the action of remarkably precise nucleotide-specific genetic mechanisms for tuning the de novo mutation landscape in mammals and underscore the genetic complexity of mutation rate control.
Collapse
|
72
|
Qiu W, Chuong CM, Lei M. Regulation of melanocyte stem cells in the pigmentation of skin and its appendages: Biological patterning and therapeutic potentials. Exp Dermatol 2019; 28:395-405. [PMID: 30537004 DOI: 10.1111/exd.13856] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 11/27/2018] [Accepted: 12/05/2018] [Indexed: 12/13/2022]
Abstract
Skin evolves essential appendages and indispensable types of cells that synergistically insulate the body from environmental insults. Residing in the specific regions in the skin such as epidermis, dermis and hair follicle, melanocytes perform an array of vital functions including defending the ultraviolet radiation and diversifying animal appearance. As one of the adult stem cells, melanocyte stem cells in the hair follicle bulge niche can proliferate, differentiate and keep quiescence to control and coordinate tissue homeostasis, repair and regeneration. In synchrony with hair follicle stem cells, melanocyte stem cells in the hair follicles undergo cyclic activation, degeneration and resting phases, to pigment the hairs and to preserve the stem cells. Disorder of melanocytes results in severe skin problems such as canities, vitiligo and even melanoma. Here, we compare and summarize recent discoveries about melanocyte in the skin, particularly in the hair follicle. A better understanding of the physiological and pathological regulation of melanocyte and melanocyte stem cell behaviours will help to guide the clinical applications in regenerative medicine.
Collapse
Affiliation(s)
- Weiming Qiu
- Department of Dermatology, Wuhan General Hospital of Chinese People's Liberation Army, Wuhan, China
| | - Cheng-Ming Chuong
- Department of Pathology, University of Southern California, Los Angeles, California.,Integrative Stem Cell Center, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Mingxing Lei
- Integrative Stem Cell Center, China Medical University Hospital, China Medical University, Taichung, Taiwan.,Institute of New Drug Development, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, Taiwan
| |
Collapse
|
73
|
Folch J, Busquets O, Ettcheto M, Sánchez-López E, Pallàs M, Beas-Zarate C, Marin M, Casadesus G, Olloquequi J, Auladell C, Camins A. Experimental Models for Aging and their Potential for Novel Drug Discovery. Curr Neuropharmacol 2018; 16:1466-1483. [PMID: 28685671 PMCID: PMC6295931 DOI: 10.2174/1570159x15666170707155345] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 06/22/2017] [Accepted: 06/30/2017] [Indexed: 02/07/2023] Open
Abstract
Background: An interesting area of scientific research is the development of potential antiaging drugs. In order to pursue this goal, it is necessary to gather the specific knowledge about the adequate preclinical models that are available to evaluate the beneficial effects of new potential drugs. This review is focused on invertebrate and vertebrate preclinical models used to evaluate the efficacy of antiaging compounds, with the objective to extend life span and health span. Methods: Research and online content related to aging, antiaging drugs, experimental aging models is reviewed. Moreover, in this review, the main experimental preclinical models of organisms that have contributed to the research in the pharmacol-ogy of lifespan extension and the understanding of the aging process are discussed. Results: Dietary restriction (DR) constitutes a common experimental process to extend life span in all organisms. Besides, classical antiaging drugs such as resveratrol, rapamycin and metformin denominated as DR mimetics are also discussed. Likewise, the main therapeutic targets of these drugs include sirtuins, IGF-1, and mTOR, all of them being modulated by DR. Conclusion: Advances in molecular biology have uncovered the potential molecular pathways involved in the aging process. Due to their characteristics, invertebrate models are mainly used for drug screening. The National Institute on Aging (NIA) developed the Interventions Testing Program (ITP). At the pre-clinical level, the ITP uses Heterogeneous mouse model (HET) which is probably the most suitable rodent model to study potential drugs against aging prevention. The accelerated-senescence mouse P8 is also a mammalian rodent model for aging research. However, when evaluating the effect of drugs on a preclinical level, the evaluation must be done in non-human primates since it is the mammalian specie closest to humans. Research is needed to investigate the impact of new potential drugs for the increase of human quality of
Collapse
Affiliation(s)
- Jaume Folch
- Unitat de Bioquimica i Biotecnologia, Facultat de Medicina i Ciencies de la Salut, Universitat Rovira i Virgili, Reus, Tarragona, Spain.,Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Oriol Busquets
- Unitat de Bioquimica i Biotecnologia, Facultat de Medicina i Ciencies de la Salut, Universitat Rovira i Virgili, Reus, Tarragona, Spain.,Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Departament Deaprtament de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Barcelona, Spain.,Institute of Neurosciences, University of Barcelona, Barcelona, Spain
| | - Miren Ettcheto
- Unitat de Bioquimica i Biotecnologia, Facultat de Medicina i Ciencies de la Salut, Universitat Rovira i Virgili, Reus, Tarragona, Spain.,Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Departament Deaprtament de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Barcelona, Spain.,Institute of Neurosciences, University of Barcelona, Barcelona, Spain
| | - Elena Sánchez-López
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Unitat de Farmacia, Tecnologia Farmacèutica i Fisico-química, Facultat de Farmàcia, Universitat de Barcelona, Barcelona, Spain.,Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain
| | - Mercè Pallàs
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Departament Deaprtament de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Barcelona, Spain.,Institute of Neurosciences, University of Barcelona, Barcelona, Spain
| | - Carlos Beas-Zarate
- Departamento de Biologia Celulary Molecular, C.U.C.B.A., Universidad de Guadalajara and Division de Neurociencias, Sierra Mojada 800, Col. Independencia, Guadalajara, Jalisco 44340, Mexico
| | - Miguel Marin
- Centro de Biotecnologia. Universidad Nacional de Loja, Av. Pío Jaramillo Alvarado y Reinaldo Espinosa, La Argelia. Loja, Ecuador
| | - Gemma Casadesus
- Department of Biological Sciences, Kent State University, Kent, OH, United States
| | - Jordi Olloquequi
- Instituto de Ciencias Biomedicas, Facultad de Ciencias de la Salud, Universidad Autonoma de Chile, Talca, Chile
| | - Carme Auladell
- Institute of Neurosciences, University of Barcelona, Barcelona, Spain.,Departamento de Biologia Celulary Molecular, C.U.C.B.A., Universidad de Guadalajara and Division de Neurociencias, Sierra Mojada 800, Col. Independencia, Guadalajara, Jalisco 44340, Mexico.,Departament de Biologia Cellular, Fisiologia i Inmunologia, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| | - Antoni Camins
- Departament Deaprtament de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Barcelona, Spain.,Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Institute of Neurosciences, University of Barcelona, Barcelona, Spain.,Centro de Biotecnologia. Universidad Nacional de Loja, Av. Pío Jaramillo Alvarado y Reinaldo Espinosa, La Argelia. Loja, Ecuador
| |
Collapse
|
74
|
Pongpanich M, Patchsung M, Mutirangura A. Pathologic Replication-Independent Endogenous DNA Double-Strand Breaks Repair Defect in Chronological Aging Yeast. Front Genet 2018; 9:501. [PMID: 30410502 PMCID: PMC6209823 DOI: 10.3389/fgene.2018.00501] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 10/05/2018] [Indexed: 12/27/2022] Open
Abstract
Reduction of physiologic replication-independent endogenous DNA double strand breaks (Phy-RIND-EDSBs) in chronological aging yeast increases pathologic RIND-EDSBs (Path-RIND-EDSBs). Path-RIND-EDSBs can occur spontaneously in non-dividing cells without any inductive agents, and they must be repaired immediately otherwise their accumulation can lead to senescence. If yeasts have DSB repair defect, retention of Path-RIND-EDSBs can be found. Previously, we found that Path-RIND-EDSBs are not only produced but also retained in chronological aging yeast. Here, we evaluated if chronological aging yeasts have a DSB repair defect. We found a significant accumulation of Path-RIND-EDSBs around the same level in aging cells and caffeine treated cells and at a much higher level in the DSB repair mutant cells. Especially in the mutant, some unknown sequence was found inserted at the breaks. In addition, % difference of cell viability between HO induced and non-induced cells was significantly greater in aging cells. Our results suggested that RIND-EDSBs repair efficiency declines, but is not absent, in chronological aging yeast which might promote senescence phenotype. When a repair protein is deficient, an alternative pathway might be employed or an end modification process might occur as inserted sequences at the breaks were observed. Restoring repair defects might slow down the deterioration of cells from chronological aging.
Collapse
Affiliation(s)
- Monnat Pongpanich
- Department of Mathematics and Computer Science, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
- Center for Excellence in Molecular Genetics of Cancer and Human Diseases, Chulalongkorn University, Bangkok, Thailand
| | - Maturada Patchsung
- Center for Excellence in Molecular Genetics of Cancer and Human Diseases, Chulalongkorn University, Bangkok, Thailand
| | - Apiwat Mutirangura
- Center for Excellence in Molecular Genetics of Cancer and Human Diseases, Chulalongkorn University, Bangkok, Thailand
- Department of Anatomy, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
75
|
Michalska E, Koppolu A, Dobrzańska A, Płoski R, Gruszfeld D. A case of severe trichothiodystrophy 3 in a neonate due to mutation in the GTF2H5 gene: Clinical report. Eur J Med Genet 2018; 62:103557. [PMID: 30359777 DOI: 10.1016/j.ejmg.2018.10.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 10/12/2018] [Accepted: 10/20/2018] [Indexed: 01/08/2023]
Abstract
Trichothiodystrophy (TTD) is a group of predominantly autosomal recessive disorders characterized by sulfur-deficient brittle hair. Clinical features of TTD consist of variable neuroectodermal symptoms including ichthyosis, nail abnormalities, mental retardation, short stature, decreased fertility and proneness to infections. Approximately half of the reported patients with TTD have clinical and cellular photosensitivity associated with mutations in three subunits (ERCC3, ERCC2, GTF2H5) of the basal transcription factor TFHII, which is involved in transcription and nucleotide excision repair. We report on a case of a male neonate with a novel GTF2H5 gene mutation, detected by whole exome sequencing. The GTF2H5 gene's role is to provide stability to the entire TFHII complex. The reported patient was born at 33 weeks' gestation from a pregnancy complicated by intrauterine growth restriction and premature rupture of membranes. His main clinical problems included severe congenital ichthyosis and proneness to infections with episodes of multiorgan failure. The infant's history displays the most severe clinical manifestations among patients with GTF2H5 gene mutations that have so far been reported.
Collapse
Affiliation(s)
- Eliza Michalska
- Department of Neonatal Intensive Care, The Children's Memorial Health Institute, Poland.
| | - Agnieszka Koppolu
- Department of Medical Genetics, Warsaw Medical University, Poland; Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Anna Dobrzańska
- Department of Neonatal Intensive Care, The Children's Memorial Health Institute, Poland
| | - Rafał Płoski
- Department of Medical Genetics, Warsaw Medical University, Poland
| | - Dariusz Gruszfeld
- Department of Neonatal Intensive Care, The Children's Memorial Health Institute, Poland
| |
Collapse
|
76
|
Guo J, Huang Y, Bian S, Zhao C, Jin Y, Yu D, Wu X, Zhang D, Cao W, Jing F, Chen G. Associations of urinary polycyclic aromatic hydrocarbons with bone mass density and osteoporosis in U.S. adults, NHANES 2005-2010. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2018; 240:209-218. [PMID: 29738949 DOI: 10.1016/j.envpol.2018.04.108] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 04/23/2018] [Accepted: 04/23/2018] [Indexed: 05/20/2023]
Abstract
UNLABELLED Polycyclic aromatic hydrocarbons (PAHs) are environmental endocrine disruptors, which may modify the bone mineralization. However, epidemiological evidences on this issue were scant. We aimed to investigate the associations of PAHs with bone mass density (BMD) and osteoporosis based on a nationally-representative sample from general U.S. POPULATION Data utilized were extracted from the 2005-2010 National Health and Nutrition Examination Survey (NHANES). Nine urinary PAHs (U-PAHs) metabolites were measured as exposure biomarkers. Associations of specific U-PAHs with BMD and osteoporosis were estimated by multivariable adjusted linear regression models and logistic regression models, respectively. Compared with women at the first tertiles, those at the third tertiles of 1-Hydroxynapthalene, 2-Hydroxyfluorene, 3-Hydroxyphenanthrene, 2-Hydroxyphenanthrene and 9-Hydroxyfluorene had significantly decreased BMD levels [coefficient (β) = -0.023 to -0.014, p < 0.05] or increased likelihoods of osteoporosis [odds ratios (ORs) = 1.86 to 3.36, p < 0.05] at different bone sites. Whereas, elevated BMD levels (β = 0.021, p < 0.05) at trochanter and decreased likelihoods of osteoporosis (OR = 0.33, p < 0.05) at intertrochanter were observed among women at the second tertiles of 1-Hydroxypyrene and 2-Hydroxynapthalene, respectively. Similar results were found for all the population, i.e., combination of men and women. Most of the significant associations disappeared among adult men only. Furthermore, Associations between U-PAHs and BMD were stronger for postmenopausal women when compared with premenopausal group. In conclusion, associations of U-PAHs with BMD and osteoporosis varied by specific U-PAHs and bone sites, as well as menopausal status and genders in U.S. adults.
Collapse
Affiliation(s)
- Jing Guo
- Institute of Environmental Health, Department of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Yun Huang
- Key Laboratory of Reproductive Genetics (Ministry of Education), Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Suchen Bian
- Department of Clinical Medicine, Undergraduate School, Zhejiang University School of Medicine, Hangzhou, China
| | - Chuning Zhao
- Institute of Environmental Health, Department of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Yumin Jin
- Institute of Environmental Health, Department of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Dongdong Yu
- Institute of Environmental Health, Department of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Xinkai Wu
- Department of Clinical Medicine, Undergraduate School, Zhejiang University School of Medicine, Hangzhou, China
| | - Dan Zhang
- Key Laboratory of Reproductive Genetics (Ministry of Education), Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Weiming Cao
- School of Humanities and Social Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Fangyuan Jing
- Department of Epidemiology and Biostatistics, School of Public Health, Hangzhou Medical College, Hangzhou, China.
| | - Guangdi Chen
- Institute of Environmental Health, Department of Public Health, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
77
|
Farr JN, Almeida M. The Spectrum of Fundamental Basic Science Discoveries Contributing to Organismal Aging. J Bone Miner Res 2018; 33:1568-1584. [PMID: 30075061 PMCID: PMC6327947 DOI: 10.1002/jbmr.3564] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 07/25/2018] [Accepted: 07/27/2018] [Indexed: 12/22/2022]
Abstract
Aging research has undergone unprecedented advances at an accelerating rate in recent years, leading to excitement in the field as well as opportunities for imagination and innovation. Novel insights indicate that, rather than resulting from a preprogrammed series of events, the aging process is predominantly driven by fundamental non-adaptive mechanisms that are interconnected, linked, and overlap. To varying degrees, these mechanisms also manifest with aging in bone where they cause skeletal fragility. Because these mechanisms of aging can be manipulated, it might be possible to slow, delay, or alleviate multiple age-related diseases and their complications by targeting conserved genetic signaling pathways, controlled functional networks, and basic biochemical processes. Indeed, findings in various mammalian species suggest that targeting fundamental aging mechanisms (eg, via either loss-of-function or gain-of-function mutations or administration of pharmacological therapies) can extend healthspan; ie, the healthy period of life free of chronic diseases. In this review, we summarize the evidence supporting the role of the spectrum of fundamental basic science discoveries contributing to organismal aging, with emphasis on mammalian studies and in particular aging mechanisms in bone that drive skeletal fragility. These mechanisms or aging hallmarks include: genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, deregulated nutrient sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, and altered intercellular communication. Because these mechanisms are linked, interventions that ameliorate one hallmark can in theory ameliorate others. In the field of bone and mineral research, current challenges include defining the relative contributions of each aging hallmark to the natural skeletal aging process, better understanding the complex interconnections among the hallmarks, and identifying the most effective therapeutic strategies to safely target multiple hallmarks. Based on their interconnections, it may be feasible to simultaneously interfere with several fundamental aging mechanisms to alleviate a wide spectrum of age-related chronic diseases, including osteoporosis. © 2018 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Joshua N Farr
- Division of Endocrinology and Metabolism and Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Maria Almeida
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
78
|
Stringer JM, Winship A, Liew SH, Hutt K. The capacity of oocytes for DNA repair. Cell Mol Life Sci 2018; 75:2777-2792. [PMID: 29748894 PMCID: PMC11105623 DOI: 10.1007/s00018-018-2833-9] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 03/27/2018] [Accepted: 05/02/2018] [Indexed: 12/18/2022]
Abstract
Female fertility and offspring health are critically dependent on the maintenance of an adequate supply of high-quality oocytes. Like somatic cells, oocytes are subject to a variety of different types of DNA damage arising from endogenous cellular processes and exposure to exogenous genotoxic stressors. While the repair of intentionally induced DNA double strand breaks in gametes during meiotic recombination is well characterised, less is known about the ability of oocytes to repair pathological DNA damage and the relative contribution of DNA repair to oocyte quality is not well defined. This review will discuss emerging data suggesting that oocytes are in fact capable of efficient DNA repair and that DNA repair may be an important mechanism for ensuring female fertility, as well as the transmission of high-quality genetic material to subsequent generations.
Collapse
Affiliation(s)
- Jessica M Stringer
- Ovarian Biology Laboratory, Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Australia
| | - Amy Winship
- Ovarian Biology Laboratory, Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Australia
| | - Seng H Liew
- Ovarian Biology Laboratory, Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Australia
| | - Karla Hutt
- Ovarian Biology Laboratory, Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Australia.
| |
Collapse
|
79
|
Schäfer A, Mekker B, Mallick M, Vastolo V, Karaulanov E, Sebastian D, von der Lippen C, Epe B, Downes DJ, Scholz C, Niehrs C. Impaired DNA demethylation of C/EBP sites causes premature aging. Genes Dev 2018; 32:742-762. [PMID: 29884649 PMCID: PMC6049513 DOI: 10.1101/gad.311969.118] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 05/07/2018] [Indexed: 12/25/2022]
Abstract
Here, Schäfer et al. investigated whether DNA methylation alterations are involved in aging. Using knockout mice for adapter proteins for site-specific demethylation by TET methylcytosine dioxygenases Gadd45a and Ing1, they show that enhancer methylation can affect aging and imply that C/EBP proteins play an unexpected role in this process. Changes in DNA methylation are among the best-documented epigenetic alterations accompanying organismal aging. However, whether and how altered DNA methylation is causally involved in aging have remained elusive. GADD45α (growth arrest and DNA damage protein 45A) and ING1 (inhibitor of growth family member 1) are adapter proteins for site-specific demethylation by TET (ten-eleven translocation) methylcytosine dioxygenases. Here we show that Gadd45a/Ing1 double-knockout mice display segmental progeria and phenocopy impaired energy homeostasis and lipodystrophy characteristic of Cebp (CCAAT/enhancer-binding protein) mutants. Correspondingly, GADD45α occupies C/EBPβ/δ-dependent superenhancers and, cooperatively with ING1, promotes local DNA demethylation via long-range chromatin loops to permit C/EBPβ recruitment. The results indicate that enhancer methylation can affect aging and imply that C/EBP proteins play an unexpected role in this process. Our study suggests a causal nexus between DNA demethylation, metabolism, and organismal aging.
Collapse
Affiliation(s)
- Andrea Schäfer
- Institute of Molecular Biology (IMB), 55128 Mainz, Germany
| | | | | | | | | | | | - Carina von der Lippen
- Institute of Pharmacy and Biochemistry, Johannes Gutenberg University of Mainz, 55128 Mainz, Germany
| | - Bernd Epe
- Institute of Pharmacy and Biochemistry, Johannes Gutenberg University of Mainz, 55128 Mainz, Germany
| | - Damien J Downes
- Medical Research Council Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, United Kingdom
| | - Carola Scholz
- Institute of Molecular Biology (IMB), 55128 Mainz, Germany
| | - Christof Niehrs
- Institute of Molecular Biology (IMB), 55128 Mainz, Germany.,German Cancer Research Center, Division of Molecular Embryology, German Cancer Research Center-Center for Molecular Biology (DKFZ-ZMBH) Alliance, 69120 Heidelberg, Germany
| |
Collapse
|
80
|
Baar MP, Perdiguero E, Muñoz-Cánoves P, de Keizer PLJ. Musculoskeletal senescence: a moving target ready to be eliminated. Curr Opin Pharmacol 2018; 40:147-155. [DOI: 10.1016/j.coph.2018.05.007] [Citation(s) in RCA: 121] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 05/16/2018] [Indexed: 11/17/2022]
|
81
|
Saquib Q, Siddiqui MA, Ahmad J, Ansari SM, Al-Wathnani HA, Rensing C. 6-OHBDE-47 induces transcriptomic alterations of CYP1A1, XRCC2, HSPA1A, EGR1 genes and trigger apoptosis in HepG2 cells. Toxicology 2018; 400-401:40-47. [DOI: 10.1016/j.tox.2018.03.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Revised: 03/17/2018] [Accepted: 03/26/2018] [Indexed: 12/19/2022]
|
82
|
Inactivation of RAD52 and HDF1 DNA repair genes leads to premature chronological aging and cellular instability. J Biosci 2018; 42:219-230. [PMID: 28569246 DOI: 10.1007/s12038-017-9684-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The present study aims to investigate the role of radiation sensitive 52 (RAD52) and high-affinity DNA binding factor 1 (HDF1) DNA repair genes on the life span of budding yeasts during chronological aging. Wild type (wt) and rad52, hdf1, and rad52 hdf1 mutant Saccharomyces cerevisiae strains were used. Chronological aging and survival assays were studied by clonogenic assay and drop test. DNA damage was analyzed by electrophoresis after phenol extraction. Mutant analysis, colony forming units and the index of respiratory competence were studied by growing on dextrose and glycerol plates as a carbon source. Rad52 and rad52 hdf1 mutants showed a gradual decrease in surviving fraction in relation to wt and hdf1 mutant during aging. Genomic DNA was spontaneously more degraded during aging, mainly in rad52 mutants. This strain showed an increased percentage of revertant colonies. Moreover, all mutants showed a decrease in the index of respiratory competence during aging. The inactivation of RAD52 leads to premature chronological aging with an increase in DNA degradation and mutation frequency. In addition, RAD52 and HDF1 contribute to maintain the metabolic state, in a different way, during chronological aging. The results obtained could have important implications in the chronobiology of aging.
Collapse
|
83
|
Popa-Wagner A, Glavan DG, Olaru A, Olaru DG, Margaritescu O, Tica O, Surugiu R, Sandu RE. Present Status and Future Challenges of New Therapeutic Targets in Preclinical Models of Stroke in Aged Animals with/without Comorbidities. Int J Mol Sci 2018; 19:ijms19020356. [PMID: 29370078 PMCID: PMC5855578 DOI: 10.3390/ijms19020356] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 01/18/2018] [Accepted: 01/22/2018] [Indexed: 01/02/2023] Open
Abstract
The aging process, comorbidities, and age-associated diseases are closely dependent on each other. Cerebral ischemia impacts a wide range of systems in an age-dependent manner. However, the aging process has many facets which are influenced by the genetic background and epigenetic or environmental factors, which can explain why some people age differently than others. Therefore, there is an urgent need to identify age-related changes in body functions or structures that increase the risk for stroke and which are associated with a poor outcome. Multimodal imaging, electrophysiology, cell biology, proteomics, and transcriptomics, offer a useful approach to link structural and functional changes in the aging brain, with or without comorbidities, to post-stroke rehabilitation. This can help us to improve our knowledge about senescence firstly, and in this context, aids in elucidating the pathophysiology of age-related diseases that allows us to develop therapeutic strategies or prevent diseases. These processes, including potential therapeutical interventions, need to be studied first in relevant preclinical models using aged animals, with and without comorbidities. Therefore, preclinical research on ischemic stroke should consider age as the most important risk factor for cerebral ischemia. Furthermore, the identification of effective therapeutic strategies, corroborated with successful translational studies, will have a dramatic impact on the lives of millions of people with cerebrovascular diseases.
Collapse
Affiliation(s)
- Aurel Popa-Wagner
- Griffith University School of Medicine, Gold Coast Campus, QLD, Queensland Eye Institute, Brisbane, QLD 4101, Australia.
- Department of Functional Sciences, Center of Clinical and Experimental Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania.
| | - Daniela-Gabriela Glavan
- Psychiatry Clinic Hospital, University of Medicine and Pharmacy of Craiova, Petru Rares Street 2, 200349 Craiova, Romania.
| | - Andrei Olaru
- Department of Ophthalmology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania.
| | | | - Otilia Margaritescu
- Department of Neurosurgery, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania.
| | - Oana Tica
- Department of "Mother and Child", University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania.
| | - Roxana Surugiu
- Department of Functional Sciences, Center of Clinical and Experimental Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania.
| | - Raluca Elena Sandu
- Department of Functional Sciences, Center of Clinical and Experimental Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania.
| |
Collapse
|
84
|
Abbosh PH, Plimack ER. Molecular and Clinical Insights into the Role and Significance of Mutated DNA Repair Genes in Bladder Cancer. Bladder Cancer 2018; 4:9-18. [PMID: 29430503 PMCID: PMC5798524 DOI: 10.3233/blc-170129] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
DNA damage response and repair genes (DDR genes) are commonly mutated in bladder cancer. The biological impact of these mutations is an area of intense basic, translational, and clinical interest. As next generation sequencing increases its foothold in the treatment of localized and advanced bladder cancer, the role of DDR genes will continue to evolve. We review the inventory and biology of DDR genes in bladder cancer and describe known and candidate roles for loss of DDR genes to engender therapeutic susceptibilities to various anti-cancer agents.
Collapse
Affiliation(s)
- Philip H Abbosh
- Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, PA, USA.,Department of Urology, Einstein Medical Center, Philadelphia, PA, USA
| | - Elizabeth R Plimack
- Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| |
Collapse
|
85
|
Al-Hashimi F, J. Diaz-Cano S. Multi-target analysis of neoplasms for the evaluation of tumor progression: stochastic approach of biologic processes. AIMS MOLECULAR SCIENCE 2018. [DOI: 10.3934/molsci.2018.1.14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
86
|
Relevance of the p53-MDM2 axis to aging. Cell Death Differ 2017; 25:169-179. [PMID: 29192902 DOI: 10.1038/cdd.2017.187] [Citation(s) in RCA: 155] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 09/19/2017] [Accepted: 09/21/2017] [Indexed: 12/13/2022] Open
Abstract
In response to varying stress signals, the p53 tumor suppressor is able to promote repair, survival, or elimination of damaged cells - processes that have great relevance to organismal aging. Although the link between p53 and cancer is well established, the contribution of p53 to the aging process is less clear. Delineating how p53 regulates distinct aging hallmarks such as cellular senescence, genomic instability, mitochondrial dysfunction, and altered metabolic pathways will be critical. Mouse models have further revealed the centrality and complexity of the p53 network in aging processes. While naturally aged mice have linked longevity with declining p53 function, some accelerated aging mice present with chronic p53 activation, whose phenotypes can be rescued upon p53 deficiency. Further, direct modulation of the p53-MDM2 axis has correlated elevated p53 activity with either early aging or with delayed-onset aging. We speculate that p53-mediated aging phenotypes in these mice must have (1) stably active p53 due to MDM2 dysregulation or chronic stress or (2) shifted p53 outcomes. Pinpointing which p53 stressors, modifications, and outcomes drive aging processes will provide further insights into our understanding of the human aging process and could have implications for both cancer and aging therapeutics.
Collapse
|
87
|
Kim J, Vaish V, Feng M, Field K, Chatzistamou I, Shim M. Transgenic expression of cyclooxygenase-2 (COX2) causes premature aging phenotypes in mice. Aging (Albany NY) 2017; 8:2392-2406. [PMID: 27750221 PMCID: PMC5115895 DOI: 10.18632/aging.101060] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 09/25/2016] [Indexed: 12/20/2022]
Abstract
Cyclooxygenase (COX) is a key enzyme in the biosynthesis of prostanoids, lipid signaling molecules that regulate various physiological processes. COX2, one of the isoforms of COX, is highly inducible in response to a wide variety of cellular and environmental stresses. Increased COX2 expression is thought to play a role in the pathogenesis of many age-related diseases. COX2 expression is also reported to be increased in the tissues of aged humans and mice, which suggests the involvement of COX2 in the aging process. However, it is not clear whether the increased COX2 expression is causal to or a result of aging. We have now addressed this question by creating an inducible COX2 transgenic mouse model. Here we show that post-natal expression of COX2 led to a panel of aging-related phenotypes. The expression of p16, p53, and phospho-H2AX was increased in the tissues of COX2 transgenic mice. Additionally, adult mouse lung fibroblasts from COX2 transgenic mice exhibited increased expression of the senescence-associated β-galactosidase. Our study reveals that the increased COX2 expression has an impact on the aging process and suggests that modulation of COX2 and its downstream signaling may be an approach for intervention of age-related disorders.
Collapse
Affiliation(s)
- Joohwee Kim
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA.,Center for Colon Cancer Research, University of South Carolina, Columbia, SC 29208, USA
| | - Vivek Vaish
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA.,Center for Colon Cancer Research, University of South Carolina, Columbia, SC 29208, USA
| | - Mingxiao Feng
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA.,Center for Colon Cancer Research, University of South Carolina, Columbia, SC 29208, USA
| | - Kevin Field
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Ioulia Chatzistamou
- Department of Pathology, Microbiology & Immunology, School of Medicine, University of South Carolina, Columbia, SC 29209, USA
| | - Minsub Shim
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA.,Center for Colon Cancer Research, University of South Carolina, Columbia, SC 29208, USA
| |
Collapse
|
88
|
MiR-455-3p activates Nrf2/ARE signaling via HDAC2 and protects osteoblasts from oxidative stress. Int J Biol Macromol 2017; 107:2094-2101. [PMID: 29042277 DOI: 10.1016/j.ijbiomac.2017.10.080] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 10/13/2017] [Accepted: 10/13/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND The important role of miR-455-3p in the pathogenesis of bone metabolism associated diseases is gradually emerging. This study aims to ascertain the involvement of miR-455-3p and its underlying mechanisms in osteoporosis. METHODS The osteoblast cell lines MC3T3-E1 was treated with ferric ammonium citrate (FAC) to mimic a pathological environment for osteoporosis. The cytotoxic effect of iron overload was assessed by proliferation, apoptosis and oxidative stress of osteoblasts using commercial kits. Molecular biological methods, including qRT-PCR analysis, cell transfection and luciferase reporter assays were used to explain the role of miR-455-3p and its potential mechanisms in osteoblast apoptosis. RESULTS FAC dramatically inhibited the proliferation of osteoblast cells MC3T3-E1 but increased the apoptosis. We also observed that FAC significantly down-regulated miR-455-3p in MC3T3-E1 cells but enhanced HDAC2 protein level. Moreover, miR-455-3p overexpression eliminated the effects of iron overload on osteoblast cell proliferation, apoptosis and oxidative stress. In addition, miR-455-3p regulated osteoblast cell proliferation, apoptosis and oxidative stress through regulating HDAC2-Nrf2/ARE signaling pathway. MiR-455-3p overexpression alleviated the oxidative stress injury in osteoporosis mice. CONCLUSION Our results demonstrated that miR-455-3p activated Nrf2/ARE signal pathway through suppressing Keap1 via negative regulating HDAC2 protein level, thereby suppressing oxidative stress and promoting osteoblasts growth.
Collapse
|
89
|
The Impact of Oxidative Stress on the Bone System in Response to the Space Special Environment. Int J Mol Sci 2017; 18:ijms18102132. [PMID: 29023398 PMCID: PMC5666814 DOI: 10.3390/ijms18102132] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 09/28/2017] [Accepted: 10/09/2017] [Indexed: 12/25/2022] Open
Abstract
The space special environment mainly includes microgravity, radiation, vacuum and extreme temperature, which seriously threatens an astronaut’s health. Bone loss is one of the most significant alterations in mammalians after long-duration habitation in space. In this review, we summarize the crucial roles of major factors—namely radiation and microgravity—in space in oxidative stress generation in living organisms, and the inhibitory effect of oxidative stress on bone formation. We discussed the possible mechanisms of oxidative stress-induced skeletal involution, and listed some countermeasures that have therapeutic potentials for bone loss via oxidative stress antagonism. Future research for better understanding the oxidative stress caused by space environment and the development of countermeasures against oxidative damage accordingly may facilitate human beings to live more safely in space and explore deeper into the universe.
Collapse
|
90
|
Ding AJ, Zheng SQ, Huang XB, Xing TK, Wu GS, Sun HY, Qi SH, Luo HR. Current Perspective in the Discovery of Anti-aging Agents from Natural Products. NATURAL PRODUCTS AND BIOPROSPECTING 2017; 7:335-404. [PMID: 28567542 PMCID: PMC5655361 DOI: 10.1007/s13659-017-0135-9] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2017] [Accepted: 05/16/2017] [Indexed: 05/18/2023]
Abstract
Aging is a process characterized by accumulating degenerative damages, resulting in the death of an organism ultimately. The main goal of aging research is to develop therapies that delay age-related diseases in human. Since signaling pathways in aging of Caenorhabditis elegans (C. elegans), fruit flies and mice are evolutionarily conserved, compounds extending lifespan of them by intervening pathways of aging may be useful in treating age-related diseases in human. Natural products have special resource advantage and with few side effect. Recently, many compounds or extracts from natural products slowing aging and extending lifespan have been reported. Here we summarized these compounds or extracts and their mechanisms in increasing longevity of C. elegans or other species, and the prospect in developing anti-aging medicine from natural products.
Collapse
Affiliation(s)
- Ai-Jun Ding
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China
- University of Chinese Academy of Sciences, Beijing, 100039, China
| | - Shan-Qing Zheng
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China
- University of Chinese Academy of Sciences, Beijing, 100039, China
| | - Xiao-Bing Huang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China
- University of Chinese Academy of Sciences, Beijing, 100039, China
| | - Ti-Kun Xing
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China
- University of Chinese Academy of Sciences, Beijing, 100039, China
| | - Gui-Sheng Wu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China
- Key Laboratory for Aging and Regenerative Medicine, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Hua-Ying Sun
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China
| | - Shu-Hua Qi
- Guangdong Key Laboratory of Marine Material Medical, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, 510301, Guangdong, China
| | - Huai-Rong Luo
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China.
- Key Laboratory for Aging and Regenerative Medicine, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China.
- Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming Institute of Botany, Chinese Academy of Sciences, 134 Lanhei Road, Kunming, 650201, Yunnan, China.
| |
Collapse
|
91
|
Park JH, Yoo Y, Park YJ. Epigenetics: Linking Nutrition to Molecular Mechanisms in Aging. Prev Nutr Food Sci 2017; 22:81-89. [PMID: 28702424 PMCID: PMC5503416 DOI: 10.3746/pnf.2017.22.2.81] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 03/07/2017] [Indexed: 11/06/2022] Open
Abstract
Healthy aging has become a major goal of public health. Many studies have provided evidence and theories to explain molecular mechanisms of the aging process. Recent studies suggest that epigenetic mechanisms are responsible for life span and the progression of aging. Epigenetics is a fascinating field of molecular biology, which studies heritable modifications of DNA and histones that regulate gene expression without altering the DNA sequence. DNA methylation is a major epigenetic mark that shows progressive changes during aging. Recent studies have investigated aging-related DNA methylation as a biomarker that predicts cellular age. Interestingly, growing evidence proposes that nutrients play a crucial role in the regulation of epigenetic modifiers. Because various nutrients and their metabolites function as substrates or cofactors for epigenetic modifiers, nutrition can modulate or reverse epigenetic marks in the genome as well as expression patterns. Here, we will review the results on aging-associated epigenetic modifications and the possible mechanisms by which nutrition, including nutrient availability and bioactive compounds, regulate epigenetic changes and affect aging physiology.
Collapse
Affiliation(s)
- Joo Hyun Park
- Metabolism and Epigenetics Laboratory, Department of Nutritional Science and Food Management, Ewha Womans University, Seoul 03760, Korea
| | - Yeongran Yoo
- Metabolism and Epigenetics Laboratory, Department of Nutritional Science and Food Management, Ewha Womans University, Seoul 03760, Korea
| | - Yoon Jung Park
- Metabolism and Epigenetics Laboratory, Department of Nutritional Science and Food Management, Ewha Womans University, Seoul 03760, Korea
| |
Collapse
|
92
|
Baar MP, Brandt RMC, Putavet DA, Klein JDD, Derks KWJ, Bourgeois BRM, Stryeck S, Rijksen Y, van Willigenburg H, Feijtel DA, van der Pluijm I, Essers J, van Cappellen WA, van IJcken WF, Houtsmuller AB, Pothof J, de Bruin RWF, Madl T, Hoeijmakers JHJ, Campisi J, de Keizer PLJ. Targeted Apoptosis of Senescent Cells Restores Tissue Homeostasis in Response to Chemotoxicity and Aging. Cell 2017; 169:132-147.e16. [PMID: 28340339 DOI: 10.1016/j.cell.2017.02.031] [Citation(s) in RCA: 984] [Impact Index Per Article: 123.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 12/29/2016] [Accepted: 02/22/2017] [Indexed: 02/06/2023]
Abstract
The accumulation of irreparable cellular damage restricts healthspan after acute stress or natural aging. Senescent cells are thought to impair tissue function, and their genetic clearance can delay features of aging. Identifying how senescent cells avoid apoptosis allows for the prospective design of anti-senescence compounds to address whether homeostasis can also be restored. Here, we identify FOXO4 as a pivot in senescent cell viability. We designed a FOXO4 peptide that perturbs the FOXO4 interaction with p53. In senescent cells, this selectively causes p53 nuclear exclusion and cell-intrinsic apoptosis. Under conditions where it was well tolerated in vivo, this FOXO4 peptide neutralized doxorubicin-induced chemotoxicity. Moreover, it restored fitness, fur density, and renal function in both fast aging XpdTTD/TTD and naturally aged mice. Thus, therapeutic targeting of senescent cells is feasible under conditions where loss of health has already occurred, and in doing so tissue homeostasis can effectively be restored.
Collapse
Affiliation(s)
- Marjolein P Baar
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, Wytemaweg 80, 3015CN, Rotterdam, the Netherlands
| | - Renata M C Brandt
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, Wytemaweg 80, 3015CN, Rotterdam, the Netherlands
| | - Diana A Putavet
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, Wytemaweg 80, 3015CN, Rotterdam, the Netherlands
| | - Julian D D Klein
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, Wytemaweg 80, 3015CN, Rotterdam, the Netherlands
| | - Kasper W J Derks
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, Wytemaweg 80, 3015CN, Rotterdam, the Netherlands
| | - Benjamin R M Bourgeois
- Institute of Molecular Biology & Biochemistry, Center of Molecular Medicine, Medical University of Graz, 8010 Graz, Austria
| | - Sarah Stryeck
- Institute of Molecular Biology & Biochemistry, Center of Molecular Medicine, Medical University of Graz, 8010 Graz, Austria
| | - Yvonne Rijksen
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, Wytemaweg 80, 3015CN, Rotterdam, the Netherlands
| | - Hester van Willigenburg
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, Wytemaweg 80, 3015CN, Rotterdam, the Netherlands
| | - Danny A Feijtel
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, Wytemaweg 80, 3015CN, Rotterdam, the Netherlands
| | - Ingrid van der Pluijm
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, Wytemaweg 80, 3015CN, Rotterdam, the Netherlands; Department of Vascular Surgery, Erasmus University Medical Center Rotterdam, Wytemaweg 80, 3015CN, Rotterdam, the Netherlands
| | - Jeroen Essers
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, Wytemaweg 80, 3015CN, Rotterdam, the Netherlands; Department of Vascular Surgery, Erasmus University Medical Center Rotterdam, Wytemaweg 80, 3015CN, Rotterdam, the Netherlands; Department of Radiation Oncology, Erasmus University Medical Center Rotterdam, Wytemaweg 80, 3015CN, Rotterdam, the Netherlands
| | - Wiggert A van Cappellen
- Erasmus Optical Imaging Center and Department of Pathology, Erasmus University Medical Center Rotterdam, Wytemaweg 80, 3015CN, Rotterdam, the Netherlands
| | - Wilfred F van IJcken
- Department of Cell Biology, Erasmus University Medical Center Rotterdam, Wytemaweg 80, 3015CN, Rotterdam, the Netherlands
| | - Adriaan B Houtsmuller
- Erasmus Optical Imaging Center and Department of Pathology, Erasmus University Medical Center Rotterdam, Wytemaweg 80, 3015CN, Rotterdam, the Netherlands
| | - Joris Pothof
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, Wytemaweg 80, 3015CN, Rotterdam, the Netherlands
| | - Ron W F de Bruin
- Department of Surgery, Erasmus University Medical Center Rotterdam, Wytemaweg 80, 3015CN, Rotterdam, the Netherlands
| | - Tobias Madl
- Institute of Molecular Biology & Biochemistry, Center of Molecular Medicine, Medical University of Graz, 8010 Graz, Austria
| | - Jan H J Hoeijmakers
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, Wytemaweg 80, 3015CN, Rotterdam, the Netherlands
| | - Judith Campisi
- The Buck Institute for Research on Aging, 8001 Redwood Blvd., Novato, CA 94945, USA; Lawrence Berkeley National Laboratories, Berkeley, CA 94720, USA
| | - Peter L J de Keizer
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, Wytemaweg 80, 3015CN, Rotterdam, the Netherlands; The Buck Institute for Research on Aging, 8001 Redwood Blvd., Novato, CA 94945, USA.
| |
Collapse
|
93
|
Peregrim I. Why we age — a new evolutionary view. Biologia (Bratisl) 2017. [DOI: 10.1515/biolog-2017-0064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
94
|
Chatzinikolaou G, Apostolou Z, Aid-Pavlidis T, Ioannidou A, Karakasilioti I, Papadopoulos GL, Aivaliotis M, Tsekrekou M, Strouboulis J, Kosteas T, Garinis GA. ERCC1-XPF cooperates with CTCF and cohesin to facilitate the developmental silencing of imprinted genes. Nat Cell Biol 2017; 19:421-432. [PMID: 28368372 DOI: 10.1038/ncb3499] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Accepted: 02/24/2017] [Indexed: 12/15/2022]
Abstract
Inborn defects in DNA repair are associated with complex developmental disorders whose causal mechanisms are poorly understood. Using an in vivo biotinylation tagging approach in mice, we show that the nucleotide excision repair (NER) structure-specific endonuclease ERCC1-XPF complex interacts with the insulator binding protein CTCF, the cohesin subunits SMC1A and SMC3 and with MBD2; the factors co-localize with ATRX at the promoters and control regions (ICRs) of imprinted genes during postnatal hepatic development. Loss of Ercc1 or exposure to MMC triggers the localization of CTCF to heterochromatin, the dissociation of the CTCF-cohesin complex and ATRX from promoters and ICRs, altered histone marks and the aberrant developmental expression of imprinted genes without altering DNA methylation. We propose that ERCC1-XPF cooperates with CTCF and cohesin to facilitate the developmental silencing of imprinted genes and that persistent DNA damage triggers chromatin changes that affect gene expression programs associated with NER disorders.
Collapse
Affiliation(s)
- Georgia Chatzinikolaou
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Nikolaou Plastira 100, 70013 Heraklion, Crete, Greece
| | - Zivkos Apostolou
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Nikolaou Plastira 100, 70013 Heraklion, Crete, Greece
- Department of Biology, University of Crete, Vassilika Vouton, GR71409 Heraklion, Crete, Greece
| | - Tamara Aid-Pavlidis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Nikolaou Plastira 100, 70013 Heraklion, Crete, Greece
| | - Anna Ioannidou
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Nikolaou Plastira 100, 70013 Heraklion, Crete, Greece
- Department of Biology, University of Crete, Vassilika Vouton, GR71409 Heraklion, Crete, Greece
| | - Ismene Karakasilioti
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Nikolaou Plastira 100, 70013 Heraklion, Crete, Greece
| | - Giorgio L Papadopoulos
- Department of Biology, University of Crete, Vassilika Vouton, GR71409 Heraklion, Crete, Greece
- Division of Molecular Oncology, Biomedical Sciences Research Center 'Alexander Fleming', GR 16672 Vari, Greece
| | - Michalis Aivaliotis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Nikolaou Plastira 100, 70013 Heraklion, Crete, Greece
| | - Maria Tsekrekou
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Nikolaou Plastira 100, 70013 Heraklion, Crete, Greece
- Department of Biology, University of Crete, Vassilika Vouton, GR71409 Heraklion, Crete, Greece
| | - John Strouboulis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Nikolaou Plastira 100, 70013 Heraklion, Crete, Greece
- Division of Molecular Oncology, Biomedical Sciences Research Center 'Alexander Fleming', GR 16672 Vari, Greece
| | - Theodore Kosteas
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Nikolaou Plastira 100, 70013 Heraklion, Crete, Greece
| | - George A Garinis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Nikolaou Plastira 100, 70013 Heraklion, Crete, Greece
- Department of Biology, University of Crete, Vassilika Vouton, GR71409 Heraklion, Crete, Greece
| |
Collapse
|
95
|
de Keizer PL. The Fountain of Youth by Targeting Senescent Cells? Trends Mol Med 2017; 23:6-17. [DOI: 10.1016/j.molmed.2016.11.006] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 11/16/2016] [Accepted: 11/17/2016] [Indexed: 12/19/2022]
|
96
|
Nicolai S, Rossi A, Di Daniele N, Melino G, Annicchiarico-Petruzzelli M, Raschellà G. DNA repair and aging: the impact of the p53 family. Aging (Albany NY) 2016; 7:1050-65. [PMID: 26668111 PMCID: PMC4712331 DOI: 10.18632/aging.100858] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Cells are constantly exposed to endogenous and exogenous factors that threaten the integrity of their DNA. The maintenance of genome stability is of paramount importance in the prevention of both cancer and aging processes. To deal with DNA damage, cells put into operation a sophisticated and coordinated mechanism, collectively known as DNA damage response (DDR). The DDR orchestrates different cellular processes, such as DNA repair, senescence and apoptosis. Among the key factors of the DDR, the related proteins p53, p63 and p73, all belonging to the same family of transcription factors, play multiple relevant roles. Indeed, the members of this family are directly involved in the induction of cell cycle arrest that is necessary to allow the cells to repair. Alternatively, they can promote cell death in case of prolonged or irreparable DNA damage. They also take part in a more direct task by modulating the expression of core factors involved in the process of DNA repair or by directly interacting with them. In this review we will analyze the fundamental roles of the p53 family in the aging process through their multifaceted function in DDR.
Collapse
Affiliation(s)
- Sara Nicolai
- Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Antonello Rossi
- Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Nicola Di Daniele
- Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Gerry Melino
- Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata", 00133 Rome, Italy.,Medical Research Council, Toxicology Unit, Hodgkin Building, Leicester University, Leicester LE1 9HN, UK
| | | | - Giuseppe Raschellà
- ENEA Research Center Casaccia, Laboratory of Biosafety and Risk Assessment, 00123 Rome, Italy
| |
Collapse
|
97
|
Kõks S, Dogan S, Tuna BG, González-Navarro H, Potter P, Vandenbroucke RE. Mouse models of ageing and their relevance to disease. Mech Ageing Dev 2016; 160:41-53. [PMID: 27717883 DOI: 10.1016/j.mad.2016.10.001] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 09/26/2016] [Accepted: 10/03/2016] [Indexed: 12/28/2022]
Abstract
Ageing is a process that gradually increases the organism's vulnerability to death. It affects different biological pathways, and the underlying cellular mechanisms are complex. In view of the growing disease burden of ageing populations, increasing efforts are being invested in understanding the pathways and mechanisms of ageing. We review some mouse models commonly used in studies on ageing, highlight the advantages and disadvantages of the different strategies, and discuss their relevance to disease susceptibility. In addition to addressing the genetics and phenotypic analysis of mice, we discuss examples of models of delayed or accelerated ageing and their modulation by caloric restriction.
Collapse
Affiliation(s)
- Sulev Kõks
- University of Tartu, Tartu, Estonia and Estonian University of Life Sciences, Tartu, Estonia.
| | - Soner Dogan
- Yeditepe University, School of Medicine, Department of Medical Biology, Istanbul, Turkey.
| | - Bilge Guvenc Tuna
- Yeditepe University, School of Medicine, Department of Biophysics, Istanbul, Turkey.
| | - Herminia González-Navarro
- Institute of Health Research-INCLIVA, 46010 Valencia, Spain and CIBER de Diabetes y Enfermedades Metabólicas (CIBERDEM), 28029 Madrid, Spain.
| | - Paul Potter
- Mammalian Genetics Unit, MRC Harwell, Oxfordshire, UK.
| | - Roosmarijn E Vandenbroucke
- Inflammation Research Center, VIB, Ghent, Belgium, Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.
| |
Collapse
|
98
|
Laven JS, Visser JA, Uitterlinden AG, Vermeij WP, Hoeijmakers JH. Menopause: Genome stability as new paradigm. Maturitas 2016; 92:15-23. [DOI: 10.1016/j.maturitas.2016.07.006] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 07/07/2016] [Indexed: 11/27/2022]
|
99
|
Huang J, Lu H, Lu Y, Hung P, Lin Y, Lin C, Yang C, Wong T, Lu S, Lin C. Enhancement of the genotoxicity of benzo[a]pyrene by arecoline through suppression of DNA repair in HEp-2 cells. Toxicol In Vitro 2016; 33:80-7. [DOI: 10.1016/j.tiv.2016.02.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 01/06/2016] [Accepted: 02/13/2016] [Indexed: 01/02/2023]
|
100
|
Pan MR, Li K, Lin SY, Hung WC. Connecting the Dots: From DNA Damage and Repair to Aging. Int J Mol Sci 2016; 17:ijms17050685. [PMID: 27164092 PMCID: PMC4881511 DOI: 10.3390/ijms17050685] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 04/19/2016] [Accepted: 05/03/2016] [Indexed: 01/15/2023] Open
Abstract
Mammalian cells evolve a delicate system, the DNA damage response (DDR) pathway, to monitor genomic integrity and to prevent the damage from both endogenous end exogenous insults. Emerging evidence suggests that aberrant DDR and deficient DNA repair are strongly associated with cancer and aging. Our understanding of the core program of DDR has made tremendous progress in the past two decades. However, the long list of the molecules involved in the DDR and DNA repair continues to grow and the roles of the new “dots” are under intensive investigation. Here, we review the connection between DDR and DNA repair and aging and discuss the potential mechanisms by which deficient DNA repair triggers systemic effects to promote physiological or pathological aging.
Collapse
Affiliation(s)
- Mei-Ren Pan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsoung Medical University, Kaohsiung 807, Taiwan.
| | - Kaiyi Li
- The Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Shiaw-Yih Lin
- Department of Systems Biology, MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Wen-Chun Hung
- National Institute of Cancer Research, National Health Research Institutes, Tainan 704, Taiwan.
| |
Collapse
|