51
|
Blagosklonny MV. DNA- and telomere-damage does not limit lifespan: evidence from rapamycin. Aging (Albany NY) 2021; 13:3167-3175. [PMID: 33578394 PMCID: PMC7906135 DOI: 10.18632/aging.202674] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 02/10/2021] [Indexed: 12/13/2022]
Abstract
Failure of rapamycin to extend lifespan in DNA repair mutant and telomerase-knockout mice, while extending lifespan in normal mice, indicates that neither DNA damage nor telomere shortening limits normal lifespan or causes normal aging.
Collapse
|
52
|
Birkisdóttir MB, Jaarsma D, Brandt RMC, Barnhoorn S, Vliet N, Imholz S, Oostrom CT, Nagarajah B, Portilla Fernández E, Roks AJM, Elgersma Y, Steeg H, Ferreira JA, Pennings JLA, Hoeijmakers JHJ, Vermeij WP, Dollé MET. Unlike dietary restriction, rapamycin fails to extend lifespan and reduce transcription stress in progeroid DNA repair-deficient mice. Aging Cell 2021; 20:e13302. [PMID: 33484480 PMCID: PMC7884048 DOI: 10.1111/acel.13302] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 11/03/2020] [Accepted: 12/07/2020] [Indexed: 12/31/2022] Open
Abstract
Dietary restriction (DR) and rapamycin extend healthspan and life span across multiple species. We have recently shown that DR in progeroid DNA repair‐deficient mice dramatically extended healthspan and trippled life span. Here, we show that rapamycin, while significantly lowering mTOR signaling, failed to improve life span nor healthspan of DNA repair‐deficient Ercc1∆/− mice, contrary to DR tested in parallel. Rapamycin interventions focusing on dosage, gender, and timing all were unable to alter life span. Even genetically modifying mTOR signaling failed to increase life span of DNA repair‐deficient mice. The absence of effects by rapamycin on P53 in brain and transcription stress in liver is in sharp contrast with results obtained by DR, and appoints reducing DNA damage and transcription stress as an important mode of action of DR, lacking by rapamycin. Together, this indicates that mTOR inhibition does not mediate the beneficial effects of DR in progeroid mice, revealing that DR and rapamycin strongly differ in their modes of action.
Collapse
Affiliation(s)
- María B. Birkisdóttir
- Princess Máxima Center for Pediatric Oncology, Genome Instability and Nutrition ONCODE Institute Utrecht The Netherlands
| | - Dick Jaarsma
- Department of Neuroscience Erasmus MC Rotterdam The Netherlands
| | | | - Sander Barnhoorn
- Department of Molecular Genetics Erasmus MC Rotterdam The Netherlands
| | - Nicole Vliet
- Department of Molecular Genetics Erasmus MC Rotterdam The Netherlands
| | - Sandra Imholz
- Centre for Health Protection National Institute for Public Health and the Environment (RIVM Bilthoven The Netherlands
| | - Conny T. Oostrom
- Centre for Health Protection National Institute for Public Health and the Environment (RIVM Bilthoven The Netherlands
| | - Bhawani Nagarajah
- Centre for Health Protection National Institute for Public Health and the Environment (RIVM Bilthoven The Netherlands
| | - Eliana Portilla Fernández
- Division of Vascular Medicine and Pharmacology Department of Internal Medicine Erasmus MC Rotterdam The Netherlands
| | - Anton J. M. Roks
- Division of Vascular Medicine and Pharmacology Department of Internal Medicine Erasmus MC Rotterdam The Netherlands
| | - Ype Elgersma
- Department of Neuroscience Erasmus MC Rotterdam The Netherlands
| | - Harry Steeg
- Centre for Health Protection National Institute for Public Health and the Environment (RIVM Bilthoven The Netherlands
| | - José A. Ferreira
- Department of Statistics, Informatics and Modelling National Institute for Public Health and the Environment (RIVM Bilthoven The Netherlands
| | - Jeroen L. A. Pennings
- Centre for Health Protection National Institute for Public Health and the Environment (RIVM Bilthoven The Netherlands
| | - Jan H. J. Hoeijmakers
- Princess Máxima Center for Pediatric Oncology, Genome Instability and Nutrition ONCODE Institute Utrecht The Netherlands
- Department of Molecular Genetics Erasmus MC Rotterdam The Netherlands
- CECAD Forschungszentrum Köln Germany
| | - Wilbert P. Vermeij
- Princess Máxima Center for Pediatric Oncology, Genome Instability and Nutrition ONCODE Institute Utrecht The Netherlands
| | - Martijn E. T. Dollé
- Centre for Health Protection National Institute for Public Health and the Environment (RIVM Bilthoven The Netherlands
| |
Collapse
|
53
|
Dingler FA, Wang M, Mu A, Millington CL, Oberbeck N, Watcham S, Pontel LB, Kamimae-Lanning AN, Langevin F, Nadler C, Cordell RL, Monks PS, Yu R, Wilson NK, Hira A, Yoshida K, Mori M, Okamoto Y, Okuno Y, Muramatsu H, Shiraishi Y, Kobayashi M, Moriguchi T, Osumi T, Kato M, Miyano S, Ito E, Kojima S, Yabe H, Yabe M, Matsuo K, Ogawa S, Göttgens B, Hodskinson MRG, Takata M, Patel KJ. Two Aldehyde Clearance Systems Are Essential to Prevent Lethal Formaldehyde Accumulation in Mice and Humans. Mol Cell 2020; 80:996-1012.e9. [PMID: 33147438 PMCID: PMC7758861 DOI: 10.1016/j.molcel.2020.10.012] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/20/2020] [Accepted: 10/08/2020] [Indexed: 01/04/2023]
Abstract
Reactive aldehydes arise as by-products of metabolism and are normally cleared by multiple families of enzymes. We find that mice lacking two aldehyde detoxifying enzymes, mitochondrial ALDH2 and cytoplasmic ADH5, have greatly shortened lifespans and develop leukemia. Hematopoiesis is disrupted profoundly, with a reduction of hematopoietic stem cells and common lymphoid progenitors causing a severely depleted acquired immune system. We show that formaldehyde is a common substrate of ALDH2 and ADH5 and establish methods to quantify elevated blood formaldehyde and formaldehyde-DNA adducts in tissues. Bone-marrow-derived progenitors actively engage DNA repair but also imprint a formaldehyde-driven mutation signature similar to aging-associated human cancer mutation signatures. Furthermore, we identify analogous genetic defects in children causing a previously uncharacterized inherited bone marrow failure and pre-leukemic syndrome. Endogenous formaldehyde clearance alone is therefore critical for hematopoiesis and in limiting mutagenesis in somatic tissues.
Collapse
Affiliation(s)
- Felix A Dingler
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Meng Wang
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK; Department of Haematology, University of Cambridge, Cambridge, UK
| | - Anfeng Mu
- Laboratory of DNA Damage Signaling, Department of Late Effects Studies, Radiation Biology Center, Kyoto University, Kyoto, Japan; Department of Genome Biology, Graduate School of Biostudies, Kyoto University, Kyoto, Japan; Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | | | - Nina Oberbeck
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Sam Watcham
- Department of Haematology, University of Cambridge, Cambridge, UK; Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
| | - Lucas B Pontel
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK; Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET, Polo Científico Tecnológico, Godoy Cruz 2390, C1425FQD Buenos Aires, Argentina
| | | | - Frederic Langevin
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Camille Nadler
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Rebecca L Cordell
- Department of Chemistry, University of Leicester, Leicester LE1 7RH, UK
| | - Paul S Monks
- Department of Chemistry, University of Leicester, Leicester LE1 7RH, UK
| | - Rui Yu
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Nicola K Wilson
- Department of Haematology, University of Cambridge, Cambridge, UK; Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
| | - Asuka Hira
- Laboratory of DNA Damage Signaling, Department of Late Effects Studies, Radiation Biology Center, Kyoto University, Kyoto, Japan; Department of Genome Biology, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Kenichi Yoshida
- Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Minako Mori
- Laboratory of DNA Damage Signaling, Department of Late Effects Studies, Radiation Biology Center, Kyoto University, Kyoto, Japan; Department of Genome Biology, Graduate School of Biostudies, Kyoto University, Kyoto, Japan; Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yusuke Okamoto
- Laboratory of DNA Damage Signaling, Department of Late Effects Studies, Radiation Biology Center, Kyoto University, Kyoto, Japan; Department of Genome Biology, Graduate School of Biostudies, Kyoto University, Kyoto, Japan; Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yusuke Okuno
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hideki Muramatsu
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuichi Shiraishi
- Section of Genome Analysis Platform, Center for Cancer Genomic and Advanced Therapeutics, National Cancer Center, Tokyo, Japan
| | - Masayuki Kobayashi
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Department of Hematology, Kyoto Katsura Hospital, Kyoto, Japan
| | | | - Tomoo Osumi
- Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan
| | - Motohiro Kato
- Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan
| | - Satoru Miyano
- Laboratory of DNA Information Analysis, Human Genome Center, The Institute of Medical Science, University of Tokyo, Tokyo Japan
| | - Etsuro Ito
- Department of Pediatrics, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Seiji Kojima
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiromasa Yabe
- Department of Innovative Medical Science, Tokai University School of Medicine, Isehara, Japan
| | - Miharu Yabe
- Department of Innovative Medical Science, Tokai University School of Medicine, Isehara, Japan
| | - Keitaro Matsuo
- Division of Cancer Epidemiology and Prevention, Aichi Cancer Center Research Institute, Nagoya, Japan; Division of Analytical Cancer Epidemiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Seishi Ogawa
- Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Department of Medicine, Center for Hematology and Regenerative Medicine, Karolinska Institute, Sweden; Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
| | - Berthold Göttgens
- Department of Haematology, University of Cambridge, Cambridge, UK; Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
| | | | - Minoru Takata
- Laboratory of DNA Damage Signaling, Department of Late Effects Studies, Radiation Biology Center, Kyoto University, Kyoto, Japan; Department of Genome Biology, Graduate School of Biostudies, Kyoto University, Kyoto, Japan.
| | - Ketan J Patel
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK; Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 2QQ, UK; MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, UK.
| |
Collapse
|
54
|
Chen Y, Geng A, Zhang W, Qian Z, Wan X, Jiang Y, Mao Z. Fight to the bitter end: DNA repair and aging. Ageing Res Rev 2020; 64:101154. [PMID: 32977059 DOI: 10.1016/j.arr.2020.101154] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 08/05/2020] [Accepted: 08/19/2020] [Indexed: 12/11/2022]
Abstract
DNA carries the genetic information that directs complex biological processes; thus, maintaining a stable genome is critical for individual growth and development and for human health. DNA repair is a fundamental and conserved mechanism responsible for mending damaged DNA and restoring genomic stability, while its deficiency is closely related to multiple human disorders. In recent years, remarkable progress has been made in the field of DNA repair and aging. Here, we will extensively discuss the relationship among DNA damage, DNA repair, aging and aging-associated diseases based on the latest research. In addition, the possible role of DNA repair in several potential rejuvenation strategies will be discussed. Finally, we will also review the emerging methods that may facilitate future research on DNA repair.
Collapse
|
55
|
Badawi Y, Nishimune H. Impairment Mechanisms and Intervention Approaches for Aged Human Neuromuscular Junctions. Front Mol Neurosci 2020; 13:568426. [PMID: 33328881 PMCID: PMC7717980 DOI: 10.3389/fnmol.2020.568426] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 10/16/2020] [Indexed: 12/19/2022] Open
Abstract
The neuromuscular junction (NMJ) is a chemical synapse formed between a presynaptic motor neuron and a postsynaptic muscle cell. NMJs in most vertebrate species share many essential features; however, some differences distinguish human NMJs from others. This review will describe the pre- and postsynaptic structures of human NMJs and compare them to NMJs of laboratory animals. We will focus on age-dependent declines in function and changes in the structure of human NMJs. Furthermore, we will describe insights into the aging process revealed from mouse models of accelerated aging. In addition, we will compare aging phenotypes to other human pathologies that cause impairments of pre- and postsynaptic structures at NMJs. Finally, we will discuss potential intervention approaches for attenuating age-related NMJ dysfunction and sarcopenia in humans.
Collapse
Affiliation(s)
- Yomna Badawi
- Department of Anatomy and Cell Biology, University of Kansas School of Medicine, Kansas City, KS, United States
| | - Hiroshi Nishimune
- Department of Anatomy and Cell Biology, University of Kansas School of Medicine, Kansas City, KS, United States.,Neurobiology of Aging, Tokyo Metropolitan Institute of Gerontology, Itabashi, Japan
| |
Collapse
|
56
|
Do JP, Defensor EB, Ichim CV, Lim MA, Mechanic JA, Rabe MD, Schaevitz LR. Automated and Continuous Monitoring of Animal Welfare through Digital Alerting. Comp Med 2020; 70:313-327. [PMID: 32414427 PMCID: PMC7446638 DOI: 10.30802/aalas-cm-19-000090] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
A primary goal in preclinical animal research is respectful and responsible care aimed toward minimizing stress and discomfort while enhancing collection of accurate and reproducible scientific data. Researchers use hands-on clinical observations and measurements as part of routine husbandry procedures or study protocols to monitor animal welfare. Although frequent assessments ensure the timely identification of animals with declining health, increased handling can result in additional stress on the animal and increased study variability. We investigated whether automated alerting regarding changes in behavior and physiology can complement existing welfare assessments to improve the identification of animals in pain or distress. Using historical data collected from a diverse range of therapeutic models, we developed algorithms that detect changes in motion and breathing rate frequently associated with sick animals but rare in healthy controls. To avoid introducing selec- tion bias, we evaluated the performance of these algorithms by using retrospective analysis of all studies occurring over a 31-d period in our vivarium. Analyses revealed that the majority of the automated alerts occurred prior to or simultaneously with technicians' observations of declining health in animals. Additional analyses performed across the entire duration of 2 studies (animal models of rapid aging and lung metastasis) demonstrated the sensitivity, accuracy, and utility of automated alerting for detecting unhealthy subjects and those eligible for humane endpoints. The percentage of alerts per total subject days ranged between 0% and 24%, depending on the animal model. Automated alerting effectively complements standard clinical observations to enhance animal welfare and promote responsible scientific advancement.
Collapse
|
57
|
Local endothelial DNA repair deficiency causes aging-resembling endothelial-specific dysfunction. Clin Sci (Lond) 2020; 134:727-746. [PMID: 32202295 DOI: 10.1042/cs20190124] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 01/21/2020] [Accepted: 03/23/2020] [Indexed: 02/07/2023]
Abstract
We previously identified genomic instability as a causative factor for vascular aging. In the present study, we determined which vascular aging outcomes are due to local endothelial DNA damage, which was accomplished by genetic removal of ERCC1 (excision repair cross-complementation group 1) DNA repair in mice (EC-knockout (EC-KO) mice). EC-KO showed a progressive decrease in microvascular dilation of the skin, increased microvascular leakage in the kidney, decreased lung perfusion, and increased aortic stiffness compared with wild-type (WT). EC-KO showed expression of DNA damage and potential senescence marker p21 exclusively in the endothelium, as demonstrated in aorta. Also the kidney showed p21-positive cells. Vasodilator responses measured in organ baths were decreased in aorta, iliac and coronary artery EC-KO compared with WT, of which coronary artery was the earliest to be affected. Nitric oxide-mediated endothelium-dependent vasodilation was abolished in aorta and coronary artery, whereas endothelium-derived hyperpolarization and responses to exogenous nitric oxide (NO) were intact. EC-KO showed increased superoxide production compared with WT, as measured in lung tissue, rich in endothelial cells (ECs). Arterial systolic blood pressure (BP) was increased at 3 months, but normal at 5 months, at which age cardiac output (CO) was decreased. Since no further signs of cardiac dysfunction were detected, this decrease might be an adaptation to prevent an increase in BP. In summary, a selective DNA repair defect in the endothelium produces features of age-related endothelial dysfunction, largely attributed to loss of endothelium-derived NO. Increased superoxide generation might contribute to the observed changes affecting end organ perfusion, as demonstrated in kidney and lung.
Collapse
|
58
|
Golshiri K, Ataei Ataabadi E, Brandt R, van der Pluijm I, de Vries R, Danser AHJ, Roks A. Chronic Sildenafil Treatment Improves Vasomotor Function in a Mouse Model of Accelerated Aging. Int J Mol Sci 2020; 21:ijms21134667. [PMID: 32630010 PMCID: PMC7369923 DOI: 10.3390/ijms21134667] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/17/2020] [Accepted: 06/22/2020] [Indexed: 12/26/2022] Open
Abstract
Aging leads to a loss of vasomotor control. Both vasodilation and vasoconstriction are affected. Decreased nitric oxide–cGMP-mediated relaxation is a hallmark of aging. It contributes to vascular disease, notably hypertension, infarction, and dementia. Decreased vasodilation can be caused by aging independently from cardiovascular risk factors. This process that can be mimicked in mice in an accelerated way by activation of the DNA damage response. Genetic deletion of the DNA repair enzyme ERCC1 endonuclease in mice, as in the case of Ercc1Δ/- mice, can be used as a tool to accelerate aging. Ercc1Δ/- mice develop age-dependent vasomotor dysfunction from two months after birth. In the present study we tested if chronic treatment with sildenafil, a phosphodiesterase 5 inhibitor that augments NO–cGMP signaling, can reduce the development of vasomotor dysfunction in Ercc1Δ/- mice. Ercc1Δ/- mice and wild-type littermates were treated with 10 mg/kg/d of sildenafil from the age of 6 to the age of 14 weeks. Blood pressure and in vivo and ex vivo vasomotor responses were measured at the end of the treatment period. Ercc1Δ/- mice developed decreased reactive hyperemia, and diminished NO–cGMP-dependent acetylcholine responses. The diminished acetylcholine response involved both endothelial and vascular smooth muscle cell signaling. Chronic sildenafil exclusively improved NO–cGMP signaling in VSMC, and had no effect on endothelium-derived hyperpolarization. Sildenafil also improved KCl hypocontractility in Ercc1Δ/- mice. All effects were blood pressure-independent. The findings might be of clinical importance for prevention of morbidities related to vascular aging as well as for progeria patients with a high risk of cardiovascular disease.
Collapse
Affiliation(s)
- Keivan Golshiri
- Department of Internal Medicine, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands; (K.G.); (E.A.A.); (R.d.V.); (A.H.J.D.)
| | - Ehsan Ataei Ataabadi
- Department of Internal Medicine, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands; (K.G.); (E.A.A.); (R.d.V.); (A.H.J.D.)
| | - Renata Brandt
- Department of Molecular Genetics, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands; (R.B.); (I.v.d.P.)
| | - Ingrid van der Pluijm
- Department of Molecular Genetics, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands; (R.B.); (I.v.d.P.)
- Department of Vascular Surgery, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands
| | - René de Vries
- Department of Internal Medicine, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands; (K.G.); (E.A.A.); (R.d.V.); (A.H.J.D.)
| | - A. H. Jan Danser
- Department of Internal Medicine, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands; (K.G.); (E.A.A.); (R.d.V.); (A.H.J.D.)
| | - Anton Roks
- Department of Internal Medicine, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands; (K.G.); (E.A.A.); (R.d.V.); (A.H.J.D.)
- Correspondence:
| |
Collapse
|
59
|
Mitochondrial phosphatase PGAM5 modulates cellular senescence by regulating mitochondrial dynamics. Nat Commun 2020; 11:2549. [PMID: 32439975 PMCID: PMC7242393 DOI: 10.1038/s41467-020-16312-7] [Citation(s) in RCA: 155] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 04/27/2020] [Indexed: 12/15/2022] Open
Abstract
Mitochondria undergo dynamic fusion/fission, biogenesis and mitophagy in response to stimuli or stresses. Disruption of mitochondrial homeostasis could lead to cell senescence, although the underlying mechanism remains unclear. We show that deletion of mitochondrial phosphatase PGAM5 leads to accelerated retinal pigment epithelial (RPE) senescence in vitro and in vivo. Mechanistically, PGAM5 is required for mitochondrial fission through dephosphorylating DRP1. PGAM5 deletion leads to increased mitochondrial fusion and decreased mitochondrial turnover. As results, cellular ATP and reactive oxygen species (ROS) levels are elevated, mTOR and IRF/IFN-β signaling pathways are enhanced, leading to cellular senescence. Overexpression of Drp1 K38A or S637A mutant phenocopies or rescues mTOR activation and senescence in PGAM5-/- cells, respectively. Young but not aging Pgam5-/- mice are resistant to sodium iodate-induced RPE cell death. Our studies establish a link between defective mitochondrial fission, cellular senescence and age-dependent oxidative stress response, which have implications in age-related diseases.
Collapse
|
60
|
Vessoni AT, Guerra CCC, Kajitani GS, Nascimento LLS, Garcia CCM. Cockayne Syndrome: The many challenges and approaches to understand a multifaceted disease. Genet Mol Biol 2020; 43:e20190085. [PMID: 32453336 PMCID: PMC7250278 DOI: 10.1590/1678-4685-gmb-2019-0085] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 01/15/2020] [Indexed: 01/04/2023] Open
Abstract
The striking and complex phenotype of Cockayne syndrome (CS) patients combines progeria-like features with developmental deficits. Since the establishment of the in vitro culture of skin fibroblasts derived from patients with CS in the 1970s, significant progress has been made in the understanding of the genetic alterations associated with the disease and their impact on molecular, cellular, and organismal functions. In this review, we provide a historic perspective on the research into CS by revisiting seminal papers in this field. We highlighted the great contributions of several researchers in the last decades, ranging from the cloning and characterization of CS genes to the molecular dissection of their roles in DNA repair, transcription, redox processes and metabolism control. We also provide a detailed description of all pathological mutations in genes ERCC6 and ERCC8 reported to date and their impact on CS-related proteins. Finally, we review the contributions (and limitations) of many genetic animal models to the study of CS and how cutting-edge technologies, such as cell reprogramming and state-of-the-art genome editing, are helping us to address unanswered questions.
Collapse
Affiliation(s)
| | - Camila Chaves Coelho Guerra
- Universidade Federal de Ouro Preto, Instituto de Ciências Exatas e
Biológicas, Núcleo de Pesquisa em Ciências Biológicas & Departamento de Ciências
Biológicas, Ouro Preto, MG, Brazil
| | - Gustavo Satoru Kajitani
- Universidade Federal de Ouro Preto, Instituto de Ciências Exatas e
Biológicas, Núcleo de Pesquisa em Ciências Biológicas & Departamento de Ciências
Biológicas, Ouro Preto, MG, Brazil
- Universidade de São Paulo, Instituto de Ciências Biomédicas,
Departamento de Microbiologia, São Paulo,SP, Brazil
| | - Livia Luz Souza Nascimento
- Universidade de São Paulo, Instituto de Ciências Biomédicas,
Departamento de Microbiologia, São Paulo,SP, Brazil
| | - Camila Carrião Machado Garcia
- Universidade Federal de Ouro Preto, Instituto de Ciências Exatas e
Biológicas, Núcleo de Pesquisa em Ciências Biológicas & Departamento de Ciências
Biológicas, Ouro Preto, MG, Brazil
| |
Collapse
|
61
|
De Majo F, Hegenbarth JC, Rühle F, Bär C, Thum T, de Boer M, Duncker DJ, Schroen B, Armand AS, Stoll M, De Windt LJ. Dichotomy between the transcriptomic landscape of naturally versus accelerated aged murine hearts. Sci Rep 2020; 10:8136. [PMID: 32424227 PMCID: PMC7235007 DOI: 10.1038/s41598-020-65115-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 04/29/2020] [Indexed: 11/12/2022] Open
Abstract
We investigated the transcriptomic landscape of the murine myocardium along the course of natural aging and in three distinct mouse models of premature aging with established aging-related cardiac dysfunction. Genome-wide total RNA-seq was performed and the expression patterns of protein-coding genes and non-coding RNAs were compared between hearts from naturally aging mice, mice with cardiac-specific deficiency of a component of the DNA repair machinery, mice with reduced mitochondrial antioxidant capacity and mice with reduced telomere length. Our results demonstrate that no dramatic changes are evident in the transcriptomes of naturally senescent murine hearts until two years of age, in contrast to the transcriptome of accelerated aged mice. Additionally, these mice displayed model-specific alterations of the expression levels of protein-coding and non-coding genes with hardly any overlap with age-related signatures. Our data demonstrate very limited similarities between the transcriptomes of all our murine aging models and question their reliability to study human cardiovascular senescence.
Collapse
Affiliation(s)
- Federica De Majo
- Department of Molecular Genetics, Faculty of Science and Engineering; Maastricht University, Maastricht, The Netherlands.,CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences; Maastricht University, Maastricht, The Netherlands
| | - Jana-Charlotte Hegenbarth
- Department of Molecular Genetics, Faculty of Science and Engineering; Maastricht University, Maastricht, The Netherlands.,CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences; Maastricht University, Maastricht, The Netherlands
| | - Frank Rühle
- Bioinformatics Core Facility, Institute of Molecular Biology (IMB), Mainz, Germany.,Department of Genetic Epidemiology, Institute of Human Genetics, University Hospital Münster, Münster, Germany
| | - Christian Bär
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany.,REBIRTH Excellence Cluster, Hannover Medical School, Hannover, Germany
| | - Martine de Boer
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Dirk J Duncker
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Blanche Schroen
- CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences; Maastricht University, Maastricht, The Netherlands
| | - Anne-Sophie Armand
- Institut Necker Enfants Malades, Inserm U1151, Paris, France; Universite Paris Descartes, Sorbonne Paris Cite, Paris, France
| | - Monika Stoll
- Department of Genetic Epidemiology, Institute of Human Genetics, University Hospital Münster, Münster, Germany.,Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Leon J De Windt
- Department of Molecular Genetics, Faculty of Science and Engineering; Maastricht University, Maastricht, The Netherlands. .,CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences; Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|
62
|
Mulderrig L, Garaycoechea JI. XPF-ERCC1 protects liver, kidney and blood homeostasis outside the canonical excision repair pathways. PLoS Genet 2020; 16:e1008555. [PMID: 32271760 PMCID: PMC7144963 DOI: 10.1371/journal.pgen.1008555] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 12/05/2019] [Indexed: 01/02/2023] Open
Abstract
Loss of the XPF-ERCC1 endonuclease causes a dramatic phenotype that results in progeroid features associated with liver, kidney and bone marrow dysfunction. As this nuclease is involved in multiple DNA repair transactions, it is plausible that this severe phenotype results from the simultaneous inactivation of both branches of nucleotide excision repair (GG- and TC-NER) and Fanconi anaemia (FA) inter-strand crosslink (ICL) repair. Here we use genetics in human cells and mice to investigate the interaction between the canonical NER and ICL repair pathways and, subsequently, how their joint inactivation phenotypically overlaps with XPF-ERCC1 deficiency. We find that cells lacking TC-NER are sensitive to crosslinking agents and that there is a genetic interaction between NER and FA in the repair of certain endogenous crosslinking agents. However, joint inactivation of GG-NER, TC-NER and FA crosslink repair cannot account for the hypersensitivity of XPF-deficient cells to classical crosslinking agents nor is it sufficient to explain the extreme phenotype of Ercc1-/- mice. These analyses indicate that XPF-ERCC1 has important functions outside of its central role in NER and FA crosslink repair which are required to prevent endogenous DNA damage. Failure to resolve such damage leads to loss of tissue homeostasis in mice and humans.
Collapse
Affiliation(s)
- Lee Mulderrig
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge, United Kingdom
| | - Juan I. Garaycoechea
- Hubrecht Institute–KNAW, University Medical Center Utrecht, Uppsalalaan, CT Utrecht, Netherlands
| |
Collapse
|
63
|
Kim DE, Dollé MET, Vermeij WP, Gyenis A, Vogel K, Hoeijmakers JHJ, Wiley CD, Davalos AR, Hasty P, Desprez P, Campisi J. Deficiency in the DNA repair protein ERCC1 triggers a link between senescence and apoptosis in human fibroblasts and mouse skin. Aging Cell 2020; 19:e13072. [PMID: 31737985 PMCID: PMC7059167 DOI: 10.1111/acel.13072] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 10/07/2019] [Accepted: 10/30/2019] [Indexed: 12/11/2022] Open
Abstract
ERCC1 (excision repair cross complementing‐group 1) is a mammalian endonuclease that incises the damaged strand of DNA during nucleotide excision repair and interstrand cross‐link repair. Ercc1−/Δ mice, carrying one null and one hypomorphic Ercc1 allele, have been widely used to study aging due to accelerated aging phenotypes in numerous organs and their shortened lifespan. Ercc1−/Δ mice display combined features of human progeroid and cancer‐prone syndromes. Although several studies report cellular senescence and apoptosis associated with the premature aging of Ercc1−/Δ mice, the link between these two processes and their physiological relevance in the phenotypes of Ercc1−/Δ mice are incompletely understood. Here, we show that ERCC1 depletion, both in cultured human fibroblasts and the skin of Ercc1−/Δ mice, initially induces cellular senescence and, importantly, increased expression of several SASP (senescence‐associated secretory phenotype) factors. Cellular senescence induced by ERCC1 deficiency was dependent on activity of the p53 tumor‐suppressor protein. In turn, TNFα secreted by senescent cells induced apoptosis, not only in neighboring ERCC1‐deficient nonsenescent cells, but also cell autonomously in the senescent cells themselves. In addition, expression of the stem cell markers p63 and Lgr6 was significantly decreased in Ercc1−/Δ mouse skin, where the apoptotic cells are localized, compared to age‐matched wild‐type skin, possibly due to the apoptosis of stem cells. These data suggest that ERCC1‐depleted cells become susceptible to apoptosis via TNFα secreted from neighboring senescent cells. We speculate that parts of the premature aging phenotypes and shortened health‐ or lifespan may be due to stem cell depletion through apoptosis promoted by senescent cells.
Collapse
Affiliation(s)
- Dong Eun Kim
- Buck Institute for Research on Aging Novato CA USA
| | - Martijn E. T. Dollé
- Centre for Health Protection Research National Institute of Public Health and the Environment (RIVM) Bilthoven The Netherlands
| | - Wilbert P. Vermeij
- Department of Molecular Genetics Erasmus University Medical Center Rotterdam The Netherlands
- Princess Máxima Center for Pediatric Oncology ONCODE Institute Utrecht The Netherlands
| | | | | | - Jan H. J. Hoeijmakers
- Department of Molecular Genetics Erasmus University Medical Center Rotterdam The Netherlands
- Princess Máxima Center for Pediatric Oncology ONCODE Institute Utrecht The Netherlands
- CECAD Forschungszentrum Köln Germany
| | | | | | - Paul Hasty
- Department of Molecular Medicine Sam and Ann Barshop Institute for Longevity and Aging Studies University of Texas Health Science Center San Antonio TX USA
| | | | - Judith Campisi
- Buck Institute for Research on Aging Novato CA USA
- Lawrence Berkeley National Laboratory Berkeley CA USA
| |
Collapse
|
64
|
Cougnoux A, Drummond RA, Fellmeth M, Navid F, Collar AL, Iben J, Kulkarni AB, Pickel J, Schiffmann R, Wassif CA, Cawley NX, Lionakis MS, Porter FD. Unique molecular signature in mucolipidosis type IV microglia. J Neuroinflammation 2019; 16:276. [PMID: 31883529 PMCID: PMC6935239 DOI: 10.1186/s12974-019-1672-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 12/09/2019] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Lysosomal storage diseases (LSD) are a large family of inherited disorders characterized by abnormal endolysosomal accumulation of cellular material due to catabolic enzyme and transporter deficiencies. Depending on the affected metabolic pathway, LSD manifest with somatic or central nervous system (CNS) signs and symptoms. Neuroinflammation is a hallmark feature of LSD with CNS involvement such as mucolipidosis type IV, but not of others like Fabry disease. METHODS We investigated the properties of microglia from LSD with and without major CNS involvement in 2-month-old mucolipidosis type IV (Mcoln1-/-) and Fabry disease (Glay/-) mice, respectively, by using a combination of flow cytometric, RNA sequencing, biochemical, in vitro and immunofluorescence analyses. RESULTS We characterized microglia activation and transcriptome from mucolipidosis type IV and Fabry disease mice to determine if impaired lysosomal function is sufficient to prime these brain-resident immune cells. Consistent with the neurological pathology observed in mucolipidosis type IV, Mcoln1-/- microglia demonstrated an activation profile with a mixed neuroprotective/neurotoxic expression pattern similar to the one we previously observed in Niemann-Pick disease, type C1, another LSD with significant CNS involvement. In contrast, the Fabry disease microglia transcriptome revealed minimal alterations, consistent with the relative lack of CNS symptoms in this disease. The changes observed in Mcoln1-/- microglia showed significant overlap with alterations previously reported for other common neuroinflammatory disorders including Alzheimer's, Parkinson's, and Huntington's diseases. Indeed, our comparison of microglia transcriptomes from Alzheimer's disease, amyotrophic lateral sclerosis, Niemann-Pick disease, type C1 and mucolipidosis type IV mouse models showed an enrichment in "disease-associated microglia" pattern among these diseases. CONCLUSIONS The similarities in microglial transcriptomes and features of neuroinflammation and microglial activation in rare monogenic disorders where the primary metabolic disturbance is known may provide novel insights into the immunopathogenesis of other more common neuroinflammatory disorders. TRIAL REGISTRATION ClinicalTrials.gov, NCT01067742, registered on February 12, 2010.
Collapse
Affiliation(s)
- Antony Cougnoux
- Division of Translational Medicine, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, DHHS, 10CRC, Rm 5-2571, 10 Center Dr, Bethesda, MD, 20892, USA
| | - Rebecca A Drummond
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Mason Fellmeth
- Division of Translational Medicine, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, DHHS, 10CRC, Rm 5-2571, 10 Center Dr, Bethesda, MD, 20892, USA
| | - Fatemeh Navid
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, MD, 20892, USA
| | - Amanda L Collar
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, 20892, USA
| | - James Iben
- Molecular Genomics Core, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20879, USA
| | - Ashok B Kulkarni
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20879, USA
| | - James Pickel
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20879, USA
| | | | - Christopher A Wassif
- Division of Translational Medicine, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, DHHS, 10CRC, Rm 5-2571, 10 Center Dr, Bethesda, MD, 20892, USA
| | - Niamh X Cawley
- Division of Translational Medicine, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, DHHS, 10CRC, Rm 5-2571, 10 Center Dr, Bethesda, MD, 20892, USA
| | - Michail S Lionakis
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Forbes D Porter
- Division of Translational Medicine, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, DHHS, 10CRC, Rm 5-2571, 10 Center Dr, Bethesda, MD, 20892, USA.
| |
Collapse
|
65
|
Milanese C, Bombardieri CR, Sepe S, Barnhoorn S, Payán-Goméz C, Caruso D, Audano M, Pedretti S, Vermeij WP, Brandt RMC, Gyenis A, Wamelink MM, de Wit AS, Janssens RC, Leen R, van Kuilenburg ABP, Mitro N, Hoeijmakers JHJ, Mastroberardino PG. DNA damage and transcription stress cause ATP-mediated redesign of metabolism and potentiation of anti-oxidant buffering. Nat Commun 2019; 10:4887. [PMID: 31653834 PMCID: PMC6814737 DOI: 10.1038/s41467-019-12640-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 09/22/2019] [Indexed: 12/13/2022] Open
Abstract
Accumulation of DNA lesions causing transcription stress is associated with natural and accelerated aging and culminates with profound metabolic alterations. Our understanding of the mechanisms governing metabolic redesign upon genomic instability, however, is highly rudimentary. Using Ercc1-defective mice and Xpg knock-out mice, we demonstrate that combined defects in transcription-coupled DNA repair (TCR) and in nucleotide excision repair (NER) directly affect bioenergetics due to declined transcription, leading to increased ATP levels. This in turn inhibits glycolysis allosterically and favors glucose rerouting through the pentose phosphate shunt, eventually enhancing production of NADPH-reducing equivalents. In NER/TCR-defective mutants, augmented NADPH is not counterbalanced by increased production of pro-oxidants and thus pentose phosphate potentiation culminates in an over-reduced redox state. Skin fibroblasts from the TCR disease Cockayne syndrome confirm results in animal models. Overall, these findings unravel a mechanism connecting DNA damage and transcriptional stress to metabolic redesign and protective antioxidant defenses. ERCC1 is involved in a number of DNA repair pathways including nucleotide excision repair. Here the authors showed that reduced transcription in Ercc1-deficient mouse livers and cells increases ATP levels, suppressing glycolysis and rerouting glucose into the pentose phosphate shunt that generates reductive stress.
Collapse
Affiliation(s)
- Chiara Milanese
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Cíntia R Bombardieri
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Sara Sepe
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Sander Barnhoorn
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - César Payán-Goméz
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands.,Facultad de Ciencias Naturales y Matemáticas, Universidad del Rosario, Bogotá, Colombia
| | - Donatella Caruso
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Matteo Audano
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Silvia Pedretti
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Wilbert P Vermeij
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Renata M C Brandt
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Akos Gyenis
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands.,Cologne Excellence Cluster for Cellular Stress Responses in Ageing-Associated Diseases (CECAD) and Systems Biology of Ageing Cologne, University of Cologne, Cologne, Germany
| | - Mirjam M Wamelink
- Department of Clinical Chemistry, VU University Medical Center, Amsterdam, the Netherlands
| | - Annelieke S de Wit
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Roel C Janssens
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - René Leen
- Laboratory of Genetic Metabolic Diseases, Academic Medical Center, Amsterdam, the Netherlands
| | | | - Nico Mitro
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Jan H J Hoeijmakers
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.,Cologne Excellence Cluster for Cellular Stress Responses in Ageing-Associated Diseases (CECAD) and Systems Biology of Ageing Cologne, University of Cologne, Cologne, Germany.,Oncode Institute, Princess Máxima Center, Utrecht, Netherlands
| | - Pier G Mastroberardino
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands. .,Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy.
| |
Collapse
|
66
|
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
|
67
|
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
|
68
|
Kawara H, Akahori R, Wakasugi M, Sancar A, Matsunaga T. DCAF7 is required for maintaining the cellular levels of ERCC1-XPF and nucleotide excision repair. Biochem Biophys Res Commun 2019; 519:204-210. [PMID: 31493872 DOI: 10.1016/j.bbrc.2019.08.147] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 08/26/2019] [Indexed: 02/03/2023]
Abstract
The ERCC1-XPF heterodimer is a structure-specific endonuclease and plays multiple roles in various DNA repair pathways including nucleotide excision repair and also telomere maintenance. The dimer formation, which is mediated by their C-terminal helix-hairpin-helix regions, is essential for their endonuclease activity as well as the stability of each protein. However, the detailed mechanism of how a cellular level of ERCC1-XPF is regulated still remains elusive. Here, we report the identification of DDB1- and CUL4-associated factor 7 (DCAF7, also known as WDR68/HAN11) as a novel interacting protein of ERCC1-XPF by mass spectrometry after tandem purification. Immunoprecipitation experiments confirmed their interaction and suggested dominant association of DCAF7 with XPF but not ERCC1. Interestingly, siRNA-mediated knockdown of DCAF7, but not DDB1, attenuated the cellular level of ERCC1-XPF, which is partly dependent on proteasome. The depletion of TCP1α, one of components of the molecular chaperon TRiC/CCT known to interact with DCAF7 and promote its folding, also reduced ERCC1-XPF level. Finally, we show that the depletion of DCAF7 causes inefficient repair of UV-induced (6-4) photoproducts, which can be rescued by ectopic overexpression of XPF or ERCC1-XPF. Altogether, our results strongly suggest that DCAF7 is a novel regulator of ERCC1-XPF protein level and cellular nucleotide excision repair activity.
Collapse
Affiliation(s)
- Hiroaki Kawara
- Laboratory of Human Molecular Genetics, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan; Department of Biochemistry and Biophysics, School of Medicine, University of North Carolina at Chapel Hill, NC, 27599, USA
| | - Ryo Akahori
- Laboratory of Human Molecular Genetics, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Mitsuo Wakasugi
- Laboratory of Human Molecular Genetics, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Aziz Sancar
- Department of Biochemistry and Biophysics, School of Medicine, University of North Carolina at Chapel Hill, NC, 27599, USA
| | - Tsukasa Matsunaga
- Laboratory of Human Molecular Genetics, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan.
| |
Collapse
|
69
|
Yousefzadeh MJ, Melos KI, Angelini L, Burd CE, Robbins PD, Niedernhofer LJ. Mouse Models of Accelerated Cellular Senescence. Methods Mol Biol 2019; 1896:203-230. [PMID: 30474850 DOI: 10.1007/978-1-4939-8931-7_17] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Senescent cells accumulate in multiple tissues as virtually all vertebrate organisms age. Senescence is a highly conserved response to many forms of cellular stress intended to block the propagation of damaged cells. Senescent cells have been demonstrated to play a causal role in aging via their senescence-associated secretory phenotype and by impeding tissue regeneration. Depletion of senescent cells either through genetic or pharmacologic methods has been demonstrated to extend murine lifespan and delay the onset of age-related diseases. Measuring the burden and location of senescent cells in vivo remains challenging, as there is no marker unique to senescent cells. Here, we describe multiple methods to detect the presence and extent of cellular senescence in preclinical models, with a special emphasis on murine models of accelerated aging that exhibit a more rapid onset of cellular senescence.
Collapse
Affiliation(s)
- Matthew J Yousefzadeh
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, USA
| | - Kendra I Melos
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, USA
| | - Luise Angelini
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, USA
| | - Christin E Burd
- Department of Molecular Genetics, The Ohio State University, Columbus, OH, USA
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, USA
| | - Paul D Robbins
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, USA
| | - Laura J Niedernhofer
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
70
|
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
|
71
|
Jager M, Blokzijl F, Kuijk E, Bertl J, Vougioukalaki M, Janssen R, Besselink N, Boymans S, de Ligt J, Pedersen JS, Hoeijmakers J, Pothof J, van Boxtel R, Cuppen E. Deficiency of nucleotide excision repair is associated with mutational signature observed in cancer. Genome Res 2019; 29:1067-1077. [PMID: 31221724 PMCID: PMC6633256 DOI: 10.1101/gr.246223.118] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 06/07/2019] [Indexed: 12/24/2022]
Abstract
Nucleotide excision repair (NER) is one of the main DNA repair pathways that protect cells against genomic damage. Disruption of this pathway can contribute to the development of cancer and accelerate aging. Mutational characteristics of NER-deficiency may reveal important diagnostic opportunities, as tumors deficient in NER are more sensitive to certain treatments. Here, we analyzed the genome-wide somatic mutational profiles of adult stem cells (ASCs) from NER-deficient Ercc1 -/Δ mice. Our results indicate that NER-deficiency increases the base substitution load twofold in liver but not in small intestinal ASCs, which coincides with the tissue-specific aging pathology observed in these mice. Moreover, NER-deficient ASCs of both tissues show an increased contribution of Signature 8 mutations, which is a mutational pattern with unknown etiology that is recurrently observed in various cancer types. The scattered genomic distribution of the base substitutions indicates that deficiency of global-genome NER (GG-NER) underlies the observed mutational consequences. In line with this, we observe increased Signature 8 mutations in a GG-NER-deficient human organoid culture, in which XPC was deleted using CRISPR-Cas9 gene-editing. Furthermore, genomes of NER-deficient breast tumors show an increased contribution of Signature 8 mutations compared with NER-proficient tumors. Elevated levels of Signature 8 mutations could therefore contribute to a predictor of NER-deficiency based on a patient's mutational profile.
Collapse
Affiliation(s)
- Myrthe Jager
- Center for Molecular Medicine and Oncode Institute, University Medical Center Utrecht, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Francis Blokzijl
- Center for Molecular Medicine and Oncode Institute, University Medical Center Utrecht, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Ewart Kuijk
- Center for Molecular Medicine and Oncode Institute, University Medical Center Utrecht, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Johanna Bertl
- Department of Molecular Medicine, Aarhus University, 8200 Aarhus N, Denmark
| | | | - Roel Janssen
- Center for Molecular Medicine and Oncode Institute, University Medical Center Utrecht, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Nicolle Besselink
- Center for Molecular Medicine and Oncode Institute, University Medical Center Utrecht, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Sander Boymans
- Center for Molecular Medicine and Oncode Institute, University Medical Center Utrecht, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Joep de Ligt
- Center for Molecular Medicine and Oncode Institute, University Medical Center Utrecht, Utrecht University, 3584 CG Utrecht, The Netherlands
| | | | | | - Joris Pothof
- Erasmus Medical Center, 3015 CN Rotterdam, The Netherlands
| | - Ruben van Boxtel
- Center for Molecular Medicine and Oncode Institute, University Medical Center Utrecht, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Edwin Cuppen
- Center for Molecular Medicine and Oncode Institute, University Medical Center Utrecht, Utrecht University, 3584 CG Utrecht, The Netherlands
| |
Collapse
|
72
|
Alyodawi K, Vermeij WP, Omairi S, Kretz O, Hopkinson M, Solagna F, Joch B, Brandt RMC, Barnhoorn S, van Vliet N, Ridwan Y, Essers J, Mitchell R, Morash T, Pasternack A, Ritvos O, Matsakas A, Collins-Hooper H, Huber TB, Hoeijmakers JHJ, Patel K. Compression of morbidity in a progeroid mouse model through the attenuation of myostatin/activin signalling. J Cachexia Sarcopenia Muscle 2019; 10:662-686. [PMID: 30916493 PMCID: PMC6596402 DOI: 10.1002/jcsm.12404] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 12/17/2018] [Accepted: 01/09/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND One of the principles underpinning our understanding of ageing is that DNA damage induces a stress response that shifts cellular resources from growth towards maintenance. A contrasting and seemingly irreconcilable view is that prompting growth of, for example, skeletal muscle confers systemic benefit. METHODS To investigate the robustness of these axioms, we induced muscle growth in a murine progeroid model through the use of activin receptor IIB ligand trap that dampens myostatin/activin signalling. Progeric mice were then investigated for neurological and muscle function as well as cellular profiling of the muscle, kidney, liver, and bone. RESULTS We show that muscle of Ercc1Δ/- progeroid mice undergoes severe wasting (decreases in hind limb muscle mass of 40-60% compared with normal mass), which is largely protected by attenuating myostatin/activin signalling using soluble activin receptor type IIB (sActRIIB) (increase of 30-62% compared with untreated progeric). sActRIIB-treated progeroid mice maintained muscle activity (distance travel per hour: 5.6 m in untreated mice vs. 13.7 m in treated) and increased specific force (19.3 mN/mg in untreated vs. 24.0 mN/mg in treated). sActRIIb treatment of progeroid mice also improved satellite cell function especially their ability to proliferate on their native substrate (2.5 cells per fibre in untreated progeroids vs. 5.4 in sActRIIB-treated progeroids after 72 h in culture). Besides direct protective effects on muscle, we show systemic improvements to other organs including the structure and function of the kidneys; there was a major decrease in the protein content in urine (albumin/creatinine of 4.9 sActRIIB treated vs. 15.7 in untreated), which is likely to be a result in the normalization of podocyte foot processes, which constitute the filtration apparatus (glomerular basement membrane thickness reduced from 224 to 177 nm following sActRIIB treatment). Treatment of the progeric mice with the activin ligand trap protected against the development of liver abnormalities including polyploidy (18.3% untreated vs. 8.1% treated) and osteoporosis (trabecular bone volume; 0.30 mm3 in treated progeroid mice vs. 0.14 mm3 in untreated mice, cortical bone volume; 0.30 mm3 in treated progeroid mice vs. 0.22 mm3 in untreated mice). The onset of neurological abnormalities was delayed (by ~5 weeks) and their severity reduced, overall sustaining health without affecting lifespan. CONCLUSIONS This study questions the notion that tissue growth and maintaining tissue function during ageing are incompatible mechanisms. It highlights the need for future investigations to assess the potential of therapies based on myostatin/activin blockade to compress morbidity and promote healthy ageing.
Collapse
Affiliation(s)
- Khalid Alyodawi
- School of Biological Sciences, University of Reading, Reading, UK.,College of Medicine, Wasit University, Kut, Iraq
| | - Wilbert P Vermeij
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands.,Princess Máxima Center, Oncode Institute, Utrecht, The Netherlands
| | - Saleh Omairi
- School of Biological Sciences, University of Reading, Reading, UK.,College of Medicine, Wasit University, Kut, Iraq
| | - Oliver Kretz
- Medizinische Klinik, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany.,Department of Medicine IV, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Department of Neuroanatomy, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | | - Francesca Solagna
- Department of Medicine IV, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Barbara Joch
- Department of Neuroanatomy, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Renata M C Brandt
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Sander Barnhoorn
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Nicole van Vliet
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Yanto Ridwan
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands.,Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Jeroen Essers
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands.,Department of Radiation Oncology, Erasmus MC, Rotterdam, The Netherlands.,Department of Vascular Surgery, Erasmus MC, Rotterdam, The Netherlands
| | - Robert Mitchell
- School of Biological Sciences, University of Reading, Reading, UK
| | - Taryn Morash
- School of Biological Sciences, University of Reading, Reading, UK
| | - Arja Pasternack
- Department of Bacteriology and Immunology, University of Helsinki, Helsinki, Finland
| | - Olli Ritvos
- Department of Bacteriology and Immunology, University of Helsinki, Helsinki, Finland.,Institute of Molecular Medicine, University of Health Science Center, Houston, TX, USA
| | | | | | - Tobias B Huber
- Medizinische Klinik, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany.,Department of Medicine IV, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,BIOSS Center for Biological Signalling Studies, University of Freiburg, Freiburg, Germany.,Freiburg Institute for Advanced Studies and Center for Biological System Analysis, Freiburg, Germany
| | - Jan H J Hoeijmakers
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands.,Princess Máxima Center, Oncode Institute, Utrecht, The Netherlands.,CECAD Forschungszentrum, Universität zu Köln, Cologne, Germany
| | - Ketan Patel
- School of Biological Sciences, University of Reading, Reading, UK.,Freiburg Institute for Advanced Studies and Center for Biological System Analysis, Freiburg, Germany
| |
Collapse
|
73
|
Functional Comparison of XPF Missense Mutations Associated to Multiple DNA Repair Disorders. Genes (Basel) 2019; 10:genes10010060. [PMID: 30658521 PMCID: PMC6357085 DOI: 10.3390/genes10010060] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 01/11/2019] [Accepted: 01/11/2019] [Indexed: 11/23/2022] Open
Abstract
XPF endonuclease is one of the most important DNA repair proteins. Encoded by XPF/ERCC4, XPF provides the enzymatic activity of XPF-ERCC1 heterodimer, an endonuclease that incises at the 5’ side of various DNA lesions. XPF is essential for nucleotide excision repair (NER) and interstrand crosslink repair (ICLR). XPF/ERCC4 mutations are associated with several human diseases: Xeroderma Pigmentosum (XP), Segmental Progeria (XFE), Fanconi Anemia (FA), Cockayne Syndrome (CS), and XP/CS combined disease (XPCSCD). Most affected individuals are compound heterozygotes for XPF/ERCC4 mutations complicating the identification of genotype/phenotype correlations. We report a detailed overview of NER and ICLR functional studies in human XPF-KO (knock-out) isogenic cells expressing six disease-specific pathogenic XPF amino acid substitution mutations. Ultraviolet (UV) sensitivity and unscheduled DNA synthesis (UDS) assays provide the most reliable information to discern mutations associated with ICLR impairment from mutations related to NER deficiency, whereas recovery of RNA synthesis (RRS) assays results hint to a possible role of XPF in resolving R-loops. Our functional studies demonstrate that a defined cellular phenotype cannot be easily correlated to each XPF mutation. Substituted positions along XPF sequences are not predictive of cellular phenotype nor reflect a particular disease. Therefore, in addition to mutation type, allelic interactions, protein stability and intracellular distribution of mutant proteins may also contribute to alter DNA repair pathways balance leading to clinically distinct disorders.
Collapse
|
74
|
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
|
75
|
Czerwińska J, Nowak M, Wojtczak P, Dziuban-Lech D, Cieśla JM, Kołata D, Gajewska B, Barańczyk-Kuźma A, Robinson AR, Shane HL, Gregg SQ, Rigatti LH, Yousefzadeh MJ, Gurkar AU, McGowan SJ, Kosicki K, Bednarek M, Zarakowska E, Gackowski D, Oliński R, Speina E, Niedernhofer LJ, Tudek B. ERCC1-deficient cells and mice are hypersensitive to lipid peroxidation. Free Radic Biol Med 2018; 124:79-96. [PMID: 29860127 PMCID: PMC6098728 DOI: 10.1016/j.freeradbiomed.2018.05.088] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 05/28/2018] [Accepted: 05/29/2018] [Indexed: 01/01/2023]
Abstract
Lipid peroxidation (LPO) products are relatively stable and abundant metabolites, which accumulate in tissues of mammals with aging, being able to modify all cellular nucleophiles, creating protein and DNA adducts including crosslinks. Here, we used cells and mice deficient in the ERCC1-XPF endonuclease required for nucleotide excision repair and the repair of DNA interstrand crosslinks to ask if specifically LPO-induced DNA damage contributes to loss of cell and tissue homeostasis. Ercc1-/- mouse embryonic fibroblasts were more sensitive than wild-type (WT) cells to the LPO products: 4-hydroxy-2-nonenal (HNE), crotonaldehyde and malondialdehyde. ERCC1-XPF hypomorphic mice were hypersensitive to CCl4 and a diet rich in polyunsaturated fatty acids, two potent inducers of endogenous LPO. To gain insight into the mechanism of how LPO influences DNA repair-deficient cells, we measured the impact of the major endogenous LPO product, HNE, on WT and Ercc1-/- cells. HNE inhibited proliferation, stimulated ROS and LPO formation, induced DNA base damage, strand breaks, error-prone translesion DNA synthesis and cellular senescence much more potently in Ercc1-/- cells than in DNA repair-competent control cells. HNE also deregulated base excision repair and energy production pathways. Our observations that ERCC1-deficient cells and mice are hypersensitive to LPO implicates LPO-induced DNA damage in contributing to cellular demise and tissue degeneration, notably even when the source of LPO is dietary polyunsaturated fats.
Collapse
Affiliation(s)
- Jolanta Czerwińska
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland.
| | - Małgorzata Nowak
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, Warsaw, Poland.
| | - Patrycja Wojtczak
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, Warsaw, Poland.
| | - Dorota Dziuban-Lech
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland.
| | - Jarosław M Cieśla
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland.
| | - Daria Kołata
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, Warsaw, Poland.
| | - Beata Gajewska
- Department of Biochemistry, Medical University of Warsaw, Warsaw, Poland.
| | | | - Andria R Robinson
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, PA, USA.
| | - Hillary L Shane
- University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, PA, USA.
| | - Siobhán Q Gregg
- University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, PA, USA; Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Lora H Rigatti
- University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, PA, USA.
| | - Matthew J Yousefzadeh
- Department of Molecular Medicine, Center on Aging, The Scripps Research Institute, Jupiter, FL, USA.
| | - Aditi U Gurkar
- Department of Molecular Medicine, Center on Aging, The Scripps Research Institute, Jupiter, FL, USA.
| | - Sara J McGowan
- Department of Molecular Medicine, Center on Aging, The Scripps Research Institute, Jupiter, FL, USA.
| | - Konrad Kosicki
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, Warsaw, Poland.
| | - Małgorzata Bednarek
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, Warsaw, Poland.
| | - Ewelina Zarakowska
- Department of Clinical Biochemistry, Faculty of Pharmacy, Collegium Medicum, Nicolaus Copernicus University in Toruń, Bydgoszcz, Poland.
| | - Daniel Gackowski
- Department of Clinical Biochemistry, Faculty of Pharmacy, Collegium Medicum, Nicolaus Copernicus University in Toruń, Bydgoszcz, Poland.
| | - Ryszard Oliński
- Department of Clinical Biochemistry, Faculty of Pharmacy, Collegium Medicum, Nicolaus Copernicus University in Toruń, Bydgoszcz, Poland.
| | - Elżbieta Speina
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland.
| | - Laura J Niedernhofer
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, PA, USA; Department of Molecular Medicine, Center on Aging, The Scripps Research Institute, Jupiter, FL, USA.
| | - Barbara Tudek
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland; Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, Warsaw, Poland.
| |
Collapse
|
76
|
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
|
77
|
|
78
|
Lee MB, Kaeberlein M. Translational Geroscience: From invertebrate models to companion animal and human interventions. TRANSLATIONAL MEDICINE OF AGING 2018; 2:15-29. [PMID: 32368707 PMCID: PMC7198054 DOI: 10.1016/j.tma.2018.08.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Translational geroscience is an interdisciplinary field descended from basic gerontology that seeks to identify, validate, and clinically apply interventions to maximize healthy, disease-free lifespan. In this review, we describe a research pipeline for the identification and validation of lifespan extending interventions. Beginning in invertebrate model systems, interventions are discovered and then characterized using other invertebrate model systems (evolutionary translation), models of genetic diversity, and disease models. Vertebrate model systems, particularly mice, can then be utilized to validate interventions in mammalian systems. Collaborative, multi-site efforts, like the Interventions Testing Program (ITP), provide a key resource to assess intervention robustness in genetically diverse mice. Mouse disease models provide a tool to understand the broader utility of longevity interventions. Beyond mouse models, we advocate for studies in companion pets. The Dog Aging Project is an exciting example of translating research in dogs, both to develop a model system and to extend their healthy lifespan as a goal in itself. Finally, we discuss proposed and ongoing intervention studies in humans, unmet needs for validating interventions in humans, and speculate on how differences in survival among human populations may influence intervention efficacy.
Collapse
Affiliation(s)
- Mitchell B Lee
- Department of Pathology, University of Washington, Seattle, WA USA
| | - Matt Kaeberlein
- Department of Pathology, University of Washington, Seattle, WA USA
| |
Collapse
|
79
|
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
|
80
|
Li C, Zhu X, Chen S, Chen L, Zhao Y, Jiang Y, Gao S, Wang F, Liu Z, Fan R, Sun L, Zhou X. Melatonin promotes the proliferation of GC-1 spg cells by inducing metallothionein-2 expression through ERK1/2 signaling pathway activation. Oncotarget 2017; 8:65627-65641. [PMID: 29029459 PMCID: PMC5630359 DOI: 10.18632/oncotarget.20019] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 07/25/2017] [Indexed: 12/31/2022] Open
Abstract
Synthesized by the pineal gland, melatonin is a neurohormone implicated in diverse physiological functions via several mechanisms. However, the role of melatonin in spermatogenesis and its underlying mechanisms have yet to be completely understood. In the present study, transcriptome sequencing was performed to characterize the mechanism of melatonin-induced GC-1 spg proliferation. Gene ontology (GO) enrichment and pathway analyses were also conducted to identify the signaling pathways and biological processes involved in differential mRNA expression. Results revealed 28 differential genes. Of these genes, 11 were upregulated and 17 were downregulated. Melatonin increased the expression of metallothionein-2 (Mt2), a gene that acts as a protector to sequester nonessential toxic heavy metals. Functional investigations demonstrated that Mt2 overexpression promoted the proliferation of GC-1 spg cells, but Mt2 knockdown significantly suppressed their proliferation and increased their apoptosis. Mechanistic analysis indicated that the extracellular-signal-regulated kinase 1/2 (ERK1/2) pathway participated in melatonin-promoted proliferation of GC-1 spg cells. Therefore, melatonin induces the proliferation of GC-spg 1 cells by stimulating Mt2 expression, and this process is mediated by the ERK1/2 signaling pathway.
Collapse
Affiliation(s)
- Chunjin Li
- College of Animal Sciences, Jilin Provincial Key Laboratory of Animal Embryo Engineering, Jilin University, Changchun, Jilin, 130062, P.R. China
| | - Xiaoling Zhu
- College of Animal Sciences, Jilin Provincial Key Laboratory of Animal Embryo Engineering, Jilin University, Changchun, Jilin, 130062, P.R. China
| | - Shuxiong Chen
- College of Animal Sciences, Jilin Provincial Key Laboratory of Animal Embryo Engineering, Jilin University, Changchun, Jilin, 130062, P.R. China
| | - Lu Chen
- College of Animal Sciences, Jilin Provincial Key Laboratory of Animal Embryo Engineering, Jilin University, Changchun, Jilin, 130062, P.R. China
| | - Yun Zhao
- College of Animal Sciences, Jilin Provincial Key Laboratory of Animal Embryo Engineering, Jilin University, Changchun, Jilin, 130062, P.R. China
| | - Yanwen Jiang
- College of Animal Sciences, Jilin Provincial Key Laboratory of Animal Embryo Engineering, Jilin University, Changchun, Jilin, 130062, P.R. China
| | - Shan Gao
- College of Animal Sciences, Jilin Provincial Key Laboratory of Animal Embryo Engineering, Jilin University, Changchun, Jilin, 130062, P.R. China
| | - Fengge Wang
- College of Animal Sciences, Jilin Provincial Key Laboratory of Animal Embryo Engineering, Jilin University, Changchun, Jilin, 130062, P.R. China
| | - Zhuo Liu
- College of Animal Sciences, Jilin Provincial Key Laboratory of Animal Embryo Engineering, Jilin University, Changchun, Jilin, 130062, P.R. China
| | - Rong Fan
- College of Animal Sciences, Jilin Provincial Key Laboratory of Animal Embryo Engineering, Jilin University, Changchun, Jilin, 130062, P.R. China
| | - Liting Sun
- College of Animal Sciences, Jilin Provincial Key Laboratory of Animal Embryo Engineering, Jilin University, Changchun, Jilin, 130062, P.R. China
| | - Xu Zhou
- College of Animal Sciences, Jilin Provincial Key Laboratory of Animal Embryo Engineering, Jilin University, Changchun, Jilin, 130062, P.R. China
| |
Collapse
|
81
|
Northcott JM, Czubryt MP, Wigle JT. Vascular senescence and ageing: a role for the MEOX proteins in promoting endothelial dysfunction. Can J Physiol Pharmacol 2017; 95:1067-1077. [PMID: 28727928 DOI: 10.1139/cjpp-2017-0149] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
In the vascular system, ageing is accompanied by the accrual of senescent cells and is associated with an increased risk of vascular disease. Endothelial cell (EC) dysfunction is a hallmark of vascular disease and is characterized by decreased angiogenic potential, reduced nitric oxide bioavailability, impaired vasodilation, increased production of ROS, and enhanced inflammation. In ECs, the major producer of nitric oxide is the endothelial nitric oxide synthase (eNOS) enzyme that is encoded by the NOS3 gene. NOS3/eNOS function is tightly regulated at both the transcriptional and post-transcriptional levels to maintain normal vascular function. A key transcriptional regulator of eNOS expression is p53, which has been shown to play a central role in mediating cellular senescence and thereby vascular dysfunction. Herein, we show that, in ECs, the MEOX homeodomain transcription factors decrease the expression of genes involved in angiogenesis, repress eNOS expression at the mRNA and protein levels, and increase the expression of p53. These findings support a role for the MEOX proteins in promoting endothelial dysfunction.
Collapse
Affiliation(s)
- Josette M Northcott
- a Institute of Cardiovascular Sciences, St. Boniface Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada.,b Department of Biochemistry and Medical Genetics, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Michael P Czubryt
- a Institute of Cardiovascular Sciences, St. Boniface Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada.,c Department of Physiology and Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Jeffrey T Wigle
- a Institute of Cardiovascular Sciences, St. Boniface Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada.,b Department of Biochemistry and Medical Genetics, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| |
Collapse
|
82
|
Takayama K, Kawakami Y, Lavasani M, Mu X, Cummins JH, Yurube T, Kuroda R, Kurosaka M, Fu FH, Robbins PD, Niedernhofer LJ, Huard J. mTOR signaling plays a critical role in the defects observed in muscle-derived stem/progenitor cells isolated from a murine model of accelerated aging. J Orthop Res 2017; 35:1375-1382. [PMID: 27572850 PMCID: PMC5516198 DOI: 10.1002/jor.23409] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 08/26/2016] [Indexed: 02/04/2023]
Abstract
Mice expressing reduced levels of ERCC1-XPF (Ercc1-/Δ mice) demonstrate premature onset of age-related changes due to decreased repair of DNA damage. Muscle-derived stem/progenitor cells (MDSPCs) isolated from Ercc1-/Δ mice have an impaired capacity for cell differentiation. The mammalian target of rapamycin (mTOR) is a critical regulator of cell growth in response to nutrient, hormone, and oxygen levels. Inhibition of the mTOR pathway extends the lifespan of several species. Here, we examined the role of mTOR in regulating the MDSPC dysfunction that occurs with accelerated aging. We show that mTOR signaling pathways are activated in Ercc1-/Δ MDSPCs compared with wild-type (WT) MDSPCs. Additionally, inhibiting mTOR with rapamycin promoted autophagy and improved the myogenic differentiation capacity of the Ercc1-/Δ MDSPCs. The percent of apoptotic and senescent cells in Ercc1-/Δ MDSPC cultures was decreased upon mTOR inhibition. These results establish that mTOR signaling contributes to stem cell dysfunction and cell fate decisions in response to endogenous DNA damage. Therefore, mTOR represents a potential therapeutic target for improving defective, aged stem cells. © 2016 The Authors. Journal of Orthopaedic Research Published by Wiley Periodicals, Inc. on behalf of Orthopaedic Research Society. J Orthop Res 35:1375-1382, 2017.
Collapse
Affiliation(s)
- Koji Takayama
- Department of Orthopaedic SurgeryUniversity of PittsburghPittsburgh 15213Pennsylvania,Stem Cell Research CenterUniversity of PittsburghPittsburgh 15219Pennsylvania,Department of Orthopaedic SurgeryKobe University Graduate School of MedicineKobe 650‐0017Japan
| | - Yohei Kawakami
- Department of Orthopaedic SurgeryUniversity of PittsburghPittsburgh 15213Pennsylvania,Stem Cell Research CenterUniversity of PittsburghPittsburgh 15219Pennsylvania,Department of Orthopaedic SurgeryKobe University Graduate School of MedicineKobe 650‐0017Japan
| | - Mitra Lavasani
- Department of Orthopaedic SurgeryUniversity of PittsburghPittsburgh 15213Pennsylvania,Stem Cell Research CenterUniversity of PittsburghPittsburgh 15219Pennsylvania
| | - Xiaodong Mu
- Department of Orthopaedic SurgeryUniversity of PittsburghPittsburgh 15213Pennsylvania,Stem Cell Research CenterUniversity of PittsburghPittsburgh 15219Pennsylvania,Department of Orthopaedic SurgeryMcGovern Medical SchoolUniversity of Texas Health Science Center at HoustonSouth Campus Research Building #3, 1881 East Rd (3SCR) 6th Floor; Room 3708Houston 77054Texas,Center for Regenerative Sports MedicineSteadman Philippon Research InstituteVail 81657Colorado
| | - James H. Cummins
- Department of Orthopaedic SurgeryUniversity of PittsburghPittsburgh 15213Pennsylvania,Stem Cell Research CenterUniversity of PittsburghPittsburgh 15219Pennsylvania,Department of Orthopaedic SurgeryMcGovern Medical SchoolUniversity of Texas Health Science Center at HoustonSouth Campus Research Building #3, 1881 East Rd (3SCR) 6th Floor; Room 3708Houston 77054Texas
| | - Takashi Yurube
- Department of Orthopaedic SurgeryUniversity of PittsburghPittsburgh 15213Pennsylvania
| | - Ryosuke Kuroda
- Department of Orthopaedic SurgeryKobe University Graduate School of MedicineKobe 650‐0017Japan
| | - Masahiro Kurosaka
- Department of Orthopaedic SurgeryKobe University Graduate School of MedicineKobe 650‐0017Japan
| | - Freddie H. Fu
- Department of Orthopaedic SurgeryUniversity of PittsburghPittsburgh 15213Pennsylvania
| | - Paul D. Robbins
- Department of Metabolism and AgingThe Scripps Research Institute FloridaJupiter 33458Florida
| | - Laura J. Niedernhofer
- Department of Metabolism and AgingThe Scripps Research Institute FloridaJupiter 33458Florida
| | - Johnny Huard
- Department of Orthopaedic SurgeryUniversity of PittsburghPittsburgh 15213Pennsylvania,Stem Cell Research CenterUniversity of PittsburghPittsburgh 15219Pennsylvania,Department of Orthopaedic SurgeryMcGovern Medical SchoolUniversity of Texas Health Science Center at HoustonSouth Campus Research Building #3, 1881 East Rd (3SCR) 6th Floor; Room 3708Houston 77054Texas,Center for Regenerative Sports MedicineSteadman Philippon Research InstituteVail 81657Colorado
| |
Collapse
|
83
|
Deng XD, Gao Q, Zhang W, Zhang B, Ma Y, Zhang LX, Muer C, Xie Y, Liu Y. The age-related expression decline of ERCC1 and XPF for forensic age estimation: A preliminary study. J Forensic Leg Med 2017; 49:15-19. [DOI: 10.1016/j.jflm.2017.05.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Revised: 03/29/2017] [Accepted: 05/01/2017] [Indexed: 01/28/2023]
|
84
|
Das D, Faridounnia M, Kovacic L, Kaptein R, Boelens R, Folkers GE. Single-stranded DNA Binding by the Helix-Hairpin-Helix Domain of XPF Protein Contributes to the Substrate Specificity of the ERCC1-XPF Protein Complex. J Biol Chem 2016; 292:2842-2853. [PMID: 28028171 DOI: 10.1074/jbc.m116.747857] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 12/24/2016] [Indexed: 11/06/2022] Open
Abstract
The nucleotide excision repair protein complex ERCC1-XPF is required for incision of DNA upstream of DNA damage. Functional studies have provided insights into the binding of ERCC1-XPF to various DNA substrates. However, because no structure for the ERCC1-XPF-DNA complex has been determined, the mechanism of substrate recognition remains elusive. Here we biochemically characterize the substrate preferences of the helix-hairpin-helix (HhH) domains of XPF and ERCC-XPF and show that the binding to single-stranded DNA (ssDNA)/dsDNA junctions is dependent on joint binding to the DNA binding domain of ERCC1 and XPF. We reveal that the homodimeric XPF is able to bind various ssDNA sequences but with a clear preference for guanine-containing substrates. NMR titration experiments and in vitro DNA binding assays also show that, within the heterodimeric ERCC1-XPF complex, XPF specifically recognizes ssDNA. On the other hand, the HhH domain of ERCC1 preferentially binds dsDNA through the hairpin region. The two separate non-overlapping DNA binding domains in the ERCC1-XPF heterodimer jointly bind to an ssDNA/dsDNA substrate and, thereby, at least partially dictate the incision position during damage removal. Based on structural models, NMR titrations, DNA-binding studies, site-directed mutagenesis, charge distribution, and sequence conservation, we propose that the HhH domain of ERCC1 binds to dsDNA upstream of the damage, and XPF binds to the non-damaged strand within a repair bubble.
Collapse
Affiliation(s)
- Devashish Das
- From the Bijvoet Center for Biomolecular Research, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands and
| | - Maryam Faridounnia
- From the Bijvoet Center for Biomolecular Research, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands and
| | - Lidija Kovacic
- the Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Jamova cesta 39, 1000 Ljubljana, Slovenia
| | - Robert Kaptein
- From the Bijvoet Center for Biomolecular Research, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands and
| | - Rolf Boelens
- From the Bijvoet Center for Biomolecular Research, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands and
| | - Gert E Folkers
- From the Bijvoet Center for Biomolecular Research, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands and
| |
Collapse
|
85
|
Ma S, Upneja A, Galecki A, Tsai YM, Burant CF, Raskind S, Zhang Q, Zhang ZD, Seluanov A, Gorbunova V, Clish CB, Miller RA, Gladyshev VN. Cell culture-based profiling across mammals reveals DNA repair and metabolism as determinants of species longevity. eLife 2016; 5:e19130. [PMID: 27874830 PMCID: PMC5148604 DOI: 10.7554/elife.19130] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Accepted: 11/21/2016] [Indexed: 12/30/2022] Open
Abstract
Mammalian lifespan differs by >100 fold, but the mechanisms associated with such longevity differences are not understood. Here, we conducted a study on primary skin fibroblasts isolated from 16 species of mammals and maintained under identical cell culture conditions. We developed a pipeline for obtaining species-specific ortholog sequences, profiled gene expression by RNA-seq and small molecules by metabolite profiling, and identified genes and metabolites correlating with species longevity. Cells from longer lived species up-regulated genes involved in DNA repair and glucose metabolism, down-regulated proteolysis and protein transport, and showed high levels of amino acids but low levels of lysophosphatidylcholine and lysophosphatidylethanolamine. The amino acid patterns were recapitulated by further analyses of primate and bird fibroblasts. The study suggests that fibroblast profiling captures differences in longevity across mammals at the level of global gene expression and metabolite levels and reveals pathways that define these differences.
Collapse
Affiliation(s)
- Siming Ma
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, United States
| | - Akhil Upneja
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, United States
| | - Andrzej Galecki
- Department of Pathology, University of Michigan Medical School, Ann Arbor, United States
- Geriatrics Center, University of Michigan Medical School, Ann Arbor, United States
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, United States
| | - Yi-Miau Tsai
- Department of Pathology, University of Michigan Medical School, Ann Arbor, United States
- Geriatrics Center, University of Michigan Medical School, Ann Arbor, United States
| | - Charles F Burant
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, United States
| | - Sasha Raskind
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, United States
| | - Quanwei Zhang
- Department of Genetics, Albert Einstein College of Medicine, Bronx, United States
| | - Zhengdong D Zhang
- Department of Genetics, Albert Einstein College of Medicine, Bronx, United States
| | - Andrei Seluanov
- Department of Biology, University of Rochester, Rochester, United States
| | - Vera Gorbunova
- Department of Biology, University of Rochester, Rochester, United States
| | | | - Richard A Miller
- Department of Pathology, University of Michigan Medical School, Ann Arbor, United States
- Geriatrics Center, University of Michigan Medical School, Ann Arbor, United States
| | - Vadim N Gladyshev
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, United States
| |
Collapse
|
86
|
van Beek AA, Sovran B, Hugenholtz F, Meijer B, Hoogerland JA, Mihailova V, van der Ploeg C, Belzer C, Boekschoten MV, Hoeijmakers JHJ, Vermeij WP, de Vos P, Wells JM, Leenen PJM, Nicoletti C, Hendriks RW, Savelkoul HFJ. Supplementation with Lactobacillus plantarum WCFS1 Prevents Decline of Mucus Barrier in Colon of Accelerated Aging Ercc1-/Δ7 Mice. Front Immunol 2016; 7:408. [PMID: 27774093 PMCID: PMC5054004 DOI: 10.3389/fimmu.2016.00408] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 09/22/2016] [Indexed: 11/23/2022] Open
Abstract
Although it is clear that probiotics improve intestinal barrier function, little is known about the effects of probiotics on the aging intestine. We investigated effects of 10-week bacterial supplementation of Lactobacillus plantarum WCFS1, Lactobacillus casei BL23, or Bifidobacterium breve DSM20213 on gut barrier and immunity in 16-week-old accelerated aging Ercc1−/Δ7 mice, which have a median lifespan of ~20 weeks, and their wild-type littermates. The colonic barrier in Ercc1−/Δ7 mice was characterized by a thin (< 10 μm) mucus layer. L. plantarum prevented this decline in mucus integrity in Ercc1−/Δ7 mice, whereas B. breve exacerbated it. Bacterial supplementations affected the expression of immune-related genes, including Toll-like receptor 4. Regulatory T cell frequencies were increased in the mesenteric lymph nodes of L. plantarum- and L. casei-treated Ercc1−/Δ7 mice. L. plantarum- and L. casei-treated Ercc1−/Δ7 mice showed increased specific antibody production in a T cell-dependent immune response in vivo. By contrast, the effects of bacterial supplementation on wild-type control mice were negligible. Thus, supplementation with L. plantarum – but not with L. casei and B. breve – prevented the decline in the mucus barrier in Ercc1−/Δ7 mice. Our data indicate that age is an important factor influencing beneficial or detrimental effects of candidate probiotics. These findings also highlight the need for caution in translating beneficial effects of probiotics observed in young animals or humans to the elderly.
Collapse
Affiliation(s)
- Adriaan A van Beek
- Cell Biology and Immunology Group, Wageningen University, Wageningen, Netherlands; Top Institute Food and Nutrition, Wageningen, Netherlands; Gut Health and Food Safety, Institute of Food Research, Norwich, UK
| | - Bruno Sovran
- Top Institute Food and Nutrition, Wageningen, Netherlands; Host Microbe Interactomics, Wageningen University, Wageningen, Netherlands
| | - Floor Hugenholtz
- Top Institute Food and Nutrition, Wageningen, Netherlands; Laboratory of Microbiology, Wageningen University, Wageningen, Netherlands
| | - Ben Meijer
- Cell Biology and Immunology Group, Wageningen University , Wageningen , Netherlands
| | - Joanne A Hoogerland
- Cell Biology and Immunology Group, Wageningen University , Wageningen , Netherlands
| | - Violeta Mihailova
- Cell Biology and Immunology Group, Wageningen University , Wageningen , Netherlands
| | - Corine van der Ploeg
- Cell Biology and Immunology Group, Wageningen University , Wageningen , Netherlands
| | - Clara Belzer
- Top Institute Food and Nutrition, Wageningen, Netherlands; Laboratory of Microbiology, Wageningen University, Wageningen, Netherlands
| | - Mark V Boekschoten
- Top Institute Food and Nutrition, Wageningen, Netherlands; Human Nutrition, Wageningen University, Wageningen, Netherlands
| | - Jan H J Hoeijmakers
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, Netherlands; CECAD Forschungszentrum, Universität zu Köln, Köln, Germany
| | - Wilbert P Vermeij
- Department of Molecular Genetics, Erasmus University Medical Center , Rotterdam , Netherlands
| | - Paul de Vos
- Top Institute Food and Nutrition, Wageningen, Netherlands; University of Groningen, Groningen, Netherlands
| | - Jerry M Wells
- Top Institute Food and Nutrition, Wageningen, Netherlands; Host Microbe Interactomics, Wageningen University, Wageningen, Netherlands
| | - Pieter J M Leenen
- Department of Immunology, Erasmus University Medical Center , Rotterdam , Netherlands
| | - Claudio Nicoletti
- Gut Health and Food Safety, Institute of Food Research, Norwich, UK; Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Rudi W Hendriks
- Department of Pulmonary Medicine, Erasmus University Medical Center , Rotterdam , Netherlands
| | - Huub F J Savelkoul
- Cell Biology and Immunology Group, Wageningen University, Wageningen, Netherlands; Top Institute Food and Nutrition, Wageningen, Netherlands
| |
Collapse
|
87
|
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
|
88
|
Vermeij WP, Dollé MET, Reiling E, Jaarsma D, Payan-Gomez C, Bombardieri CR, Wu H, Roks AJM, Botter SM, van der Eerden BC, Youssef SA, Kuiper RV, Nagarajah B, van Oostrom CT, Brandt RMC, Barnhoorn S, Imholz S, Pennings JLA, de Bruin A, Gyenis Á, Pothof J, Vijg J, van Steeg H, Hoeijmakers JHJ. Restricted diet delays accelerated ageing and genomic stress in DNA-repair-deficient mice. Nature 2016; 537:427-431. [PMID: 27556946 PMCID: PMC5161687 DOI: 10.1038/nature19329] [Citation(s) in RCA: 225] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 07/25/2016] [Indexed: 12/27/2022]
Abstract
Mice deficient in the DNA excision-repair gene Ercc1 (Ercc1∆/-) show numerous accelerated ageing features that limit their lifespan to 4-6 months. They also exhibit a 'survival response', which suppresses growth and enhances cellular maintenance. Such a response resembles the anti-ageing response induced by dietary restriction (also known as caloric restriction). Here we report that a dietary restriction of 30% tripled the median and maximal remaining lifespans of these progeroid mice, strongly retarding numerous aspects of accelerated ageing. Mice undergoing dietary restriction retained 50% more neurons and maintained full motor function far beyond the lifespan of mice fed ad libitum. Other DNA-repair-deficient, progeroid Xpg-/- (also known as Ercc5-/-) mice, a model of Cockayne syndrome, responded similarly. The dietary restriction response in Ercc1∆/- mice closely resembled the effects of dietary restriction in wild-type animals. Notably, liver tissue from Ercc1∆/- mice fed ad libitum showed preferential extinction of the expression of long genes, a phenomenon we also observed in several tissues ageing normally. This is consistent with the accumulation of stochastic, transcription-blocking lesions that affect long genes more than short ones. Dietary restriction largely prevented this declining transcriptional output and reduced the number of γH2AX DNA damage foci, indicating that dietary restriction preserves genome function by alleviating DNA damage. Our findings establish the Ercc1∆/- mouse as a powerful model organism for health-sustaining interventions, reveal potential for reducing endogenous DNA damage, facilitate a better understanding of the molecular mechanism of dietary restriction and suggest a role for counterintuitive dietary-restriction-like therapy for human progeroid genome instability syndromes and possibly neurodegeneration in general.
Collapse
Affiliation(s)
- W P Vermeij
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - M E T Dollé
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), PO Box 1, 3720 BA Bilthoven, The Netherlands
| | - E Reiling
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands.,Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), PO Box 1, 3720 BA Bilthoven, The Netherlands
| | - D Jaarsma
- Department of Neuroscience, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - C Payan-Gomez
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands.,Facultad de Ciencias Naturales y Matemáticas, Universidad del Rosario, Carrera 24, 63C-69 Bogotá, Colombia
| | - C R Bombardieri
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - H Wu
- Department of Internal Medicine, Division of Vascular Medicine and Pharmacology, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - A J M Roks
- Department of Internal Medicine, Division of Vascular Medicine and Pharmacology, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - S M Botter
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands.,Laboratory for Orthopedic Research, Balgrist University Hospital, Forchstrasse 340, 8008, Zürich, Switzerland
| | - B C van der Eerden
- Department of Internal Medicine, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - S A Youssef
- Dutch Molecular Pathology Center, Department of Pathobiology, Faculty of Veterinary Medicine, Utrecht University, PO Box 80125, 3508 TC Utrecht, The Netherlands
| | - R V Kuiper
- Dutch Molecular Pathology Center, Department of Pathobiology, Faculty of Veterinary Medicine, Utrecht University, PO Box 80125, 3508 TC Utrecht, The Netherlands
| | - B Nagarajah
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), PO Box 1, 3720 BA Bilthoven, The Netherlands
| | - C T van Oostrom
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), PO Box 1, 3720 BA Bilthoven, The Netherlands
| | - R M C Brandt
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - S Barnhoorn
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - S Imholz
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), PO Box 1, 3720 BA Bilthoven, The Netherlands
| | - J L A Pennings
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), PO Box 1, 3720 BA Bilthoven, The Netherlands
| | - A de Bruin
- Dutch Molecular Pathology Center, Department of Pathobiology, Faculty of Veterinary Medicine, Utrecht University, PO Box 80125, 3508 TC Utrecht, The Netherlands.,Department of Pediatrics, Division Molecular Genetics, University Medical Center Groningen, PO Box 30001, 9700 RB Groningen, The Netherlands
| | - Á Gyenis
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - J Pothof
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - J Vijg
- Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York 10461, USA
| | - H van Steeg
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), PO Box 1, 3720 BA Bilthoven, The Netherlands.,Department of Human Genetics, Leiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands
| | - J H J Hoeijmakers
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands.,CECAD Forschungszentrum, Universität zu Köln, Joseph-Stelzmann-Straße 26, 50931 Köln, Germany
| |
Collapse
|
89
|
Jokić M, Vlašić I, Rinneburger M, Klümper N, Spiro J, Vogel W, Offermann A, Kümpers C, Fritz C, Schmitt A, Riabinska A, Wittersheim M, Michels S, Ozretić L, Florin A, Welcker D, Akyuz MD, Nowak M, Erkel M, Wolf J, Büttner R, Schumacher B, Thomale J, Persigehl T, Maintz D, Perner S, Reinhardt HC. Ercc1 Deficiency Promotes Tumorigenesis and Increases Cisplatin Sensitivity in a Tp53 Context-Specific Manner. Mol Cancer Res 2016; 14:1110-1123. [PMID: 27514406 DOI: 10.1158/1541-7786.mcr-16-0094] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 06/18/2016] [Accepted: 07/06/2016] [Indexed: 11/16/2022]
Abstract
KRAS-mutant lung adenocarcinoma is among the most common cancer entities and, in advanced stages, typically displays poor prognosis due to acquired resistance against chemotherapy, which is still largely based on cisplatin-containing combination regimens. Mechanisms of cisplatin resistance have been extensively investigated, and ERCC1 has emerged as a key player due to its central role in the repair of cisplatin-induced DNA lesions. However, clinical data have not unequivocally confirmed ERCC1 status as a predictor of the response to cisplatin treatment. Therefore, we employed an autochthonous mouse model of Kras-driven lung adenocarcinoma resembling human lung adenocarcinoma to investigate the role of Ercc1 in the response to cisplatin treatment. Our data show that Ercc1 deficiency in Tp53-deficient murine lung adenocarcinoma induces a more aggressive tumor phenotype that displays enhanced sensitivity to cisplatin treatment. Furthermore, tumors that relapsed after cisplatin treatment in our model develop a robust etoposide sensitivity that is independent of the Ercc1 status and depends solely on previous cisplatin exposure. Our results provide a solid rationale for further investigation of the possibility of preselection of lung adenocarcinoma patients according to the functional ERCC1- and mutational TP53 status, where functionally ERCC1-incompetent patients might benefit from sequential cisplatin and etoposide chemotherapy. IMPLICATIONS This study provides a solid rationale for the stratification of lung adenocarcinoma patients according to the functional ERCC1- and mutational TP53 status, where functionally ERCC1-incompetent patients could benefit from sequential cisplatin and etoposide chemotherapy. Mol Cancer Res; 14(11); 1110-23. ©2016 AACR.
Collapse
Affiliation(s)
- Mladen Jokić
- Department I of Internal Medicine, University Hospital of Cologne, Weyertal 115B, 50931, Cologne, Germany. .,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases, University of Cologne, Weyertal 115B, 50931, Cologne, Germany
| | - Ignacija Vlašić
- Department I of Internal Medicine, University Hospital of Cologne, Weyertal 115B, 50931, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases, University of Cologne, Weyertal 115B, 50931, Cologne, Germany
| | - Miriam Rinneburger
- Department I of Internal Medicine, University Hospital of Cologne, Weyertal 115B, 50931, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases, University of Cologne, Weyertal 115B, 50931, Cologne, Germany
| | - Niklas Klümper
- Pathology of the University Medical Center Schleswig-Holstein, Campus Luebeck and the Research Center Borstel, Leibniz Center for Medicine and Biosciences, 23538 Luebeck and 23845 Borstel, Germany
| | - Judith Spiro
- Department of Radiology, University Hospital of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Wenzel Vogel
- Pathology of the University Medical Center Schleswig-Holstein, Campus Luebeck and the Research Center Borstel, Leibniz Center for Medicine and Biosciences, 23538 Luebeck and 23845 Borstel, Germany
| | - Anne Offermann
- Pathology of the University Medical Center Schleswig-Holstein, Campus Luebeck and the Research Center Borstel, Leibniz Center for Medicine and Biosciences, 23538 Luebeck and 23845 Borstel, Germany
| | - Christiane Kümpers
- Pathology of the University Medical Center Schleswig-Holstein, Campus Luebeck and the Research Center Borstel, Leibniz Center for Medicine and Biosciences, 23538 Luebeck and 23845 Borstel, Germany
| | - Christian Fritz
- Department I of Internal Medicine, University Hospital of Cologne, Weyertal 115B, 50931, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases, University of Cologne, Weyertal 115B, 50931, Cologne, Germany
| | - Anna Schmitt
- Department I of Internal Medicine, University Hospital of Cologne, Weyertal 115B, 50931, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases, University of Cologne, Weyertal 115B, 50931, Cologne, Germany
| | - Arina Riabinska
- Department I of Internal Medicine, University Hospital of Cologne, Weyertal 115B, 50931, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases, University of Cologne, Weyertal 115B, 50931, Cologne, Germany
| | - Maike Wittersheim
- Institute of Pathology, University Hospital of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Sebastian Michels
- Department I of Internal Medicine, University Hospital of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Luka Ozretić
- Institute of Pathology, University Hospital of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Alexandra Florin
- Institute of Pathology, University Hospital of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Daniela Welcker
- Department I of Internal Medicine, University Hospital of Cologne, Weyertal 115B, 50931, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases, University of Cologne, Weyertal 115B, 50931, Cologne, Germany.,Department II of Internal Medicine, University Hospital Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Mehmet Deniz Akyuz
- Institute for genome stability in ageing and disease, CECAD Research Center, Joseph-Stelzmann-Str. 26, 50931, Cologne, Germany
| | - Michael Nowak
- Institute of Pathology, University Hospital Bonn, Sigmund-Freud-Str. 25, 53127, Bonn, Germany
| | - Martin Erkel
- Institute for Cell Biology, University Hospital Essen, Hufelandstraβe 55, 45122, Essen, Germany
| | - Jürgen Wolf
- Department I of Internal Medicine, University Hospital of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Reinhard Büttner
- Institute of Pathology, University Hospital of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Björn Schumacher
- Institute for genome stability in ageing and disease, CECAD Research Center, Joseph-Stelzmann-Str. 26, 50931, Cologne, Germany
| | - Jürgen Thomale
- Institute for Cell Biology, University Hospital Essen, Hufelandstraβe 55, 45122, Essen, Germany
| | - Thorsten Persigehl
- Department of Radiology, University Hospital of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - David Maintz
- Department of Radiology, University Hospital of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Sven Perner
- Pathology of the University Medical Center Schleswig-Holstein, Campus Luebeck and the Research Center Borstel, Leibniz Center for Medicine and Biosciences, 23538 Luebeck and 23845 Borstel, Germany
| | - Hans Christian Reinhardt
- Department I of Internal Medicine, University Hospital of Cologne, Weyertal 115B, 50931, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases, University of Cologne, Weyertal 115B, 50931, Cologne, Germany
| |
Collapse
|
90
|
van Beek AA, Hugenholtz F, Meijer B, Sovran B, Perdijk O, Vermeij WP, Brandt RMC, Barnhoorn S, Hoeijmakers JHJ, de Vos P, Leenen PJM, Hendriks RW, Savelkoul HFJ. Frontline Science: Tryptophan restriction arrests B cell development and enhances microbial diversity in WT and prematurely aging Ercc1-/Δ7 mice. J Leukoc Biol 2016; 101:811-821. [PMID: 27418353 DOI: 10.1189/jlb.1hi0216-062rr] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 06/09/2016] [Accepted: 06/14/2016] [Indexed: 12/13/2022] Open
Abstract
With aging, tryptophan metabolism is affected. Tryptophan has a crucial role in the induction of immune tolerance and the maintenance of gut microbiota. We, therefore, studied the effect of dietary tryptophan restriction in young wild-type (WT) mice (118-wk life span) and in DNA-repair deficient, premature-aged (Ercc1-/Δ7 ) mice (20-wk life span). First, we found that the effect of aging on the distribution of B and T cells in bone marrow (BM) and in the periphery of 16-wk-old Ercc1-/Δ7 mice was comparable to that in 18-mo-old WT mice. Dietary tryptophan restriction caused an arrest of B cell development in the BM, accompanied by diminished B cell frequencies in the periphery. In general, old Ercc1-/Δ7 mice showed similar responses to tryptophan restriction compared with young WT mice, indicative of age-independent effects. Dietary tryptophan restriction increased microbial diversity and made the gut microbiota composition of old Ercc1-/Δ7 mice more similar to that of young WT mice. The decreased abundances of Alistipes and Akkermansia spp. after dietary tryptophan restriction correlated significantly with decreased B cell precursor numbers. In conclusion, we report that dietary tryptophan restriction arrests B cell development and concomitantly changes gut microbiota composition. Our study suggests a beneficial interplay between dietary tryptophan, B cell development, and gut microbial composition on several aspects of age-induced changes.
Collapse
Affiliation(s)
- Adriaan A van Beek
- Top Institute Food and Nutrition, Wageningen, The Netherlands; .,Cell Biology and Immunology Group, Wageningen University, Wageningen, The Netherlands.,Department of Immunology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Floor Hugenholtz
- Top Institute Food and Nutrition, Wageningen, The Netherlands.,Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
| | - Ben Meijer
- Cell Biology and Immunology Group, Wageningen University, Wageningen, The Netherlands
| | - Bruno Sovran
- Top Institute Food and Nutrition, Wageningen, The Netherlands.,Host-Microbe Interactomics Group, Wageningen University, Wageningen, The Netherlands
| | - Olaf Perdijk
- Cell Biology and Immunology Group, Wageningen University, Wageningen, The Netherlands
| | - Wilbert P Vermeij
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Renata M C Brandt
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Sander Barnhoorn
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Jan H J Hoeijmakers
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Paul de Vos
- Top Institute Food and Nutrition, Wageningen, The Netherlands.,Pathology and Medical Biology, University of Groningen, Groningen, The Netherlands; and
| | - Pieter J M Leenen
- Department of Immunology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Rudi W Hendriks
- Department of Pulmonary Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Huub F J Savelkoul
- Cell Biology and Immunology Group, Wageningen University, Wageningen, The Netherlands
| |
Collapse
|
91
|
Sepe S, Milanese C, Gabriels S, Derks KWJ, Payan-Gomez C, van IJcken WFJ, Rijksen YMA, Nigg AL, Moreno S, Cerri S, Blandini F, Hoeijmakers JHJ, Mastroberardino PG. Inefficient DNA Repair Is an Aging-Related Modifier of Parkinson's Disease. Cell Rep 2016; 15:1866-75. [PMID: 27210754 PMCID: PMC4893155 DOI: 10.1016/j.celrep.2016.04.071] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 02/01/2016] [Accepted: 04/19/2016] [Indexed: 11/27/2022] Open
Abstract
The underlying relation between Parkinson’s disease (PD) etiopathology and its major risk factor, aging, is largely unknown. In light of the causative link between genome stability and aging, we investigate a possible nexus between DNA damage accumulation, aging, and PD by assessing aging-related DNA repair pathways in laboratory animal models and humans. We demonstrate that dermal fibroblasts from PD patients display flawed nucleotide excision repair (NER) capacity and that Ercc1 mutant mice with mildly compromised NER exhibit typical PD-like pathological alterations, including decreased striatal dopaminergic innervation, increased phospho-synuclein levels, and defects in mitochondrial respiration. Ercc1 mouse mutants are also more sensitive to the prototypical PD toxin MPTP, and their transcriptomic landscape shares important similarities with that of PD patients. Our results demonstrate that specific defects in DNA repair impact the dopaminergic system and are associated with human PD pathology and might therefore constitute an age-related risk factor for PD. Ercc1-mediated DNA repair is necessary for preservation of dopaminergic neurons Mouse mutants with mild Ercc1 defects display signs of dopaminergic pathology Mild Ercc1 dysfunction is sensitized to the prototypical PD neurotoxin MPTP PD patients’ peripheral cells exhibit inefficient nucleotide excision repair
Collapse
Affiliation(s)
- Sara Sepe
- Department of Molecular Genetics, Erasmus Medical Center, 3015 Rotterdam, the Netherlands
| | - Chiara Milanese
- Department of Molecular Genetics, Erasmus Medical Center, 3015 Rotterdam, the Netherlands; Ri.Med Foundation, 90133 Palermo, Italy
| | - Sylvia Gabriels
- Department of Molecular Genetics, Erasmus Medical Center, 3015 Rotterdam, the Netherlands
| | - Kasper W J Derks
- Department of Molecular Genetics, Erasmus Medical Center, 3015 Rotterdam, the Netherlands
| | - Cesar Payan-Gomez
- Department of Molecular Genetics, Erasmus Medical Center, 3015 Rotterdam, the Netherlands; Facultad de Ciencias Naturales y Matemáticas, Universidad del Rosario, 111711 Bogotá, Colombia
| | | | - Yvonne M A Rijksen
- Department of Molecular Genetics, Erasmus Medical Center, 3015 Rotterdam, the Netherlands
| | - Alex L Nigg
- Optical Imaging Center, Erasmus Medical Centre, 3015 Rotterdam, the Netherlands
| | | | - Silvia Cerri
- Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, C. Mondino National Neurological Institute, 27100 Pavia, Italy
| | - Fabio Blandini
- Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, C. Mondino National Neurological Institute, 27100 Pavia, Italy
| | - Jan H J Hoeijmakers
- Department of Molecular Genetics, Erasmus Medical Center, 3015 Rotterdam, the Netherlands
| | - Pier G Mastroberardino
- Department of Molecular Genetics, Erasmus Medical Center, 3015 Rotterdam, the Netherlands.
| |
Collapse
|
92
|
Bautista-Niño PK, Portilla-Fernandez E, Vaughan DE, Danser AHJ, Roks AJM. DNA Damage: A Main Determinant of Vascular Aging. Int J Mol Sci 2016; 17:E748. [PMID: 27213333 PMCID: PMC4881569 DOI: 10.3390/ijms17050748] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 05/04/2016] [Accepted: 05/10/2016] [Indexed: 01/16/2023] Open
Abstract
Vascular aging plays a central role in health problems and mortality in older people. Apart from the impact of several classical cardiovascular risk factors on the vasculature, chronological aging remains the single most important determinant of cardiovascular problems. The causative mechanisms by which chronological aging mediates its impact, independently from classical risk factors, remain to be elucidated. In recent years evidence has accumulated that unrepaired DNA damage may play an important role. Observations in animal models and in humans indicate that under conditions during which DNA damage accumulates in an accelerated rate, functional decline of the vasculature takes place in a similar but more rapid or more exaggerated way than occurs in the absence of such conditions. Also epidemiological studies suggest a relationship between DNA maintenance and age-related cardiovascular disease. Accordingly, mouse models of defective DNA repair are means to study the mechanisms involved in biological aging of the vasculature. We here review the evidence of the role of DNA damage in vascular aging, and present mechanisms by which genomic instability interferes with regulation of the vascular tone. In addition, we present potential remedies against vascular aging induced by genomic instability. Central to this review is the role of diverse types of DNA damage (telomeric, non-telomeric and mitochondrial), of cellular changes (apoptosis, senescence, autophagy), mediators of senescence and cell growth (plasminogen activator inhibitor-1 (PAI-1), cyclin-dependent kinase inhibitors, senescence-associated secretory phenotype (SASP)/senescence-messaging secretome (SMS), insulin and insulin-like growth factor 1 (IGF-1) signaling), the adenosine monophosphate-activated protein kinase (AMPK)-mammalian target of rapamycin (mTOR)-nuclear factor kappa B (NFκB) axis, reactive oxygen species (ROS) vs. endothelial nitric oxide synthase (eNOS)-cyclic guanosine monophosphate (cGMP) signaling, phosphodiesterase (PDE) 1 and 5, transcription factor NF-E2-related factor-2 (Nrf2), and diet restriction.
Collapse
Affiliation(s)
- Paula K Bautista-Niño
- Department of Internal Medicine, Division of Vascular Medicine and Pharmacology, Erasmus Medical Center, Rotterdam 3015 CN, The Netherlands.
- Department of Epidemiology, Erasmus Medical Center, Rotterdam 3015 CN, The Netherlands.
| | - Eliana Portilla-Fernandez
- Department of Internal Medicine, Division of Vascular Medicine and Pharmacology, Erasmus Medical Center, Rotterdam 3015 CN, The Netherlands.
- Department of Epidemiology, Erasmus Medical Center, Rotterdam 3015 CN, The Netherlands.
| | - Douglas E Vaughan
- Department of Medicine & Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| | - A H Jan Danser
- Department of Internal Medicine, Division of Vascular Medicine and Pharmacology, Erasmus Medical Center, Rotterdam 3015 CN, The Netherlands.
| | - Anton J M Roks
- Department of Internal Medicine, Division of Vascular Medicine and Pharmacology, Erasmus Medical Center, Rotterdam 3015 CN, The Netherlands.
| |
Collapse
|
93
|
Visser WE, Bombardieri CR, Zevenbergen C, Barnhoorn S, Ottaviani A, van der Pluijm I, Brandt R, Kaptein E, van Heerebeek R, van Toor H, Garinis GA, Peeters RP, Medici M, van Ham W, Vermeij WP, de Waard MC, de Krijger RR, Boelen A, Kwakkel J, Kopchick JJ, List EO, Melis JPM, Darras VM, Dollé MET, van der Horst GTJ, Hoeijmakers JHJ, Visser TJ. Tissue-Specific Suppression of Thyroid Hormone Signaling in Various Mouse Models of Aging. PLoS One 2016; 11:e0149941. [PMID: 26953569 PMCID: PMC4783069 DOI: 10.1371/journal.pone.0149941] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 02/07/2016] [Indexed: 01/24/2023] Open
Abstract
DNA damage contributes to the process of aging, as underscored by premature aging syndromes caused by defective DNA repair. Thyroid state changes during aging, but underlying mechanisms remain elusive. Since thyroid hormone (TH) is a key regulator of metabolism, changes in TH signaling have widespread effects. Here, we reveal a significant common transcriptomic signature in livers from hypothyroid mice, DNA repair-deficient mice with severe (Csbm/m/Xpa-/-) or intermediate (Ercc1-/Δ-7) progeria and naturally aged mice. A strong induction of TH-inactivating deiodinase D3 and decrease of TH-activating D1 activities are observed in Csbm/m/Xpa-/- livers. Similar findings are noticed in Ercc1-/Δ-7, in naturally aged animals and in wild-type mice exposed to a chronic subtoxic dose of DNA-damaging agents. In contrast, TH signaling in muscle, heart and brain appears unaltered. These data show a strong suppression of TH signaling in specific peripheral organs in premature and normal aging, probably lowering metabolism, while other tissues appear to preserve metabolism. D3-mediated TH inactivation is unexpected, given its expression mainly in fetal tissues. Our studies highlight the importance of DNA damage as the underlying mechanism of changes in thyroid state. Tissue-specific regulation of deiodinase activities, ensuring diminished TH signaling, may contribute importantly to the protective metabolic response in aging.
Collapse
Affiliation(s)
- W. Edward Visser
- Dept of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
- * E-mail:
| | - Cíntia R. Bombardieri
- MGC Dept of Genetics, Cancer Genomics Center, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Chantal Zevenbergen
- Dept of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Sander Barnhoorn
- MGC Dept of Genetics, Cancer Genomics Center, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Alexandre Ottaviani
- MGC Dept of Genetics, Cancer Genomics Center, Erasmus Medical Center, Rotterdam, The Netherlands
- Institute for Research on Cancer and Aging, Nice (IRCAN), UMR 7284 CNRS U1081 INSERM UNS, 28 avenue de Valombrose Faculté de Médecine, Nice, France
| | - Ingrid van der Pluijm
- MGC Dept of Genetics, Cancer Genomics Center, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Renata Brandt
- MGC Dept of Genetics, Cancer Genomics Center, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ellen Kaptein
- Dept of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Hans van Toor
- Dept of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - George A. Garinis
- MGC Dept of Genetics, Cancer Genomics Center, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Robin P. Peeters
- Dept of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Marco Medici
- Dept of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Willy van Ham
- Laboratory of Comparative Endocrinology, Biology Department, KULeuven, Leuven, Belgium
| | - Wilbert P. Vermeij
- MGC Dept of Genetics, Cancer Genomics Center, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Monique C. de Waard
- MGC Dept of Genetics, Cancer Genomics Center, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Anita Boelen
- Dept of Endocrinology and Metabolism, Academic Medical Center, Amsterdam, The Netherlands
| | - Joan Kwakkel
- Dept of Endocrinology and Metabolism, Academic Medical Center, Amsterdam, The Netherlands
| | - John J. Kopchick
- Dept of Biomedical Sciences, Edison Biotechnology Institute, Ohio University, Athens, Ohio, United States of America
| | - Edward O. List
- Dept of Biomedical Sciences, Edison Biotechnology Institute, Ohio University, Athens, Ohio, United States of America
| | - Joost P. M. Melis
- Dept of Toxicogenetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Veerle M. Darras
- Laboratory of Comparative Endocrinology, Biology Department, KULeuven, Leuven, Belgium
| | - Martijn E. T. Dollé
- Centre for Health Protection Research, National Institute of Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | | | - Jan H. J. Hoeijmakers
- MGC Dept of Genetics, Cancer Genomics Center, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Theo J. Visser
- Dept of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
94
|
Andriani GA, Faggioli F, Baker D, Dollé MET, Sellers RS, Hébert JM, Van Steeg H, Hoeijmakers J, Vijg J, Montagna C. Whole chromosome aneuploidy in the brain of Bub1bH/H and Ercc1-/Δ7 mice. Hum Mol Genet 2016; 25:755-65. [PMID: 26681803 PMCID: PMC4743693 DOI: 10.1093/hmg/ddv612] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Revised: 12/07/2015] [Accepted: 12/14/2015] [Indexed: 12/16/2022] Open
Abstract
High levels of aneuploidy have been observed in disease-free tissues, including post-mitotic tissues such as the brain. Using a quantitative interphase-fluorescence in situ hybridization approach, we previously reported a chromosome-specific, age-related increase in aneuploidy in the mouse cerebral cortex. Increased aneuploidy has been associated with defects in DNA repair and the spindle assembly checkpoint, which in turn can lead to premature aging. Here, we quantified the frequency of aneuploidy of three autosomes in the cerebral cortex and cerebellum of adult and developing brain of Bub1b(H/H) mice, which have a faulty mitotic checkpoint, and Ercc1(-/Δ7) mice, defective in nucleotide excision repair and inter-strand cross-link repair. Surprisingly, the level of aneuploidy in the brain of these murine models of accelerated aging remains as low as in the young adult brains from control animals, i.e. <1% in the cerebral cortex and ∼0.1% in the cerebellum. Therefore, based on aneuploidy, these adult mice with reduced life span and accelerated progeroid features are indistinguishable from age-matched, normal controls. Yet, during embryonic development, we found that Bub1b(H/H), but not Ercc1(-/Δ7) mice, have a significantly higher frequency of aneuploid nuclei relative to wild-type controls in the cerebral cortex, reaching a frequency as high as 40.3% for each chromosome tested. Aneuploid cells in these mutant mice are likely eliminated early in development through apoptosis and/or immune-mediated clearance mechanisms, which would explain the low levels of aneuploidy during adulthood in the cerebral cortex of Bub1b(H/H) mice. These results shed light on the mechanisms of removal of aneuploidy cells in vivo.
Collapse
Affiliation(s)
| | | | - Darren Baker
- Department of Pediatric and Adolescent Medicine, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Martijn E T Dollé
- National Institute of Public Health and the Environment, Bilthoven, The Netherlands and
| | | | - Jean M Hébert
- Department of Genetics, Dominick P. Purpura Department of Neuroscience
| | - Harry Van Steeg
- National Institute of Public Health and the Environment, Bilthoven, The Netherlands and
| | - Jan Hoeijmakers
- MGC Department of Genetics, CBG Cancer Genomics Center, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Jan Vijg
- Department of Genetics, Department Ophthalmology and Visual Science and Department of Obstetrics and Gynecology and Women's Health, Albert Einstein College of Medicine, Yeshiva University, Bronx, NY 10461, USA
| | | |
Collapse
|
95
|
Gorbunova V, Seluanov A. DNA double strand break repair, aging and the chromatin connection. Mutat Res 2016; 788:2-6. [PMID: 26923716 DOI: 10.1016/j.mrfmmm.2016.02.004] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2016] [Revised: 02/01/2016] [Accepted: 02/10/2016] [Indexed: 01/07/2023]
Abstract
Are DNA damage and mutations possible causes or consequences of aging? This question has been hotly debated by biogerontologists for decades. The importance of DNA damage as a possible driver of the aging process went from being widely recognized to then forgotten, and is now slowly making a comeback. DNA double strand breaks (DSBs) are particularly relevant to aging because of their toxicity, increased frequency with age and the association of defects in their repair with premature aging. Recent studies expand the potential impact of DNA damage and mutations on aging by linking DNA DSB repair and age-related chromatin changes. There is overwhelming evidence that increased DNA damage and mutations accelerate aging. However, an ultimate proof of causality would be to show that enhanced genome and epigenome stability delays aging. This is not an easy task, as improving such complex biological processes is infinitely more difficult than disabling it. We will discuss the possibility that animal models with enhanced DNA repair and epigenome maintenance will be generated in the near future.
Collapse
Affiliation(s)
- Vera Gorbunova
- University of Rochester, Department of Biology, Hutchison Hall, RC, Rochester, NY 14627, USA.
| | - Andrei Seluanov
- University of Rochester, Department of Biology, Hutchison Hall, RC, Rochester, NY 14627, USA
| |
Collapse
|
96
|
Abstract
Progeroid mouse models display phenotypes in multiple organ systems that suggest premature aging and resemble features of natural aging of both mice and humans. The prospect of a significant increase in the global elderly population within the next decades has led to the emergence of "geroscience," which aims at elucidating the molecular mechanisms involved in aging. Progeroid mouse models are frequently used in geroscience as they provide insight into the molecular mechanisms that are involved in the highly complex process of natural aging. This review provides an overview of the most commonly reported nonneoplastic macroscopic and microscopic pathologic findings in progeroid mouse models (eg, osteoporosis, osteoarthritis, degenerative joint disease, intervertebral disc degeneration, kyphosis, sarcopenia, cutaneous atrophy, wound healing, hair loss, alopecia, lymphoid atrophy, cataract, corneal endothelial dystrophy, retinal degenerative diseases, and vascular remodeling). Furthermore, several shortcomings in pathologic analysis and descriptions of these models are discussed. Progeroid mouse models are valuable models for aging, but thorough knowledge of both the mouse strain background and the progeria-related phenotype is required to guide interpretation and translation of the pathology data.
Collapse
Affiliation(s)
- L Harkema
- Dutch Molecular Pathology Center, Department of Pathobiology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - S A Youssef
- Dutch Molecular Pathology Center, Department of Pathobiology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - A de Bruin
- Dutch Molecular Pathology Center, Department of Pathobiology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands Department of Pediatrics, Division of Molecular Genetics, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
97
|
Vermeij WP, Hoeijmakers JHJ, Pothof J. Genome Integrity in Aging: Human Syndromes, Mouse Models, and Therapeutic Options. Annu Rev Pharmacol Toxicol 2015; 56:427-45. [PMID: 26514200 DOI: 10.1146/annurev-pharmtox-010814-124316] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Human syndromes and mouse mutants that exhibit accelerated but bona fide aging in multiple organs and tissues have been invaluable for the identification of nine denominators of aging: telomere attrition, genome instability, epigenetic alterations, mitochondrial dysfunction, deregulated nutrient sensing, altered intercellular communication, loss of proteostasis, cellular senescence and adult stem cell exhaustion. However, whether and how these instigators of aging interrelate or whether they have one root cause is currently largely unknown. Rare human progeroid syndromes and corresponding mouse mutants with resolved genetic defects highlight the dominant importance of genome maintenance for aging. A second class of aging-related disorders reveals a cross connection with metabolism. As genome maintenance and metabolism are closely interconnected, they may constitute the main underlying biology of aging. This review focuses on the role of genome stability in aging, its crosstalk with metabolism, and options for nutritional and/or pharmaceutical interventions that delay age-related pathology.
Collapse
Affiliation(s)
- Wilbert P Vermeij
- Department of Genetics, Erasmus University Medical Center, Postbus 2040, 3000 CA, Rotterdam, The Netherlands; , ,
| | - Jan H J Hoeijmakers
- Department of Genetics, Erasmus University Medical Center, Postbus 2040, 3000 CA, Rotterdam, The Netherlands; , ,
| | - Joris Pothof
- Department of Genetics, Erasmus University Medical Center, Postbus 2040, 3000 CA, Rotterdam, The Netherlands; , ,
| |
Collapse
|
98
|
Keane M, Semeiks J, Webb AE, Li YI, Quesada V, Craig T, Madsen LB, van Dam S, Brawand D, Marques PI, Michalak P, Kang L, Bhak J, Yim HS, Grishin NV, Nielsen NH, Heide-Jørgensen MP, Oziolor EM, Matson CW, Church GM, Stuart GW, Patton JC, George JC, Suydam R, Larsen K, López-Otín C, O'Connell MJ, Bickham JW, Thomsen B, de Magalhães JP. Insights into the evolution of longevity from the bowhead whale genome. Cell Rep 2015; 10:112-22. [PMID: 25565328 PMCID: PMC4536333 DOI: 10.1016/j.celrep.2014.12.008] [Citation(s) in RCA: 243] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Revised: 11/21/2014] [Accepted: 12/03/2014] [Indexed: 01/01/2023] Open
Abstract
The bowhead whale (Balaena mysticetus) is estimated to live over 200 years and is possibly the longest-living mammal. These animals should possess protective molecular adaptations relevant to age-related diseases, particularly cancer. Here, we report the sequencing and comparative analysis of the bowhead whale genome and two transcriptomes from different populations. Our analysis identifies genes under positive selection and bowhead-specific mutations in genes linked to cancer and aging. In addition, we identify gene gain and loss involving genes associated with DNA repair, cell-cycle regulation, cancer, and aging. Our results expand our understanding of the evolution of mammalian longevity and suggest possible players involved in adaptive genetic changes conferring cancer resistance. We also found potentially relevant changes in genes related to additional processes, including thermoregulation, sensory perception, dietary adaptations, and immune response. Our data are made available online (http://www.bowhead-whale.org) to facilitate research in this long-lived species.
Collapse
Affiliation(s)
- Michael Keane
- Integrative Genomics of Ageing Group, Institute of Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | - Jeremy Semeiks
- Howard Hughes Medical Institute and Departments of Biophysics and Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390-9050, USA
| | - Andrew E Webb
- Bioinformatics and Molecular Evolution Group, School of Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Yang I Li
- MRC Functional Genomics Unit, University of Oxford, Oxford OX1 3QX, UK
| | - Víctor Quesada
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, 33006 Oviedo, Spain
| | - Thomas Craig
- Integrative Genomics of Ageing Group, Institute of Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | - Lone Bruhn Madsen
- Department of Molecular Biology and Genetics, Aarhus University, 8830 Tjele, Denmark
| | - Sipko van Dam
- Integrative Genomics of Ageing Group, Institute of Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | - David Brawand
- MRC Functional Genomics Unit, University of Oxford, Oxford OX1 3QX, UK
| | - Patrícia I Marques
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, 33006 Oviedo, Spain
| | - Pawel Michalak
- Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, VA 24061, USA
| | - Lin Kang
- Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, VA 24061, USA
| | - Jong Bhak
- Personal Genomics Institute, Genome Research Foundation, Suwon 443-270, Republic of Korea
| | - Hyung-Soon Yim
- KIOST, Korea Institute of Ocean Science and Technology, Ansan 426-744, Republic of Korea
| | - Nick V Grishin
- Howard Hughes Medical Institute and Departments of Biophysics and Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390-9050, USA
| | | | | | - Elias M Oziolor
- Department of Environmental Science, Center for Reservoir and Aquatic Systems Research (CRASR) and Institute for Biomedical Studies, Baylor University, Waco, TX 76798, USA
| | - Cole W Matson
- Department of Environmental Science, Center for Reservoir and Aquatic Systems Research (CRASR) and Institute for Biomedical Studies, Baylor University, Waco, TX 76798, USA
| | - George M Church
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Gary W Stuart
- The Center for Genomic Advocacy (TCGA) and Department of Biology, Indiana State University, Terre Haute, IN 47809, USA
| | - John C Patton
- Department of Forestry and Natural Resources, Purdue University, West Lafayette, IN 47907, USA
| | - J Craig George
- North Slope Borough, Department of Wildlife Management, Barrow, AK 99723, USA
| | - Robert Suydam
- North Slope Borough, Department of Wildlife Management, Barrow, AK 99723, USA
| | - Knud Larsen
- Department of Molecular Biology and Genetics, Aarhus University, 8830 Tjele, Denmark
| | - Carlos López-Otín
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, 33006 Oviedo, Spain
| | - Mary J O'Connell
- Bioinformatics and Molecular Evolution Group, School of Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - John W Bickham
- Battelle Memorial Institute, Houston, TX 77079, USA; Department of Wildlife and Fisheries Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Bo Thomsen
- Department of Molecular Biology and Genetics, Aarhus University, 8830 Tjele, Denmark
| | - João Pedro de Magalhães
- Integrative Genomics of Ageing Group, Institute of Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK.
| |
Collapse
|
99
|
Genome Instability in Development and Aging: Insights from Nucleotide Excision Repair in Humans, Mice, and Worms. Biomolecules 2015; 5:1855-69. [PMID: 26287260 PMCID: PMC4598778 DOI: 10.3390/biom5031855] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 08/06/2015] [Accepted: 08/07/2015] [Indexed: 12/12/2022] Open
Abstract
DNA damage causally contributes to aging and cancer. Congenital defects in nucleotide excision repair (NER) lead to distinct cancer-prone and premature aging syndromes. The genetics of NER mutations have provided important insights into the distinct consequences of genome instability. Recent work in mice and C. elegans has shed new light on the mechanisms through which developing and aging animals respond to persistent DNA damage. The various NER mouse mutants have served as important disease models for Xeroderma pigmentosum (XP), Cockayne syndrome (CS), and trichothiodystrophy (TTD), while the traceable genetics of C. elegans have allowed the mechanistic delineation of the distinct outcomes of genome instability in metazoan development and aging. Intriguingly, highly conserved longevity assurance mechanisms respond to transcription-blocking DNA lesions in mammals as well as in worms and counteract the detrimental consequences of persistent DNA damage. The insulin-like growth factor signaling (IIS) effector transcription factor DAF-16 could indeed overcome DNA damage-driven developmental growth delay and functional deterioration even when DNA damage persists. Longevity assurance mechanisms might thus delay DNA damage-driven aging by raising the threshold when accumulating DNA damage becomes detrimental for physiological tissue functioning.
Collapse
|
100
|
Phosphodiesterase 1 regulation is a key mechanism in vascular aging. Clin Sci (Lond) 2015; 129:1061-75. [PMID: 26464516 DOI: 10.1042/cs20140753] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 06/25/2015] [Indexed: 12/31/2022]
Abstract
Reduced nitric oxide (NO)/cGMP signalling is observed in age-related vascular disease. We hypothesize that this disturbed signalling involves effects of genomic instability, a primary causal factor in aging, on vascular smooth muscle cells (VSMCs) and that the underlying mechanism plays a role in human age-related vascular disease. To test our hypothesis, we combined experiments in mice with genomic instability resulting from the defective nucleotide excision repair gene ERCC1 (Ercc1(d/-) mice), human VSMC cultures and population genome-wide association studies (GWAS). Aortic rings of Ercc1(d/-) mice showed 43% reduced responses to the soluble guanylate cyclase (sGC) stimulator sodium nitroprusside (SNP). Inhibition of phosphodiesterase (PDE) 1 and 5 normalized SNP-relaxing effects in Ercc1(d/-) to wild-type (WT) levels. PDE1C levels were increased in lung and aorta. cGMP hydrolysis by PDE in lungs was higher in Ercc1(d/-) mice. No differences in activity or levels of cGMP-dependent protein kinase 1 or sGC were observed in Ercc1(d/-) mice compared with WT. Senescent human VSMC showed elevated PDE1A and PDE1C and PDE5 mRNA levels (11.6-, 9- and 2.3-fold respectively), which associated with markers of cellular senescence. Conversely, PDE1 inhibition lowered expression of these markers. Human genetic studies revealed significant associations of PDE1A single nucleotide polymorphisms with diastolic blood pressure (DBP; β=0.28, P=2.47×10(-5)) and carotid intima-media thickness (cIMT; β=-0.0061, P=2.89×10(-5)). In summary, these results show that genomic instability and cellular senescence in VSMCs increase PDE1 expression. This might play a role in aging-related loss of vasodilator function, VSMC senescence, increased blood pressure and vascular hypertrophy.
Collapse
|