1
|
Radak M, Fallahi H. Zbp1 gene: a modulator of multiple aging hallmarks as potential therapeutic target for age-related diseases. Biogerontology 2023; 24:831-844. [PMID: 37199888 DOI: 10.1007/s10522-023-10039-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 05/07/2023] [Indexed: 05/19/2023]
Abstract
The Zbp1 gene has recently emerged as a potential therapeutic target for age-related diseases. Multiple studies have reported that Zbp1 plays a key role in regulating several aging hallmarks, including cellular senescence, chronic inflammation, DNA damage response, and mitochondrial dysfunction. Regarding cellular senescence, Zbp1 appears to regulate the onset and progression of senescence by controlling the expression of key markers such as p16INK4a and p21CIP1/WAF1. Similarly, evidence suggests that Zbp1 plays a role in regulating inflammation by promoting the production of pro-inflammatory cytokines, such as IL-6 and IL-1β, through activation of the NLRP3 inflammasome. Furthermore, Zbp1 seems to be involved in the DNA damage response, coordinating the cellular response to DNA damage by regulating the expression of genes such as p53 and ATM. Additionally, Zbp1 appears to regulate mitochondrial function, which is crucial for energy production and cellular homeostasis. Given the involvement of Zbp1 in multiple aging hallmarks, targeting this gene represents a potential strategy to prevent or treat age-related diseases. For example, inhibiting Zbp1 activity could be a promising approach to reduce cellular senescence and chronic inflammation, two critical hallmarks of aging associated with various age-related diseases. Similarly, modulating Zbp1 expression or activity could also improve DNA damage response and mitochondrial function, thus delaying or preventing the development of age-related diseases. Overall, the Zbp1 gene appears to be a promising therapeutic target for age-related diseases. In the current review, we have discussed the molecular mechanisms underlying the involvement of Zbp1 in aging hallmarks and proposed to develop effective strategies to target this gene for therapeutic purposes.
Collapse
Affiliation(s)
- Mehran Radak
- Department of Biology, School of Sciences, Razi University, Baq-e-Abrisham, Kermanshah, 6714967346, Islamic Republic of Iran
| | - Hossein Fallahi
- Department of Biology, School of Sciences, Razi University, Baq-e-Abrisham, Kermanshah, 6714967346, Islamic Republic of Iran.
| |
Collapse
|
2
|
Liang A, Fang Y, Ye L, Meng J, Wang X, Chen J, Xu X. Signaling pathways in hair aging. Front Cell Dev Biol 2023; 11:1278278. [PMID: 38033857 PMCID: PMC10687558 DOI: 10.3389/fcell.2023.1278278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 11/06/2023] [Indexed: 12/02/2023] Open
Abstract
Hair follicle (HF) homeostasis is regulated by various signaling pathways. Disruption of such homeostasis leads to HF disorders, such as alopecia, pigment loss, and hair aging, which is causing severe health problems and aesthetic concerns. Among these disorders, hair aging is characterized by hair graying, hair loss, hair follicle miniaturization (HFM), and structural changes to the hair shaft. Hair aging occurs under physiological conditions, while premature hair aging is often associated with certain pathological conditions. Numerous investigations have been made to determine the mechanisms and explore treatments to prevent hair aging. The most well-known hypotheses about hair aging include oxidative stress, hormonal disorders, inflammation, as well as DNA damage and repair defects. Ultimately, these factors pose threats to HF cells, especially stem cells such as hair follicle stem cells, melanocyte stem cells, and mesenchymal stem cells, which hamper hair regeneration and pigmentation. Here, we summarize previous studies investigating the above mechanisms and the existing therapeutic methods for hair aging. We also provide insights into hair aging research and discuss the limitations and outlook.
Collapse
Affiliation(s)
- Aishi Liang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Yingshan Fang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Lan Ye
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Jianda Meng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Xusheng Wang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Jinsong Chen
- Endocrinology Department, First People’s Hospital of Foshan, Foshan, China
| | - Xuejuan Xu
- Endocrinology Department, First People’s Hospital of Foshan, Foshan, China
| |
Collapse
|
3
|
A Validation Study on Immunophenotypic Differences in T-lymphocyte Chromosomal Radiosensitivity between Newborns and Adults in South Africa. RADIATION 2021. [DOI: 10.3390/radiation2010001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Children have an increased risk of developing radiation-induced secondary malignancies compared to adults, due to their high radiosensitivity and longer life expectancy. In contrast to the epidemiological evidence, there is only a handful of radiobiology studies which investigate the difference in radiosensitivity between children and adults at a cellular level. In this study, the previous results on the potential age dependency in chromosomal radiosensitivity were validated again by means of the cytokinesis-block micronucleus (CBMN) assay in T-lymphocytes isolated from the umbilical cord and adult peripheral blood of a South African population. The isolated cells were irradiated with 60Co γ-rays at doses ranging from 0.5 Gy to 4 Gy. Increased radiosensitivities of 34%, 42%, 29%, 26% and 16% were observed for newborns compared to adults at 0.5, 1, 2, 3 and 4 Gy, respectively. An immunophenotypic evaluation with flow cytometry revealed a significant change in the fraction of naïve (CD45RA+) T-lymphocytes in CD4+ and CD8+ T-lymphocytes with age. Newborns co-expressed an average of 91.05% CD45RA+ (range: 80.80–98.40%) of their CD4+ cells, while this fraction decreased to an average of 39.08% (range: 12.70–58.90%) for adults. Similar observations were made for CD8+ cells. This agrees with previous published results that the observed differences in chromosomal radiosensitivity between newborn and adult T-lymphocytes could potentially be linked to their immunophenotypic profiles.
Collapse
|
4
|
Rübe CE, Bäumert C, Schuler N, Isermann A, Schmal Z, Glanemann M, Mann C, Scherthan H. Human skin aging is associated with increased expression of the histone variant H2A.J in the epidermis. NPJ Aging Mech Dis 2021; 7:7. [PMID: 33795696 PMCID: PMC8016850 DOI: 10.1038/s41514-021-00060-z] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 01/26/2021] [Indexed: 01/08/2023] Open
Abstract
Cellular senescence is an irreversible growth arrest that occurs as a result of damaging stimuli, including DNA damage and/or telomere shortening. Here, we investigate histone variant H2A.J as a new biomarker to detect senescent cells during human skin aging. Skin biopsies from healthy volunteers of different ages (18–90 years) were analyzed for H2A.J expression and other parameters involved in triggering and/or maintaining cellular senescence. In the epidermis, the proportions of H2A.J-expressing keratinocytes increased from ≈20% in young to ≈60% in aged skin. Inverse correlations between Ki67- and H2A.J staining in germinative layers may reflect that H2A.J-expressing cells having lost their capacity to divide. As cellular senescence is triggered by DNA-damage signals, persistent 53BP1-foci, telomere lengths, and telomere-associated damage foci were analyzed in epidermal keratinocytes. Only slight age-related telomere attrition and few persistent nuclear 53BP1-foci, occasionally colocalizing with telomeres, suggest that unprotected telomeres are not a significant cause of senescence during skin aging. Quantification of integrin-α6+ basal cells suggests that the number and function of stem/progenitor cells decreased during aging and their altered proliferation capacities resulted in diminished tissue renewal with epidermal thinning. Collectively, our findings suggest that H2A.J is a sensitive marker of epidermal aging in human skin.
Collapse
Affiliation(s)
- Claudia E Rübe
- Saarland University Hospital, Department of Radiation Oncology, Homburg/Saar, Germany.
| | - Caroline Bäumert
- Saarland University Hospital, Department of Radiation Oncology, Homburg/Saar, Germany
| | - Nadine Schuler
- Saarland University Hospital, Department of Radiation Oncology, Homburg/Saar, Germany
| | - Anna Isermann
- Saarland University Hospital, Department of Radiation Oncology, Homburg/Saar, Germany
| | - Zoé Schmal
- Saarland University Hospital, Department of Radiation Oncology, Homburg/Saar, Germany
| | - Matthias Glanemann
- Saarland University Hospital, Department of Visceral Surgery, Homburg/Saar, Germany
| | - Carl Mann
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette Cedex, France
| | - Harry Scherthan
- Bundeswehr Inst. of Radiobiology affiliated to the Univ. of Ulm, München, Germany
| |
Collapse
|
5
|
DNA damage in aging, the stem cell perspective. Hum Genet 2019; 139:309-331. [PMID: 31324975 DOI: 10.1007/s00439-019-02047-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 07/05/2019] [Indexed: 02/07/2023]
Abstract
DNA damage is one of the most consistent cellular process proposed to contribute to aging. The maintenance of genomic and epigenomic integrity is critical for proper function of cells and tissues throughout life, and this homeostasis is under constant strain from both extrinsic and intrinsic insults. Considering the relationship between lifespan and genotoxic burden, it is plausible that the longest-lived cellular populations would face an accumulation of DNA damage over time. Tissue-specific stem cells are multipotent populations residing in localized niches and are responsible for maintaining all lineages of their resident tissue/system throughout life. However, many of these stem cells are impacted by genotoxic stress. Several factors may dictate the specific stem cell population response to DNA damage, including the niche location, life history, and fate decisions after damage accrual. This leads to differential handling of DNA damage in different stem cell compartments. Given the importance of adult stem cells in preserving normal tissue function during an individual's lifetime, DNA damage sensitivity and accumulation in these compartments could have crucial implications for aging. Despite this, more support for direct functional effects driven by accumulated DNA damage in adult stem cell compartments is needed. This review will present current evidence for the accumulation and potential influence of DNA damage in adult tissue-specific stem cells and propose inquiry directions that could benefit individual healthspan.
Collapse
|
6
|
Mani C, Reddy PH, Palle K. DNA repair fidelity in stem cell maintenance, health, and disease. Biochim Biophys Acta Mol Basis Dis 2019; 1866:165444. [PMID: 30953688 DOI: 10.1016/j.bbadis.2019.03.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 12/20/2018] [Accepted: 01/06/2019] [Indexed: 12/13/2022]
Abstract
Stem cells are a sub population of cell types that form the foundation of our body, and have the potential to replicate, replenish and repair limitlessly to maintain the tissue and organ homeostasis. Increased lifetime and frequent replication set them vulnerable for both exogenous and endogenous agents-induced DNA damage compared to normal cells. To counter these damages and preserve genetic information, stem cells have evolved with various DNA damage response and repair mechanisms. Furthermore, upon experiencing irreparable DNA damage, stem cells mostly prefer early senescence or apoptosis to avoid the accumulation of damages. However, the failure of these mechanisms leads to various diseases, including cancer. Especially, given the importance of stem cells in early development, DNA repair deficiency in stem cells leads to various disabilities like developmental delay, premature aging, sensitivity to DNA damaging agents, degenerative diseases, etc. In this review, we have summarized the recent update about how DNA repair mechanisms are regulated in stem cells and their association with disease progression and pathogenesis.
Collapse
Affiliation(s)
- Chinnadurai Mani
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Centre, Lubbock, TX 79430, United States of America
| | - P Hemachandra Reddy
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Centre, Lubbock, TX 79430, United States of America
| | - Komaraiah Palle
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Centre, Lubbock, TX 79430, United States of America.
| |
Collapse
|
7
|
Schuler N, Timm S, Rübe CE. Hair Follicle Stem Cell Faith Is Dependent on Chromatin Remodeling Capacity Following Low-Dose Radiation. Stem Cells 2018; 36:574-588. [PMID: 29282803 DOI: 10.1002/stem.2768] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 11/21/2017] [Accepted: 12/03/2017] [Indexed: 12/17/2022]
Abstract
The main function of the skin, to protect against the environment, is supported by the activity of different stem cell populations. The main focus of this study was elucidating the coping mechanisms of stem cells against the stimulation of constant exposure to genotoxic stresses, both endogenous and exogenous, to ensure long-term function. Investigation of various mouse strains, differing in their DNA repair capacity, enables us to clarify fractionated low-dose irradiation (LDR)-induced consequences for different stem cell populations of the murine hair follicle (HF) in their physiological stem cell niche. Using microscopic techniques combined with flow cytometry, we could show that LDR induces accumulation of persisting; pKu70-independent 53BP1-foci ("chromatin-alterations") in heterochromatic regions of the HF stem cells (HFSCs). These remaining chromatin-alterations result in varying stem cell consequences. CD34-positive HFSCs react by ataxia telangiectasia mutated-dependent, premature senescence, which correlates with global chromatin compaction, whereby apoptosis is prevented by the activity of DNA-dependent protein kinase catalytic subunit. However, distinctively highly damaged HFSCs seem to be sorted out of the niche by differentiation, transferring their chromatin-alterations to more proliferative G protein-coupled receptor 5-positive stem cells. Consequentially, the loss of basal HFSCs is compensated by increased proliferation within the stem cell pool. Despite the initial success of these mechanisms in stem cell population maintenance, the combined effect of the chromatin-alterations and the modification in stem cell pool composition may lead to downstream long-term functional loss of tissue or organs. Stem Cells 2018;36:574-588.
Collapse
Affiliation(s)
- Nadine Schuler
- Department of Radiation Oncology, Saarland University, Homburg/Saar, Germany
| | - Sara Timm
- Department of Radiation Oncology, Saarland University, Homburg/Saar, Germany
| | - Claudia E Rübe
- Department of Radiation Oncology, Saarland University, Homburg/Saar, Germany
| |
Collapse
|
8
|
König A, Zöller N, Kippenberger S, Bernd A, Kaufmann R, Layer PG, Heselich A. Non-thermal near-infrared exposure photobiomodulates cellular responses to ionizing radiation in human full thickness skin models. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2018; 178:115-123. [DOI: 10.1016/j.jphotobiol.2017.11.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 10/18/2017] [Accepted: 11/01/2017] [Indexed: 12/13/2022]
|
9
|
Contrepois K, Coudereau C, Benayoun BA, Schuler N, Roux PF, Bischof O, Courbeyrette R, Carvalho C, Thuret JY, Ma Z, Derbois C, Nevers MC, Volland H, Redon CE, Bonner WM, Deleuze JF, Wiel C, Bernard D, Snyder MP, Rübe CE, Olaso R, Fenaille F, Mann C. Histone variant H2A.J accumulates in senescent cells and promotes inflammatory gene expression. Nat Commun 2017; 8:14995. [PMID: 28489069 PMCID: PMC5436145 DOI: 10.1038/ncomms14995] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 02/20/2017] [Indexed: 01/02/2023] Open
Abstract
The senescence of mammalian cells is characterized by a proliferative arrest in response to stress and the expression of an inflammatory phenotype. Here we show that histone H2A.J, a poorly studied H2A variant found only in mammals, accumulates in human fibroblasts in senescence with persistent DNA damage. H2A.J also accumulates in mice with aging in a tissue-specific manner and in human skin. Knock-down of H2A.J inhibits the expression of inflammatory genes that contribute to the senescent-associated secretory phenotype (SASP), and over expression of H2A.J increases the expression of some of these genes in proliferating cells. H2A.J accumulation may thus promote the signalling of senescent cells to the immune system, and it may contribute to chronic inflammation and the development of aging-associated diseases.
Collapse
Affiliation(s)
- Kévin Contrepois
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 91198 Gif-sur-Yvette cedex, France
- Department of Genetics, Stanford University, Stanford, California 94305-5120, USA
| | - Clément Coudereau
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 91198 Gif-sur-Yvette cedex, France
| | - Bérénice A. Benayoun
- Department of Genetics, Stanford University, Stanford, California 94305-5120, USA
- Paul F. Glenn Laboratories for the Biology of Aging, Stanford University, Stanford, California 94305-5120, USA
| | - Nadine Schuler
- Department of Radiation Oncology, Saarland University, 66421 Homburg (Saar), Germany
| | - Pierre-François Roux
- Institut Pasteur/INSERM U933, Laboratory of Nuclear Organization and Oncogenesis, Department of Cell Biology and Infection, 75015 Paris, France
| | - Oliver Bischof
- Institut Pasteur/INSERM U933, Laboratory of Nuclear Organization and Oncogenesis, Department of Cell Biology and Infection, 75015 Paris, France
| | - Régis Courbeyrette
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 91198 Gif-sur-Yvette cedex, France
| | - Cyril Carvalho
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 91198 Gif-sur-Yvette cedex, France
| | - Jean-Yves Thuret
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 91198 Gif-sur-Yvette cedex, France
| | - Zhihai Ma
- Department of Genetics, Stanford University, Stanford, California 94305-5120, USA
| | | | - Marie-Claire Nevers
- CEA, Service de Pharmacologie et Immunoanalyse (SPI), INRA, Université Paris-Saclay, F-91191 Gif-sur-Yvette, France
| | - Hervé Volland
- CEA, Service de Pharmacologie et Immunoanalyse (SPI), INRA, Université Paris-Saclay, F-91191 Gif-sur-Yvette, France
| | - Christophe E. Redon
- Laboratory of Molecular Pharmacology, C.C.R., N.C.I., N.I.H., Bethesda, Maryland 20892, USA
| | - William M. Bonner
- Laboratory of Molecular Pharmacology, C.C.R., N.C.I., N.I.H., Bethesda, Maryland 20892, USA
| | | | - Clotilde Wiel
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, CNRS UMR5286, Centre Léon Bérard, Université de Lyon 69008, Lyon, France
| | - David Bernard
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, CNRS UMR5286, Centre Léon Bérard, Université de Lyon 69008, Lyon, France
| | - Michael P. Snyder
- Department of Genetics, Stanford University, Stanford, California 94305-5120, USA
| | - Claudia E. Rübe
- Department of Radiation Oncology, Saarland University, 66421 Homburg (Saar), Germany
| | | | - François Fenaille
- CEA, IBITECS, Service de Pharmacologie et d'Immunoanalyse, UMR 0496, Laboratoire d'Etude du Métabolisme des Médicaments, MetaboHUB-Paris, Université Paris Saclay, F-91191 Gif-sur-Yvette cedex, France
| | - Carl Mann
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 91198 Gif-sur-Yvette cedex, France
| |
Collapse
|
10
|
Revenco T, Lapouge G, Moers V, Brohée S, Sotiropoulou PA. Low Dose Radiation Causes Skin Cancer in Mice and Has a Differential Effect on Distinct Epidermal Stem Cells. Stem Cells 2017; 35:1355-1364. [PMID: 28100039 DOI: 10.1002/stem.2571] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 01/02/2017] [Indexed: 12/22/2022]
Abstract
The carcinogenic effect of ionizing radiation has been evaluated based on limited populations accidently exposed to high dose radiation. In contrast, insufficient data are available on the effect of low dose radiation (LDR), such as radiation deriving from medical investigations and interventions, as well as occupational exposure that concern a large fraction of western populations. Using mouse skin epidermis as a model, we showed that LDR results in DNA damage in sebaceous gland (SG) and bulge epidermal stem cells (SCs). While the first commit apoptosis upon low dose irradiation, the latter survive. Bulge SC survival coincides with higher HIF-1α expression and a metabolic switch upon LDR. Knocking down HIF-1α sensitizes bulge SCs to LDR-induced apoptosis, while upregulation of HIF-1α in the epidermis, including SG SCs, rescues cell death. Most importantly, we show that LDR results in cancer formation with full penetrance in the radiation-sensitive Patched1 heterozygous mice. Overall, our results demonstrate for the first time that LDR can be a potent carcinogen in individuals predisposed to cancer. Stem Cells 2017;35:1355-1364.
Collapse
Affiliation(s)
| | - Gaelle Lapouge
- IRIBHM, Université Libre de Bruxelles, Brussels, Belgium
| | - Virginie Moers
- IRIBHM, Université Libre de Bruxelles, Brussels, Belgium
| | - Sylvain Brohée
- IRIBHM, Université Libre de Bruxelles, Brussels, Belgium
| | | |
Collapse
|
11
|
Waaijer MEC, Croco E, Westendorp RGJ, Slagboom PE, Sedivy JM, Lorenzini A, Maier AB. DNA damage markers in dermal fibroblasts in vitro reflect chronological donor age. Aging (Albany NY) 2016; 8:147-57. [PMID: 26830451 PMCID: PMC4761719 DOI: 10.18632/aging.100890] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The aging process is accompanied by an accumulation of cellular damage, which compromises the viability and function of cells and tissues. We aim to further explore the association between in vitro DNA damage markers and the chronological age of the donor, as well as long-lived family membership and presence of cardiovascular diseases. Therefore, numbers of 53BP1 foci, telomere-associated foci (TAF) and micronuclei were measured in cultured dermal fibroblasts obtained from three age groups of donors (mean age 22, 63 and 90 years). Fibroblasts were cultured without a stressor and with 0.6 μM rotenone for 3 days. We found that 53BP1 foci and TAF were more frequently present in fibroblasts of old donors compared to middle-aged and young donors. No association between micronuclei and donor age was found. Within the fibroblasts of the middle-aged donors we did not find associations between DNA damage markers and long-lived family membership or cardiovascular disease. Results were comparable when fibroblasts were stressed in vitro with rotenone. In conclusion, we found that DNA damage foci of cultured fibroblasts are significantly associated with the chronological age, but not biological age, of the donor.
Collapse
Affiliation(s)
- Mariëtte E C Waaijer
- Department of Gerontology and Geriatrics, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Eleonora Croco
- Department for Life Quality Studies, University of Bologna, 40126 Bologna, Italy
| | - Rudi G J Westendorp
- Department of Public Health and Center for Healthy Aging, Faculty of Health and Medical Sciences, University of Copenhagen, 1123 Copenhagen, Denmark
| | - P Eline Slagboom
- Department of Molecular Epidemiology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands.,Netherlands Consortium for Healthy Aging, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - John M Sedivy
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI 02903, USA
| | - Antonello Lorenzini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
| | - Andrea B Maier
- Department of Internal Medicine, Section of Gerontology and Geriatrics, VU University Medical Center, 1007 MB Amsterdam, The Netherlands.,Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville VIC 3050, Australia
| |
Collapse
|
12
|
Moehrle BM, Geiger H. Aging of hematopoietic stem cells: DNA damage and mutations? Exp Hematol 2016; 44:895-901. [PMID: 27402537 DOI: 10.1016/j.exphem.2016.06.253] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 06/27/2016] [Indexed: 12/15/2022]
Abstract
Aging in the hematopoietic system and the stem cell niche contributes to aging-associated phenotypes of hematopoietic stem cells (HSCs), including leukemia and aging-associated immune remodeling. Among others, the DNA damage theory of aging of HSCs is well established, based on the detection of a significantly larger amount of γH2AX foci and a higher tail moment in the comet assay, both initially thought to be associated with DNA damage in aged HSCs compared with young cells, and bone marrow failure in animals devoid of DNA repair factors. Novel data on the increase in and nature of DNA mutations in the hematopoietic system with age, the quality of the DNA damage response in aged HSCs, and the nature of γH2AX foci question a direct link between DNA damage and the DNA damage response and aging of HSCs, and rather favor changes in epigenetics, splicing-factors or three-dimensional architecture of the cell as major cell intrinsic factors of HSCs aging. Aging of HSCs is also driven by a strong contribution of aging of the niche. This review discusses the DNA damage theory of HSC aging in the light of these novel mechanisms of aging of HSCs.
Collapse
Affiliation(s)
| | - Hartmut Geiger
- Institute for Molecular Medicine, Ulm University, Ulm, Germany; Aging Research Center, Ulm University, Ulm, Germany; Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH.
| |
Collapse
|
13
|
Vandevoorde C, Vral A, Vandekerckhove B, Philippé J, Thierens H. Radiation Sensitivity of Human CD34+Cells Versus Peripheral Blood T Lymphocytes of Newborns and Adults: DNA Repair and Mutagenic Effects. Radiat Res 2016; 185:580-90. [DOI: 10.1667/rr14109.1] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
14
|
Ultraviolet Radiation-Induced Skin Aging: The Role of DNA Damage and Oxidative Stress in Epidermal Stem Cell Damage Mediated Skin Aging. Stem Cells Int 2016; 2016:7370642. [PMID: 27148370 PMCID: PMC4842382 DOI: 10.1155/2016/7370642] [Citation(s) in RCA: 198] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 03/14/2016] [Indexed: 12/11/2022] Open
Abstract
Skin is the largest human organ. Skin continually reconstructs itself to ensure its viability, integrity, and ability to provide protection for the body. Some areas of skin are continuously exposed to a variety of environmental stressors that can inflict direct and indirect damage to skin cell DNA. Skin homeostasis is maintained by mesenchymal stem cells in inner layer dermis and epidermal stem cells (ESCs) in the outer layer epidermis. Reduction of skin stem cell number and function has been linked to impaired skin homeostasis (e.g., skin premature aging and skin cancers). Skin stem cells, with self-renewal capability and multipotency, are frequently affected by environment. Ultraviolet radiation (UVR), a major cause of stem cell DNA damage, can contribute to depletion of stem cells (ESCs and mesenchymal stem cells) and damage of stem cell niche, eventually leading to photoinduced skin aging. In this review, we discuss the role of UV-induced DNA damage and oxidative stress in the skin stem cell aging in order to gain insights into the pathogenesis and develop a way to reduce photoaging of skin cells.
Collapse
|
15
|
Moehrle BM, Nattamai K, Brown A, Florian MC, Ryan M, Vogel M, Bliederhaeuser C, Soller K, Prows DR, Abdollahi A, Schleimer D, Walter D, Milsom MD, Stambrook P, Porteus M, Geiger H. Stem Cell-Specific Mechanisms Ensure Genomic Fidelity within HSCs and upon Aging of HSCs. Cell Rep 2015; 13:2412-2424. [PMID: 26686632 DOI: 10.1016/j.celrep.2015.11.030] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 08/13/2015] [Accepted: 11/08/2015] [Indexed: 01/22/2023] Open
Abstract
Whether aged hematopoietic stem and progenitor cells (HSPCs) have impaired DNA damage repair is controversial. Using a combination of DNA mutation indicator assays, we observe a 2- to 3-fold increase in the number of DNA mutations in the hematopoietic system upon aging. Young and aged hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs) do not show an increase in mutation upon irradiation-induced DNA damage repair, and young and aged HSPCs respond very similarly to DNA damage with respect to cell-cycle checkpoint activation and apoptosis. Both young and aged HSPCs show impaired activation of the DNA-damage-induced G1-S checkpoint. Induction of chronic DNA double-strand breaks by zinc-finger nucleases suggests that HSPCs undergo apoptosis rather than faulty repair. These data reveal a protective mechanism in both the young and aged hematopoietic system against accumulation of mutations in response to DNA damage.
Collapse
Affiliation(s)
- Bettina M Moehrle
- Institute of Molecular Medicine, University of Ulm, 89081 Ulm, Germany
| | - Kalpana Nattamai
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, OH 45229, USA
| | - Andreas Brown
- Institute of Molecular Medicine, University of Ulm, 89081 Ulm, Germany
| | - Maria C Florian
- Institute of Molecular Medicine, University of Ulm, 89081 Ulm, Germany
| | - Marnie Ryan
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, OH 45229, USA
| | - Mona Vogel
- Institute of Molecular Medicine, University of Ulm, 89081 Ulm, Germany
| | | | - Karin Soller
- Institute of Molecular Medicine, University of Ulm, 89081 Ulm, Germany
| | - Daniel R Prows
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, OH 45229, USA
| | - Amir Abdollahi
- German Cancer Consortium (DKTK), 69120 Heidelberg, Germany; Molecular and Translational Radiation Oncology, Heidelberg Ion Therapy Center (HIT), 69120 Heidelberg, Germany
| | - David Schleimer
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, OH 45229, USA
| | - Dagmar Walter
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine gGmbH (HI-STEM), 69120 Heidelberg, Germany
| | - Michael D Milsom
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine gGmbH (HI-STEM), 69120 Heidelberg, Germany; Deutsches Krebsforschungszentrum (DKFZ), Division of Stem Cells and Cancer, Experimental Hematology Group, 69120 Heidelberg, Germany
| | - Peter Stambrook
- Department of Molecular Genetics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Matthew Porteus
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
| | - Hartmut Geiger
- Institute of Molecular Medicine, University of Ulm, 89081 Ulm, Germany; Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, OH 45229, USA.
| |
Collapse
|
16
|
Bigot N, Mouche A, Preti M, Loisel S, Renoud ML, Le Guével R, Sensebé L, Tarte K, Pedeux R. Hypoxia Differentially Modulates the Genomic Stability of Clinical-Grade ADSCs and BM-MSCs in Long-Term Culture. Stem Cells 2015; 33:3608-20. [PMID: 26422646 DOI: 10.1002/stem.2195] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 08/02/2015] [Indexed: 12/12/2022]
Abstract
Long-term cultures under hypoxic conditions have been demonstrated to maintain the phenotype of mesenchymal stromal/stem cells (MSCs) and to prevent the emergence of senescence. According to several studies, hypoxia has frequently been reported to drive genomic instability in cancer cells and in MSCs by hindering the DNA damage response and DNA repair. Thus, we evaluated the occurrence of DNA damage and repair events during the ex vivo expansion of clinical-grade adipose-derived stromal cells (ADSCs) and bone marrow (BM)-derived MSCs cultured with platelet lysate under 21% (normoxia) or 1% (hypoxia) O2 conditions. Hypoxia did not impair cell survival after DNA damage, regardless of MSC origin. However, ADSCs, unlike BM-MSCs, displayed altered γH2AX signaling and increased ubiquitylated γH2AX levels under hypoxic conditions, indicating an impaired resolution of DNA damage-induced foci. Moreover, hypoxia specifically promoted BM-MSC DNA integrity, with increased Ku80, TP53BP1, BRCA1, and RAD51 expression levels and more efficient nonhomologous end joining and homologous recombination repair. We further observed that hypoxia favored mtDNA stability and maintenance of differentiation potential after genotoxic stress. We conclude that long-term cultures under 1% O2 were more suitable for BM-MSCs as suggested by improved genomic stability compared with ADSCs.
Collapse
Affiliation(s)
- Nicolas Bigot
- INSERM U917, Microenvironnement et Cancer, Rennes, France.,Université de Rennes 1, Rennes, France.,Etablissement Français du Sang Bretagne, Rennes, France
| | - Audrey Mouche
- INSERM U917, Microenvironnement et Cancer, Rennes, France.,Université de Rennes 1, Rennes, France.,Etablissement Français du Sang Bretagne, Rennes, France
| | - Milena Preti
- Etablissement Français du Sang Pyrénées Méditerranée, Toulouse, France.,Université Paul Sabatier, Toulouse, France.,UMR5273-INSERM U1031, Toulouse, France
| | - Séverine Loisel
- INSERM U917, Microenvironnement et Cancer, Rennes, France.,Université de Rennes 1, Rennes, France.,Etablissement Français du Sang Bretagne, Rennes, France
| | - Marie-Laure Renoud
- Etablissement Français du Sang Pyrénées Méditerranée, Toulouse, France.,Université Paul Sabatier, Toulouse, France.,UMR5273-INSERM U1031, Toulouse, France
| | - Rémy Le Guével
- Université de Rennes 1, Rennes, France.,ImPACcell, SFR Biosit, Université de Rennes 1, Rennes, France
| | - Luc Sensebé
- Etablissement Français du Sang Pyrénées Méditerranée, Toulouse, France.,Université Paul Sabatier, Toulouse, France.,UMR5273-INSERM U1031, Toulouse, France
| | - Karin Tarte
- INSERM U917, Microenvironnement et Cancer, Rennes, France.,Université de Rennes 1, Rennes, France.,Etablissement Français du Sang Bretagne, Rennes, France.,Service ITeCH, CHU Pontchaillou, Rennes, France
| | - Rémy Pedeux
- INSERM U917, Microenvironnement et Cancer, Rennes, France.,Université de Rennes 1, Rennes, France.,Etablissement Français du Sang Bretagne, Rennes, France
| |
Collapse
|
17
|
|
18
|
Suzuki E, Nishimatsu H, Homma Y. Stem cell therapy for erectile dysfunction. World J Clin Urol 2014; 3:272-282. [DOI: 10.5410/wjcu.v3.i3.272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 05/03/2014] [Accepted: 10/10/2014] [Indexed: 02/06/2023] Open
Abstract
Erectile dysfunction (ED) is an important health problem that has commonly been clinically treated using phosphodiesterase type 5 inhibitors (PDE5Is). However, PDE5Is are less effective when the structure of the cavernous body has been severely injured, and thus regeneration is required. Stem cell therapy has been investigated as a possible means for regenerating the injured cavernous body. Stem cells are classified into embryonic stem cells and adult stem cells (ASCs), and the intracavernous injection of ASCs has been explored as a therapy in animal ED models. Bone marrow-derived mesenchymal stem cells and adipose tissue-derived stem cells are major sources of ASCs used for the treatment of ED, and accumulated evidence now suggests that ASCs are useful in the restoration of erectile function and the regeneration of the cavernous body. However, the mechanisms by which ASCs recover erectile function remain controversial. Some studies indicated that ASCs were differentiated into the vascular endothelial cells, vascular smooth muscle cells, and nerve cells that originally resided in the cavernous body, whereas other studies have suggested that ASCs improved erectile function via the secretion of anti-apoptotic and/or proangiogenic cytokines rather than differentiation into other cell types. In this paper, we reviewed the characteristics of stem cells used for the treatment of ED, and the possible mechanisms by which these cells exert their effects. We also discussed the problems to be solved before implementation in the clinical setting.
Collapse
|
19
|
Vahidi Ferdousi L, Rocheteau P, Chayot R, Montagne B, Chaker Z, Flamant P, Tajbakhsh S, Ricchetti M. More efficient repair of DNA double-strand breaks in skeletal muscle stem cells compared to their committed progeny. Stem Cell Res 2014; 13:492-507. [PMID: 25262445 DOI: 10.1016/j.scr.2014.08.005] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Revised: 07/14/2014] [Accepted: 08/15/2014] [Indexed: 01/17/2023] Open
Abstract
The loss of genome integrity in adult stem cells results in accelerated tissue aging and is possibly cancerogenic. Adult stem cells in different tissues appear to react robustly to DNA damage. We report that adult skeletal stem (satellite) cells do not primarily respond to radiation-induced DNA double-strand breaks (DSBs) via differentiation and exhibit less apoptosis compared to other myogenic cells. Satellite cells repair these DNA lesions more efficiently than their committed progeny. Importantly, non-proliferating satellite cells and post-mitotic nuclei in the fiber exhibit dramatically distinct repair efficiencies. Altogether, reduction of the repair capacity appears to be more a function of differentiation than of the proliferation status of the muscle cell. Notably, satellite cells retain a high efficiency of DSB repair also when isolated from the natural niche. Finally, we show that repair of DSB substrates is not only very efficient but, surprisingly, also very accurate in satellite cells and that accurate repair depends on the key non-homologous end-joining factor DNA-PKcs.
Collapse
Affiliation(s)
- Leyla Vahidi Ferdousi
- Institut Pasteur, Yeast Molecular Genetics, Dept. of Genomes and Genetics, 25 rue du Dr. Roux, 75724 Paris Cedex 15, France; CNRS UMR 3525, Team Stability of Nuclear and Mitochondrial DNA, Paris, France; Sorbonne Universités, UPMC, University of Paris 06, IFD-ED 515, Place Jussieu, Paris, 72252, France
| | - Pierre Rocheteau
- Institut Pasteur, Stem Cells & Development, Dept. of Developmental and Stem Cell Biology, 25 rue du Dr. Roux, 75724 Paris Cedex 15, France; CNRS URA 2578, Paris, France
| | - Romain Chayot
- Institut Pasteur, Yeast Molecular Genetics, Dept. of Genomes and Genetics, 25 rue du Dr. Roux, 75724 Paris Cedex 15, France; CNRS UMR 3525, Team Stability of Nuclear and Mitochondrial DNA, Paris, France
| | - Benjamin Montagne
- Institut Pasteur, Yeast Molecular Genetics, Dept. of Genomes and Genetics, 25 rue du Dr. Roux, 75724 Paris Cedex 15, France; CNRS UMR 3525, Team Stability of Nuclear and Mitochondrial DNA, Paris, France
| | - Zayna Chaker
- Institut Pasteur, Yeast Molecular Genetics, Dept. of Genomes and Genetics, 25 rue du Dr. Roux, 75724 Paris Cedex 15, France; CNRS UMR 3525, Team Stability of Nuclear and Mitochondrial DNA, Paris, France
| | - Patricia Flamant
- Institut Pasteur, Stem Cells & Development, Dept. of Developmental and Stem Cell Biology, 25 rue du Dr. Roux, 75724 Paris Cedex 15, France; CNRS URA 2578, Paris, France
| | - Shahragim Tajbakhsh
- Institut Pasteur, Stem Cells & Development, Dept. of Developmental and Stem Cell Biology, 25 rue du Dr. Roux, 75724 Paris Cedex 15, France; CNRS URA 2578, Paris, France
| | - Miria Ricchetti
- Institut Pasteur, Yeast Molecular Genetics, Dept. of Genomes and Genetics, 25 rue du Dr. Roux, 75724 Paris Cedex 15, France; CNRS UMR 3525, Team Stability of Nuclear and Mitochondrial DNA, Paris, France.
| |
Collapse
|
20
|
Benavides M, Berciano-Guerrero M. Elderly patients with metastatic colorectal cancer: overall issues and first-line chemotherapy options. COLORECTAL CANCER 2013. [DOI: 10.2217/crc.13.69] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
SUMMARY The aging phenomenon is resulting in an ever greater incidence of colorectal cancer (CRC) in the elderly. Chronologic age is not the best or only way to define elderly patients because aging varies greatly. Comprehensive geriatric assessment has proved beneficial for more appropriate therapeutic options although its influence on treatment decisions and outcomes remains to be validated. Fit elderly patients with metastatic CRC derive similar benefits to their younger counterparts, but only one Phase III trial exists to define the best treatment. New strategies such as maintenance therapies, which are particularly appropriate in these patients, are needed. As very few data are available for the vulnerable/frail elderly population, it is important to better define these terms and the efficacy (if any) of treatment modalities in this group. Translational research in geriatric oncology must be improved in this heterogeneous population to identify biological and clinical correlates of cancer and aging, ameliorating personalized treatment in elderly metastatic CRC patients.
Collapse
Affiliation(s)
- Manuel Benavides
- Medical Oncology Department, Hospital Regional Universitario Carlos Haya, Málaga, Spain
| | | |
Collapse
|
21
|
Abstract
Technical improvements in electron microscopy, both instrumental and preparative, permit increasingly accurate analyses. Digital images for transmission electron microscopy (TEM) can be processed by software programs that automate tasks and create custom tools that allow for image enhancement for brightness, contrast and coloration; for creation of rectangular, ellipsoidal or irregular area selections; and for measurement of mean area and standard deviation. Sample preparation remains a source of error since organelles and spatial arrangements of macromolecules rapidly change after anoxia. Guidelines for maintaining consistency in preparation, examination and interpretation are presented for different electron microscopy (EM) modalities.
Collapse
Affiliation(s)
- N. F. Cheville
- Department of Veterinary Pathology, Iowa State University, Ames, IA, USA
| | - J. Stasko
- Microscopy Services, National Animal Disease Center, Ames, IA, USA
| |
Collapse
|