1
|
Zhao P, Chang J, Chen Y, Sun X, Ma X, Zhou C, Zhou L, Wang Y, Yang Y. Cellular Senescence-Related Long Non-coding RNA Signatures Predict Prognosis in Juvenile Osteosarcoma. PHENOMICS (CHAM, SWITZERLAND) 2024; 4:430-452. [PMID: 39723224 PMCID: PMC11666862 DOI: 10.1007/s43657-023-00132-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 08/29/2023] [Accepted: 09/01/2023] [Indexed: 12/28/2024]
Abstract
Osteosarcoma is the most common malignant bone tumor and is frequently diagnosed in juvenile. Cellular senescence is a fundamental hallmark of osteosarcoma and plays a vital role in the initiation and progression of aging and tumorigenesis. Long non-coding RNAs (lncRNAs) are implicated in tumorigenesis. In this study, six cellular senescence-related lncRNAs with independent prognostic significance in juvenile osteosarcoma patients were identified through univariate Cox regression analysis, least absolute shrinkage and selection operator (LASSO) regression analysis, and multivariate Cox regression analysis. Prognostic significance was further confirmed by Kaplan-Meier (KM) survival curves, co-expression interaction networks, and sankey diagrams. A prognostic model of cellular senescence-related genes in juvenile osteosarcoma patients was then constructed using multivariate Cox regression analysis based on these six genes. High- and low-risk groups were identified according to the median risk score calculated by the prognostic model. The favorable prognostic significance of this model was demonstrated through survival curves, receiver operating characteristic (ROC) curves, distribution scatter plots and lncRNA expression heatmaps. Furthermore, cellular senescence-related lncRNAs were validated by enrichment analysis, immunological correlation analysis, m6A correlation analysis, and drug sensitivity correlation analysis. These findings are important for improving the prognosis of juvenile osteosarcoma patients and understanding the mechanisms underlying cellular senescence in juvenile osteosarcoma development.
Collapse
Affiliation(s)
- Peng Zhao
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032 China
- Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai, 200032 China
| | - Junli Chang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032 China
- Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai, 200032 China
| | - YeKai Chen
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032 China
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032 China
| | - Xingyuan Sun
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032 China
- Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai, 200032 China
| | - Xiaoping Ma
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032 China
- Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai, 200032 China
| | - Chujie Zhou
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032 China
- Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai, 200032 China
| | - Lei Zhou
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032 China
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032 China
| | - Yongjun Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032 China
- Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai, 200032 China
| | - Yanping Yang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032 China
- Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai, 200032 China
| |
Collapse
|
2
|
Yamamoto Y, Takahashi RU, Kinehara M, Yano K, Kuramoto T, Shimamoto A, Tahara H. Downregulation of Histone H3.3 Induces p53-Dependent Cellular Senescence in Human Diploid Fibroblasts. Genes (Basel) 2024; 15:543. [PMID: 38790171 PMCID: PMC11121134 DOI: 10.3390/genes15050543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/19/2024] [Accepted: 04/22/2024] [Indexed: 05/26/2024] Open
Abstract
Cellular senescence is an irreversible growth arrest that acts as a barrier to cancer initiation and progression. Histone alteration is one of the major events during replicative senescence. However, little is known about the function of H3.3 in cellular senescence. Here we found that the downregulation of H3.3 induced growth suppression with senescence-like phenotypes such as senescence-associated heterochromatin foci (SAHF) and β-galactosidase (SA-β-gal) activity. Furthermore, H3.3 depletion induced senescence-like phenotypes with the p53/p21-depedent pathway. In addition, we identified miR-22-3p, tumor suppressive miRNA, as an upstream regulator of the H3F3B (H3 histone, family 3B) gene which is the histone variant H3.3 and replaces conventional H3 in active genes. Therefore, our results reveal for the first time the molecular mechanisms for cellular senescence which are regulated by H3.3 abundance. Taken together, our studies suggest that H3.3 exerts functional roles in regulating cellular senescence and is a promising target for cancer therapy.
Collapse
Affiliation(s)
- Yuki Yamamoto
- Department of Cellular and Molecular Biology, Basic Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan; (Y.Y.); (R.-u.T.)
| | - Ryou-u Takahashi
- Department of Cellular and Molecular Biology, Basic Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan; (Y.Y.); (R.-u.T.)
| | - Masaki Kinehara
- Department of Cellular and Molecular Biology, Basic Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan; (Y.Y.); (R.-u.T.)
| | - Kimiyoshi Yano
- Laboratory of Genome Stress Signaling, National Cancer Center Research Institute, Tokyo 104-0045, Japan;
| | - Tatsuya Kuramoto
- Department of Cellular and Molecular Biology, Basic Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan; (Y.Y.); (R.-u.T.)
| | - Akira Shimamoto
- Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Sanyo Onoda 756-0884, Japan;
| | - Hidetoshi Tahara
- Department of Cellular and Molecular Biology, Basic Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan; (Y.Y.); (R.-u.T.)
| |
Collapse
|
3
|
Wang X, Yang J, Li D, Teng L, Chen Y, Meng J, Yang C, Yin Z, Li C. Pazopanib stimulates senescence of renal carcinoma cells through targeting nuclear factor E2-related factor 2 (Nrf2). J Biochem Mol Toxicol 2024; 38:e23689. [PMID: 38613465 DOI: 10.1002/jbt.23689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 02/16/2024] [Accepted: 03/07/2024] [Indexed: 04/15/2024]
Abstract
Renal cell carcinoma (RCC) is the most common kidney cancer with high mortality rate. Pazopanib has been approved for the treatment of RCC. However, the underlying mechanism is not clear. Here, we report a novel finding by showing that treatment with Pazopanib could promote cellular senescence of the human RCC cell line ACHN. Cells were stimulated with 5, 10, and 20 μM Pazopanib, respectively. Cellular senescence was measured using senescence-associated β-galactosidase (SA-β-Gal) staining. Western blot analysis and real-time polymerase chain reaction were used to measure the mRNA and protein expression of nuclear factor E2-related factor 2 (Nrf2), γH2AX, human telomerase reverse transcriptase (hTERT), telomeric repeat binding factor 2 (TERF2), p53 and plasminogen activator inhibitor (PAI). First, we found that exposure to Pazopanib reduced the cell viability of ACHN cells. Additionally, Pazopanib induced oxidative stress by increasing the production of reactive oxygen species, reducing the levels of glutathione peroxidase, and promoting nuclear translocation of Nrf2. Interestingly, Pazopanib exposure resulted in DNA damage by increasing the expression of γH2AX. Importantly, Pazopanib increased cellular senescence and reduced telomerase activity. Pazopanib also reduced the gene expression of hTERT but increased the gene expression of TERF2. Correspondingly, we found that Pazopanib increased the expression of p53 and PAI at both the mRNA and protein levels. To elucidate the underlying mechanism, the expression of Nrf2 was knocked down by transduction with Ad- Nrf2 shRNA. Results indicate that silencing of Nrf2 in ACHN cells abolished the effects of Pazopanib in stimulating cellular senescence and reducing telomerase activity. Consistently, knockdown of Nrf2 restored the expression of p53 and PAI in ACHN cells. Based on these results, we explored a novel mechanism whereby which Pazopanib displays a cytotoxicity effect in RCC cells through promoting cellular senescence mediated by Nrf2.
Collapse
Affiliation(s)
- Xingyuan Wang
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Jing Yang
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Dechao Li
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Lichen Teng
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yongsheng Chen
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Jie Meng
- Department of Orthognathic Surgery, Heilongjiang Provincial Hospital, Harbin, China
| | - Chen Yang
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Zhihao Yin
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Changfu Li
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
4
|
Jing X, Jia S, Teng M, Day BW, Afolayan AJ, Jarzembowski JA, Lin CW, Hessner MJ, Pritchard KA, Naylor S, Konduri GG, Teng RJ. Cellular Senescence Contributes to the Progression of Hyperoxic Bronchopulmonary Dysplasia. Am J Respir Cell Mol Biol 2024; 70:94-109. [PMID: 37874230 DOI: 10.1165/rcmb.2023-0038oc] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 10/23/2023] [Indexed: 10/25/2023] Open
Abstract
Oxidative stress, inflammation, and endoplasmic reticulum (ER) stress sequentially occur in bronchopulmonary dysplasia (BPD), and all result in DNA damage. When DNA damage becomes irreparable, tumor suppressors increase, followed by apoptosis or senescence. Although cellular senescence contributes to wound healing, its persistence inhibits growth. Therefore, we hypothesized that cellular senescence contributes to BPD progression. Human autopsy lungs were obtained. Sprague-Dawley rat pups exposed to 95% oxygen between Postnatal Day 1 (P1) and P10 were used as the BPD phenotype. N-acetyl-lysyltyrosylcysteine-amide (KYC), tauroursodeoxycholic acid (TUDCA), and Foxo4 dri were administered intraperitoneally to mitigate myeloperoxidase oxidant generation, ER stress, and cellular senescence, respectively. Lungs were examined by histology, transcriptomics, and immunoblotting. Cellular senescence increased in rat and human BPD lungs, as evidenced by increased oxidative DNA damage, tumor suppressors, GL-13 stain, and inflammatory cytokines with decreased cell proliferation and lamin B expression. Cellular senescence-related transcripts in BPD rat lungs were enriched at P10 and P21. Single-cell RNA sequencing showed increased cellular senescence in several cell types, including type 2 alveolar cells. In addition, Foxo4-p53 binding increased in BPD rat lungs. Daily TUDCA or KYC, administered intraperitoneally, effectively decreased cellular senescence, improved alveolar complexity, and partially maintained the numbers of type 2 alveolar cells. Foxo4 dri administered at P4, P6, P8, and P10 led to outcomes similar to TUDCA and KYC. Our data suggest that cellular senescence plays an essential role in BPD after initial inducement by hyperoxia. Reducing myeloperoxidase toxic oxidant production, ER stress, and attenuating cellular senescence are potential therapeutic strategies for halting BPD progression.
Collapse
Affiliation(s)
- Xigang Jing
- Department of Pediatrics
- Children's Research Institute
| | - Shuang Jia
- Department of Pediatrics
- Children's Research Institute
| | - Maggie Teng
- Department of Anthropology, Washington University in St. Louis, St. Louis, Missouri; and
| | | | | | | | - Chien-Wei Lin
- Division of Biostatistics, Institute for Health and Equity, and
| | | | - Kirkwood A Pritchard
- Children's Research Institute
- Department of Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin
- ReNeuroGen LLC, Milwaukee, Wisconsin
| | | | | | - Ru-Jeng Teng
- Department of Pediatrics
- Children's Research Institute
| |
Collapse
|
5
|
Robledo E, Benito Rodriguez PG, Vega IA, Colombo MI, Aguilera MO. Staphylococcus aureus phagocytosis is affected by senescence. FRONTIERS IN AGING 2023; 4:1198241. [PMID: 37584054 PMCID: PMC10423838 DOI: 10.3389/fragi.2023.1198241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 07/03/2023] [Indexed: 08/17/2023]
Abstract
Senescent cells accumulate in multicellular animals with aging, resulting in organ or tissue dysfunction. These alterations increase the incidence of a variety of illnesses, including infectious diseases, and, in certain instances, its severity. In search of a rationale for this phenomenon, we focused on the endophagocytic pathway in senescent cells. We first described the endocytic vesicle populations at different stages of maturation using confocal microscopy. There was an increase in the number of vacuoles per cell, which was partially explained by an increase in cell size. No changes in vesicle maturation or degradation capacities were determined by microscopy or Western blot assays. Also, we studied the internalization of various endophagocytic cargoes in senescent cells and observed only a decrease in the intracellular recovery of bacteria such as Staphylococcus aureus. Afterwards, we studied the intracellular traffic of S. aureus, and observed no differences in the infection between control and senescent cells. In addition we quantified the recovery of bacteria from control and senescent cells infected in the presence of several inhibitors of endophagosomal maturation, and no changes were observed. These results suggest that bacterial internalization is affected in senescent cells. Indeed, we confirmed this hypothesis by determining minor bacterial adherence and internalization by confocal microscopy. Furthermore, it is important to highlight that we found very similar results with cells from aged animals, specifically BMDMs. This alteration in senescent cells enlightens the diminished bacterial clearance and may be a factor that increases the propensity to suffer severe infectious conditions in the elderly.
Collapse
Affiliation(s)
- Esteban Robledo
- Instituto de Histología y Embriología (IHEM) “Dr. Mario H. Burgos” CONICET, Universidad Nacional de Cuyo Mendoza, Mendoza, Argentina
- Departamento Bases Científicas en Salud-Facultad de Ciencias Médicas, Facultad de Medicina, Biología Celular y Molecular, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Paula Guadalupe Benito Rodriguez
- Instituto de Histología y Embriología (IHEM) “Dr. Mario H. Burgos” CONICET, Universidad Nacional de Cuyo Mendoza, Mendoza, Argentina
| | - Israel Aníbal Vega
- Departamento de Biología, Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - María Isabel Colombo
- Instituto de Histología y Embriología (IHEM) “Dr. Mario H. Burgos” CONICET, Universidad Nacional de Cuyo Mendoza, Mendoza, Argentina
- Departamento Bases Científicas en Salud-Facultad de Ciencias Médicas, Facultad de Medicina, Biología Celular y Molecular, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Milton Osmar Aguilera
- Departamento Bases Científicas en Salud-Facultad de Ciencias Médicas, Facultad de Medicina, Biología Celular y Molecular, Universidad Nacional de Cuyo, Mendoza, Argentina
- Facultad de Odontología, Microbiología, Parasitología e Inmunología, Universidad Nacional de Cuyo, Mendoza, Argentina
| |
Collapse
|
6
|
Torres-Arciga K, Flores-León M, Ruiz-Pérez S, Trujillo-Pineda M, González-Barrios R, Herrera LA. Histones and their chaperones: Adaptive remodelers of an ever-changing chromatinic landscape. Front Genet 2022; 13:1057846. [PMID: 36468032 PMCID: PMC9709290 DOI: 10.3389/fgene.2022.1057846] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/02/2022] [Indexed: 07/29/2023] Open
Abstract
Chromatin maintenance and remodeling are processes that take place alongside DNA repair, replication, or transcription to ensure the survival and adaptability of a cell. The environment and the needs of the cell dictate how chromatin is remodeled; particularly where and which histones are deposited, thus changing the canonical histone array to regulate chromatin structure and gene expression. Chromatin is highly dynamic, and histone variants and their chaperones play a crucial role in maintaining the epigenetic regulation at different genomic regions. Despite the large number of histone variants reported to date, studies on their roles in physiological processes and pathologies are emerging but continue to be scarce. Here, we present recent advances in the research on histone variants and their chaperones, with a focus on their importance in molecular mechanisms such as replication, transcription, and DNA damage repair. Additionally, we discuss the emerging role they have in transposable element regulation, aging, and chromatin remodeling syndromes. Finally, we describe currently used methods and their limitations in the study of these proteins and highlight the importance of improving the experimental approaches to further understand this epigenetic machinery.
Collapse
Affiliation(s)
- Karla Torres-Arciga
- Doctorado en Ciencias Biológicas, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología (INCan)-Instituto de Investigaciones Biomédicas (IIBO), Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Manuel Flores-León
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas (IIBO), Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Samuel Ruiz-Pérez
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología (INCan)-Instituto de Investigaciones Biomédicas (IIBO), Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Magalli Trujillo-Pineda
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología (INCan)-Instituto de Investigaciones Biomédicas (IIBO), Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Rodrigo González-Barrios
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología (INCan)-Instituto de Investigaciones Biomédicas (IIBO), Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Luis A. Herrera
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología (INCan)-Instituto de Investigaciones Biomédicas (IIBO), Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
- Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| |
Collapse
|
7
|
Abstract
Cellular senescence is implicated in a wide range of physiological and pathological conditions throughout an organism's entire lifetime. In particular, it has become evident that senescence plays a causative role in aging and age-associated disorders. This is not due simply to the loss of function of senescent cells. Instead, the substantial alterations of the cellular activities of senescent cells, especially the array of secretory factors, impact the surrounding tissues or even entire organisms. Such non-cell-autonomous functionality is largely coordinated by tissue-specific genes, constituting a cell fate-determining state. Senescence can be viewed as a gain-of-function phenotype or a process of cell identity shift. Cellular functionality or lineage-specific gene expression is tightly linked to the cell type-specific epigenetic landscape, reinforcing the heterogeneity of senescence across cell types. Here, we aim to define the senescence cellular functionality and epigenetic features that may contribute to the gain-of-function phenotype.
Collapse
Affiliation(s)
- Ioana Olan
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom; ,
| | - Masashi Narita
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom; ,
| |
Collapse
|
8
|
Viegas D, Pereira CD, Martins F, Mateus T, da Cruz e Silva OAB, Herdeiro MT, Rebelo S. Nuclear Envelope Alterations in Myotonic Dystrophy Type 1 Patient-Derived Fibroblasts. Int J Mol Sci 2022; 23:522. [PMID: 35008948 PMCID: PMC8745202 DOI: 10.3390/ijms23010522] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 12/29/2021] [Accepted: 12/30/2021] [Indexed: 02/01/2023] Open
Abstract
Myotonic dystrophy type 1 (DM1) is a hereditary and multisystemic disease characterized by myotonia, progressive distal muscle weakness and atrophy. The molecular mechanisms underlying this disease are still poorly characterized, although there are some hypotheses that envisage to explain the multisystemic features observed in DM1. An emergent hypothesis is that nuclear envelope (NE) dysfunction may contribute to muscular dystrophies, particularly to DM1. Therefore, the main objective of the present study was to evaluate the nuclear profile of DM1 patient-derived and control fibroblasts and to determine the protein levels and subcellular distribution of relevant NE proteins in these cell lines. Our results demonstrated that DM1 patient-derived fibroblasts exhibited altered intracellular protein levels of lamin A/C, LAP1, SUN1, nesprin-1 and nesprin-2 when compared with the control fibroblasts. In addition, the results showed an altered location of these NE proteins accompanied by the presence of nuclear deformations (blebs, lobes and/or invaginations) and an increased number of nuclear inclusions. Regarding the nuclear profile, DM1 patient-derived fibroblasts had a larger nuclear area and a higher number of deformed nuclei and micronuclei than control-derived fibroblasts. These results reinforce the evidence that NE dysfunction is a highly relevant pathological characteristic observed in DM1.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Sandra Rebelo
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, 3810-193 Aveiro, Portugal; (D.V.); (C.D.P.); (F.M.); (T.M.); (O.A.B.d.C.e.S.); (M.T.H.)
| |
Collapse
|
9
|
Tomimatsu K, Bihary D, Olan I, Parry AJ, Schoenfelder S, Chan ASL, Slater GSC, Ito Y, Rugg-Gunn PJ, Kirschner K, Bermejo-Rodriguez C, Seko T, Kugoh H, Shiraishi K, Sayama K, Kimura H, Fraser P, Narita M, Samarajiwa SA, Narita M. Locus-specific induction of gene expression from heterochromatin loci during cellular senescence. NATURE AGING 2022; 2:31-45. [PMID: 37118356 DOI: 10.1038/s43587-021-00147-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 11/04/2021] [Indexed: 04/30/2023]
Abstract
Senescence is a fate-determined state, accompanied by reorganization of heterochromatin. Although lineage-appropriate genes can be temporarily repressed through facultative heterochromatin, stable silencing of lineage-inappropriate genes often involves the constitutive heterochromatic mark, histone H3 lysine 9 trimethylation (H3K9me3). The fate of these heterochromatic genes during senescence is unclear. In the present study, we show that a small number of lineage-inappropriate genes, exemplified by the LCE2 skin genes, are derepressed during senescence from H3K9me3 regions in fibroblasts. DNA FISH experiments reveal that these gene loci, which are condensed at the nuclear periphery in proliferative cells, are decompacted during senescence. Decompaction of the locus is not sufficient for LCE2 expression, which requires p53 and C/EBPβ signaling. NLRP3, which is predominantly expressed in macrophages from an open topologically associated domain (TAD), is also derepressed in senescent fibroblasts due to the local disruption of the H3K9me3-rich TAD that contains it. NLRP3 has been implicated in the amplification of inflammatory cytokine signaling in senescence and aging, highlighting the functional relevance of gene induction from 'permissive' H3K9me3 regions in senescent cells.
Collapse
Affiliation(s)
- Kosuke Tomimatsu
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
- Shiga University of Medical Science, Shiga, Japan
- Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Dóra Bihary
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Cambridge, UK
- VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium
| | - Ioana Olan
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Aled J Parry
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
- Epigenetics Programme, The Babraham Institute, Cambridge, UK
| | - Stefan Schoenfelder
- Epigenetics Programme, The Babraham Institute, Cambridge, UK
- Nuclear Dynamics Programme, The Babraham Institute, Cambridge, UK
| | - Adelyne S L Chan
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Guy St C Slater
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Yoko Ito
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
- International University of Health and Welfare, Tochigi, Japan
| | | | - Kristina Kirschner
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
- Institute for Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Camino Bermejo-Rodriguez
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - Tomomi Seko
- Department of Biomedical Science, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Science, Tottori University, Yonago, Japan
- Chromosome Engineering Research Center, Tottori University, Yonago, Japan
| | - Hiroyuki Kugoh
- Department of Biomedical Science, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Science, Tottori University, Yonago, Japan
- Chromosome Engineering Research Center, Tottori University, Yonago, Japan
| | - Ken Shiraishi
- Department of Dermatology, Graduate School of Medicine, Ehime University, Toon, Japan
| | - Koji Sayama
- Department of Dermatology, Graduate School of Medicine, Ehime University, Toon, Japan
| | - Hiroshi Kimura
- Tokyo Tech World Research Hub Initiative and Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan
| | - Peter Fraser
- Nuclear Dynamics Programme, The Babraham Institute, Cambridge, UK
- Department of Biological Science, Florida State University, Tallahassee, FL, USA
| | - Masako Narita
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK.
| | - Shamith A Samarajiwa
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Cambridge, UK.
| | - Masashi Narita
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK.
- Tokyo Tech World Research Hub Initiative and Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan.
| |
Collapse
|
10
|
Guo G, Watterson S, Zhang SD, Bjourson A, McGilligan V, Peace A, Rai TS. The role of senescence in the pathogenesis of atrial fibrillation: A target process for health improvement and drug development. Ageing Res Rev 2021; 69:101363. [PMID: 34023420 DOI: 10.1016/j.arr.2021.101363] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 01/24/2021] [Accepted: 05/12/2021] [Indexed: 12/11/2022]
Abstract
Cellular senescence is a state of growth arrest that occurs after cells encounter various stresses. Senescence contributes to tumour suppression, embryonic development, and wound healing. It impacts on the pathology of various diseases by secreting inflammatory chemokines, immune modulators and other bioactive factors. These secretory biosignatures ultimately cause inflammation, tissue fibrosis, immunosenescence and many ageing-related diseases such as atrial fibrillation (AF). Because the molecular mechanisms underpinning AF development remain unclear, current treatments are suboptimal and have serious side effects. In this review, we summarize recent results describing the role of senescence in AF. We propose that senescence factors induce AF and have a causative role. Hence, targeting senescence and its secretory phenotype may attenuate AF.
Collapse
|
11
|
Napoletani G, Protto V, Marcocci ME, Nencioni L, Palamara AT, De Chiara G. Recurrent Herpes Simplex Virus Type 1 (HSV-1) Infection Modulates Neuronal Aging Marks in In Vitro and In Vivo Models. Int J Mol Sci 2021; 22:6279. [PMID: 34208020 PMCID: PMC8230621 DOI: 10.3390/ijms22126279] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/20/2021] [Accepted: 06/08/2021] [Indexed: 01/21/2023] Open
Abstract
Herpes simplex virus 1 (HSV-1) is a widespread neurotropic virus establishing a life-long latent infection in neurons with periodic reactivations. Recent studies linked HSV-1 to neurodegenerative processes related to age-related disorders such as Alzheimer's disease. Here, we explored whether recurrent HSV-1 infection might accelerate aging in neurons, focusing on peculiar marks of aged cells, such as the increase in histone H4 lysine (K) 16 acetylation (ac) (H4K16ac); the decrease of H3K56ac, and the modified expression of Sin3/HDAC1 and HIRA proteins. By exploiting both in vitro and in vivo models of recurrent HSV-1 infection, we found a significant increase in H4K16ac, Sin3, and HDAC1 levels, suggesting that the neuronal response to virus latency and reactivation includes the upregulation of these aging markers. On the contrary, we found a significant decrease in H3K56ac that was specifically linked to viral reactivation and apparently not related to aging-related markers. A complex modulation of HIRA expression and localization was found in the brain from HSV-1 infected mice suggesting a specific role of this protein in viral latency and reactivation. Overall, our results pointed out novel molecular mechanisms through which recurrent HSV-1 infection may affect neuronal aging, likely contributing to neurodegeneration.
Collapse
Affiliation(s)
- Giorgia Napoletani
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia–Fondazione Cenci Bolognetti, 00185 Rome, Italy; (G.N.); (V.P.); (M.E.M.); (L.N.); (A.T.P.)
| | - Virginia Protto
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia–Fondazione Cenci Bolognetti, 00185 Rome, Italy; (G.N.); (V.P.); (M.E.M.); (L.N.); (A.T.P.)
| | - Maria Elena Marcocci
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia–Fondazione Cenci Bolognetti, 00185 Rome, Italy; (G.N.); (V.P.); (M.E.M.); (L.N.); (A.T.P.)
| | - Lucia Nencioni
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia–Fondazione Cenci Bolognetti, 00185 Rome, Italy; (G.N.); (V.P.); (M.E.M.); (L.N.); (A.T.P.)
| | - Anna Teresa Palamara
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia–Fondazione Cenci Bolognetti, 00185 Rome, Italy; (G.N.); (V.P.); (M.E.M.); (L.N.); (A.T.P.)
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Giovanna De Chiara
- Institute of Translational Pharmacology, National Research Council (CNR), 00133 Rome, Italy
| |
Collapse
|
12
|
Chen L, Liao F, Wu J, Wang Z, Jiang Z, Zhang C, Luo P, Ma L, Gong Q, Wang Y, Wang Q, Luo M, Yang Z, Han S, Shi C. Acceleration of ageing via disturbing mTOR-regulated proteostasis by a new ageing-associated gene PC4. Aging Cell 2021; 20:e13370. [PMID: 33957702 PMCID: PMC8208792 DOI: 10.1111/acel.13370] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 02/21/2021] [Accepted: 03/31/2021] [Indexed: 01/08/2023] Open
Abstract
Research on ageing‐associated genes is important for investigating ageing and anti‐ageing strategies. Here, we firstly reported that the human positive cofactor 4 (PC4), a multifunctional and highly conserved nucleoprotein, is accumulated and activated during ageing and causes global accelerated ageing process by disrupting proteostasis. Mechanistically, PC4 interacts with Sin3‐HDAC complex and inhibits its deacetylated activity, leads to hyper‐acetylation of the histones at the promoters of mTOR‐related genes and causes mTOR signalling activation. Accordingly, mTOR activation causes excessive protein synthesis, resulting in impaired proteostasis and accelerated senescence. These results reveal a new biological function of PC4 in vivo, recognizes PC4 as a new ageing‐associated gene and provides a genetically engineered mouse model to simulate natural ageing. More importantly, our findings also indicate that PC4 is involved in histone acetylation and serves as a potential target to improve proteostasis and delay ageing.
Collapse
Affiliation(s)
- Long Chen
- Institute of Rocket Force Medicine State Key Laboratory of Trauma, Burns and Combined Injury Third Military Medical University Chongqing China
| | - Fengying Liao
- Institute of Rocket Force Medicine State Key Laboratory of Trauma, Burns and Combined Injury Third Military Medical University Chongqing China
| | - Jie Wu
- Institute of Rocket Force Medicine State Key Laboratory of Trauma, Burns and Combined Injury Third Military Medical University Chongqing China
| | - Ziwen Wang
- Institute of Rocket Force Medicine State Key Laboratory of Trauma, Burns and Combined Injury Third Military Medical University Chongqing China
- Department of Cardiology Geriatric Cardiovascular Disease Research and Treatment Center 252 Hospital of PLA (82nd Group Army Hospital of PLA) Baoding China
| | - Zhongyong Jiang
- Institute of Rocket Force Medicine State Key Laboratory of Trauma, Burns and Combined Injury Third Military Medical University Chongqing China
| | - Chi Zhang
- Institute of Rocket Force Medicine State Key Laboratory of Trauma, Burns and Combined Injury Third Military Medical University Chongqing China
| | - Peng Luo
- Institute of Rocket Force Medicine State Key Laboratory of Trauma, Burns and Combined Injury Third Military Medical University Chongqing China
| | - Le Ma
- Institute of Rocket Force Medicine State Key Laboratory of Trauma, Burns and Combined Injury Third Military Medical University Chongqing China
| | - Qiang Gong
- Department of Hematology Southwest Hospital Third Military Medical University Chongqing China
| | - Yang Wang
- Institute of Rocket Force Medicine State Key Laboratory of Trauma, Burns and Combined Injury Third Military Medical University Chongqing China
| | - Qing Wang
- Institute of Rocket Force Medicine State Key Laboratory of Trauma, Burns and Combined Injury Third Military Medical University Chongqing China
| | - Min Luo
- Institute of Rocket Force Medicine State Key Laboratory of Trauma, Burns and Combined Injury Third Military Medical University Chongqing China
| | - Zeyu Yang
- Breast and Thyroid Surgical Department Chongqing General Hospital University of Chinese Academy of Sciences Chongqing China
| | - Shiqian Han
- Institute of Tropical Medicine Third Military Medical University Chongqing China
| | - Chunmeng Shi
- Institute of Rocket Force Medicine State Key Laboratory of Trauma, Burns and Combined Injury Third Military Medical University Chongqing China
| |
Collapse
|
13
|
Mehta IS, Riyahi K, Pereira RT, Meaburn KJ, Figgitt M, Kill IR, Eskiw CH, Bridger JM. Interphase Chromosomes in Replicative Senescence: Chromosome Positioning as a Senescence Biomarker and the Lack of Nuclear Motor-Driven Chromosome Repositioning in Senescent Cells. Front Cell Dev Biol 2021; 9:640200. [PMID: 34113611 PMCID: PMC8185894 DOI: 10.3389/fcell.2021.640200] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 02/25/2021] [Indexed: 01/10/2023] Open
Abstract
This study demonstrates, and confirms, that chromosome territory positioning is altered in primary senescent human dermal fibroblasts (HDFs). The chromosome territory positioning pattern is very similar to that found in HDFs made quiescent either by serum starvation or confluence; but not completely. A few chromosomes are found in different locations. One chromosome in particular stands out, chromosome 10, which is located in an intermediate location in young proliferating HDFs, but is found at the nuclear periphery in quiescent cells and in an opposing location of the nuclear interior in senescent HDFs. We have previously demonstrated that individual chromosome territories can be actively and rapidly relocated, with 15 min, after removal of serum from the culture media. These chromosome relocations require nuclear motor activity through the presence of nuclear myosin 1β (NM1β). We now also demonstrate rapid chromosome movement in HDFs after heat-shock at 42°C. Others have shown that heat shock genes are actively relocated using nuclear motor protein activity via actin or NM1β (Khanna et al., 2014; Pradhan et al., 2020). However, this current study reveals, that in senescent HDFs, chromosomes can no longer be relocated to expected nuclear locations upon these two types of stimuli. This coincides with a entirely different organisation and distribution of NM1β within senescent HDFs.
Collapse
Affiliation(s)
- Ishita S Mehta
- Centre for Genome Engineering and Maintenance, Division of Biosciences, Department of Life Sciences, College of Health, Medicine and Life Sciences, Kingston Lane, Brunel University London, Uxbridge, United Kingdom.,Tata Institute of Fundamental Research, Mumbai, India
| | - Kumars Riyahi
- Centre for Genome Engineering and Maintenance, Division of Biosciences, Department of Life Sciences, College of Health, Medicine and Life Sciences, Kingston Lane, Brunel University London, Uxbridge, United Kingdom
| | - Rita Torres Pereira
- Centre for Genome Engineering and Maintenance, Division of Biosciences, Department of Life Sciences, College of Health, Medicine and Life Sciences, Kingston Lane, Brunel University London, Uxbridge, United Kingdom
| | - Karen J Meaburn
- Centre for Genome Engineering and Maintenance, Division of Biosciences, Department of Life Sciences, College of Health, Medicine and Life Sciences, Kingston Lane, Brunel University London, Uxbridge, United Kingdom
| | - Martin Figgitt
- Centre for Genome Engineering and Maintenance, Division of Biosciences, Department of Life Sciences, College of Health, Medicine and Life Sciences, Kingston Lane, Brunel University London, Uxbridge, United Kingdom.,Department of Life Sciences, Birmingham City University, Birmingham, United Kingdom
| | - Ian R Kill
- Centre for Genome Engineering and Maintenance, Division of Biosciences, Department of Life Sciences, College of Health, Medicine and Life Sciences, Kingston Lane, Brunel University London, Uxbridge, United Kingdom
| | - Christopher H Eskiw
- Department of Food and Bioproduct Sciences, University of Saskatchewan, Saskatoon, SK, Canada
| | - Joanna M Bridger
- Centre for Genome Engineering and Maintenance, Division of Biosciences, Department of Life Sciences, College of Health, Medicine and Life Sciences, Kingston Lane, Brunel University London, Uxbridge, United Kingdom
| |
Collapse
|
14
|
Wang J, Zheng B, Yang S, Zhou D, Wang J. Olmesartan Prevents Oligomerized Amyloid β (Aβ)-Induced Cellular Senescence in Neuronal Cells. ACS Chem Neurosci 2021; 12:1162-1169. [PMID: 33710861 DOI: 10.1021/acschemneuro.0c00775] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease with high morbidity. The deposition of oligomerized amyloid β (Aβ) is the pathological feature of AD. The Aβ-caused neuronal oxidative stress and cellular senescence play an important role in the development and progression of AD. Olmesartan is a novel angiotensin receptor blocker with promising antihypertensive properties and has recently been reported to exert anti-inflammatory and antioxidative stress effects. Blood pressure control using Angiotensin receptor blockers has shown multiple benefits in Alzheimer's disease models. In the present study, the effect of Olmesartan on oligomerized amyloid β (Aβ)-induced cellular senescence was investigated in cultured M17 neuronal cells. Our results show that Olmesartan treatment significantly ameliorates oligomerized Aβ-elevated ROS and MDA levels, as well as the induced senescent cells number. At the molecular level, Olmesartan inhibits the elevated expression of senescence biomarkers (p16 and p21). Furthermore, Olmesartan potently reversed the increased K382 acetylation of p53 and the downregulation of SIRT1. Moreover, we show that the effect of Olmesartan against cell senescence and deacetylation of p53 was abolished by inhibition of SIRT1, either by using nicotinamide or by transfection with SIRT1 siRNA. In conclusion, Olmesartan prevents oligomerized Aβ-induced cellular senescence in neuronal cells by downregulating p16 and p21 through a SIRT1 dependent deacetylation of p53; our finding indicates that Olmesartan has a protective effect in Aβ-induced neurotoxicity.
Collapse
Affiliation(s)
- Jian Wang
- Department of Neurology, Ya’an Peoples Hospital, Ya’an, Sichuan 625000, China
| | - Bo Zheng
- Department of Neurology, Ya’an Peoples Hospital, Ya’an, Sichuan 625000, China
| | - Shu Yang
- Department of Neurology, The Affiliated Hospital of University of Electronic Science and Technology, Sichuan Provincial People’s Hospital, Chengdu, Sichuan 610000, China
| | - Duoqiang Zhou
- Department of Neurology, Hospital of Traditional Chinese Medicine, Qiannan Bouyei and Miao Autonomous Prefecture, Duyun, Guizhou 558000, China
| | - Jianhong Wang
- Department of Neurology, The Affiliated Hospital of University of Electronic Science and Technology, Sichuan Provincial People’s Hospital, Chengdu, Sichuan 610000, China
| |
Collapse
|
15
|
Garnett S, de Bruyns A, Provencher-Tom V, Dutchak K, Shu R, Dankort D. Metabolic Regulator IAPP (Amylin) Is Required for BRAF and RAS Oncogene-Induced Senescence. Mol Cancer Res 2021; 19:874-885. [PMID: 33500359 DOI: 10.1158/1541-7786.mcr-20-0879] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 12/17/2020] [Accepted: 01/21/2021] [Indexed: 11/16/2022]
Abstract
Cellular senescence is characterized by a prolonged and predominantly irreversible cell-cycle arrest state, which is linked to loss of tissue function and aging in mammals. Moreover, in response to aberrant oncogenic signals such as those from oncogenic RAS or BRAF, senescence functions as an intrinsic tumor suppressor mechanism restraining tumor progression. In addition to this durable proliferative block, senescent cells adopt altered morphologies, transcriptional profiles, and metabolism, while often possessing unusual heterochromatin formation termed senescence-associated heterochromatic foci. To uncover genes that are required to permit proliferation in the face of sustained oncogene signaling, we conducted an shRNA-based genetic screen in primary cells expressing inducible BRAF. Here we show that depletion of a known glycolysis regulator, islet amylin polypeptide (IAPP also known as amylin), prevents RAS and BRAF oncogene-induced senescence (OIS) in human cells. Importantly, depletion of IAPP resulted in changes of the cells' metabolome and this metabolic reprogramming was associated with widespread alterations in chromatin modifications compared with senescent cells. Conversely, exogenous treatment of IAPP-depleted cells with amylin restored OIS. Together, our results demonstrate that the metabolic regulator IAPP is important regulator of OIS. Moreover, they suggest that IAPP analog treatment or activation of IAPP signaling in RAS/BRAF mutant tumors may have therapeutic potential through senescence induction. IMPLICATIONS: These findings demonstrate that IAPP is a novel metabolic regulator of oncogene-induced senescence and use of IAPP analogs may be therapeutically effective to restore growth arrest to BRAF and/or RAS mutant cancers.
Collapse
Affiliation(s)
- Sam Garnett
- Department of Biology, McGill University, Montréal QC, Canada
| | | | | | - Kendall Dutchak
- Department of Biology, McGill University, Montréal QC, Canada
| | - Ran Shu
- Department of Biology, McGill University, Montréal QC, Canada
| | - David Dankort
- Department of Biology, McGill University, Montréal QC, Canada. .,Goodman Cancer Research Centre, Montréal QC, Canada
| |
Collapse
|
16
|
Olan I, Parry AJ, Schoenfelder S, Narita M, Ito Y, Chan ASL, Slater GSC, Bihary D, Bando M, Shirahige K, Kimura H, Samarajiwa SA, Fraser P, Narita M. Transcription-dependent cohesin repositioning rewires chromatin loops in cellular senescence. Nat Commun 2020; 11:6049. [PMID: 33247104 PMCID: PMC7695716 DOI: 10.1038/s41467-020-19878-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 10/30/2020] [Indexed: 12/19/2022] Open
Abstract
Senescence is a state of stable proliferative arrest, generally accompanied by the senescence-associated secretory phenotype, which modulates tissue homeostasis. Enhancer-promoter interactions, facilitated by chromatin loops, play a key role in gene regulation but their relevance in senescence remains elusive. Here, we use Hi-C to show that oncogenic RAS-induced senescence in human diploid fibroblasts is accompanied by extensive enhancer-promoter rewiring, which is closely connected with dynamic cohesin binding to the genome. We find de novo cohesin peaks often at the 3' end of a subset of active genes. RAS-induced de novo cohesin peaks are transcription-dependent and enriched for senescence-associated genes, exemplified by IL1B, where de novo cohesin binding is involved in new loop formation. Similar IL1B induction with de novo cohesin appearance and new loop formation are observed in terminally differentiated macrophages, but not TNFα-treated cells. These results suggest that RAS-induced senescence represents a cell fate determination-like process characterised by a unique gene expression profile and 3D genome folding signature, mediated in part through cohesin redistribution on chromatin.
Collapse
Affiliation(s)
- Ioana Olan
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, UK
| | - Aled J Parry
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, UK
- Epigenetics Programme, The Babraham Institute, Babraham Research Campus, Cambridge, UK
| | - Stefan Schoenfelder
- Epigenetics Programme, The Babraham Institute, Babraham Research Campus, Cambridge, UK
- Nuclear Dynamics Programme, The Babraham Institute, Babraham Research Campus, Cambridge, UK
| | - Masako Narita
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, UK
| | - Yoko Ito
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, UK
| | - Adelyne S L Chan
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, UK
| | - Guy St C Slater
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, UK
| | - Dóra Bihary
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Masashige Bando
- Laboratory of Genome Structure and Function, Institute of Molecular and Cellular Biosciences, The University of Tokyo, Tokyo, Japan
| | - Katsuhiko Shirahige
- Laboratory of Genome Structure and Function, Institute of Molecular and Cellular Biosciences, The University of Tokyo, Tokyo, Japan
| | - Hiroshi Kimura
- Cell Biology Centre, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan
| | - Shamith A Samarajiwa
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Peter Fraser
- Nuclear Dynamics Programme, The Babraham Institute, Babraham Research Campus, Cambridge, UK.
- Department of Biological Science, Florida State University, Tallahassee, FL, USA.
| | - Masashi Narita
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, UK.
- Tokyo Tech World Research Hub Initiative (WRHI), Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan.
| |
Collapse
|
17
|
Mizi A, Zhang S, Papantonis A. Genome folding and refolding in differentiation and cellular senescence. Curr Opin Cell Biol 2020; 67:56-63. [PMID: 32911122 DOI: 10.1016/j.ceb.2020.08.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 08/02/2020] [Accepted: 08/03/2020] [Indexed: 12/11/2022]
Abstract
The spatial conformation of chromatin within the confines of eukaryotic cell nuclei is now acknowledged as a decisive epigenetic mechanism for the modulation of such cellular functions as gene expression regulation, DNA replication or DNA damage repair. Of course, these processes are tightly regulated during organismal development and markedly affected by cellular ageing. Thus, the question that arises is to what extent does folding or refolding of the genome in three-dimensional space underlie the progression of development or ageing? Herein, we discuss recent experimental and modelling evidence to address this question and revisit how these seemingly different processed might represent two sides of the same coin.
Collapse
Affiliation(s)
- Athanasia Mizi
- Institute of Pathology, University Medical Center Göttingen, 37075, Göttingen, Germany
| | - Shu Zhang
- Institute of Pathology, University Medical Center Göttingen, 37075, Göttingen, Germany
| | - Argyris Papantonis
- Institute of Pathology, University Medical Center Göttingen, 37075, Göttingen, Germany.
| |
Collapse
|
18
|
Shang D, Hong Y, Xie W, Tu Z, Xu J. Interleukin-1β Drives Cellular Senescence of Rat Astrocytes Induced by Oligomerized Amyloid β Peptide and Oxidative Stress. Front Neurol 2020; 11:929. [PMID: 33013631 PMCID: PMC7493674 DOI: 10.3389/fneur.2020.00929] [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: 04/15/2020] [Accepted: 07/17/2020] [Indexed: 01/10/2023] Open
Abstract
Background: Alzheimer's disease (AD) is the leading cause of dementia. With no reliable treatment that delays or reverses the progress of AD, effective medical drugs, and interventions for AD treatment are in urgent need. Clinical success for patients thus relies on gaining a clearer understanding of AD pathogenesis to feed the development of novel and potent therapy strategies. It is well-established that inflammatory processes are involved in the pathology of AD, and recent studies implicated senescence of glial cells as an important player in the progression of AD. Methods: We did a preliminary screen in rat astrocytes for the five most abundant inflammatory factors in neuroinflammation, namely IL-1β, IL-6, IL-8, TGF-β1, and TNF-α, and found that IL-1β could efficiently induce cellular senescence. After that, SA-β-gal staining, immunofluorescence, ELISA, qRT-PCR, and immunoblotting were used to explore the underlying mechanism through which IL-1β mediates cellular senescence of rat astrocytes. Results: IL-1β-induced cellular senescence of rat astrocytes was accompanied by increased total and phosphorylated tau. Further experiments showed that both oligomerized amyloid β (Aβ) and H2O2 treatment can induce cellular senescence in rat astrocytes and increase the production and secretion of IL-1β from these cells. Subsequent mechanistic study revealed that activation of NLRP3 mediates Aβ and H2O2-induced maturation and secretion of IL-1β. Conclusion: Our results suggest that IL-1β mediates senescence in rat astrocytes induced by several common adverse stimuli in AD, implicating IL-1β and NLRP3 as valuable diagnostic biomarkers and therapeutic targets for AD.
Collapse
Affiliation(s)
- Dongsheng Shang
- Institute of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Yin Hong
- China National Clinical Research Center for Neurological Diseases (NCRC-ND), Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Wangwang Xie
- Institute of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Zhigang Tu
- Institute of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Jun Xu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
19
|
Hussain F, Basu S, Heng JJH, Loo LH, Zink D. Predicting direct hepatocyte toxicity in humans by combining high-throughput imaging of HepaRG cells and machine learning-based phenotypic profiling. Arch Toxicol 2020; 94:2749-2767. [PMID: 32533217 DOI: 10.1007/s00204-020-02778-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 05/05/2020] [Indexed: 02/07/2023]
Abstract
Accurate prediction of drug- and chemical-induced hepatotoxicity remains to be a problem for pharmaceutical companies as well as other industries and regulators. The goal of the current study was to develop an in vitro/in silico method for the rapid and accurate prediction of drug- and chemical-induced hepatocyte injury in humans. HepaRG cells were employed for high-throughput imaging in combination with phenotypic profiling. A reference set of 69 drugs and chemicals was screened at a range of 7 concentrations, and the cellular response values were used for training a supervised classifier and for determining assay performance by using tenfold cross-validation. The results showed that the best performing phenotypic features were related to nuclear translocation of RELA (RELA proto-oncogene, NF-kB subunit; also known as NF-kappa B p65), DNA organization, and the F-actin cytoskeleton. Using a subset of 30 phenotypic features, direct hepatocyte toxicity in humans could be predicted with a test sensitivity, specificity and balanced accuracy of 73%, 92%, and 83%, respectively. The method was applied to another set of 26 drugs and chemicals with unclear annotation and their hepatocyte toxicity in humans was predicted. The results also revealed that the identified discriminative phenotypic changes were related to cell death and cellular senescence. Whereas cell death-related endpoints are widely applied in in vitro toxicology, cellular senescence-related endpoints are not, although cellular senescence can be induced by various drugs and other small molecule compounds and plays an important role in liver injury and disease. These findings show how phenotypic profiling can reveal unexpected chemical-induced mechanisms in toxicology.
Collapse
Affiliation(s)
- Faezah Hussain
- NanoBio Lab and Institute of Bioengineering and Nanotechnology (IBN), 31 Biopolis Way, The Nanos, Singapore, 138669, Singapore
| | - Sreetama Basu
- Bioinformatics Institute, 30 Biopolis Street, #07-01 Matrix, Singapore, 138671, Singapore
| | - Javen Jun Hao Heng
- NanoBio Lab and Institute of Bioengineering and Nanotechnology (IBN), 31 Biopolis Way, The Nanos, Singapore, 138669, Singapore
| | - Lit-Hsin Loo
- Bioinformatics Institute, 30 Biopolis Street, #07-01 Matrix, Singapore, 138671, Singapore. .,Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore, 117600, Singapore.
| | - Daniele Zink
- NanoBio Lab and Institute of Bioengineering and Nanotechnology (IBN), 31 Biopolis Way, The Nanos, Singapore, 138669, Singapore.
| |
Collapse
|
20
|
de Mera-Rodríguez JA, Álvarez-Hernán G, Gañán Y, Martín-Partido G, Rodríguez-León J, Francisco-Morcillo J. Senescence-associated β-galactosidase activity in the developing avian retina. Dev Dyn 2019; 248:850-865. [PMID: 31226225 DOI: 10.1002/dvdy.74] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 05/22/2019] [Accepted: 06/15/2019] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Senescence-associated β-galactosidase (SA-β-GAL) histochemistry is the most commonly used biomarker of cellular senescence. These SA-β-GAL-positive cells are senescent embryonic cells that are usually removed by apoptosis from the embryo, followed by macrophage-mediated clearance. RESULTS Some authors have proposed that SA-β-GAL activity in differentiated neurons from young and adult mammals cannot be uniquely attributed to cell senescence, whether in vivo or in vitro. Using the developing visual system of the chicken as a model, the present study found that SA-β-GAL detected in the developing retina corresponded to lysosomal β-galactosidase activity, and that SA-β-GAL activity did not correlate with the chronotopographical distribution of apoptotic cells. However, SA-β-GAL staining in the undifferentiated retina coincided with the appearance of early differentiating neurons. In the laminated retina, SA-β-GAL staining was concentrated in the ganglion, amacrine, and horizontal cell layers. The photoreceptors and pigment epithelial cells also exhibited SA-β-GAL activity throughout retinal development. We have also found that SA-β-GAL staining strongly correlated p21 immunoreactivity. CONCLUSION In conclusion, the results clearly show that SA-β-GAL activity cannot be regarded as a specific marker of senescence during retinal development, and that it is mainly expressed in subpopulations of postmitotic neurons, which are nonproliferative cells, even at early stages of cell differentiation.
Collapse
Affiliation(s)
- José Antonio de Mera-Rodríguez
- Área de Anatomía Humana, Departamento de Anatomía, Biología Celular y Zoología, Facultad de Medicina, Universidad de Extremadura, Badajoz, Spain
| | - Guadalupe Álvarez-Hernán
- Área de Biología Celular, Departamento de Anatomía, Biología Celular y Zoología, Facultad de Ciencias, Universidad de Extremadura, Badajoz, Spain
| | - Yolanda Gañán
- Área de Anatomía Humana, Departamento de Anatomía, Biología Celular y Zoología, Facultad de Medicina, Universidad de Extremadura, Badajoz, Spain
| | - Gervasio Martín-Partido
- Área de Biología Celular, Departamento de Anatomía, Biología Celular y Zoología, Facultad de Ciencias, Universidad de Extremadura, Badajoz, Spain
| | - Joaquín Rodríguez-León
- Área de Anatomía Humana, Departamento de Anatomía, Biología Celular y Zoología, Facultad de Medicina, Universidad de Extremadura, Badajoz, Spain
| | - Javier Francisco-Morcillo
- Área de Biología Celular, Departamento de Anatomía, Biología Celular y Zoología, Facultad de Ciencias, Universidad de Extremadura, Badajoz, Spain
| |
Collapse
|
21
|
Chronic Infections: A Possible Scenario for Autophagy and Senescence Cross-Talk. Cells 2018; 7:cells7100162. [PMID: 30308990 PMCID: PMC6210027 DOI: 10.3390/cells7100162] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 10/04/2018] [Accepted: 10/07/2018] [Indexed: 02/07/2023] Open
Abstract
Multiple tissues and systems in the organism undergo modifications during aging due to an accumulation of damaged proteins, lipids, and genetic material. To counteract this process, the cells are equipped with specific mechanisms, such as autophagy and senescence. Particularly, the immune system undergoes a process called immunosenescence, giving rise to a chronic inflammatory status of the organism, with a decreased ability to counteract antigens. The obvious result of this process is a reduced defence capacity. Currently, there is evidence that some pathogens are able to accelerate the immunosenescence process for their own benefit. Although to date numerous reports show the autophagy–senescence relationship, or the connection between pathogens with autophagy or senescence, the link between the three actors remains unexplored. In this review, we have summarized current knowledge about important issues related to aging, senescence, and autophagy.
Collapse
|
22
|
A Heterochromatin Domain Forms Gradually at a New Telomere and Is Dynamic at Stable Telomeres. Mol Cell Biol 2018; 38:MCB.00393-17. [PMID: 29784772 PMCID: PMC6048312 DOI: 10.1128/mcb.00393-17] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 05/09/2018] [Indexed: 02/03/2023] Open
Abstract
Heterochromatin domains play important roles in chromosome biology, organismal development, and aging, including centromere function, mammalian female X chromosome inactivation, and senescence-associated heterochromatin foci. In the fission yeast Schizosaccharomyces pombe and metazoans, heterochromatin contains histone H3 that is dimethylated at lysine 9. Heterochromatin domains play important roles in chromosome biology, organismal development, and aging, including centromere function, mammalian female X chromosome inactivation, and senescence-associated heterochromatin foci. In the fission yeast Schizosaccharomyces pombe and metazoans, heterochromatin contains histone H3 that is dimethylated at lysine 9. While factors required for heterochromatin have been identified, the dynamics of heterochromatin formation are poorly understood. Telomeres convert adjacent chromatin into heterochromatin. To form a new heterochromatic region in S. pombe, an inducible DNA double-strand break (DSB) was engineered next to 48 bp of telomere repeats in euchromatin, which caused formation of a new telomere and the establishment and gradual spreading of a new heterochromatin domain. However, spreading was dynamic even after the telomere had reached its stable length, with reporter genes within the heterochromatin domain showing variegated expression. The system also revealed the presence of repeats located near the boundaries of euchromatin and heterochromatin that are oriented to allow the efficient healing of a euchromatic DSB to cap the chromosome end with a new telomere. Telomere formation in S. pombe therefore reveals novel aspects of heterochromatin dynamics and fail-safe mechanisms to repair subtelomeric breaks, with implications for similar processes in metazoan genomes.
Collapse
|
23
|
Laville V, Le Clerc S, Ezzedine K, Jdid R, Taing L, Labib T, Coulonges C, Ulveling D, Galan P, Guinot C, Fezeu L, Morizot F, Latreille J, Malvy D, Tschachler E, Zagury J. A genome wide association study identifies new genes potentially associated with eyelid sagging. Exp Dermatol 2018; 28:892-898. [DOI: 10.1111/exd.13559] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2018] [Indexed: 01/06/2023]
Affiliation(s)
- Vincent Laville
- Équipe GénomiqueBioinformatique et ApplicationsChaire de BioinformatiqueConservatoire National des Arts et Métiers Paris France
| | - Sigrid Le Clerc
- Équipe GénomiqueBioinformatique et ApplicationsChaire de BioinformatiqueConservatoire National des Arts et Métiers Paris France
| | - Khaled Ezzedine
- Department of DermatologyHenri Mondor Hospital and EA EpiDermE (Epidémiologie en Dermatologie et Evaluation des Thérapeutiques)UPEC‐Université Paris‐Est Créteil France
| | - Randa Jdid
- Department of Skin Knowledge & Women BeautyChanel R & T Pantin France
| | - Lieng Taing
- Équipe GénomiqueBioinformatique et ApplicationsChaire de BioinformatiqueConservatoire National des Arts et Métiers Paris France
| | - Toufik Labib
- Équipe GénomiqueBioinformatique et ApplicationsChaire de BioinformatiqueConservatoire National des Arts et Métiers Paris France
| | - Cédric Coulonges
- Équipe GénomiqueBioinformatique et ApplicationsChaire de BioinformatiqueConservatoire National des Arts et Métiers Paris France
| | - Damien Ulveling
- Équipe GénomiqueBioinformatique et ApplicationsChaire de BioinformatiqueConservatoire National des Arts et Métiers Paris France
| | - Pilar Galan
- Université Paris 13Equipe de Recherche en Epidémiologie Nutritionnelle (EREN)Centre d’Epidemiologie et Biostatistiques Sorbonne Paris Cité (CRESS)Inserm U1153, Inra U1125Cnam, COMUE Sorbonne‐Paris‐Cité Bobigny France
| | - Christiane Guinot
- Computer Science LaboratoryUniversity François Rabelais of Tours Tours France
| | - Leopold Fezeu
- Université Paris 13Equipe de Recherche en Epidémiologie Nutritionnelle (EREN)Centre d’Epidemiologie et Biostatistiques Sorbonne Paris Cité (CRESS)Inserm U1153, Inra U1125Cnam, COMUE Sorbonne‐Paris‐Cité Bobigny France
| | | | - Julie Latreille
- Department of Skin Knowledge & Women BeautyChanel R & T Pantin France
| | - Denis Malvy
- Université Paris 13Equipe de Recherche en Epidémiologie Nutritionnelle (EREN)Centre d’Epidemiologie et Biostatistiques Sorbonne Paris Cité (CRESS)Inserm U1153, Inra U1125Cnam, COMUE Sorbonne‐Paris‐Cité Bobigny France
- Department of Internal Medicine and Tropical DiseasesHôpital Saint‐André Bordeaux France
| | - Erwin Tschachler
- Department of DermatologyUniversity of Vienna Medical School Vienna Austria
| | - Jean‐François Zagury
- Équipe GénomiqueBioinformatique et ApplicationsChaire de BioinformatiqueConservatoire National des Arts et Métiers Paris France
| |
Collapse
|
24
|
Zirkel A, Nikolic M, Sofiadis K, Mallm JP, Brackley CA, Gothe H, Drechsel O, Becker C, Altmüller J, Josipovic N, Georgomanolis T, Brant L, Franzen J, Koker M, Gusmao EG, Costa IG, Ullrich RT, Wagner W, Roukos V, Nürnberg P, Marenduzzo D, Rippe K, Papantonis A. HMGB2 Loss upon Senescence Entry Disrupts Genomic Organization and Induces CTCF Clustering across Cell Types. Mol Cell 2018; 70:730-744.e6. [PMID: 29706538 DOI: 10.1016/j.molcel.2018.03.030] [Citation(s) in RCA: 137] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 02/19/2018] [Accepted: 03/25/2018] [Indexed: 11/30/2022]
Abstract
Processes like cellular senescence are characterized by complex events giving rise to heterogeneous cell populations. However, the early molecular events driving this cascade remain elusive. We hypothesized that senescence entry is triggered by an early disruption of the cells' three-dimensional (3D) genome organization. To test this, we combined Hi-C, single-cell and population transcriptomics, imaging, and in silico modeling of three distinct cells types entering senescence. Genes involved in DNA conformation maintenance are suppressed upon senescence entry across all cell types. We show that nuclear depletion of the abundant HMGB2 protein occurs early on the path to senescence and coincides with the dramatic spatial clustering of CTCF. Knocking down HMGB2 suffices for senescence-induced CTCF clustering and for loop reshuffling, while ectopically expressing HMGB2 rescues these effects. Our data suggest that HMGB2-mediated genomic reorganization constitutes a primer for the ensuing senescent program.
Collapse
Affiliation(s)
- Anne Zirkel
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany
| | - Milos Nikolic
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany
| | - Konstantinos Sofiadis
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany
| | - Jan-Philipp Mallm
- German Cancer Research Center and Bioquant, 69120 Heidelberg, Germany
| | - Chris A Brackley
- School of Physics and Astronomy, University of Edinburgh, EH9 3FD Edinburgh, UK
| | - Henrike Gothe
- Institute of Molecular Biology, 55128 Mainz, Germany
| | | | - Christian Becker
- Cologne Center for Genomics, University of Cologne, 50931 Cologne, Germany
| | - Janine Altmüller
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany; Cologne Center for Genomics, University of Cologne, 50931 Cologne, Germany
| | - Natasa Josipovic
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany
| | | | - Lilija Brant
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany
| | - Julia Franzen
- Helmholtz Institute for Biomedical Engineering, RWTH Aachen University Medical School, 52074 Aachen, Germany
| | - Mirjam Koker
- Clinic I of Internal Medicine and Center for Integrated Oncology, University Hospital Cologne, 50931 Cologne, Germany
| | - Eduardo G Gusmao
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany; Interdisciplinary Centre for Clinical Research, RWTH Aachen University Medical School, 52062 Aachen, Germany
| | - Ivan G Costa
- Interdisciplinary Centre for Clinical Research, RWTH Aachen University Medical School, 52062 Aachen, Germany
| | - Roland T Ullrich
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany; Clinic I of Internal Medicine and Center for Integrated Oncology, University Hospital Cologne, 50931 Cologne, Germany
| | - Wolfgang Wagner
- Helmholtz Institute for Biomedical Engineering, RWTH Aachen University Medical School, 52074 Aachen, Germany
| | | | - Peter Nürnberg
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany; Cologne Center for Genomics, University of Cologne, 50931 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany
| | - Davide Marenduzzo
- School of Physics and Astronomy, University of Edinburgh, EH9 3FD Edinburgh, UK
| | - Karsten Rippe
- German Cancer Research Center and Bioquant, 69120 Heidelberg, Germany
| | - Argyris Papantonis
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany.
| |
Collapse
|
25
|
Shang D, Wu Y, Ding Y, Lu Z, Shen Y, Zhu F, Liu H, Zhu C, Tu Z. Identification of a pyridine derivative inducing senescence in ovarian cancer cell lines via P21 activation. Clin Exp Pharmacol Physiol 2017; 45:452-460. [DOI: 10.1111/1440-1681.12891] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 10/04/2017] [Accepted: 10/26/2017] [Indexed: 01/15/2023]
Affiliation(s)
- Dongsheng Shang
- School of Pharmacy; Jiangsu University; Zhenjiang China
- Institute of Life Sciences; Jiangsu University; Zhenjiang China
| | - Yanfang Wu
- Institute of Life Sciences; Jiangsu University; Zhenjiang China
| | - Ya Ding
- School of Chemistry & Chemical Engineering; Jiangsu University; Zhenjiang China
| | - Ziwen Lu
- School of Pharmacy; Jiangsu University; Zhenjiang China
| | - Yanting Shen
- School of Pharmacy; Jiangsu University; Zhenjiang China
| | - Feifei Zhu
- Institute of Life Sciences; Jiangsu University; Zhenjiang China
| | - Hanqing Liu
- School of Pharmacy; Jiangsu University; Zhenjiang China
| | - Chunyin Zhu
- School of Chemistry & Chemical Engineering; Jiangsu University; Zhenjiang China
| | - Zhigang Tu
- Institute of Life Sciences; Jiangsu University; Zhenjiang China
| |
Collapse
|
26
|
Lujambio A. To clear, or not to clear (senescent cells)? That is the question. Bioessays 2017; 38 Suppl 1:S56-64. [PMID: 27417123 DOI: 10.1002/bies.201670910] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 11/30/2015] [Accepted: 12/09/2015] [Indexed: 12/22/2022]
Abstract
Cellular senescence is an anti-proliferative program that restricts the propagation of cells subjected to different kinds of stress. Cellular senescence was initially described as a cell-autonomous tumor suppressor mechanism that triggers an irreversible cell cycle arrest that prevents the proliferation of damaged cells at risk of neoplastic transformation. However, discoveries during the last decade have established that senescent cells can also impact the surrounding tissue microenvironment and the neighboring cells in a non-cell-autonomous manner. These non-cell-autonomous activities are, in part, mediated by the selective secretion of extracellular matrix degrading enzymes, cytokines, chemokines and immune modulators, which collectively constitute the senescence-associated secretory phenotype. One of the key functions of the senescence-associated secretory phenotype is to attract immune cells, which in turn can orchestrate the elimination of senescent cells. Interestingly, the clearance of senescent cells seems to be critical to dictate the net effects of cellular senescence. As a general rule, the successful elimination of senescent cells takes place in processes that are considered beneficial, such as tumor suppression, tissue remodeling and embryonic development, while the chronic accumulation of senescent cells leads to more detrimental consequences, namely, cancer and aging. Nevertheless, exceptions to this rule may exist. Now that cellular senescence is in the spotlight for both anti-cancer and anti-aging therapies, understanding the precise underpinnings of senescent cell removal will be essential to exploit cellular senescence to its full potential.
Collapse
Affiliation(s)
- Amaia Lujambio
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
27
|
Abstract
During aging, the mechanisms that normally maintain health and stress resistance strikingly decline, resulting in decrepitude, frailty, and ultimately death. Exactly when and how this decline occurs is unknown. Changes in transcriptional networks and chromatin state lie at the heart of age-dependent decline. These epigenomic changes are not only observed during aging but also profoundly affect cellular function and stress resistance, thereby contributing to the progression of aging. We propose that the dysregulation of transcriptional and chromatin networks is a crucial component of aging. Understanding age-dependent epigenomic changes will yield key insights into how aging begins and progresses and should lead to the development of new therapeutics that delay or even reverse aging and age-related diseases.
Collapse
Affiliation(s)
- Lauren N Booth
- Department of Genetics, Stanford University, 300 Pasteur Drive, Stanford, CA 94305, USA
| | - Anne Brunet
- Department of Genetics, Stanford University, 300 Pasteur Drive, Stanford, CA 94305, USA; Glenn Laboratories for the Biology of Aging, Stanford University, 300 Pasteur Drive, Stanford, CA 94305, USA.
| |
Collapse
|
28
|
Botchkarev VA. Second International Symposium-Epigenetic Regulation of Skin Regeneration and Aging: From Chromatin Biology towards the Understanding of Epigenetic Basis of Skin Diseases. J Invest Dermatol 2017; 137:1604-1608. [PMID: 28583676 DOI: 10.1016/j.jid.2017.01.037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2016] [Revised: 01/03/2017] [Accepted: 01/10/2017] [Indexed: 12/31/2022]
Affiliation(s)
- Vladimir A Botchkarev
- Centre for Skin Sciences, Faculty of Life Sciences, University of Bradford, Bradford, UK; Department of Dermatology, Boston University School of Medicine, Boston, Massachusetts, USA.
| |
Collapse
|
29
|
Valenzuela N, Soibam B, Li L, Wang J, Byers LA, Liu Y, Schwartz RJ, Stewart MD. HIRA deficiency in muscle fibers causes hypertrophy and susceptibility to oxidative stress. J Cell Sci 2017; 130:2551-2563. [PMID: 28600325 DOI: 10.1242/jcs.200642] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Accepted: 06/05/2017] [Indexed: 01/14/2023] Open
Abstract
Nucleosome assembly proceeds through DNA replication-coupled or replication-independent mechanisms. For skeletal myocytes, whose nuclei have permanently exited the cell cycle, replication-independent assembly is the only mode available for chromatin remodeling. For this reason, any nucleosome composition alterations accompanying transcriptional responses to physiological signals must occur through a DNA replication-independent pathway. HIRA is the histone chaperone primarily responsible for replication-independent incorporation of histone variant H3.3 across gene bodies and regulatory regions. Thus, HIRA would be expected to play an important role in epigenetically regulating myocyte gene expression. The objective of this study was to determine the consequence of eliminating HIRA from mouse skeletal myocytes. At 6 weeks of age, myofibers lacking HIRA showed no pathological abnormalities; however, genes involved in transcriptional regulation were downregulated. By 6 months of age, myofibers lacking HIRA exhibited hypertrophy, sarcolemmal perforation and oxidative damage. Genes involved in muscle growth and development were upregulated, but those associated with responses to cellular stresses were downregulated. These data suggest that elimination of HIRA produces a hypertrophic response in skeletal muscle and leaves myofibers susceptible to stress-induced degeneration.
Collapse
Affiliation(s)
- Nicolas Valenzuela
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Benjamin Soibam
- Department of Computer Science and Engineering Technology, University of Houston-Downtown, Houston, TX 77002, USA
| | - Lerong Li
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jing Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Lauren A Byers
- Department of Thoracic Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yu Liu
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Robert J Schwartz
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA.,Stem Cell Engineering Department, Texas Heart Institute at St. Luke's Episcopal Hospital, Houston, TX 77030, USA
| | - M David Stewart
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA .,Stem Cell Engineering Department, Texas Heart Institute at St. Luke's Episcopal Hospital, Houston, TX 77030, USA
| |
Collapse
|
30
|
Dillinger S, Straub T, Németh A. Nucleolus association of chromosomal domains is largely maintained in cellular senescence despite massive nuclear reorganisation. PLoS One 2017; 12:e0178821. [PMID: 28575119 PMCID: PMC5456395 DOI: 10.1371/journal.pone.0178821] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 05/21/2017] [Indexed: 01/10/2023] Open
Abstract
Mammalian chromosomes are organized in structural and functional domains of 0.1–10 Mb, which are characterized by high self-association frequencies in the nuclear space and different contact probabilities with nuclear sub-compartments. They exhibit distinct chromatin modification patterns, gene expression levels and replication timing. Recently, nucleolus-associated chromosomal domains (NADs) have been discovered, yet their precise genomic organization and dynamics are still largely unknown. Here, we use nucleolus genomics and single-cell experiments to address these questions in human embryonic fibroblasts during replicative senescence. Genome-wide mapping reveals 1,646 NADs in proliferating cells, which cover about 38% of the annotated human genome. They are mainly heterochromatic and correlate with late replicating loci. Using Hi-C data analysis, we show that interactions of NADs dominate interphase chromosome contacts in the 10–50 Mb distance range. Interestingly, only minute changes in nucleolar association are observed upon senescence. These spatial rearrangements in subdomains smaller than 100 kb are accompanied with local transcriptional changes. In contrast, large centromeric and pericentromeric satellite repeat clusters extensively dissociate from nucleoli in senescent cells. Accordingly, H3K9me3-marked heterochromatin gets remodelled at the perinucleolar space as revealed by immunofluorescence analyses. Collectively, this study identifies connections between the nucleolus, 3D genome structure, and cellular aging at the level of interphase chromosome organization.
Collapse
Affiliation(s)
- Stefan Dillinger
- Biochemistry Center Regensburg, University of Regensburg, Regensburg, Germany
| | - Tobias Straub
- Biomedical Center, Bioinformatics, Ludwig-Maximilians-University, Munich, Germany
| | - Attila Németh
- Biochemistry Center Regensburg, University of Regensburg, Regensburg, Germany
- * E-mail: ,
| |
Collapse
|
31
|
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
|
32
|
Davaapil H, Brockes JP, Yun MH. Conserved and novel functions of programmed cellular senescence during vertebrate development. Development 2017; 144:106-114. [PMID: 27888193 PMCID: PMC5278627 DOI: 10.1242/dev.138222] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 11/10/2017] [Indexed: 12/14/2022]
Abstract
Cellular senescence, a form of stable cell cycle arrest that is traditionally associated with tumour suppression, has been recently found to occur during mammalian development. Here, we show that cell senescence is an intrinsic part of the developmental programme in amphibians. Programmed senescence occurs in specific structures during defined time windows during amphibian development. It contributes to the physiological degeneration of the amphibian pronephros and to the development of the cement gland and oral cavity. In both contexts, senescence depends on TGFβ but is independent of ERK/MAPK activation. Furthermore, elimination of senescent cells through temporary TGFβ inhibition leads to developmental defects. Our findings uncover conserved and new roles of senescence in vertebrate organogenesis and support the view that cellular senescence may have arisen in evolution as a developmental mechanism.
Collapse
Affiliation(s)
- Hongorzul Davaapil
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London WC1E 6BT, UK
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK
| | - Jeremy P Brockes
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London WC1E 6BT, UK
| | - Maximina H Yun
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London WC1E 6BT, UK
| |
Collapse
|
33
|
Valenzuela N, Fan Q, Fa'ak F, Soibam B, Nagandla H, Liu Y, Schwartz RJ, McConnell BK, Stewart MD. Cardiomyocyte-specific conditional knockout of the histone chaperone HIRA in mice results in hypertrophy, sarcolemmal damage and focal replacement fibrosis. Dis Model Mech 2016; 9:335-45. [PMID: 26935106 PMCID: PMC4833330 DOI: 10.1242/dmm.022889] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
HIRA is the histone chaperone responsible for replication-independent incorporation of histone variant H3.3 within gene bodies and regulatory regions of actively transcribed genes, and within the bivalent promoter regions of developmentally regulated genes. The HIRA gene lies within the 22q11.2 deletion syndrome critical region; individuals with this syndrome have multiple congenital heart defects. Because terminally differentiated cardiomyocytes have exited the cell cycle, histone variants should be utilized for the bulk of chromatin remodeling. Thus, HIRA is likely to play an important role in epigenetically defining the cardiac gene expression program. In this study, we determined the consequence of HIRA deficiency in cardiomyocytes in vivo by studying the phenotype of cardiomyocyte-specific Hira conditional-knockout mice. Loss of HIRA did not perturb heart development, but instead resulted in cardiomyocyte hypertrophy and susceptibility to sarcolemmal damage. Cardiomyocyte degeneration gave way to focal replacement fibrosis and impaired cardiac function. Gene expression was widely altered in Hira conditional-knockout hearts. Significantly affected pathways included responses to cellular stress, DNA repair and transcription. Consistent with heart failure, fetal cardiac genes were re-expressed in the Hira conditional knockout. Our results suggest that transcriptional regulation by HIRA is crucial for cardiomyocyte homeostasis.
Collapse
Affiliation(s)
- Nicolas Valenzuela
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Qiying Fan
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA
| | - Faisal Fa'ak
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA
| | - Benjamin Soibam
- Department of Computer Science and Engineering Technology, University of Houston-Downtown, Houston, TX 77002, USA
| | - Harika Nagandla
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Yu Liu
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Robert J Schwartz
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA Stem Cell Engineering Department, Texas Heart Institute at St Luke's Episcopal Hospital, Houston, TX 77030, USA
| | - Bradley K McConnell
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA
| | - M David Stewart
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| |
Collapse
|
34
|
Yang R, Wu L, Chen J, Chen W, Zhang L, Zhang L, You R, Yin L, Li CH, Guan YQ. Effects of Differentiation and Antisenescence from BMSCs to Hepatocy-Like Cells of the PAAm-IGF-1/TNF-α Biomaterial. ACS APPLIED MATERIALS & INTERFACES 2016; 8:26638-26647. [PMID: 27668443 DOI: 10.1021/acsami.6b10377] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Aiming at the cells' differentiation phenomenon and senescence problem in liver tissue engineering, this work is designed to synthesize three different chargeable polymers (polypropylene acid (PAAc), polyethylene glycol (PEG), and polypropylene amine (PAAm)) coimmobilized by the insulin-like growth factor 1 (IGF-1) and tumor necrosis factor-α (TNF-α). We explore the hepatocyte differentiation effect and the antisenecence effect of PSt-PAAm-IGF-1/TNF-α biomaterial which was selected from the three different chargeable polymers in bone marrow mesenchymal stem cells (BMSCs). Our work will establish a model for studying the biochemical molecular regulation mechanism and signal transduction pathway of cell senescence in liver tissue engineering, which provide a molecular basis for developing biomaterials for liver tissue engineering.
Collapse
Affiliation(s)
- Runcai Yang
- School of Life Science, South China Normal University , Guangzhou 510631, China
| | - Lifang Wu
- School of Life Science, South China Normal University , Guangzhou 510631, China
| | - Jiehong Chen
- School of Life Science, South China Normal University , Guangzhou 510631, China
| | - Wuya Chen
- School of Life Science, South China Normal University , Guangzhou 510631, China
| | - Lin Zhang
- School of Life Science, South China Normal University , Guangzhou 510631, China
| | - Li Zhang
- School of Life Science, South China Normal University , Guangzhou 510631, China
| | - Rong You
- School of Life Science, South China Normal University , Guangzhou 510631, China
| | - Liang Yin
- School of Life Science, South China Normal University , Guangzhou 510631, China
| | - Chu-Hua Li
- School of Life Science, South China Normal University , Guangzhou 510631, China
| | - Yan-Qing Guan
- School of Life Science, South China Normal University , Guangzhou 510631, China
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University , Guangzhou 510631, China
| |
Collapse
|
35
|
Chandra T, Kirschner K. Chromosome organisation during ageing and senescence. Curr Opin Cell Biol 2016; 40:161-167. [PMID: 27101466 DOI: 10.1016/j.ceb.2016.03.020] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 03/08/2016] [Accepted: 03/28/2016] [Indexed: 02/01/2023]
Abstract
Acute cellular stress caused by oncogene activation or high levels of DNA damage can engage a tumour suppressive response, which can lead to cellular senescence. Chronic cellular stress evoked by low levels of DNA damage or telomere erosion is involved in the ageing process. In oncogene induced senescence in fibroblasts, a dramatic rearrangement of heterochromatin into foci and accumulation of constitutive heterochromatin is well documented. In contrast, a loss of heterochromatin has been described in replicative senescence and premature ageing syndromes. The distinct nuclear phenotypes that accompany the stress response highlight the differences between acute and chronic stress models, and this review will address the differences and similarities between these models with a focus on chromosome organisation and heterochromatin.
Collapse
Affiliation(s)
- Tamir Chandra
- Epigenetics Programme, The Babraham Institute, Cambridge CB22 3AT, UK; The Wellcome Trust Sanger Institute, Cambridge CB10 1SA, UK.
| | - Kristina Kirschner
- Cambridge Institute for Medical Research, Wellcome Trust/MRC Stem Cell Institute and Department of Haematology, University of Cambridge, Hills Road, Cambridge CB2 0XY, UK.
| |
Collapse
|
36
|
Borghesan M, Fusilli C, Rappa F, Panebianco C, Rizzo G, Oben JA, Mazzoccoli G, Faulkes C, Pata I, Agodi A, Rezaee F, Minogue S, Warren A, Peterson A, Sedivy JM, Douet J, Buschbeck M, Cappello F, Mazza T, Pazienza V, Vinciguerra M. DNA Hypomethylation and Histone Variant macroH2A1 Synergistically Attenuate Chemotherapy-Induced Senescence to Promote Hepatocellular Carcinoma Progression. Cancer Res 2016; 76:594-606. [PMID: 26772755 DOI: 10.1158/0008-5472.can-15-1336] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 11/02/2015] [Indexed: 12/11/2022]
Abstract
Aging is a major risk factor for progression of liver diseases to hepatocellular carcinoma (HCC). Cellular senescence contributes to age-related tissue dysfunction, but the epigenetic basis underlying drug-induced senescence remains unclear. macroH2A1, a variant of histone H2A, is a marker of senescence-associated heterochromatic foci that synergizes with DNA methylation to silence tumor-suppressor genes in human fibroblasts. In this study, we investigated the relationship between macroH2A1 splice variants, macroH2A1.1 and macroH2A1.2, and liver carcinogenesis. We found that protein levels of both macroH2A1 isoforms were increased in the livers of very elderly rodents and humans, and were robust immunohistochemical markers of human cirrhosis and HCC. In response to the chemotherapeutic and DNA-demethylating agent 5-aza-deoxycytidine (5-aza-dC), transgenic expression of macroH2A1 isoforms in HCC cell lines prevented the emergence of a senescent-like phenotype and induced synergistic global DNA hypomethylation. Conversely, macroH2A1 depletion amplified the antiproliferative effects of 5-aza-dC in HCC cells, but failed to enhance senescence. Senescence-associated secretory phenotype and whole-transcriptome analyses implicated the p38 MAPK/IL8 pathway in mediating macroH2A1-dependent escape of HCC cells from chemotherapy-induced senescence. Furthermore, chromatin immunoprecipitation sequencing revealed that this hepatic antisenescence state also required active transcription that could not be attributed to genomic occupancy of these histones. Collectively, our findings reveal a new mechanism by which drug-induced senescence is epigenetically regulated by macroH2A1 and DNA methylation and suggest macroH2A1 as a novel biomarker of hepatic senescence that could potentially predict prognosis and disease progression.
Collapse
Affiliation(s)
- Michela Borghesan
- Institute for Liver and Digestive Health, Royal Free Hospital, University College London, London, United Kingdom. Division of Internal Medicine, Department of Medical Sciences, IRCCS "Casa Sollievo della Sofferenza" Hospital, San Giovanni Rotondo, Italy
| | - Caterina Fusilli
- Bioinformatics Unit, IRCCS "Casa Sollievo della Sofferenza"-Mendel Laboratory, Rome, Italy
| | - Francesca Rappa
- Department of Experimental Biomedicine and Clinical Neurosciences, Section of Human Anatomy, University of Palermo, Palermo, Italy
| | - Concetta Panebianco
- Gastroenterology Unit, IRCCS "Casa Sollievo della Sofferenza" Hospital, San Giovanni Rotondo, Italy
| | - Giovanni Rizzo
- Institute for Liver and Digestive Health, Royal Free Hospital, University College London, London, United Kingdom
| | - Jude A Oben
- Institute for Liver and Digestive Health, Royal Free Hospital, University College London, London, United Kingdom
| | - Gianluigi Mazzoccoli
- Division of Internal Medicine, Department of Medical Sciences, IRCCS "Casa Sollievo della Sofferenza" Hospital, San Giovanni Rotondo, Italy
| | - Chris Faulkes
- School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom
| | - Illar Pata
- Department of Gene Technology, Tallinn University of Technology (TTU), IVEX Lab, Tallinn, Estonia
| | - Antonella Agodi
- Department GF Ingrassia, University of Catania, Catania, Italy
| | - Farhad Rezaee
- Department of Cell Biology, University Medical Center Groningen, Groningen, the Netherlands
| | - Shane Minogue
- Institute for Liver and Digestive Health, Royal Free Hospital, University College London, London, United Kingdom
| | - Alessandra Warren
- Institute for Liver and Digestive Health, Royal Free Hospital, University College London, London, United Kingdom. Centre for Education and Research on Aging (CERA) and the ANZAC Research Institute, Concord RG Hospital, University of Sydney, Sydney, Australia
| | - Abigail Peterson
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island
| | - John M Sedivy
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island
| | - Julien Douet
- Institute for Predictive and Personalized Medicine of Cancer, Campus Can Ruti, Badalona, Spain. Josep Carreras Institute for Leukaemia Research, Campus ICO-HGTP, Campus Can Ruti, Badalona, Spain
| | - Marcus Buschbeck
- Institute for Predictive and Personalized Medicine of Cancer, Campus Can Ruti, Badalona, Spain. Josep Carreras Institute for Leukaemia Research, Campus ICO-HGTP, Campus Can Ruti, Badalona, Spain
| | - Francesco Cappello
- Department of Experimental Biomedicine and Clinical Neurosciences, Section of Human Anatomy, University of Palermo, Palermo, Italy. Euro-Mediterranean Institute of Science and Technology (IEMEST), Palermo, Italy
| | - Tommaso Mazza
- Bioinformatics Unit, IRCCS "Casa Sollievo della Sofferenza"-Mendel Laboratory, Rome, Italy
| | - Valerio Pazienza
- Gastroenterology Unit, IRCCS "Casa Sollievo della Sofferenza" Hospital, San Giovanni Rotondo, Italy
| | - Manlio Vinciguerra
- Institute for Liver and Digestive Health, Royal Free Hospital, University College London, London, United Kingdom. Gastroenterology Unit, IRCCS "Casa Sollievo della Sofferenza" Hospital, San Giovanni Rotondo, Italy. Euro-Mediterranean Institute of Science and Technology (IEMEST), Palermo, Italy. School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom.
| |
Collapse
|
37
|
The role of the chromatin assembly complex (CAF-1) and its p60 subunit (CHAF1b) in homeostasis and disease. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2015; 1849:979-86. [PMID: 26066981 DOI: 10.1016/j.bbagrm.2015.05.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 05/22/2015] [Accepted: 05/25/2015] [Indexed: 11/23/2022]
Abstract
Nucleosome assembly following DNA synthesis is critical for maintaining genomic stability. The proteins directly responsible for shuttling newly synthesized histones H3 and H4 from the cytoplasm to the assembly fork during DNA replication comprise the Chromatin Assembly Factor 1 complex (CAF-1). Whereas the diverse functions of the large (CAF-1-p150, CHAF1a) and small (RbAp48, p48) subunits of the CAF-1 complex have been well-characterized in many tissues and extend beyond histone chaperone activity, the contributions of the medium subunit (CAF-1-p60, CHAF1b) are much less well understood. Although it is known that CHAF1b has multiple functional domains (7× WD repeat domain, B-like domain, and a PEST domain), how these components come together to elicit the functions of this protein are still unclear. Here, we review the biology of the CAF-1 complex, with an emphasis on CHAF1b, including its structure, regulation, and function. In addition, we discuss the possible contributions of CHAF1b and the CAF-1 complex to human diseases. Of note, CHAF1b is located within the Down syndrome critical region (DSCR) of chromosome 21. Therefore, we also address the putative contributions of its trisomy to the various manifestations of DS.
Collapse
|
38
|
Saade E, Pirozhkova I, Aimbetov R, Lipinski M, Ogryzko V. Molecular turnover, the H3.3 dilemma and organismal aging (hypothesis). Aging Cell 2015; 14:322-33. [PMID: 25720734 PMCID: PMC4406661 DOI: 10.1111/acel.12332] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/28/2015] [Indexed: 12/22/2022] Open
Abstract
The H3.3 histone variant has been a subject of increasing interest in the field of chromatin studies due to its two distinguishing features. First, its incorporation into chromatin is replication independent unlike the replication-coupled deposition of its canonical counterparts H3.1/2. Second, H3.3 has been consistently associated with an active state of chromatin. In accordance, this histone variant should be expected to be causally involved in the regulation of gene expression, or more generally, its incorporation should have downstream consequences for the structure and function of chromatin. This, however, leads to an apparent paradox: In cells that slowly replicate in the organism, H3.3 will accumulate with time, opening the way to aberrant effects on heterochromatin. Here, we review the indications that H3.3 is expected both to be incorporated in the heterochromatin of slowly replicating cells and to retain its functional downstream effects. Implications for organismal aging are discussed.
Collapse
Affiliation(s)
- Evelyne Saade
- Faculty of Public Health Lebanese University LU Beirut Lebanon
| | - Iryna Pirozhkova
- Institute Gustave Roussy University Paris SUD 114, rue Edouard Vaillant Villejuif 94805France
| | - Rakhan Aimbetov
- Institute Gustave Roussy University Paris SUD 114, rue Edouard Vaillant Villejuif 94805France
| | - Marc Lipinski
- Institute Gustave Roussy University Paris SUD 114, rue Edouard Vaillant Villejuif 94805France
| | - Vasily Ogryzko
- Institute Gustave Roussy University Paris SUD 114, rue Edouard Vaillant Villejuif 94805France
| |
Collapse
|
39
|
Piano A, Titorenko VI. The Intricate Interplay between Mechanisms Underlying Aging and Cancer. Aging Dis 2015; 6:56-75. [PMID: 25657853 PMCID: PMC4306474 DOI: 10.14336/ad.2014.0209] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Revised: 01/30/2014] [Accepted: 02/09/2014] [Indexed: 12/15/2022] Open
Abstract
Age is the major risk factor in the incidence of cancer, a hyperplastic disease associated with aging. Here, we discuss the complex interplay between mechanisms underlying aging and cancer as a reciprocal relationship. This relationship progresses with organismal age, follows the history of cell proliferation and senescence, is driven by common or antagonistic causes underlying aging and cancer in an age-dependent fashion, and is maintained via age-related convergent and divergent mechanisms. We summarize our knowledge of these mechanisms, outline the most important unanswered questions and suggest directions for future research.
Collapse
Affiliation(s)
- Amanda Piano
- Department of Biology, Concordia University, Montreal, Quebec, Canada
| | | |
Collapse
|
40
|
Duarte LF, Young ARJ, Wang Z, Wu HA, Panda T, Kou Y, Kapoor A, Hasson D, Mills NR, Ma’ayan A, Narita M, Bernstein E. Histone H3.3 and its proteolytically processed form drive a cellular senescence programme. Nat Commun 2014; 5:5210. [PMID: 25394905 PMCID: PMC4235654 DOI: 10.1038/ncomms6210] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 09/09/2014] [Indexed: 01/24/2023] Open
Abstract
The process of cellular senescence generates a repressive chromatin environment, however, the role of histone variants and histone proteolytic cleavage in senescence remains unclear. Here, using models of oncogene-induced and replicative senescence, we report novel histone H3 tail cleavage events mediated by the protease Cathepsin L. We find that cleaved forms of H3 are nucleosomal and the histone variant H3.3 is the preferred cleaved form of H3. Ectopic expression of H3.3 and its cleavage product (H3.3cs1), which lacks the first 21 amino acids of the H3 tail, is sufficient to induce senescence. Further, H3.3cs1 chromatin incorporation is mediated by the HUCA histone chaperone complex. Genome-wide transcriptional profiling revealed that H3.3cs1 facilitates transcriptional silencing of cell cycle regulators including RB/E2F target genes, likely via the permanent removal of H3K4me3. Collectively, our study identifies histone H3.3 and its proteolytically processed forms as key regulators of cellular senescence.
Collapse
Affiliation(s)
- Luis F. Duarte
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Andrew R. J. Young
- Cancer Research UK Cambridge Institute, University of Cambridge, The Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - Zichen Wang
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Department of Pharmacology and Systems Therapeutics and Systems Biology Center New York (SBCNY); Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Hsan-Au Wu
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Taniya Panda
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Yan Kou
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Department of Pharmacology and Systems Therapeutics and Systems Biology Center New York (SBCNY); Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Avnish Kapoor
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Dan Hasson
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Nicholas R. Mills
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Avi Ma’ayan
- Department of Pharmacology and Systems Therapeutics and Systems Biology Center New York (SBCNY); Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Masashi Narita
- Cancer Research UK Cambridge Institute, University of Cambridge, The Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - Emily Bernstein
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| |
Collapse
|
41
|
Evertts AG, Manning AL, Wang X, Dyson NJ, Garcia BA, Coller HA. H4K20 methylation regulates quiescence and chromatin compaction. Mol Biol Cell 2013; 24:3025-37. [PMID: 23924899 PMCID: PMC3784377 DOI: 10.1091/mbc.e12-07-0529] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The methylation state of K20 on histone H4 is important for proper cell cycle control and chromatin compaction in human fibroblasts. High levels of dimethylated and trimethylated K20 are associated with quiescence, and loss of these modifications causes a more open chromatin conformation and defects in cell cycle progression and exit. The transition between proliferation and quiescence is frequently associated with changes in gene expression, extent of chromatin compaction, and histone modifications, but whether changes in chromatin state actually regulate cell cycle exit with quiescence is unclear. We find that primary human fibroblasts induced into quiescence exhibit tighter chromatin compaction. Mass spectrometry analysis of histone modifications reveals that H4K20me2 and H4K20me3 increase in quiescence and other histone modifications are present at similar levels in proliferating and quiescent cells. Analysis of cells in S, G2/M, and G1 phases shows that H4K20me1 increases after S phase and is converted to H4K20me2 and H4K20me3 in quiescence. Knockdown of the enzyme that creates H4K20me3 results in an increased fraction of cells in S phase, a defect in exiting the cell cycle, and decreased chromatin compaction. Overexpression of Suv4-20h1, the enzyme that creates H4K20me2 from H4K20me1, results in G2 arrest, consistent with a role for H4K20me1 in mitosis. The results suggest that the same lysine on H4K20 may, in its different methylation states, facilitate mitotic functions in M phase and promote chromatin compaction and cell cycle exit in quiescent cells.
Collapse
Affiliation(s)
- Adam G Evertts
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544 Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA 02129 Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA 19104 Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, and Department of Biological Chemistry, David Geffen School of Medicine, Los Angeles, CA 90095
| | | | | | | | | | | |
Collapse
|
42
|
Sasai N, Saitoh N, Saitoh H, Nakao M. The transcriptional cofactor MCAF1/ATF7IP is involved in histone gene expression and cellular senescence. PLoS One 2013; 8:e68478. [PMID: 23935871 PMCID: PMC3728336 DOI: 10.1371/journal.pone.0068478] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2013] [Accepted: 05/31/2013] [Indexed: 12/27/2022] Open
Abstract
Cellular senescence is post-mitotic or oncogene-induced events combined with nuclear remodeling. MCAF1 (also known as hAM or ATF7IP), a transcriptional cofactor that is overexpressed in various cancers, functions in gene activation or repression, depending on interacting partners. In this study, we found that MCAF1 localizes to PML nuclear bodies in human fibroblasts and non-cancerous cells. Interestingly, depletion of MCAF1 in fibroblasts induced premature senescence that was characterized by cell cycle arrest, SA-β-gal activity, and senescence-associated heterochromatic foci (SAHF) formation. Under this condition, core histones and the linker histone H1 significantly decreased at both mRNA and protein levels, resulting in reduced nucleosome formation. Consistently, in activated Ras-induced senescent fibroblasts, the accumulation of MCAF1 in PML bodies was enhanced via the binding of this protein to SUMO molecules, suggesting that sequestration of MCAF1 to PML bodies promotes cellular senescence. Collectively, these results reveal that MCAF1 is an essential regulator of cellular senescence.
Collapse
Affiliation(s)
- Nobuhiro Sasai
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Noriko Saitoh
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Hisato Saitoh
- Department of Biological Sciences, Graduate School of Science and Technology, Kumamoto University, Kumamoto, Japan
| | - Mitsuyoshi Nakao
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
- Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency, Tokyo, Japan
- * E-mail:
| |
Collapse
|
43
|
Terweij M, van Leeuwen F. Histone exchange: sculpting the epigenome. FRONTIERS IN LIFE SCIENCE 2013. [DOI: 10.1080/21553769.2013.838193] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|