1
|
Granger SL, Sharma R, Kaushik V, Razzaghi M, Honda M, Gaur P, Bhat DS, Labenz SM, Heinen JE, Williams BA, Tabei SMA, Wlodarski MW, Antony E, Spies M. Human hnRNPA1 reorganizes telomere-bound replication protein A. Nucleic Acids Res 2024:gkae834. [PMID: 39329264 DOI: 10.1093/nar/gkae834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 09/04/2024] [Accepted: 09/11/2024] [Indexed: 09/28/2024] Open
Abstract
Human replication protein A (RPA) is a heterotrimeric ssDNA binding protein responsible for many aspects of cellular DNA metabolism. Dynamic interactions of the four RPA DNA binding domains (DBDs) with DNA control replacement of RPA by downstream proteins in various cellular metabolic pathways. RPA plays several important functions at telomeres where it binds to and melts telomeric G-quadruplexes, non-canonical DNA structures formed at the G-rich telomeric ssDNA overhangs. Here, we combine single-molecule total internal reflection fluorescence microscopy (smTIRFM) and mass photometry (MP) with biophysical and biochemical analyses to demonstrate that heterogeneous nuclear ribonucleoprotein A1 (hnRNPA1) specifically remodels RPA bound to telomeric ssDNA by dampening the RPA configurational dynamics and forming a ternary complex. Uniquely, among hnRNPA1 target RNAs, telomeric repeat-containing RNA (TERRA) is selectively capable of releasing hnRNPA1 from the RPA-telomeric DNA complex. We speculate that this telomere specific RPA-DNA-hnRNPA1 complex is an important structure in telomere protection.
Collapse
Affiliation(s)
- Sophie L Granger
- Department of Biochemistry and Molecular Biology, University of Iowa Carver College of Medicine, 51 Newton Road, IA City, IA 52242, USA
| | - Richa Sharma
- Department of Hematology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Vikas Kaushik
- Department of Biochemistry and Molecular Biology, St. Louis University School of Medicine, 1250 Carr Lane, St. Louis, MO 63104, USA
| | - Mortezaali Razzaghi
- Department of Biochemistry and Molecular Biology, University of Iowa Carver College of Medicine, 51 Newton Road, IA City, IA 52242, USA
| | - Masayoshi Honda
- Department of Biochemistry and Molecular Biology, University of Iowa Carver College of Medicine, 51 Newton Road, IA City, IA 52242, USA
| | - Paras Gaur
- Department of Biochemistry and Molecular Biology, University of Iowa Carver College of Medicine, 51 Newton Road, IA City, IA 52242, USA
| | - Divya S Bhat
- Department of Biochemistry and Molecular Biology, University of Iowa Carver College of Medicine, 51 Newton Road, IA City, IA 52242, USA
| | - Sabryn M Labenz
- Department of Physics, University of Northern Iowa, Cedar Falls, IA 50614, USA
| | - Jenna E Heinen
- Department of Physics, University of Northern Iowa, Cedar Falls, IA 50614, USA
| | - Blaine A Williams
- Department of Physics, University of Northern Iowa, Cedar Falls, IA 50614, USA
| | - S M Ali Tabei
- Department of Physics, University of Northern Iowa, Cedar Falls, IA 50614, USA
| | - Marcin W Wlodarski
- Department of Hematology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Edwin Antony
- Department of Biochemistry and Molecular Biology, St. Louis University School of Medicine, 1250 Carr Lane, St. Louis, MO 63104, USA
| | - Maria Spies
- Department of Biochemistry and Molecular Biology, University of Iowa Carver College of Medicine, 51 Newton Road, IA City, IA 52242, USA
| |
Collapse
|
2
|
Kallingal A, Krzemieniecki R, Maciejewska N, Brankiewicz-Kopcińska W, Baginski M. TRF1 and TRF2: pioneering targets in telomere-based cancer therapy. J Cancer Res Clin Oncol 2024; 150:353. [PMID: 39012375 PMCID: PMC11252209 DOI: 10.1007/s00432-024-05867-3] [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: 04/30/2024] [Accepted: 06/25/2024] [Indexed: 07/17/2024]
Abstract
This article presents an in-depth exploration of the roles of Telomere Repeat-binding Factors 1 and 2 (TRF1 and TRF2), and the shelterin complex, in the context of cancer biology. It emphasizes their emerging significance as potential biomarkers and targets for therapeutic intervention. Central to the shelterin complex, TRF1 and TRF2 are crucial in maintaining telomere integrity and genomic stability, their dysregulation often being a hallmark of cancerous cells. The article delves into the diagnostic and prognostic capabilities of TRF1 and TRF2 across various cancer types, highlighting their sensitivity and specificity. Furthermore, it reviews current strides in drug discovery targeting the shelterin complex, detailing specific compounds and their modes of action. The review candidly addresses the challenges in developing therapies aimed at the shelterin complex, including drug resistance, off-target effects, and issues in drug delivery. By synthesizing recent research findings, the article sheds light on the intricate relationship between telomere biology and cancer development. It underscores the urgency for continued research to navigate the existing challenges and fully leverage the therapeutic potential of TRF1, TRF2, and the shelterin complex in the realm of cancer treatment.
Collapse
Affiliation(s)
- Anoop Kallingal
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdansk University of Technology, Gdansk, 80-233, Poland.
| | - Radosław Krzemieniecki
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdansk University of Technology, Gdansk, 80-233, Poland
| | - Natalia Maciejewska
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdansk University of Technology, Gdansk, 80-233, Poland
| | | | - Maciej Baginski
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdansk University of Technology, Gdansk, 80-233, Poland
| |
Collapse
|
3
|
Stuart A, de Lange T. Replicative senescence is ATM driven, reversible, and accelerated by hyperactivation of ATM at normoxia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.24.600514. [PMID: 38979390 PMCID: PMC11230194 DOI: 10.1101/2024.06.24.600514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Programmed telomere shortening limits tumorigenesis through the induction of replicative senescence. Here we address three long-standing questions concerning senescence. First, we show that the ATM kinase is solely responsible for the induction of replicative senescence. Senescence was delayed by ATM inhibition (ATMi) or overexpression of TRF2, the shelterin subunit dedicated to ATM repression. In contrast, there was no evidence for ATR signaling contributing to replicative senescence even when ATMi was combined with ATR inhibition. Second, we show ATMi can induce apparently normal cell divisions in a subset of senescent cells, indicating that senescence can be reversed. Third, we show that the extended replicative life span at low (physiological) oxygen is due to diminished ATM activity. At low oxygen, cells show a decreased ATM response to dysfunctional telomeres and genome-wide DSBs compared to 20% oxygen. As this effect could be reversed by NAC, the attenuated response of ATM to critically short telomeres and the resulting extended life span at low oxygen is likely due to ROS-induced formation of cysteine disulfide-bridges that crosslink ATM dimers into a form that is not activated by DSBs. These findings show how primary human cells detect shortened telomeres and reveal the molecular mechanism underlying the telomere tumor suppressor pathway.
Collapse
Affiliation(s)
- Alexander Stuart
- Laboratory for Cell Biology and Genetics, Rockefeller University, New York, USA
| | - Titia de Lange
- Laboratory for Cell Biology and Genetics, Rockefeller University, New York, USA
| |
Collapse
|
4
|
Goyer ML, Desaulniers-Langevin C, Sonn A, Mansour Nehmo G, Lisi V, Benabdallah B, Raynal NJM, Beauséjour C. Induced Pluripotent Stem Cell-Derived Fibroblasts Efficiently Engage Senescence Pathways but Show Increased Sensitivity to Stress Inducers. Cells 2024; 13:849. [PMID: 38786071 PMCID: PMC11119907 DOI: 10.3390/cells13100849] [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: 04/24/2024] [Revised: 05/10/2024] [Accepted: 05/13/2024] [Indexed: 05/25/2024] Open
Abstract
The risk of aberrant growth of induced pluripotent stem cell (iPSC)-derived cells in response to DNA damage is a potential concern as the tumor suppressor genes TP53 and CDKN2A are transiently inactivated during reprogramming. Herein, we evaluate the integrity of cellular senescence pathways and DNA double-strand break (DSB) repair in Sendai virus reprogrammed iPSC-derived human fibroblasts (i-HF) compared to their parental skin fibroblasts (HF). Using transcriptomics analysis and a variety of functional assays, we show that the capacity of i-HF to enter senescence and repair DSB is not compromised after damage induced by ionizing radiation (IR) or the overexpression of H-RASV12. Still, i-HF lines are transcriptionally different from their parental lines, showing enhanced metabolic activity and higher expression of p53-related effector genes. As a result, i-HF lines generally exhibit increased sensitivity to various stresses, have an elevated senescence-associated secretory phenotype (SASP), and cannot be immortalized unless p53 expression is knocked down. In conclusion, while our results suggest that i-HF are not at a greater risk of transformation, their overall hyperactivation of senescence pathways may impede their function as a cell therapy product.
Collapse
Affiliation(s)
- Marie-Lyn Goyer
- Centre de Recherche du CHU Sainte-Justine, 3175 Côte Sainte-Catherine, Montréal, QC H3T 1C5, Canada; (M.-L.G.); (C.D.-L.); (A.S.); (G.M.N.); (V.L.); (B.B.); (N.J.-M.R.)
- Département de Pharmacologie et Physiologie, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Cynthia Desaulniers-Langevin
- Centre de Recherche du CHU Sainte-Justine, 3175 Côte Sainte-Catherine, Montréal, QC H3T 1C5, Canada; (M.-L.G.); (C.D.-L.); (A.S.); (G.M.N.); (V.L.); (B.B.); (N.J.-M.R.)
- Département de Pharmacologie et Physiologie, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Anthony Sonn
- Centre de Recherche du CHU Sainte-Justine, 3175 Côte Sainte-Catherine, Montréal, QC H3T 1C5, Canada; (M.-L.G.); (C.D.-L.); (A.S.); (G.M.N.); (V.L.); (B.B.); (N.J.-M.R.)
- Département de Pharmacologie et Physiologie, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Georgio Mansour Nehmo
- Centre de Recherche du CHU Sainte-Justine, 3175 Côte Sainte-Catherine, Montréal, QC H3T 1C5, Canada; (M.-L.G.); (C.D.-L.); (A.S.); (G.M.N.); (V.L.); (B.B.); (N.J.-M.R.)
- Département de Pharmacologie et Physiologie, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Véronique Lisi
- Centre de Recherche du CHU Sainte-Justine, 3175 Côte Sainte-Catherine, Montréal, QC H3T 1C5, Canada; (M.-L.G.); (C.D.-L.); (A.S.); (G.M.N.); (V.L.); (B.B.); (N.J.-M.R.)
| | - Basma Benabdallah
- Centre de Recherche du CHU Sainte-Justine, 3175 Côte Sainte-Catherine, Montréal, QC H3T 1C5, Canada; (M.-L.G.); (C.D.-L.); (A.S.); (G.M.N.); (V.L.); (B.B.); (N.J.-M.R.)
| | - Noël J.-M. Raynal
- Centre de Recherche du CHU Sainte-Justine, 3175 Côte Sainte-Catherine, Montréal, QC H3T 1C5, Canada; (M.-L.G.); (C.D.-L.); (A.S.); (G.M.N.); (V.L.); (B.B.); (N.J.-M.R.)
- Département de Pharmacologie et Physiologie, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Christian Beauséjour
- Centre de Recherche du CHU Sainte-Justine, 3175 Côte Sainte-Catherine, Montréal, QC H3T 1C5, Canada; (M.-L.G.); (C.D.-L.); (A.S.); (G.M.N.); (V.L.); (B.B.); (N.J.-M.R.)
- Département de Pharmacologie et Physiologie, Université de Montréal, Montréal, QC H3T 1J4, Canada
| |
Collapse
|
5
|
Granger SL, Sharma R, Kaushik V, Razzaghi M, Honda M, Gaur P, Bhat DS, Labenz SM, Heinen JE, Williams BA, Tabei SMA, Wlodarski MW, Antony E, Spies M. Human hnRNPA1 reorganizes telomere-bound Replication Protein A. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.05.09.540056. [PMID: 37214874 PMCID: PMC10197631 DOI: 10.1101/2023.05.09.540056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Human replication protein A (RPA) is a heterotrimeric ssDNA binding protein responsible for many aspects of cellular DNA metabolism. Dynamic interactions of the four RPA DNA binding domains (DBDs) with DNA control replacement of RPA by downstream proteins in various cellular metabolic pathways. RPA plays several important functions at telomeres where it binds to and melts telomeric G-quadruplexes, non-canonical DNA structures formed at the G-rich telomeric ssDNA overhangs. Here, we combine single-molecule total internal reflection fluorescence microscopy (smTIRFM) and mass photometry (MP) with biophysical and biochemical analyses to demonstrate that heterogeneous nuclear ribonucleoprotein A1 (hnRNPA1) specifically remodels RPA bound to telomeric ssDNA by dampening the RPA configurational dynamics and forming a ternary complex. Uniquely, among hnRNPA1 target RNAs, telomeric repeat-containing RNA (TERRA) is selectively capable of releasing hnRNPA1 from the RPA-telomeric DNA complex. We speculate that this telomere specific RPA-DNA-hnRNPA1 complex is an important structure in telomere protection. One Sentence Summary At the single-stranded ends of human telomeres, the heterogeneous nuclear ribonucleoprotein A1 (hnRNPA1) binds to and modulates conformational dynamics of the ssDNA binding protein RPA forming a ternary complex which is controlled by telomeric repeat-containing RNA (TERRA).
Collapse
|
6
|
Rodseth E, Sumasgutner P, Tate G, Nilsson JF, Watson H, Maritz MF, Ingle RA, Amar A. Pleiotropic effects of melanin pigmentation: haemoparasite infection intensity but not telomere length is associated with plumage morph in black sparrowhawks. ROYAL SOCIETY OPEN SCIENCE 2024; 11:230370. [PMID: 38577209 PMCID: PMC10987988 DOI: 10.1098/rsos.230370] [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: 03/23/2023] [Revised: 09/21/2023] [Accepted: 02/13/2024] [Indexed: 04/06/2024]
Abstract
There is increasing recognition of the potential pleiotropic effects of melanin pigmentation, particularly on immunity, with reports of variation in haemoparasite infection intensity and immune responses between the morphs of colour-polymorphic bird species. In a population of the black sparrowhawk (Accipiter melanoleucus) in western South Africa, light morphs have a higher haemoparasite infection intensity, but no physiological effects of this are apparent. Here, we investigate the possible effects of haemoparasite infection on telomere length in this species and explore whether relative telomere length is associated with either plumage morph or sex. Using quantitative polymerase chain reaction analysis, we confirmed that dark morphs had a lower haemoparasite infection intensity than light morphs. However, we found no differences in telomere length associated with either the haemoparasite infection status or morph in adults, although males have longer telomeres than females. While differences in haemoparasite intensity between morphs are consistent with pleiotropic effects of melanin pigmentation in the black sparrowhawk, we found no evidence that telomere length was associated with haemoparasite infection. Further work is needed to investigate the implications of possible pleiotropic effects of plumage morph and their potential role in the maintenance of colour polymorphism in this species.
Collapse
Affiliation(s)
- Edmund Rodseth
- Department of Molecular and Cell Biology, University of Cape Town, Cape Town, South Africa
| | - Petra Sumasgutner
- FitzPatrick Institute of African Ornithology, University of Cape Town, Cape Town, South Africa
- Konrad Lorenz Research Centre, Department of Behavioural and Cognitive Biology, University of Vienna, Vienna, Austria
| | - Gareth Tate
- FitzPatrick Institute of African Ornithology, University of Cape Town, Cape Town, South Africa
- Birds of Prey Programme, Endangered Wildlife Trust, Midrand, South Africa
| | - Johan F. Nilsson
- Evolutionary Ecology, Department of Biology, Lund University, Lund, Sweden
| | - Hannah Watson
- Evolutionary Ecology, Department of Biology, Lund University, Lund, Sweden
| | - Michelle F. Maritz
- Department of Molecular and Cell Biology, University of Cape Town, Cape Town, South Africa
| | - Robert A. Ingle
- Department of Molecular and Cell Biology, University of Cape Town, Cape Town, South Africa
| | - Arjun Amar
- FitzPatrick Institute of African Ornithology, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
7
|
Yang B, Lin Y, Huang Y, Shen YQ, Chen Q. Thioredoxin (Trx): A redox target and modulator of cellular senescence and aging-related diseases. Redox Biol 2024; 70:103032. [PMID: 38232457 PMCID: PMC10827563 DOI: 10.1016/j.redox.2024.103032] [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: 10/14/2023] [Revised: 12/03/2023] [Accepted: 01/04/2024] [Indexed: 01/19/2024] Open
Abstract
Thioredoxin (Trx) is a compact redox-regulatory protein that modulates cellular redox state by reducing oxidized proteins. Trx exhibits dual functionality as an antioxidant and a cofactor for diverse enzymes and transcription factors, thereby exerting influence over their activity and function. Trx has emerged as a pivotal biomarker for various diseases, particularly those associated with oxidative stress, inflammation, and aging. Recent clinical investigations have underscored the significance of Trx in disease diagnosis, treatment, and mechanistic elucidation. Despite its paramount importance, the intricate interplay between Trx and cellular senescence-a condition characterized by irreversible growth arrest induced by multiple aging stimuli-remains inadequately understood. In this review, our objective is to present a comprehensive and up-to-date overview of the structure and function of Trx, its involvement in redox signaling pathways and cellular senescence, its association with aging and age-related diseases, as well as its potential as a therapeutic target. Our review aims to elucidate the novel and extensive role of Trx in senescence while highlighting its implications for aging and age-related diseases.
Collapse
Affiliation(s)
- Bowen Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Yumeng Lin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Yibo Huang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Ying-Qiang Shen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Qianming Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
8
|
Lipskaia L, Breau M, Cayrou C, Churikov D, Braud L, Jacquet J, Born E, Fouillade C, Curras-Alonso S, Bauwens S, Jourquin F, Fiore F, Castellano R, Josselin E, Sánchez-Ferrer C, Giovinazzo G, Lachaud C, Gilson E, Flores I, Londono-Vallejo A, Adnot S, Géli V. mTert induction in p21-positive cells counteracts capillary rarefaction and pulmonary emphysema. EMBO Rep 2024; 25:1650-1684. [PMID: 38424230 PMCID: PMC10933469 DOI: 10.1038/s44319-023-00041-1] [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: 10/06/2023] [Revised: 12/12/2023] [Accepted: 12/14/2023] [Indexed: 03/02/2024] Open
Abstract
Lung diseases develop when telomeres shorten beyond a critical point. We constructed a mouse model in which the catalytic subunit of telomerase (mTert), or its catalytically inactive form (mTertCI), is expressed from the p21Cdkn1a locus. Expression of either TERT or TERTCI reduces global p21 levels in the lungs of aged mice, highlighting TERT non-canonical function. However, only TERT reduces accumulation of very short telomeres, oxidative damage, endothelial cell (ECs) senescence and senile emphysema in aged mice. Single-cell analysis of the lung reveals that p21 (and hence TERT) is expressed mainly in the capillary ECs. We report that a fraction of capillary ECs marked by CD34 and endowed with proliferative capacity declines drastically with age, and this is counteracted by TERT but not TERTCI. Consistently, only TERT counteracts decline of capillary density. Natural aging effects are confirmed using the experimental model of emphysema induced by VEGFR2 inhibition and chronic hypoxia. We conclude that catalytically active TERT prevents exhaustion of the putative CD34 + EC progenitors with age, thus protecting against capillary vessel loss and pulmonary emphysema.
Collapse
Affiliation(s)
- Larissa Lipskaia
- Institute for Lung Health, Justus Liebig University, Giessen, Germany
- INSERM U955 and Département de Physiologie, Hôpital Henri Mondor, FHU SENEC, AP-HP, 94010, Créteil, and Université Paris-Est Créteil (UPEC), Paris, France
| | - Marielle Breau
- Marseille Cancer Research Centre (CRCM), U1068 INSERM, UMR7258 CNRS, UM105 Aix-Marseille University, Institut Paoli-Calmettes, Ligue Nationale Contre le Cancer (Equipe labellisée), Team Telomeres and Chromatin, Marseille, France
| | - Christelle Cayrou
- Marseille Cancer Research Centre (CRCM), U1068 INSERM, UMR7258 CNRS, UM105 Aix-Marseille University, Institut Paoli-Calmettes, Ligue Nationale Contre le Cancer (Equipe labellisée), Team Telomeres and Chromatin, Marseille, France
| | - Dmitri Churikov
- Marseille Cancer Research Centre (CRCM), U1068 INSERM, UMR7258 CNRS, UM105 Aix-Marseille University, Institut Paoli-Calmettes, Ligue Nationale Contre le Cancer (Equipe labellisée), Team Telomeres and Chromatin, Marseille, France
| | - Laura Braud
- Marseille Cancer Research Centre (CRCM), U1068 INSERM, UMR7258 CNRS, UM105 Aix-Marseille University, Institut Paoli-Calmettes, Ligue Nationale Contre le Cancer (Equipe labellisée), Team Telomeres and Chromatin, Marseille, France
| | - Juliette Jacquet
- Institute for Lung Health, Justus Liebig University, Giessen, Germany
| | - Emmanuelle Born
- Institute for Lung Health, Justus Liebig University, Giessen, Germany
| | - Charles Fouillade
- Institut Curie, Inserm U1021, CNRS UMR 3347, University Paris-Saclay, PSL Research University, Orsay, France
| | - Sandra Curras-Alonso
- Institut Curie, PSL Research University, CNRS UMR3244, Sorbonne Université, Telomeres and Cancer, 75005, Paris, France
| | - Serge Bauwens
- Université Côte d'Azur, CNRS, Inserm, IRCAN, Faculty of Medicine, Nice, France
| | - Frederic Jourquin
- Marseille Cancer Research Centre (CRCM), U1068 INSERM, UMR7258 CNRS, UM105 Aix-Marseille University, Institut Paoli-Calmettes, Ligue Nationale Contre le Cancer (Equipe labellisée), Team Telomeres and Chromatin, Marseille, France
| | - Frederic Fiore
- Centre d'Immunophénomique, Aix Marseille Université, INSERM, CNRS UMR, Marseille, France
| | - Rémy Castellano
- Marseille Cancer Research Centre (CRCM), TrGET Preclinical Platform, Institut Paoli-Calmettes, Inserm, CNRS, Aix Marseille Université, Marseille, France
| | - Emmanuelle Josselin
- Marseille Cancer Research Centre (CRCM), TrGET Preclinical Platform, Institut Paoli-Calmettes, Inserm, CNRS, Aix Marseille Université, Marseille, France
| | | | - Giovanna Giovinazzo
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, 28029, Madrid, Spain
| | - Christophe Lachaud
- Marseille Cancer Research Centre (CRCM), U1068 INSERM, UMR7258 CNRS, UM105 Aix-Marseille University, Institut Paoli-Calmettes, Team DNA Interstrand Crosslink Lesions and Blood Disorders, Marseille, France
| | - Eric Gilson
- Université Côte d'Azur, CNRS, Inserm, IRCAN, Faculty of Medicine, Nice, France
| | - Ignacio Flores
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, 28029, Madrid, Spain
- Centro de Biologia Molecular Severo Ochoa, CSIC-UAM, Cantoblanco, Madrid, Spain
| | - Arturo Londono-Vallejo
- Institut Curie, PSL Research University, CNRS UMR3244, Sorbonne Université, Telomeres and Cancer, 75005, Paris, France
| | - Serge Adnot
- Institute for Lung Health, Justus Liebig University, Giessen, Germany.
- INSERM U955 and Département de Physiologie, Hôpital Henri Mondor, FHU SENEC, AP-HP, 94010, Créteil, and Université Paris-Est Créteil (UPEC), Paris, France.
| | - Vincent Géli
- Marseille Cancer Research Centre (CRCM), U1068 INSERM, UMR7258 CNRS, UM105 Aix-Marseille University, Institut Paoli-Calmettes, Ligue Nationale Contre le Cancer (Equipe labellisée), Team Telomeres and Chromatin, Marseille, France.
| |
Collapse
|
9
|
Lim CJ. Telomere C-Strand Fill-In Machinery: New Insights into the Human CST-DNA Polymerase Alpha-Primase Structures and Functions. Subcell Biochem 2024; 104:73-100. [PMID: 38963484 DOI: 10.1007/978-3-031-58843-3_5] [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] [Indexed: 07/05/2024]
Abstract
Telomeres at the end of eukaryotic chromosomes are extended by a specialized set of enzymes and telomere-associated proteins, collectively termed here the telomere "replisome." The telomere replisome acts on a unique replicon at each chromosomal end of the telomeres, the 3' DNA overhang. This telomere replication process is distinct from the replisome mechanism deployed to duplicate the human genome. The G-rich overhang is first extended before the complementary C-strand is filled in. This overhang is extended by telomerase, a specialized ribonucleoprotein and reverse transcriptase. The overhang extension process is terminated when telomerase is displaced by CTC1-STN1-TEN1 (CST), a single-stranded DNA-binding protein complex. CST then recruits DNA polymerase α-primase to complete the telomere replication process by filling in the complementary C-strand. In this chapter, the recent structure-function insights into the human telomere C-strand fill-in machinery (DNA polymerase α-primase and CST) will be discussed.
Collapse
Affiliation(s)
- Ci Ji Lim
- Department of Biochemistry, College of Agricultural and Life Sciences, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
10
|
Vazifehmand R, Ali DS, Homaie FM, Jalalvand FM, Othman Z, Deming C, Stanslas J, Sekawi Z. Effects of HSV-G47Δ Oncolytic Virus on Telomerase and Telomere Length Alterations in Glioblastoma Multiforme Cancer Stem Cells Under Hypoxia and Normoxia Conditions. Curr Cancer Drug Targets 2024; 24:1262-1274. [PMID: 38357955 DOI: 10.2174/0115680096274769240115165344] [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: 09/02/2023] [Revised: 12/07/2023] [Accepted: 01/01/2024] [Indexed: 02/16/2024]
Abstract
BACKGROUND Due to the existence of tumor stem cells with tumorigenicity properties and resistance patterns, treatment of glioblastoma is not easy. Hypoxia is a major concern in glioblastoma therapy. Telomerase activity and telomere length alterations have been known to play a critical role in glioblastoma progression and invasion. OBJECTIVE This study aimed to investigate the effects of HSV-G47Δ oncolytic virus on telomerase and telomere length alterations in U251GBMCSCs (U251-Glioblastoma cancer stem cells) under hypoxia and normoxia conditions. METHODS U251-CSCs were exposed to the HSV-G47Δ virus in optimized MOI (Multiplicity of infection= 1/14 hours). An absolute telomere length and gene expression of telomerase subunits were determined using an absolute human telomere length quantification PCR assay. Furthermore, a bioinformatics pathway analysis was carried out to evaluate physical and genetic interactions between dysregulated genes with other potential genes and pathways. RESULTS Data revealed that U251CSCs had longer telomeres when exposed to HSV-G47Δ in normoxic conditions but had significantly shorter telomeres in hypoxic conditions. Furthermore, hTERC, DKC1, and TEP1 genes were significantly dysregulated in hypoxic and normoxic microenvironments. The analysis revealed that the expression of TERF2 was significantly reduced in both microenvironments, and two critical genes from the MRN complex, MER11 and RAD50, were significantly upregulated in normoxic conditions. RAD50 showed a significant downregulation pattern in the hypoxic niche. Our results suggested that repair complex in the telomeric structure could be targeted by HSV-G47Δ in both microenvironments. CONCLUSION In the glioblastoma treatment strategy, telomerase and telomere complex could be potential targets for HSV-G47Δ in both microenvironments.
Collapse
Affiliation(s)
- Reza Vazifehmand
- Department of Medical Microbiology & Parasitology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia (UPM), 43400, Serdang, Selangor Darul Ehsan, Malaysia
| | - Dhuha Saeed Ali
- Halal Products Research Institute, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor, Malaysia
| | | | | | - Zulkefley Othman
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia (UPM), 43400, Serdang, Selangor Darul Ehsan, Malaysia
| | - Chau Deming
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia (UPM), 43400, Serdang, Selangor Darul Ehsan, Malaysia
| | - Johnson Stanslas
- Pharmacotherapeutics Unit, Department of Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor, Malaysia
| | - Zamberi Sekawi
- Department of Medical Microbiology & Parasitology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia (UPM), 43400, Serdang, Selangor Darul Ehsan, Malaysia
| |
Collapse
|
11
|
Sobinoff AP, Di Maro S, Low RRJ, Benedetti R, Tomassi S, D'Aniello A, Russo R, Baglivo I, Chianese U, Pedone PV, Chambery A, Cesare AJ, Altucci L, Pickett HA, Cosconati S. Irreversible inhibition of TRF2 TRFH recruiting functions by a covalent cyclic peptide induces telomeric replication stress in cancer cells. Cell Chem Biol 2023; 30:1652-1665.e6. [PMID: 38065101 DOI: 10.1016/j.chembiol.2023.11.008] [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: 07/25/2022] [Revised: 09/28/2023] [Accepted: 11/13/2023] [Indexed: 12/24/2023]
Abstract
The TRF2 shelterin component is an essential regulator of telomere homeostasis and genomic stability. Mutations in the TRF2TRFH domain physically impair t-loop formation and prevent the recruitment of several factors that promote efficient telomere replication, causing telomeric DNA damage. Here, we design, synthesize, and biologically test covalent cyclic peptides that irreversibly target the TRF2TRFH domain. We identify APOD53 as our most promising compound, as it consistently induces a telomeric DNA damage response in cancer cell lines. APOD53 forms a covalent adduct with a reactive cysteine residue present in the TRF2TRFH domain and induces phenotypes consistent with TRF2TRFH domain mutants. These include induction of a telomeric DNA damage response, increased telomeric replication stress, and impaired recruitment of RTEL1 and SLX4 to telomeres. We demonstrate that APOD53 impairs cancer cell growth and find that co-treatment with APOD53 can exacerbate telomere replication stress caused by the G4 stabilizer RHPS4 and low dose aphidicolin (APH).
Collapse
Affiliation(s)
- Alexander P Sobinoff
- Telomere Length Regulation Unit, Children's Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, NSW 2145, Australia
| | - Salvatore Di Maro
- DiSTABiF, University of Campania "Luigi Vanvitelli", Via Vivaldi 43, 81100 Caserta, Italy
| | - Ronnie R J Low
- Genome Integrity Unit, Children's Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, NSW 2145, Australia
| | - Rosaria Benedetti
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Vico L. De Crecchio 7, 80138 Naples, Italy; Azienda Ospedaliera Universitaria "Luigi Vanvitelli", Medical Epigenetics Program
| | - Stefano Tomassi
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, 80131 Naples, Italy
| | - Antonia D'Aniello
- DiSTABiF, University of Campania "Luigi Vanvitelli", Via Vivaldi 43, 81100 Caserta, Italy
| | - Rosita Russo
- DiSTABiF, University of Campania "Luigi Vanvitelli", Via Vivaldi 43, 81100 Caserta, Italy
| | - Ilaria Baglivo
- DiSTABiF, University of Campania "Luigi Vanvitelli", Via Vivaldi 43, 81100 Caserta, Italy
| | - Ugo Chianese
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Vico L. De Crecchio 7, 80138 Naples, Italy
| | - Paolo V Pedone
- DiSTABiF, University of Campania "Luigi Vanvitelli", Via Vivaldi 43, 81100 Caserta, Italy
| | - Angela Chambery
- DiSTABiF, University of Campania "Luigi Vanvitelli", Via Vivaldi 43, 81100 Caserta, Italy
| | - Anthony J Cesare
- Genome Integrity Unit, Children's Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, NSW 2145, Australia
| | - Lucia Altucci
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Vico L. De Crecchio 7, 80138 Naples, Italy; BIOGEM, 83031 Ariano Irpino, Italy; Azienda Ospedaliera Universitaria "Luigi Vanvitelli", Medical Epigenetics Program; IEOS CNR, Napoli, Italy
| | - Hilda A Pickett
- Telomere Length Regulation Unit, Children's Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, NSW 2145, Australia.
| | - Sandro Cosconati
- DiSTABiF, University of Campania "Luigi Vanvitelli", Via Vivaldi 43, 81100 Caserta, Italy.
| |
Collapse
|
12
|
Westin ER, Khodadadi-Jamayran A, Pham LK, Tung ML, Goldman FD. CRISPR screen identifies CEBPB as contributor to dyskeratosis congenita fibroblast senescence via augmented inflammatory gene response. G3 (BETHESDA, MD.) 2023; 13:jkad207. [PMID: 37717172 PMCID: PMC10627266 DOI: 10.1093/g3journal/jkad207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/16/2023] [Accepted: 08/17/2023] [Indexed: 09/18/2023]
Abstract
Aging is the consequence of intra- and extracellular events that promote cellular senescence. Dyskeratosis congenita (DC) is an example of a premature aging disorder caused by underlying telomere/telomerase-related mutations. Cells from these patients offer an opportunity to study telomere-related aging and senescence. Our previous work has found that telomere shortening stimulates DNA damage responses (DDRs) and increases reactive oxygen species (ROS), thereby promoting entry into senescence. This work also found that telomere elongation via TERT expression, the catalytic component of the telomere-elongating enzyme telomerase, or p53 shRNA could decrease ROS by disrupting this telomere-DDR-ROS pathway. To further characterize this pathway, we performed a CRISPR/Cas9 knockout screen to identify genes that extend life span in DC cells. Of the cellular clones isolated due to increased life span, 34% had a guide RNA (gRNA) targeting CEBPB, while gRNAs targeting WSB1, MED28, and p73 were observed multiple times. CEBPB is a transcription factor associated with activation of proinflammatory response genes suggesting that inflammation may be present in DC cells. The inflammatory response was investigated using RNA sequencing to compare DC and control cells. Expression of inflammatory genes was found to be significantly elevated (P < 0.0001) in addition to a key subset of these inflammation-related genes [IL1B, IL6, IL8, IL12A, CXCL1 (GROa), CXCL2 (GROb), and CXCL5]. which are regulated by CEBPB. Exogenous TERT expression led to downregulation of RNA/protein CEBPB expression and the inflammatory response genes suggesting a telomere length-dependent mechanism to regulate CEBPB. Furthermore, unlike exogenous TERT and p53 shRNA, CEBPB shRNA did not significantly decrease ROS suggesting that CEBPB's contribution in DC cells' senescence is ROS independent. Our findings demonstrate a key role for CEBPB in engaging senescence by mobilizing an inflammatory response within DC cells.
Collapse
Affiliation(s)
- Erik R Westin
- Department of Pediatrics, Division of Hematology Oncology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Department of Cancer Precision Medicine, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA 70808, USA
| | - Alireza Khodadadi-Jamayran
- Genome Technology Center, Applied Bioinformatics Laboratories, NYU Langone Medical Center, New York, NY 10016, USA
| | - Linh K Pham
- Department of Pediatrics, Division of Hematology Oncology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Moon Ley Tung
- Stead Family Department of Pediatrics, Division of Medical Genetics and Genomics, University of Iowa, Iowa City, IA 52242, USA
| | - Frederick D Goldman
- Department of Pediatrics, Division of Hematology Oncology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
13
|
Aquilanti E, Kageler L, Watson J, Baird DM, Jones RE, Hodges M, Szegletes ZM, Doench JG, Strathdee CA, Figueroa JRMF, Ligon KL, Beck M, Wen PY, Meyerson M. Telomerase inhibition is an effective therapeutic strategy in TERT promoter-mutant glioblastoma models with low tumor volume. Neuro Oncol 2023; 25:1275-1285. [PMID: 36694348 PMCID: PMC10326479 DOI: 10.1093/neuonc/noad024] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Glioblastoma is one of the most lethal forms of cancer, with 5-year survival rates of only 6%. Glioblastoma-targeted therapeutics have been challenging to develop due to significant inter- and intra-tumoral heterogeneity. Telomerase reverse transcriptase gene (TERT) promoter mutations are the most common known clonal oncogenic mutations in glioblastoma. Telomerase is therefore considered to be a promising therapeutic target against this tumor. However, an important limitation of this strategy is that cell death does not occur immediately after telomerase ablation, but rather after several cell divisions required to reach critically short telomeres. We, therefore, hypothesize that telomerase inhibition would only be effective in glioblastomas with low tumor burden. METHODS We used CRISPR interference to knock down TERT expression in TERT promoter-mutant glioblastoma cell lines and patient-derived models. We then measured viability using serial proliferation assays. We also assessed for features of telomere crisis by measuring telomere length and chromatin bridge formation. Finally, we used a doxycycline-inducible CRISPR interference system to knock down TERT expression in vivo early and late in tumor development. RESULTS Upon TERT inactivation, glioblastoma cells lose their proliferative ability over time and exhibit telomere shortening and chromatin bridge formation. In vivo, survival is only prolonged when TERT knockdown is induced shortly after tumor implantation, but not when the tumor burden is high. CONCLUSIONS Our results support the idea that telomerase inhibition would be most effective at treating glioblastomas with low tumor burden, for example in the adjuvant setting after surgical debulking and chemoradiation.
Collapse
Affiliation(s)
- Elisa Aquilanti
- Division of Neuro Oncology, Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Lauren Kageler
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Jacqueline Watson
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Duncan M Baird
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, UK
| | - Rhiannon E Jones
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, UK
| | - Marie Hodges
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, UK
| | - Zsofia M Szegletes
- Genetic Perturbation Platform, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - John G Doench
- Genetic Perturbation Platform, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Craig A Strathdee
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | | | - Keith L Ligon
- Department of Pathology, Brigham and Women’s Hospital, Boston Children’s Hospital, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Matthew Beck
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Patrick Y Wen
- Division of Neuro Oncology, Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Matthew Meyerson
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Center for Cancer Genomics, Dana Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Genetics and Medicine, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
14
|
Choi YJ, Kim MS, Rhoades JH, Johnson NM, Berry CT, Root S, Chen Q, Tian Y, Fernandez RJ, Cramer Z, Adams-Tzivelekidis S, Li N, Johnson FB, Lengner CJ. Patient-Induced Pluripotent Stem Cell-Derived Hepatostellate Organoids Establish a Basis for Liver Pathologies in Telomeropathies. Cell Mol Gastroenterol Hepatol 2023; 16:451-472. [PMID: 37302654 PMCID: PMC10404563 DOI: 10.1016/j.jcmgh.2023.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 06/02/2023] [Accepted: 06/05/2023] [Indexed: 06/13/2023]
Abstract
BACKGROUND & AIMS Dyskeratosis congenita (DC) is a telomere biology disorder caused primarily by mutations in the DKC1 gene. Patients with DC and related telomeropathies resulting from premature telomere dysfunction experience multiorgan failure. In the liver, DC patients present with nodular hyperplasia, steatosis, inflammation, and cirrhosis. However, the mechanism responsible for telomere dysfunction-induced liver disease remains unclear. METHODS We used isogenic human induced pluripotent stem cells (iPSCs) harboring a causal DC mutation in DKC1 or a CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats/Cas9)-corrected control allele to model DC liver pathologies. We differentiated these iPSCs into hepatocytes (HEPs) or hepatic stellate cells (HSCs) followed by generation of genotype-admixed hepatostellate organoids. Single-cell transcriptomics were applied to hepatostellate organoids to understand cell type-specific genotype-phenotype relationships. RESULTS Directed differentiation of iPSCs into HEPs and stellate cells and subsequent hepatostellate organoid formation revealed a dominant phenotype in the parenchyma, with DC HEPs becoming hyperplastic and also eliciting a pathogenic hyperplastic, proinflammatory response in stellate cells independent of stellate cell genotype. Pathogenic phenotypes in DKC1-mutant HEPs and hepatostellate organoids could be rescued via suppression of serine/threonine kinase AKT (protein kinase B) activity, a central regulator of MYC-driven hyperplasia downstream of DKC1 mutation. CONCLUSIONS Isogenic iPSC-derived admixed hepatostellate organoids offer insight into the liver pathologies in telomeropathies and provide a framework for evaluating emerging therapies.
Collapse
Affiliation(s)
- Young-Jun Choi
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Melissa S Kim
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Joshua H Rhoades
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Nicolette M Johnson
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Corbett T Berry
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sarah Root
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Qijun Chen
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Yuhua Tian
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Rafael J Fernandez
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Zvi Cramer
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Stephanie Adams-Tzivelekidis
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ning Li
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - F Brad Johnson
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| | - Christopher J Lengner
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
15
|
BORZONE FRANCISCORAÚL, GIORELLO MARÍABELÉN, MARTINEZ LEANDROMARCELO, SANMARTIN MARÍACECILIA, FELDMAN LEONARDO, DIMASE FEDERICO, BATAGELJ EMILIO, YANNARELLI GUSTAVO, CHASSEING NORMAALEJANDRA. Senescent mesenchymal stem/stromal cells in pre-metastatic bone marrow of untreated advanced breast cancer patients. Oncol Res 2023; 31:361-374. [PMID: 37305388 PMCID: PMC10229310 DOI: 10.32604/or.2023.028104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 03/30/2023] [Indexed: 06/13/2023] Open
Abstract
Breast cancer is the predominant form of carcinoma among women worldwide, with 70% of advanced patients developing bone metastases, with a high mortality rate. In this sense, the bone marrow (BM) mesenchymal stem/stromal cells (MSCs) are critical for BM/bone homeostasis, and failures in their functionality, transform the BM into a pre-metastatic niche (PMN). We previously found that BM-MSCs from advanced breast cancer patients (BCPs, infiltrative ductal carcinoma, stage III-B) have an abnormal profile. This work aims to study some of the metabolic and molecular mechanisms underlying MSCs shift from a normal to an abnormal profile in this group of patients. A comparative analysis was undertaken, which included self-renewal capacity, morphology, proliferation capacity, cell cycle, reactive oxygen species (ROS) levels, and senescence-associated β‑galactosidase (SA‑β‑gal) staining of BM-derived MSCs isolated from 14 BCPs and 9 healthy volunteers (HVs). Additionally, the expression and activity of the telomerase subunit TERT, as well as telomere length, were measured. Expression levels of pluripotency, osteogenic, and osteoclastogenic genes (OCT-4, SOX-2, M-CAM, RUNX-2, BMP-2, CCL-2, M-CSF, and IL-6) were also determined. The results showed that MSCs from BCPs had reduced ,self-renewal and proliferation capacity. These cells also exhibited inhibited cell cycle progression and phenotypic changes, such as an enlarged and flattened appearance. Additionally, there was an increase in ROS and senescence levels and a decrease in the functional capacity of TERT to preserve telomere length. We also found an increase in pro-inflammatory/pro-osteoclastogenic gene expression and a decrease in pluripotency gene expression. We conclude that these changes could be responsible for the abnormal functional profile that MSCs show in this group of patients.
Collapse
Affiliation(s)
- FRANCISCO RAÚL BORZONE
- Laboratorio de Inmunohematología, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - MARÍA BELÉN GIORELLO
- Laboratorio de Inmunohematología, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - LEANDRO MARCELO MARTINEZ
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medical College, New York, USA
| | - MARÍA CECILIA SANMARTIN
- Laboratorio de Inmunohematología, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Laboratorio de Regulación Génica y Células Madre, Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMeTTyB), Universidad Favaloro-CONICET, Buenos Aires, Argentina
| | - LEONARDO FELDMAN
- Facultad de Ciencias de la Salud, Universidad Nacional del Centro de la Provincia de Buenos Aires (UNCPB), Tandil, Buenos Aires, Argentina
| | - FEDERICO DIMASE
- Servicio de Hematología, Hospital Militar Central, Buenos Aires, Argentina
| | - EMILIO BATAGELJ
- Servicio de Oncología, Hospital Militar Central, Buenos Aires, Argentina
| | - GUSTAVO YANNARELLI
- Laboratorio de Regulación Génica y Células Madre, Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMeTTyB), Universidad Favaloro-CONICET, Buenos Aires, Argentina
| | - NORMA ALEJANDRA CHASSEING
- Laboratorio de Inmunohematología, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
16
|
Sharma K, Sarkar J, Trisal A, Ghosh R, Dixit A, Singh AK. Targeting mitochondrial dysfunction to salvage cellular senescence for managing neurodegeneration. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 136:309-337. [PMID: 37437982 DOI: 10.1016/bs.apcsb.2023.02.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
Aging is an inevitable phenomenon that causes a decline in bodily functions over time. One of the most important processes that play a role in aging is senescence. Senescence is characterized by accumulation of cells that are no longer functional but elude the apoptotic pathway. These cells secrete inflammatory molecules that comprise the senescence associated secretory phenotype (SASP). Several essential molecules such as p53, Rb, and p16INK4a regulate the senescence process. Mitochondrial regulation has been found to play an important role in senescence. Reactive oxygen species (ROS) generated from mitochondria can affect cellular senescence by inducing the persistent DNA damage response, thus stabilizing the senescence. Evidently, senescence plays a major contributory role to the development of age-related neurological disorders. In this chapter, we discuss the role of senescence in the progression and onset of several neurodegenerative diseases including Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis. Moreover, we also discuss the efficacy of certain molecules like MitoQ, SkQ1, and Latrepirdine that could be proven therapeutics with respect to these disorders by regulating mitochondrial activity.
Collapse
Affiliation(s)
- Komal Sharma
- Amity Institute of Neuropsychology and Neurosciences, Amity University Uttar Pradesh, Noida, India
| | - Joyobrata Sarkar
- Amity Institute of Neuropsychology and Neurosciences, Amity University Uttar Pradesh, Noida, India
| | - Anchal Trisal
- Amity Institute of Neuropsychology and Neurosciences, Amity University Uttar Pradesh, Noida, India
| | - Rishika Ghosh
- Amity Institute of Neuropsychology and Neurosciences, Amity University Uttar Pradesh, Noida, India
| | - Anubhuti Dixit
- Amity Institute of Neuropsychology and Neurosciences, Amity University Uttar Pradesh, Noida, India.
| | - Abhishek Kumar Singh
- Amity Institute of Neuropsychology and Neurosciences, Amity University Uttar Pradesh, Noida, India.
| |
Collapse
|
17
|
Storchova R, Palek M, Palkova N, Veverka P, Brom T, Hofr C, Macurek L. Phosphorylation of TRF2 promotes its interaction with TIN2 and regulates DNA damage response at telomeres. Nucleic Acids Res 2023; 51:1154-1172. [PMID: 36651296 PMCID: PMC9943673 DOI: 10.1093/nar/gkac1269] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 11/25/2022] [Accepted: 12/23/2022] [Indexed: 01/19/2023] Open
Abstract
Protein phosphatase magnesium-dependent 1 delta (PPM1D) terminates the cell cycle checkpoint by dephosphorylating the tumour suppressor protein p53. By targeting additional substrates at chromatin, PPM1D contributes to the control of DNA damage response and DNA repair. Using proximity biotinylation followed by proteomic analysis, we identified a novel interaction between PPM1D and the shelterin complex that protects telomeric DNA. In addition, confocal microscopy revealed that endogenous PPM1D localises at telomeres. Further, we found that ATR phosphorylated TRF2 at S410 after induction of DNA double strand breaks at telomeres and this modification increased after inhibition or loss of PPM1D. TRF2 phosphorylation stimulated its interaction with TIN2 both in vitro and at telomeres. Conversely, induced expression of PPM1D impaired localisation of TIN2 and TPP1 at telomeres. Finally, recruitment of the DNA repair factor 53BP1 to the telomeric breaks was strongly reduced after inhibition of PPM1D and was rescued by the expression of TRF2-S410A mutant. Our results suggest that TRF2 phosphorylation promotes the association of TIN2 within the shelterin complex and regulates DNA repair at telomeres.
Collapse
Affiliation(s)
- Radka Storchova
- Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague CZ-14220, Czech Republic
| | - Matous Palek
- Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague CZ-14220, Czech Republic
| | - Natalie Palkova
- Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague CZ-14220, Czech Republic
| | - Pavel Veverka
- LifeB, Functional Genomics and Proteomics, National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Brno CZ-62500, Czech Republic
| | - Tomas Brom
- LifeB, Functional Genomics and Proteomics, National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Brno CZ-62500, Czech Republic
| | - Ctirad Hofr
- LifeB, Functional Genomics and Proteomics, National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Brno CZ-62500, Czech Republic
| | - Libor Macurek
- Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague CZ-14220, Czech Republic
| |
Collapse
|
18
|
Yang Q, Nie Z, Zhu Y, Hao M, Liu S, Ding X, Wang F, Wang F, Geng X. Inhibition of TRF2 Leads to Ferroptosis, Autophagic Death, and Apoptosis by Causing Telomere Dysfunction. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:6897268. [PMID: 37113742 PMCID: PMC10129434 DOI: 10.1155/2023/6897268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 10/23/2022] [Accepted: 02/04/2023] [Indexed: 04/29/2023]
Abstract
Background Gastric cancer (GC) is an aggressive malignancy with a high mortality rate and poor prognosis. Telomeric repeat-binding factor 2 (TRF2) is a critical telomere protection protein. Emerging evidence indicates that TRF2 may be an essential treatment option for GC; however, the exact mechanism remains largely unknown. Objective We aimed to explore the role of TRF2 in GC cells. The function and molecular mechanisms of TRF2 in the pathogenesis of GC were mainly discussed in this study. Methods Relevant data from GEPIA and TCGA databases regarding TRF2 gene expression and its prognostic significance in GC samples were analyzed. Analysis of 53BP1 foci at telomeres by immunofluorescence, metaphase spreads, and telomere-specific FISH analysis was carried out to explore telomere damage and dysfunction after TRF2 depletion. CCK8 cell proliferation, trypan blue staining, and colony formation assay were performed to evaluate cell survival. Apoptosis and cell migration were determined with flow cytometry and scratch-wound healing assay, respectively. qRT-PCR and Western blotting were carried out to analyze the mRNA and protein expression levels after TRF2 depletion on apoptosis, autophagic death, and ferroptosis. Results By searching with GEPIA and TCGA databases, the results showed that the expression levels of TRF2 were obviously elevated in the samples of GC patients, which was associated with adverse prognosis. Knockdown of TRF2 suppressed the cell growth, proliferation, and migration in GC cells, causing significant telomere dysfunction. Apoptosis, autophagic death, and ferroptosis were also triggered in this process. The pretreatment of chloroquine (autophagy inhibitor) and ferrostatin-1 (ferroptosis inhibitor) improved the survival phenotypes of GC cells. Conclusion Our data suggest that TRF2 depletion can inhibit cell growth, proliferation, and migration through the combined action of ferroptosis, autophagic death, and apoptosis in GC cells. The results indicate that TRF2 might be used as a potential target to develop therapeutic strategies for treating GC.
Collapse
Affiliation(s)
- Qiuhui Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Medical University, Tianjin 300070, China
| | - Ziyang Nie
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Medical University, Tianjin 300070, China
- School of Life Sciences, Central China Normal University, Hubei Province, China
| | - Yukun Zhu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Medical University, Tianjin 300070, China
- Fuyang Hospital Affiliated to Anhui Medical University, Anhui Province 236000, China
| | - Mingying Hao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Medical University, Tianjin 300070, China
| | - Siqi Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Medical University, Tianjin 300070, China
| | - Xuelu Ding
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Medical University, Tianjin 300070, China
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University, General Hospital, Tianjin 300052, China
| | - Feng Wang
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Fei Wang
- Department of Neurology, General Hospital, Tianjin Medical University, Tianjin 300052, China
| | - Xin Geng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Medical University, Tianjin 300070, China
| |
Collapse
|
19
|
Muoio D, Laspata N, Fouquerel E. Functions of ADP-ribose transferases in the maintenance of telomere integrity. Cell Mol Life Sci 2022; 79:215. [PMID: 35348914 PMCID: PMC8964661 DOI: 10.1007/s00018-022-04235-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/03/2022] [Accepted: 03/07/2022] [Indexed: 12/21/2022]
Abstract
The ADP-ribose transferase (ART) family comprises 17 enzymes that catalyze mono- or poly-ADP-ribosylation, a post-translational modification of proteins. Present in all subcellular compartments, ARTs are implicated in a growing number of biological processes including DNA repair, replication, transcription regulation, intra- and extra-cellular signaling, viral infection and cell death. Five members of the family, PARP1, PARP2, PARP3, tankyrase 1 and tankyrase 2 are mainly described for their crucial functions in the maintenance of genome stability. It is well established that the most describedrole of PARP1, 2 and 3 is the repair of DNA lesions while tankyrases 1 and 2 are crucial for maintaining the integrity of telomeres. Telomeres, nucleoprotein complexes located at the ends of eukaryotic chromosomes, utilize their unique structure and associated set of proteins to orchestrate the mechanisms necessary for their own protection and replication. While the functions of tankyrases 1 and 2 at telomeres are well known, several studies have also brought PARP1, 2 and 3 to the forefront of telomere protection. The singular quality of the telomeric environment has highlighted protein interactions and molecular pathways distinct from those described throughout the genome. The aim of this review is to provide an overview of the current knowledge on the multiple roles of PARP1, PARP2, PARP3, tankyrase 1 and tankyrase 2 in the maintenance and preservation of telomere integrity.
Collapse
Affiliation(s)
- Daniela Muoio
- UPMC Cancer Institute and Department of Pharmacology and Chemical Biology at the University of Pittsburgh, Hillman Cancer Center, 5115 Centre Avenue, Pittsburgh, PA, 15213, USA
| | - Natalie Laspata
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, 233 S. 10th street, Philadelphia, PA, 19107, USA
| | - Elise Fouquerel
- UPMC Cancer Institute and Department of Pharmacology and Chemical Biology at the University of Pittsburgh, Hillman Cancer Center, 5115 Centre Avenue, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
20
|
Ghadaouia S, Olivier MA, Martinez A, Kientega T, Qin J, Lambert-Lanteigne P, Cardin GB, Autexier C, Malaquin N, Rodier F. Homologous recombination-mediated irreversible genome damage underlies telomere-induced senescence. Nucleic Acids Res 2021; 49:11690-11707. [PMID: 34725692 PMCID: PMC8599762 DOI: 10.1093/nar/gkab965] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/28/2021] [Accepted: 10/12/2021] [Indexed: 12/12/2022] Open
Abstract
Loss of telomeric DNA leads to telomere uncapping, which triggers a persistent, p53-centric DNA damage response that sustains a stable senescence-associated proliferation arrest. Here, we show that in normal cells telomere uncapping triggers a focal telomeric DNA damage response accompanied by a transient cell cycle arrest. Subsequent cell division with dysfunctional telomeres resulted in sporadic telomeric sister chromatid fusions that gave rise to next-mitosis genome instability, including non-telomeric DNA lesions responsible for a stable, p53-mediated, senescence-associated proliferation arrest. Unexpectedly, the blocking of Rad51/RPA-mediated homologous recombination, but not non-homologous end joining (NHEJ), prevented senescence despite multiple dysfunctional telomeres. When cells approached natural replicative senescence, interphase senescent cells displayed genome instability, whereas near-senescent cells that underwent mitosis despite the presence of uncapped telomeres did not. This suggests that these near-senescent cells had not yet acquired irreversible telomeric fusions. We propose a new model for telomere-initiated senescence where tolerance of telomere uncapping eventually results in irreversible non-telomeric DNA lesions leading to stable senescence. Paradoxically, our work reveals that senescence-associated tumor suppression from telomere shortening requires irreversible genome instability at the single-cell level, which suggests that interventions to repair telomeres in the pre-senescent state could prevent senescence and genome instability.
Collapse
Affiliation(s)
- Sabrina Ghadaouia
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, H2X 0A9, Canada.,Institut du cancer de Montréal, Montreal, QC, H2X 0A9, Canada
| | - Marc-Alexandre Olivier
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, H2X 0A9, Canada.,Institut du cancer de Montréal, Montreal, QC, H2X 0A9, Canada
| | - Aurélie Martinez
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, H2X 0A9, Canada.,Institut du cancer de Montréal, Montreal, QC, H2X 0A9, Canada
| | - Tibila Kientega
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, H2X 0A9, Canada.,Institut du cancer de Montréal, Montreal, QC, H2X 0A9, Canada
| | - Jian Qin
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC, H3A 0C7, Canada.,Jewish General Hospital, Lady Davis Institute, Montreal, QC, H3T 1E2, Canada
| | | | - Guillaume B Cardin
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, H2X 0A9, Canada.,Institut du cancer de Montréal, Montreal, QC, H2X 0A9, Canada
| | - Chantal Autexier
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC, H3A 0C7, Canada.,Jewish General Hospital, Lady Davis Institute, Montreal, QC, H3T 1E2, Canada
| | - Nicolas Malaquin
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, H2X 0A9, Canada.,Institut du cancer de Montréal, Montreal, QC, H2X 0A9, Canada
| | - Francis Rodier
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, H2X 0A9, Canada.,Institut du cancer de Montréal, Montreal, QC, H2X 0A9, Canada.,Department of Radiology, Radio-Oncology and Nuclear Medicine, Université de Montréal, Montreal, QC, H3T 1J4, Canada
| |
Collapse
|
21
|
Mojiri A, Walther BK, Jiang C, Matrone G, Holgate R, Xu Q, Morales E, Wang G, Gu J, Wang R, Cooke JP. Telomerase therapy reverses vascular senescence and extends lifespan in progeria mice. Eur Heart J 2021; 42:4352-4369. [PMID: 34389865 PMCID: PMC8603239 DOI: 10.1093/eurheartj/ehab547] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 03/29/2021] [Accepted: 08/12/2021] [Indexed: 12/28/2022] Open
Abstract
AIMS Hutchinson-Gilford progeria syndrome (HGPS) is an accelerated ageing syndrome associated with premature vascular disease and death due to heart attack and stroke. In HGPS a mutation in lamin A (progerin) alters nuclear morphology and gene expression. Current therapy increases the lifespan of these children only modestly. Thus, greater understanding of the underlying mechanisms of HGPS is required to improve therapy. Endothelial cells (ECs) differentiated from induced pluripotent stem cells (iPSCs) derived from these patients exhibit hallmarks of senescence including replication arrest, increased expression of inflammatory markers, DNA damage, and telomere erosion. We hypothesized that correction of shortened telomeres may reverse these measures of vascular ageing. METHODS AND RESULTS We generated ECs from iPSCs belonging to children with HGPS and their unaffected parents. Telomerase mRNA (hTERT) was used to treat HGPS ECs. Endothelial morphology and functions were assessed, as well as proteomic and transcriptional profiles with attention to inflammatory markers, DNA damage, and EC identity genes. In a mouse model of HGPS, we assessed the effects of lentiviral transfection of mTERT on measures of senescence, focusing on the EC phenotype in various organs. hTERT treatment of human HGPS ECs improved replicative capacity; restored endothelial functions such as nitric oxide generation, acetylated low-density lipoprotein uptake and angiogenesis; and reduced the elaboration of inflammatory cytokines. In addition, hTERT treatment improved cellular and nuclear morphology, in association with a normalization of the transcriptional profile, effects that may be mediated in part by a reduction in progerin expression and an increase in sirtuin 1 (SIRT1). Progeria mice treated with mTERT lentivirus manifested similar improvements, with a reduction in inflammatory and DNA damage markers and increased SIRT1 in their vasculature and other organs. Furthermore, mTERT therapy increased the lifespan of HGPS mice. CONCLUSION Vascular rejuvenation using telomerase mRNA is a promising approach for progeria and other age-related diseases.
Collapse
Affiliation(s)
- Anahita Mojiri
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, 6670 Bertner Ave., R10-South, Houston, TX 77030, USA
| | - Brandon K Walther
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, 6670 Bertner Ave., R10-South, Houston, TX 77030, USA
- Department of Biomedical Engineering, Texas A&M University, 101 Bizzell St., College Station, TX 77840, USA
| | - Chongming Jiang
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Gianfranco Matrone
- British Heart Foundation Centre for Cardiovascular Science, Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Rhonda Holgate
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, 6670 Bertner Ave., R10-South, Houston, TX 77030, USA
| | - Qiu Xu
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, 6670 Bertner Ave., R10-South, Houston, TX 77030, USA
| | - Elisa Morales
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, 6670 Bertner Ave., R10-South, Houston, TX 77030, USA
| | - Guangyu Wang
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, 6670 Bertner Ave., R10-South, Houston, TX 77030, USA
- Center for Bioinformatics and Computational Biology, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Jianhua Gu
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, 6670 Bertner Ave., R10-South, Houston, TX 77030, USA
| | - Rongfu Wang
- Department of Medicine, and Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - John P Cooke
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, 6670 Bertner Ave., R10-South, Houston, TX 77030, USA
| |
Collapse
|
22
|
Pennarun G, Picotto J, Etourneaud L, Redavid AR, Certain A, Gauthier LR, Fontanilla-Ramirez P, Busso D, Chabance-Okumura C, Thézé B, Boussin FD, Bertrand P. Increase in lamin B1 promotes telomere instability by disrupting the shelterin complex in human cells. Nucleic Acids Res 2021; 49:9886-9905. [PMID: 34469544 PMCID: PMC8464066 DOI: 10.1093/nar/gkab761] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 08/04/2021] [Accepted: 08/31/2021] [Indexed: 12/19/2022] Open
Abstract
Telomere maintenance is essential to preserve genomic stability and involves telomere-specific proteins, DNA replication and repair proteins. Lamins are key components of the nuclear envelope and play numerous roles, including maintenance of the nuclear integrity, regulation of transcription, and DNA replication. Elevated levels of lamin B1, one of the major lamins, have been observed in some human pathologies and several cancers. Yet, the effect of lamin B1 dysregulation on telomere maintenance remains unknown. Here, we unveil that lamin B1 overexpression drives telomere instability through the disruption of the shelterin complex. Indeed, lamin B1 dysregulation leads to an increase in telomere dysfunction-induced foci, telomeric fusions and telomere losses in human cells. Telomere aberrations were preceded by mislocalizations of TRF2 and its binding partner RAP1. Interestingly, we identified new interactions between lamin B1 and these shelterin proteins, which are strongly enhanced at the nuclear periphery upon lamin B1 overexpression. Importantly, chromosomal fusions induced by lamin B1 in excess were rescued by TRF2 overexpression. These data indicated that lamin B1 overexpression triggers telomere instability through a mislocalization of TRF2. Altogether our results point to lamin B1 as a new interacting partner of TRF2, that is involved in telomere stability.
Collapse
Affiliation(s)
- Gaëlle Pennarun
- Université de Paris and Université Paris-Saclay, INSERM, iRCM/IBFJ CEA, UMR Stabilité Génétique Cellules Souches et Radiations, F-92265 Fontenay-aux-Roses, France
- “DNA Repair and Ageing” Team, iRCM/IBFJ, DRF, CEA, Fontenay-aux-Roses, France
| | - Julien Picotto
- Université de Paris and Université Paris-Saclay, INSERM, iRCM/IBFJ CEA, UMR Stabilité Génétique Cellules Souches et Radiations, F-92265 Fontenay-aux-Roses, France
- “DNA Repair and Ageing” Team, iRCM/IBFJ, DRF, CEA, Fontenay-aux-Roses, France
| | - Laure Etourneaud
- Université de Paris and Université Paris-Saclay, INSERM, iRCM/IBFJ CEA, UMR Stabilité Génétique Cellules Souches et Radiations, F-92265 Fontenay-aux-Roses, France
- “DNA Repair and Ageing” Team, iRCM/IBFJ, DRF, CEA, Fontenay-aux-Roses, France
| | - Anna-Rita Redavid
- Université de Paris and Université Paris-Saclay, INSERM, iRCM/IBFJ CEA, UMR Stabilité Génétique Cellules Souches et Radiations, F-92265 Fontenay-aux-Roses, France
- “DNA Repair and Ageing” Team, iRCM/IBFJ, DRF, CEA, Fontenay-aux-Roses, France
| | - Anaïs Certain
- Université de Paris and Université Paris-Saclay, INSERM, iRCM/IBFJ CEA, UMR Stabilité Génétique Cellules Souches et Radiations, F-92265 Fontenay-aux-Roses, France
- “DNA Repair and Ageing” Team, iRCM/IBFJ, DRF, CEA, Fontenay-aux-Roses, France
| | - Laurent R Gauthier
- Université de Paris and Université Paris-Saclay, INSERM, iRCM/IBFJ CEA, UMR Stabilité Génétique Cellules Souches et Radiations, F-92265 Fontenay-aux-Roses, France
- “Radiopathology” Team, iRCM/IBFJ, DRF, CEA, Fontenay-aux-Roses, France
| | - Paula Fontanilla-Ramirez
- Université de Paris and Université Paris-Saclay, INSERM, iRCM/IBFJ CEA, UMR Stabilité Génétique Cellules Souches et Radiations, F-92265 Fontenay-aux-Roses, France
- “DNA Repair and Ageing” Team, iRCM/IBFJ, DRF, CEA, Fontenay-aux-Roses, France
| | - Didier Busso
- Université de Paris and Université Paris-Saclay, INSERM, iRCM/IBFJ CEA, UMR Stabilité Génétique Cellules Souches et Radiations, F-92265 Fontenay-aux-Roses, France
- Genetic Engineering and Expression Platform (CIGEX), iRCM, DRF, CEA, Fontenay-aux-Roses, France
| | - Caroline Chabance-Okumura
- Université de Paris and Université Paris-Saclay, INSERM, iRCM/IBFJ CEA, UMR Stabilité Génétique Cellules Souches et Radiations, F-92265 Fontenay-aux-Roses, France
- “DNA Repair and Ageing” Team, iRCM/IBFJ, DRF, CEA, Fontenay-aux-Roses, France
| | - Benoît Thézé
- Université de Paris and Université Paris-Saclay, INSERM, iRCM/IBFJ CEA, UMR Stabilité Génétique Cellules Souches et Radiations, F-92265 Fontenay-aux-Roses, France
- “DNA Repair and Ageing” Team, iRCM/IBFJ, DRF, CEA, Fontenay-aux-Roses, France
| | - François D Boussin
- Université de Paris and Université Paris-Saclay, INSERM, iRCM/IBFJ CEA, UMR Stabilité Génétique Cellules Souches et Radiations, F-92265 Fontenay-aux-Roses, France
- “Radiopathology” Team, iRCM/IBFJ, DRF, CEA, Fontenay-aux-Roses, France
| | - Pascale Bertrand
- Université de Paris and Université Paris-Saclay, INSERM, iRCM/IBFJ CEA, UMR Stabilité Génétique Cellules Souches et Radiations, F-92265 Fontenay-aux-Roses, France
- “DNA Repair and Ageing” Team, iRCM/IBFJ, DRF, CEA, Fontenay-aux-Roses, France
| |
Collapse
|
23
|
H2B Type 1-K Accumulates in Senescent Fibroblasts with Persistent DNA Damage along with Methylated and Phosphorylated Forms of HMGA1. Proteomes 2021; 9:proteomes9020030. [PMID: 34205514 PMCID: PMC8293446 DOI: 10.3390/proteomes9020030] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/11/2021] [Accepted: 06/17/2021] [Indexed: 11/27/2022] Open
Abstract
Cellular senescence is a state of terminal proliferative arrest that plays key roles in aging by preventing stem cell renewal and by inducing the expression of a series of inflammatory factors including many secreted proteins with paracrine effects. The in vivo identification of senescent cells is difficult due to the absence of universal biomarkers. Chromatin modifications are key aspects of the senescence transition and may provide novel biomarkers. We used a combined protein profiling and bottom-up mass spectrometry approach to characterize the isoforms and post-translational modifications of chromatin proteins over time in post-mitotic human fibroblasts in vitro. We show that the H2B type 1-K variant is specifically enriched in deep senescent cells with persistent DNA damage. This accumulation was not observed in quiescent cells or in cells induced into senescence without DNA damage by expression of the RAF kinase. Similarly, HMGA1a di-methylated and HMGA1b tri-phosphorylated forms accumulated exclusively in the chromatin of cells in deep senescent conditions with persistent DNA damage. H2B type 1-K and modified HMGA1 may thus represent novel biomarkers of senescent cells containing persistent DNA damage.
Collapse
|
24
|
Vessoni AT, Zhang T, Quinet A, Jeong HC, Munroe M, Wood M, Tedone E, Vindigni A, Shay JW, Greenberg RA, Batista LF. Telomere erosion in human pluripotent stem cells leads to ATR-mediated mitotic catastrophe. J Cell Biol 2021; 220:211982. [PMID: 33851958 PMCID: PMC8050844 DOI: 10.1083/jcb.202011014] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 03/03/2021] [Accepted: 03/15/2021] [Indexed: 12/14/2022] Open
Abstract
It is well established that short telomeres activate an ATM-driven DNA damage response that leads to senescence in terminally differentiated cells. However, technical limitations have hampered our understanding of how telomere shortening is signaled in human stem cells. Here, we show that telomere attrition induces ssDNA accumulation (G-strand) at telomeres in human pluripotent stem cells (hPSCs), but not in their differentiated progeny. This led to a unique role for ATR in the response of hPSCs to telomere shortening that culminated in an extended S/G2 cell cycle phase and a longer period of mitosis, which was associated with aneuploidy and mitotic catastrophe. Loss of p53 increased resistance to death, at the expense of increased mitotic abnormalities in hPSCs. Taken together, our data reveal an unexpected dominant role of ATR in hPSCs, combined with unique cell cycle abnormalities and, ultimately, consequences distinct from those observed in their isogenic differentiated counterparts.
Collapse
Affiliation(s)
| | - Tianpeng Zhang
- Department of Cancer Biology, Penn Center for Genome Integrity, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Annabel Quinet
- Department of Medicine, Washington University in St. Louis, St. Louis, MO
| | - Ho-Chang Jeong
- Department of Medicine, Washington University in St. Louis, St. Louis, MO
| | - Michael Munroe
- Department of Medicine, Washington University in St. Louis, St. Louis, MO
| | - Matthew Wood
- Department of Medicine, Washington University in St. Louis, St. Louis, MO
| | - Enzo Tedone
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX
| | | | - Jerry W. Shay
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX
| | - Roger A. Greenberg
- Department of Cancer Biology, Penn Center for Genome Integrity, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Luis F.Z. Batista
- Department of Medicine, Washington University in St. Louis, St. Louis, MO
- Center of Regenerative Medicine, Washington University in St. Louis, St. Louis, MO
- Correspondence to Luis F.Z. Batista:
| |
Collapse
|
25
|
van Batenburg AA, Kazemier KM, van Oosterhout MFM, van der Vis JJ, Grutters JC, Goldschmeding R, van Moorsel CHM. Telomere shortening and DNA damage in culprit cells of different types of progressive fibrosing interstitial lung disease. ERJ Open Res 2021; 7:00691-2020. [PMID: 34084786 PMCID: PMC8165375 DOI: 10.1183/23120541.00691-2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 12/14/2020] [Indexed: 12/11/2022] Open
Abstract
Pulmonary fibrosis is strongly associated with telomere shortening and increased DNA damage. Key cells in the pathogenesis involve alveolar type 2 (AT2) cells, club cells and myofibroblasts; however, to what extent these cells are affected by telomere shortening and DNA damage is not yet known. We sought to determine the degree of, and correlation between, telomere shortening and DNA damage in different cell types involved in the pathogenesis of progressive fibrosing interstitial lung disease. Telomere length and DNA damage were quantified, using combined fluorescence in situ hybridisation and immunofluorescence staining techniques, in AT2 cells, club cells and myofibroblasts of controls and patients with pulmonary fibrosis and a telomerase reverse transcriptase mutation (TERT-PF), idiopathic pulmonary fibrosis (IPF) and fibrotic hypersensitivity pneumonitis (fHP). In IPF and TERT-PF lungs, AT2 cells contained shorter telomeres and expressed higher DNA damage signals than club cells and myofibroblasts. In fHP lungs, club cells contained highly elevated levels of DNA damage, while telomeres were not obviously short. In vitro, we found significantly shorter telomeres and higher DNA damage levels only in AT2 surrogate cell lines treated with telomerase inhibitor BIBR1532. Our study demonstrated that in IPF and TERT-PF lungs, telomere shortening and accumulation of DNA damage primarily affects AT2 cells, further supporting the importance of AT2 cells in these diseases, while in fHP the particularly high telomere-independent DNA damage signals in club cells underscores its bronchiolocentric pathogenesis. These findings suggest that cell type-specific telomere shortening and DNA damage may help to discriminate between different drivers of fibrogenesis.
Collapse
Affiliation(s)
- Aernoud A van Batenburg
- Dept of Pulmonology, St Antonius ILD Center of Excellence, St Antonius Hospital, Nieuwegein, The Netherlands
| | - Karin M Kazemier
- Center of Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands.,Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Matthijs F M van Oosterhout
- Dept of Pathology, Pathology DNA, St Antonius ILD Center of Excellence, St Antonius Hospital, Nieuwegein, The Netherlands
| | - Joanne J van der Vis
- Dept of Pulmonology, St Antonius ILD Center of Excellence, St Antonius Hospital, Nieuwegein, The Netherlands.,Dept of Clinical Chemistry, St Antonius ILD Center of Excellence, St Antonius Hospital, Nieuwegein, The Netherlands
| | - Jan C Grutters
- Dept of Pulmonology, St Antonius ILD Center of Excellence, St Antonius Hospital, Nieuwegein, The Netherlands.,Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Roel Goldschmeding
- Dept of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Coline H M van Moorsel
- Dept of Pulmonology, St Antonius ILD Center of Excellence, St Antonius Hospital, Nieuwegein, The Netherlands.,Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
26
|
Abdelgawad IY, Sadak KT, Lone DW, Dabour MS, Niedernhofer LJ, Zordoky BN. Molecular mechanisms and cardiovascular implications of cancer therapy-induced senescence. Pharmacol Ther 2021; 221:107751. [PMID: 33275998 PMCID: PMC8084867 DOI: 10.1016/j.pharmthera.2020.107751] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 11/16/2020] [Accepted: 11/23/2020] [Indexed: 12/11/2022]
Abstract
Cancer treatment has been associated with accelerated aging that can lead to early-onset health complications typically experienced by older populations. In particular, cancer survivors have an increased risk of developing premature cardiovascular complications. In the last two decades, cellular senescence has been proposed as an important mechanism of premature cardiovascular diseases. Cancer treatments, specifically anthracyclines and radiation, have been shown to induce senescence in different types of cardiovascular cells. Additionally, clinical studies identified increased systemic markers of senescence in cancer survivors. Preclinical research has demonstrated the potential of several approaches to mitigate cancer therapy-induced senescence. However, strategies to prevent and/or treat therapy-induced cardiovascular senescence have not yet been translated to the clinic. In this review, we will discuss how therapy-induced senescence can contribute to cardiovascular complications. Thereafter, we will summarize the current in vitro, in vivo, and clinical evidence regarding cancer therapy-induced cardiovascular senescence. Then, we will discuss interventional strategies that have the potential to protect against therapy-induced cardiovascular senescence. To conclude, we will highlight challenges and future research directions to mitigate therapy-induced cardiovascular senescence in cancer survivors.
Collapse
Affiliation(s)
- Ibrahim Y Abdelgawad
- Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, MN 55455, USA
| | - Karim T Sadak
- Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN 55455, USA; University of Minnesota Masonic Children's Hospital, Minneapolis, MN 55455, USA; University of Minnesota Masonic Cancer Center, Minneapolis, MN 55455, USA
| | - Diana W Lone
- University of Minnesota Masonic Children's Hospital, Minneapolis, MN 55455, USA
| | - Mohamed S Dabour
- Clinical Pharmacy Department, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt
| | - Laura J Niedernhofer
- Institute on the Biology of Aging and Metabolism and Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Beshay N Zordoky
- Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, MN 55455, USA.
| |
Collapse
|
27
|
Willaume S, Rass E, Fontanilla-Ramirez P, Moussa A, Wanschoor P, Bertrand P. A Link between Replicative Stress, Lamin Proteins, and Inflammation. Genes (Basel) 2021; 12:genes12040552. [PMID: 33918867 PMCID: PMC8070205 DOI: 10.3390/genes12040552] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/23/2021] [Accepted: 04/08/2021] [Indexed: 12/12/2022] Open
Abstract
Double-stranded breaks (DSB), the most toxic DNA lesions, are either a consequence of cellular metabolism, programmed as in during V(D)J recombination, or induced by anti-tumoral therapies or accidental genotoxic exposure. One origin of DSB sources is replicative stress, a major source of genome instability, especially when the integrity of the replication forks is not properly guaranteed. To complete stalled replication, restarting the fork requires complex molecular mechanisms, such as protection, remodeling, and processing. Recently, a link has been made between DNA damage accumulation and inflammation. Indeed, defects in DNA repair or in replication can lead to the release of DNA fragments in the cytosol. The recognition of this self-DNA by DNA sensors leads to the production of inflammatory factors. This beneficial response activating an innate immune response and destruction of cells bearing DNA damage may be considered as a novel part of DNA damage response. However, upon accumulation of DNA damage, a chronic inflammatory cellular microenvironment may lead to inflammatory pathologies, aging, and progression of tumor cells. Progress in understanding the molecular mechanisms of DNA damage repair, replication stress, and cytosolic DNA production would allow to propose new therapeutical strategies against cancer or inflammatory diseases associated with aging. In this review, we describe the mechanisms involved in DSB repair, the replicative stress management, and its consequences. We also focus on new emerging links between key components of the nuclear envelope, the lamins, and DNA repair, management of replicative stress, and inflammation.
Collapse
|
28
|
Kim S, Kim C. Transcriptomic Analysis of Cellular Senescence: One Step Closer to Senescence Atlas. Mol Cells 2021; 44:136-145. [PMID: 33795532 PMCID: PMC8019598 DOI: 10.14348/molcells.2021.2239] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 03/05/2021] [Accepted: 03/12/2021] [Indexed: 12/19/2022] Open
Abstract
Senescent cells that gradually accumulate during aging are one of the leading causes of aging. While senolytics can improve aging in humans as well as mice by specifically eliminating senescent cells, the effect of the senolytics varies in different cell types, suggesting variations in senescence. Various factors can induce cellular senescence, and the rate of accumulation of senescent cells differ depending on the organ. In addition, since the heterogeneity is due to the spatiotemporal context of senescent cells, in vivo studies are needed to increase the understanding of senescent cells. Since current methods are often unable to distinguish senescent cells from other cells, efforts are being made to find markers commonly expressed in senescent cells using bulk RNA-sequencing. Moreover, single-cell RNA (scRNA) sequencing, which analyzes the transcripts of each cell, has been utilized to understand the in vivo characteristics of the rare senescent cells. Recently, transcriptomic cell atlases for each organ using this technology have been published in various species. Novel senescent cells that do not express previously established marker genes have been discovered in some organs. However, there is still insufficient information on senescent cells due to the limited throughput of the scRNA sequencing technology. Therefore, it is necessary to improve the throughput of the scRNA sequencing technology or develop a way to enrich the rare senescent cells. The in vivo senescent cell atlas that is established using rapidly developing single-cell technologies will contribute to the precise rejuvenation by specifically removing senescent cells in each tissue and individual.
Collapse
Affiliation(s)
- Sohee Kim
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea
- Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon 34113, Korea
| | - Chuna Kim
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea
| |
Collapse
|
29
|
Abstract
Decades of study on cell cycle regulation have provided great insight into human cellular life span barriers, as well as their dysregulation during tumorigenesis. Telomeres, the extremities of linear chromosomes, perform an essential role in implementing these proliferative boundaries and preventing the propagation of potentially cancerous cells. The tumor-suppressive function of telomeres relies on their ability to initiate DNA damage signaling pathways and downstream cellular events, ranging from cell cycle perturbation to inflammation and cell death. While the tumor-suppressor role of telomeres is undoubtable, recent advances have pointed to telomeres as a major source of many of the genomic aberrations found in both early- and late-stage cancers, including the most recently discovered mutational phenomenon of chromothripsis. Telomere shortening appears as a double-edged sword that can function in opposing directions in carcinogenesis. This review focuses on the current knowledge of the dual role of telomeres in cancer and suggests a new perspective to reconcile the paradox of telomeres and their implications in cancer etiology.
Collapse
Affiliation(s)
- Joe Nassour
- Molecular and Cell Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, California 92037, USA
| | - Tobias T Schmidt
- Molecular and Cell Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, California 92037, USA
| | - Jan Karlseder
- Molecular and Cell Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, California 92037, USA
| |
Collapse
|
30
|
Swift ML, Beishline K, Flashner S, Azizkhan-Clifford J. DSB repair pathway choice is regulated by recruitment of 53BP1 through cell cycle-dependent regulation of Sp1. Cell Rep 2021; 34:108840. [PMID: 33730584 DOI: 10.1016/j.celrep.2021.108840] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 09/13/2020] [Accepted: 02/17/2021] [Indexed: 12/14/2022] Open
Abstract
Although many of the factors, epigenetic changes, and cell cycle stages that distinguish repair of double-strand breaks (DSBs) by homologous recombination (HR) from non-homologous end joining (NHEJ) are known, the underlying mechanisms that determine pathway choice are incompletely understood. Previously, we found that the transcription factor Sp1 is recruited to DSBs and is necessary for repair. Here, we demonstrate that Sp1 localizes to DSBs in G1 and is necessary for recruitment of the NHEJ repair factor, 53BP1. Phosphorylation of Sp1-S59 in early S phase evicts Sp1 and 53BP1 from the break site; inhibition of that phosphorylation results in 53BP1 and Sp1 remaining at DSBs in S phase cells, precluding BRCA1 binding and suppressing HR. Expression of Sp1-S59A increases sensitivity of BRCA1+/+ cells to poly (ADP-ribose) polymerase (PARP) inhibition similar to BRCA1 deficiency. These data demonstrate how Sp1 integrates the cell cycle and DSB repair pathway choice to favor NHEJ.
Collapse
Affiliation(s)
- Michelle L Swift
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Kate Beishline
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Samuel Flashner
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Jane Azizkhan-Clifford
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
31
|
Sullivan DI, Jiang M, Hinchie AM, Roth MG, Bahudhanapati H, Nouraie M, Liu J, McDyer JF, Mallampalli RK, Zhang Y, Kass DJ, Finkel T, Alder JK. Transcriptional and Proteomic Characterization of Telomere-Induced Senescence in a Human Alveolar Epithelial Cell Line. Front Med (Lausanne) 2021; 8:600626. [PMID: 33634147 PMCID: PMC7902064 DOI: 10.3389/fmed.2021.600626] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 01/05/2021] [Indexed: 01/16/2023] Open
Abstract
Cellular senescence due to telomere dysfunction has been hypothesized to play a role in age-associated diseases including idiopathic pulmonary fibrosis (IPF). It has been postulated that paracrine mediators originating from senescent alveolar epithelia signal to surrounding mesenchymal cells and contribute to disease pathogenesis. However, murine models of telomere-induced alveolar epithelial senescence fail to display the canonical senescence-associated secretory phenotype (SASP) that is observed in senescent human cells. In an effort to understand human-specific responses to telomere dysfunction, we modeled telomere dysfunction-induced senescence in a human alveolar epithelial cell line. We hypothesized that this system would enable us to probe for differences in transcriptional and proteomic senescence pathways in vitro and to identify novel secreted protein (secretome) changes that potentially contribute to the pathogenesis of IPF. Following induction of telomere dysfunction, a robust senescence phenotype was observed. RNA-seq analysis of the senescent cells revealed the SASP and comparisons to previous murine data highlighted differences in response to telomere dysfunction. We conducted a proteomic analysis of the senescent cells using a novel biotin ligase capable of labeling secreted proteins. Candidate biomarkers selected from our transcriptional and secretome data were then evaluated in IPF and control patient plasma. Four novel proteins were found to be differentially expressed between the patient groups: stanniocalcin-1, contactin-1, tenascin C, and total inhibin. Our data show that human telomere-induced, alveolar epithelial senescence results in a transcriptional SASP that is distinct from that seen in analogous murine cells. Our findings suggest that studies in animal models should be carefully validated given the possibility of species-specific responses to telomere dysfunction. We also describe a pragmatic approach for the study of the consequences of telomere-induced alveolar epithelial cell senescence in humans.
Collapse
Affiliation(s)
- Daniel I. Sullivan
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, United States,Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, Pittsburgh, PA, United States
| | - Mao Jiang
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, United States,The Third Xiangya Hospital, Central South University, Changsha, China
| | - Angela M. Hinchie
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Mark G. Roth
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Harinath Bahudhanapati
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, United States,Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, Pittsburgh, PA, United States
| | - Mehdi Nouraie
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, United States,Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, Pittsburgh, PA, United States
| | - Jie Liu
- Aging Institute, University of Pittsburgh, Pittsburgh, PA, United States,University of Pittsburgh Medical Center, Pittsburgh, PA, United States,Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - John F. McDyer
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Rama K. Mallampalli
- Department of Internal Medicine, The Ohio State University, Columbus, OH, United States
| | - Yingze Zhang
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, United States,Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, Pittsburgh, PA, United States
| | - Daniel J. Kass
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, United States,Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, Pittsburgh, PA, United States
| | - Toren Finkel
- Aging Institute, University of Pittsburgh, Pittsburgh, PA, United States,University of Pittsburgh Medical Center, Pittsburgh, PA, United States,Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Jonathan K. Alder
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, United States,Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, Pittsburgh, PA, United States,*Correspondence: Jonathan K. Alder
| |
Collapse
|
32
|
Wang X, Bootsma H, Terpstra J, Vissink A, van der Vegt B, Spijkervet FKL, Kroese FGM, Pringle S. Progenitor cell niche senescence reflects pathology of the parotid salivary gland in primary Sjögren's syndrome. Rheumatology (Oxford) 2021; 59:3003-3013. [PMID: 32159757 PMCID: PMC7516109 DOI: 10.1093/rheumatology/keaa012] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 12/20/2019] [Indexed: 12/13/2022] Open
Abstract
Objective Salivary gland (SG) progenitor cells (SGPCs) maintain SG homeostasis. We have previously shown that in primary Sjögren’s syndrome (pSS), SGPCs are likely to be senescent, and may underpin SG dysfunction. This study assessed the extent of senescence of cells in a SGPC niche in pSS patients’ SGs, and its correlation with functional and clinical parameters. Methods The expression of p16 and p21 as markers of senescence in both total SG epithelium and a SGPC niche (basal striated duct cells, BSD) was examined in SGs of pSS (n = 35), incomplete pSS (n = 13) (patients with some signs of pSS, but not fulfilling all classification criteria) and non-SS sicca control (n = 21) patients. This was correlated with functional and clinical parameters. Results pSS patient SGs contained significantly more p16+ cells both in the epithelium in general (P <0.01) and in the BSD layer (P <0.001), than non-SS SGs. Significant correlations were found in pSS patients between p16+ BSD cells and secretion of unstimulated whole saliva, stimulated whole saliva, stimulated parotid saliva, CD45+ infiltrate, ultrasound total score and ACR-EULAR classification score, but not with EULAR Sjögren’s syndrome disease activity index (ESSDAI) and EULAR Sjögren’s Syndrome Patient Reported Index (ESSPRI) scores. Correlations with total epithelium p16+ cells were weaker. Incomplete pSS patients also had increased numbers of p16+ epithelial and BSD cells. Based on protein and mRNA expression, p21+ appears not to play a significant role in the SG in pSS. Conclusion These findings suggest SGPC senescence may be an early feature of primary Sjögren’s syndrome and may contribute to defective SG function in pSS but not to systemic disease activity.
Collapse
Affiliation(s)
- Xiaoyan Wang
- Department of Rheumatology and Clinical ImmunologyUniversity Medical Center Groningen, Groningen, The Netherlands
| | - Hendrika Bootsma
- Department of Rheumatology and Clinical ImmunologyUniversity Medical Center Groningen, Groningen, The Netherlands
| | - Janneke Terpstra
- Department of Rheumatology and Clinical ImmunologyUniversity Medical Center Groningen, Groningen, The Netherlands
| | - Arjan Vissink
- Department of Oral and Maxillofacial Surgery, University Medical Center Groningen, Groningen, The Netherlands
| | - Bert van der Vegt
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Fred K L Spijkervet
- Department of Oral and Maxillofacial Surgery, University Medical Center Groningen, Groningen, The Netherlands
| | - Frans G M Kroese
- Department of Rheumatology and Clinical ImmunologyUniversity Medical Center Groningen, Groningen, The Netherlands
| | - Sarah Pringle
- Department of Rheumatology and Clinical ImmunologyUniversity Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
33
|
Sławińska N, Krupa R. Molecular Aspects of Senescence and Organismal Ageing-DNA Damage Response, Telomeres, Inflammation and Chromatin. Int J Mol Sci 2021; 22:ijms22020590. [PMID: 33435578 PMCID: PMC7827783 DOI: 10.3390/ijms22020590] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/30/2020] [Accepted: 01/03/2021] [Indexed: 02/07/2023] Open
Abstract
Cells can become senescent in response to stress. Senescence is a process characterised by a stable proliferative arrest. Sometimes it can be beneficial—for example, it can suppress tumour development or take part in tissue repair. On the other hand, studies show that it is also involved in the ageing process. DNA damage response (DDR) is triggered by DNA damage or telomere shortening during cell division. When left unresolved, it may lead to the activation of senescence. Senescent cells secrete certain proteins in larger quantities. This phenomenon is referred to as senescence-associated secretory phenotype (SASP). SASP can induce senescence in other cells; evidence suggests that overabundance of senescent cells contributes to ageing. SASP proteins include proinflammatory cytokines and metalloproteinases, which degrade the extracellular matrix. Shortening of telomeres is another feature associated with organismal ageing. Older organisms have shorter telomeres. Restoring telomerase activity in mice not only slowed but also partially reversed the symptoms of ageing. Changes in chromatin structure during senescence include heterochromatin formation or decondensation and loss of H1 histones. During organismal ageing, cells can experience heterochromatin loss, DNA demethylation and global histone loss. Cellular and organismal ageing are both complex processes with many aspects that are often related. The purpose of this review is to bring some of these aspects forward and provide details regarding them.
Collapse
|
34
|
Engin AB, Engin A. The Connection Between Cell Fate and Telomere. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1275:71-100. [PMID: 33539012 DOI: 10.1007/978-3-030-49844-3_3] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Abolition of telomerase activity results in telomere shortening, a process that eventually destabilizes the ends of chromosomes, leading to genomic instability and cell growth arrest or death. Telomere shortening leads to the attainment of the "Hayflick limit", and the transition of cells to state of senescence. If senescence is bypassed, cells undergo crisis through loss of checkpoints. This process causes massive cell death concomitant with further telomere shortening and spontaneous telomere fusions. In functional telomere of mammalian cells, DNA contains double-stranded tandem repeats of TTAGGG. The Shelterin complex, which is composed of six different proteins, is required for the regulation of telomere length and stability in cells. Telomere protection by telomeric repeat binding protein 2 (TRF2) is dependent on DNA damage response (DDR) inhibition via formation of T-loop structures. Many protein kinases contribute to the DDR activated cell cycle checkpoint pathways, and prevent DNA replication until damaged DNA is repaired. Thereby, the connection between cell fate and telomere length-associated telomerase activity is regulated by multiple protein kinase activities. Contrarily, inactivation of DNA damage checkpoint protein kinases in senescent cells can restore cell-cycle progression into S phase. Therefore, telomere-initiated senescence is a DNA damage checkpoint response that is activated with a direct contribution from dysfunctional telomeres. In this review, in addition to the above mentioned, the choice of main repair pathways, which comprise non-homologous end joining and homologous recombination in telomere uncapping telomere dysfunctions, are discussed.
Collapse
Affiliation(s)
- Ayse Basak Engin
- Department of Toxicology, Faculty of Pharmacy, Gazi University, Ankara, Turkey.
| | - Atilla Engin
- Department of General Surgery, Faculty of Medicine, Gazi University, Ankara, Turkey
| |
Collapse
|
35
|
Schmutz I, Mensenkamp AR, Takai KK, Haadsma M, Spruijt L, de Voer RM, Choo SS, Lorbeer FK, van Grinsven EJ, Hockemeyer D, Jongmans MCJ, de Lange T. TINF2 is a haploinsufficient tumor suppressor that limits telomere length. eLife 2020; 9:e61235. [PMID: 33258446 PMCID: PMC7707837 DOI: 10.7554/elife.61235] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 11/11/2020] [Indexed: 12/20/2022] Open
Abstract
Telomere shortening is a presumed tumor suppressor pathway that imposes a proliferative barrier (the Hayflick limit) during tumorigenesis. This model predicts that excessively long somatic telomeres predispose to cancer. Here, we describe cancer-prone families with two unique TINF2 mutations that truncate TIN2, a shelterin subunit that controls telomere length. Patient lymphocyte telomeres were unusually long. We show that the truncated TIN2 proteins do not localize to telomeres, suggesting that the mutations create loss-of-function alleles. Heterozygous knock-in of the mutations or deletion of one copy of TINF2 resulted in excessive telomere elongation in clonal lines, indicating that TINF2 is haploinsufficient for telomere length control. In contrast, telomere protection and genome stability were maintained in all heterozygous clones. The data establish that the TINF2 truncations predispose to a tumor syndrome. We conclude that TINF2 acts as a haploinsufficient tumor suppressor that limits telomere length to ensure a timely Hayflick limit.
Collapse
Affiliation(s)
- Isabelle Schmutz
- Laboratory for Cell Biology and Genetics, Rockefeller UniversityNew YorkUnited States
| | - Arjen R Mensenkamp
- Department of Human Genetics, Radboud University Medical CenterNijmegenNetherlands
| | - Kaori K Takai
- Laboratory for Cell Biology and Genetics, Rockefeller UniversityNew YorkUnited States
| | - Maaike Haadsma
- Department of Human Genetics, Radboud University Medical CenterNijmegenNetherlands
| | - Liesbeth Spruijt
- Department of Human Genetics, Radboud University Medical CenterNijmegenNetherlands
| | - Richarda M de Voer
- Department of Human Genetics, Radboud University Medical CenterNijmegenNetherlands
| | - Seunga Sara Choo
- Department of Molecular and Cellular Biology, University of California, BerkeleyBerkeleyUnited States
| | - Franziska K Lorbeer
- Department of Molecular and Cellular Biology, University of California, BerkeleyBerkeleyUnited States
| | - Emma J van Grinsven
- Department of Molecular and Cellular Biology, University of California, BerkeleyBerkeleyUnited States
| | - Dirk Hockemeyer
- Department of Molecular and Cellular Biology, University of California, BerkeleyBerkeleyUnited States
- Chan Zuckerberg BiohubSan FranciscoUnited States
| | | | - Titia de Lange
- Laboratory for Cell Biology and Genetics, Rockefeller UniversityNew YorkUnited States
| |
Collapse
|
36
|
Naikawadi RP, Green G, Jones KD, Achtar-Zadeh N, Mieleszko JE, Arnould I, Kukreja J, Greenland JR, Wolters PJ. Airway Epithelial Telomere Dysfunction Drives Remodeling Similar to Chronic Lung Allograft Dysfunction. Am J Respir Cell Mol Biol 2020; 63:490-501. [PMID: 32551854 DOI: 10.1165/rcmb.2019-0374oc] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Telomere dysfunction is associated with multiple fibrotic lung processes, including chronic lung allograft dysfunction (CLAD)-the major limitation to long-term survival following lung transplantation. Although shorter donor telomere lengths are associated with an increased risk of CLAD, it is unknown whether short telomeres are a cause or consequence of CLAD pathology. Our objective was to test whether telomere dysfunction contributes to the pathologic changes observed in CLAD. Histopathologic and molecular analysis of human CLAD lungs demonstrated shortened telomeres in lung epithelial cells quantified by teloFISH, increased numbers of surfactant protein C immunoreactive type II alveolar epithelial cells, and increased expression of senescence markers (β-galactosidase, p16, p53, and p21) in lung epithelial cells. TRF1F/F (telomere repeat binding factor 1 flox/flox) mice were crossed with tamoxifen-inducible SCGB1a1-cre mice to generate SCGB1a1-creTRF1F/F mice. Following 9 months of tamoxifen-induced deletion of TRF1 in club cells, mice developed mixed obstructive and restrictive lung physiology, small airway obliteration on microcomputed tomography, a fourfold decrease in telomere length in airway epithelial cells, collagen deposition around bronchioles and adjacent lung parenchyma, increased type II aveolar epithelial cell numbers, expression of senescence-associated β-galactosidase in epithelial cells, and decreased SCGB1a1 expression in airway epithelial cells. These findings demonstrate that telomere dysfunction isolated to airway epithelial cells leads to airway-centric lung remodeling and fibrosis similar to that observed in patients with CLAD and suggest that lung epithelial cell telomere dysfunction may be a molecular driver of CLAD.
Collapse
Affiliation(s)
- Ram P Naikawadi
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine
| | - Gary Green
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine
| | | | - Natalia Achtar-Zadeh
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine
| | - Julia E Mieleszko
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine
| | - Isabel Arnould
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine
| | - Jasleen Kukreja
- Department of Surgery, University of California, San Francisco, California; and
| | - John R Greenland
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine.,Medical Service, Veterans Affairs Health Care System, San Francisco, California
| | - Paul J Wolters
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine
| |
Collapse
|
37
|
Fundamental insights into the interaction between telomerase/TERT and intracellular signaling pathways. Biochimie 2020; 181:12-24. [PMID: 33232793 DOI: 10.1016/j.biochi.2020.11.015] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 11/07/2020] [Accepted: 11/16/2020] [Indexed: 12/12/2022]
Abstract
Telomerase activity is critical for cancer cells to provide unrestricted proliferation and cellular immortality through maintaining telomeres. Telomerase enzymatic activity is regulatable at the level of DNA, mRNA, post translational modifications, cellular transport and enzyme assembly. More recent studies confirm the interaction of the telomerase with various intracellular signaling pathways including PI3K/AKT/mTOR, NF-κB and Wnt/β-catenin which mainly participating in inflammation, epithelial to mesenchymal transition (EMT) and tumor cell invasion and metastasis. Furthermore, hTERT protein has been detected in non-nuclear sites such as the mitochondria and cytoplasm in cells. Mitochondrial TERT indicates various non-telomere-related functions such as decreasing reactive oxygen species (ROS) generation, boosting the respiration rate, protecting mtDNA by direct binding, interacting with mitochondrial tRNAs and increasing mitochondrial membrane potential which can lead to higher chemoresistance rate in cancer cells during therapies. Understanding the molecular mechanisms of the TERT function and depended interactions in tumor cells can suggest novel therapeutic approaches. Hence, in this review we will explain the telomerase activity regulation in translational and post translational levels besides the established correlations with various cell signaling pathways with possible pathways for therapeutic targeting.
Collapse
|
38
|
Resnik SR, Egger A, Abdo Abujamra B, Jozic I. Clinical Implications of Cellular Senescence on Wound Healing. CURRENT DERMATOLOGY REPORTS 2020. [DOI: 10.1007/s13671-020-00320-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
39
|
Drosopoulos WC, Deng Z, Twayana S, Kosiyatrakul ST, Vladimirova O, Lieberman PM, Schildkraut CL. TRF2 Mediates Replication Initiation within Human Telomeres to Prevent Telomere Dysfunction. Cell Rep 2020; 33:108379. [PMID: 33176153 PMCID: PMC7790361 DOI: 10.1016/j.celrep.2020.108379] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 09/15/2020] [Accepted: 10/20/2020] [Indexed: 02/06/2023] Open
Abstract
The telomeric shelterin protein telomeric repeat-binding factor 2 (TRF2) recruits origin recognition complex (ORC) proteins, the foundational building blocks of DNA replication origins, to telomeres. We seek to determine whether TRF2-recruited ORC proteins give rise to functional origins in telomere repeat tracts. We find that reduction of telomeric recruitment of ORC2 by expression of an ORC interaction-defective TRF2 mutant significantly reduces telomeric initiation events in human cells. This reduction in initiation events is accompanied by telomere repeat loss, telomere aberrations and dysfunction. We demonstrate that telomeric origins are activated by induced replication stress to provide a key rescue mechanism for completing compromised telomere replication. Importantly, our studies also indicate that the chromatin remodeler SNF2H promotes telomeric initiation events by providing access for ORC2. Collectively, our findings reveal that active recruitment of ORC by TRF2 leads to formation of functional origins, providing an important mechanism for avoiding telomere dysfunction and rescuing challenged telomere replication.
Collapse
Affiliation(s)
- William C Drosopoulos
- Department of Cell Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA.
| | - Zhong Deng
- Gene Expression and Regulation Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Shyam Twayana
- Department of Cell Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - Settapong T Kosiyatrakul
- Department of Cell Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - Olga Vladimirova
- Gene Expression and Regulation Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Paul M Lieberman
- Gene Expression and Regulation Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Carl L Schildkraut
- Department of Cell Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA.
| |
Collapse
|
40
|
Fernandes SG, Dsouza R, Pandya G, Kirtonia A, Tergaonkar V, Lee SY, Garg M, Khattar E. Role of Telomeres and Telomeric Proteins in Human Malignancies and Their Therapeutic Potential. Cancers (Basel) 2020; 12:E1901. [PMID: 32674474 PMCID: PMC7409176 DOI: 10.3390/cancers12071901] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/10/2020] [Accepted: 07/13/2020] [Indexed: 12/19/2022] Open
Abstract
Telomeres are the ends of linear chromosomes comprised of repetitive nucleotide sequences in humans. Telomeres preserve chromosomal stability and genomic integrity. Telomere length shortens with every cell division in somatic cells, eventually resulting in replicative senescence once telomere length becomes critically short. Telomere shortening can be overcome by telomerase enzyme activity that is undetectable in somatic cells, while being active in germline cells, stem cells, and immune cells. Telomeres are bound by a shelterin complex that regulates telomere lengthening as well as protects them from being identified as DNA damage sites. Telomeres are transcribed by RNA polymerase II, and generate a long noncoding RNA called telomeric repeat-containing RNA (TERRA), which plays a key role in regulating subtelomeric gene expression. Replicative immortality and genome instability are hallmarks of cancer and to attain them cancer cells exploit telomere maintenance and telomere protection mechanisms. Thus, understanding the role of telomeres and their associated proteins in cancer initiation, progression and treatment is very important. The present review highlights the critical role of various telomeric components with recently established functions in cancer. Further, current strategies to target various telomeric components including human telomerase reverse transcriptase (hTERT) as a therapeutic approach in human malignancies are discussed.
Collapse
Affiliation(s)
- Stina George Fernandes
- Sunandan Divatia School of Science, SVKM’s NMIMS (Deemed to be University), Vile Parle West, Mumbai 400056, India; (S.G.F.); (R.D.)
| | - Rebecca Dsouza
- Sunandan Divatia School of Science, SVKM’s NMIMS (Deemed to be University), Vile Parle West, Mumbai 400056, India; (S.G.F.); (R.D.)
| | - Gouri Pandya
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University Uttar Pradesh, Noida 201313, India; (G.P.); (A.K.)
| | - Anuradha Kirtonia
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University Uttar Pradesh, Noida 201313, India; (G.P.); (A.K.)
| | - Vinay Tergaonkar
- Laboratory of NF-κB Signaling, Institute of Molecular and Cell Biology (IMCB), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore; (V.T.); (S.Y.L.)
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore 117597, Singapore
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore 117597, Singapore
| | - Sook Y. Lee
- Laboratory of NF-κB Signaling, Institute of Molecular and Cell Biology (IMCB), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore; (V.T.); (S.Y.L.)
| | - Manoj Garg
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University Uttar Pradesh, Noida 201313, India; (G.P.); (A.K.)
| | - Ekta Khattar
- Sunandan Divatia School of Science, SVKM’s NMIMS (Deemed to be University), Vile Parle West, Mumbai 400056, India; (S.G.F.); (R.D.)
| |
Collapse
|
41
|
Gilbert-Girard S, Gravel A, Collin V, Wight DJ, Kaufer BB, Lazzerini-Denchi E, Flamand L. Role for the shelterin protein TRF2 in human herpesvirus 6A/B chromosomal integration. PLoS Pathog 2020; 16:e1008496. [PMID: 32320442 PMCID: PMC7197865 DOI: 10.1371/journal.ppat.1008496] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 05/04/2020] [Accepted: 03/24/2020] [Indexed: 02/07/2023] Open
Abstract
Human herpesviruses 6A and 6B (HHV-6A/B) are unique among human herpesviruses in their ability to integrate their genome into host chromosomes. Viral integration occurs at the ends of chromosomes within the host telomeres. The ends of the HHV-6A/B genomes contain telomeric repeats that facilitate the integration process. Here, we report that productive infections are associated with a massive increase in telomeric sequences of viral origin. The majority of the viral telomeric signals can be detected within viral replication compartments (VRC) that contain the viral DNA processivity factor p41 and the viral immediate-early 2 (IE2) protein. Components of the shelterin protein complex present at telomeres, including TRF1 and TRF2 are also recruited to VRC during infection. Biochemical, immunofluorescence coupled with in situ hybridization and chromatin immunoprecipitation demonstrated the binding of TRF2 to the HHV-6A/B telomeric repeats. In addition, approximately 60% of the viral IE2 protein localize at cellular telomeres during infection. Transient knockdown of TRF2 resulted in greatly reduced (13%) localization of IE2 at cellular telomeres (p<0.0001). Lastly, TRF2 knockdown reduced HHV-6A/B integration frequency (p<0.05), while no effect was observed on the infection efficiency. Overall, our study identified that HHV-6A/B IE2 localizes to telomeres during infection and highlight the role of TRF2 in HHV-6A/B infection and chromosomal integration.
Collapse
Affiliation(s)
- Shella Gilbert-Girard
- Division of Infectious Disease and Immunity, CHU de Québec Research Center, Quebec City, Quebec, Canada
| | - Annie Gravel
- Division of Infectious Disease and Immunity, CHU de Québec Research Center, Quebec City, Quebec, Canada
| | - Vanessa Collin
- Division of Infectious Disease and Immunity, CHU de Québec Research Center, Quebec City, Quebec, Canada
| | - Darren J. Wight
- Institut für Virologie, Freie Universität Berlin, Berlin, Germany
| | | | - Eros Lazzerini-Denchi
- Laboratory of Genome Integrity, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Louis Flamand
- Division of Infectious Disease and Immunity, CHU de Québec Research Center, Quebec City, Quebec, Canada
- Department of microbiology, infectious diseases and immunology, Faculty of Medicine, Université Laval, Quebec City, Québec, Canada
- * E-mail:
| |
Collapse
|
42
|
Sándor S, Kubinyi E. Genetic Pathways of Aging and Their Relevance in the Dog as a Natural Model of Human Aging. Front Genet 2019; 10:948. [PMID: 31681409 PMCID: PMC6813227 DOI: 10.3389/fgene.2019.00948] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 09/05/2019] [Indexed: 12/15/2022] Open
Abstract
Aging research has experienced a burst of scientific efforts in the last decades as the growing ratio of elderly people has begun to pose an increased burden on the healthcare and pension systems of developed countries. Although many breakthroughs have been reported in understanding the cellular mechanisms of aging, the intrinsic and extrinsic factors that contribute to senescence on higher biological levels are still barely understood. The dog, Canis familiaris, has already served as a valuable model of human physiology and disease. The possible role the dog could play in aging research is still an open question, although utilization of dogs may hold great promises as they naturally develop age-related cognitive decline, with behavioral and histological characteristics very similar to those of humans. In this regard, family dogs may possess unmatched potentials as models for investigations on the complex interactions between environmental, behavioral, and genetic factors that determine the course of aging. In this review, we summarize the known genetic pathways in aging and their relevance in dogs, putting emphasis on the yet barely described nature of certain aging pathways in canines. Reasons for highlighting the dog as a future aging and gerontology model are also discussed, ranging from its unique evolutionary path shared with humans, its social skills, and the fact that family dogs live together with their owners, and are being exposed to the same environmental effects.
Collapse
Affiliation(s)
- Sára Sándor
- Department of Ethology, Eötvös Loránd University, Budapest, Hungary
| | | |
Collapse
|
43
|
Transient induction of telomerase expression mediates senescence and reduces tumorigenesis in primary fibroblasts. Proc Natl Acad Sci U S A 2019; 116:18983-18993. [PMID: 31481614 PMCID: PMC6754593 DOI: 10.1073/pnas.1907199116] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Telomerase is an enzymatic ribonucleoprotein complex that acts as a reverse transcriptase in the elongation of telomeres. Telomerase activity is well documented in embryonic stem cells and the vast majority of tumor cells, but its role in somatic cells remains to be understood. Here, we report an unexpected function of telomerase during cellular senescence and tumorigenesis. We crossed Tert heterozygous knockout mice (mTert +/- ) for 26 generations, during which time there was progressive shortening of telomeres, and obtained primary skin fibroblasts from mTert +/+ and mTert -/- progeny of the 26th cross. As a consequence of insufficient telomerase activities in prior generations, both mTert +/+ and mTert -/- fibroblasts showed comparable and extremely short telomere length. However, mTert -/- cells approached cellular senescence faster and exhibited a significantly higher rate of malignant transformation than mTert +/+ cells. Furthermore, an evident up-regulation of telomerase reverse-transcriptase (TERT) expression was detected in mTert +/+ cells at the presenescence stage. Moreover, removal or down-regulation of TERT expression in mTert +/+ and human primary fibroblast cells via CRISPR/Cas9 or shRNA recapitulated mTert -/- phenotypes of accelerated senescence and transformation, and overexpression of TERT in mTert -/- cells rescued these phenotypes. Taking these data together, this study suggests that TERT has a previously underappreciated, protective role in buffering senescence stresses due to short, dysfunctional telomeres, and preventing malignant transformation.
Collapse
|
44
|
Wallace HA, Rana V, Nguyen HQ, Bosco G. Condensin II subunit NCAPH2 associates with shelterin protein TRF1 and is required for telomere stability. J Cell Physiol 2019; 234:20755-20768. [PMID: 31026066 PMCID: PMC6767372 DOI: 10.1002/jcp.28681] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 03/19/2019] [Indexed: 12/24/2022]
Abstract
Condensin II subunits are known to be expressed and localized to interphase nuclei of eukaryotic cells. Although some studies have shown that condensin II likely exerts axial compaction forces, organizes chromosome territories, and has possible transcriptional modulatory functions, the full range of condensin II interphase activities are not known. In particular, it is not known if condensin II interphase activities are generally genome‐wide or if they have additional local activities unique to specific chromosomal structures such as telomeres. Here, we find that NCAPH2 interacts with TRF1 and these two proteins co‐localize at telomeres. Depletion of NCAPH2 leads to ATR‐dependent accumulation of 53BP1 and γH2AX DNA damage foci, including damage specific to telomeres. Furthermore, depletion of NCAPH2 results in a fragile telomere phenotype and apparent sister‐telomere fusions only days after NCAPH2 depletion. Taken together these observations suggest that NCAPH2 promotes telomere stability, possibly through a direct interaction with the TRF1 shelterin component, and prevents telomere dysfunction resulting from impaired DNA replication. Because proper telomere function is essential for chromosome integrity these observations reveal a previously unappreciated function for NCAPH2 in ensuring genome and telomere stability.
Collapse
Affiliation(s)
| | - Vibhuti Rana
- Department of Genetics, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Huy Q Nguyen
- Department of Genetics, Harvard Medical School, Boston, Massachusetts
| | - Giovanni Bosco
- Department of Genetics, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| |
Collapse
|
45
|
Zhang Z, Zhang T, Ge Y, Tang M, Ma W, Zhang Q, Gong S, Wright WE, Shay J, Liu H, Zhao Y. 2D gel electrophoresis reveals dynamics of t-loop formation during the cell cycle and t-loop in maintenance regulated by heterochromatin state. J Biol Chem 2019; 294:6645-6656. [PMID: 30819801 DOI: 10.1074/jbc.ra119.007677] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 02/20/2019] [Indexed: 12/15/2022] Open
Abstract
Linear chromosome ends are capped by telomeres that have been previously reported to adopt a t-loop structure. The lack of simple methods for detecting t-loops has hindered progress in understanding the dynamics of t-loop formation and its function in protecting chromosome ends. Here, we employed a classical two-dimensional agarose gel method (2D gel method) to innovatively apply to t-loop detection. Briefly, restriction fragments of genomic DNA were separated in a 2D gel, and the telomere sequence was detected by in-gel hybridization with telomeric probe. Using this method, we found that t-loops are present throughout the cell cycle, and t-loop formation tightly couples to telomere replication. We also observed that t-loop abundance positively correlates with chromatin condensation, i.e. cells with less compact telomeric chromatin (ALT cells and trichostatin A (TSA)-treated HeLa cells) exhibited fewer t-loops. Moreover, we observed that telomere dysfunction-induced foci, ALT-associated promyelocytic leukemia bodies, and telomere sister chromatid exchanges are activated upon TSA-induced loss of t-loops. These findings confirm the importance of the t-loop in protecting linear chromosomes from damage or illegitimate recombination.
Collapse
Affiliation(s)
- Zepeng Zhang
- From the MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Tianpeng Zhang
- From the MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Yuanlong Ge
- From the MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Mengfan Tang
- From the MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Wenbin Ma
- From the MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Qinfen Zhang
- From the MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Shengzhao Gong
- Department of Chemical Engineering, Guangdong Industry Technical College, Guangzhou 510006, China
| | - Woodring E Wright
- the Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, and
| | - Jerry Shay
- the Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, and
| | - Haiying Liu
- From the MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510006, China,
| | - Yong Zhao
- From the MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510006, China,
| |
Collapse
|
46
|
Nassrally MS, Lau A, Wise K, John N, Kotecha S, Lee KL, Brooks RF. Cell cycle arrest in replicative senescence is not an immediate consequence of telomere dysfunction. Mech Ageing Dev 2019; 179:11-22. [PMID: 30710559 DOI: 10.1016/j.mad.2019.01.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 12/19/2018] [Accepted: 01/28/2019] [Indexed: 11/15/2022]
Abstract
In replicative senescence, cells with critically-short telomeres activate a DNA-damage response leading to cell-cycle arrest, while those without telomere dysfunction would be expected to cycle normally. However, population growth declines more gradually than such a simple binary switch between cycling and non-cycling states would predict. We show here that late-passage cultures of human fibroblasts are not a simple mixture of cycling and non-cycling cells. Rather, although some cells had short cycle times comparable to those of younger cells, others continued to divide but with greatly extended cycle times, indicating a more-gradual approach to permanent arrest. Remarkably, in late passage cells, the majority showed prominent DNA-damage foci positive for 53BP1, yet many continued to divide. Evidently, the DNA-damage-response elicited by critically-short telomeres is not initially strong enough for complete cell-cycle arrest. A similar continuation of the cell cycle in the face of an active DNA-damage response was also seen in cells treated with a low dose of doxorubicin sufficient to produce multiple 53BP1 foci in all nuclei. Cell cycle checkpoint engagement in response to DNA damage is thus weaker than generally supposed, explaining why an accumulation of dysfunctional telomeres is needed before marked cell cycle elongation or permanent arrest is achieved.
Collapse
Affiliation(s)
- M Shamim Nassrally
- King's College London, Faculty of Life Sciences & Medicine, Department of Anatomy, Guy's Campus, LONDON SE1 1UL, UK
| | - Ashley Lau
- King's College London, Faculty of Life Sciences & Medicine, Department of Anatomy, Guy's Campus, LONDON SE1 1UL, UK
| | - Katherine Wise
- King's College London, Faculty of Life Sciences & Medicine, Department of Anatomy, Guy's Campus, LONDON SE1 1UL, UK
| | - Noah John
- King's College London, Faculty of Life Sciences & Medicine, Department of Anatomy, Guy's Campus, LONDON SE1 1UL, UK
| | - Sanjeev Kotecha
- King's College London, Faculty of Life Sciences & Medicine, Department of Anatomy, Guy's Campus, LONDON SE1 1UL, UK
| | - Kar Lai Lee
- King's College London, Faculty of Life Sciences & Medicine, Department of Anatomy, Guy's Campus, LONDON SE1 1UL, UK
| | - Robert F Brooks
- King's College London, Faculty of Life Sciences & Medicine, Department of Anatomy, Guy's Campus, LONDON SE1 1UL, UK; St George's, University of London, Molecular and Clinical Sciences Research Institute, Mailpoint J2A, Cranmer Terrace, London, SW17 0RE, UK.
| |
Collapse
|
47
|
Genome-wide Control of Heterochromatin Replication by the Telomere Capping Protein TRF2. Mol Cell 2019; 70:449-461.e5. [PMID: 29727617 DOI: 10.1016/j.molcel.2018.03.036] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 02/20/2018] [Accepted: 03/29/2018] [Indexed: 11/22/2022]
Abstract
Hard-to-replicate regions of chromosomes (e.g., pericentromeres, centromeres, and telomeres) impede replication fork progression, eventually leading, in the event of replication stress, to chromosome fragility, aging, and cancer. Our knowledge of the mechanisms controlling the stability of these regions is essentially limited to telomeres, where fragility is counteracted by the shelterin proteins. Here we show that the shelterin subunit TRF2 ensures progression of the replication fork through pericentromeric heterochromatin, but not centromeric chromatin. In a process involving its N-terminal basic domain, TRF2 binds to pericentromeric Satellite III sequences during S phase, allowing the recruitment of the G-quadruplex-resolving helicase RTEL1 to facilitate fork progression. We also show that TRF2 is required for the stability of other heterochromatic regions localized throughout the genome, paving the way for future research on heterochromatic replication and its relationship with aging and cancer.
Collapse
|
48
|
Jiang CM, Liu X, Li CX, Qian HC, Chen D, Lai CQ, Shen LR. Anti-senescence effect and molecular mechanism of the major royal jelly proteins on human embryonic lung fibroblast (HFL-I) cell line. J Zhejiang Univ Sci B 2018; 19:960-972. [PMID: 30507079 PMCID: PMC6305251 DOI: 10.1631/jzus.b1800257] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Revised: 09/09/2018] [Accepted: 09/09/2018] [Indexed: 12/15/2022]
Abstract
Royal jelly (RJ) from honeybee has been widely used as a health promotion supplement. The major royal jelly proteins (MRJPs) have been identified as the functional component of RJ. However, the question of whether MRJPs have anti-senescence activity for human cells remains. Human embryonic lung fibroblast (HFL-I) cells were cultured in media containing no MRJPs (A), MRJPs at 0.1 mg/ml (B), 0.2 mg/ml (C), or 0.3 mg/ml (D), or bovine serum albumin (BSA) at 0.2 mg/ml (E). The mean population doubling levels of cells in media B, C, D, and E were increased by 12.4%, 31.2%, 24.0%, and 10.4%, respectively, compared with that in medium A. The cells in medium C also exhibited the highest relative proliferation activity, the lowest senescence, and the longest telomeres. Moreover, MRJPs up-regulated the expression of superoxide dismutase-1 (SOD1) and down-regulated the expression of mammalian target of rapamycin (MTOR), catenin beta like-1 (CTNNB1), and tumor protein p53 (TP53). Raman spectra analysis showed that there were two unique bands related to DNA synthesis materials, amide carbonyl group vibrations and aromatic hydrogens. These results suggest that MRJPs possess anti-senescence activity for the HFL-I cell line, and provide new knowledge illustrating the molecular mechanism of MRJPs as anti-senescence factors.
Collapse
Affiliation(s)
- Chen-min Jiang
- College of Biosystems Engineering and Food Science, Zhejiang University / Key Laboratory of Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs / Zhejiang Key Laboratory for Agri-Food Processing, Hangzhou 310058, China
| | - Xin Liu
- College of Biosystems Engineering and Food Science, Zhejiang University / Key Laboratory of Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs / Zhejiang Key Laboratory for Agri-Food Processing, Hangzhou 310058, China
| | - Chun-xue Li
- College of Biosystems Engineering and Food Science, Zhejiang University / Key Laboratory of Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs / Zhejiang Key Laboratory for Agri-Food Processing, Hangzhou 310058, China
| | - Hao-cheng Qian
- College of Biosystems Engineering and Food Science, Zhejiang University / Key Laboratory of Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs / Zhejiang Key Laboratory for Agri-Food Processing, Hangzhou 310058, China
| | - Di Chen
- College of Biosystems Engineering and Food Science, Zhejiang University / Key Laboratory of Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs / Zhejiang Key Laboratory for Agri-Food Processing, Hangzhou 310058, China
| | - Chao-qiang Lai
- USDA ARS Nutritional Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, Massachusetts 02111, the United States
| | - Li-rong Shen
- College of Biosystems Engineering and Food Science, Zhejiang University / Key Laboratory of Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs / Zhejiang Key Laboratory for Agri-Food Processing, Hangzhou 310058, China
| |
Collapse
|
49
|
Wang H, Xu X, Nguyen CM, Liu Y, Gao Y, Lin X, Daley T, Kipniss NH, La Russa M, Qi LS. CRISPR-Mediated Programmable 3D Genome Positioning and Nuclear Organization. Cell 2018; 175:1405-1417.e14. [PMID: 30318144 PMCID: PMC6239909 DOI: 10.1016/j.cell.2018.09.013] [Citation(s) in RCA: 141] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 08/09/2018] [Accepted: 09/10/2018] [Indexed: 12/20/2022]
Abstract
Programmable control of spatial genome organization is a powerful approach for studying how nuclear structure affects gene regulation and cellular function. Here, we develop a versatile CRISPR-genome organization (CRISPR-GO) system that can efficiently control the spatial positioning of genomic loci relative to specific nuclear compartments, including the nuclear periphery, Cajal bodies, and promyelocytic leukemia (PML) bodies. CRISPR-GO is chemically inducible and reversible, enabling interrogation of real-time dynamics of chromatin interactions with nuclear compartments in living cells. Inducible repositioning of genomic loci to the nuclear periphery allows for dissection of mitosis-dependent and -independent relocalization events and also for interrogation of the relationship between gene position and gene expression. CRISPR-GO mediates rapid de novo formation of Cajal bodies at desired chromatin loci and causes significant repression of endogenous gene expression over long distances (30-600 kb). The CRISPR-GO system offers a programmable platform to investigate large-scale spatial genome organization and function.
Collapse
Affiliation(s)
- Haifeng Wang
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Xiaoshu Xu
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Cindy M Nguyen
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Yanxia Liu
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Yuchen Gao
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Cancer Biology Program, Stanford University, Stanford, CA 94305, USA
| | - Xueqiu Lin
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Timothy Daley
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Department of Statistics, Stanford University, Stanford, CA 94305, USA
| | - Nathan H Kipniss
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Marie La Russa
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Lei S Qi
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA; Stanford ChEM-H, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
50
|
Mukherjee AK, Sharma S, Sengupta S, Saha D, Kumar P, Hussain T, Srivastava V, Roy SD, Shay JW, Chowdhury S. Telomere length-dependent transcription and epigenetic modifications in promoters remote from telomere ends. PLoS Genet 2018; 14:e1007782. [PMID: 30439955 PMCID: PMC6264879 DOI: 10.1371/journal.pgen.1007782] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 11/29/2018] [Accepted: 10/23/2018] [Indexed: 12/03/2022] Open
Abstract
Telomere-binding proteins constituting the shelterin complex have been studied primarily for telomeric functions. However, mounting evidence shows non-telomeric binding and gene regulation by shelterin factors. This raises a key question—do telomeres impact binding of shelterin proteins at distal non-telomeric sites? Here we show that binding of the telomere-repeat-binding-factor-2 (TRF2) at promoters ~60 Mb from telomeres depends on telomere length in human cells. Promoter TRF2 occupancy was depleted in cells with elongated telomeres resulting in altered TRF2-mediated transcription of distal genes. In addition, histone modifications—activation (H3K4me1 and H3K4me3) as well as silencing marks (H3K27me3)—at distal promoters were telomere length-dependent. These demonstrate that transcription, and the epigenetic state, of telomere-distal promoters can be influenced by telomere length. Molecular links between telomeres and the extra-telomeric genome, emerging from findings here, might have important implications in telomere-related physiology, particularly ageing and cancer. Telomeres (special DNA-protein assemblies that protect chromosome ends) affect ageing and diseases such as cancer. Although this has been recognized for many years, biological processes that connect telomeres to ageing, cancer and other cellular functions remain to be fully understood. Certain proteins, believed to be only telomere-associated, engage DNA outside telomeres. This raises an interesting question. Does telomere length influence how telomere-binding proteins associate with DNA at regions distal from telomeres. If so, how does this impact function? Motivated by these questions, in the present studies we tested if extra-telomeric binding of the well-known telomere-repeat-binding-actor-2 (TRF2) depends on telomere length. Our results show that the level of DNA-bound TRF2 at telomere-distal sites changes as telomeres shorten or elongate. Consequently, TRF2-mediated gene regulation affects many genes. Notably, histone modifications that dictate chromatin compaction and access to regulatory factors, at sites distant from telomere ends also depended on telomere length. Together, this links the state of telomeres to gene regulation and epigenetics directly in ways not previously appreciated that might impact a more complete understanding of molecular processes underlying ageing and cancer.
Collapse
Affiliation(s)
- Ananda Kishore Mukherjee
- Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
- Academy of Scientific and Innovative Research, CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Shalu Sharma
- Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
- Academy of Scientific and Innovative Research, CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Suman Sengupta
- Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Dhurjhoti Saha
- Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Pankaj Kumar
- Academy of Scientific and Innovative Research, CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
- G.N.R. Knowledge Centre for Genome Informatics, CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Tabish Hussain
- Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Vivek Srivastava
- Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Sumitabho Deb Roy
- Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Jerry W. Shay
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Shantanu Chowdhury
- Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
- Academy of Scientific and Innovative Research, CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
- G.N.R. Knowledge Centre for Genome Informatics, CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
- * E-mail:
| |
Collapse
|