1
|
Fernández-Varas B, Manguan-García C, Rodriguez-Centeno J, Mendoza-Lupiáñez L, Calatayud J, Perona R, Martín-Martínez M, Gutierrez-Rodriguez M, Benítez-Buelga C, Sastre L. Clinical mutations in the TERT and TERC genes coding for telomerase components induced oxidative stress, DNA damage at telomeres and cell apoptosis besides decreased telomerase activity. Hum Mol Genet 2024; 33:818-834. [PMID: 38641551 PMCID: PMC11031360 DOI: 10.1093/hmg/ddae015] [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: 10/02/2023] [Revised: 01/16/2024] [Accepted: 01/17/2024] [Indexed: 04/21/2024] Open
Abstract
Telomeres are nucleoprotein structures at the end of chromosomes that maintain their integrity. Mutations in genes coding for proteins involved in telomere protection and elongation produce diseases such as dyskeratosis congenita or idiopathic pulmonary fibrosis known as telomeropathies. These diseases are characterized by premature telomere shortening, increased DNA damage and oxidative stress. Genetic diagnosis of telomeropathy patients has identified mutations in the genes TERT and TERC coding for telomerase components but the functional consequences of many of these mutations still have to be experimentally demonstrated. The activity of twelve TERT and five TERC mutants, five of them identified in Spanish patients, has been analyzed. TERT and TERC mutants were expressed in VA-13 human cells that express low telomerase levels and the activity induced was analyzed. The production of reactive oxygen species, DNA oxidation and TRF2 association at telomeres, DNA damage response and cell apoptosis were determined. Most mutations presented decreased telomerase activity, as compared to wild-type TERT and TERC. In addition, the expression of several TERT and TERC mutants induced oxidative stress, DNA oxidation, DNA damage, decreased recruitment of the shelterin component TRF2 to telomeres and increased apoptosis. These observations might indicate that the increase in DNA damage and oxidative stress observed in cells from telomeropathy patients is dependent on their TERT or TERC mutations. Therefore, analysis of the effect of TERT and TERC mutations of unknown function on DNA damage and oxidative stress could be of great utility to determine the possible pathogenicity of these variants.
Collapse
Affiliation(s)
- Beatriz Fernández-Varas
- Instituto de Investigaciones Biomedicas Sols/Morreale CSIC/UAM, Arturo Duperier 4, 28029 Madrid, Spain
| | - Cristina Manguan-García
- Instituto de Investigaciones Biomedicas Sols/Morreale CSIC/UAM, Arturo Duperier 4, 28029 Madrid, Spain
- Centro de Investigacion Biomedica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III. C. Melchor Fernandez de Almagro, 3, 28029 Madrid, Spain
| | - Javier Rodriguez-Centeno
- Instituto de Investigaciones Biomedicas Sols/Morreale CSIC/UAM, Arturo Duperier 4, 28029 Madrid, Spain
| | - Lucía Mendoza-Lupiáñez
- Instituto de Investigaciones Biomedicas Sols/Morreale CSIC/UAM, Arturo Duperier 4, 28029 Madrid, Spain
| | - Joaquin Calatayud
- Departamento de Biología y Geología, Física y Química inorgánica. ESCET, Universidad Rey Juan Carlos, C/Tulipán s/n, Móstoles, C.P. 28933 Madrid, Spain
| | - Rosario Perona
- Centro de Investigacion Biomedica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III. C. Melchor Fernandez de Almagro, 3, 28029 Madrid, Spain
- Instituto de Salud Carlos III. Calle Monforte de Lemos 5, 28029 Madrid, Spain
| | | | | | - Carlos Benítez-Buelga
- Instituto de Investigaciones Biomedicas Sols/Morreale CSIC/UAM, Arturo Duperier 4, 28029 Madrid, Spain
| | - Leandro Sastre
- Instituto de Investigaciones Biomedicas Sols/Morreale CSIC/UAM, Arturo Duperier 4, 28029 Madrid, Spain
- Centro de Investigacion Biomedica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III. C. Melchor Fernandez de Almagro, 3, 28029 Madrid, Spain
| |
Collapse
|
2
|
Wang Y, Zhang Z, He H, Song J, Cui Y, Chen Y, Zhuang Y, Zhang X, Li M, Zhang X, Zhang MQ, Shi M, Yi C, Wang J. Aging-induced pseudouridine synthase 10 impairs hematopoietic stem cells. Haematologica 2023; 108:2677-2689. [PMID: 37165848 PMCID: PMC10542847 DOI: 10.3324/haematol.2022.282211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 05/04/2023] [Indexed: 05/12/2023] Open
Abstract
Aged hematopoietic stem cells (HSC) exhibit compromised reconstitution capacity and differentiation-bias towards myeloid lineage, however, the molecular mechanism behind it remains not fully understood. In this study, we observed that the expression of pseudouridine (Ψ) synthase 10 is increased in aged hematopoietic stem and progenitor cells (HSPC) and enforced protein of Ψ synthase 10 (PUS10) recapitulates the phenotype of aged HSC, which is not achieved by its Ψ synthase activity. Consistently, we observed no difference of transcribed RNA pseudouridylation profile between young and aged HSPC. No significant alteration of hematopoietic homeostasis and HSC function is observed in young Pus10-/- mice, while aged Pus10-/- mice exhibit mild alteration of hematopoietic homeostasis and HSC function. Moreover, we observed that PUS10 is ubiquitinated by E3 ubiquitin ligase CRL4DCAF1 complex and the increase of PUS10 in aged HSPC is due to aging-declined CRL4DCAF1- mediated ubiquitination degradation signaling. Taken together, this study for the first time evaluated the role of PUS10 in HSC aging and function, and provided a novel insight into HSC rejuvenation and its clinical application.
Collapse
Affiliation(s)
- Yuqian Wang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084
| | | | - Hanqing He
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084
| | - Jinghui Song
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093
| | - Yang Cui
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084
| | - Yunan Chen
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing
| | - Yuan Zhuang
- Department of Basic Medical Sciences, School of Medicine, Institute for Immunology, Beijing Key Lab for Immunological Research on Chronic Diseases, THU-PKU Center for Life Sciences, Tsinghua University, Beijing
| | - Xiaoting Zhang
- Department of Basic Medical Sciences, School of Medicine, Institute for Immunology, Beijing Key Lab for Immunological Research on Chronic Diseases, THU-PKU Center for Life Sciences, Tsinghua University, Beijing
| | - Mo Li
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191
| | - Xinxiang Zhang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing
| | - Michael Q Zhang
- School of Medicine, Tsinghua University, Beijing 100084, China; MOE Key Laboratory of Bioinformatics; Bioinformatics Division and Center for Synthetic and Systems Biology, BNRist; Department of Automation, Tsinghua University, Beijing 100084, China; Department of Biological Sciences, Center for Systems Biology, the University of Texas, Richardson, TX 75080-3021.
| | - Minglei Shi
- School of Medicine, Tsinghua University, Beijing 100084.
| | - Chengqi Yi
- Department of Basic Medical Sciences, School of Medicine, Institute for Immunology, Beijing Key Lab for Immunological Research on Chronic Diseases, THU-PKU Center for Life Sciences, Tsinghua University, Beijing.
| | - Jianwei Wang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084.
| |
Collapse
|
3
|
Jerome MS, Nanjappa DP, Chakraborty A, Chakrabarty S. Molecular etiology of defective nuclear and mitochondrial ribosome biogenesis: Clinical phenotypes and therapy. Biochimie 2023; 207:122-136. [PMID: 36336106 DOI: 10.1016/j.biochi.2022.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 10/27/2022] [Accepted: 11/01/2022] [Indexed: 11/06/2022]
Abstract
Ribosomopathies are rare congenital disorders associated with defective ribosome biogenesis due to pathogenic variations in genes that encode proteins related to ribosome function and biogenesis. Defects in ribosome biogenesis result in a nucleolar stress response involving the TP53 tumor suppressor protein and impaired protein synthesis leading to a deregulated translational output. Despite the accepted notion that ribosomes are omnipresent and essential for all cells, most ribosomopathies show tissue-specific phenotypes affecting blood cells, hair, spleen, or skin. On the other hand, defects in mitochondrial ribosome biogenesis are associated with a range of clinical manifestations affecting more than one organ. Intriguingly, the deregulated ribosomal function is also a feature in several human malignancies with a selective upregulation or downregulation of specific ribosome components. Here, we highlight the clinical conditions associated with defective ribosome biogenesis in the nucleus and mitochondria with a description of the affected genes and the implicated pathways, along with a note on the treatment strategies currently available for these disorders.
Collapse
Affiliation(s)
- Maria Sona Jerome
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Dechamma Pandyanda Nanjappa
- Division of Molecular Genetics and Cancer, Nitte University Centre for Science Education and Research (NUCSER), NITTE (Deemed to Be University), Deralakate, Mangaluru, 575018, India
| | - Anirban Chakraborty
- Division of Molecular Genetics and Cancer, Nitte University Centre for Science Education and Research (NUCSER), NITTE (Deemed to Be University), Deralakate, Mangaluru, 575018, India.
| | - Sanjiban Chakrabarty
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India.
| |
Collapse
|
4
|
Nelson N, Feurstein S, Niaz A, Truong J, Holien JK, Lucas S, Fairfax K, Dickinson J, Bryan TM. Functional genomics for curation of variants in telomere biology disorder associated genes: A systematic review. Genet Med 2023; 25:100354. [PMID: 36496180 DOI: 10.1016/j.gim.2022.11.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 11/28/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
PURPOSE Patients with an underlying telomere biology disorder (TBD) have variable clinical presentations, and they can be challenging to diagnose clinically. A genomic diagnosis for patients presenting with TBD is vital for optimal treatment. Unfortunately, many variants identified during diagnostic testing are variants of uncertain significance. This complicates management decisions, delays treatment, and risks nonuptake of potentially curative therapies. Improved application of functional genomic evidence may reduce variants of uncertain significance classifications. METHODS We systematically searched the literature for published functional assays interrogating TBD gene variants. When possible, established likely benign/benign and likely pathogenic/pathogenic variants were used to estimate the assay sensitivity, specificity, positive predictive value, negative predictive value, and odds of pathogenicity. RESULTS In total, 3131 articles were screened and 151 met inclusion criteria. Sufficient data to enable a PS3/BS3 recommendation were available for TERT variants only. We recommend that PS3 and BS3 can be applied at a moderate and supportive level, respectively. PS3/BS3 application was limited by a lack of assay standardization and limited inclusion of benign variants. CONCLUSION Further assay standardization and assessment of benign variants are required for optimal use of the PS3/BS3 criterion for TBD gene variant classification.
Collapse
Affiliation(s)
- Niles Nelson
- The Menzies Institute for Medical Research, College of Health and Medicine, The University of Tasmania, Hobart, Tasmania, Australia; Department of Molecular Medicine, The Royal Hobart Hospital, Hobart, Tasmania, Australia; Department of Molecular Haematology, The Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.
| | - Simone Feurstein
- Section of Hematology, Oncology, and Rheumatology, Department of Internal Medicine, Heidelberg University Hospital, Heidelberg, Germany
| | - Aram Niaz
- Children's Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, New South Wales, Australia
| | - Jia Truong
- School of Science, STEM College, RMIT University, Bundoora, Victoria, Australia
| | - Jessica K Holien
- School of Science, STEM College, RMIT University, Bundoora, Victoria, Australia
| | - Sionne Lucas
- The Menzies Institute for Medical Research, College of Health and Medicine, The University of Tasmania, Hobart, Tasmania, Australia
| | - Kirsten Fairfax
- The Menzies Institute for Medical Research, College of Health and Medicine, The University of Tasmania, Hobart, Tasmania, Australia
| | - Joanne Dickinson
- The Menzies Institute for Medical Research, College of Health and Medicine, The University of Tasmania, Hobart, Tasmania, Australia
| | - Tracy M Bryan
- Children's Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, New South Wales, Australia
| |
Collapse
|
5
|
Emerging Evidence of the Significance of Thioredoxin-1 in Hematopoietic Stem Cell Aging. Antioxidants (Basel) 2022; 11:antiox11071291. [PMID: 35883782 PMCID: PMC9312246 DOI: 10.3390/antiox11071291] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/22/2022] [Accepted: 06/28/2022] [Indexed: 02/04/2023] Open
Abstract
The United States is undergoing a demographic shift towards an older population with profound economic, social, and healthcare implications. The number of Americans aged 65 and older will reach 80 million by 2040. The shift will be even more dramatic in the extremes of age, with a projected 400% increase in the population over 85 years old in the next two decades. Understanding the molecular and cellular mechanisms of ageing is crucial to reduce ageing-associated disease and to improve the quality of life for the elderly. In this review, we summarized the changes associated with the ageing of hematopoietic stem cells (HSCs) and what is known about some of the key underlying cellular and molecular pathways. We focus here on the effects of reactive oxygen species and the thioredoxin redox homeostasis system on ageing biology in HSCs and the HSC microenvironment. We present additional data from our lab demonstrating the key role of thioredoxin-1 in regulating HSC ageing.
Collapse
|
6
|
Gueiderikh A, Maczkowiak-Chartois F, Rosselli F. A new frontier in Fanconi anemia: From DNA repair to ribosome biogenesis. Blood Rev 2021; 52:100904. [PMID: 34750031 DOI: 10.1016/j.blre.2021.100904] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 10/14/2021] [Accepted: 10/26/2021] [Indexed: 12/27/2022]
Abstract
Described by Guido Fanconi almost 100 years ago, Fanconi anemia (FA) is a rare genetic disease characterized by developmental abnormalities, bone marrow failure (BMF) and cancer predisposition. The proteins encoded by FA-mutated genes (FANC proteins) and assembled in the so-called FANC/BRCA pathway have key functions in DNA repair and replication safeguarding, which loss leads to chromosome structural aberrancies. Therefore, since the 1980s, FA has been considered a genomic instability and chromosome fragility syndrome. However, recent findings have demonstrated new and unexpected roles of FANC proteins in nucleolar homeostasis and ribosome biogenesis, the alteration of which impacts cellular proteostasis. Here, we review the different cellular, biochemical and molecular anomalies associated with the loss of function of FANC proteins and discuss how these anomalies contribute to BMF by comparing FA to other major inherited BMF syndromes. Our aim is to determine the extent to which alterations in the DNA damage response in FA contribute to BMF compared to the consequences of the loss of function of the FANC/BRCA pathway on the other roles of the pathway.
Collapse
Affiliation(s)
- Anna Gueiderikh
- CNRS - UMR9019, Équipe labellisée "La Ligue contre le Cancer", 94805 Villejuif, France; Gustave Roussy Cancer Center, 94805 Villejuif, France; Université Paris-Saclay - Paris Sud, Orsay, France.
| | - Frédérique Maczkowiak-Chartois
- CNRS - UMR9019, Équipe labellisée "La Ligue contre le Cancer", 94805 Villejuif, France; Gustave Roussy Cancer Center, 94805 Villejuif, France; Université Paris-Saclay - Paris Sud, Orsay, France.
| | - Filippo Rosselli
- CNRS - UMR9019, Équipe labellisée "La Ligue contre le Cancer", 94805 Villejuif, France; Gustave Roussy Cancer Center, 94805 Villejuif, France; Université Paris-Saclay - Paris Sud, Orsay, France.
| |
Collapse
|
7
|
Vieri M, Brümmendorf TH, Beier F. Treatment of telomeropathies. Best Pract Res Clin Haematol 2021; 34:101282. [PMID: 34404536 DOI: 10.1016/j.beha.2021.101282] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/22/2021] [Accepted: 06/25/2021] [Indexed: 10/21/2022]
Abstract
Telomeropathies or telomere biology disorders (TBDs) are a group of rare diseases characterised by altered telomere maintenance. Most patients with TBDs show pathogenic variants of genes that encode factors involved in the prevention of telomere shortening. Particularly in adults, TBDs mostly present themselves with heterogeneous clinical features that often include bone marrow failure, hepatopathies, interstitial lung disease and other organ sites. Different degrees of severity are also observed among patients with TBDs, ranging from very severe syndromes manifesting themselves in early childhood, such as Revesz syndrome, Hoyeraal-Hreidarsson syndrome, and Coats plus disease, to dyskeratosis congenita (DKC) and adult-onset "cryptic" forms of TBD, which often affect fewer organ systems. Overall, the most relevant clinical complications of TBD are bone marrow failure, lung fibrosis, and liver cirrhosis. In this review, we summarise recent advances in the management and treatment of TBD and provide a brief overview of the various treatment approaches.
Collapse
Affiliation(s)
- Margherita Vieri
- Department of Hematology, Oncology, Hemostaseology, Stem Cell Transplantation, Medical Faculty, RWTH Aachen University, Aachen, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Germany.
| | - Tim H Brümmendorf
- Department of Hematology, Oncology, Hemostaseology, Stem Cell Transplantation, Medical Faculty, RWTH Aachen University, Aachen, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Germany.
| | - Fabian Beier
- Department of Hematology, Oncology, Hemostaseology, Stem Cell Transplantation, Medical Faculty, RWTH Aachen University, Aachen, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Germany.
| |
Collapse
|
8
|
Shan M, Ji X, Janssen K, Silverman IM, Humenik J, Garcia BA, Liebhaber SA, Gregory BD. Dynamic changes in RNA-protein interactions and RNA secondary structure in mammalian erythropoiesis. Life Sci Alliance 2021; 4:4/9/e202000659. [PMID: 34315813 PMCID: PMC8321672 DOI: 10.26508/lsa.202000659] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 07/06/2021] [Accepted: 07/07/2021] [Indexed: 11/24/2022] Open
Abstract
Two features of eukaryotic RNA molecules that regulate their post-transcriptional fates are RNA secondary structure and RNA-binding protein (RBP) interaction sites. However, a comprehensive global overview of the dynamic nature of these sequence features during erythropoiesis has never been obtained. Here, we use our ribonuclease-mediated structure and RBP-binding site mapping approach to reveal the global landscape of RNA secondary structure and RBP-RNA interaction sites and the dynamics of these features during this important developmental process. We identify dynamic patterns of RNA secondary structure and RBP binding throughout the process and determine a set of corresponding protein-bound sequence motifs along with their dynamic structural and RBP-binding contexts. Finally, using these dynamically bound sequences, we identify a number of RBPs that have known and putative key functions in post-transcriptional regulation during mammalian erythropoiesis. In total, this global analysis reveals new post-transcriptional regulators of mammalian blood cell development.
Collapse
Affiliation(s)
- Mengge Shan
- Department of Biology, University of Pennsylvania, Philadelphia, PA, USA.,Genomics and Computational Biology Graduate Group, Perelman School of Medicine, Epigenetics Institute, University of Pennsylvania, PA, USA
| | - Xinjun Ji
- Department of Genetics, Perelman School of Medicine, Epigenetics Institute, University of Pennsylvania, PA, USA
| | - Kevin Janssen
- Department of Biochemistry and Biophysics, Perelman School of Medicine, Epigenetics Institute, University of Pennsylvania, PA, USA
| | - Ian M Silverman
- Department of Genetics, Perelman School of Medicine, Epigenetics Institute, University of Pennsylvania, PA, USA
| | - Jesse Humenik
- Department of Genetics, Perelman School of Medicine, Epigenetics Institute, University of Pennsylvania, PA, USA
| | - Ben A Garcia
- Department of Biochemistry and Biophysics, Perelman School of Medicine, Epigenetics Institute, University of Pennsylvania, PA, USA
| | - Stephen A Liebhaber
- Department of Genetics, Perelman School of Medicine, Epigenetics Institute, University of Pennsylvania, PA, USA .,Department of Medicine, Perelman School of Medicine, Epigenetics Institute, University of Pennsylvania, PA, USA
| | - Brian D Gregory
- Department of Biology, University of Pennsylvania, Philadelphia, PA, USA .,Genomics and Computational Biology Graduate Group, Perelman School of Medicine, Epigenetics Institute, University of Pennsylvania, PA, USA
| |
Collapse
|
9
|
McMahon M, Forester C, Buffenstein R. Aging through an epitranscriptomic lens. NATURE AGING 2021; 1:335-346. [PMID: 37117595 DOI: 10.1038/s43587-021-00058-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 03/08/2021] [Indexed: 04/30/2023]
Abstract
The mechanistic causes of aging, the time-related decline in function and good health that leads to increased mortality, remain poorly understood. Here we propose that age-dependent alteration of the epitranscriptome, encompassing more than 150 chemically distinct post-transcriptional modifications or editing events, warrants exploration as an important modulator of aging. The epitranscriptome is a potent regulator of RNA function, diverse cellular processes and tissue regenerative capacity. To date, only a few studies link alterations in the epitranscriptome to molecular and physiological changes during aging; however, epitranscriptome dysfunction is associated with and underlies several age-associated pathologies, including cancer and neurodegenerative, cardiovascular and autoimmune diseases. For example, changes in RNA modifications (such as N6-methyladenosine and inosine) impact cardiac physiology and are linked to cardiac fibrosis. Although an uncharted research focus, mapping epitranscriptome alterations in the context of aging may elucidate novel predictors of both health and lifespan, and may identify therapeutic targets for attenuating aging and abrogating age-related diseases.
Collapse
Affiliation(s)
- Mary McMahon
- Calico Life Sciences LLC, South San Francisco, CA, USA.
| | - Craig Forester
- Department of Pediatrics, University of Colorado, Denver, CO, USA
- Children's Hospital Colorado, Division of Pediatric Hematology/Oncology/Bone Marrow Transplant, Colorado, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | |
Collapse
|
10
|
Telomere length shortening in hospitalized preterm infants: A pilot study. PLoS One 2021; 16:e0243468. [PMID: 33471805 PMCID: PMC7817026 DOI: 10.1371/journal.pone.0243468] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 11/21/2020] [Indexed: 12/18/2022] Open
Abstract
Leukocyte telomere length is a biomarker of aging-related health risks. Hospitalized preterm infants frequently experience elevated oxidative stress and inflammation, both of which contribute to telomere shortening. Our aim was to examine changes in telomere length during neonatal intensive care unit (NICU) hospitalization in a cohort of preterm infants <32 weeks' gestation. We conducted a longitudinal study of 10 infants (mean gestational age 27 weeks, range 23.5 to 29, at birth). We isolated DNA from dried blood spots and used Real Time Quantitative PCR to measure relative leukocyte telomere length in triplicate at three time points for each participant. From birth to discharge, infants experienced an average decline in relative telomere length of 0.021 units per week (95% CI -0.040, -0.0020; p = 0.03), after adjustment for gestational age at birth. Our results suggest a measurable decline in telomere length during NICU hospitalization. We speculate that telomere length change may convey information about NICU exposures that carry short- and long-term health risks.
Collapse
|
11
|
AlSabbagh MM. Dyskeratosis congenita: ein Literaturüberblick. J Dtsch Dermatol Ges 2020; 18:943-968. [PMID: 32985809 DOI: 10.1111/ddg.14268_g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 01/29/2020] [Indexed: 02/06/2023]
|
12
|
AlSabbagh MM. Dyskeratosis congenita: a literature review. J Dtsch Dermatol Ges 2020; 18:943-967. [PMID: 32930426 DOI: 10.1111/ddg.14268] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 01/29/2020] [Indexed: 12/26/2022]
Abstract
Dyskeratosis congenita is a rare hereditary disease that occurs predominantly in males and manifests clinically as the classic triad of reticulate hyperpigmentation, nail dystrophy and leukoplakia. It increases the risk of malignancy and other potentially lethal complications such as bone marrow failure, lung and liver diseases. Mutations in 19 genes are associated with dyskeratosis congenita, and a fifth of the pathogenic mutations are found in DKC1, the gene coding for dyskerin. This review aims to address the clinical and genetic aspects of the disease.
Collapse
|
13
|
Breitbach ME, Greenspan S, Resnick NM, Perera S, Gurkar AU, Absher D, Levine AS. Exonic Variants in Aging-Related Genes Are Predictive of Phenotypic Aging Status. Front Genet 2019; 10:1277. [PMID: 31921313 PMCID: PMC6931058 DOI: 10.3389/fgene.2019.01277] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 11/19/2019] [Indexed: 01/31/2023] Open
Abstract
Background: Recent studies investigating longevity have revealed very few convincing genetic associations with increased lifespan. This is, in part, due to the complexity of biological aging, as well as the limited power of genome-wide association studies, which assay common single nucleotide polymorphisms (SNPs) and require several thousand subjects to achieve statistical significance. To overcome such barriers, we performed comprehensive DNA sequencing of a panel of 20 genes previously associated with phenotypic aging in a cohort of 200 individuals, half of whom were clinically defined by an "early aging" phenotype, and half of whom were clinically defined by a "late aging" phenotype based on age (65-75 years) and the ability to walk up a flight of stairs or walk for 15 min without resting. A validation cohort of 511 late agers was used to verify our results. Results: We found early agers were not enriched for more total variants in these 20 aging-related genes than late agers. Using machine learning methods, we identified the most predictive model of aging status, both in our discovery and validation cohorts, to be a random forest model incorporating damaging exon variants [Combined Annotation-Dependent Depletion (CADD) > 15]. The most heavily weighted variants in the model were within poly(ADP-ribose) polymerase 1 (PARP1) and excision repair cross complementation group 5 (ERCC5), both of which are involved in a canonical aging pathway, DNA damage repair. Conclusion: Overall, this study implemented a framework to apply machine learning to identify sequencing variants associated with complex phenotypes such as aging. While the small sample size making up our cohort inhibits our ability to make definitive conclusions about the ability of these genes to accurately predict aging, this study offers a unique method for exploring polygenic associations with complex phenotypes.
Collapse
Affiliation(s)
- Megan E. Breitbach
- HudsonAlpha Institute for Biotechnology, Hunstville, AL, United States
- Department of Biotechnology Science and Engineering, University of Alabama in Huntsville, Hunstville, AL, United States
| | - Susan Greenspan
- Division of Geriatric Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Neil M. Resnick
- Division of Geriatric Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Institute on Aging of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Subashan Perera
- Division of Geriatric Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Biostatistics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, United States
| | - Aditi U. Gurkar
- Division of Geriatric Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Institute on Aging of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Devin Absher
- HudsonAlpha Institute for Biotechnology, Hunstville, AL, United States
| | - Arthur S. Levine
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- UPMC Hillman Cancer Center, Pittsburgh, PA, United States
| |
Collapse
|
14
|
Belli V, Matrone N, Sagliocchi S, Incarnato R, Conte A, Pizzo E, Turano M, Angrisani A, Furia M. A dynamic link between H/ACA snoRNP components and cytoplasmic stress granules. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:118529. [DOI: 10.1016/j.bbamcr.2019.118529] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 07/08/2019] [Accepted: 08/05/2019] [Indexed: 02/06/2023]
|
15
|
Dyskerin Mutations Present in Dyskeratosis Congenita Patients Increase Oxidative Stress and DNA Damage Signalling in Dictyostelium Discoideum. Cells 2019; 8:cells8111406. [PMID: 31717312 PMCID: PMC6912284 DOI: 10.3390/cells8111406] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 11/04/2019] [Accepted: 11/05/2019] [Indexed: 12/17/2022] Open
Abstract
Dyskerin is a protein involved in the formation of small nucleolar and small Cajal body ribonucleoproteins. These complexes participate in RNA pseudouridylation and are also components of the telomerase complex required for telomere elongation. Dyskerin mutations cause a rare disease, X-linked dyskeratosis congenita, with no curative treatment. The social amoeba Dictyostelium discoideum contains a gene coding for a dyskerin homologous protein. In this article D. discoideum mutant strains that have mutations corresponding to mutations found in dyskeratosis congenita patients are described. The phenotype of the mutant strains has been studied and no alterations were observed in pseudouridylation activity and telomere structure. Mutant strains showed increased proliferation on liquid culture but reduced growth feeding on bacteria. The results obtained indicated the existence of increased DNA damage response and reactive oxygen species, as also reported in human Dyskeratosis congenita cells and some other disease models. These data, together with the haploid character of D. discoideum vegetative cells, that resemble the genomic structure of the human dyskerin gene, located in the X chromosome, support the conclusion that D. discoideum can be a good model system for the study of this disease.
Collapse
|
16
|
Akieda Y, Ogamino S, Furuie H, Ishitani S, Akiyoshi R, Nogami J, Masuda T, Shimizu N, Ohkawa Y, Ishitani T. Cell competition corrects noisy Wnt morphogen gradients to achieve robust patterning in the zebrafish embryo. Nat Commun 2019; 10:4710. [PMID: 31624259 PMCID: PMC6797755 DOI: 10.1038/s41467-019-12609-4] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 09/18/2019] [Indexed: 12/31/2022] Open
Abstract
Morphogen signalling forms an activity gradient and instructs cell identities in a signalling strength-dependent manner to pattern developing tissues. However, developing tissues also undergo dynamic morphogenesis, which may produce cells with unfit morphogen signalling and consequent noisy morphogen gradients. Here we show that a cell competition-related system corrects such noisy morphogen gradients. Zebrafish imaging analyses of the Wnt/β-catenin signalling gradient, which acts as a morphogen to establish embryonic anterior-posterior patterning, identify that unfit cells with abnormal Wnt/β-catenin activity spontaneously appear and produce noise in the gradient. Communication between unfit and neighbouring fit cells via cadherin proteins stimulates apoptosis of the unfit cells by activating Smad signalling and reactive oxygen species production. This unfit cell elimination is required for proper Wnt/β-catenin gradient formation and consequent anterior-posterior patterning. Because this gradient controls patterning not only in the embryo but also in adult tissues, this system may support tissue robustness and disease prevention. Gradients of morphogens such as Wnt provide instructive cues for cell identities during development. Here, the authors report that in the developing zebrafish embryo, cell competition and elimination of unfit cells are required for proper Wnt gradient formation.
Collapse
Affiliation(s)
- Yuki Akieda
- Laboratory of Integrated Signaling Systems, Department of Molecular Medicine, Institute for Molecular & Cellular Regulation, Gunma University, Gunma, 371-8512, Japan.,Department of Homeostatic Regulation, Division of Cellular and Molecular Biology, Research Institute for Microbial Diseases, Osaka University, Osaka, 565-0871, Japan
| | - Shohei Ogamino
- Laboratory of Integrated Signaling Systems, Department of Molecular Medicine, Institute for Molecular & Cellular Regulation, Gunma University, Gunma, 371-8512, Japan
| | - Hironobu Furuie
- Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582, Japan.,Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Shizuka Ishitani
- Laboratory of Integrated Signaling Systems, Department of Molecular Medicine, Institute for Molecular & Cellular Regulation, Gunma University, Gunma, 371-8512, Japan
| | - Ryutaro Akiyoshi
- Biological Evaluation Technology 2, Research and Development, Olympus Corp., Tokyo, 192-8512, Japan
| | - Jumpei Nogami
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582, Japan
| | - Takamasa Masuda
- Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582, Japan
| | - Nobuyuki Shimizu
- Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582, Japan
| | - Yasuyuki Ohkawa
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582, Japan
| | - Tohru Ishitani
- Laboratory of Integrated Signaling Systems, Department of Molecular Medicine, Institute for Molecular & Cellular Regulation, Gunma University, Gunma, 371-8512, Japan. .,Department of Homeostatic Regulation, Division of Cellular and Molecular Biology, Research Institute for Microbial Diseases, Osaka University, Osaka, 565-0871, Japan. .,Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582, Japan.
| |
Collapse
|
17
|
Herrero D, Tomé M, Cañón S, Cruz FM, Carmona RM, Fuster E, Roche E, Bernad A. Redox-dependent BMI1 activity drives in vivo adult cardiac progenitor cell differentiation. Cell Death Differ 2018; 25:809-822. [PMID: 29323265 DOI: 10.1038/s41418-017-0022-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 10/20/2017] [Accepted: 10/23/2017] [Indexed: 01/04/2023] Open
Abstract
Accumulation of reactive oxygen species (ROS) is associated with several cardiovascular pathologies and with cell cycle exit by neonanatal cardiomyocytes, a key limiting factor in the regenerative capacity of the adult mammalian heart. The polycomb complex component BMI1 is linked to adult progenitors and is an important partner in DNA repair and redox regulation. We found that high BMI1 expression is associated with an adult Sca1+ cardiac progenitor sub-population with low ROS levels. In homeostasis, BMI1 repressed cell fate genes, including a cardiogenic differentiation program. Oxidative damage nonetheless modified BMI1 activity in vivo by derepressing canonical target genes in favor of their antioxidant and anticlastogenic functions. This redox-mediated mechanism is not restricted to damage situations, however, and we report ROS-associated differentiation of cardiac progenitors in steady state. These findings demonstrate how redox status influences the cardiac progenitor response, and identify redox-mediated BMI1 regulation with implications in maintenance of cellular identity in vivo.
Collapse
Affiliation(s)
- Diego Herrero
- Department of Immunology and Oncology, Spanish National Center for Biotechnology (CNB-CSIC), Madrid, Spain
| | - María Tomé
- Cardiovascular Development and Repair Department, Spanish National Cardiovascular Research Center (CNIC), Madrid, Spain
| | - Susana Cañón
- Department of Immunology and Oncology, Spanish National Center for Biotechnology (CNB-CSIC), Madrid, Spain.,Cardiovascular Development and Repair Department, Spanish National Cardiovascular Research Center (CNIC), Madrid, Spain
| | - Francisco M Cruz
- Cardiovascular Development and Repair Department, Spanish National Cardiovascular Research Center (CNIC), Madrid, Spain
| | - Rosa María Carmona
- Department of Immunology and Oncology, Spanish National Center for Biotechnology (CNB-CSIC), Madrid, Spain
| | - Encarna Fuster
- Department of Applied Biology-Nutrition and Institute of Bioengineering, University Miguel Hernández, Institute for Health and Biomedical Research (ISABIAL-FISABIO Fundation), Alicante, Spain
| | - Enrique Roche
- CIBERobn (Physiopathology of Obesity and Nutrition CB12/03/30038), Carlos III Health Research Institute (ISCIII), Madrid, Spain.,Department of Applied Biology-Nutrition and Institute of Bioengineering, University Miguel Hernández, Institute for Health and Biomedical Research (ISABIAL-FISABIO Fundation), Alicante, Spain
| | - Antonio Bernad
- Department of Immunology and Oncology, Spanish National Center for Biotechnology (CNB-CSIC), Madrid, Spain. .,Cardiovascular Development and Repair Department, Spanish National Cardiovascular Research Center (CNIC), Madrid, Spain.
| |
Collapse
|
18
|
Ibáñez-Cabellos JS, Pérez-Machado G, Seco-Cervera M, Berenguer-Pascual E, García-Giménez JL, Pallardó FV. Acute telomerase components depletion triggers oxidative stress as an early event previous to telomeric shortening. Redox Biol 2017; 14:398-408. [PMID: 29055871 PMCID: PMC5650655 DOI: 10.1016/j.redox.2017.10.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 10/04/2017] [Accepted: 10/05/2017] [Indexed: 01/13/2023] Open
Abstract
Loss of function of dyskerin (DKC1), NOP10 and TIN2 are responsible for different inheritance patterns of Dyskeratosis congenita (DC; ORPHA1775). They are key components of telomerase (DKC1 and NOP10) and shelterin (TIN2), and play an important role in telomere homeostasis. They participate in several fundamental cellular processes by contributing to Dyskeratosis congenita through mechanisms that are not fully understood. Presence of oxidative stress was postulated to result from telomerase ablation. However, the resulting disturbed redox status can promote telomere attrition by generating a vicious circle, which promotes cellular senescence. This fact prompted us to study if acute loss of DKC1, NOP10 and TINF2 can promote redox disequilibrium as an early event when telomere shortening has not yet taken place. We generated siRNA-mediated (DKC1, NOP10 and TINF2) cell lines by RNA interference, which was confirmed by mRNA and protein expression analyses. No telomere shortening occurred in any silenced cell line. Depletion of H/ACA ribonucleoproteins DKC1 and NOP10 diminished telomerase activity via TERC down-regulation, and produced alterations in pseudouridylation and ribosomal biogenesis. An increase in the GSSG/GSH ratio, carbonylated proteins and oxidized peroxiredoxin-6 was observed, in addition to MnSOD and TRX1 overexpression in the siRNA DC cells. Likewise, high PARylation levels and high PARP1 protein expression were detected. In contrast, the silenced TINF2 cells did not alter any evaluated oxidative stress marker. Altogether these findings lead us to conclude that loss of DKC1 and NOP10 functions induces oxidative stress in a telomere shortening independent manner. Transient silencing of DKC1 and NOP10 genes produce oxidative stress. Cells depleted of DKC1 and NOP10 are susceptible to DNA damage. Acute DKC1 and NOP10 depletion disrupts RNA maturation. Oxidative stress is an early event previous to telomere shortening.
Collapse
Affiliation(s)
- José Santiago Ibáñez-Cabellos
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Institute of Health Carlos III, Valencia, Spain; Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, Valencia, Spain; INCLIVA Biomedical Research Institute, Valencia, Spain.
| | - Giselle Pérez-Machado
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, Valencia, Spain; INCLIVA Biomedical Research Institute, Valencia, Spain.
| | - Marta Seco-Cervera
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Institute of Health Carlos III, Valencia, Spain; Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, Valencia, Spain; INCLIVA Biomedical Research Institute, Valencia, Spain.
| | - Ester Berenguer-Pascual
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, Valencia, Spain.
| | - José Luis García-Giménez
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Institute of Health Carlos III, Valencia, Spain; Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, Valencia, Spain; INCLIVA Biomedical Research Institute, Valencia, Spain.
| | - Federico V Pallardó
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Institute of Health Carlos III, Valencia, Spain; Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, Valencia, Spain; INCLIVA Biomedical Research Institute, Valencia, Spain.
| |
Collapse
|
19
|
Biechonski S, Yassin M, Milyavsky M. DNA-damage response in hematopoietic stem cells: an evolutionary trade-off between blood regeneration and leukemia suppression. Carcinogenesis 2017; 38:367-377. [PMID: 28334174 DOI: 10.1093/carcin/bgx002] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 01/11/2017] [Indexed: 12/12/2022] Open
Abstract
Self-renewing and multipotent hematopoietic stem cells (HSCs) maintain lifelong hematopoiesis. Their enormous regenerative potential coupled with lifetime persistence in the body, in contrast with the Progenitors, demand tight control of HSCs genome stability. Indeed, failure to accurately repair DNA damage in HSCs is associated with bone marrow failure and accelerated leukemogenesis. Recent observations exposed remarkable differences in several DNA-damage response (DDR) aspects between HSCs and Progenitors, especially in their DNA-repair capacities and susceptibility to apoptosis. Human HSCs in comparison with Progenitors exhibit delayed DNA double-strand break rejoining, persistent DDR signaling activation, higher sensitivity to the cytotoxic effects of ionizing radiation and attenuated expression of DNA-repair genes. Importantly, the distinct DDR of HSCs was also documented in mouse models. Nevertheless, physiological significance and the molecular basis of the HSCs-specific DDR features are only partially understood. Taking radiation-induced DDR as a paradigm, this review will focus on the current advances in understanding the role of cell-intrinsic DDR regulators and the cellular microenvironment in balancing stemness with genome stability. Pre-leukemia HSCs and clonal hematopoiesis evolvement will be discussed as an evolutionary compromise between the need for lifelong blood regeneration and DDR. Uniquely for this review, we outline the differences in HSCs-related DDR as highlighted by various experimental systems and attempt to provide their critical analysis.
Collapse
Affiliation(s)
- Shahar Biechonski
- Department of Pathology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Muhammad Yassin
- Department of Pathology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Michael Milyavsky
- Department of Pathology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel
| |
Collapse
|
20
|
Abstract
Cells adapt to their environment by linking external stimuli to an intricate network of transcriptional, post-transcriptional and translational processes. Among these, mechanisms that couple environmental cues to the regulation of protein translation are not well understood. Chemical modifications of RNA allow rapid cellular responses to external stimuli by modulating a wide range of fundamental biochemical properties and processes, including the stability, splicing and translation of messenger RNA. In this Review, we focus on the occurrence of N6-methyladenosine (m6A), 5-methylcytosine (m5C) and pseudouridine (Ψ) in RNA, and describe how these RNA modifications are implicated in regulating pluripotency, stem cell self-renewal and fate specification. Both post-transcriptional modifications and the enzymes that catalyse them modulate stem cell differentiation pathways and are essential for normal development.
Collapse
Affiliation(s)
- Michaela Frye
- Wellcome Trust - Medical Research Council Cambridge Stem Cell Institute, Department of Genetics, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Sandra Blanco
- Wellcome Trust - Medical Research Council Cambridge Stem Cell Institute, Department of Genetics, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
- CIC bioGUNE, Bizkaia Technology Park, Building 801A, Derio, Bizkaia 48160, Spain
| |
Collapse
|
21
|
Pereboeva L, Hubbard M, Goldman FD, Westin ER. Robust DNA Damage Response and Elevated Reactive Oxygen Species in TINF2-Mutated Dyskeratosis Congenita Cells. PLoS One 2016; 11:e0148793. [PMID: 26859482 PMCID: PMC4747510 DOI: 10.1371/journal.pone.0148793] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 01/22/2016] [Indexed: 12/13/2022] Open
Abstract
Dyskeratosis Congenita (DC) is an inherited multisystem premature aging disorder with characteristic skin and mucosal findings as well as a predisposition to cancer and bone marrow failure. DC arises due to gene mutations associated with the telomerase complex or telomere maintenance, resulting in critically shortened telomeres. The pathogenesis of DC, as well as several congenital bone marrow failure (BMF) syndromes, converges on the DNA damage response (DDR) pathway and subsequent elevation of reactive oxygen species (ROS). Historically, DC patients have had poor outcomes following bone marrow transplantation (BMT), perhaps as a consequence of an underlying DNA hypersensitivity to cytotoxic agents. Previously, we demonstrated an activated DDR and increased ROS, augmented by chemotherapy and radiation, in somatic cells isolated from DC patients with a mutation in the RNA component of telomerase, TERC. The current study was undertaken to determine whether previous findings related to ROS and DDR in TERC patients’ cells could be extended to other DC mutations. Of particular interest was whether an antioxidant approach could counter increased ROS and decrease DC pathologies. To test this, we examined lymphocytes from DC patients from different DC mutations (TERT, TINF2, and TERC) for the presence of an active DDR and increased ROS. All DC mutations led to increased steady-state p53 (2-fold to 10-fold) and ROS (1.5-fold to 2-fold). Upon exposure to ionizing radiation (XRT), DC cells increased in both DDR and ROS to a significant degree. Exposing DC cells to hydrogen peroxide also revealed that DC cells maintain a significant oxidant burden compared to controls (1.5-fold to 3-fold). DC cell culture supplemented with N-acetylcysteine, or alternatively grown in low oxygen, afforded significant proliferative benefits (proliferation: maximum 2-fold increase; NAC: 5-fold p53 decrease; low oxygen: maximum 3.5-fold p53 decrease). Together, our data supports a mechanism whereby telomerase deficiency and subsequent shortened telomeres initiate a DDR and create a pro-oxidant environment, especially in cells carrying the TINF2 mutations. Finally, the ameliorative effects of antioxidants in vitro suggest this could translate to therapeutic benefits in DC patients.
Collapse
Affiliation(s)
- Larisa Pereboeva
- Department of Medicine, Division of Hematology Oncology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Meredith Hubbard
- Department of Pediatrics, Division of Hematology Oncology, University of Alabama, Birmingham, Alabama, United States of America
| | - Frederick D. Goldman
- Department of Pediatrics, Division of Hematology Oncology, University of Alabama, Birmingham, Alabama, United States of America
| | - Erik R. Westin
- Department of Pediatrics, Division of Hematology Oncology, University of Alabama, Birmingham, Alabama, United States of America
- * E-mail:
| |
Collapse
|
22
|
GSE4, a Small Dyskerin- and GSE24.2-Related Peptide, Induces Telomerase Activity, Cell Proliferation and Reduces DNA Damage, Oxidative Stress and Cell Senescence in Dyskerin Mutant Cells. PLoS One 2015; 10:e0142980. [PMID: 26571381 PMCID: PMC4646510 DOI: 10.1371/journal.pone.0142980] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 10/29/2015] [Indexed: 02/04/2023] Open
Abstract
Dyskeratosis congenita is an inherited disease caused by mutations in genes coding for telomeric components. It was previously reported that expression of a dyskerin-derived peptide, GSE24.2, increases telomerase activity, regulates gene expression and decreases DNA damage and oxidative stress in dyskeratosis congenita patient cells. The biological activity of short peptides derived from GSE24.2 was tested and one of them, GSE4, that probed to be active, was further characterized in this article. Expression of this eleven amino acids long peptide increased telomerase activity and reduced DNA damage, oxidative stress and cell senescence in dyskerin-mutated cells. GSE4 expression also activated c-myc and TERT promoters and increase of c-myc, TERT and TERC expression. The level of biological activity of GSE4 was similar to that obtained by GSE24.2 expression. Incorporation of a dyskerin nuclear localization signal to GSE24.2 did not change its activity on promoter regulation and DNA damage protection. However, incorporation of a signal that increases the rate of nucleolar localization impaired GSE24.2 activity. Incorporation of the dyskerin nuclear localization signal to GSE4 did not alter its biological activity. Mutation of the Aspartic Acid residue that is conserved in the pseudouridine synthase domain present in GSE4 did not impair its activity, except for the repression of c-myc promoter activity and the decrease of c-myc, TERT and TERC gene expression in dyskerin-mutated cells. These results indicated that GSE4 could be of great therapeutic interest for treatment of dyskeratosis congenita patients.
Collapse
|
23
|
Reeves DA, Gu BW, Bessler M, Mason PJ. Variations in reactive oxygen species between mouse strains. Blood Cells Mol Dis 2015; 55:189-90. [PMID: 26227841 DOI: 10.1016/j.bcmd.2015.06.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 06/20/2015] [Indexed: 12/11/2022]
Affiliation(s)
- D A Reeves
- Comprehensive Bone Marrow Failure Center, Division of Hematology, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, 3615 Civic Center Blvd., Philadelphia, PA 19104, USA.
| | - B W Gu
- Comprehensive Bone Marrow Failure Center, Division of Hematology, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, 3615 Civic Center Blvd., Philadelphia, PA 19104, USA
| | - M Bessler
- Comprehensive Bone Marrow Failure Center, Division of Hematology, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, 3615 Civic Center Blvd., Philadelphia, PA 19104, USA; Department of Internal Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - P J Mason
- Comprehensive Bone Marrow Failure Center, Division of Hematology, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, 3615 Civic Center Blvd., Philadelphia, PA 19104, USA.
| |
Collapse
|
24
|
Manguan-Garcia C, Pintado-Berninches L, Carrillo J, Machado-Pinilla R, Sastre L, Pérez-Quilis C, Esmoris I, Gimeno A, García-Giménez JL, Pallardó FV, Perona R. Expression of the genetic suppressor element 24.2 (GSE24.2) decreases DNA damage and oxidative stress in X-linked dyskeratosis congenita cells. PLoS One 2014; 9:e101424. [PMID: 24987982 PMCID: PMC4079255 DOI: 10.1371/journal.pone.0101424] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 06/06/2014] [Indexed: 01/10/2023] Open
Abstract
The predominant X-linked form of Dyskeratosis congenita results from mutations in DKC1, which encodes dyskerin, a protein required for ribosomal RNA modification that is also a component of the telomerase complex. We have previously found that expression of an internal fragment of dyskerin (GSE24.2) rescues telomerase activity in X-linked dyskeratosis congenita (X-DC) patient cells. Here we have found that an increased basal and induced DNA damage response occurred in X-DC cells in comparison with normal cells. DNA damage that is also localized in telomeres results in increased heterochromatin formation and senescence. Expression of a cDNA coding for GSE24.2 rescues both global and telomeric DNA damage. Furthermore, transfection of bacterial purified or a chemically synthesized GSE24.2 peptide is able to rescue basal DNA damage in X-DC cells. We have also observed an increase in oxidative stress in X-DC cells and expression of GSE24.2 was able to diminish it. Altogether our data indicated that supplying GSE24.2, either from a cDNA vector or as a peptide reduces the pathogenic effects of Dkc1 mutations and suggests a novel therapeutic approach.
Collapse
Affiliation(s)
- Cristina Manguan-Garcia
- Instituto de Investigaciones Biomédicas CSIC/UAM, Madrid, Spain
- CIBER de Enfermedades Raras, Valencia, Spain
| | | | - Jaime Carrillo
- Instituto de Investigaciones Biomédicas CSIC/UAM, Madrid, Spain
| | - Rosario Machado-Pinilla
- Instituto de Investigaciones Biomédicas CSIC/UAM, Madrid, Spain
- CIBER de Enfermedades Raras, Valencia, Spain
| | - Leandro Sastre
- Instituto de Investigaciones Biomédicas CSIC/UAM, Madrid, Spain
- CIBER de Enfermedades Raras, Valencia, Spain
| | - Carme Pérez-Quilis
- Biomedical Research Institute INCLIVA, Valencia, Spain
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, Valencia, Spain
| | - Isabel Esmoris
- Biomedical Research Institute INCLIVA, Valencia, Spain
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, Valencia, Spain
| | - Amparo Gimeno
- Biomedical Research Institute INCLIVA, Valencia, Spain
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, Valencia, Spain
| | - Jose Luis García-Giménez
- CIBER de Enfermedades Raras, Valencia, Spain
- Biomedical Research Institute INCLIVA, Valencia, Spain
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, Valencia, Spain
| | - Federico V. Pallardó
- CIBER de Enfermedades Raras, Valencia, Spain
- Biomedical Research Institute INCLIVA, Valencia, Spain
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, Valencia, Spain
| | - Rosario Perona
- Instituto de Investigaciones Biomédicas CSIC/UAM, Madrid, Spain
- CIBER de Enfermedades Raras, Valencia, Spain
- * E-mail:
| |
Collapse
|
25
|
Tang Q, Koh LK, Jiang D, Schwarz H. CD137 ligand reverse signaling skews hematopoiesis towards myelopoiesis during aging. Aging (Albany NY) 2014; 5:643-52. [PMID: 23945137 PMCID: PMC3808697 DOI: 10.18632/aging.100588] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
CD137 is a costimulatory molecule expressed on activated T cells. Its ligand, CD137L, is expressed on the surface of hematopoietic progenitor cells, and upon binding to CD137 induces reverse signaling into hematopoietic progenitor cells promoting their activation, proliferation and myeloid differentiation. Since aging is associated with an increasing number of myeloid cells we investigated the role of CD137 and CD137L on myelopoiesis during aging. Comparing 3 and 12 months old WT, CD137−/− and CD137L−/− mice we found significantly more granulocytes and monocytes in the bone marrow of older WT mice, while this age-dependent increase was absent in CD137−/− and CD137L−/− mice. Instead, the bone marrow of 12 months old CD137−/− and CD137L−/− mice was characterized by an accumulation of hematopoietic progenitor cells, suggesting that the differentiation of hematopoietic progenitor cells became arrested in the absence of CD137L signaling. CD137L signaling is initiated by activated CD137-expressing, CD4+ T cells. These data identify a novel molecular mechanisms underlying immune aging by demonstrating that CD137-expressing CD4+ T cells in the bone marrow engage CD137L on hematopoietic progenitor cells, and that this CD137L signaling biases hematopoiesis towards myelopoiesis during aging.
Collapse
|
26
|
Alawi F, Lin P. Dyskerin localizes to the mitotic apparatus and is required for orderly mitosis in human cells. PLoS One 2013; 8:e80805. [PMID: 24303026 PMCID: PMC3841160 DOI: 10.1371/journal.pone.0080805] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Accepted: 10/15/2013] [Indexed: 11/18/2022] Open
Abstract
Dyskerin is a highly conserved, nucleolar RNA-binding protein with established roles in small nuclear ribonucleoprotein biogenesis, telomerase and telomere maintenance and precursor rRNA processing. Telomerase is functional during S phase and the bulk of rRNA maturation occurs during G1 and S phases; both processes are inactivated during mitosis. Yet, we show that during the course of cell cycle progression, human dyskerin expression peaks during G2/M in parallel with the upregulation of pro-mitotic factors. Dyskerin redistributed from the nucleolus in interphase cells to the perichromosomal region during prometaphase, metaphase and anaphase. With continued anaphase progression, dyskerin also localized to the cytoplasm within the mid-pole region. Loss of dyskerin function via siRNA-mediated depletion promoted G2/M accumulation and this was accompanied by an increased mitotic index and activation of the spindle assembly checkpoint. Live cell imaging further revealed an array of mitotic defects including delayed prometaphase progression, a significantly increased incidence of multi-polar spindles, and anaphase bridges culminating in micronucleus formation. Together, these findings suggest that dyskerin is a highly dynamic protein throughout the cell cycle and increases the repertoire of fundamental cellular processes that are disrupted by absence of its normal function.
Collapse
Affiliation(s)
- Faizan Alawi
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| | - Ping Lin
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
27
|
Pereboeva L, Westin E, Patel T, Flaniken I, Lamb L, Klingelhutz A, Goldman F. DNA damage responses and oxidative stress in dyskeratosis congenita. PLoS One 2013; 8:e76473. [PMID: 24124565 PMCID: PMC3790691 DOI: 10.1371/journal.pone.0076473] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Accepted: 08/27/2013] [Indexed: 12/23/2022] Open
Abstract
Dyskeratosis congenita (DC) is an inherited multisystem disorder of premature aging, cancer predisposition, and bone marrow failure caused by selective exhaustion of highly proliferative cell pools. DC patients also have a poor tolerance to chemo/radiotherapy and bone marrow transplantation. Although critically shortened telomeres and defective telomere maintenance contribute to DC pathology, other mechanisms likely exist. We investigate the link between telomere dysfunction and oxidative and DNA damage response pathways and assess the effects of antioxidants. In vitro studies employed T lymphocytes from DC subjects with a hTERC mutation and age-matched controls. Cells were treated with cytotoxic agents, including Paclitaxel, Etoposide, or ionizing radiation. Apoptosis and reactive oxygen species (ROS) were assessed by flow cytometry, and Western blotting was used to measure expression of DNA damage response (DDR) proteins, including total p53, p53S15, and p21(WAF). N-acetyl-cysteine (NAC), an antioxidant, was used to modulate cell growth and ROS. In stimulated culture, DC lymphocytes displayed a stressed phenotype, characterized by elevated levels of ROS, DDR and apoptotic markers as well as a proliferative defect that was more pronounced after exposure to cytotoxic agents. NAC partially ameliorated the growth disadvantage of DC cells and decreased radiation-induced apoptosis and oxidative stress. These findings suggest that oxidative stress may play a role in the pathogenesis of DC and that pharmacologic intervention to correct this pro-oxidant imbalance may prove useful in the clinical setting, potentially alleviating untoward toxicities associated with current cytotoxic treatments.
Collapse
Affiliation(s)
- Larisa Pereboeva
- Department of Medicine, Division of Hematology Oncology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Erik Westin
- Department of Pediatrics, Division of Hematology Oncology, Children's Hospital of Alabama, Birmingham, Alabama, United States of America
| | - Toral Patel
- Department of Pediatrics, Division of Hematology Oncology, Children's Hospital of Alabama, Birmingham, Alabama, United States of America
| | - Ian Flaniken
- Department of Pediatrics, Division of Hematology Oncology, Children's Hospital of Alabama, Birmingham, Alabama, United States of America
| | - Lawrence Lamb
- Department of Medicine, Division of Hematology Oncology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Aloysius Klingelhutz
- Department of Microbiology, University of Iowa, Iowa City, Iowa, United States of America
| | - Frederick Goldman
- Department of Pediatrics, Division of Hematology Oncology, Children's Hospital of Alabama, Birmingham, Alabama, United States of America
- * E-mail:
| |
Collapse
|
28
|
Abstract
A critical question in human health is the malleability of telomere length. Telomere length, sampled at one point during adult life, is predictive of certain types of cancer and other immune and metabolic-related diseases. We now know from basic studies that the telomere/telomerase maintenance system plays a causal role in accelerating biologic aging and promoting disease processes. One can develop short telomeres for a multitude of reasons. Historical factors such as genetics, prenatal conditions, and early adversity, contribute to adult telomere length; however, current stress and lifestyle are also associated. If these modifiable predictors are causal factors in telomere shortening, there is a tremendous opportunity to improve maintenance and possibly even lengthen telomeres with behavioral interventions. This minireview discusses our current understanding of telomere lengthening and questions facing the field. Several small-scale stress reduction/wellness studies show promising findings, suggesting that cell aging can be slowed or reversed in vivo over short periods. Moreover, possible mechanisms are discussed, that take into account actual telomeric lengthening, such as that which occurs through telomerase-mediated elongation, or mechanisms resulting in "pseudo-telomeric lengthening" as might occur from changes in cell type distribution. There is a strong need for more translational clinical to bench research to address mechanistic questions in experimental models. In addition, well-designed intervention research that examines both telomeres and potential mediators of change can further enhance our understanding of malleability, mechanism, and clinical implications of telomere lengthening. Cancer Prev Res; 5(10); 1163-8. ©2012 AACR.
Collapse
Affiliation(s)
- Elissa Epel
- University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
29
|
Cho J, Kook S, Robinson AR, Niedernhofer LJ, Lee BC. Cell autonomous and nonautonomous mechanisms drive hematopoietic stem/progenitor cell loss in the absence of DNA repair. Stem Cells 2013; 31:511-25. [PMID: 23097336 PMCID: PMC3582850 DOI: 10.1002/stem.1261] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Accepted: 09/23/2012] [Indexed: 12/24/2022]
Abstract
Daily, cells incur tens of thousands of DNA lesions caused by endogenous processes. Due to their long-lived nature, adult stem cells may be particularly susceptible to the negative impact of this constant genotoxic stress. Indeed, in murine models of DNA repair deficiencies, there is accumulation of DNA damage in hematopoietic stem cells and premature loss of function. Herein, we demonstrate that mice expressing reduced levels of ERCC1-XPF DNA repair endonuclease (Ercc1-/Δ mice) spontaneously display a progressive decline in the number and function of hematopoietic stem/progenitor cells (HSPCs). This was accompanied by increased cell death, expression of senescence markers, reactive oxygen species, and DNA damage in HSPC populations, illustrating cell autonomous mechanisms that contribute to loss of function. In addition, the bone marrow microenvironment of Ercc1-/Δ mice was not permissive for the engraftment of transplanted normal stem cells. Bones from Ercc1-/Δ mice displayed excessive osteoclastic activity, which alters the microenvironment in a way that is unfavorable to HSPC maintenance. This was accompanied by increased proinflammatory cytokines in the bone marrow of Ercc1-/Δ mice. These data provide novel evidence that spontaneous, endogenous DNA damage, if not repaired, promotes progressive attrition of adult stem cells via both cell autonomous and nonautonomous mechanisms.
Collapse
Affiliation(s)
- JoonSeok Cho
- University of Pittsburgh Cancer Institute, 5117 Centre Ave, Hillman Cancer Center, Pittsburgh, PA 15213 USA
- Department of Medicine, University of Pittsburgh, 1218 Scaife Hall, 3550 Terrace Street, Pittsburgh, PA, 15216
| | - SungHo Kook
- University of Pittsburgh Cancer Institute, 5117 Centre Ave, Hillman Cancer Center, Pittsburgh, PA 15213 USA
- Department of Medicine, University of Pittsburgh, 1218 Scaife Hall, 3550 Terrace Street, Pittsburgh, PA, 15216
| | - Andria Rasile Robinson
- University of Pittsburgh Cancer Institute, 5117 Centre Ave, Hillman Cancer Center, Pittsburgh, PA 15213 USA
- Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, 130 DeSoto Street, Pittsburgh, PA 15261 USA
| | - Laura J. Niedernhofer
- University of Pittsburgh Cancer Institute, 5117 Centre Ave, Hillman Cancer Center, Pittsburgh, PA 15213 USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, 523 Bridgeside Point II, 450 Technology Drive, Pittsburgh, PA 15219 USA
| | - Byeong-Chel Lee
- University of Pittsburgh Cancer Institute, 5117 Centre Ave, Hillman Cancer Center, Pittsburgh, PA 15213 USA
- Department of Medicine, University of Pittsburgh, 1218 Scaife Hall, 3550 Terrace Street, Pittsburgh, PA, 15216
| |
Collapse
|
30
|
Cai Z, Yan LJ, Ratka A. Telomere Shortening and Alzheimer’s Disease. Neuromolecular Med 2012; 15:25-48. [PMID: 23161153 DOI: 10.1007/s12017-012-8207-9] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Accepted: 11/02/2012] [Indexed: 10/27/2022]
|
31
|
Agostini A, Mondragón L, Bernardos A, Martínez-Máñez R, Marcos MD, Sancenón F, Soto J, Costero A, Manguan-García C, Perona R, Moreno-Torres M, Aparicio-Sanchis R, Murguía JR. Targeted Cargo Delivery in Senescent Cells Using Capped Mesoporous Silica Nanoparticles. Angew Chem Int Ed Engl 2012. [DOI: 10.1002/ange.201204663] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
32
|
Agostini A, Mondragón L, Bernardos A, Martínez-Máñez R, Marcos MD, Sancenón F, Soto J, Costero A, Manguan-García C, Perona R, Moreno-Torres M, Aparicio-Sanchis R, Murguía JR. Targeted Cargo Delivery in Senescent Cells Using Capped Mesoporous Silica Nanoparticles. Angew Chem Int Ed Engl 2012; 51:10556-60. [DOI: 10.1002/anie.201204663] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Revised: 07/31/2012] [Indexed: 12/12/2022]
|
33
|
Tümpel S, Rudolph KL. The role of telomere shortening in somatic stem cells and tissue aging: lessons from telomerase model systems. Ann N Y Acad Sci 2012; 1266:28-39. [DOI: 10.1111/j.1749-6632.2012.06547.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
34
|
Liu J, Liu M, Ye X, Liu K, Huang J, Wang L, Ji G, Liu N, Tang X, Baltz JM, Keefe DL, Liu L. Delay in oocyte aging in mice by the antioxidant N-acetyl-L-cysteine (NAC). Hum Reprod 2012; 27:1411-20. [PMID: 22357770 DOI: 10.1093/humrep/des019] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Ovarian aging is associated with declining numbers and quality of oocytes and follicles. Oxidative stress by reactive oxygen species (ROS) contributes to somatic aging in general, and also has been implicated in reproductive aging. Telomere shortening is also involved in aging, and telomeres are particularly susceptible to ROS-induced damage. Previously, we have shown that antioxidant N-acetyl-L-cysteine (NAC) effectively rescues oocytes and embryos from ROS-induced telomere shortening and apoptosis in vitro. Using mice as models, we tested the hypothesis that reducing oxidative stress by NAC might prevent or delay ovarian aging in vivo. METHODS Initially, young females were treated with NAC in drinking water for 2 months and the quality of fertilized oocytes and early embryo development were evaluated. Next, young mice 1-1½ months old were treated for 1 year with NAC added in drinking water, and their fertility was analyzed starting at 6 months, as indicated by litter size, oocyte number and quality. The ovaries were also examined for telomere activity and length and the expression of selected genes related to aging and DNA damage. RESULTS Short-term treatment of mice for 2 months with NAC demonstrated that NAC improved the quality of fertilized oocytes and early embryo development. Mice treated with a long-term low concentration (0.1 mM) of NAC had increased litter sizes at the ages of 7-10 months compared with age-matched controls without NAC treatment. NAC also increased the quality of the oocytes from these older mice. Moreover, the expression of sirtuins was increased, telomerase activity was higher and telomere length was longer in the ovaries of mice treated with NAC compared with those of the control group. CONCLUSIONS These data suggest that appropriate treatment with the antioxidant NAC postpones the process of oocyte aging in mice.
Collapse
Affiliation(s)
- Jinmiao Liu
- Department of Cell Biology and Genetics, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Zhu JY, Ye Q, Zhao QP, Ming ZP, Grevelding CG, Jiang MS, Dong HF. Effects of protein extract from head-foot tissue of Oncomelania hupensis on the growth and gene expression of mother sporocysts of Schistosoma japonicum. Parasitol Res 2011; 110:721-31. [PMID: 21800125 DOI: 10.1007/s00436-011-2548-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2011] [Accepted: 07/01/2011] [Indexed: 10/17/2022]
Abstract
Oncomelania hupensis is the intermediate host of Schistosoma japonicum. In the present study, we investigated the effects of protein extracts from head-foot or gland tissue of O. hupensis on mother sporocysts of S. japonicum cultured in vitro. In the presence of head-foot protein extract of snails from the native province Hunan, in-vitro-transformed mother sporocysts presented not only a longer survival time and stronger motility, but also a bigger size than parasites cultured with protein extracts of glands of the same snail or head-foot tissue of a non-native snail from the Hubei province. Using suppression subtractive hybridization, two subtractive libraries were constructed on the basis of RNA of sporocysts cultured with or without native snail head-foot protein extract. A number of 31 transcripts were found to be up-regulated. Sequence analyses revealed that they represented genes involved among others in metabolic process, electron transport chain, response to chemical stimulus, and oxidation-reduction processes. Opposite to that 20 down-regulated transcripts were among others related to pseudouridine synthesis, RNA processing, and ribosome biogenesis. The differential expression of three of these transcripts, encoding cytochrome c oxidase subunit 2 (Cox2), NADH-ubiquinone oxidoreductase (ND1), and dyskeratosis congenita 1 protein (DKC1), were confirmed by real-time PCR. The promoted development and the differential gene expression of cultured sporocysts under the influence of head-foot protein extract of native O. hupensis implied not only its ability to improve in vitro culture conditions for intramolluscan stages, it may also represent a priming result with respect to the identification and characterization of factors involved in the parasite-host interplay between S. japonicum and O. hupensis.
Collapse
Affiliation(s)
- Jun Yong Zhu
- Department of Parasitology, School of Basic Medical Science, Wuhan University, Wuhan, Hubei Province, China
| | | | | | | | | | | | | |
Collapse
|