1
|
Kvarnung M, Pettersson M, Chun-On P, Rafati M, McReynolds LJ, Norberg A, Moura PL, Pesonen I, Chaireti R, Grönros Söderholm B, Burlin J, Rydén J, Lindberg EH, Giri N, Savage SA, Agarwal S, Nordgren A, Tesi B. Identification of biallelic POLA2 variants in two families with an autosomal recessive telomere biology disorder. Eur J Hum Genet 2025; 33:580-587. [PMID: 39616267 PMCID: PMC12048608 DOI: 10.1038/s41431-024-01722-8] [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: 01/09/2024] [Revised: 09/11/2024] [Accepted: 10/17/2024] [Indexed: 05/04/2025] Open
Abstract
POLA2 encodes the accessory subunit of DNA polymerase α (polα)/primase, which is crucial for telomere C-strand fill-in. Incomplete fill-in of the C-rich telomeric strand after DNA replication has been proposed as a mechanism for Coats plus syndrome, a phenotype within the broader spectrum of telomere biology disorders (TBD). Coats plus syndrome has so far been associated with pathogenic variants in POT1, CTC1, and STN1. Here we report the findings of biallelic deleterious rare variants in POLA2 gene detected by whole genome sequencing and segregation analysis in five young adults from two unrelated families. All five individuals displayed abnormally short telomeres and a clinical phenotype suggesting a TBD disorder with Coats plus features including retinal and gastrointestinal telangiectasias. Our results suggest POLA2 as a novel autosomal recessive gene for a TBD with Coats plus features.
Collapse
Affiliation(s)
- Malin Kvarnung
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.
- Clinical Genetics and Genomics, Karolinska University Hospital, Stockholm, Sweden.
| | - Maria Pettersson
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Clinical Genetics and Genomics, Karolinska University Hospital, Stockholm, Sweden
| | - Pattra Chun-On
- Division of Hematology/Oncology, Department of Pediatrics, Boston Children's Hospital; Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Maryam Rafati
- Division of Cancer Epidemiology and Genetics, Clinical Genetics Branch, National Cancer Institute, Bethesda, MD, USA
| | - Lisa J McReynolds
- Division of Cancer Epidemiology and Genetics, Clinical Genetics Branch, National Cancer Institute, Bethesda, MD, USA
| | - Anna Norberg
- Department of Medical Biosciences, Medical and Clinical Genetics, Umeå University, Umeå, Sweden
| | - Pedro Luis Moura
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ida Pesonen
- Department of Medicine Solna, Respiratory Medicine Unit, Karolinska Institutet, Stockholm, Sweden
- Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm, Sweden
| | - Roza Chaireti
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Department of Hematology, Karolinska University Hospital, Stockholm, Sweden
- Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | | | - Julia Burlin
- Division of Nephrology, Danderyd University Hospital, Stockholm, Sweden
| | - Jenny Rydén
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Hematology, Karolinska University Hospital, Stockholm, Sweden
| | - Eva Hellström Lindberg
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Hematology, Karolinska University Hospital, Stockholm, Sweden
| | - Neelam Giri
- Division of Cancer Epidemiology and Genetics, Clinical Genetics Branch, National Cancer Institute, Bethesda, MD, USA
| | - Sharon A Savage
- Division of Cancer Epidemiology and Genetics, Clinical Genetics Branch, National Cancer Institute, Bethesda, MD, USA
| | - Suneet Agarwal
- Division of Hematology/Oncology, Department of Pediatrics, Boston Children's Hospital; Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Ann Nordgren
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Clinical Genetics and Genomics, Karolinska University Hospital, Stockholm, Sweden
- Department of Clinical Genetics and Genomics, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Genomic Medicine Center Karolinska, Karolinska University Hospital, Stockholm, Sweden
| | - Bianca Tesi
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Clinical Genetics and Genomics, Karolinska University Hospital, Stockholm, Sweden
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
2
|
Sanz-Moreno A, Becker L, Xie K, da Silva-Buttkus P, Dragano NRV, Aguilar-Pimentel A, Amarie OV, Calzada-Wack J, Kraiger M, Leuchtenberger S, Seisenberger C, Marschall S, Rathkolb B, Scifo E, Liu T, Thanabalasingam A, Sanchez-Vazquez R, Martinez P, Blasco MA, Savage SA, Fuchs H, Ehninger D, Gailus-Durner V, de Angelis MH. Loss of Ten1 in mice induces telomere shortening and models human dyskeratosis congenita. SCIENCE ADVANCES 2025; 11:eadp8093. [PMID: 40215293 PMCID: PMC11988282 DOI: 10.1126/sciadv.adp8093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 03/07/2025] [Indexed: 04/14/2025]
Abstract
Telomere length regulation is essential for genome stability as short telomeres can trigger cellular senescence and apoptosis constituting an integral aspect of biological aging. Telomere biology disorders (TBDs) such as dyskeratosis congenita (DC) are rare, inherited diseases with known mutations in at least 16 different genes encoding components of the telomere maintenance complexes. The precise role of TEN1, part of the CST complex (CTC1, STN1, and TEN1), and the consequences of its loss of function in vivo are not yet known. We investigated the first viable murine model of Ten1 deficiency created by CRISPR-Cas9-mediated exon 3 deletion. Ten1 homozygous knockout mice present with telomere attrition, short life span, skin hyperpigmentation, aplastic anemia, and cerebellar hypoplasia. Molecular analyses revealed a reduction of proliferating cells, increased apoptosis, and stem cell depletion with activation of the p53/p21 signaling pathway. Our data demonstrate that Ten1 deficiency causes telomere shortening and associates with accelerated aging.
Collapse
Affiliation(s)
- Adrián Sanz-Moreno
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Lore Becker
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Kan Xie
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127 Bonn, Germany
| | - Patricia da Silva-Buttkus
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Nathalia R. V. Dragano
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
- German Center for Diabetes Research (DZD), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Antonio Aguilar-Pimentel
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Oana V. Amarie
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Julia Calzada-Wack
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Markus Kraiger
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Stefanie Leuchtenberger
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Claudia Seisenberger
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Susan Marschall
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Birgit Rathkolb
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
- German Center for Diabetes Research (DZD), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-University München, Feodor-Lynen Str. 25, 81377 Munich, Germany
| | - Enzo Scifo
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127 Bonn, Germany
| | - Ting Liu
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127 Bonn, Germany
| | - Anoja Thanabalasingam
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127 Bonn, Germany
| | - Raul Sanchez-Vazquez
- Telomeres and Telomerase Group–Fundación Humanismo y Ciencia, Molecular Oncology Program, Spanish National Cancer Centre (CNIO), Melchor Fernández Almagro 3, Madrid, E-28029, Spain
| | - Paula Martinez
- Telomeres and Telomerase Group–Fundación Humanismo y Ciencia, Molecular Oncology Program, Spanish National Cancer Centre (CNIO), Melchor Fernández Almagro 3, Madrid, E-28029, Spain
| | - Maria A. Blasco
- Telomeres and Telomerase Group–Fundación Humanismo y Ciencia, Molecular Oncology Program, Spanish National Cancer Centre (CNIO), Melchor Fernández Almagro 3, Madrid, E-28029, Spain
| | - Sharon A. Savage
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Helmut Fuchs
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Dan Ehninger
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127 Bonn, Germany
| | - Valérie Gailus-Durner
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Martin Hrabê de Angelis
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
- German Center for Diabetes Research (DZD), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
- Chair of Experimental Genetics, TUM School of Life Sciences, Technische Universität München, Alte Akademie 8, 85354 Freising, Germany
| |
Collapse
|
3
|
Jones-Weinert C, Mainz L, Karlseder J. Telomere function and regulation from mouse models to human ageing and disease. Nat Rev Mol Cell Biol 2025; 26:297-313. [PMID: 39614014 DOI: 10.1038/s41580-024-00800-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/16/2024] [Indexed: 12/01/2024]
Abstract
Telomeres protect the ends of chromosomes but shorten following cell division in the absence of telomerase activity. When telomeres become critically short or damaged, a DNA damage response is activated. Telomeres then become dysfunctional and trigger cellular senescence or death. Telomere shortening occurs with ageing and may contribute to associated maladies such as infertility, neurodegeneration, cancer, lung dysfunction and haematopoiesis disorders. Telomere dysfunction (sometimes without shortening) is associated with various diseases, known as telomere biology disorders (also known as telomeropathies). Telomere biology disorders include dyskeratosis congenita, Høyeraal-Hreidarsson syndrome, Coats plus syndrome and Revesz syndrome. Although mouse models have been invaluable in advancing telomere research, full recapitulation of human telomere-related diseases in mice has been challenging, owing to key differences between the species. In this Review, we discuss telomere protection, maintenance and damage. We highlight the differences between human and mouse telomere biology that may contribute to discrepancies between human diseases and mouse models. Finally, we discuss recent efforts to generate new 'humanized' mouse models to better model human telomere biology. A better understanding of the limitations of mouse telomere models will pave the road for more human-like models and further our understanding of telomere biology disorders, which will contribute towards the development of new therapies.
Collapse
Affiliation(s)
| | - Laura Mainz
- The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Jan Karlseder
- The Salk Institute for Biological Studies, La Jolla, CA, USA.
| |
Collapse
|
4
|
Sertori R, Truong B, Singh MK, Shinton S, Price R, Sharo A, Shultes P, Sunderam U, Rana S, Srinivasan R, Datta S, Font-Burgada J, Brenner SE, Puck JM, Wiest DL. Disruption of the moonlighting function of CTF18 in a patient with T-lymphopenia. Front Immunol 2025; 16:1539848. [PMID: 40028343 PMCID: PMC11868726 DOI: 10.3389/fimmu.2025.1539848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 01/22/2025] [Indexed: 03/05/2025] Open
Abstract
Introduction Newborn screening for immunodeficiency has led to the identification of numerous cases for which the causal etiology is unknown. Methods Here we report the diagnosis of T lymphopenia of unknown etiology in a male proband. Whole exome sequencing (WES) was employed to nominate candidate variants, which were then analyzed functionally in zebrafish and in mice bearing orthologous mutations. Results WES revealed missense mutations in CHTF18 that were inherited in an autosomal recessive manner. CTF18, encoded by the CHTF18 gene, is a component of a secondary clamp loader, which is primarily thought to function by promoting DNA replication. We determined that the patient's variants in CHTF18 (CTF18 R751W and E851Q) were damaging to function and severely attenuated the capacity of CTF18 to support hematopoiesis and lymphoid development, strongly suggesting that they were responsible for his T lymphopenia; however, the function of CTF18 appeared to be unrelated to its role as a clamp loader. DNA-damage, expected when replication is impaired, was not evident by expression profiling in murine Chtf18 mutant hematopoietic stem and progenitor cells (HSPC), nor was development of Ctf18-deficient progenitors rescued by p53 loss. Instead, we observed an expression signature suggesting disruption of HSPC positioning and migration. Indeed, the positioning of HSPC in ctf18 morphant zebrafish embryos was perturbed, suggesting that HSPC function was impaired through disrupted positioning in hematopoietic organs. Discussion Accordingly, we propose that T lymphopenia in our patient resulted from disturbed cell-cell contacts and migration of HSPC, caused by a non-canonical function of CHTF18 in regulating gene expression.
Collapse
Affiliation(s)
- Robert Sertori
- Nuclear Dynamics and Cancer Program, Fox Chase Cancer Center, Philadelphia, PA, United States
| | - Billy Truong
- Nuclear Dynamics and Cancer Program, Fox Chase Cancer Center, Philadelphia, PA, United States
| | - Manoj K. Singh
- Nuclear Dynamics and Cancer Program, Fox Chase Cancer Center, Philadelphia, PA, United States
| | - Susan Shinton
- Nuclear Dynamics and Cancer Program, Fox Chase Cancer Center, Philadelphia, PA, United States
| | - Rachael Price
- Nuclear Dynamics and Cancer Program, Fox Chase Cancer Center, Philadelphia, PA, United States
| | - Andrew Sharo
- Center for Computational Biology, University of California, Berkeley, Berkeley, CA, United States
| | - Paulameena Shultes
- Center for Computational Biology, University of California, Berkeley, Berkeley, CA, United States
| | - Uma Sunderam
- Innovation Labs, Tata Consultancy Services, Hyderabad, India
| | - Sadhna Rana
- Innovation Labs, Tata Consultancy Services, Hyderabad, India
| | | | - Sutapa Datta
- Innovation Labs, Tata Consultancy Services, Hyderabad, India
| | - Joan Font-Burgada
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, United States
| | - Steven E. Brenner
- Center for Computational Biology, University of California, Berkeley, Berkeley, CA, United States
| | - Jennifer M. Puck
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, United States
- Department of Pediatrics, University of California, San Francisco (UCSF) and UCSF Benioff Children’s Hospital, San Francisco, CA, United States
| | - David L. Wiest
- Nuclear Dynamics and Cancer Program, Fox Chase Cancer Center, Philadelphia, PA, United States
| |
Collapse
|
5
|
Muiño E, Carcel-Marquez J, Llucià-Carol L, Gallego-Fabrega C, Cullell N, Lledós M, Martín-Campos JM, Villatoro-González P, Sierra-Marcos A, Ros-Castelló V, Aguilera-Simón A, Marti-Fabregas J, Fernandez-Cadenas I. Identification of Genetic Loci Associated With Intracerebral Hemorrhage Using a Multitrait Analysis Approach. Neurology 2024; 103:e209666. [PMID: 39298701 PMCID: PMC11446162 DOI: 10.1212/wnl.0000000000209666] [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: 02/05/2024] [Accepted: 05/15/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Genome-wide association studies (GWASs) have only 2 loci associated with spontaneous intracerebral hemorrhage (ICH): APOE for lobar and 1q22 for nonlobar ICH. We aimed to discover new loci through an analysis that combines correlated traits (multi-trait analysis of GWAS [MTAG]) and explore a gene-based analysis, transcriptome-wide association study (TWAS), and proteome-wide association study (PWAS) to understand the biological mechanisms of spontaneous ICH providing potential therapeutic targets. METHODS We use the published MTAG of ICH (patients with spontaneous intraparenchymal bleeding) and small-vessel ischemic stroke. For all ICH, lobar ICH, and nonlobar ICH, a pairwise MTAG combined ICH with traits related to cardiovascular risk factors, cerebrovascular diseases, or Alzheimer disease (AD). For the analysis, we assembled those traits with a genetic correlation ≥0.3. A new MTAG combining multiple traits was performed with those traits whose pairwise MTAG yielded new GWAS-significant single nucleotide polymorphisms (SNPs), with a posterior-probability of model 3 (GWAS-pairwise) ≥0.6. We perform TWAS and PWAS that correlate the genetic component of expression or protein levels with the genetic component of a trait. We use the ICH cohort from UK Biobank as replication. RESULTS For all ICH (1,543 ICH, 1,711 controls), the mean age was 72 ± 2 in cases and 70 ± 2 in controls, and half of them were women. Replication cohort: 700 ICH and 399,717 controls. Novel loci were found only for all ICH (the trait containing lobar and nonlobar ICH), combining data of ICH and small vessel stroke, white matter hyperintensities volume, fractional anisotropy, mean diffusivity, and AD. We replicated 6 SNPs belonging to 2q33.2 (ICA1L, β = 0.20, SE = 0.03, p value = 8.91 × 10-12), 10q24.33 (OBFC1, β = -0.12, SE = 0.02, p value = 1.67 × 10-8), 13q34 (COL4A2, β = 0.02, SE = 0.02, p value = 2.34 × 10-11), and 19q13.32 (APOC1, β = -0.19, SE = 0.03, p value = 1.38 × 10-12; APOE, β = 0.21, SE = 0.03, p value = 2.70 × 10-11; PVRL2:CTB-129P6.4, β = 0.15, SE = 0.03, p value = 1.38 × 10-8); 2 genes (SH3PXD2A, Z-score = 4.83, p value = 6.67 × 10-7; and APOC1, Z-score: = 5.11, p value = 1.60 × 10-7); and ICA1L transcript (Z-score = 6.8, p value = 9.1 × 10-12) and protein levels (Z-score = -5.8, p value = 6.7 × 10-9). DISCUSSION Our results reinforce the role of APOE in ICH risk, replicate previous ICH-associated loci (2q33 and 13q34), and point to new ICH associations with OBFC1, PVRL2:CTB-129P6.4, APOC1, and SH3PXD2A. Our study used data from European subjects, our main limitation. These molecules could be potential targets for future studies for modulating ICH risk.
Collapse
Affiliation(s)
- Elena Muiño
- From the Stroke Pharmacogenomics and Genetics Group (E.M., J.C.-M., L.L.-C., C.G.-F., N.C., M.L.L., J.M.M.-C., P.V.-G., I.F.-C.), Biomedical Research Institute Sant Pau (IIB SANT PAU); Epilepsy Unit (E.M., A.S.-M., V.R.-C.), Neurology Service, Hospital de la Santa Creu i Sant Pau, Barcelona; Stroke Pharmacogenomics and Genetics (N.C.), Fundació MútuaTerrassa per la Docència i la Recerca; and Department of Neurology (C.G.-F., A.A.-S., J.M.-F.), Hospital de la Santa Creu i Sant Pau, IIB SANT PAU, Barcelona, Spain
| | - Jara Carcel-Marquez
- From the Stroke Pharmacogenomics and Genetics Group (E.M., J.C.-M., L.L.-C., C.G.-F., N.C., M.L.L., J.M.M.-C., P.V.-G., I.F.-C.), Biomedical Research Institute Sant Pau (IIB SANT PAU); Epilepsy Unit (E.M., A.S.-M., V.R.-C.), Neurology Service, Hospital de la Santa Creu i Sant Pau, Barcelona; Stroke Pharmacogenomics and Genetics (N.C.), Fundació MútuaTerrassa per la Docència i la Recerca; and Department of Neurology (C.G.-F., A.A.-S., J.M.-F.), Hospital de la Santa Creu i Sant Pau, IIB SANT PAU, Barcelona, Spain
| | - Laia Llucià-Carol
- From the Stroke Pharmacogenomics and Genetics Group (E.M., J.C.-M., L.L.-C., C.G.-F., N.C., M.L.L., J.M.M.-C., P.V.-G., I.F.-C.), Biomedical Research Institute Sant Pau (IIB SANT PAU); Epilepsy Unit (E.M., A.S.-M., V.R.-C.), Neurology Service, Hospital de la Santa Creu i Sant Pau, Barcelona; Stroke Pharmacogenomics and Genetics (N.C.), Fundació MútuaTerrassa per la Docència i la Recerca; and Department of Neurology (C.G.-F., A.A.-S., J.M.-F.), Hospital de la Santa Creu i Sant Pau, IIB SANT PAU, Barcelona, Spain
| | - Cristina Gallego-Fabrega
- From the Stroke Pharmacogenomics and Genetics Group (E.M., J.C.-M., L.L.-C., C.G.-F., N.C., M.L.L., J.M.M.-C., P.V.-G., I.F.-C.), Biomedical Research Institute Sant Pau (IIB SANT PAU); Epilepsy Unit (E.M., A.S.-M., V.R.-C.), Neurology Service, Hospital de la Santa Creu i Sant Pau, Barcelona; Stroke Pharmacogenomics and Genetics (N.C.), Fundació MútuaTerrassa per la Docència i la Recerca; and Department of Neurology (C.G.-F., A.A.-S., J.M.-F.), Hospital de la Santa Creu i Sant Pau, IIB SANT PAU, Barcelona, Spain
| | - Natalia Cullell
- From the Stroke Pharmacogenomics and Genetics Group (E.M., J.C.-M., L.L.-C., C.G.-F., N.C., M.L.L., J.M.M.-C., P.V.-G., I.F.-C.), Biomedical Research Institute Sant Pau (IIB SANT PAU); Epilepsy Unit (E.M., A.S.-M., V.R.-C.), Neurology Service, Hospital de la Santa Creu i Sant Pau, Barcelona; Stroke Pharmacogenomics and Genetics (N.C.), Fundació MútuaTerrassa per la Docència i la Recerca; and Department of Neurology (C.G.-F., A.A.-S., J.M.-F.), Hospital de la Santa Creu i Sant Pau, IIB SANT PAU, Barcelona, Spain
| | - Miquel Lledós
- From the Stroke Pharmacogenomics and Genetics Group (E.M., J.C.-M., L.L.-C., C.G.-F., N.C., M.L.L., J.M.M.-C., P.V.-G., I.F.-C.), Biomedical Research Institute Sant Pau (IIB SANT PAU); Epilepsy Unit (E.M., A.S.-M., V.R.-C.), Neurology Service, Hospital de la Santa Creu i Sant Pau, Barcelona; Stroke Pharmacogenomics and Genetics (N.C.), Fundació MútuaTerrassa per la Docència i la Recerca; and Department of Neurology (C.G.-F., A.A.-S., J.M.-F.), Hospital de la Santa Creu i Sant Pau, IIB SANT PAU, Barcelona, Spain
| | - Jesús M Martín-Campos
- From the Stroke Pharmacogenomics and Genetics Group (E.M., J.C.-M., L.L.-C., C.G.-F., N.C., M.L.L., J.M.M.-C., P.V.-G., I.F.-C.), Biomedical Research Institute Sant Pau (IIB SANT PAU); Epilepsy Unit (E.M., A.S.-M., V.R.-C.), Neurology Service, Hospital de la Santa Creu i Sant Pau, Barcelona; Stroke Pharmacogenomics and Genetics (N.C.), Fundació MútuaTerrassa per la Docència i la Recerca; and Department of Neurology (C.G.-F., A.A.-S., J.M.-F.), Hospital de la Santa Creu i Sant Pau, IIB SANT PAU, Barcelona, Spain
| | - Paula Villatoro-González
- From the Stroke Pharmacogenomics and Genetics Group (E.M., J.C.-M., L.L.-C., C.G.-F., N.C., M.L.L., J.M.M.-C., P.V.-G., I.F.-C.), Biomedical Research Institute Sant Pau (IIB SANT PAU); Epilepsy Unit (E.M., A.S.-M., V.R.-C.), Neurology Service, Hospital de la Santa Creu i Sant Pau, Barcelona; Stroke Pharmacogenomics and Genetics (N.C.), Fundació MútuaTerrassa per la Docència i la Recerca; and Department of Neurology (C.G.-F., A.A.-S., J.M.-F.), Hospital de la Santa Creu i Sant Pau, IIB SANT PAU, Barcelona, Spain
| | - Alba Sierra-Marcos
- From the Stroke Pharmacogenomics and Genetics Group (E.M., J.C.-M., L.L.-C., C.G.-F., N.C., M.L.L., J.M.M.-C., P.V.-G., I.F.-C.), Biomedical Research Institute Sant Pau (IIB SANT PAU); Epilepsy Unit (E.M., A.S.-M., V.R.-C.), Neurology Service, Hospital de la Santa Creu i Sant Pau, Barcelona; Stroke Pharmacogenomics and Genetics (N.C.), Fundació MútuaTerrassa per la Docència i la Recerca; and Department of Neurology (C.G.-F., A.A.-S., J.M.-F.), Hospital de la Santa Creu i Sant Pau, IIB SANT PAU, Barcelona, Spain
| | - Victoria Ros-Castelló
- From the Stroke Pharmacogenomics and Genetics Group (E.M., J.C.-M., L.L.-C., C.G.-F., N.C., M.L.L., J.M.M.-C., P.V.-G., I.F.-C.), Biomedical Research Institute Sant Pau (IIB SANT PAU); Epilepsy Unit (E.M., A.S.-M., V.R.-C.), Neurology Service, Hospital de la Santa Creu i Sant Pau, Barcelona; Stroke Pharmacogenomics and Genetics (N.C.), Fundació MútuaTerrassa per la Docència i la Recerca; and Department of Neurology (C.G.-F., A.A.-S., J.M.-F.), Hospital de la Santa Creu i Sant Pau, IIB SANT PAU, Barcelona, Spain
| | - Ana Aguilera-Simón
- From the Stroke Pharmacogenomics and Genetics Group (E.M., J.C.-M., L.L.-C., C.G.-F., N.C., M.L.L., J.M.M.-C., P.V.-G., I.F.-C.), Biomedical Research Institute Sant Pau (IIB SANT PAU); Epilepsy Unit (E.M., A.S.-M., V.R.-C.), Neurology Service, Hospital de la Santa Creu i Sant Pau, Barcelona; Stroke Pharmacogenomics and Genetics (N.C.), Fundació MútuaTerrassa per la Docència i la Recerca; and Department of Neurology (C.G.-F., A.A.-S., J.M.-F.), Hospital de la Santa Creu i Sant Pau, IIB SANT PAU, Barcelona, Spain
| | - Joan Marti-Fabregas
- From the Stroke Pharmacogenomics and Genetics Group (E.M., J.C.-M., L.L.-C., C.G.-F., N.C., M.L.L., J.M.M.-C., P.V.-G., I.F.-C.), Biomedical Research Institute Sant Pau (IIB SANT PAU); Epilepsy Unit (E.M., A.S.-M., V.R.-C.), Neurology Service, Hospital de la Santa Creu i Sant Pau, Barcelona; Stroke Pharmacogenomics and Genetics (N.C.), Fundació MútuaTerrassa per la Docència i la Recerca; and Department of Neurology (C.G.-F., A.A.-S., J.M.-F.), Hospital de la Santa Creu i Sant Pau, IIB SANT PAU, Barcelona, Spain
| | - Israel Fernandez-Cadenas
- From the Stroke Pharmacogenomics and Genetics Group (E.M., J.C.-M., L.L.-C., C.G.-F., N.C., M.L.L., J.M.M.-C., P.V.-G., I.F.-C.), Biomedical Research Institute Sant Pau (IIB SANT PAU); Epilepsy Unit (E.M., A.S.-M., V.R.-C.), Neurology Service, Hospital de la Santa Creu i Sant Pau, Barcelona; Stroke Pharmacogenomics and Genetics (N.C.), Fundació MútuaTerrassa per la Docència i la Recerca; and Department of Neurology (C.G.-F., A.A.-S., J.M.-F.), Hospital de la Santa Creu i Sant Pau, IIB SANT PAU, Barcelona, Spain
| |
Collapse
|
6
|
Diaz Escarcega R, Marshall P, Tsvetkov AS. G-quadruplex DNA and RNA in cellular senescence. FRONTIERS IN AGING 2024; 5:1491389. [PMID: 39444378 PMCID: PMC11496277 DOI: 10.3389/fragi.2024.1491389] [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: 09/04/2024] [Accepted: 09/25/2024] [Indexed: 10/25/2024]
Abstract
Normal cells divide, are damaged, and are repaired across their lifetime. As cells age, they enter cellular senescence, characterized by a permanent state of cell-cycle arrest triggered by various stressors. The molecular mechanisms that regulate senescent phenotypes have been actively investigated over the last several decades; however, one area that has been neglected is how G-quadruplex (G4) DNA and RNA (G4-DNA and G4-RNA) mediate senescence. These non-canonical four-stranded DNA and RNA structures regulate most normative DNA and RNA-dependent processes, such as transcription, replication, and translation, as well as pathogenic mechanisms, including genomic instability and abnormal stress granule function. This review also highlights the contribution of G4s to sex differences in age-associated diseases and emphasizes potential translational approaches to target senescence and anti-aging mechanisms through G4 manipulation.
Collapse
Affiliation(s)
- Rocio Diaz Escarcega
- Department of Neurology, University of Texas McGovern Medical School, Houston, TX, United States
| | - Paul Marshall
- College of Health and Medicine, The Australian National University, Canberra, ACT, Australia
| | - Andrey S. Tsvetkov
- Department of Neurology, University of Texas McGovern Medical School, Houston, TX, United States
- The University of Texas Graduate School of Biomedical Sciences, Houston, TX, United States
- UTHealth Consortium on Aging, The University of Texas McGovern Medical School, Houston, TX, United States
| |
Collapse
|
7
|
Tummala H, Walne AJ, Badat M, Patel M, Walne AM, Alnajar J, Chow CC, Albursan I, Frost JM, Ballard D, Killick S, Szitányi P, Kelly AM, Raghavan M, Powell C, Raymakers R, Todd T, Mantadakis E, Polychronopoulou S, Pontikos N, Liao T, Madapura P, Hossain U, Vulliamy T, Dokal I. The evolving genetic landscape of telomere biology disorder dyskeratosis congenita. EMBO Mol Med 2024; 16:2560-2582. [PMID: 39198715 PMCID: PMC11473520 DOI: 10.1038/s44321-024-00118-x] [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: 04/10/2024] [Revised: 07/12/2024] [Accepted: 07/18/2024] [Indexed: 09/01/2024] Open
Abstract
Dyskeratosis congenita (DC) is a rare inherited bone marrow failure syndrome, caused by genetic mutations that principally affect telomere biology. Approximately 35% of cases remain uncharacterised at the genetic level. To explore the genetic landscape, we conducted genetic studies on a large collection of clinically diagnosed cases of DC as well as cases exhibiting features resembling DC, referred to as 'DC-like' (DCL). This led us to identify several novel pathogenic variants within known genetic loci and in the novel X-linked gene, POLA1. In addition, we have also identified several novel variants in POT1 and ZCCHC8 in multiple cases from different families expanding the allelic series of DC and DCL phenotypes. Functional characterisation of novel POLA1 and POT1 variants, revealed pathogenic effects on protein-protein interactions with primase, CTC1-STN1-TEN1 (CST) and shelterin subunit complexes, that are critical for telomere maintenance. ZCCHC8 variants demonstrated ZCCHC8 deficiency and signs of pervasive transcription, triggering inflammation in patients' blood. In conclusion, our studies expand the current genetic architecture and broaden our understanding of disease mechanisms underlying DC and DCL disorders.
Collapse
Affiliation(s)
- Hemanth Tummala
- Centre for Genomics and Child Health, Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, Newark Street, London, E12AT, UK.
- Barts Health NHS Trust, London, UK.
| | - Amanda J Walne
- Centre for Genomics and Child Health, Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, Newark Street, London, E12AT, UK
| | - Mohsin Badat
- Centre for Genomics and Child Health, Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, Newark Street, London, E12AT, UK
- Barts Health NHS Trust, London, UK
| | - Manthan Patel
- Centre for Genomics and Child Health, Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, Newark Street, London, E12AT, UK
| | - Abigail M Walne
- Centre for Genomics and Child Health, Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, Newark Street, London, E12AT, UK
| | - Jenna Alnajar
- Centre for Genomics and Child Health, Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, Newark Street, London, E12AT, UK
| | - Chi Ching Chow
- Centre for Genomics and Child Health, Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, Newark Street, London, E12AT, UK
| | - Ibtehal Albursan
- Centre for Genomics and Child Health, Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, Newark Street, London, E12AT, UK
| | - Jennifer M Frost
- Centre for Genomics and Child Health, Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, Newark Street, London, E12AT, UK
| | - David Ballard
- Department of Analytical, Environmental & Forensic Sciences, Kings College London, Franklin-Wilkins Building, Stamford Street, London, SE1 9NH, UK
| | - Sally Killick
- Department of Haematology, Royal Bournemouth Hospital NHS Foundation Trust, Bournemouth, BH7 7DW, UK
| | - Peter Szitányi
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 2, 128 08 Praha 2, Prague, Czech Republic
| | - Anne M Kelly
- Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
| | - Manoj Raghavan
- Clinical Haematology, Queen Elizabeth Hospital, Edgbaston, Birmingham, B15 2TH, UK
| | - Corrina Powell
- Clinical Genetics, Birmingham Women's and Children's NHS Foundation Trust, Birmingham, B15 2TG, UK
| | - Reinier Raymakers
- University Medical Center Utrecht, 3508 GA, Utrecht, The Netherlands
| | - Tony Todd
- Department of Haematology, Royal Devon and Exeter Hospital, Exeter, EX2 5DW, UK
| | - Elpis Mantadakis
- Department of Pediatrics' University General Hospital of Alexandroupolis, Democritus University of Thrace Faculty of Medicine, 6th Kilometer Alexandroupolis-Makris, 68 100 Alexandroupolis, Thrace, Greece
| | - Sophia Polychronopoulou
- Department of Pediatric Hematology-Oncology, Aghia Sophia Children's Hospital, Athens, Greece
| | - Nikolas Pontikos
- Institute of Ophthalmology, Faculty of Brain Sciences, University College London, Gower St, London, WC1E 6BT, UK
| | - Tianyi Liao
- Centre for Genomics and Child Health, Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, Newark Street, London, E12AT, UK
| | - Pradeep Madapura
- Centre for Genomics and Child Health, Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, Newark Street, London, E12AT, UK
| | - Upal Hossain
- Centre for Genomics and Child Health, Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, Newark Street, London, E12AT, UK
- Barts Health NHS Trust, London, UK
| | - Tom Vulliamy
- Centre for Genomics and Child Health, Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, Newark Street, London, E12AT, UK
| | - Inderjeet Dokal
- Centre for Genomics and Child Health, Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, Newark Street, London, E12AT, UK
- Barts Health NHS Trust, London, UK
| |
Collapse
|
8
|
Rolles B, Tometten M, Meyer R, Kirschner M, Beier F, Brümmendorf TH. Inherited Telomere Biology Disorders: Pathophysiology, Clinical Presentation, Diagnostics, and Treatment. Transfus Med Hemother 2024; 51:292-309. [PMID: 39371255 PMCID: PMC11452174 DOI: 10.1159/000540109] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 06/25/2024] [Indexed: 10/08/2024] Open
Abstract
Background Telomeres are the end-capping structures of all eukaryotic chromosomes thereby protecting the genome from damage and degradation. During the aging process, telomeres shorten continuously with each cell division until critically short telomeres prevent further proliferation whereby cells undergo terminal differentiation, senescence, or apoptosis. Premature aging due to critically short telomere length (TL) can also result from pathogenic germline variants in the telomerase complex or related genes that typically counteract replicative telomere shortening in germline and certain somatic cell populations, e.g., hematopoetic stem cells. Inherited diseases that result in altered telomere maintenance are summarized under the term telomere biology disorder (TBD). Summary Since TL both reflects but more importantly restricts the replicative capacity of various human tissues, a sufficient telomere reserve is particularly important in cells with high proliferative activity (e.g., hematopoiesis, immune cells, intestinal cells, liver, lung, and skin). Consequently, altered telomere maintenance as observed in TBDs typically results in premature replicative cellular exhaustion in the respective organ systems eventually leading to life-threatening complications such as bone marrow failure (BMF), pulmonary fibrosis, and liver cirrhosis. Key Messages The recognition of a potential congenital origin in approximately 10% of adult patients with clinical BMF is of utmost importance for the proper diagnosis, appropriate patient and family counseling, to prevent the use of inefficient treatment and to avoid therapy-related toxicities including appropriate donor selection when patients have to undergo stem cell transplantation from related donors. This review summarizes the current state of knowledge about TBDs with particular focus on the clinical manifestation patterns in children (termed early onset TBD) compared to adults (late-onset TBD) including typical treatment- and disease course-related complications as well as their prognosis and adequate therapy. Thereby, it aims to raise awareness for a disease group that is currently still highly underdiagnosed particularly when it first manifests itself in adulthood.
Collapse
Affiliation(s)
- Benjamin Rolles
- Division of Hematology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Medical Faculty, RWTH Aachen University, Aachen, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Cologne, Germany
| | - Mareike Tometten
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Medical Faculty, RWTH Aachen University, Aachen, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Cologne, Germany
| | - Robert Meyer
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Cologne, Germany
- Institute for Human Genetics and Genomic Medicine, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Martin Kirschner
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Medical Faculty, RWTH Aachen University, Aachen, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Cologne, Germany
| | - Fabian Beier
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Medical Faculty, RWTH Aachen University, Aachen, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Cologne, Germany
| | - Tim H. Brümmendorf
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Medical Faculty, RWTH Aachen University, Aachen, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Cologne, Germany
| |
Collapse
|
9
|
Khan S, Martin CA, Scarboro S, Wood E, Harper CA. Improvement in Cystoid Macular Edema Secondary to Systemic Bevacizumab in a Patient With Coats Plus Syndrome. JOURNAL OF VITREORETINAL DISEASES 2024; 9:24741264241276601. [PMID: 39539839 PMCID: PMC11556374 DOI: 10.1177/24741264241276601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Purpose: To report a pediatric case of Coats plus syndrome that initially presented resembling familial exudative vitreoretinopathy (FEVR). Methods: A single case was analyzed. Results: A pediatric patient was referred at 2 years of age to the retina clinic for exotropia and decreased visual acuity in the right eye and was found to have a dense vitreous hemorrhage. She later developed persistent gastrointestinal bleeding requiring regular blood transfusions and intravenous bevacizumab. Treatment with systemic bevacizumab resolved the patient's cystoid macular edema (CME). Although her presentation and examination were initially suggestive of FEVR, genetic analysis revealed heterozygous biallelic mutations in the STN1 gene, mutations that are known to be associated with Coats plus syndrome. Conclusions: Coats plus syndrome is a rare and life-threatening microangiopathy that affects the retina, central nervous system, and gastrointestinal system. The patient's resulting CME significantly improved with intravenous bevacizumab.
Collapse
Affiliation(s)
- Saima Khan
- Austin Retina Associates, The University of Texas at Austin, Dell Medical School, Austin, TX, USA
| | - Cole A. Martin
- Austin Retina Associates, The University of Texas at Austin, Dell Medical School, Austin, TX, USA
| | - Shannon Scarboro
- Austin Retina Associates, The University of Texas at Austin, Dell Medical School, Austin, TX, USA
| | - Edward Wood
- Austin Retina Associates, The University of Texas at Austin, Dell Medical School, Austin, TX, USA
| | - Clio A. Harper
- Austin Retina Associates, The University of Texas at Austin, Dell Medical School, Austin, TX, USA
| |
Collapse
|
10
|
Serpa-Irizarry M, Ruiz-Medina P, Del Valle-Segarra A, Zequeira-Diaz J. Role of Laparoscopy in Severe Gastrointestinal Bleeding Secondary to Coats Plus Syndrome. Cureus 2024; 16:e69158. [PMID: 39268024 PMCID: PMC11391663 DOI: 10.7759/cureus.69158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2024] [Indexed: 09/15/2024] Open
Abstract
Coats plus syndrome (CPS) is an exceedingly rare genetic disorder associated with premature telomere shortening. The syndrome, also known as cerebroretinal microangiopathy with calcifications and cysts, has a multisystemic manifestation. It may present as brain abnormalities, seizures, osteopenia, prenatal and postnatal growth deficiency, and portal hypertension, among others. Up to 40% of affected individuals manifest recurrent gastrointestinal (GI) bleeding which can be life-threatening in some cases. Treatment for GI bleeding is not standardized and is therefore individualized based on the patient's clinical status, comorbidities, and resource availability. We herein present a case of a 20-year-old female with CPS and a two-year history of severe recurrent GI bleeding unable to be identified by conventional endoscopy. This report highlights successful laparoscopic assisted enteroscopy with enterectomy as a novel diagnostic and therapeutic modality in this population.
Collapse
Affiliation(s)
| | - Pedro Ruiz-Medina
- General Surgery, University of Puerto Rico Medical Sciences Campus, San Juan, PRI
| | | | - Jorge Zequeira-Diaz
- General Surgery, University of Puerto Rico, Medical Sciences Campus, San Juan, PRI
| |
Collapse
|
11
|
Cai SW, Takai H, Zaug AJ, Dilgen TC, Cech TR, Walz T, de Lange T. POT1 recruits and regulates CST-Polα/primase at human telomeres. Cell 2024; 187:3638-3651.e18. [PMID: 38838667 PMCID: PMC11246235 DOI: 10.1016/j.cell.2024.05.002] [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: 06/12/2023] [Revised: 03/12/2024] [Accepted: 05/01/2024] [Indexed: 06/07/2024]
Abstract
Telomere maintenance requires the extension of the G-rich telomeric repeat strand by telomerase and the fill-in synthesis of the C-rich strand by Polα/primase. At telomeres, Polα/primase is bound to Ctc1/Stn1/Ten1 (CST), a single-stranded DNA-binding complex. Like mutations in telomerase, mutations affecting CST-Polα/primase result in pathological telomere shortening and cause a telomere biology disorder, Coats plus (CP). We determined cryogenic electron microscopy structures of human CST bound to the shelterin heterodimer POT1/TPP1 that reveal how CST is recruited to telomeres by POT1. Our findings suggest that POT1 hinge phosphorylation is required for CST recruitment, and the complex is formed through conserved interactions involving several residues mutated in CP. Our structural and biochemical data suggest that phosphorylated POT1 holds CST-Polα/primase in an inactive, autoinhibited state until telomerase has extended the telomere ends. We propose that dephosphorylation of POT1 releases CST-Polα/primase into an active state that completes telomere replication through fill-in synthesis.
Collapse
Affiliation(s)
- Sarah W Cai
- Laboratory of Cell Biology and Genetics, The Rockefeller University, New York, NY 10065, USA; Laboratory of Molecular Electron Microscopy, The Rockefeller University, New York, NY 10065, USA
| | - Hiroyuki Takai
- Laboratory of Cell Biology and Genetics, The Rockefeller University, New York, NY 10065, USA
| | - Arthur J Zaug
- Department of Biochemistry, University of Colorado Boulder, Boulder, CO 80303, USA; BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA; Howard Hughes Medical Institute, University of Colorado Boulder, Boulder, CO 80303, USA
| | - Teague C Dilgen
- Laboratory of Cell Biology and Genetics, The Rockefeller University, New York, NY 10065, USA
| | - Thomas R Cech
- Department of Biochemistry, University of Colorado Boulder, Boulder, CO 80303, USA; BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA; Howard Hughes Medical Institute, University of Colorado Boulder, Boulder, CO 80303, USA
| | - Thomas Walz
- Laboratory of Molecular Electron Microscopy, The Rockefeller University, New York, NY 10065, USA.
| | - Titia de Lange
- Laboratory of Cell Biology and Genetics, The Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|
12
|
Knowles S, Chai W. Conditional Depletion of STN1 in Mouse Embryonic Fibroblasts. Bio Protoc 2024; 14:e4977. [PMID: 38686350 PMCID: PMC11056013 DOI: 10.21769/bioprotoc.4977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 03/20/2024] [Accepted: 03/21/2024] [Indexed: 05/02/2024] Open
Abstract
The CTC1-STN1-TEN1 (CST) complex is a single-strand DNA-binding protein complex that plays an important role in genome maintenance in various model eukaryotes. Dysfunction of CST is the underlying cause of the rare genetic disorder known as Coats plus disease. In addition, down regulation of STN1 promotes colorectal cancer development in mice. While prior studies have utilized RNAi to knock down CST components in mammalian cells, this approach is associated with off-target effects. Attempts to employ CRISPR/Cas9-based knockout of CST components in somatic cell lines have been unsuccessful due to CST's indispensable role in DNA replication and cell proliferation. To address these challenges, we outline a novel approach utilizing a Cre-loxP-based conditional knockout in mouse embryonic fibroblasts (MEFs). This method offers an alternative means to investigate the function and characteristics of the CST complex in mammalian systems, potentially shedding new light on its roles in genome maintenance. Key features • Conditional depletion of mammalian STN1 using mouse embryonic fibroblast (MEFs). • Analysis of oxidative damage sensitivity using STN1-depleted MEFs. • This protocol requires Stn1flox/flox mice.
Collapse
Affiliation(s)
- Sara Knowles
- Department of Cancer Biology, Cardinal Bernardin Cancer Center, Loyola University Chicago Stritch School of Medicine, Maywood, IL, USA
- Center for Genetic Diseases, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Weihang Chai
- Department of Cancer Biology, Cardinal Bernardin Cancer Center, Loyola University Chicago Stritch School of Medicine, Maywood, IL, USA
- Center for Genetic Diseases, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| |
Collapse
|
13
|
Olson CL, Wuttke DS. Guardians of the Genome: How the Single-Stranded DNA-Binding Proteins RPA and CST Facilitate Telomere Replication. Biomolecules 2024; 14:263. [PMID: 38540683 PMCID: PMC10968030 DOI: 10.3390/biom14030263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/02/2024] [Accepted: 02/20/2024] [Indexed: 04/26/2024] Open
Abstract
Telomeres act as the protective caps of eukaryotic linear chromosomes; thus, proper telomere maintenance is crucial for genome stability. Successful telomere replication is a cornerstone of telomere length regulation, but this process can be fraught due to the many intrinsic challenges telomeres pose to the replication machinery. In addition to the famous "end replication" problem due to the discontinuous nature of lagging strand synthesis, telomeres require various telomere-specific steps for maintaining the proper 3' overhang length. Bulk telomere replication also encounters its own difficulties as telomeres are prone to various forms of replication roadblocks. These roadblocks can result in an increase in replication stress that can cause replication forks to slow, stall, or become reversed. Ultimately, this leads to excess single-stranded DNA (ssDNA) that needs to be managed and protected for replication to continue and to prevent DNA damage and genome instability. RPA and CST are single-stranded DNA-binding protein complexes that play key roles in performing this task and help stabilize stalled forks for continued replication. The interplay between RPA and CST, their functions at telomeres during replication, and their specialized features for helping overcome replication stress at telomeres are the focus of this review.
Collapse
Affiliation(s)
- Conner L. Olson
- Department of Biochemistry, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Deborah S. Wuttke
- Department of Biochemistry, University of Colorado Boulder, Boulder, CO 80309, USA
| |
Collapse
|
14
|
Lasho T, Patnaik MM. Adaptive and Maladaptive Clonal Hematopoiesis in Telomere Biology Disorders. Curr Hematol Malig Rep 2024; 19:35-44. [PMID: 38095828 DOI: 10.1007/s11899-023-00719-2] [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] [Accepted: 11/24/2023] [Indexed: 01/30/2024]
Abstract
PURPOSE OF REVIEW Telomere biology disorders (TBDs) are germline-inherited conditions characterized by reduction in telomerase function, accelerated shortening of telomeres, predisposition to organ-failure syndromes, and increased risk of neoplasms, especially myeloid malignancies. In normal cells, critically short telomeres trigger apoptosis and/or cellular senescence. However, the evolutionary mechanism by which TBD-related telomerase-deficient cells can overcome this fitness constraint remains elusive. RECENT FINDINGS Preliminary data suggests the existence of adaptive somatic mosaic states characterized by variants in TBD-related genes and maladaptive somatic mosaic states that attempt to overcome hematopoietic fitness constraints by alternative methods leading to clonal hematopoiesis. TBDs are both rare and highly heterogeneous in presentation, and the association of TBD with malignant transformation is unclear. Understanding the clonal complexity and mechanisms behind TBD-associated molecular signatures that lead to somatic adaptation in the setting of defective hematopoiesis will help inform therapy and treatment for this set of diseases.
Collapse
Affiliation(s)
- Terra Lasho
- Division of Hematology, Mayo Clinic Rochester, 200 First Street SW, Rochester, MN, 55905, USA
| | - Mrinal M Patnaik
- Division of Hematology, Mayo Clinic Rochester, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
15
|
Livingston JH. Childhood-inherited white matter disorders with calcification. HANDBOOK OF CLINICAL NEUROLOGY 2024; 204:95-109. [PMID: 39322397 DOI: 10.1016/b978-0-323-99209-1.00013-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
Intracranial calcification (ICC) occurs in many neurologic disorders both acquired and genetic. In some inherited white matter disorders, it is a common or even invariable feature where the presence and pattern of calcification provides an important pointer to the specific diagnosis. This is particularly the case for the Aicardi-Goutières syndrome (AGS) and for Coats plus (CP) and leukoencephalopathy with calcifications and cysts (LCC), which are discussed in detail in this chapter. AGS is a genetic disorder of type 1 interferon regulation, caused by mutations in any of the nine genes identified to date. In its classic form, AGS has very characteristic clinical and neuroimaging features which will be discussed here. LCC is a purely neurologic disorder caused by mutations in the SNORD118 gene, whereas CP is a multisystem disorder of telomere function that may result from mutations in the CTC1, POT1, or STN genes. In spite of the different pathogenetic basis for LCC and CP, they share remarkably similar neuroimaging and neuropathologic features. Cockayne syndrome, in which ICC is usually present, is discussed elsewhere in this volume. ICC may occur as an occasional feature of many other white matter diseases, including Alexander disease, Krabbe disease, X-ALD, and occulodentodigital dysplasia.
Collapse
Affiliation(s)
- John H Livingston
- Professor of Paediatric Neurology, University of Leeds, Leeds, United Kingdom; Department of Paediatric Neurology, Leeds Teaching Hospitals, Leeds, United Kingdom.
| |
Collapse
|
16
|
Ongie L, Raj HA, Stevens KB. Genetic Counseling and Family Screening Recommendations in Patients with Telomere Biology Disorders. Curr Hematol Malig Rep 2023; 18:273-283. [PMID: 37787873 DOI: 10.1007/s11899-023-00713-8] [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] [Accepted: 09/11/2023] [Indexed: 10/04/2023]
Abstract
PURPOSE OF REVIEW Telomere biology disorders (TBDs) encompass a spectrum of genetic diseases with a common pathogenesis of defects in telomerase function and telomere maintenance causing extremely short telomere lengths. Here, we review the current literature surrounding genetic testing strategies, cascade testing, reproductive implications, and the role of genetic counseling. RECENT FINDINGS The understanding of the genetic causes and clinical symptoms of TBDs continues to expand while genetic testing and telomere length testing are nuanced tools utilized in the diagnosis of this condition. Access to genetic counseling is becoming more abundant and is valuable in supporting patients and their families in making informed decisions. Patient resources and support groups are valuable to this community. Defining which populations should be offered genetic counseling and testing is imperative to provide proper diagnoses and medical management for not only the primary patient, but also their biological relatives.
Collapse
Affiliation(s)
| | - Hannah A Raj
- Team Telomere, Inc., New York, NY, USA
- College of Medicine, University of Illinois, Chicago, IL, USA
| | | |
Collapse
|
17
|
Jaiswal RK, Lei KH, Chastain M, Wang Y, Shiva O, Li S, You Z, Chi P, Chai W. CaMKK2 and CHK1 phosphorylate human STN1 in response to replication stress to protect stalled forks from aberrant resection. Nat Commun 2023; 14:7882. [PMID: 38036565 PMCID: PMC10689503 DOI: 10.1038/s41467-023-43685-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 11/16/2023] [Indexed: 12/02/2023] Open
Abstract
Keeping replication fork stable is essential for safeguarding genome integrity; hence, its protection is highly regulated. The CTC1-STN1-TEN1 (CST) complex protects stalled forks from aberrant MRE11-mediated nascent strand DNA degradation (NSD). However, the activation mechanism for CST at forks is unknown. Here, we report that STN1 is phosphorylated in its intrinsic disordered region. Loss of STN1 phosphorylation reduces the replication stress-induced STN1 localization to stalled forks, elevates NSD, increases MRE11 access to stalled forks, and decreases RAD51 localization at forks, leading to increased genome instability under perturbed DNA replication condition. STN1 is phosphorylated by both the ATR-CHK1 and the calcium-sensing kinase CaMKK2 in response to hydroxyurea/aphidicolin treatment or elevated cytosolic calcium concentration. Cancer-associated STN1 variants impair STN1 phosphorylation, conferring inability of fork protection. Collectively, our study uncovers that CaMKK2 and ATR-CHK1 target STN1 to enable its fork protective function, and suggests an important role of STN1 phosphorylation in cancer development.
Collapse
Affiliation(s)
- Rishi Kumar Jaiswal
- Department of Cancer Biology, Cardinal Bernardin Cancer Center, Loyola University Chicago Stritch School of Medicine, Maywood, IL, USA
| | - Kai-Hang Lei
- Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| | - Megan Chastain
- Office of Research, Washington State University, Spokane, WA, USA
| | - Yuan Wang
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Olga Shiva
- Office of Research, Washington State University, Spokane, WA, USA
| | - Shan Li
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Zhongsheng You
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Peter Chi
- Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Weihang Chai
- Department of Cancer Biology, Cardinal Bernardin Cancer Center, Loyola University Chicago Stritch School of Medicine, Maywood, IL, USA.
| |
Collapse
|
18
|
Liao P, Yan B, Wang C, Lei P. Telomeres: Dysfunction, Maintenance, Aging and Cancer. Aging Dis 2023; 15:2595-2631. [PMID: 38270117 PMCID: PMC11567242 DOI: 10.14336/ad.2023.1128] [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: 10/17/2023] [Accepted: 11/28/2023] [Indexed: 01/26/2024] Open
Abstract
Aging has emerged at the forefront of scientific research due to the growing social and economic costs associated with the growing aging global population. The defining features of aging involve a variety of molecular processes and cellular systems, which are interconnected and collaboratively contribute to the aging process. Herein, we analyze how telomere dysfunction potentially amplifies or accelerates the molecular and biochemical mechanisms underpinning each feature of aging and contributes to the emergence of age-associated illnesses, including cancer and neurodegeneration, via the perspective of telomere biology. Furthermore, the recently identified novel mechanistic actions for telomere maintenance offer a fresh viewpoint and approach to the management of telomeres and associated disorders. Telomeres and the defining features of aging are intimately related, which has implications for therapeutic and preventive approaches to slow aging and reduce the prevalence of age-related disorders.
Collapse
Affiliation(s)
- Pan Liao
- The School of Medicine, Nankai University, Tianjin, China.
- Haihe Laboratory of Cell Ecosystem, Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China.
- Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin, China.
| | - Bo Yan
- Haihe Laboratory of Cell Ecosystem, Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China.
- Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin, China.
| | - Conglin Wang
- Haihe Laboratory of Cell Ecosystem, Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China.
- Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin, China.
| | - Ping Lei
- The School of Medicine, Nankai University, Tianjin, China.
- Haihe Laboratory of Cell Ecosystem, Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China.
- Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin, China.
| |
Collapse
|
19
|
Kayarian FB, Cohen SM, Cohen ML, Sammartino DE. Coats Plus Syndrome Presenting in an Adult. JOURNAL OF VITREORETINAL DISEASES 2023; 7:562-564. [PMID: 37974921 PMCID: PMC10649444 DOI: 10.1177/24741264231171465] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
Abstract
Purpose: To present a case of retinal vascular disease characterized primarily by capillary nonperfusion in an adult with Coats plus syndrome (CPS). Methods: A case and its findings were analyzed. Results: A 38-year-old woman with a history of poliosis, thrombocytopenia, seizures, and white-matter brain lesions was referred for evaluation of bilateral blurred central vision. Fluorescein angiography showed extensive bilateral retinal capillary nonperfusion with retinal arteriolitis in the right eye. Genetic testing found 2 pathological mutations in the conserved telomere maintenance component 1 (CTC1) gene, diagnostic of CPS. Conclusions: Genetic testing may be diagnostic in patients who present with retinal vascular disease and systemic disease suggestive of CPS.
Collapse
Affiliation(s)
- Fae B. Kayarian
- Rush Medical College of Rush University Medical Center, Chicago, IL, USA
| | - Steven M. Cohen
- University of South Florida, Tampa, FL, USA
- Retina Vitreous Associates of Florida, Clearwater, FL, USA
| | - Mark L. Cohen
- National Prion Disease Pathology Surveillance Center, Case Western Reserve University, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | | |
Collapse
|
20
|
Li T, Zhang M, Li Y, Han X, Tang L, Ma T, Zhao X, Zhao R, Wang Y, Bai X, Zhang K, Geng X, Sui L, Feng X, Zhang Q, Zhao Y, Liu Y, Stewart JA, Wang F. Cooperative interaction of CST and RECQ4 resolves G-quadruplexes and maintains telomere stability. EMBO Rep 2023; 24:e55494. [PMID: 37493024 PMCID: PMC10481657 DOI: 10.15252/embr.202255494] [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: 05/26/2022] [Revised: 07/06/2023] [Accepted: 07/14/2023] [Indexed: 07/27/2023] Open
Abstract
Human CST (CTC1-STN1-TEN1) is a ssDNA-binding complex that interacts with the replisome to aid in stalled fork rescue. We previously found that CST promotes telomere replication to maintain genomic integrity via G-quadruplex (G4) resolution. However, the detailed mechanism by which CST resolves G4s in vivo and whether additional factors are involved remains unclear. Here, we identify RECQ4 as a novel CST-interacting partner and show that RECQ4 can unwind G4 structures in vitro using a FRET assay. Moreover, G4s accumulate at the telomere after RECQ4 depletion, resulting in telomere dysfunction, including the formation of MTSs, SFEs, and TIFs, suggesting that RECQ4 is crucial for telomere integrity. Furthermore, CST is also required for RECQ4 telomere or chromatin localization in response to G4 stabilizers. RECQ4 is involved in preserving genomic stability by CST and RECQ4 disruption impairs restart of replication forks stalled by G4s. Overall, our findings highlight the essential roles of CST and RECQ4 in resolving G-rich regions, where they collaborate to resolve G4-induced replication deficiencies and maintain genomic homeostasis.
Collapse
Affiliation(s)
- Tingfang Li
- Department of Genetics, School of Basic Medical Sciences & The Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical Epigenetics, Geriatrics Institute General Hospital, School and Hospital of StomatologyTianjin Medical UniversityTianjinChina
| | - Miaomiao Zhang
- Medical Research CenterAffiliated Hospital of Jining Medical UniversityJiningChina
| | - Yanjing Li
- Department of Prosthodontics, School and Hospital of StomatologyTianjin Medical UniversityTianjinChina
| | - Xinyu Han
- Department of Genetics, School of Basic Medical Sciences & The Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical Epigenetics, Geriatrics Institute General Hospital, School and Hospital of StomatologyTianjin Medical UniversityTianjinChina
| | - Lu Tang
- Department of Prosthodontics, School and Hospital of StomatologyTianjin Medical UniversityTianjinChina
| | - Tengfei Ma
- Institute of Precision MedicineThe First Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Xiaotong Zhao
- Department of Radiobiology, Institute of Radiation MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Rui Zhao
- Department of Genetics, School of Basic Medical Sciences & The Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical Epigenetics, Geriatrics Institute General Hospital, School and Hospital of StomatologyTianjin Medical UniversityTianjinChina
| | - Yuwen Wang
- Department of Genetics, School of Basic Medical Sciences & The Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical Epigenetics, Geriatrics Institute General Hospital, School and Hospital of StomatologyTianjin Medical UniversityTianjinChina
| | - Xue Bai
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences & The Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical EpigeneticsTianjin Medical UniversityTianjinChina
| | - Kai Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences & The Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical EpigeneticsTianjin Medical UniversityTianjinChina
| | - Xin Geng
- Department of Biochemistry and Molecular Biology, School of Basic Medical SciencesTianjin Medical UniversityTianjinChina
| | - Lei Sui
- Department of Prosthodontics, School and Hospital of StomatologyTianjin Medical UniversityTianjinChina
| | - Xuyang Feng
- Institute of Precision MedicineThe First Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Qiang Zhang
- Department of Geriatrics, Tianjin Medical University General HospitalTianjin Geriatrics InstituteTianjinChina
| | - Yang Zhao
- Department of Radiology, Tianjin Institute of UrologyThe Second Hospital of Tianjin Medical UniversityTianjinChina
| | - Yang Liu
- Department of Radiobiology, Institute of Radiation MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Jason A Stewart
- Department of BiologyWestern Kentucky UniversityBowling GreenKYUSA
- Department of Biological SciencesUniversity of South CarolinaColumbiaSCUSA
| | - Feng Wang
- Department of Genetics, School of Basic Medical Sciences & The Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical Epigenetics, Geriatrics Institute General Hospital, School and Hospital of StomatologyTianjin Medical UniversityTianjinChina
| |
Collapse
|
21
|
Abstract
It has been known for decades that telomerase extends the 3' end of linear eukaryotic chromosomes and dictates the telomeric repeat sequence based on the template in its RNA. However, telomerase does not mitigate sequence loss at the 5' ends of chromosomes, which results from lagging strand DNA synthesis and nucleolytic processing. Therefore, a second enzyme is needed to keep telomeres intact: DNA polymerase α/Primase bound to Ctc1-Stn1-Ten1 (CST). CST-Polα/Primase maintains telomeres through a fill-in reaction that replenishes the lost sequences at the 5' ends. CST not only serves to maintain telomeres but also determines their length by keeping telomerase from overelongating telomeres. Here we discuss recent data on the evolution, structure, function, and recruitment of mammalian CST-Polα/Primase, highlighting the role of this complex and telomere length control in human disease.
Collapse
Affiliation(s)
- Sarah W Cai
- Laboratory for Cell Biology and Genetics, The Rockefeller University, New York, New York 10065, USA
| | - Titia de Lange
- Laboratory for Cell Biology and Genetics, The Rockefeller University, New York, New York 10065, USA
| |
Collapse
|
22
|
Vaurs M, Naiman K, Bouabboune C, Rai S, Ptasińska K, Rives M, Matmati S, Carr AM, Géli V, Coulon S. Stn1-Ten1 and Taz1 independently promote replication of subtelomeric fragile sequences in fission yeast. Cell Rep 2023; 42:112537. [PMID: 37243596 DOI: 10.1016/j.celrep.2023.112537] [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: 09/15/2022] [Revised: 03/01/2023] [Accepted: 05/03/2023] [Indexed: 05/29/2023] Open
Abstract
Efficient replication of terminal DNA is crucial to maintain telomere stability. In fission yeast, Taz1 and the Stn1-Ten1 (ST) complex play prominent roles in DNA-ends replication. However, their function remains elusive. Here, we have analyzed genome-wide replication and show that ST does not affect genome-wide replication but is crucial for the efficient replication of a subtelomeric region called STE3-2. We further show that, when ST function is compromised, a homologous recombination (HR)-based fork restart mechanism becomes necessary for STE3-2 stability. While both Taz1 and Stn1 bind to STE3-2, we find that the STE3-2 replication function of ST is independent of Taz1 but relies on its association with the shelterin proteins Pot1-Tpz1-Poz1. Finally, we demonstrate that the firing of an origin normally inhibited by Rif1 can circumvent the replication defect of subtelomeres when ST function is compromised. Our results help illuminate why fission yeast telomeres are terminal fragile sites.
Collapse
Affiliation(s)
- Mélina Vaurs
- CNRS, INSERM, Aix Marseille Univ, Institut Paoli-Calmettes, CRCM, Ligue Nationale Contre le Cancer (équipe labellisée), Marseille, France
| | - Karel Naiman
- CNRS, INSERM, Aix Marseille Univ, Institut Paoli-Calmettes, CRCM, Ligue Nationale Contre le Cancer (équipe labellisée), Marseille, France; Genome Damage and Stability Centre, School of Life Sciences, University of Sussex, Falmer BN1 9RQ, UK
| | - Chaïnez Bouabboune
- CNRS, INSERM, Aix Marseille Univ, Institut Paoli-Calmettes, CRCM, Ligue Nationale Contre le Cancer (équipe labellisée), Marseille, France
| | - Sudhir Rai
- CNRS, INSERM, Aix Marseille Univ, Institut Paoli-Calmettes, CRCM, Ligue Nationale Contre le Cancer (équipe labellisée), Marseille, France
| | - Katarzyna Ptasińska
- Genome Damage and Stability Centre, School of Life Sciences, University of Sussex, Falmer BN1 9RQ, UK
| | - Marion Rives
- CNRS, INSERM, Aix Marseille Univ, Institut Paoli-Calmettes, CRCM, Ligue Nationale Contre le Cancer (équipe labellisée), Marseille, France
| | - Samah Matmati
- CNRS, INSERM, Aix Marseille Univ, Institut Paoli-Calmettes, CRCM, Ligue Nationale Contre le Cancer (équipe labellisée), Marseille, France
| | - Antony M Carr
- Genome Damage and Stability Centre, School of Life Sciences, University of Sussex, Falmer BN1 9RQ, UK
| | - Vincent Géli
- CNRS, INSERM, Aix Marseille Univ, Institut Paoli-Calmettes, CRCM, Ligue Nationale Contre le Cancer (équipe labellisée), Marseille, France.
| | - Stéphane Coulon
- CNRS, INSERM, Aix Marseille Univ, Institut Paoli-Calmettes, CRCM, Ligue Nationale Contre le Cancer (équipe labellisée), Marseille, France.
| |
Collapse
|
23
|
Vijay Kumar MJ, Morales R, Tsvetkov AS. G-quadruplexes and associated proteins in aging and Alzheimer's disease. FRONTIERS IN AGING 2023; 4:1164057. [PMID: 37323535 PMCID: PMC10267416 DOI: 10.3389/fragi.2023.1164057] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 05/17/2023] [Indexed: 06/17/2023]
Abstract
Aging is a prominent risk factor for many neurodegenerative disorders, such as Alzheimer's disease (AD). Alzheimer's disease is characterized by progressive cognitive decline, memory loss, and neuropsychiatric and behavioral symptoms, accounting for most of the reported dementia cases. This disease is now becoming a major challenge and burden on modern society, especially with the aging population. Over the last few decades, a significant understanding of the pathophysiology of AD has been gained by studying amyloid deposition, hyperphosphorylated tau, synaptic dysfunction, oxidative stress, calcium dysregulation, and neuroinflammation. This review focuses on the role of non-canonical secondary structures of DNA/RNA G-quadruplexes (G4s, G4-DNA, and G4-RNA), G4-binding proteins (G4BPs), and helicases, and their roles in aging and AD. Being critically important for cellular function, G4s are involved in the regulation of DNA and RNA processes, such as replication, transcription, translation, RNA localization, and degradation. Recent studies have also highlighted G4-DNA's roles in inducing DNA double-strand breaks that cause genomic instability and G4-RNA's participation in regulating stress granule formation. This review emphasizes the significance of G4s in aging processes and how their homeostatic imbalance may contribute to the pathophysiology of AD.
Collapse
Affiliation(s)
- M. J. Vijay Kumar
- The Department of Neurology, The University of Texas McGovern Medical School at Houston, Houston, TX, United States
| | - Rodrigo Morales
- The Department of Neurology, The University of Texas McGovern Medical School at Houston, Houston, TX, United States
- Centro Integrativo de Biologia y Quimica Aplicada (CIBQA), Universidad Bernardo O’Higgins, Santiago, Chile
| | - Andrey S. Tsvetkov
- The Department of Neurology, The University of Texas McGovern Medical School at Houston, Houston, TX, United States
- The University of Texas Graduate School of Biomedical Sciences, Houston, TX, United States
- UTHealth Consortium on Aging, The University of Texas McGovern Medical School, Houston, TX, United States
| |
Collapse
|
24
|
Nguyen DD, Kim E, Le NT, Ding X, Jaiswal RK, Kostlan RJ, Nguyen TNT, Shiva O, Le MT, Chai W. Deficiency in mammalian STN1 promotes colon cancer development via inhibiting DNA repair. SCIENCE ADVANCES 2023; 9:eadd8023. [PMID: 37163605 PMCID: PMC10171824 DOI: 10.1126/sciadv.add8023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 04/05/2023] [Indexed: 05/12/2023]
Abstract
Despite the high lethality of colorectal cancers (CRCs), only a limited number of genetic risk factors are identified. The mammalian ssDNA-binding protein complex CTC1-STN1-TEN1 protects genome stability, yet its role in tumorigenesis is unknown. Here, we show that attenuated CTC1/STN1 expression is common in CRCs. We generated an inducible STN1 knockout mouse model and found that STN1 deficiency in young adult mice increased CRC incidence, tumor size, and tumor load. CRC tumors exhibited enhanced proliferation, reduced apoptosis, and elevated DNA damage and replication stress. We found that STN1 deficiency down-regulated multiple DNA glycosylases, resulting in defective base excision repair (BER) and accumulation of oxidative damage. Collectively, this study identifies STN1 deficiency as a risk factor for CRC and implicates the previously unknown STN1-BER axis in protecting colon tissues from oxidative damage, therefore providing insights into the CRC tumor-suppressing mechanism.
Collapse
Affiliation(s)
- Dinh Duc Nguyen
- Department of Cancer Biology, Cardinal Bernardin Cancer Center, Loyola University Chicago Stritch School of Medicine, Maywood, IL, USA
| | - Eugene Kim
- Department of Cancer Biology, Cardinal Bernardin Cancer Center, Loyola University Chicago Stritch School of Medicine, Maywood, IL, USA
| | - Nhat Thong Le
- School of Biotechnology, International University, Ho Chi Minh City, Vietnam
| | - Xianzhong Ding
- Department of Pathology, Loyola University Chicago Stritch School of Medicine, Maywood, IL, USA
| | - Rishi Kumar Jaiswal
- Department of Cancer Biology, Cardinal Bernardin Cancer Center, Loyola University Chicago Stritch School of Medicine, Maywood, IL, USA
| | - Raymond Joseph Kostlan
- Department of Cancer Biology, Cardinal Bernardin Cancer Center, Loyola University Chicago Stritch School of Medicine, Maywood, IL, USA
| | - Thi Ngoc Thanh Nguyen
- Department of Cancer Biology, Cardinal Bernardin Cancer Center, Loyola University Chicago Stritch School of Medicine, Maywood, IL, USA
| | - Olga Shiva
- Office of Research, Washington State University-Spokane, Spokane, WA, USA
| | - Minh Thong Le
- School of Biotechnology, International University, Ho Chi Minh City, Vietnam
| | - Weihang Chai
- Department of Cancer Biology, Cardinal Bernardin Cancer Center, Loyola University Chicago Stritch School of Medicine, Maywood, IL, USA
| |
Collapse
|
25
|
Duperron MG, Knol MJ, Le Grand Q, Evans TE, Mishra A, Tsuchida A, Roshchupkin G, Konuma T, Trégouët DA, Romero JR, Frenzel S, Luciano M, Hofer E, Bourgey M, Dueker ND, Delgado P, Hilal S, Tankard RM, Dubost F, Shin J, Saba Y, Armstrong NJ, Bordes C, Bastin ME, Beiser A, Brodaty H, Bülow R, Carrera C, Chen C, Cheng CY, Deary IJ, Gampawar PG, Himali JJ, Jiang J, Kawaguchi T, Li S, Macalli M, Marquis P, Morris Z, Muñoz Maniega S, Miyamoto S, Okawa M, Paradise M, Parva P, Rundek T, Sargurupremraj M, Schilling S, Setoh K, Soukarieh O, Tabara Y, Teumer A, Thalamuthu A, Trollor JN, Valdés Hernández MC, Vernooij MW, Völker U, Wittfeld K, Wong TY, Wright MJ, Zhang J, Zhao W, Zhu YC, Schmidt H, Sachdev PS, Wen W, Yoshida K, Joutel A, Satizabal CL, Sacco RL, Bourque G, Lathrop M, Paus T, Fernandez-Cadenas I, Yang Q, Mazoyer B, Boutinaud P, Okada Y, Grabe HJ, Mather KA, Schmidt R, Joliot M, Ikram MA, Matsuda F, Tzourio C, Wardlaw JM, Seshadri S, Adams HHH, Debette S. Genomics of perivascular space burden unravels early mechanisms of cerebral small vessel disease. Nat Med 2023; 29:950-962. [PMID: 37069360 PMCID: PMC10115645 DOI: 10.1038/s41591-023-02268-w] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 02/15/2023] [Indexed: 04/19/2023]
Abstract
Perivascular space (PVS) burden is an emerging, poorly understood, magnetic resonance imaging marker of cerebral small vessel disease, a leading cause of stroke and dementia. Genome-wide association studies in up to 40,095 participants (18 population-based cohorts, 66.3 ± 8.6 yr, 96.9% European ancestry) revealed 24 genome-wide significant PVS risk loci, mainly in the white matter. These were associated with white matter PVS already in young adults (N = 1,748; 22.1 ± 2.3 yr) and were enriched in early-onset leukodystrophy genes and genes expressed in fetal brain endothelial cells, suggesting early-life mechanisms. In total, 53% of white matter PVS risk loci showed nominally significant associations (27% after multiple-testing correction) in a Japanese population-based cohort (N = 2,862; 68.3 ± 5.3 yr). Mendelian randomization supported causal associations of high blood pressure with basal ganglia and hippocampal PVS, and of basal ganglia PVS and hippocampal PVS with stroke, accounting for blood pressure. Our findings provide insight into the biology of PVS and cerebral small vessel disease, pointing to pathways involving extracellular matrix, membrane transport and developmental processes, and the potential for genetically informed prioritization of drug targets.
Collapse
Affiliation(s)
- Marie-Gabrielle Duperron
- Bordeaux Population Health Research Center, UMR 1219, University of Bordeaux, Inserm, Bordeaux, France
- Department of Neurology, Institute of Neurodegenerative Diseases, Bordeaux University Hospital, Bordeaux, France
| | - Maria J Knol
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Quentin Le Grand
- Bordeaux Population Health Research Center, UMR 1219, University of Bordeaux, Inserm, Bordeaux, France
| | - Tavia E Evans
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, the Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Aniket Mishra
- Bordeaux Population Health Research Center, UMR 1219, University of Bordeaux, Inserm, Bordeaux, France
| | - Ami Tsuchida
- Bordeaux Population Health Research Center, UMR 1219, University of Bordeaux, Inserm, Bordeaux, France
- Groupe d'Imagerie Neurofonctionelle - Institut des maladies neurodégénératives (GIN-IMN), UMR 5293, University of Bordeaux, CNRS, CEA, Bordeaux, France
| | - Gennady Roshchupkin
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Takahiro Konuma
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
| | - David-Alexandre Trégouët
- Bordeaux Population Health Research Center, UMR 1219, University of Bordeaux, Inserm, Bordeaux, France
| | - Jose Rafael Romero
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- The Framingham Heart Study, Framingham, MA, USA
| | - Stefan Frenzel
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | | | - Edith Hofer
- Clinical Division of Neurogeriatrics, Department of Neurology, Medical University of Graz, Graz, Austria
- Institute for Medical Informatics, Statistics and Documentation, Medical University of Graz, Graz, Austria
| | - Mathieu Bourgey
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
- Victor Phillip Dahdaleh Institute of Genomic Medicine at McGill University, Montreal, Quebec, Canada
- Canadian Centre for Computational Genomics, McGill University, Montreal, Quebec, Canada
| | - Nicole D Dueker
- John P. Hussman Institute for Human Genomics, University of Miami, Miami, FL, USA
| | - Pilar Delgado
- Institut de Recerca Vall d'hebron, Neurovascular Research Lab, Universitat Autònoma de Barcelona, Barcelona, Spain
- Hospital Universitari Vall d'Hebron, Neurology Department, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Saima Hilal
- Memory Aging and Cognition Center, National University Health System, Singapore, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Singapore
| | - Rick M Tankard
- Department of Mathematics and Statistics, Curtin University, Perth, Western Australia, Australia
| | - Florian Dubost
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, the Netherlands
- Department of Medical Informatics, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Jean Shin
- The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
- Departments of Physiology and Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Yasaman Saba
- Bordeaux Population Health Research Center, UMR 1219, University of Bordeaux, Inserm, Bordeaux, France
- Institute for Molecular Biology & Biochemistry, Gottfried Schatz Research Center (for Cell Signaling, Metabolism and Aging), Medical University of Graz, Graz, Austria
| | - Nicola J Armstrong
- Department of Mathematics and Statistics, Curtin University, Perth, Western Australia, Australia
| | - Constance Bordes
- Bordeaux Population Health Research Center, UMR 1219, University of Bordeaux, Inserm, Bordeaux, France
| | - Mark E Bastin
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Alexa Beiser
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- The Framingham Heart Study, Framingham, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Henry Brodaty
- Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry & Mental Health, University of New South Wales, Sydney, New South Wales, Australia
- Dementia Collaborative Research Centre Assessment and Better Care, UNSW, Sydney, New South Wales, Australia
| | - Robin Bülow
- Institute for Radiology and Neuroradiology, University Medicine Greifswald, Greifswald, Germany
| | - Caty Carrera
- Stroke Pharmacogenomics and Genetics Group, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
| | - Christopher Chen
- Memory Aging and Cognition Center, National University Health System, Singapore, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Ching-Yu Cheng
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Center for Innovation and Precision Eye Health, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Ophthalmology & Visual Sciences Academic Clinical Program, Duke-NUS Medical School, Singapore, Singapore
| | - Ian J Deary
- School of Psychology, University of Edinburgh, Edinburgh, UK
| | - Piyush G Gampawar
- Institute for Molecular Biology & Biochemistry, Gottfried Schatz Research Center (for Cell Signaling, Metabolism and Aging), Medical University of Graz, Graz, Austria
| | - Jayandra J Himali
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- The Framingham Heart Study, Framingham, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, UT Health San Antonio, San Antonio, TX, USA
- Department of Population Health Sciences, UT Health San Antonio, San Antonio, TX, USA
| | - Jiyang Jiang
- Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry & Mental Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Takahisa Kawaguchi
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Shuo Li
- The Framingham Heart Study, Framingham, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Melissa Macalli
- Bordeaux Population Health Research Center, UMR 1219, University of Bordeaux, Inserm, Bordeaux, France
| | - Pascale Marquis
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
- Victor Phillip Dahdaleh Institute of Genomic Medicine at McGill University, Montreal, Quebec, Canada
- Canadian Centre for Computational Genomics, McGill University, Montreal, Quebec, Canada
| | - Zoe Morris
- Neuroimaging, Department of Clinical Neurosciences, Royal Infirmary of Edinburgh, Edinburgh, UK
| | - Susana Muñoz Maniega
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute Centre at the University of Edinburgh, Edinburgh, UK
| | | | - Masakazu Okawa
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Matthew Paradise
- Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry & Mental Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Pedram Parva
- The Framingham Heart Study, Framingham, MA, USA
- Radiology Department, Boston University School of Medicine, Boston, MA, USA
- Department of Radiology, Harvard Medical School, Boston, MA, USA
| | - Tatjana Rundek
- Department of Neurology, Miller School of Medicine, University of Miami, Miami, FL, USA
- Evelyn F. McKnight Brain Institute, Department of Neurology, University of Miami, Miami, FL, USA
| | | | - Sabrina Schilling
- Bordeaux Population Health Research Center, UMR 1219, University of Bordeaux, Inserm, Bordeaux, France
| | - Kazuya Setoh
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Graduate School of Public Health, Shizuoka Graduate University of Public Health, Shizuoka, Japan
| | - Omar Soukarieh
- Bordeaux Population Health Research Center, UMR 1219, University of Bordeaux, Inserm, Bordeaux, France
| | - Yasuharu Tabara
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Graduate School of Public Health, Shizuoka Graduate University of Public Health, Shizuoka, Japan
| | - Alexander Teumer
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Anbupalam Thalamuthu
- Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry & Mental Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Julian N Trollor
- Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry & Mental Health, University of New South Wales, Sydney, New South Wales, Australia
- Department of Developmental Disability Neuropsychiatry, UNSW, Sydney, New South Wales, Australia
| | - Maria C Valdés Hernández
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- Row Fogo Centre for Research into Ageing and the Brain, University of Edinburgh, Edinburgh, UK
| | - Meike W Vernooij
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Katharina Wittfeld
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
- German Center for Neurodegenerative Diseases (DZNE), Site Rostock/Greifswald, Greifswald, Germany
| | - Tien Yin Wong
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Tsinghua Medicine, Tsinghua University, Beijing, China
| | - Margaret J Wright
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia
- Centre for Advanced Imaging, University of Queensland, Brisbane, Queensland, Australia
| | - Junyi Zhang
- Department of Neurology, Peking Union Medical College Hospital, Beijing, China
| | - Wanting Zhao
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- The Centre for Quantitative Medicine, Duke-NUS Medical School, Singapore, Singapore
| | - Yi-Cheng Zhu
- Department of Neurology, Peking Union Medical College Hospital, Beijing, China
| | - Helena Schmidt
- Institute for Molecular Biology & Biochemistry, Gottfried Schatz Research Center (for Cell Signaling, Metabolism and Aging), Medical University of Graz, Graz, Austria
| | - Perminder S Sachdev
- Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry & Mental Health, University of New South Wales, Sydney, New South Wales, Australia
- Neuropsychiatric Institute, the Prince of Wales Hospital, Sydney, New South Wales, Australia
| | - Wei Wen
- Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry & Mental Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Kazumichi Yoshida
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Anne Joutel
- Institut de Psychiatrie et Neurosciences de Paris, Université Paris Cité, Inserm, France
| | - Claudia L Satizabal
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- The Framingham Heart Study, Framingham, MA, USA
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, UT Health San Antonio, San Antonio, TX, USA
- Department of Population Health Sciences, UT Health San Antonio, San Antonio, TX, USA
| | - Ralph L Sacco
- Department of Neurology, Miller School of Medicine, University of Miami, Miami, FL, USA
- Evelyn F. McKnight Brain Institute, Department of Neurology, University of Miami, Miami, FL, USA
- Department of Public Health Sciences, Miller School of Medicine, University of Miami, Miami, FL, USA
- Department of Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, USA
- Department of Neurosurgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Guillaume Bourque
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
- Victor Phillip Dahdaleh Institute of Genomic Medicine at McGill University, Montreal, Quebec, Canada
- Canadian Centre for Computational Genomics, McGill University, Montreal, Quebec, Canada
| | - Mark Lathrop
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
- Victor Phillip Dahdaleh Institute of Genomic Medicine at McGill University, Montreal, Quebec, Canada
| | - Tomas Paus
- University of Montreal, Faculty of Medicine, Departments of Psychiatry and Neuroscience, Montreal, Quebec, Canada
- Department of Psychology, University of Toronto, Toronto, Ontario, Canada
- Centre Hospitalier Universitaire Sainte Justine, Montreal, Quebec, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Israel Fernandez-Cadenas
- Stroke Pharmacogenomics and Genetics Group, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
- Stroke Pharmacogenomics and Genetics Group, Fundació per la Docència i la Recerca Mutua Terrassa, Terrassa, Spain
| | - Qiong Yang
- The Framingham Heart Study, Framingham, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Bernard Mazoyer
- Groupe d'Imagerie Neurofonctionelle - Institut des maladies neurodégénératives (GIN-IMN), UMR 5293, University of Bordeaux, CNRS, CEA, Bordeaux, France
- Bordeaux University Hospital, Bordeaux, France
| | | | - Yukinori Okada
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Japan
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, Japan
- Department of Genome Informatics, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Hans J Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
- German Center for Neurodegenerative Diseases (DZNE), Site Rostock/Greifswald, Greifswald, Germany
| | - Karen A Mather
- Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry & Mental Health, University of New South Wales, Sydney, New South Wales, Australia
- Neuroscience Research Australia, Sydney, New South Wales, Australia
| | - Reinhold Schmidt
- Clinical Division of Neurogeriatrics, Department of Neurology, Medical University of Graz, Graz, Austria
| | - Marc Joliot
- Groupe d'Imagerie Neurofonctionelle - Institut des maladies neurodégénératives (GIN-IMN), UMR 5293, University of Bordeaux, CNRS, CEA, Bordeaux, France
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Fumihiko Matsuda
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Christophe Tzourio
- Bordeaux Population Health Research Center, UMR 1219, University of Bordeaux, Inserm, Bordeaux, France
- Department of Medical Informatics, Bordeaux University Hospital, Bordeaux, France
| | - Joanna M Wardlaw
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute Centre at the University of Edinburgh, Edinburgh, UK
- Row Fogo Centre for Research into Ageing and the Brain, University of Edinburgh, Edinburgh, UK
| | - Sudha Seshadri
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- The Framingham Heart Study, Framingham, MA, USA
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, UT Health San Antonio, San Antonio, TX, USA
- Department of Population Health Sciences, UT Health San Antonio, San Antonio, TX, USA
| | - Hieab H H Adams
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, the Netherlands.
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, the Netherlands.
- Latin American Brain Health (BrainLat), Universidad Adolfo Ibáñez, Santiago, Chile.
| | - Stéphanie Debette
- Bordeaux Population Health Research Center, UMR 1219, University of Bordeaux, Inserm, Bordeaux, France.
- Department of Neurology, Institute of Neurodegenerative Diseases, Bordeaux University Hospital, Bordeaux, France.
| |
Collapse
|
26
|
Basheer F, Sertori R, Liongue C, Ward AC. Zebrafish: A Relevant Genetic Model for Human Primary Immunodeficiency (PID) Disorders? Int J Mol Sci 2023; 24:ijms24076468. [PMID: 37047441 PMCID: PMC10095346 DOI: 10.3390/ijms24076468] [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: 03/06/2023] [Revised: 03/28/2023] [Accepted: 03/28/2023] [Indexed: 04/14/2023] Open
Abstract
Primary immunodeficiency (PID) disorders, also commonly referred to as inborn errors of immunity, are a heterogenous group of human genetic diseases characterized by defects in immune cell development and/or function. Since these disorders are generally uncommon and occur on a variable background profile of potential genetic and environmental modifiers, animal models are critical to provide mechanistic insights as well as to create platforms to underpin therapeutic development. This review aims to review the relevance of zebrafish as an alternative genetic model for PIDs. It provides an overview of the conservation of the zebrafish immune system and details specific examples of zebrafish models for a multitude of specific human PIDs across a range of distinct categories, including severe combined immunodeficiency (SCID), combined immunodeficiency (CID), multi-system immunodeficiency, autoinflammatory disorders, neutropenia and defects in leucocyte mobility and respiratory burst. It also describes some of the diverse applications of these models, particularly in the fields of microbiology, immunology, regenerative biology and oncology.
Collapse
Affiliation(s)
- Faiza Basheer
- School of Medicine, Deakin University, Geelong, VIC 3216, Australia
- Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, VIC 3216, Australia
| | - Robert Sertori
- School of Medicine, Deakin University, Geelong, VIC 3216, Australia
| | - Clifford Liongue
- School of Medicine, Deakin University, Geelong, VIC 3216, Australia
- Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, VIC 3216, Australia
| | - Alister C Ward
- School of Medicine, Deakin University, Geelong, VIC 3216, Australia
- Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, VIC 3216, Australia
| |
Collapse
|
27
|
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
|
28
|
Mirman Z, Cai S, de Lange T. CST/Polα/primase-mediated fill-in synthesis at DSBs. Cell Cycle 2023; 22:379-389. [PMID: 36205622 PMCID: PMC9879193 DOI: 10.1080/15384101.2022.2123886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 08/27/2022] [Accepted: 09/06/2022] [Indexed: 01/29/2023] Open
Abstract
DNA double-strand breaks (DSBs) pose a major threat to the genome, so the efficient repair of such breaks is essential. DSB processing and repair is affected by 53BP1, which has been proposed to determine repair pathway choice and/or promote repair fidelity. 53BP1 and its downstream effectors, RIF1 and shieldin, control 3' overhang length, and the mechanism has been a topic of intensive research. Here, we highlight recent evidence that 3' overhang control by 53BP1 occurs through fill-in synthesis of resected DSBs by CST/Polα/primase. We focus on the crucial role of fill-in synthesis in BRCA1-deficient cells treated with PARPi and discuss the notion of fill-in synthesis in other specialized settings and in the repair of random DSBs. We argue that - in addition to other determinants - repair pathway choice may be influenced by the DNA sequence at the break which can impact CST binding and therefore the deployment of Polα/primase fill-in.
Collapse
Affiliation(s)
- Zachary Mirman
- Laboratory for Cell Biology and Genetics, The Rockefeller University, New York, NY, USA
- Department of Genetics, Harvard Medical School, Division of Genetics, Brigham and Women’s Hospital, HHMI, Boston, MA, USA
| | - Sarah Cai
- Laboratory for Cell Biology and Genetics, The Rockefeller University, New York, NY, USA
- Laboratory for Molecular Electron Microscopy, The Rockefeller University, New York, NY
| | - Titia de Lange
- Laboratory for Cell Biology and Genetics, The Rockefeller University, New York, NY, USA
| |
Collapse
|
29
|
Revy P, Kannengiesser C, Bertuch AA. Genetics of human telomere biology disorders. Nat Rev Genet 2023; 24:86-108. [PMID: 36151328 DOI: 10.1038/s41576-022-00527-z] [Citation(s) in RCA: 68] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/11/2022] [Indexed: 01/24/2023]
Abstract
Telomeres are specialized nucleoprotein structures at the ends of linear chromosomes that prevent the activation of DNA damage response and repair pathways. Numerous factors localize at telomeres to regulate their length, structure and function, to avert replicative senescence or genome instability and cell death. In humans, Mendelian defects in several of these factors can result in abnormally short or dysfunctional telomeres, causing a group of rare heterogeneous premature-ageing diseases, termed telomeropathies, short-telomere syndromes or telomere biology disorders (TBDs). Here, we review the TBD-causing genes identified so far and describe their main functions associated with telomere biology. We present molecular aspects of TBDs, including genetic anticipation, phenocopy, incomplete penetrance and somatic genetic rescue, which underlie the complexity of these diseases. We also discuss the implications of phenotypic and genetic features of TBDs on fundamental aspects related to human telomere biology, ageing and cancer, as well as on diagnostic, therapeutic and clinical approaches.
Collapse
Affiliation(s)
- Patrick Revy
- INSERM UMR 1163, Laboratory of Genome Dynamics in the Immune System, Equipe Labellisée Ligue Nationale contre le Cancer, Paris, France.
- Université Paris Cité, Imagine Institute, Paris, France.
| | - Caroline Kannengiesser
- APHP Service de Génétique, Hôpital Bichat, Paris, France
- Inserm U1152, Université Paris Cité, Paris, France
| | - Alison A Bertuch
- Departments of Paediatrics and Molecular & Human Genetics, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
30
|
Zheng B, Fu J. Telomere dysfunction in some pediatric congenital and growth-related diseases. Front Pediatr 2023; 11:1133102. [PMID: 37077333 PMCID: PMC10106694 DOI: 10.3389/fped.2023.1133102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 03/20/2023] [Indexed: 04/21/2023] Open
Abstract
Telomere wear and dysfunction may lead to aging-related diseases. Moreover, increasing evidence show that the occurrence, development, and prognosis of some pediatric diseases are also related to telomere dysfunction. In this review, we systematically analyzed the relationship between telomere biology and some pediatric congenital and growth-related diseases and proposed new theoretical basis and therapeutic targets for the treatment of these diseases.
Collapse
|
31
|
Abstract
Telomere biology was first studied in maize, ciliates, yeast, and mice, and in recent decades, it has informed understanding of common disease mechanisms with broad implications for patient care. Short telomere syndromes are the most prevalent premature aging disorders, with prominent phenotypes affecting the lung and hematopoietic system. Less understood are a newly recognized group of cancer-prone syndromes that are associated with mutations that lengthen telomeres. A large body of new data from Mendelian genetics and epidemiology now provides an opportunity to reconsider paradigms related to the role of telomeres in human aging and cancer, and in some cases, the findings diverge from what was interpreted from model systems. For example, short telomeres have been considered potent drivers of genome instability, but age-associated solid tumors are rare in individuals with short telomere syndromes, and T cell immunodeficiency explains their spectrum. More commonly, short telomeres promote clonal hematopoiesis, including somatic reversion, providing a new leukemogenesis paradigm that is independent of genome instability. Long telomeres, on the other hand, which extend the cellular life span in vitro, are now appreciated to be the most common shared germline risk factor for cancer in population studies. Through this contemporary lens, I revisit here the role of telomeres in human aging, focusing on how short and long telomeres drive cancer evolution but through distinct mechanisms.
Collapse
Affiliation(s)
- Mary Armanios
- Departments of Oncology, Genetic Medicine, Pathology, and Molecular Biology and Genetics; Telomere Center at Johns Hopkins; and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA;
| |
Collapse
|
32
|
Patient-Derived iPSCs Reveal Evidence of Telomere Instability and DNA Repair Deficiency in Coats Plus Syndrome. Genes (Basel) 2022; 13:genes13081395. [PMID: 36011306 PMCID: PMC9407572 DOI: 10.3390/genes13081395] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/26/2022] [Accepted: 08/02/2022] [Indexed: 12/11/2022] Open
Abstract
Coats plus (CP) syndrome is an inherited autosomal recessive condition that results from mutations in the conserved telomere maintenance component 1 gene (CTC1). The CTC1 protein functions as a part of the CST protein complex, a protein heterotrimer consisting of CTC1-STN1-TEN1 which promotes telomere DNA synthesis and inhibits telomerase-mediated telomere elongation. However, it is unclear how CTC1 mutations may have an effect on telomere structure and function. For that purpose, we established the very first induced pluripotent stem cell lines (iPSCs) from a compound heterozygous patient with CP carrying deleterious mutations in both alleles of CTC1. Telomere dysfunction and chromosomal instability were assessed in both circulating lymphocytes and iPSCs from the patient and from healthy controls of similar age. The circulating lymphocytes and iPSCs from the CP patient were characterized by their higher telomere length heterogeneity and telomere aberrations compared to those in control cells from healthy donors. Moreover, in contrast to iPSCs from healthy controls, the high levels of telomerase were associated with activation of the alternative lengthening of telomere (ALT) pathway in CP-iPSCs. This was accompanied by inappropriate activation of the DNA repair proteins γH2AX, 53BP1, and ATM, as well as with accumulation of DNA damage, micronuclei, and anaphase bridges. CP-iPSCs presented features of cellular senescence and increased radiation sensitivity. Clonal dicentric chromosomes were identified only in CP-iPSCs after exposure to radiation, thus mirroring the role of telomere dysfunction in their formation. These data demonstrate that iPSCs derived from CP patients can be used as a model system for molecular studies of the CP syndrome and underscores the complexity of telomere dysfunction associated with the defect of DNA repair machinery in the CP syndrome.
Collapse
|
33
|
Tummala H, Walne A, Buccafusca R, Alnajar J, Szabo A, Robinson P, McConkie-Rosell A, Wilson M, Crowley S, Kinsler V, Ewins AM, Madapura PM, Patel M, Pontikos N, Codd V, Vulliamy T, Dokal I. Germline thymidylate synthase deficiency impacts nucleotide metabolism and causes dyskeratosis congenita. Am J Hum Genet 2022; 109:1472-1483. [PMID: 35931051 PMCID: PMC9388389 DOI: 10.1016/j.ajhg.2022.06.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/23/2022] [Indexed: 11/19/2022] Open
Abstract
Dyskeratosis congenita (DC) is an inherited bone-marrow-failure disorder characterized by a triad of mucocutaneous features that include abnormal skin pigmentation, nail dystrophy, and oral leucoplakia. Despite the identification of several genetic variants that cause DC, a significant proportion of probands remain without a molecular diagnosis. In a cohort of eight independent DC-affected families, we have identified a remarkable series of heterozygous germline variants in the gene encoding thymidylate synthase (TYMS). Although the inheritance appeared to be autosomal recessive, one parent in each family had a wild-type TYMS coding sequence. Targeted genomic sequencing identified a specific haplotype and rare variants in the naturally occurring TYMS antisense regulator ENOSF1 (enolase super family 1) inherited from the other parent. Lymphoblastoid cells from affected probands have severe TYMS deficiency, altered cellular deoxyribonucleotide triphosphate pools, and hypersensitivity to the TYMS-specific inhibitor 5-fluorouracil. These defects in the nucleotide metabolism pathway resulted in genotoxic stress, defective transcription, and abnormal telomere maintenance. Gene-rescue studies in cells from affected probands revealed that post-transcriptional epistatic silencing of TYMS is occurring via elevated ENOSF1. These cell and molecular abnormalities generated by the combination of germline digenic variants at the TYMS-ENOSF1 locus represent a unique pathogenetic pathway for DC causation in these affected individuals, whereas the parents who are carriers of either of these variants in a singular fashion remain unaffected.
Collapse
Affiliation(s)
- Hemanth Tummala
- Genomics and Child Health, Blizard Institute, Queen Mary University of London, Newark Street, London E1 2AT, UK.
| | - Amanda Walne
- Genomics and Child Health, Blizard Institute, Queen Mary University of London, Newark Street, London E1 2AT, UK
| | - Roberto Buccafusca
- School of Physical and Chemical Sciences, Queen Mary University of London, Mile End, London E1 4NS, UK
| | - Jenna Alnajar
- Genomics and Child Health, Blizard Institute, Queen Mary University of London, Newark Street, London E1 2AT, UK
| | - Anita Szabo
- Institute of Ophthalmology, Faculty of Brain Sciences, University College London, 11-43 Bath St, London EC1V 9EL, UK
| | - Peter Robinson
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Dr., Farmington, CT 06032, USA
| | | | - Meredith Wilson
- Department of Clinical Genetics, The Children's Hospital at Westmead, Sydney, Australia
| | - Suzanne Crowley
- Department of Paediatrics, St George's Healthcare NHS Trust, London, UK
| | - Veronica Kinsler
- Department of Paediatric Dermatology, Great Ormond Street Hospital, The Francis Crick Institute, London, UK
| | - Anna-Maria Ewins
- Haematology/Oncology Department, Royal Hospital for Sick Children, Glasgow, UK
| | - Pradeepa M Madapura
- Genomics and Child Health, Blizard Institute, Queen Mary University of London, Newark Street, London E1 2AT, UK
| | - Manthan Patel
- Genomics and Child Health, Blizard Institute, Queen Mary University of London, Newark Street, London E1 2AT, UK
| | - Nikolas Pontikos
- Institute of Ophthalmology, Faculty of Brain Sciences, University College London, 11-43 Bath St, London EC1V 9EL, UK
| | - Veryan Codd
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Tom Vulliamy
- Genomics and Child Health, Blizard Institute, Queen Mary University of London, Newark Street, London E1 2AT, UK
| | - Inderjeet Dokal
- Genomics and Child Health, Blizard Institute, Queen Mary University of London, Newark Street, London E1 2AT, UK; Barts Health NHS Trust, London, UK
| |
Collapse
|
34
|
Tummala H, Walne A, Dokal I. The biology and management of dyskeratosis congenita and related disorders of telomeres. Expert Rev Hematol 2022; 15:685-696. [PMID: 35929966 DOI: 10.1080/17474086.2022.2108784] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 07/29/2022] [Indexed: 11/04/2022]
Abstract
BACKGROUND Dyskeratosis congenita (DC) is a multisystem syndrome characterized by mucocutaneous abnormalities, bone marrow failure, and predisposition to cancer. Studies over the last 25 years have led to the identification of 18 disease genes. These have a principal role in telomere maintenance, and patients usually have very short/abnormal telomeres. The advances have also led to the unification of DC with a number of other diseases, now collectively referred to as the telomeropathies or telomere biology disorders. WHAT IS COVERED Clinical features, genetics, and biology of the different subtypes. Expert view on diagnosis, treatment of the hematological complications and future. EXPERT VIEW As these are very pleotropic disorders affecting multiple organs, a high index of suspicion is necessary to make the diagnosis. Telomere length measurement and genetic analysis of the disease genes have become useful diagnostic tools. Although hematological defects can respond to danazol/oxymetholone, the only current curative treatment for these is hematopoietic stem cell transplantation (HSCT) using fludarabine-based conditioning protocols. New therapies are needed where danazol/oxymetholone is ineffective and HSCT is not feasible.
Collapse
Affiliation(s)
- Hemanth Tummala
- Centre for Genomics and Child Health, Blizard Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Amanda Walne
- Centre for Genomics and Child Health, Blizard Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Inderjeet Dokal
- Centre for Genomics and Child Health, Blizard Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
- Department of Haematology, Barts Health, London, UK
| |
Collapse
|
35
|
Sertori R, Lin JX, Martinez E, Rana S, Sharo A, Kazemian M, Sunderam U, Andrake M, Shinton S, Truong B, Dunbrack RM, Liu C, Srinivasan R, Brenner SE, Seroogy CM, Puck JM, Leonard WJ, Wiest DL. Investigation of the causal etiology in a patient with T-B+NK+ immunodeficiency. Front Immunol 2022; 13:928252. [PMID: 35967429 PMCID: PMC9372720 DOI: 10.3389/fimmu.2022.928252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 06/27/2022] [Indexed: 11/24/2022] Open
Abstract
Newborn screening for severe combined immunodeficiency (SCID) has not only accelerated diagnosis and improved treatment for affected infants, but also led to identification of novel genes required for human T cell development. A male proband had SCID newborn screening showing very low T cell receptor excision circles (TRECs), a biomarker for thymic output of nascent T cells. He had persistent profound T lymphopenia, but normal numbers of B and natural killer (NK) cells. Despite an allogeneic hematopoietic stem cell transplant from his brother, he failed to develop normal T cells. Targeted resequencing excluded known SCID genes; however, whole exome sequencing (WES) of the proband and parents revealed a maternally inherited X-linked missense mutation in MED14 (MED14V763A), a component of the mediator complex. Morpholino (MO)-mediated loss of MED14 function attenuated T cell development in zebrafish. Moreover, this arrest was rescued by ectopic expression of cDNA encoding the wild type human MED14 ortholog, but not by MED14V763A , suggesting that the variant impaired MED14 function. Modeling of the equivalent mutation in mouse (Med14V769A) did not disrupt T cell development at baseline. However, repopulation of peripheral T cells upon competitive bone marrow transplantation was compromised, consistent with the incomplete T cell reconstitution experienced by the proband upon transplantation with bone marrow from his healthy male sibling, who was found to have the same MED14V763A variant. Suspecting that the variable phenotypic expression between the siblings was influenced by further mutation(s), we sought to identify genetic variants present only in the affected proband. Indeed, WES revealed a mutation in the L1 cell adhesion molecule (L1CAMQ498H); however, introducing that mutation in vivo in mice did not disrupt T cell development. Consequently, immunodeficiency in the proband may depend upon additional, unidentified gene variants.
Collapse
Affiliation(s)
- Robert Sertori
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA, United States
| | - Jian-Xin Lin
- Laboratory of Molecular Immunology, Immunology Center, National Heart, Lung, and Blood Institute, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Esteban Martinez
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA, United States
| | - Sadhna Rana
- Innovation Labs, Tata Consultancy Services, Hyderabad, India
| | - Andrew Sharo
- Center for Computational Biology, University of California, Berkeley, CA, United States
| | - Majid Kazemian
- Departments of Biochemistry and Computer Science, Purdue University, West Lafayette, IN, United States
| | - Uma Sunderam
- Innovation Labs, Tata Consultancy Services, Hyderabad, India
| | - Mark Andrake
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA, United States
| | - Susan Shinton
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA, United States
| | - Billy Truong
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA, United States
| | - Roland M. Dunbrack
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA, United States
| | - Chengyu Liu
- Transgenic Core, National Heart, Lung, and Blood Institute, National Institutes of Health (NIH), Bethesda, MD, United States
| | | | - Steven E. Brenner
- Center for Computational Biology, University of California, Berkeley, CA, United States
| | - Christine M. Seroogy
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Jennifer M. Puck
- Department of Pediatrics, University of California San Francisco and UCSF Benioff Children’s Hospital, San Francisco, CA, United States
| | - Warren J. Leonard
- Laboratory of Molecular Immunology, Immunology Center, National Heart, Lung, and Blood Institute, National Institutes of Health (NIH), Bethesda, MD, United States
| | - David L. Wiest
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA, United States
| |
Collapse
|
36
|
Leach DF, Mathavan A, Mathavan A, Kaur J, Zori AG, Ataya A. A 26-Year-Old Woman With Retinal Telangiectasias, Onychodystrophy, and Persistent Dyspnea. Chest 2022; 161:e365-e369. [DOI: 10.1016/j.chest.2022.02.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 02/01/2022] [Accepted: 02/03/2022] [Indexed: 11/25/2022] Open
|
37
|
Feurstein S, Hahn CN, Mehta N, Godley LA. A practical guide to interpreting germline variants that drive hematopoietic malignancies, bone marrow failure, and chronic cytopenias. Genet Med 2022; 24:931-954. [PMID: 35063349 DOI: 10.1016/j.gim.2021.12.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 12/14/2021] [Indexed: 10/19/2022] Open
Abstract
PURPOSE The American College of Medical Genetics and Genomics and the Association for Molecular Pathology guidelines for germline variant interpretation are implemented as a broad framework by standardizing variant interpretation. These rules were designed to be specified, but this process has not been performed for most of the 200 genes associated with inherited hematopoietic malignancies, bone marrow failure, and cytopenias. Because guidelines on how to perform these gene specifications are lacking, variant interpretation is less reliable and reproducible. METHODS We have used a variety of methods such as calculations of minor allele frequencies, quasi-case-control studies to establish thresholds, proband counting, and plotting of receiver operating characteristic curves to compare different in silico prediction tools to design recommendations for variant interpretation. RESULTS We herein provide practical recommendations for the creation of thresholds for minor allele frequencies, in silico predictions, counting of probands, identification of functional domains with minimal benign variation, use of constraint Z-scores and functional evidence, prediction of nonsense-mediated decay, and assessment of phenotype specificity. CONCLUSION These guidelines can be used by anyone interpreting variants associated with inherited hematopoietic malignancies, bone marrow failure, and cytopenias to develop criteria for reliable, accurate, and reproducible germline variant interpretation.
Collapse
Affiliation(s)
- Simone Feurstein
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL; Section of Hematology, Oncology and Rheumatology, Department of Internal Medicine, Heidelberg University Hospital, Heidelberg, Germany
| | - Christopher N Hahn
- Molecular Pathology Research Laboratory, Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, South Australia, Australia; Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| | - Nikita Mehta
- Diagnostic Molecular Genetics Laboratory, Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Lucy A Godley
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL; Department of Human Genetics, The University of Chicago, Chicago, IL.
| |
Collapse
|
38
|
Wang L, Ma T, Liu W, Li H, Luo Z, Feng X. Pan-Cancer Analyses Identify the CTC1-STN1-TEN1 Complex as a Protective Factor and Predictive Biomarker for Immune Checkpoint Blockade in Cancer. Front Genet 2022; 13:859617. [PMID: 35368664 PMCID: PMC8966541 DOI: 10.3389/fgene.2022.859617] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 02/23/2022] [Indexed: 11/13/2022] Open
Abstract
The CTC1-STN1-TEN1 (CST) complex plays a crucial role in telomere replication and genome stability. However, the detailed mechanisms of CST regulation in cancer remain largely unknown. Here, we perform a comprehensive analysis of CST across 33 cancer types using multi-omic data from The Cancer Genome Atlas. In the genomic landscape, we identify CTC1/STN1 deletion and mutation and TEN1 amplification as the dominant alteration events. Expressions of CTC1 and STN1 are decreased in tumors compared to those in adjacent normal tissues. Clustering analysis based on CST expression reveals three cancer clusters displaying differences in survival, telomerase activity, cell proliferation, and genome stability. Interestingly, we find that CTC1 and STN1, but not TEN1, are co-expressed and associated with better survival. CTC1-STN1 is positively correlated with CD8 T cells and B cells and predicts a better response to immune checkpoint blockade in external datasets of cancer immunotherapy. Pathway analysis shows that MYC targets are negatively correlated with CTC1-STN1. We experimentally validated that knockout of CTC1 increased the mRNA level of c-MYC. Furthermore, CTC1 and STN1 are repressed by miRNAs and lncRNAs. Finally, by mining the connective map database, we discover a number of potential drugs that may target CST. In sum, this study illustrates CTC1-STN1 as a protective factor and provides broad molecular signatures for further functional and therapeutic studies of CST in cancer.
Collapse
Affiliation(s)
- Lishuai Wang
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Department of Medical Oncology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Tengfei Ma
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Weijin Liu
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Heping Li
- Department of Medical Oncology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- *Correspondence: Heping Li, ; Zhenhua Luo, ; Xuyang Feng,
| | - Zhenhua Luo
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- *Correspondence: Heping Li, ; Zhenhua Luo, ; Xuyang Feng,
| | - Xuyang Feng
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- *Correspondence: Heping Li, ; Zhenhua Luo, ; Xuyang Feng,
| |
Collapse
|
39
|
Abstract
Cerebral small vessel disease (cSVD) is a leading cause of ischaemic and haemorrhagic stroke and a major contributor to dementia. Covert cSVD, which is detectable with brain MRI but does not manifest as clinical stroke, is highly prevalent in the general population, particularly with increasing age. Advances in technologies and collaborative work have led to substantial progress in the identification of common genetic variants that are associated with cSVD-related stroke (ischaemic and haemorrhagic) and MRI-defined covert cSVD. In this Review, we provide an overview of collaborative studies - mostly genome-wide association studies (GWAS) - that have identified >50 independent genetic loci associated with the risk of cSVD. We describe how these associations have provided novel insights into the biological mechanisms involved in cSVD, revealed patterns of shared genetic variation across cSVD traits, and shed new light on the continuum between rare, monogenic and common, multifactorial cSVD. We consider how GWAS summary statistics have been leveraged for Mendelian randomization studies to explore causal pathways in cSVD and provide genetic evidence for drug effects, and how the combination of findings from GWAS with gene expression resources and drug target databases has enabled identification of putative causal genes and provided proof-of-concept for drug repositioning potential. We also discuss opportunities for polygenic risk prediction, multi-ancestry approaches and integration with other omics data.
Collapse
|
40
|
Telomeres and Cancer. Life (Basel) 2021; 11:life11121405. [PMID: 34947936 PMCID: PMC8704776 DOI: 10.3390/life11121405] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/01/2021] [Accepted: 12/06/2021] [Indexed: 12/18/2022] Open
Abstract
Telomeres cap the ends of eukaryotic chromosomes and are indispensable chromatin structures for genome protection and replication. Telomere length maintenance has been attributed to several functional modulators, including telomerase, the shelterin complex, and the CST complex, synergizing with DNA replication, repair, and the RNA metabolism pathway components. As dysfunctional telomere maintenance and telomerase activation are associated with several human diseases, including cancer, the molecular mechanisms behind telomere length regulation and protection need particular emphasis. Cancer cells exhibit telomerase activation, enabling replicative immortality. Telomerase reverse transcriptase (TERT) activation is involved in cancer development through diverse activities other than mediating telomere elongation. This review describes the telomere functions, the role of functional modulators, the implications in cancer development, and the future therapeutic opportunities.
Collapse
|
41
|
Lei KH, Yang HL, Chang HY, Yeh HY, Nguyen DD, Lee TY, Lyu X, Chastain M, Chai W, Li HW, Chi P. Crosstalk between CST and RPA regulates RAD51 activity during replication stress. Nat Commun 2021; 12:6412. [PMID: 34741010 PMCID: PMC8571288 DOI: 10.1038/s41467-021-26624-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 10/06/2021] [Indexed: 11/09/2022] Open
Abstract
Replication stress causes replication fork stalling, resulting in an accumulation of single-stranded DNA (ssDNA). Replication protein A (RPA) and CTC1-STN1-TEN1 (CST) complex bind ssDNA and are found at stalled forks, where they regulate RAD51 recruitment and foci formation in vivo. Here, we investigate crosstalk between RPA, CST, and RAD51. We show that CST and RPA localize in close proximity in cells. Although CST stably binds to ssDNA with a high affinity at low ionic strength, the interaction becomes more dynamic and enables facilitated dissociation at high ionic strength. CST can coexist with RPA on the same ssDNA and target RAD51 to RPA-coated ssDNA. Notably, whereas RPA-coated ssDNA inhibits RAD51 activity, RAD51 can assemble a functional filament and exhibit strand-exchange activity on CST-coated ssDNA at high ionic strength. Our findings provide mechanistic insights into how CST targets and tethers RAD51 to RPA-coated ssDNA in response to replication stress.
Collapse
Affiliation(s)
- Kai-Hang Lei
- Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| | - Han-Lin Yang
- Department of Chemistry, National Taiwan University, Taipei, Taiwan
| | - Hao-Yen Chang
- Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| | - Hsin-Yi Yeh
- Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| | - Dinh Duc Nguyen
- Department of Cancer Biology, Cardinal Bernardin Cancer Center, Loyola University Chicago Stritch School of Medicine, Maywood, IL, USA
| | - Tzu-Yu Lee
- Department of Chemistry, National Taiwan University, Taipei, Taiwan
| | - Xinxing Lyu
- Department of Cancer Biology, Cardinal Bernardin Cancer Center, Loyola University Chicago Stritch School of Medicine, Maywood, IL, USA
| | - Megan Chastain
- Office of Research, Washington State University, Spokane, WA, USA
| | - Weihang Chai
- Department of Cancer Biology, Cardinal Bernardin Cancer Center, Loyola University Chicago Stritch School of Medicine, Maywood, IL, USA
| | - Hung-Wen Li
- Department of Chemistry, National Taiwan University, Taipei, Taiwan.
| | - Peter Chi
- Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan. .,Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
42
|
The Intrinsically Disordered Region in the Human STN1 OB-Fold Domain Is Important for Protecting Genome Stability. BIOLOGY 2021; 10:biology10100977. [PMID: 34681076 PMCID: PMC8533325 DOI: 10.3390/biology10100977] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/17/2021] [Accepted: 09/24/2021] [Indexed: 11/17/2022]
Abstract
Simple Summary The human CTC1–STN1–TEN1 (CST) complex is an ssDNA-binding protein complex that is thought to be related to the RPA70/RPA32/RPA14 complex. While recent studies have shown that CST plays key roles in multiple genome maintenance pathways, including protecting fork stability under perturbed replication, promoting efficient replication of difficult-to-replicate DNA, repairing DNA double-stranded breaks, and maintaining telomere integrity, it is poorly understood how CST function is regulated in genome maintenance. In this study, we identify an intrinsically disordered region (IDR) in the OB domain of STN1 and analyze the functions of cancer-associated IDR variants and a number of alanine substitutions of individual polar or hydrophilic residues in this IDR. We observe that these variants confer replication-associated genome instability, reduced cellular viability, and increased HU sensitivity. Analysis of protein–protein interactions using IDR variants and IDR deletion shows that the IDR is critical for STN1–POLα interaction, but not CST–RAD51 interaction or CST complex formation. Together, our results identify the IDR in STN1-OB as an important element modulating CST function in protecting genome stability under replication stress. Abstract The mammalian CTC1–STN1–TEN1 (CST) complex is an ssDNA-binding protein complex that has emerged as an important player in protecting genome stability and preserving telomere integrity. Studies have shown that CST localizes at stalled replication forks and is critical for protecting the stability of nascent strand DNA. Recent cryo-EM analysis reveals that CST subunits possess multiple OB-fold domains that can form a decameric supercomplex. While considered to be RPA-like, CST acts distinctly from RPA to protect genome stability. Here, we report that while the OB domain of STN1 shares structural similarity with the OB domain of RPA32, the STN1-OB domain contains an intrinsically disordered region (IDR) that is important for maintaining genome stability under replication stress. Single mutations in multiple positions in this IDR, including cancer-associated mutations, cause genome instabilities that are elevated by replication stress and display reduced cellular viability and increased HU sensitivity. While IDR mutations do not impact CST complex formation or CST interaction with its binding partner RAD51, they diminish RAD51 foci formation when replication is perturbed. Interestingly, the IDR is critical for STN1–POLα interaction. Collectively, our results identify the STN1 IDR as an important element in regulating CST function in genome stability maintenance.
Collapse
|
43
|
Kara B, Uyguner O, Maraş Genç H, İşlek EE, Kasap M, Toksoy G, Akpınar G, Uyur Yalçın E, Anık Y, Üstek D. BEND4 as a Candidate Gene for an Infection-Induced Acute Encephalopathy Characterized by a Cyst and Calcification of the Pons and Cerebellar Atrophy. Mol Syndromol 2021; 13:12-22. [DOI: 10.1159/000517541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 06/01/2021] [Indexed: 11/19/2022] Open
Abstract
Three siblings born to Turkish parents from the same village had normal brain development until acute neurological deterioration between 12 months and 8 years of age. Consequent loss of all acquired motor, social, and language functions following infections was associated with a pontine cyst, calcification, and cerebellar atrophy. Exome sequencing revealed a homozygous c.1297G>A (p.Gly433Ser) alteration in <i>BEND4</i>, which was predicted to be deleterious in in silico analysis tools and segregated in multiple affected individuals in the family. <i>BEND4</i> has not been associated with any existing disease. Immunofluorescence microscopy analysis of wild-type and mutant BEND4 expressing Vero cells showed nuclear and cytoplasmic localization. Wild-type BEND4 displayed a network-like distribution, whereas mutant BEND4 showed a juxtanuclear distribution pattern. Differential proteome analysis of Vero cells expressing BEND4 revealed that mutant BEND4 expression caused selective increase in reticulocalbin-1 and endoplasmic reticulum resident protein-29. Both proteins are associated with the endoplasmic reticulum and are primarily involved in protein processing and folding pathways. Any defect or stress in protein folding creates stress on cells and may cause chronic damage. This is the first study showing that pathogenic <i>BEND4</i> variants may lead to an infection-induced acute necrotizing encephalopathy as demonstrated in characteristic neuroimaging findings.
Collapse
|
44
|
Tabor N, Ngwa C, Mitteaux J, Meyer MD, Moruno-Manchon JF, Zhu L, Liu F, Monchaud D, McCullough LD, Tsvetkov AS. Differential responses of neurons, astrocytes, and microglia to G-quadruplex stabilization. Aging (Albany NY) 2021; 13:15917-15941. [PMID: 34139671 PMCID: PMC8266374 DOI: 10.18632/aging.203222] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 06/12/2021] [Indexed: 12/29/2022]
Abstract
The G-quadruplex (G4-DNA or G4) is a secondary DNA structure formed by DNA sequences containing multiple runs of guanines. While it is now firmly established that stabilized G4s lead to enhanced genomic instability in cancer cells, whether and how G4s contribute to genomic instability in brain cells is still not clear. We previously showed that, in cultured primary neurons, small-molecule G4 stabilizers promote formation of DNA double-strand breaks (DSBs) and downregulate the Brca1 gene. Here, we determined if G4-dependent Brca1 downregulation is unique to neurons or if the effects in neurons also occur in astrocytes and microglia. We show that primary neurons, astrocytes and microglia basally exhibit different G4 landscapes. Stabilizing G4-DNA with the G4 ligand pyridostatin (PDS) differentially modifies chromatin structure in these cell types. Intriguingly, PDS promotes DNA DSBs in neurons, astrocytes and microglial cells, but fails to downregulate Brca1 in astrocytes and microglia, indicating differences in DNA damage and repair pathways between brain cell types. Taken together, our findings suggest that stabilized G4-DNA contribute to genomic instability in the brain and may represent a novel senescence pathway in brain aging.
Collapse
Affiliation(s)
- Natalie Tabor
- Department of Neurology, The University of Texas McGovern Medical School at Houston, Houston, TX 77030, USA
| | - Conelius Ngwa
- Department of Neurology, The University of Texas McGovern Medical School at Houston, Houston, TX 77030, USA
| | - Jeremie Mitteaux
- Institut de Chimie Moléculaire (ICMUB), UBFC Dijon, CNRS UMR6302, Dijon, France
| | - Matthew D. Meyer
- Shared Equipment Authority, Rice University, Houston, TX 77005, USA
| | - Jose F. Moruno-Manchon
- Department of Neurology, The University of Texas McGovern Medical School at Houston, Houston, TX 77030, USA
| | - Liang Zhu
- Biostatistics and Epidemiology Research Design Core Center for Clinical and Translational Sciences, The University of Texas McGovern Medical School at Houston, Houston, TX 77030, USA
- Department of Internal Medicine, The University of Texas McGovern Medical School at Houston, Houston, TX 77030, USA
| | - Fudong Liu
- Department of Neurology, The University of Texas McGovern Medical School at Houston, Houston, TX 77030, USA
| | - David Monchaud
- Institut de Chimie Moléculaire (ICMUB), UBFC Dijon, CNRS UMR6302, Dijon, France
| | - Louise D. McCullough
- Department of Neurology, The University of Texas McGovern Medical School at Houston, Houston, TX 77030, USA
- The University of Texas Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Andrey S. Tsvetkov
- Department of Neurology, The University of Texas McGovern Medical School at Houston, Houston, TX 77030, USA
- The University of Texas Graduate School of Biomedical Sciences, Houston, TX 77030, USA
- UTHealth Consortium on Aging, The University of Texas McGovern Medical School, Houston, TX 77030, USA
| |
Collapse
|
45
|
Acharya T, Firth HV, Dugar S, Grammatikopoulos T, Seabra L, Walters A, Crow YJ, Parker APJ. Novel compound heterozygous STN1 variants are associated with Coats Plus syndrome. Mol Genet Genomic Med 2021; 9:e1708. [PMID: 34110109 PMCID: PMC8683631 DOI: 10.1002/mgg3.1708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 01/20/2021] [Accepted: 04/13/2021] [Indexed: 11/16/2022] Open
Abstract
Aim Coats plus syndrome (CP) is a rare autosomal recessive disorder, characterised by retinal telangiectasia exudates (Coats disease), leukodystrophy, distinctive intracranial calcification and cysts, as well as extra‐neurological features including abnormal vasculature of the gastrointestinal tract, portal hypertension and osteopenia with a tendency to fractures. CP most frequently occurs due to loss‐of‐function mutations in CTC1. The encoded protein CTC1 constitutes part of the CST (CTC1‐STN1‐TEN1) complex, and three patients have been described with CP due to biallelic mutations in STN1. Together with the identification of homozygosity for a specific loss‐of‐function mutation in POT1 in a sibling pair, these observations highlight a defect in the maintenance of telomere integrity as the cause of CP, although the precise mechanism leading to the micro‐vasculopathy seen at a pathological level remains unclear. Here, we present the investigation of a fourth child who presented to us with retinal exudates, intracranial calcifications and developmental delay, in keeping with a diagnosis of CP, and later went on to develop pancytopenia and gastrointestinal bleeding. Genome sequencing revealed compound heterozygous variants in STN1 as the likely genetic cause of CP in this present case. Methods We assessed the phenotype to be CP and undertook targeted sequencing. Results Whilst sequencing of CTC1 and POT1 was normal, we identified novel compound heterozygous variants in STN1 (previous gene symbol OBFC1): one loss‐of‐function––c.894dup (p.(Asp299Argfs*58)); and one missense––c.707T>C (p.(Leu236Pro)). Conclusion Given the clinical phenotype and identified variants we suggest that this is only the fourth patient reported to date with CP due to mutations in STN1.
Collapse
Affiliation(s)
- Tanvi Acharya
- School of Clinical Medicine, Cambridge University, Cambridge, UK
| | - Helen V Firth
- Department of Clinical Genetics, Addenbrooke's Hospital, Cambridge, UK
| | - Shilpa Dugar
- Paediatric Liver, GI and Nutrition Centre and Mowat Labs, King's College Hospital NHS Foundation Trust, London, UK
| | - Tassos Grammatikopoulos
- Paediatric Liver, GI and Nutrition Centre and Mowat Labs, King's College Hospital NHS Foundation Trust, London, UK
| | - Luis Seabra
- Laboratory of Neurogenetics and Neuroinflammation, Institut Imagine, Paris, France
| | - Angharad Walters
- Cambridgeshire Community Services, Brooksfield Hospital, Cambridge, UK
| | - Yanick J Crow
- Centre for Genomic and Experimental Medicine, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | | |
Collapse
|
46
|
Lyu X, Sang PB, Chai W. CST in maintaining genome stability: Beyond telomeres. DNA Repair (Amst) 2021; 102:103104. [PMID: 33780718 PMCID: PMC8081025 DOI: 10.1016/j.dnarep.2021.103104] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/17/2021] [Accepted: 03/18/2021] [Indexed: 12/12/2022]
Abstract
The human CST (CTC1-STN1-TEN1) complex is an RPA-like single-stranded DNA binding protein complex. While its telomeric functions have been well investigated, numerous studies have revealed that hCST also plays important roles in maintaining genome stability beyond telomeres. Here, we review and discuss recent discoveries on CST in various global genome maintenance pathways, including findings on the CST supercomplex structure, its functions in unperturbed DNA replication, stalled replication, double-strand break repair, and the ATR-CHK1 activation pathway. By summarizing these recent discoveries, we hope to offer new insights into genome maintenance mechanisms and the pathogenesis of CST mutation-associated diseases.
Collapse
Affiliation(s)
- Xinxing Lyu
- Institute of Basic Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250062, China; Department of Cancer Biology, Cardinal Bernardin Cancer Center, Loyola University Chicago Stritch School of Medicine, Maywood, IL, 60153, United States
| | - Pau Biak Sang
- Department of Cancer Biology, Cardinal Bernardin Cancer Center, Loyola University Chicago Stritch School of Medicine, Maywood, IL, 60153, United States
| | - Weihang Chai
- Department of Cancer Biology, Cardinal Bernardin Cancer Center, Loyola University Chicago Stritch School of Medicine, Maywood, IL, 60153, United States.
| |
Collapse
|
47
|
Bonnell E, Pasquier E, Wellinger RJ. Telomere Replication: Solving Multiple End Replication Problems. Front Cell Dev Biol 2021; 9:668171. [PMID: 33869233 PMCID: PMC8047117 DOI: 10.3389/fcell.2021.668171] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 03/10/2021] [Indexed: 12/19/2022] Open
Abstract
Eukaryotic genomes are highly complex and divided into linear chromosomes that require end protection from unwarranted fusions, recombination, and degradation in order to maintain genomic stability. This is accomplished through the conserved specialized nucleoprotein structure of telomeres. Due to the repetitive nature of telomeric DNA, and the unusual terminal structure, namely a protruding single stranded 3' DNA end, completing telomeric DNA replication in a timely and efficient manner is a challenge. For example, the end replication problem causes a progressive shortening of telomeric DNA at each round of DNA replication, thus telomeres eventually lose their protective capacity. This phenomenon is counteracted by the recruitment and the activation at telomeres of the specialized reverse transcriptase telomerase. Despite the importance of telomerase in providing a mechanism for complete replication of telomeric ends, the majority of telomere replication is in fact carried out by the conventional DNA replication machinery. There is significant evidence demonstrating that progression of replication forks is hampered at chromosomal ends due to telomeric sequences prone to form secondary structures, tightly DNA-bound proteins, and the heterochromatic nature of telomeres. The telomeric loop (t-loop) formed by invasion of the 3'-end into telomeric duplex sequences may also impede the passage of replication fork. Replication fork stalling can lead to fork collapse and DNA breaks, a major cause of genomic instability triggered notably by unwanted repair events. Moreover, at chromosomal ends, unreplicated DNA distal to a stalled fork cannot be rescued by a fork coming from the opposite direction. This highlights the importance of the multiple mechanisms involved in overcoming fork progression obstacles at telomeres. Consequently, numerous factors participate in efficient telomeric DNA duplication by preventing replication fork stalling or promoting the restart of a stalled replication fork at telomeres. In this review, we will discuss difficulties associated with the passage of the replication fork through telomeres in both fission and budding yeasts as well as mammals, highlighting conserved mechanisms implicated in maintaining telomere integrity during replication, thus preserving a stable genome.
Collapse
Affiliation(s)
| | | | - Raymund J. Wellinger
- Department of Microbiology and Infectious Diseases, Faculty of Medicine and Health Sciences, Cancer Research Pavilion, Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
48
|
Teixeira da Silva JA. Multiple co-first authors, co-corresponding authors and co-supervisors: a synthesis of shared authorship credit. ONLINE INFORMATION REVIEW 2021. [DOI: 10.1108/oir-06-2020-0219] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
PurposeAuthorship is the ultimate status of intellectual recognition in academic publishing. Although fairly robust guidelines have already been in place for a considerable amount of time regarding authorship criteria and credit, such as those by the International Committee of Medical Journal Editors or Contributor Roles Taxonomy, the lack of reliable verification techniques hamper their accuracy, thereby reducing the validity of authorship claims in such statements. This paper aims to focus on the authorship status and responsibilities of co-first authors and co-corresponding authors.Design/methodology/approachTo appreciate authorship responsibilities in this subset of authors, the broader academic authorship literature, as well as position statements, rules and guidelines, were consulted.FindingsAcademic publishing that relies on metrics is a global multi-billion-dollar business, so strict measures to assess and confirm authorship, which can be intellectually or financially “profitable” among academics that game such metrics, are needed. The current assessment is that there are inconsistent rules for equally credited authors such as co-first authors, co-corresponding authors and co-supervisors. In shared and collaborative authorship, there are also shared authorship-related responsibilities, but these are infrequently discussed, or tend to only be dealt with broadly.Originality/valueWithin the wider, and important, discussion about authorship, which is one of the most central issues in academic publishing, there has been a limited focus on equally credited authors such as co-first authors, co-corresponding authors and co-supervisors. This paper expands and fortifies that discussion.
Collapse
|
49
|
Himes RW, Chiou EH, Queliza K, Shouval DS, Somech R, Agarwal S, Jajoo K, Ziegler DS, Kratz CP, Huang J, Lucas TL, Myers KC, Nelson AS, DiNardo CD, Alter BP, Giri N, Khincha PP, McReynolds LJ, Dufour C, Pierri F, Goldman FD, Sherif Y, Savage SA, Miloh T, Bertuch AA. Gastrointestinal Hemorrhage: A Manifestation of the Telomere Biology Disorders. J Pediatr 2021; 230:55-61.e4. [PMID: 32971146 DOI: 10.1016/j.jpeds.2020.09.038] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 08/17/2020] [Accepted: 09/16/2020] [Indexed: 12/20/2022]
Abstract
OBJECTIVE To describe the clinical features, therapeutic interventions, and patient outcomes of gastrointestinal (GI) hemorrhage in individuals with a telomere biology disorder, including dyskeratosis congenita, Hoyeraal-Hreidarsson syndrome, Revesz syndrome, and Coats plus. STUDY DESIGN Clinical Care Consortium for Telomere Associated Ailments members were invited to contribute data on individuals with telomere biology disorders at their institutions who experienced GI bleeding. Patient demographic, laboratory, imaging, procedural, and treatment information and outcomes were extracted from the medical record. RESULTS Sixteen patients who experienced GI hemorrhage were identified at 11 centers. Among 14 patients who underwent genetic testing, 8 had mutations in TINF2, 4 had mutations in CTC1 or STN1, and 1 patient each had a mutation in TERC and RTEL1. Ten patients had a history of hematopoietic cell transplantation. The patients with Coats plus and those without Coats plus had similar clinical features and courses. Angiodysplasia of the stomach and/or small bowel was described in 8 of the 12 patients who underwent endoscopy; only 4 had esophageal varices. Various medical interventions were trialed. No single intervention was uniformly associated with cessation of bleeding, although 1 patient had a sustained response to treatment with bevacizumab. Recurrence was common, and the overall long-term outcome for affected patients was poor. CONCLUSIONS GI bleeding in patients with telomere biology disorders is associated with significant morbidity and with vascular ectasias rather than varices.
Collapse
Affiliation(s)
- Ryan W Himes
- Department of Pediatric Gastroenterology, Hepatology and Nutrition, Baylor College of Medicine and Texas Children's Hospital, Houston, TX; Section of Gastroenterology and Hepatology, Department of Pediatrics, Ochsner Health, New Orleans, LA.
| | - Eric H Chiou
- Department of Pediatric Gastroenterology, Hepatology and Nutrition, Baylor College of Medicine and Texas Children's Hospital, Houston, TX
| | - Karen Queliza
- Department of Pediatric Gastroenterology, Hepatology and Nutrition, Baylor College of Medicine and Texas Children's Hospital, Houston, TX; Gastroenterology, Hepatology and Nutrition Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Dror S Shouval
- Pediatric Gastroenterology Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Raz Somech
- Pediatric Gastroenterology Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel
| | - Suneet Agarwal
- Boston Children's Hospital, Dana-Farber Cancer Institute, and Harvard Medical School, Boston, MA
| | - Kunal Jajoo
- Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - David S Ziegler
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, NSW, Australia
| | - Christian P Kratz
- Department of Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
| | - James Huang
- Division of Hematology/Oncology, Department of Pediatrics, UCSF Benioff Children's Hospital, University of California, San Francisco, CA
| | - Tiffany L Lucas
- Division of Hematology/Oncology, Department of Pediatrics, UCSF Benioff Children's Hospital, University of California, San Francisco, CA
| | - Kasiani C Myers
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Adam S Nelson
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | | | - Blanche P Alter
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD
| | - Neelam Giri
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD
| | - Payal P Khincha
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD
| | - Lisa J McReynolds
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD
| | - Carlo Dufour
- Hemato-Oncology-Stem Cell Transplant Pole, IRCCS Giannina Gaslini, Genoa, Italy
| | - Filomena Pierri
- Hemato-Oncology-Stem Cell Transplant Pole, IRCCS Giannina Gaslini, Genoa, Italy
| | | | - Youmna Sherif
- Department of Surgery, Baylor College of Medicine and Texas Children's Hospital, Houston, TX
| | - Sharon A Savage
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD
| | - Tamir Miloh
- Department of Pediatric Gastroenterology, Hepatology and Nutrition, Baylor College of Medicine and Texas Children's Hospital, Houston, TX; Miami Transplant Institute, University of Miami, Miami, FL
| | - Alison A Bertuch
- Section of Hematology/Oncology, Department of Pediatrics, Baylor College of Medicine and Texas Children's Hospital, Houston, TX
| |
Collapse
|
50
|
Michniacki TF, Weyand AC. Gastrointestinal Bleeding: Expanding the Shortened Telomere Disorder Phenotype. J Pediatr 2021; 230:12-14. [PMID: 33121962 DOI: 10.1016/j.jpeds.2020.10.057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 10/25/2020] [Indexed: 10/23/2022]
Affiliation(s)
- Thomas F Michniacki
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, University of Michigan, Ann Arbor, Michigan
| | - Angela C Weyand
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|