1
|
Thompson AS, Niewisch MR, Giri N, McReynolds LJ, Savage SA. Germline RTEL1 Variants in Telomere Biology Disorders. Am J Med Genet A 2025; 197:e63882. [PMID: 39279436 DOI: 10.1002/ajmg.a.63882] [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: 06/21/2024] [Revised: 08/22/2024] [Accepted: 09/02/2024] [Indexed: 09/18/2024]
Abstract
Rare germline variation in regulator of telomere elongation helicase 1 (RTEL1) is associated with telomere biology disorders (TBDs). Biallelic RTEL1 variants result in childhood onset dyskeratosis congenita and Hoyeraal-Hreidarsson syndrome whereas heterozygous individuals usually present later in life with pulmonary fibrosis or bone marrow failure. We compiled all TBD-associated RTEL1 variants in the literature and assessed phenotypes and outcomes of 44 individuals from 14 families with mono- or biallelic RTEL1 variants enrolled in clinical trial NCT00027274. Variants were classified by adapting ACMG-AMP guidelines using clinical information, telomere length, and variant allele frequency data. Compared with heterozygotes, individuals with biallelic RTEL1 variants had an earlier age at diagnosis (median age 35.5 vs. 5.1 years, p < 0.01) and worse overall survival (median age 66.5 vs. 22.9 years, p < 0.001). There were 257 unique RTEL1 variants reported in 47 publications, and 209 had a gnomAD minor allele frequency <1%. Only 38.3% (80/209) met pathogenic/likely pathogenic criteria. Notably, 8 of 209 reported disease-associated variants were benign or likely benign and the rest were variants of uncertain significance. Given the considerable differences in outcomes of TBDs associated with RTEL1 germline variants and the extent of variation in the gene, systematic functional studies and standardization of variant curation are urgently needed to inform clinical management.
Collapse
Affiliation(s)
- Ashley S Thompson
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, USA
| | - Marena R Niewisch
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, USA
- Department of Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
| | - Neelam Giri
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, USA
| | - Lisa J McReynolds
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, USA
| | - Sharon A Savage
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, USA
| |
Collapse
|
2
|
Pedroza LA, van den Haak F, Frumovitz A, Hernandez E, Hegewisch-Solloa E, Orange TK, Sheehan KB, Prockop S, Bodansky A, Chinn IK, Lupski JR, Posey JE, Mace EM, Li Y, Orange JS. The Golgi complex governs natural killer cell lytic granule positioning to promote directionality in cytotoxicity. Cell Rep 2025; 44:115156. [PMID: 39813120 PMCID: PMC11844255 DOI: 10.1016/j.celrep.2024.115156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 08/26/2024] [Accepted: 12/13/2024] [Indexed: 01/18/2025] Open
Abstract
Cytotoxic immune cells mediate precise attacks against diseased cells to maintain organismal health. Their operational unit of killing and host defense is lytic granules (LGs), which are specialized lysosomal-related organelles. Precision in cytotoxicity is achieved by converging the many LGs to the microtubule-organizing center (MTOC) and polarizing these to the diseased cell for secretion. We identify unappreciated intimate relationships between the Golgi, MTOC, and LGs after cytotoxic cell activation, as well as the trans-Golgin protein GCC2 on the LG surface. GCC2 serves to tether LGs to the Golgi following convergence, and both GCC2 and the Golgi are required for the persistence of convergence. GCC2 allows LGs to utilize the Golgi as a docking station preventing LG dispersion and innocent bystander killing in complex three-dimensional environments. We also identify GCC2 variants causing human natural killer cell deficiency, further emphasizing the importance of LG convergence and Golgi linkage in precision targeting for human immunity.
Collapse
Affiliation(s)
- Luis A Pedroza
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | | | - Alexander Frumovitz
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Evelyn Hernandez
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | | | - Tabitha K Orange
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | | | | | - Aaron Bodansky
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Ivan K Chinn
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - James R Lupski
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jennifer E Posey
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Emily M Mace
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Yu Li
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Jordan S Orange
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
3
|
Yang C, Wang S, Gao G, Xu P, Qian M, Yin Y, Yao S, Huang Z, Bian Z. RTEL1 is upregulated in gastric cancer and promotes tumor growth. J Cancer Res Clin Oncol 2024; 151:23. [PMID: 39724284 PMCID: PMC11671550 DOI: 10.1007/s00432-024-06062-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 12/06/2024] [Indexed: 12/28/2024]
Abstract
Gastric cancer (GC) is one of the most common cancers worldwide, with increasing incidence and mortality rates. It is typically diagnosed at advanced stages, leading to a poor prognosis. GC is a highly heterogeneous disease and its progression is associated with complex interplay between genetic and environmental factors. Identifying novel genes and pathways involved in GC development is crucial for improving the therapeutic outcome. Regulator of Telomerase Length 1 (RTEL1) has been found to maintain telomere stability through its helicase activity, facilitating telomere reconstruction and repair. However, the precise role of RTEL1 in human cancers, particularly in GC, is not yet fully understood. In this study, we observed significantly increased RTEL1 expression in GC tissues, which was associated with a poor prognosis. Functionally, RTEL1 promotes GC cell proliferation both in vitro and in vivo. Additionally, RTEL1 appears to regulate multiple signaling pathways, with a particular promoting effect on the cell cycle progression. Notably, CDC23 and TRIP13 are potential downstream target genes of RTEL1, which may mediate its tumor-promoting effects in GC. These findings suggest that RTEL1 plays a critical role in GC tumorigenesis and could be a promising target for the therapy and prognosis of GC.
Collapse
Affiliation(s)
- Chunyu Yang
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, 200 Hui He Road, Wuxi, Jiangsu, 214062, China
- Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Suzeng Wang
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, 200 Hui He Road, Wuxi, Jiangsu, 214062, China
- Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Ge Gao
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, 200 Hui He Road, Wuxi, Jiangsu, 214062, China
- Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Peiwen Xu
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, 200 Hui He Road, Wuxi, Jiangsu, 214062, China
- Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Mengyuan Qian
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, 200 Hui He Road, Wuxi, Jiangsu, 214062, China
- Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Yuan Yin
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, 200 Hui He Road, Wuxi, Jiangsu, 214062, China
- Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Surui Yao
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, 200 Hui He Road, Wuxi, Jiangsu, 214062, China
- Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Zhaohui Huang
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, 200 Hui He Road, Wuxi, Jiangsu, 214062, China.
- Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214122, China.
| | - Zehua Bian
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, 200 Hui He Road, Wuxi, Jiangsu, 214062, China.
- Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214122, China.
| |
Collapse
|
4
|
Dixit S, Nagraj T, Bhattacharya D, Saxena S, Sahoo S, Chittela RK, Somyajit K, Nagaraju G. RTEL1 helicase counteracts RAD51-mediated homologous recombination and fork reversal to safeguard replicating genomes. Cell Rep 2024; 43:114594. [PMID: 39116203 DOI: 10.1016/j.celrep.2024.114594] [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: 10/05/2023] [Revised: 05/23/2024] [Accepted: 07/22/2024] [Indexed: 08/10/2024] Open
Abstract
Homologous recombination (HR) plays an essential role in the repair of DNA double-strand breaks (DSBs), replication stress responses, and genome maintenance. However, unregulated HR during replication can impair genome duplication and compromise genome stability. The mechanisms underlying HR regulation during DNA replication are obscure. Here, we find that RTEL1 helicase, RAD51, and RAD51 paralogs are enriched at stalled replication sites. The absence of RTEL1 leads to an increase in the RAD51-mediated HR and fork reversal during replication and affects genome-wide replication, which can be rescued by co-depleting RAD51 and RAD51 paralogs. Interestingly, co-depletion of fork remodelers such as SMARCAL1/ZRANB3/HLTF/FBH1 and expression of HR-defective RAD51 mutants also rescues replication defects in RTEL1-deficient cells. The anti-recombinase function of RTEL1 during replication depends on its interaction with PCNA and helicase activity. Together, our data identify the role of RTEL1 helicase in restricting RAD51-mediated fork reversal and HR activity to facilitate error-free genome duplication.
Collapse
Affiliation(s)
- Suruchi Dixit
- Department of Biochemistry Indian Institute of Science, Bangalore 560012, India
| | - Tarun Nagraj
- Department of Biochemistry Indian Institute of Science, Bangalore 560012, India
| | | | - Sneha Saxena
- Department of Biochemistry Indian Institute of Science, Bangalore 560012, India
| | - Satyaranjan Sahoo
- Department of Biochemistry Indian Institute of Science, Bangalore 560012, India
| | - Rajani Kant Chittela
- Applied Genomics Section, Bioscience Group, Bhabha Atomic Research Centre, Mumbai 400085, India
| | - Kumar Somyajit
- Department of Biochemistry Indian Institute of Science, Bangalore 560012, India; Functional Genomics & Metabolism Research Unit, Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, Odense M 5230, Denmark.
| | - Ganesh Nagaraju
- Department of Biochemistry Indian Institute of Science, Bangalore 560012, India.
| |
Collapse
|
5
|
Chaudhari S, Acharya LP, Jasti DB, Ware AP, Gorthi SP, Satyamoorthy K. Discovery of a Novel Shared Variant Among RTEL1 Gene and RTEL1-TNFRSF6B lncRNA at Chromosome 20q13.33 in Familial Progressive Myoclonus Epilepsy. Int J Genomics 2024; 2024:7518528. [PMID: 39156922 PMCID: PMC11330336 DOI: 10.1155/2024/7518528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 04/17/2024] [Accepted: 07/20/2024] [Indexed: 08/20/2024] Open
Abstract
Background: Progressive myoclonus epilepsy (PME) is a neurodegenerative disorder marked by recurrent seizures and progressive myoclonus. To date, based on the phenotypes and causal genes, more than 40 subtypes of PMEs have been identified, and more remain to be characterized. Our study is aimed at identifying the aberrant gene(s) possibly associated with PMEs in two siblings born to asymptomatic parents, in the absence of known genetic mutations. Methods: Clinical assessments and molecular analyses, such as the repeat expansion test for CSTB; SCA1, 2, 3, 6, and 7; whole exome sequencing (WES); and mitochondrial genome sequencing coupled with computational analysis, were performed. Results: A family-based segregation analysis of WES data was performed to identify novel genes associated with PMEs. The potassium channel, KCNH8 [c.298T>C; (p.Tyr100His)], a DNA repair gene, regulator of telomere elongation helicase 1 (RTEL1) [c.691G>T; (p.Asp231Tyr)] and long noncoding RNA, RTEL1-TNFRSF6B [chr20:62298898_G>T; NR_037882.1, hg19] were among the candidate genes that were found to be associated with PMEs. These homozygous variations in siblings belong to genes with a loss-of-function intolerant (pLI) score of ≤ 0.86, expected to be detrimental by multiple computational analyses, and were heterozygous in parents. Additionally, computational analysis and the expression of RTEL1 and RTEL1-TNFRSF6B revealed that RTEL1-TNFRSF6B may modulate RTEL1 via hsa-miR-3529-3p. In the patient with the severe phenotype, a further deleterious mutation in SLC22A17 was identified. No de novo variants specific to these probands were identified in the mitochondrial genome. Conclusions: Our study is the first to report variants in KCNH8, RTEL1, and RTEL1-TNFRSF6B among PME cases. These genes when characterized fully may shed light on pathogenicity and have the potential to be used in the diagnosis of PME.
Collapse
Affiliation(s)
- Sima Chaudhari
- Department of Cell and Molecular BiologyManipal School of Life SciencesManipal Academy of Higher Education 576104, Manipal, Karnataka, India
| | - Lavanya Prakash Acharya
- Department of Cell and Molecular BiologyManipal School of Life SciencesManipal Academy of Higher Education 576104, Manipal, Karnataka, India
| | - Dushyanth Babu Jasti
- Department of NeurologyKasturba Medical College 576104, Manipal, Karnataka, India
| | - Akshay Pramod Ware
- Department of BioinformaticsManipal School of Life SciencesManipal Academy of Higher Education 576104, Manipal, Karnataka, India
| | - Sankar Prasad Gorthi
- Department of NeurologyKasturba Medical College 576104, Manipal, Karnataka, India
- Department of NeurologyBharati Hospital and Research CenterBharati Vidyapeeth (Deemed to Be University) Medical College and Hospital, Dhankawadi 411043, Pune, Maharashtra, India
| | - Kapaettu Satyamoorthy
- Department of Cell and Molecular BiologyManipal School of Life SciencesManipal Academy of Higher Education 576104, Manipal, Karnataka, India
- SDM College of Medical Sciences and HospitalShri Dharmasthala Manjunatheshwara (SDM) University, Manjushree Nagar, Sattur 580009, Dharwad, Karnataka, India
| |
Collapse
|
6
|
Luo Q, Wang J, Ge W, Li Z, Mao Y, Wang C, Zhang L. Exploration of the potential causative genes for inflammatory bowel disease: Transcriptome-wide association analysis, Mendelian randomization analysis and Bayesian colocalisation. Heliyon 2024; 10:e28944. [PMID: 38617957 PMCID: PMC11015108 DOI: 10.1016/j.heliyon.2024.e28944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/27/2024] [Accepted: 03/27/2024] [Indexed: 04/16/2024] Open
Abstract
Background Inflammatory bowel disease (IBD) poses a complex challenge due to its intricate underlying mechanisms, and curative treatments remain elusive. Consequently, there is an urgent need to identify genes causally associated with IBD. Methods We extracted blood eQTL data from the GTExv8.ALL.Whole_Blood database, genome-wide association studies (GWAS) summary statistics of IBD from the IEU GWAS database, and performed a three-fold analysis protocol, including transcriptome-wide association analysis, Mendelian randomisation analysis, Bayesian colocalisation, and subsequent potential therapeutic agents identification. Results We identified four pathogenic genes, namely CARD9, RTEL1, STMN3 and ARFRP1, that promote the development of IBD, encompassing both ulcerative colitis (UC) and Crohn's disease (CD). Notably, ARFRP1 exhibited the ability to suppress IBD (encompassing UC and CD) development. Regarding drug prediction, cyclophosphamide emerged as a promising novel therapeutic option for IBD, encompassing UC and CD. Conclusion We identified several potential genes related to IBD (UC and CD), including CARD9, RTEL1, STMN3 and ARFRP1, warranting further investigation in functional studies to elucidate underlying disease mechanisms. Additionally, clinical studies exploring the potential of cyclophosphamide as a treatment avenue for IBD are warranted.
Collapse
Affiliation(s)
- Qinghua Luo
- Clinical Medical College, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Jiawen Wang
- Department of Proctology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wei Ge
- Department of Proctology, Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, China
| | - Zihao Li
- Office of the President, Jiangmen Wuyi Hospital of Traditional Chinese Medicine, Jiangmen, China
| | - Yuanting Mao
- Clinical Medical College, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Chen Wang
- Department of Proctology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Leichang Zhang
- Formula-Pattern Research Center, Jiangxi University of Chinese Medicine, Jiangxi, China
| |
Collapse
|
7
|
Hourvitz N, Awad A, Tzfati Y. The many faces of the helicase RTEL1 at telomeres and beyond. Trends Cell Biol 2024; 34:109-121. [PMID: 37532653 DOI: 10.1016/j.tcb.2023.07.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 07/01/2023] [Accepted: 07/05/2023] [Indexed: 08/04/2023]
Abstract
Regulator of telomere elongation 1 (RTEL1) is known as a DNA helicase that is important for telomeres and genome integrity. However, the diverse phenotypes of RTEL1 dysfunction, the wide spectrum of symptoms caused by germline RTEL1 mutations, and the association of RTEL1 mutations with cancers suggest that RTEL1 is a complex machine that interacts with DNA, RNA, and proteins, and functions in diverse cellular pathways. We summarize the proposed functions of RTEL1 and discuss their implications for telomere maintenance. Studying RTEL1 is crucial for understanding the complex interplay between telomere maintenance and other nuclear pathways, and how compromising these pathways causes telomere biology diseases, various aging-associated pathologies, and cancer.
Collapse
Affiliation(s)
- Noa Hourvitz
- Department of Genetics, The Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Safra Campus, Jerusalem 91904, Israel
| | - Aya Awad
- Department of Genetics, The Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Safra Campus, Jerusalem 91904, Israel
| | - Yehuda Tzfati
- Department of Genetics, The Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Safra Campus, Jerusalem 91904, Israel.
| |
Collapse
|
8
|
Schmit MM, Baxley RM, Wang L, Hinderlie P, Kaufman M, Simon E, Raju A, Miller JS, Bielinsky AK. A critical threshold of MCM10 is required to maintain genome stability during differentiation of induced pluripotent stem cells into natural killer cells. Open Biol 2024; 14:230407. [PMID: 38262603 PMCID: PMC10805602 DOI: 10.1098/rsob.230407] [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: 11/01/2023] [Accepted: 11/23/2023] [Indexed: 01/25/2024] Open
Abstract
Natural killer (NK) cell deficiency (NKD) is a rare disease in which NK cell function is reduced, leaving affected individuals susceptible to repeated viral infections and cancer. Recently, a patient with NKD was identified carrying compound heterozygous variants of MCM10 (minichromosome maintenance protein 10), an essential gene required for DNA replication, that caused a significant decrease in the amount of functional MCM10. NKD in this patient presented as loss of functionally mature late-stage NK cells. To understand how MCM10 deficiency affects NK cell development, we generated MCM10 heterozygous (MCM10+/-) induced pluripotent stem cell (iPSC) lines. Analyses of these cell lines demonstrated that MCM10 was haploinsufficient, similar to results in other human cell lines. Reduced levels of MCM10 in mutant iPSCs was associated with impaired clonogenic survival and increased genomic instability, including micronuclei formation and telomere erosion. The severity of these phenotypes correlated with the extent of MCM10 depletion. Significantly, MCM10+/- iPSCs displayed defects in NK cell differentiation, exhibiting reduced yields of hematopoietic stem cells (HSCs). Although MCM10+/- HSCs were able to give rise to lymphoid progenitors, these did not generate mature NK cells. The lack of mature NK cells coincided with telomere erosion, suggesting that NKD caused by these MCM10 variants arose from the accumulation of genomic instability including degradation of chromosome ends.
Collapse
Affiliation(s)
- Megan M. Schmit
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Ryan M. Baxley
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Liangjun Wang
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Peter Hinderlie
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Marissa Kaufman
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Emily Simon
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Anjali Raju
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Jeffrey S. Miller
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Anja-Katrin Bielinsky
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
9
|
Schertzer M, Jullien L, Pinto AL, Calado RT, Revy P, Londoño-Vallejo A. Human RTEL1 Interacts with KPNB1 (Importin β) and NUP153 and Connects Nuclear Import to Nuclear Envelope Stability in S-Phase. Cells 2023; 12:2798. [PMID: 38132118 PMCID: PMC10741959 DOI: 10.3390/cells12242798] [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: 11/07/2023] [Revised: 11/29/2023] [Accepted: 12/03/2023] [Indexed: 12/23/2023] Open
Abstract
Regulator of TElomere Length Helicase 1 (RTEL1) is a helicase required for telomere maintenance and genome replication and repair. RTEL1 has been previously shown to participate in the nuclear export of small nuclear RNAs. Here we show that RTEL1 deficiency leads to a nuclear envelope destabilization exclusively in cells entering S-phase and in direct connection to origin firing. We discovered that inhibiting protein import also leads to similar, albeit non-cell cycle-related, nuclear envelope disruptions. Remarkably, overexpression of wild-type RTEL1, or of its C-terminal part lacking the helicase domain, protects cells against nuclear envelope anomalies mediated by protein import inhibition. We identified distinct domains in the C-terminus of RTEL1 essential for the interaction with KPNB1 (importin β) and NUP153, respectively, and we demonstrated that, on its own, the latter domain can promote the dynamic nuclear internalization of peptides that freely diffuse through the nuclear pore. Consistent with putative functions exerted in protein import, RTEL1 can be visualized on both sides of the nuclear pore using high-resolution microscopy. In all, our work points to an unanticipated, helicase-independent, role of RTEL1 in connecting both nucleocytoplasmic trafficking and nuclear envelope integrity to genome replication initiation in S-phase.
Collapse
Affiliation(s)
- Michael Schertzer
- Institut Curie, PSL Research University, CNRS, UMR3244, F-75005 Paris, France;
- Sorbonne Universités, CNRS, UMR3244, F-75005 Paris, France
| | - Laurent Jullien
- INSERM UMR 1163, Laboratory of Genome Dynamics in the Immune System, Equipe Labellisée Ligue Contre le Cancer, F-75006 Paris, France; (L.J.); (P.R.)
- Paris Descartes–Sorbonne Paris Cité University, Imagine Institute, F-75015 Paris, France
| | - André L. Pinto
- Department of Medical Imaging, Hematology, and Oncology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto 14049-900, Brazil; (A.L.P.); (R.T.C.)
| | - Rodrigo T. Calado
- Department of Medical Imaging, Hematology, and Oncology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto 14049-900, Brazil; (A.L.P.); (R.T.C.)
| | - Patrick Revy
- INSERM UMR 1163, Laboratory of Genome Dynamics in the Immune System, Equipe Labellisée Ligue Contre le Cancer, F-75006 Paris, France; (L.J.); (P.R.)
- Paris Descartes–Sorbonne Paris Cité University, Imagine Institute, F-75015 Paris, France
| | - Arturo Londoño-Vallejo
- Institut Curie, PSL Research University, CNRS, UMR3244, F-75005 Paris, France;
- Sorbonne Universités, CNRS, UMR3244, F-75005 Paris, France
| |
Collapse
|
10
|
Wu Z, Gong Z, Li C, Huang Z. RTEL1 is upregulated in colorectal cancer and promotes tumor progression. Pathol Res Pract 2023; 252:154958. [PMID: 37988793 DOI: 10.1016/j.prp.2023.154958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 10/23/2023] [Accepted: 11/16/2023] [Indexed: 11/23/2023]
Abstract
As one of the most common cancers worldwide, the incidence of colorectal cancer (CRC) continues to increase. Metastasis is the leading cause of death for this malignant disease. Regulator of telomere elongation helicase 1 (RTEL1) is a key factor that maintains telomere stability and contributes to DNA repair. Recent advances have suggested that RTEL1 exerts other functions through various mechanisms. However, little is known about its role in human cancers, including CRC. In this study, we revealed that the copy number of RTEL1 was significantly higher in CRC tissues, especially in metastatic CRC tissues, than in paired normal tissues, which was associated with increased expression. Increased RTEL1 expression was significantly correlated with CRC progression and poor survival. Functionally, RTEL1 knockdown suppressed the proliferation and metastasis of CRC both in vitro and in vivo. In addition, multiple signalling pathways, including the mitotic cell cycle, DNA repair, and EMT, were potentially regulated by RTEL1. Notably, GPR17 appeared to be a candidate downstream target gene that partially mediated the tumor-promoting effects of RTEL1 in CRC. Altogether, our results indicate that RTEL1 plays a crucial role in CRC progression and appears to be a promising therapeutic target and prognosis for CRC.
Collapse
Affiliation(s)
- Zhuochao Wu
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi 214062, Jiangsu, China; Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi 214062, Jiangsu, China
| | - Zhicheng Gong
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi 214062, Jiangsu, China; Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi 214062, Jiangsu, China
| | - Chaoqun Li
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi 214062, Jiangsu, China; Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi 214062, Jiangsu, China
| | - Zhaohui Huang
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi 214062, Jiangsu, China; Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi 214062, Jiangsu, China.
| |
Collapse
|
11
|
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
|
12
|
Alsultan A, Abujoub R, Alsudairy R, Memon S, Jarrar MS, Alafghani S, Aldaama S, Ballourah W, Almanjomi F, Essa MF. Human leucocyte antigen-matched related haematopoietic stem cell transplantation using low-dose cyclophosphamide, fludarabine and thymoglobulin in children with severe aplastic anaemia. Br J Haematol 2023; 203:255-263. [PMID: 37491781 DOI: 10.1111/bjh.19004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/17/2023] [Accepted: 07/10/2023] [Indexed: 07/27/2023]
Abstract
When human leucocyte antigen-matched related donors are available, haematopoietic stem cell transplantation (HSCT) in children with severe aplastic anaemia (SAA) represents the standard of care. Cyclophosphamide (Cy) 200 mg/kg and anti-thymocyte globulin (ATG) are frequently administered, but to-date, no standard conditioning regimen exists. In this study, we investigated the efficacy of a unified HSCT conditioning protocol consisting of low-dose Cy 80 mg/kg, fludarabine and ATG. Data were reviewed from children aged ≤14 years with either acquired SAA or non-Fanconi anaemia inherited bone marrow failure syndrome (IBMFS) between 2011 and 2022 at various Saudi institutions. Graft-versus-host disease (GVHD) prophylaxis included mycophenolate mofetil and calcineurin inhibitors. HSCT was performed in 32 children (17 females and 15 males). Nine patients had deleterious mutations (two ERCC6L2, two ANKRD26, two TINF2, one LZTFL1, one RTEL1 and one DNAJC21). Four patients had short telomeres. All 32 patients engrafted successfully. At 3 years post-transplant, the event-free survival was 93% and overall survival was 95%. Two patients experienced secondary graft failure or myelodysplastic syndrome. A low probability of GVHD was observed (one acute GVHD II and one mild chronic GVHD). These data highlight how HSCT using low-dose Cy as part of a fludarabine-based regimen is safe and effective in SAA/non-Fanconi anaemia IBMFS.
Collapse
Affiliation(s)
- Abdulrahman Alsultan
- Department of Pediatrics, College of Medicine, King Saud University, Riyadh, Saudi Arabia
- Oncology Center, King Saud University Medical City, Riyadh, Saudi Arabia
- Department of Pediatric Hematology/Oncology and Stem Cell Transplantation, Comprehensive Cancer Center, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Rodaina Abujoub
- Department of Nursing, King Abdullah Specialist Children's Hospital, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
- Department of Pediatric Hematology/Oncology, King Abdullah Specialist Children's Hospital, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Reem Alsudairy
- Department of Pediatric Hematology/Oncology, King Abdullah Specialist Children's Hospital, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Shahbaz Memon
- Department of Pediatric Hematology/Oncology, King Abdullah Specialist Children's Hospital, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Mohammad S Jarrar
- Schulich School of Medicine, University of Western Ontario, London, Ontario, Canada
- Windsor Regional Hospital and Cancer Center, Windsor, Ontario, Canada
| | - Sameera Alafghani
- Department of Pediatric Hematology/Oncology and Stem Cell Transplantation, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Saad Aldaama
- Department of Pediatric Hematology/Oncology and Stem Cell Transplantation, King Fahad Specialist Hospital, Dammam, Saudi Arabia
| | - Walid Ballourah
- Department of Pediatric Hematology/Oncology and Stem Cell Transplantation, Comprehensive Cancer Center, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Fahd Almanjomi
- Department of Pediatric Hematology/Oncology and Stem Cell Transplantation, Comprehensive Cancer Center, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Mohammed F Essa
- Department of Pediatric Hematology/Oncology, King Abdullah Specialist Children's Hospital, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| |
Collapse
|
13
|
Kawashima N, Bezzerri V, Corey SJ. The Molecular and Genetic Mechanisms of Inherited Bone Marrow Failure Syndromes: The Role of Inflammatory Cytokines in Their Pathogenesis. Biomolecules 2023; 13:1249. [PMID: 37627314 PMCID: PMC10452082 DOI: 10.3390/biom13081249] [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: 07/18/2023] [Revised: 08/09/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
Inherited bone marrow failure syndromes (IBMFSs) include Fanconi anemia, Diamond-Blackfan anemia, Shwachman-Diamond syndrome, dyskeratosis congenita, severe congenital neutropenia, and other rare entities such as GATA2 deficiency and SAMD9/9L mutations. The IBMFS monogenic disorders were first recognized by their phenotype. Exome sequencing has validated their classification, with clusters of gene mutations affecting DNA damage response (Fanconi anemia), ribosome structure (Diamond-Blackfan anemia), ribosome assembly (Shwachman-Diamond syndrome), or telomere maintenance/stability (dyskeratosis congenita). The pathogenetic mechanisms of IBMFSs remain to be characterized fully, but an overarching hypothesis states that different stresses elicit TP53-dependent growth arrest and apoptosis of hematopoietic stem, progenitor, and precursor cells. Here, we review the IBMFSs and propose a role for pro-inflammatory cytokines, such as TGF-β, IL-1β, and IFN-α, in mediating the cytopenias. We suggest a pathogenic role for cytokines in the transformation to myeloid neoplasia and hypothesize a role for anti-inflammatory therapies.
Collapse
Affiliation(s)
- Nozomu Kawashima
- Departments of Pediatrics and Cancer Biology, Cleveland Clinic, Cleveland, OH 44195, USA;
| | - Valentino Bezzerri
- Cystic Fibrosis Center, Azienda Ospedaliera Universitaria Integrata, 37126 Verona, Italy;
| | - Seth J. Corey
- Departments of Pediatrics and Cancer Biology, Cleveland Clinic, Cleveland, OH 44195, USA;
| |
Collapse
|
14
|
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
|
15
|
Tobai H, Endo M, Ishimura M, Moriya K, Yano J, Kanamori K, Sato N, Amanuma F, Maruyama H, Muramatsu H, Shibahara J, Narita M, Fumoto S, Peltier D, Ohga S. Neonatal intestinal obstruction in Hoyeraal-Hreidarsson syndrome with novel RTEL1 variants. Pediatr Blood Cancer 2023; 70:e30250. [PMID: 36776130 DOI: 10.1002/pbc.30250] [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: 11/03/2022] [Revised: 01/22/2023] [Accepted: 01/24/2023] [Indexed: 02/14/2023]
Affiliation(s)
- Hiromi Tobai
- Division of Pediatrics, Iwate Prefectural Iwai Hospital, Ichinoseki, Japan.,Department of Pediatrics, Graduate School of Medical Sciences, Kyusyu University, Fukuoka, Japan
| | - Mikiya Endo
- Department of Pediatrics, Iwate Medical University, School of Medicine, Morioka, Japan
| | - Masataka Ishimura
- Department of Pediatrics, Graduate School of Medical Sciences, Kyusyu University, Fukuoka, Japan
| | - Kunihiko Moriya
- Division of Pediatrics, Iwate Prefectural Iwai Hospital, Ichinoseki, Japan.,Department of Pediatrics, National Defense Medical College, Tokorozawa, Japan
| | - Jun Yano
- Division of Pediatrics, Iwate Prefectural Iwai Hospital, Ichinoseki, Japan
| | - Keita Kanamori
- Division of Pediatrics, Iwate Prefectural Iwai Hospital, Ichinoseki, Japan
| | - Norio Sato
- Division of Pediatrics, Iwate Prefectural Iwai Hospital, Ichinoseki, Japan
| | - Fumitaka Amanuma
- Division of Pediatrics, Iwate Prefectural Iwai Hospital, Ichinoseki, Japan
| | - Hidekazu Maruyama
- Division of Pediatrics, Iwate Prefectural Iwai Hospital, Ichinoseki, Japan
| | - Hideki Muramatsu
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Junji Shibahara
- Department of Pathology, Kyorin University School of Medicine, Mitaka, Japan
| | - Masami Narita
- Department of Pediatrics, Kyorin University School of Medicine, Mitaka, Japan
| | - Seiko Fumoto
- Department of Pediatrics, Kyorin University School of Medicine, Mitaka, Japan
| | - Daniel Peltier
- Pediatric Hematology and Oncology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Shouichi Ohga
- Department of Pediatrics, Graduate School of Medical Sciences, Kyusyu University, Fukuoka, Japan
| |
Collapse
|
16
|
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
|
17
|
Nisar H, Khan M, Chaudhry QUN, Iftikhar R, Ghafoor T. Case report: A novel mutation in RTEL1 gene in dyskeratosis congenita. Front Oncol 2023; 13:1098876. [PMID: 36937416 PMCID: PMC10017992 DOI: 10.3389/fonc.2023.1098876] [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: 11/15/2022] [Accepted: 02/13/2023] [Indexed: 03/06/2023] Open
Abstract
Dyskeratosis congenita (DKC), also known as Zinsser-Cole-Engman syndrome, is a telomeropathy typically presenting as a triad of leukoplakia, nail dystrophy, and reticular hyperpigmentation. Reported genetic mutations linked to DKC include DKC1, TINF2, TERC, TERT, C16orf57, NOLA2, NOLA3, WRAP53/TCAB1, and RTEL1. Homozygous, compound heterozygous, and heterozygous mutations in RTEL1 (RTEL1, regulator of telomere elongation helicase 1) gene on chromosome 20q13 are known to cause autosomal dominant as well as recessive DKC. Pathogenic variants of RTEL1 gene in DKC patients include c.2288G>T (p. Gly763Val), c.3791G>A (p. Arg1264His), and RTEL p. Arg981Trp. We report a novel homozygous variant of RTEL1, transcript ID: ENST00000360203.11, exon 24, c.2060C>T (p.Ala687Val), in a patient of DKC presenting with leukoplakia, dystrophic nails, reticulate pigmentation, and positive family history of a similar phenotype. The novel variant, reported as a variant of uncertain significance, may therefore be considered diagnostic for DKC in a Pakistani population.
Collapse
Affiliation(s)
- Haider Nisar
- Adult and Pediatric Transplant Unit, Armed Forces Bone Marrow Transplant Center/National Institute of Bone Marrow Transplant, Rawalpindi, Pakistan
| | - Memoona Khan
- Pathology Department and Stem Cell Research Lab, Armed Forces Bone Marrow Transplant Center/National Institute of Bone Marrow Transplant, Rawalpindi, Pakistan
- *Correspondence: Memoona Khan,
| | - Qamar Un Nisa Chaudhry
- Adult and Pediatric Transplant Unit, Armed Forces Bone Marrow Transplant Center/National Institute of Bone Marrow Transplant, Rawalpindi, Pakistan
- Pathology Department and Stem Cell Research Lab, Armed Forces Bone Marrow Transplant Center/National Institute of Bone Marrow Transplant, Rawalpindi, Pakistan
| | - Raheel Iftikhar
- Adult and Pediatric Transplant Unit, Armed Forces Bone Marrow Transplant Center/National Institute of Bone Marrow Transplant, Rawalpindi, Pakistan
| | - Tariq Ghafoor
- Adult and Pediatric Transplant Unit, Armed Forces Bone Marrow Transplant Center/National Institute of Bone Marrow Transplant, Rawalpindi, Pakistan
| |
Collapse
|
18
|
Hassani MA, Murid J, Yan J. Regulator of telomere elongation helicase 1 gene and its association with malignancy. Cancer Rep (Hoboken) 2022; 6:e1735. [PMID: 36253342 PMCID: PMC9875622 DOI: 10.1002/cnr2.1735] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 09/26/2022] [Accepted: 09/28/2022] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND With the progression of next-generation sequencing technologies, researchers have identified numerous variants of the regulator of telomere elongation helicase 1 (RTEL1) gene that are associated with a broad spectrum of phenotypic manifestations, including malignancies. At the molecular level, RTEL1 is involved in the regulation of the repair, replication, and transcription of deoxyribonucleic acid (DNA) and the maintenance of telomere length. RTEL1 can act both as a promotor and inhibitor of tumorigenesis. Here, we review the potential mechanisms implicated in the malignant transformation of tissues under conditions of RTEL1 deficiency or its aberrant overexpression. RECENT FINDINGS A major hemostatic challenge during RTEL1 dysfunction could arise from its unbalanced activity for unwinding guanine-rich quadruplex DNA (G4-DNA) structures. In contrast, RTEL1 deficiency leads to alterations in telomeric and genome-wide DNA maintenance mechanisms, ribonucleoprotein metabolism, and the creation of an inflammatory and immune-deficient microenvironment, all promoting malignancy. Additionally, we hypothesize that functionally similar molecules could act to compensate for the deteriorated functions of RTEL1, thereby facilitating the survival of malignant cells. On the contrary, RTEL1 over-expression was directed toward G4-unwinding, by promoting replication fork progression and maintaining intact telomeres, may facilitate malignant transformation and proliferation of various pre-malignant cellular compartments. CONCLUSIONS Therefore, restoring the equilibrium of RTEL1 functions could serve as a therapeutic approach for preventing and treating malignancies.
Collapse
Affiliation(s)
- Mohammad Arian Hassani
- Department of Hematology, Liaoning Medical Center for Hematopoietic Stem Cell Transplantation, Liaoning Key Laboratory of Hematopoietic Stem Cell Transplantation and Translational Medicine, Dalian Key Laboratory of HematologySecond Hospital of Dalian Medical UniversityDalianChina,Department of Hematology, Endocrinology and Rheumatology, Ali Abad Teaching HospitalKabul University of Medical SciencesJamal menaKabulAfghanistan
| | - Jamshid Murid
- Department of Hematology, Endocrinology and Rheumatology, Ali Abad Teaching HospitalKabul University of Medical SciencesJamal menaKabulAfghanistan
| | - Jinsong Yan
- Department of Hematology, Liaoning Medical Center for Hematopoietic Stem Cell Transplantation, Liaoning Key Laboratory of Hematopoietic Stem Cell Transplantation and Translational Medicine, Dalian Key Laboratory of HematologySecond Hospital of Dalian Medical UniversityDalianChina,Diamond Bay Institute of HematologySecond Hospital of Dalian Medical UniversityDalianChina
| |
Collapse
|
19
|
Ouahed JD. Understanding inborn errors of immunity: A lens into the pathophysiology of monogenic inflammatory bowel disease. Front Immunol 2022; 13:1026511. [PMID: 36248828 PMCID: PMC9556666 DOI: 10.3389/fimmu.2022.1026511] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 09/13/2022] [Indexed: 11/13/2022] Open
Abstract
Inflammatory bowel diseases (IBD) are chronic inflammatory conditions of the gastrointestinal tract, including Crohn’s disease, ulcerative colitis and inflammatory bowel disease-undefined (IBD-U). IBD are understood to be multifactorial, involving genetic, immune, microbial and environmental factors. Advances in next generation sequencing facilitated the growing identification of over 80 monogenic causes of IBD, many of which overlap with Inborn errors of immunity (IEI); Approximately a third of currently identified IEI result in gastrointestinal manifestations, many of which are inflammatory in nature, such as IBD. Indeed, the gastrointestinal tract represents an opportune system to study IEI as it consists of the largest mass of lymphoid tissue in the body and employs a thin layer of intestinal epithelial cells as the critical barrier between the intestinal lumen and the host. In this mini-review, a selection of pertinent IEI resulting in monogenic IBD is described involving disorders in the intestinal epithelial barrier, phagocytosis, T and B cell defects, as well as those impairing central and peripheral tolerance. The contribution of disrupted gut-microbiota-host interactions in disturbing intestinal homeostasis among patients with intestinal disease is also discussed. The molecular mechanisms driving pathogenesis are reviewed along with the personalized therapeutic interventions and investigational avenues this growing knowledge has enabled.
Collapse
|
20
|
Kohzaki M. Mammalian Resilience Revealed by a Comparison of Human Diseases and Mouse Models Associated With DNA Helicase Deficiencies. Front Mol Biosci 2022; 9:934042. [PMID: 36032672 PMCID: PMC9403131 DOI: 10.3389/fmolb.2022.934042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 06/23/2022] [Indexed: 12/01/2022] Open
Abstract
Maintaining genomic integrity is critical for sustaining individual animals and passing on the genome to subsequent generations. Several enzymes, such as DNA helicases and DNA polymerases, are involved in maintaining genomic integrity by unwinding and synthesizing the genome, respectively. Indeed, several human diseases that arise caused by deficiencies in these enzymes have long been known. In this review, the author presents the DNA helicases associated with human diseases discovered to date using recent analyses, including exome sequences. Since several mouse models that reflect these human diseases have been developed and reported, this study also summarizes the current knowledge regarding the outcomes of DNA helicase deficiencies in humans and mice and discusses possible mechanisms by which DNA helicases maintain genomic integrity in mammals. It also highlights specific diseases that demonstrate mammalian resilience, in which, despite the presence of genomic instability, patients and mouse models have lifespans comparable to those of the general population if they do not develop cancers; finally, this study discusses future directions for therapeutic applications in humans that can be explored using these mouse models.
Collapse
|
21
|
Wang F, Li Y, Zhao S, Chen Z, Xu Z, Wang L, Zhang TJ, Yan J, Cao L, Wang P, Li A, Zhong Y, Wu Z, Qi X, Zhang M, Wu N. The utility of hierarchical genetic testing in paediatric liver disease. Liver Int 2022; 42:1097-1108. [PMID: 35257483 DOI: 10.1111/liv.15235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 10/20/2021] [Accepted: 11/01/2021] [Indexed: 02/13/2023]
Abstract
BACKGROUND & AIMS Genetic factors underlie a substantial proportion of paediatric liver diseases. Hereditary liver diseases have considerable genetic heterogeneity and variable clinical manifestations, which bring great challenges to clinical and molecular diagnoses. In this study, we investigated a group of paediatric patients with varying degrees of liver dysfunction using a hierarchical genetic testing strategy. METHODS We first applied a panel encompassing 166 known causal genes of liver disease. We then used exome sequencing (ES) in those patients whose cases remained undiagnosed to identify the genetic aetiology of their symptoms. RESULTS In total, we enrolled 131 unrelated paediatric patients with liver disease of Chinese Han ethnicity. We first applied targeted gene sequencing of 166 genes to all patients and yielded a diagnostic rate of 35.9% (47 of 131). Eighty-four patients who remained undiagnosed after target gene sequencing were subjected to ES. As a result, eight (8/84, 9.5%) of them obtained molecular diagnoses, including four patients suspected of abnormal bilirubin metabolism and four idiopathic cases. Non-typical genetic findings, including digenic inheritance and dual molecular diagnosis, were also identified. Through a comprehensive assessment of novel candidate variants of uncertain disease association, 11 patients of the remaining undiagnosed patients were able to obtain likely molecular diagnoses. CONCLUSIONS Our study presents evidence for the diagnostic utility of sequential genetic testing in a cohort of patients with paediatric liver disease. Our findings expand the understanding of the phenotypic and mutational spectrum underlying this heterogeneous group of diseases.
Collapse
Affiliation(s)
- Fuchuan Wang
- Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yaqi Li
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of Big Data for Spinal Deformities, Chinese Academy of Medical Sciences, Beijing, China.,Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, China
| | - Sen Zhao
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of Big Data for Spinal Deformities, Chinese Academy of Medical Sciences, Beijing, China.,Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, China
| | - Zefu Chen
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of Big Data for Spinal Deformities, Chinese Academy of Medical Sciences, Beijing, China.,Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, China
| | - Zhiqiang Xu
- Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Lianlei Wang
- Department of Orthopedic Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, P. R. China
| | - Terry Jianguo Zhang
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of Big Data for Spinal Deformities, Chinese Academy of Medical Sciences, Beijing, China.,Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, China
| | - Jianguo Yan
- Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Lili Cao
- Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Pu Wang
- Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Aiqin Li
- Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yanwei Zhong
- Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Zhihong Wu
- Key Laboratory of Big Data for Spinal Deformities, Chinese Academy of Medical Sciences, Beijing, China.,Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, China.,Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaolong Qi
- CHESS Center, Institute of Portal Hypertension, The First Hospital of Lanzhou University, Lanzhou, China
| | - Min Zhang
- Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Nan Wu
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of Big Data for Spinal Deformities, Chinese Academy of Medical Sciences, Beijing, China.,Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, China
| |
Collapse
|
22
|
Kermasson L, Churikov D, Awad A, Smoom R, Lainey E, Touzot F, Audebert-Bellanger S, Haro S, Roger L, Costa E, Mouf M, Bottero A, Oleastro M, Abdo C, de Villartay JP, Géli V, Tzfati Y, Callebaut I, Danielian S, Soares G, Kannengiesser C, Revy P. Inherited human Apollo deficiency causes severe bone marrow failure and developmental defects. Blood 2022; 139:2427-2440. [PMID: 35007328 PMCID: PMC11022855 DOI: 10.1182/blood.2021010791] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 12/13/2021] [Indexed: 11/20/2022] Open
Abstract
Inherited bone marrow failure syndromes (IBMFSs) are a group of disorders typified by impaired production of 1 or several blood cell types. The telomere biology disorders dyskeratosis congenita (DC) and its severe variant, Høyeraal-Hreidarsson (HH) syndrome, are rare IBMFSs characterized by bone marrow failure, developmental defects, and various premature aging complications associated with critically short telomeres. We identified biallelic variants in the gene encoding the 5'-to-3' DNA exonuclease Apollo/SNM1B in 3 unrelated patients presenting with a DC/HH phenotype consisting of early-onset hypocellular bone marrow failure, B and NK lymphopenia, developmental anomalies, microcephaly, and/or intrauterine growth retardation. All 3 patients carry a homozygous or compound heterozygous (in combination with a null allele) missense variant affecting the same residue L142 (L142F or L142S) located in the catalytic domain of Apollo. Apollo-deficient cells from patients exhibited spontaneous chromosome instability and impaired DNA repair that was complemented by CRISPR/Cas9-mediated gene correction. Furthermore, patients' cells showed signs of telomere fragility that were not associated with global reduction of telomere length. Unlike patients' cells, human Apollo KO HT1080 cell lines showed strong telomere dysfunction accompanied by excessive telomere shortening, suggesting that the L142S and L142F Apollo variants are hypomorphic. Collectively, these findings define human Apollo as a genome caretaker and identify biallelic Apollo variants as a genetic cause of a hitherto unrecognized severe IBMFS that combines clinical hallmarks of DC/HH with normal telomere length.
Collapse
Affiliation(s)
- Laëtitia Kermasson
- Laboratory of Genome Dynamics in the Immune System, Laboratoire labellisé Ligue Naionale contre le Cancer, INSERM UMR 1163, Université de Paris, Imagine Institute, Paris, France
| | - Dmitri Churikov
- U1068 INSERM, Unité Mixte de Recherche (UMR) 7258 (CNRS), Equipe Labellisée Ligue Nationale Contre le Cancer, Marseille Cancer Research Center (CRCM), Institut Paoli-Calmettes, Aix Marseille University, Marseille, France
| | - Aya Awad
- Department of Genetics, The Silberman Institute of Life Science, The Hebrew University of Jerusalem, Safra Campus-Givat Ram, Jerusalem, Israel
| | - Riham Smoom
- Department of Genetics, The Silberman Institute of Life Science, The Hebrew University of Jerusalem, Safra Campus-Givat Ram, Jerusalem, Israel
| | - Elodie Lainey
- Hematology Laboratory, Robert Debré Hospital-Assistance Publique-Hôpitaux de Paris (APHP); INSERM UMR 1131-Hematology University Institute-Denis Diderot School of Medicine, Paris, France
| | - Fabien Touzot
- Department of Immunology-Rheumatology, Department of Pediatrics, Centre Hospitalier Universitaire (CHU), Sainte Justine Research Center, Université de Montréal, Montréal, Quebec, Canada
| | | | - Sophie Haro
- Department of Paediatrics and Medical Genetics, CHU de Brest, Brest, France
| | - Lauréline Roger
- Structure and Instability of Genomes laboratory, “Muséum National d'Histoire Naturelle” (MNHN), INSERM U1154, CNRS UMR 7196, Paris, France
| | - Emilia Costa
- Serviço de Pediatria, Centro Hospitalar e Universitário do Porto, Porto, Portugal
| | - Maload Mouf
- 68HAL Meddle Laboratory, Zenon Skelter Institute, Green Hills, Eggum, Norway
| | | | - Matias Oleastro
- Rheumathology and Immunology Service, Hospital Nacional de Pediatría JP Garrahan, Buenos Aires, Argentina
| | - Chrystelle Abdo
- Onco-Hematology, Assistance Publique-Hôpitaux de Paris, Université de Paris and Institut Necker Enfants Malades, Paris, France
| | - Jean-Pierre de Villartay
- Laboratory of Genome Dynamics in the Immune System, Laboratoire labellisé Ligue Naionale contre le Cancer, INSERM UMR 1163, Université de Paris, Imagine Institute, Paris, France
| | - Vincent Géli
- U1068 INSERM, Unité Mixte de Recherche (UMR) 7258 (CNRS), Equipe Labellisée Ligue Nationale Contre le Cancer, Marseille Cancer Research Center (CRCM), Institut Paoli-Calmettes, Aix Marseille University, Marseille, France
| | - Yehuda Tzfati
- Department of Genetics, The Silberman Institute of Life Science, The Hebrew University of Jerusalem, Safra Campus-Givat Ram, Jerusalem, Israel
| | - Isabelle Callebaut
- UMR CNRS 7590, Institut de Minéralogie, de Physique des Matériaux et de Cosmochimie (IMPMC), Muséum National d'Histoire Naturelle, Sorbonne Université, Paris, France
| | - Silvia Danielian
- Department of Immunology, JP Garrahan National Hospital of Pediatrics, Buenos Aires, Argentina
| | - Gabriela Soares
- Centro de Genética Médica Jacinto de Magalhães, Centro Hospitalar e Universitário do Porto, Porto, Portugal
| | - Caroline Kannengiesser
- Service de Génétique, Assistance Publique des Hôpitaux de Paris, Hôpital Bichat, Université Paris Diderot, Paris, France
| | - Patrick Revy
- Laboratory of Genome Dynamics in the Immune System, Laboratoire labellisé Ligue Naionale contre le Cancer, INSERM UMR 1163, Université de Paris, Imagine Institute, Paris, France
| |
Collapse
|
23
|
Wilkins BJ, Kelsen JR, Conrad MA. A Pattern-based Pathology Approach to Very Early-onset Inflammatory Bowel Disease: Thinking Beyond Crohn Disease and Ulcerative Colitis. Adv Anat Pathol 2022; 29:62-70. [PMID: 34813528 PMCID: PMC8665089 DOI: 10.1097/pap.0000000000000327] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Very early-onset inflammatory bowel disease (VEO-IBD), IBD diagnosed in children younger than 6 years old, is phenotypically and genetically distinct from older onset IBD. Monogenic and digenic causative defects, particularly in primary immunodeficiency and intestinal epithelial barrier genes, have been identified in a subset of patients with VEO-IBD allowing for targeted therapies and improved outcomes. However, these findings are the minority, thus strategies to correctly diagnose patients, including identification of specific histopathologic findings with correlating clinical and laboratory features may provide critical and necessary insight into mechanisms of disease pathogenesis and subsequent therapeutic options. In this article, we review the pathologic findings seen in patients with VEO-IBD and outline a pattern-based approach to diagnosis using examples from primary immunodeficiencies with gastrointestinal manifestations.
Collapse
Affiliation(s)
- Benjamin J. Wilkins
- Division of Anatomic Pathology, Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104 USA
| | - Judith R. Kelsen
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children’s Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104 USA
| | - Maire A. Conrad
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children’s Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104 USA
| |
Collapse
|
24
|
Ruiz A, Flores-Gonzalez J, Buendia-Roldan I, Chavez-Galan L. Telomere Shortening and Its Association with Cell Dysfunction in Lung Diseases. Int J Mol Sci 2021; 23:425. [PMID: 35008850 PMCID: PMC8745057 DOI: 10.3390/ijms23010425] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/22/2021] [Accepted: 12/29/2021] [Indexed: 01/10/2023] Open
Abstract
Telomeres are localized at the end of chromosomes to provide genome stability; however, the telomere length tends to be shortened with each cell division inducing a progressive telomere shortening (TS). In addition to age, other factors, such as exposure to pollutants, diet, stress, and disruptions in the shelterin protein complex or genes associated with telomerase induce TS. This phenomenon favors cellular senescence and genotoxic stress, which increases the risk of the development and progression of lung diseases such as idiopathic pulmonary fibrosis, chronic obstructive pulmonary disease, SARS-CoV-2 infection, and lung cancer. In an infectious environment, immune cells that exhibit TS are associated with severe lymphopenia and death, whereas in a noninfectious context, naïve T cells that exhibit TS are related to cancer progression and enhanced inflammatory processes. In this review, we discuss how TS modifies the function of the immune system cells, making them inefficient in maintaining homeostasis in the lung. Finally, we discuss the advances in drug and gene therapy for lung diseases where TS could be used as a target for future treatments.
Collapse
Affiliation(s)
| | | | | | - Leslie Chavez-Galan
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (A.R.); (J.F.-G.); (I.B.-R.)
| |
Collapse
|
25
|
Baxley RM, Leung W, Schmit MM, Matson JP, Yin L, Oram MK, Wang L, Taylor J, Hedberg J, Rogers CB, Harvey AJ, Basu D, Taylor JC, Pagnamenta AT, Dreau H, Craft J, Ormondroyd E, Watkins H, Hendrickson EA, Mace EM, Orange JS, Aihara H, Stewart GS, Blair E, Cook JG, Bielinsky AK. Bi-allelic MCM10 variants associated with immune dysfunction and cardiomyopathy cause telomere shortening. Nat Commun 2021; 12:1626. [PMID: 33712616 PMCID: PMC7955084 DOI: 10.1038/s41467-021-21878-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 02/11/2021] [Indexed: 12/22/2022] Open
Abstract
Minichromosome maintenance protein 10 (MCM10) is essential for eukaryotic DNA replication. Here, we describe compound heterozygous MCM10 variants in patients with distinctive, but overlapping, clinical phenotypes: natural killer (NK) cell deficiency (NKD) and restrictive cardiomyopathy (RCM) with hypoplasia of the spleen and thymus. To understand the mechanism of MCM10-associated disease, we modeled these variants in human cell lines. MCM10 deficiency causes chronic replication stress that reduces cell viability due to increased genomic instability and telomere erosion. Our data suggest that loss of MCM10 function constrains telomerase activity by accumulating abnormal replication fork structures enriched with single-stranded DNA. Terminally-arrested replication forks in MCM10-deficient cells require endonucleolytic processing by MUS81, as MCM10:MUS81 double mutants display decreased viability and accelerated telomere shortening. We propose that these bi-allelic variants in MCM10 predispose specific cardiac and immune cell lineages to prematurely arrest during differentiation, causing the clinical phenotypes observed in both NKD and RCM patients.
Collapse
Affiliation(s)
- Ryan M Baxley
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Wendy Leung
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Megan M Schmit
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Jacob Peter Matson
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Lulu Yin
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Marissa K Oram
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Liangjun Wang
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, 55455, USA
| | - John Taylor
- Oxford Medical Genetics Laboratories, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Jack Hedberg
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Colette B Rogers
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Adam J Harvey
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Debashree Basu
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Jenny C Taylor
- Wellcome Centre Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
- Oxford NIHR Biomedical Research Centre, Oxford, OX3 7BN, UK
| | - Alistair T Pagnamenta
- Wellcome Centre Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
- Oxford NIHR Biomedical Research Centre, Oxford, OX3 7BN, UK
| | - Helene Dreau
- Department of Haematology, University of Oxford, Oxford, OX3 7BN, UK
| | - Jude Craft
- Oxford Centre for Genomic Medicine, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Elizabeth Ormondroyd
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Hugh Watkins
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Eric A Hendrickson
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Emily M Mace
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Jordan S Orange
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Hideki Aihara
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Grant S Stewart
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Edward Blair
- Oxford Centre for Genomic Medicine, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Jeanette Gowen Cook
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Anja-Katrin Bielinsky
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, 55455, USA.
| |
Collapse
|
26
|
Schmit M, Bielinsky AK. Congenital Diseases of DNA Replication: Clinical Phenotypes and Molecular Mechanisms. Int J Mol Sci 2021; 22:E911. [PMID: 33477564 PMCID: PMC7831139 DOI: 10.3390/ijms22020911] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 01/13/2021] [Accepted: 01/14/2021] [Indexed: 12/19/2022] Open
Abstract
Deoxyribonucleic acid (DNA) replication can be divided into three major steps: initiation, elongation and termination. Each time a human cell divides, these steps must be reiteratively carried out. Disruption of DNA replication can lead to genomic instability, with the accumulation of point mutations or larger chromosomal anomalies such as rearrangements. While cancer is the most common class of disease associated with genomic instability, several congenital diseases with dysfunctional DNA replication give rise to similar DNA alterations. In this review, we discuss all congenital diseases that arise from pathogenic variants in essential replication genes across the spectrum of aberrant replisome assembly, origin activation and DNA synthesis. For each of these conditions, we describe their clinical phenotypes as well as molecular studies aimed at determining the functional mechanisms of disease, including the assessment of genomic stability. By comparing and contrasting these diseases, we hope to illuminate how the disruption of DNA replication at distinct steps affects human health in a surprisingly cell-type-specific manner.
Collapse
Affiliation(s)
| | - Anja-Katrin Bielinsky
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA;
| |
Collapse
|
27
|
Human mutational constraint as a tool to understand biology of rare and emerging bone marrow failure syndromes. Blood Adv 2020; 4:5232-5245. [PMID: 33104793 DOI: 10.1182/bloodadvances.2020002687] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 09/16/2020] [Indexed: 12/17/2022] Open
Abstract
Inherited bone marrow failure (IBMF) syndromes are rare blood disorders characterized by hematopoietic cell dysfunction and predisposition to hematologic malignancies. Despite advances in the understanding of molecular pathogenesis of these heterogeneous diseases, genetic variant interpretation, genotype-phenotype correlation, and outcome prognostication remain difficult. As new IBMF and other myelodysplastic syndrome (MDS) predisposition genes continue to be discovered (frequently in small kindred studies), there is an increasing need for a systematic framework to evaluate penetrance and prevalence of mutations in genes associated with IBMF phenotypes. To address this need, we analyzed population-based genomic data from >125 000 individuals in the Genome Aggregation Database for loss-of-function (LoF) variants in 100 genes associated with IBMF. LoF variants in genes associated with IBMF/MDS were present in 0.426% of individuals. Heterozygous LoF variants in genes in which haploinsufficiency is associated with IBMF/MDS were identified in 0.422% of the population; homozygous LoF variants associated with autosomal recessive IBMF/MDS diseases were identified in only .004% of the cohort. Using age distribution of LoF variants and 2 measures of mutational constraint, LOEUF ("loss-of-function observed/expected upper bound fraction") and pLI ("probability of being loss-of-function intolerance"), we evaluated the pathogenicity, tolerance, and age-related penetrance of LoF mutations in specific genes associated with IBMF syndromes. This analysis led to insights into rare IBMF diseases, including syndromes associated with DHX34, MDM4, RAD51, SRP54, and WIPF1. Our results provide an important population-based framework for the interpretation of LoF variant pathogenicity in rare and emerging IBMF syndromes.
Collapse
|
28
|
Ziv A, Werner L, Konnikova L, Awad A, Jeske T, Hastreiter M, Mitsialis V, Stauber T, Wall S, Kotlarz D, Klein C, Snapper SB, Tzfati Y, Weiss B, Somech R, Shouval DS. An RTEL1 Mutation Links to Infantile-Onset Ulcerative Colitis and Severe Immunodeficiency. J Clin Immunol 2020; 40:1010-1019. [PMID: 32710398 DOI: 10.1007/s10875-020-00829-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 07/16/2020] [Indexed: 12/27/2022]
Abstract
PURPOSE More than 50 different monogenic disorders causing inflammatory bowel disease (IBD) have been identified. Our goal was to characterize the clinical phenotype, genetic workup, and immunologic alterations in an Ashkenazi Jewish patient that presented during infancy with ulcerative colitis and unique clinical manifestations. METHODS Immune workup and whole-exome sequencing were performed, along with Sanger sequencing for confirmation. Next-generation sequencing of the TCRB and IgH was conducted for immune repertoire analysis. Telomere length was evaluated by in-gel hybridization assay. Mass cytometry was performed on patient's peripheral blood mononuclear cells, and compared with control subjects and patients with UC. RESULTS The patient presented in infancy with failure to thrive and dysmorphic features, consistent with a diagnosis of dyskeratosis congenita and Hoyeraal-Hreidarsson syndrome. Severe ulcerative colitis manifested in the first year of life and proceeded to the development of a primary immunodeficiency, presenting as Pneumocystis jiroveci pneumonia and hypogammaglobulinemia. Genetic studies identified a deleterious homozygous C.3791G>A missense mutation in the helicase regulator of telomere elongation 1 (RTEL1), leading to short telomeres in the index patient. Immune repertoire studies showed polyclonal T and B cell receptor distribution, while mass cytometry analysis demonstrated marked immunological alterations, including a predominance of naïve T cells, paucity of B cells, and a decrease in various innate immune subsets. CONCLUSIONS RTEL1 mutations are associated with significant alterations in immune landscape and can manifest with infantile-onset IBD. A high index of suspicion is required in Ashkenazi Jewish families where the carriage rate of the C.3791G>A variant is high.
Collapse
Affiliation(s)
- Alma Ziv
- Pediatric Department A, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel
- 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
| | - Lael Werner
- 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
| | - Liza Konnikova
- Division of Newborn Medicine, Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Aya Awad
- Department of Genetics, The Silberman Institute of Life Science, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Tim Jeske
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, LMU Munich, Munich, Germany
| | - Maximilian Hastreiter
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, LMU Munich, Munich, Germany
| | - Vanessa Mitsialis
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Tali Stauber
- Pediatric Department A, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Immunology Service, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel
- Jeffrey Modell Foundation Center, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel
| | - Sarah Wall
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA
| | - Daniel Kotlarz
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, LMU Munich, Munich, Germany
| | - Christoph Klein
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, LMU Munich, Munich, Germany
| | - Scott B Snapper
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Yehuda Tzfati
- Department of Genetics, The Silberman Institute of Life Science, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Batia Weiss
- 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 Department A, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Immunology Service, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel
- Jeffrey Modell Foundation Center, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel
| | - 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.
| |
Collapse
|
29
|
Zeng T, Lv G, Chen X, Yang L, Zhou L, Dou Y, Tang X, Yang J, An Y, Zhao X. CD8 + T-cell senescence and skewed lymphocyte subsets in young Dyskeratosis Congenita patients with PARN and DKC1 mutations. J Clin Lab Anal 2020; 34:e23375. [PMID: 32452087 PMCID: PMC7521304 DOI: 10.1002/jcla.23375] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 04/16/2020] [Accepted: 05/01/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Dyskeratosis congenita (DC) is a syndrome resulting from defective telomere maintenance. Immunodeficiency associated with DC can cause significant morbidity and lead to premature mortality, but the immunological characteristics and molecular hallmark of DC patients, especially young patients, have not been described in detail. METHODS We summarize the clinical data of two juvenile patients with DC. Gene mutations were identified by whole-exome and direct sequencing. Swiss-PdbViewer was used to predict the pathogenicity of identified mutations. The relative telomere length was determined by QPCR, and a comprehensive analysis of lymphocyte subsets and CD57 expression was performed by flow cytometry. RESULTS Both patients showed typical features of DC without severe infection. In addition, patient 1 (P1) was diagnosed with Hoyeraal-Hreidarsson syndrome due to cerebellar hypoplasia. Gene sequencing showed P1 had a compound heterozygous mutation (c.204G > T and c.178-245del) in PARN and P2 had a novel hemizygous mutation in DKC1 (c.1051A > G). Lymphocyte subset analysis showed B and NK cytopenia, an inverted CD4:CD8 ratio, and decreased naïve CD4 and CD8 cells. A significant increase in CD21low B cells and skewed numbers of helper T cells (Th), regulatory T cells (Treg), follicular regulatory T cells (Tfr), and follicular helper T cells (Tfh) were also detected. Short telomere lengths, increased CD57 expression, and an expansion of CD8 effector memory T cells re-expressing CD45RA (TEMRA) were also found in both patients. CONCLUSION Unique immunologic abnormalities, CD8 T-cell senescence, and shortened telomere together as a hallmark occur in young DC patients before progression to severe disease.
Collapse
Affiliation(s)
- Ting Zeng
- Pediatric Research InstituteMinistry of Education Key Laboratory of Child Development and DisordersNational Clinical Research Center for Child Health and Disorders (Chongqing)China International Science and Technology Cooperation base of Child Development and Critical DisordersChildren's Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of Child Infection and ImmunityChildren's Hospital of Chongqing Medical UniversityChongqingChina
| | - Ge Lv
- Pediatric Research InstituteMinistry of Education Key Laboratory of Child Development and DisordersNational Clinical Research Center for Child Health and Disorders (Chongqing)China International Science and Technology Cooperation base of Child Development and Critical DisordersChildren's Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of Child Infection and ImmunityChildren's Hospital of Chongqing Medical UniversityChongqingChina
| | - Xuemei Chen
- Pediatric Research InstituteMinistry of Education Key Laboratory of Child Development and DisordersNational Clinical Research Center for Child Health and Disorders (Chongqing)China International Science and Technology Cooperation base of Child Development and Critical DisordersChildren's Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of Child Infection and ImmunityChildren's Hospital of Chongqing Medical UniversityChongqingChina
| | - Lu Yang
- Pediatric Research InstituteMinistry of Education Key Laboratory of Child Development and DisordersNational Clinical Research Center for Child Health and Disorders (Chongqing)China International Science and Technology Cooperation base of Child Development and Critical DisordersChildren's Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of Child Infection and ImmunityChildren's Hospital of Chongqing Medical UniversityChongqingChina
| | - Lina Zhou
- Pediatric Research InstituteMinistry of Education Key Laboratory of Child Development and DisordersNational Clinical Research Center for Child Health and Disorders (Chongqing)China International Science and Technology Cooperation base of Child Development and Critical DisordersChildren's Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of Child Infection and ImmunityChildren's Hospital of Chongqing Medical UniversityChongqingChina
| | - Ying Dou
- Pediatric Research InstituteMinistry of Education Key Laboratory of Child Development and DisordersNational Clinical Research Center for Child Health and Disorders (Chongqing)China International Science and Technology Cooperation base of Child Development and Critical DisordersChildren's Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of Child Infection and ImmunityChildren's Hospital of Chongqing Medical UniversityChongqingChina
- Department of Hematology and OncologyChildren's Hospital of Chongqing Medical UniversityChongqingChina
| | - Xuemei Tang
- Pediatric Research InstituteMinistry of Education Key Laboratory of Child Development and DisordersNational Clinical Research Center for Child Health and Disorders (Chongqing)China International Science and Technology Cooperation base of Child Development and Critical DisordersChildren's Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of Child Infection and ImmunityChildren's Hospital of Chongqing Medical UniversityChongqingChina
- Department of Rheumatology and ImmunologyChildren's Hospital of Chongqing Medical UniversityChongqingChina
| | - Jun Yang
- Pediatric Research InstituteMinistry of Education Key Laboratory of Child Development and DisordersNational Clinical Research Center for Child Health and Disorders (Chongqing)China International Science and Technology Cooperation base of Child Development and Critical DisordersChildren's Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of Child Infection and ImmunityChildren's Hospital of Chongqing Medical UniversityChongqingChina
- Department of Rheumatology and ImmunologyShenzhen Children's HospitalShenzhenChina
| | - Yunfei An
- Pediatric Research InstituteMinistry of Education Key Laboratory of Child Development and DisordersNational Clinical Research Center for Child Health and Disorders (Chongqing)China International Science and Technology Cooperation base of Child Development and Critical DisordersChildren's Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of Child Infection and ImmunityChildren's Hospital of Chongqing Medical UniversityChongqingChina
- Department of Rheumatology and ImmunologyChildren's Hospital of Chongqing Medical UniversityChongqingChina
| | - Xiaodong Zhao
- Pediatric Research InstituteMinistry of Education Key Laboratory of Child Development and DisordersNational Clinical Research Center for Child Health and Disorders (Chongqing)China International Science and Technology Cooperation base of Child Development and Critical DisordersChildren's Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of Child Infection and ImmunityChildren's Hospital of Chongqing Medical UniversityChongqingChina
- Department of Rheumatology and ImmunologyChildren's Hospital of Chongqing Medical UniversityChongqingChina
| |
Collapse
|
30
|
Awad A, Glousker G, Lamm N, Tawil S, Hourvitz N, Smoom R, Revy P, Tzfati Y. Full length RTEL1 is required for the elongation of the single-stranded telomeric overhang by telomerase. Nucleic Acids Res 2020; 48:7239-7251. [PMID: 32542379 PMCID: PMC7367169 DOI: 10.1093/nar/gkaa503] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 05/29/2020] [Accepted: 06/02/2020] [Indexed: 12/17/2022] Open
Abstract
Telomeres cap the ends of eukaryotic chromosomes and distinguish them from broken DNA ends to suppress DNA damage response, cell cycle arrest and genomic instability. Telomeres are elongated by telomerase to compensate for incomplete replication and nuclease degradation and to extend the proliferation potential of germ and stem cells and most cancers. However, telomeres in somatic cells gradually shorten with age, ultimately leading to cellular senescence. Hoyeraal-Hreidarsson syndrome (HHS) is characterized by accelerated telomere shortening and diverse symptoms including bone marrow failure, immunodeficiency, and neurodevelopmental defects. HHS is caused by germline mutations in telomerase subunits, factors essential for its biogenesis and recruitment to telomeres, and in the helicase RTEL1. While diverse phenotypes were associated with RTEL1 deficiency, the telomeric role of RTEL1 affected in HHS is yet unknown. Inducible ectopic expression of wild-type RTEL1 in patient fibroblasts rescued the cells, enabled telomerase-dependent telomere elongation and suppressed the abnormal cellular phenotypes, while silencing its expression resulted in gradual telomere shortening. Our observations reveal an essential role of the RTEL1 C-terminus in facilitating telomerase action at the telomeric 3' overhang. Thus, the common etiology for HHS is the compromised telomerase action, resulting in telomere shortening and reduced lifespan of telomerase positive cells.
Collapse
Affiliation(s)
- Aya Awad
- Department of Genetics, The Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Givat Ram, Jerusalem 91904, Israel
| | - Galina Glousker
- Department of Genetics, The Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Givat Ram, Jerusalem 91904, Israel
| | - Noa Lamm
- Department of Genetics, The Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Givat Ram, Jerusalem 91904, Israel
| | - Shadi Tawil
- Department of Genetics, The Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Givat Ram, Jerusalem 91904, Israel
| | - Noa Hourvitz
- Department of Genetics, The Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Givat Ram, Jerusalem 91904, Israel
| | - Riham Smoom
- Department of Genetics, The Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Givat Ram, Jerusalem 91904, Israel
| | - Patrick Revy
- INSERM UMR 1163, Laboratory of Genome Dynamics in the Immune System, Equipe Labellisée Ligue contre le Cancer and Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, Paris, France
| | - Yehuda Tzfati
- Department of Genetics, The Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Givat Ram, Jerusalem 91904, Israel
| |
Collapse
|
31
|
Nonsense Suppression Therapy: New Hypothesis for the Treatment of Inherited Bone Marrow Failure Syndromes. Int J Mol Sci 2020; 21:ijms21134672. [PMID: 32630050 PMCID: PMC7369780 DOI: 10.3390/ijms21134672] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 06/29/2020] [Accepted: 06/29/2020] [Indexed: 12/13/2022] Open
Abstract
Inherited bone marrow failure syndromes (IBMFS) are a group of cancer-prone genetic diseases characterized by hypocellular bone marrow with impairment in one or more hematopoietic lineages. The pathogenesis of IBMFS involves mutations in several genes which encode for proteins involved in DNA repair, telomere biology and ribosome biogenesis. The classical IBMFS include Shwachman–Diamond syndrome (SDS), Diamond–Blackfan anemia (DBA), Fanconi anemia (FA), dyskeratosis congenita (DC), and severe congenital neutropenia (SCN). IBMFS are associated with high risk of myelodysplastic syndrome (MDS), acute myeloid leukemia (AML), and solid tumors. Unfortunately, no specific pharmacological therapies have been highly effective for IBMFS. Hematopoietic stem cell transplantation provides a cure for aplastic or myeloid neoplastic complications. However, it does not affect the risk of solid tumors. Since approximately 28% of FA, 24% of SCN, 21% of DBA, 20% of SDS, and 17% of DC patients harbor nonsense mutations in the respective IBMFS-related genes, we discuss the use of the nonsense suppression therapy in these diseases. We recently described the beneficial effect of ataluren, a nonsense suppressor drug, in SDS bone marrow hematopoietic cells ex vivo. A similar approach could be therefore designed for treating other IBMFS. In this review we explain in detail the new generation of nonsense suppressor molecules and their mechanistic roles. Furthermore, we will discuss strengths and limitations of these molecules which are emerging from preclinical and clinical studies. Finally we discuss the state-of-the-art of preclinical and clinical therapeutic studies carried out for IBMFS.
Collapse
|
32
|
Björkman A, Johansen SL, Lin L, Schertzer M, Kanellis DC, Katsori AM, Christensen ST, Luo Y, Andersen JS, Elsässer SJ, Londono-Vallejo A, Bartek J, Schou KB. Human RTEL1 associates with Poldip3 to facilitate responses to replication stress and R-loop resolution. Genes Dev 2020; 34:1065-1074. [PMID: 32561545 PMCID: PMC7397856 DOI: 10.1101/gad.330050.119] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 05/18/2020] [Indexed: 02/07/2023]
Abstract
In this study from Björkman et al., the authors sought to understand how RTEL1 helicase preserves genomic stability during replication. They demonstrate that RTEL1 and the Polδ subunit Poldip3 form a complex and are mutually dependent in chromatin binding after replication stress, and loss of RTEL1 and Poldip3 leads to marked R-loop accumulation that is confined to sites of active replication, thus highlighting a previously unknown role of RTEL1 and Poldip3 in R-loop suppression at genomic regions where transcription and replication intersect. RTEL1 helicase is a component of DNA repair and telomere maintenance machineries. While RTEL1's role in DNA replication is emerging, how RTEL1 preserves genomic stability during replication remains elusive. Here we used a range of proteomic, biochemical, cell, and molecular biology and gene editing approaches to provide further insights into potential role(s) of RTEL1 in DNA replication and genome integrity maintenance. Our results from complementary human cell culture models established that RTEL1 and the Polδ subunit Poldip3 form a complex and are/function mutually dependent in chromatin binding after replication stress. Loss of RTEL1 and Poldip3 leads to marked R-loop accumulation that is confined to sites of active replication, enhances endogenous replication stress, and fuels ensuing genomic instability. The impact of depleting RTEL1 and Poldip3 is epistatic, consistent with our proposed concept of these two proteins operating in a shared pathway involved in DNA replication control under stress conditions. Overall, our data highlight a previously unsuspected role of RTEL1 and Poldip3 in R-loop suppression at genomic regions where transcription and replication intersect, with implications for human diseases including cancer.
Collapse
Affiliation(s)
- Andrea Björkman
- Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Science for Life Laboratory, Karolinska Institute, Solna 171 77, Sweden
| | - Søren L Johansen
- Department of Cell Biology and Physiology, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Lin Lin
- Department of Biomedicine, Aarhus University, Aarhus 8200, Denmark.,Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus 8200, Denmark
| | - Mike Schertzer
- 3UMR 3244 (Telomere and Cancer Laboratory), Centre National de la Recherche Scientifique, Institut Curie, PSL Research University, Sorbonne Universités, Paris 75005, France
| | - Dimitris C Kanellis
- Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Science for Life Laboratory, Karolinska Institute, Solna 171 77, Sweden
| | - Anna-Maria Katsori
- Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Science for Life Laboratory, Karolinska Institute, Solna 171 77, Sweden
| | - Søren T Christensen
- Department of Cell Biology and Physiology, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Yonglun Luo
- Department of Biomedicine, Aarhus University, Aarhus 8200, Denmark.,Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus 8200, Denmark
| | - Jens S Andersen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, DK-5230 Odense M, Denmark
| | - Simon J Elsässer
- Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Science for Life Laboratory, Karolinska Institute, Solna 171 77, Sweden
| | - Arturo Londono-Vallejo
- 3UMR 3244 (Telomere and Cancer Laboratory), Centre National de la Recherche Scientifique, Institut Curie, PSL Research University, Sorbonne Universités, Paris 75005, France
| | - Jiri Bartek
- Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Science for Life Laboratory, Karolinska Institute, Solna 171 77, Sweden.,Danish Cancer Society Research Centre, DK-2100 Copenhagen, Denmark
| | - Kenneth B Schou
- Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Science for Life Laboratory, Karolinska Institute, Solna 171 77, Sweden
| |
Collapse
|
33
|
Kelsen JR, Sullivan KE, Rabizadeh S, Singh N, Snapper S, Elkadri A, Grossman AB. North American Society for Pediatric Gastroenterology, Hepatology, and Nutrition Position Paper on the Evaluation and Management for Patients With Very Early-onset Inflammatory Bowel Disease. J Pediatr Gastroenterol Nutr 2020; 70:389-403. [PMID: 32079889 DOI: 10.1097/mpg.0000000000002567] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The rate of pediatric inflammatory bowel disease (IBD) has been increasing over the last decade and this increase has occurred most rapidly in the youngest children diagnosed <6 years, known as very early-onset inflammatory bowel disease (VEO-IBD). These children can present with more extensive and severe disease than older children and adults. The contribution of host genetics in this population is underscored by the young age of onset and the distinct, aggressive phenotype. In fact, monogenic defects, often involving primary immunodeficiency genes, have been identified in children with VEO-IBD and have led to targeted and life-saving therapy. This position paper will discuss the phenotype of VEO-IBD and outline the approach and evaluation for these children and what factors should trigger concern for an underlying immunodeficiency. We will then review the immunological assays and genetic studies that can facilitate the identification of the underlying diagnosis in patients with VEO-IBD and how this evaluation may lead to directed therapies. The position paper will also aid the pediatric gastroenterologist in recognizing when a patient should be referred to a center specializing in the care of these patients. These guidelines are intended for pediatricians, allied health professionals caring for children, pediatric gastroenterologists, pediatric pathologists, and immunologists.
Collapse
Affiliation(s)
| | - Kathleen E Sullivan
- Division of Immunology and Allergy, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Shervin Rabizadeh
- Division of Gastroenterology, Hepatology, and Nutrition, Cedar-Sinai Medical Center, Los Angeles, CA
| | - Namita Singh
- Division of Gastroenterology, Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, WA
| | - Scott Snapper
- Division of Gastroenterology, Hepatology, and Nutrition, Boston Children's Hospital, Department of Pediatrics, Harvard Medical School
- Division of Gastroenterology, Department of Medicine, Brigham & Women's Hospital and Harvard Medical School, Boston, MA
| | - Abdul Elkadri
- Division of Gastroenterology, Hepatology, and Nutrition, Medical College of Wisconsin, Milwaukee, WI
| | | |
Collapse
|
34
|
Gutierrez-Rodrigues F, Masri N, Chouery E, Diamond C, Jalkh N, Vicente A, Kajigaya S, Abillama F, Bejjani N, Serhal W, Calado RT, Young NS, Farhat H, Coussa ML. A novel homozygous RTEL1 variant in a consanguineous Lebanese family: phenotypic heterogeneity and disease anticipation. Hum Genet 2019; 138:1323-1330. [PMID: 31677132 PMCID: PMC9809984 DOI: 10.1007/s00439-019-02076-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 10/22/2019] [Indexed: 01/06/2023]
Abstract
Phenotypic heterogeneity is often observed in patients with telomeropathies caused by pathogenic variants in telomere biology genes. However, the roles of recessive variants in these different phenotypes are not fully characterized. Our goal is to describe the biological roles of a novel homozygous RTEL1 variant identified in a consanguineous Lebanese family with unusual presentation of telomeropathies. A proband was screened for germline variants in telomere biology genes by whole exome sequencing. Leukocytes' telomere length was measured in the proband and eight relatives. We identified a novel homozygous p.E665K RTEL1 variant in the proband, his mother, and seven siblings that associated with telomere shortening and a broad spectrum of clinical manifestations, ranging from mild unspecific findings to severe phenotypes. Consanguinity in at least three family generations led to increased frequency of the homozygous p.E665K variant in the youngest generation and progressive telomere shortening. The increased frequency of the homozygous RTEL1 variant due to consanguinity in this Lebanese family allowed us to infer novel behaviors of recessive RTEL1 variants, as the expressivity and penetrance of this gene are very heterogenous between inter- and intra-generations. Progressive telomere shortening was associated with disease anticipation, first reported in recessive autosomal telomeropathies. Both genetic testing and telomere length measurement were critical for the clinical diagnosis of this family with telomere diseases marked by phenotypic heterogeneity.
Collapse
Affiliation(s)
| | - Nohad Masri
- LAU Gilbert and Rose-Marie Chagoury School of Medicine, LAUMC/RH, Zahar Street, Achrafieh, Beirut, 1110, Lebanon
| | - Eliane Chouery
- Unité de Génétique Médicale, Faculty of Medicine, Saint Joseph University, Beirut, Lebanon
| | - Carrie Diamond
- Hematology Branch, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, MD, 20892-1202, USA
| | - Nadine Jalkh
- Unité de Génétique Médicale, Faculty of Medicine, Saint Joseph University, Beirut, Lebanon
| | - Alana Vicente
- Hematology Branch, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, MD, 20892-1202, USA
| | - Sachiko Kajigaya
- Hematology Branch, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, MD, 20892-1202, USA
| | - Fayez Abillama
- LAU Gilbert and Rose-Marie Chagoury School of Medicine, LAUMC/RH, Zahar Street, Achrafieh, Beirut, 1110, Lebanon
| | - Noha Bejjani
- LAU Gilbert and Rose-Marie Chagoury School of Medicine, LAUMC/RH, Zahar Street, Achrafieh, Beirut, 1110, Lebanon
| | - Wassim Serhal
- LAU Gilbert and Rose-Marie Chagoury School of Medicine, LAUMC/RH, Zahar Street, Achrafieh, Beirut, 1110, Lebanon
| | - Rodrigo T Calado
- Department of Medical Imaging, Hematology, and Clinical Oncology, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Neal S Young
- Hematology Branch, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, MD, 20892-1202, USA
| | - Hussein Farhat
- LAU Gilbert and Rose-Marie Chagoury School of Medicine, LAUMC/RH, Zahar Street, Achrafieh, Beirut, 1110, Lebanon.
| | - Marie Louise Coussa
- LAU Gilbert and Rose-Marie Chagoury School of Medicine, LAUMC/RH, Zahar Street, Achrafieh, Beirut, 1110, Lebanon
| |
Collapse
|
35
|
Ley B, Torgerson DG, Oldham JM, Adegunsoye A, Liu S, Li J, Elicker BM, Henry TS, Golden JA, Jones KD, Dressen A, Yaspan BL, Arron JR, Noth I, Hoffmann TJ, Wolters PJ. Rare Protein-Altering Telomere-related Gene Variants in Patients with Chronic Hypersensitivity Pneumonitis. Am J Respir Crit Care Med 2019; 200:1154-1163. [PMID: 31268371 PMCID: PMC6888660 DOI: 10.1164/rccm.201902-0360oc] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Accepted: 07/02/2019] [Indexed: 12/30/2022] Open
Abstract
Rationale: Rare genetic variants in telomere-related genes have been identified in familial, idiopathic, and rheumatoid arthritis-associated pulmonary fibrosis. Short peripheral blood leukocyte (PBL) telomere length predicts poor outcomes in chronic hypersensitivity pneumonitis (CHP).Objectives: Determine the prevalence and clinical relevance of rare protein-altering variants in telomere-related genes in patients with CHP.Methods: Next-generation sequences from two CHP cohorts were analyzed to identify variants in TERT (telomerase reverse transcriptase), TERC (telomerase RNA component), DKC1 (dyskerin pseudouridine synthase 1), RTEL1 (regulator of telomere elongation helicase 1), PARN (poly[A]-specific RNase), and TINF2 (TERF1-interacting nuclear factor 2). To qualify, variants were required to have a minor allele frequency less than 0.005 and be predicted to be damaging to protein function. Variant status (binary variable) was used in statistical association tests, including Cox proportional hazard models for transplant-free survival. PBL telomere length was measured using quantitative PCR.Measurements and Main Results: Qualifying variants were identified in 16 of 144 patients (11.1%; 95% confidence interval [CI], 6.5-17.4) in the discovery cohort and 17 of 209 patients (8.1%; 95% CI, 4.8-12.7) in the replication cohort. Age- and ancestry-adjusted PBL telomere length was significantly shorter in the presence of a variant in both cohorts (discovery: -561 bp; 95% CI, -933 to -190; P = 0.003; replication: -612 bp; 95% CI, -870 to -354; P = 5.30 × 10-6). Variant status was significantly associated with transplant-free survival in both cohorts (discovery: age-, sex-, and ancestry-adjusted hazard ratio, 3.73; 95% CI, 1.92-7.28; P = 0.0001; replication: hazard ratio, 2.72; 95% CI, 1.26-5.88; P = 0.011).Conclusions: A substantial proportion of patients diagnosed with CHP have rare, protein-altering variants in telomere-related genes, which are associated with short peripheral blood telomere length and significantly reduced transplant-free survival.
Collapse
Affiliation(s)
| | - Dara G. Torgerson
- Department of Human Genetics, McGill University and Genome Quebec Innovation Centre, Montreal, Quebec, Canada
| | - Justin M. Oldham
- Department of Medicine, University of California Davis, Davis, California
| | | | - Shuo Liu
- Department of Respiratory Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, People’s Republic of China
| | - Jie Li
- University of California Davis Bioinformatics Core, Davis, California
| | | | | | | | | | - Amy Dressen
- Genentech, South San Francisco, California; and
| | | | | | - Imre Noth
- Department of Medicine, University of Virginia, Charlottesville, Virginia
| | - Thomas J. Hoffmann
- Department of Epidemiology and Biostatistics, Institute for Human Genetics, University of California San Francisco, San Francisco, California
| | | |
Collapse
|
36
|
Porreca RM, Glousker G, Awad A, Matilla Fernandez MI, Gibaud A, Naucke C, Cohen SB, Bryan TM, Tzfati Y, Draskovic I, Londoño-Vallejo A. Human RTEL1 stabilizes long G-overhangs allowing telomerase-dependent over-extension. Nucleic Acids Res 2019. [PMID: 29522136 PMCID: PMC5961080 DOI: 10.1093/nar/gky173] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Telomere maintenance protects the cell against genome instability and senescence. Accelerated telomere attrition is a characteristic of premature aging syndromes including Dyskeratosis congenita (DC). Mutations in hRTEL1 are associated with a severe form of DC called Hoyeraal-Hreidarsson syndrome (HHS). HHS patients carry short telomeres and HHS cells display telomere damage. Here we investigated how hRTEL1 contributes to telomere maintenance in human primary as well as tumor cells. Transient depletion of hRTEL1 resulted in rapid telomere shortening only in the context of telomerase-positive cells with very long telomeres and high levels of telomerase. The effect of hRTEL1 on telomere length is telomerase dependent without impacting telomerase biogenesis or targeting of the enzyme to telomeres. Instead, RTEL1 depletion led to a decrease in both G-overhang content and POT1 association with telomeres with limited telomere uncapping. Strikingly, overexpression of POT1 restored telomere length but not the overhang, demonstrating that G-overhang loss is the primary defect caused by RTEL1 depletion. We propose that hRTEL1 contributes to the maintenance of long telomeres by preserving long G-overhangs, thereby facilitating POT1 binding and elongation by telomerase.
Collapse
Affiliation(s)
- Rosa M Porreca
- Institut Curie, PSL Research University, Sorbonne Universités, CNRS UMR3244 Telomere and cancer lab, 75005 Paris, France
| | - Galina Glousker
- Department of Genetics, The Silberman Institute of Life Science, The Hebrew University of Jerusalem, Safra Campus-Givat Ram, Jerusalem 91904, Israel
| | - Aya Awad
- Department of Genetics, The Silberman Institute of Life Science, The Hebrew University of Jerusalem, Safra Campus-Givat Ram, Jerusalem 91904, Israel
| | | | - Anne Gibaud
- Institut Curie, PSL Research University, Sorbonne Universités, CNRS UMR3244 Telomere and cancer lab, 75005 Paris, France
| | - Christian Naucke
- Institut Curie, PSL Research University, Sorbonne Universités, CNRS UMR3244 Telomere and cancer lab, 75005 Paris, France
| | - Scott B Cohen
- Children's Medical Research Institute, University of Sydney, Westmead, NSW 2145, Australia
| | - Tracy M Bryan
- Children's Medical Research Institute, University of Sydney, Westmead, NSW 2145, Australia
| | - Yehuda Tzfati
- Department of Genetics, The Silberman Institute of Life Science, The Hebrew University of Jerusalem, Safra Campus-Givat Ram, Jerusalem 91904, Israel
| | - Irena Draskovic
- Institut Curie, PSL Research University, Sorbonne Universités, CNRS UMR3244 Telomere and cancer lab, 75005 Paris, France
| | - Arturo Londoño-Vallejo
- Institut Curie, PSL Research University, Sorbonne Universités, CNRS UMR3244 Telomere and cancer lab, 75005 Paris, France
| |
Collapse
|
37
|
Borie R, Bouvry D, Cottin V, Gauvain C, Cazes A, Debray MP, Cadranel J, Dieude P, Degot T, Dominique S, Gamez AS, Jaillet M, Juge PA, Londono-Vallejo A, Mailleux A, Mal H, Boileau C, Menard C, Nunes H, Prevot G, Quetant S, Revy P, Traclet J, Wemeau-Stervinou L, Wislez M, Kannengiesser C, Crestani B. Regulator of telomere length 1 ( RTEL1) mutations are associated with heterogeneous pulmonary and extra-pulmonary phenotypes. Eur Respir J 2019; 53:13993003.00508-2018. [PMID: 30523160 DOI: 10.1183/13993003.00508-2018] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 11/06/2018] [Indexed: 01/10/2023]
Abstract
Regulator of telomere length 1 (RTEL1) mutations have been evidenced in 5-9% of familial pulmonary fibrosis; however, the phenotype of patients with interstitial lung disease (ILD) and RTEL1 mutations is poorly understood.Whole exome sequencing was performed in 252 probands with ILD and we included all patients with ILD and RTEL1 mutation. RTEL1 expression was evaluated by immunochemistry in the lungs of controls, as well as in RTEL1 and telomerase reverse transcriptase (TERT) mutation carriers.We identified 35 subjects from 17 families. Median age at diagnosis of ILD was 53.1 years (range 28.0-80.6). The most frequent pulmonary diagnoses were idiopathic pulmonary fibrosis (n=20, 57%), secondary ILD (n=7, 20%) and unclassifiable fibrosis or interstitial pneumonia with autoimmune features (n=7, 20%). The median transplant-free and overall survival periods were 39.2 months and 45.3 months, respectively. Forced vital capacity at diagnosis was the only factor associated with decreased transplant-free survival. Extra-pulmonary manifestations were less frequent as compared to other telomere-related gene mutation carriers. A systematic analysis of the literature identified 110 patients with ILD and RTEL1 mutations (including this series) and confirmed the heterogeneity of the pulmonary phenotype, the prevalence of non-idiopathic diseases and the low prevalence of extra-pulmonary manifestations.Immunohistochemistry showed that RTEL1 was expressed by bronchial and alveolar epithelial cells, as well as by alveolar macrophages and lymphocytes, but not by fibroblasts.
Collapse
Affiliation(s)
- Raphael Borie
- Service de Pneumologie A, Hôpital Bichat, AP-HP, DHU FIRE, Paris, France.,Unité 1152, INSERM, Université Paris Diderot, Paris, France
| | - Diane Bouvry
- Service de Pneumologie, Hôpital Avicenne, AP-HP, Bobigny, France
| | - Vincent Cottin
- Service de Pneumologie, Hôpital Louis Pradel, Université Claude Bernard Lyon 1, Lyon, France
| | | | - Aurélie Cazes
- Unité 1152, INSERM, Université Paris Diderot, Paris, France.,Service d'Anatomopathologie, Hôpital Bichat, AP-HP, Paris, France
| | - Marie-Pierre Debray
- Unité 1152, INSERM, Université Paris Diderot, Paris, France.,Service de Radiologie, Hôpital Bichat, AP-HP, Paris, France
| | | | - Philippe Dieude
- Unité 1152, INSERM, Université Paris Diderot, Paris, France.,Service de Rhumatologie, Hôpital Bichat, AP-HP, Paris, France.,Université Paris Diderot, Paris, France
| | - Tristan Degot
- Service de Pneumologie, CHU Strasbourg, Strasbourg, France
| | | | | | | | | | - Arturo Londono-Vallejo
- UMR 3244 (Telomere and Cancer Lab), CNRS, Institut Curie, PSL Research University, Sorbonne Universités, Paris, France
| | | | - Hervé Mal
- Unité 1152, INSERM, Université Paris Diderot, Paris, France.,Service de Pneumologie B, Hôpital Bichat, AP-HP, Paris, France
| | - Catherine Boileau
- Université Paris Diderot, Paris, France.,Laboratoire de Génétique, Hôpital Bichat, AP-HP, Paris, France
| | | | - Hilario Nunes
- Service de Pneumologie, Hôpital Avicenne, AP-HP, Bobigny, France
| | | | | | - Patrick Revy
- UMR 1163 (Laboratory of Genome Dynamics in the Immune System), INSERM, Imagine Institute, Paris Descartes-Sorbonne Paris Cité University, Paris, France
| | - Julie Traclet
- Service de Pneumologie, Hôpital Tenon, AP-HP, Paris, France
| | - Lidwine Wemeau-Stervinou
- Service de Pneumologie, Centre de Compétence des Maladies Pulmonaires Rares, CHRU de Lille, Lille, France
| | - Marie Wislez
- Service de Pneumologie, Unité d'Oncologie Thoracique, Hôpital Cochin, Hôpitaux Universitaires Paris Centre, AP-HP, Paris, France
| | - Caroline Kannengiesser
- Unité 1152, INSERM, Université Paris Diderot, Paris, France.,Université Paris Diderot, Paris, France.,Laboratoire de Génétique, Hôpital Bichat, AP-HP, Paris, France
| | - Bruno Crestani
- Service de Pneumologie A, Hôpital Bichat, AP-HP, DHU FIRE, Paris, France.,Unité 1152, INSERM, Université Paris Diderot, Paris, France.,Université Paris Diderot, Paris, France
| |
Collapse
|
38
|
Girard E, Eon-Marchais S, Olaso R, Renault AL, Damiola F, Dondon MG, Barjhoux L, Goidin D, Meyer V, Le Gal D, Beauvallet J, Mebirouk N, Lonjou C, Coignard J, Marcou M, Cavaciuti E, Baulard C, Bihoreau MT, Cohen-Haguenauer O, Leroux D, Penet C, Fert-Ferrer S, Colas C, Frebourg T, Eisinger F, Adenis C, Fajac A, Gladieff L, Tinat J, Floquet A, Chiesa J, Giraud S, Mortemousque I, Soubrier F, Audebert-Bellanger S, Limacher JM, Lasset C, Lejeune-Dumoulin S, Dreyfus H, Bignon YJ, Longy M, Pujol P, Venat-Bouvet L, Bonadona V, Berthet P, Luporsi E, Maugard CM, Noguès C, Delnatte C, Fricker JP, Gesta P, Faivre L, Lortholary A, Buecher B, Caron O, Gauthier-Villars M, Coupier I, Servant N, Boland A, Mazoyer S, Deleuze JF, Stoppa-Lyonnet D, Andrieu N, Lesueur F. Familial breast cancer and DNA repair genes: Insights into known and novel susceptibility genes from the GENESIS study, and implications for multigene panel testing. Int J Cancer 2018; 144:1962-1974. [PMID: 30303537 PMCID: PMC6587727 DOI: 10.1002/ijc.31921] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Revised: 09/11/2018] [Accepted: 09/25/2018] [Indexed: 12/16/2022]
Abstract
Pathogenic variants in BRCA1 and BRCA2 only explain the underlying genetic cause of about 10% of hereditary breast and ovarian cancer families. Because of cost‐effectiveness, multigene panel testing is often performed even if the clinical utility of testing most of the genes remains questionable. The purpose of our study was to assess the contribution of rare, deleterious‐predicted variants in DNA repair genes in familial breast cancer (BC) in a well‐characterized and homogeneous population. We analyzed 113 DNA repair genes selected from either an exome sequencing or a candidate gene approach in the GENESIS study, which includes familial BC cases with no BRCA1 or BRCA2 mutation and having a sister with BC (N = 1,207), and general population controls (N = 1,199). Sequencing data were filtered for rare loss‐of‐function variants (LoF) and likely deleterious missense variants (MV). We confirmed associations between LoF and MV in PALB2, ATM and CHEK2 and BC occurrence. We also identified for the first time associations between FANCI, MAST1, POLH and RTEL1 and BC susceptibility. Unlike other associated genes, carriers of an ATM LoF had a significantly higher risk of developing BC than carriers of an ATM MV (ORLoF = 17.4 vs. ORMV = 1.6; pHet = 0.002). Hence, our approach allowed us to specify BC relative risks associated with deleterious‐predicted variants in PALB2, ATM and CHEK2 and to add MAST1, POLH, RTEL1 and FANCI to the list of DNA repair genes possibly involved in BC susceptibility. We also highlight that different types of variants within the same gene can lead to different risk estimates. What's new? Pathogenic variants in BRCA1 and BRCA2 only explain the genetic cause of about 10% of hereditary breast and ovarian cancer families, and the clinical usefulness of testing other genes following the recent introduction of cost‐effective multigene panel sequencing in diagnostics laboratories remains questionable. This large case‐control study describes genetic variation in 113 DNA repair genes and specifies breast cancer relative risks associated with rare deleterious‐predicted variants in PALB2, ATM, and CHEK2. Importantly, different types of variants within the same gene can lead to different risk estimates. The results may help improve risk prediction models and define gene‐specific consensus management guidelines.
Collapse
Affiliation(s)
- Elodie Girard
- Inserm, Paris, France.,Institut Curie, Paris, France.,Mines ParisTech, Fontainebleau, France.,PSL Research University, Paris, France
| | - Séverine Eon-Marchais
- Inserm, Paris, France.,Institut Curie, Paris, France.,Mines ParisTech, Fontainebleau, France.,PSL Research University, Paris, France
| | - Robert Olaso
- Centre National de Recherche en Génomique Humaine, Institut de Biologie François Jacob, CEA, Evry, France
| | - Anne-Laure Renault
- Inserm, Paris, France.,Institut Curie, Paris, France.,Mines ParisTech, Fontainebleau, France.,PSL Research University, Paris, France
| | | | - Marie-Gabrielle Dondon
- Inserm, Paris, France.,Institut Curie, Paris, France.,Mines ParisTech, Fontainebleau, France.,PSL Research University, Paris, France
| | - Laure Barjhoux
- Département de Biopathologie, Centre Léon Bérard, Lyon, France
| | - Didier Goidin
- Life Sciences and Diagnostics Group, Agilent Technologies France, Les Ulis, France
| | - Vincent Meyer
- Centre National de Recherche en Génomique Humaine, Institut de Biologie François Jacob, CEA, Evry, France
| | - Dorothée Le Gal
- Inserm, Paris, France.,Institut Curie, Paris, France.,Mines ParisTech, Fontainebleau, France.,PSL Research University, Paris, France
| | - Juana Beauvallet
- Inserm, Paris, France.,Institut Curie, Paris, France.,Mines ParisTech, Fontainebleau, France.,PSL Research University, Paris, France
| | - Noura Mebirouk
- Inserm, Paris, France.,Institut Curie, Paris, France.,Mines ParisTech, Fontainebleau, France.,PSL Research University, Paris, France
| | - Christine Lonjou
- Inserm, Paris, France.,Institut Curie, Paris, France.,Mines ParisTech, Fontainebleau, France.,PSL Research University, Paris, France
| | - Juliette Coignard
- Inserm, Paris, France.,Institut Curie, Paris, France.,Mines ParisTech, Fontainebleau, France.,PSL Research University, Paris, France.,Université Paris Sud, Paris, France
| | - Morgane Marcou
- Inserm, Paris, France.,Institut Curie, Paris, France.,Mines ParisTech, Fontainebleau, France.,PSL Research University, Paris, France
| | - Eve Cavaciuti
- Inserm, Paris, France.,Institut Curie, Paris, France.,Mines ParisTech, Fontainebleau, France.,PSL Research University, Paris, France
| | - Céline Baulard
- Centre National de Recherche en Génomique Humaine, Institut de Biologie François Jacob, CEA, Evry, France
| | - Marie-Thérèse Bihoreau
- Centre National de Recherche en Génomique Humaine, Institut de Biologie François Jacob, CEA, Evry, France
| | | | - Dominique Leroux
- Département de Génétique, CHU de Grenoble, Hôpital Couple-Enfant, Grenoble, France
| | - Clotilde Penet
- Consultation d'Oncogénétique, Institut Jean-Godinot & ICC Courlancy, Reims, France
| | | | - Chrystelle Colas
- Département de Génétique Groupe Hospitalier Pitié-Salpêtrière, APHP, Paris, France.,Service de Génétique, Institut Curie, Paris, France
| | - Thierry Frebourg
- Département de Génétique, Hôpital Universitaire de Rouen, Rouen, France
| | - François Eisinger
- Institut Paoli Calmette, Département d'Anticipation et de Suivi des Cancers, Oncogénétique Clinique, Institut Paoli-Calmettes & Aix Marseille Université, Inserm, IRD, SESSTIM, Marseille, France
| | - Claude Adenis
- Service de Génétique, Centre Oscar-Lambret, Lille, France
| | - Anne Fajac
- Service d'Oncogénétique, Hôpital Tenon, Paris, France
| | - Laurence Gladieff
- Service d'Oncologie Médicale, Institut Claudius Regaud - IUCT-Oncopole, Toulouse, France
| | - Julie Tinat
- Département de Génétique, Hôpital Universitaire de Rouen, Rouen, France
| | | | | | - Sophie Giraud
- Service de Génétique, Hospices Civils de Lyon, Groupement Hospitalier EST, Bron, France
| | | | | | | | | | - Christine Lasset
- Université Claude Bernard Lyon 1, Villeurbanne; CNRS UMR 5558, Unité de Prévention et Epidémiologie Génétique, Lyon, Centre, Léon Bérard, France
| | | | - Hélène Dreyfus
- Clinique Sainte Catherine, Avignon & CHU de Grenoble, Département de Génétique, Hôpital Couple-Enfant, Grenoble, France
| | - Yves-Jean Bignon
- Université Clermont Auvergne; Inserm, U1240, Centre Jean Perrin, Clermont-Ferrand, France
| | | | - Pascal Pujol
- Service de Génétique Médicale et Oncogénétique, Hôpital Arnaud de Villeneuve, CHU Montpellier & INSERM 896, CRCM Val d'Aurelle, Montpellier, France
| | | | - Valérie Bonadona
- Université Claude Bernard Lyon 1, Villeurbanne; CNRS UMR 5558, Unité de Prévention et Epidémiologie Génétique, Lyon, Centre, Léon Bérard, France
| | - Pascaline Berthet
- Unité de Pathologie Gynécologique, Centre François Baclesse, Caen, France
| | - Elisabeth Luporsi
- Service de Génétique UF4128 CHR Metz-Thionville, Hôpital de Mercy, Metz, France
| | - Christine M Maugard
- Hôpitaux Universitaires de Strasbourg, UF1422 Oncogénétique moléculaire, Laboratoire d'Oncobiologie & UF6948 Oncogénétique Evaluation familiale et suivi, Strasbourg, France
| | - Catherine Noguès
- Institut Paoli Calmette, Département d'Anticipation et de Suivi des Cancers, Oncogénétique Clinique, Institut Paoli-Calmettes & Aix Marseille Université, Inserm, IRD, SESSTIM, Marseille, France
| | - Capucine Delnatte
- Unité d'Oncogénétique, Centre René Gauducheau, Nantes, Saint Herblain, France
| | | | - Paul Gesta
- Service d'Oncogénétique Régional Poitou-Charentes, Niort, France
| | - Laurence Faivre
- Institut GIMI, CHU de Dijon, Hôpital d'Enfants, Oncogénétique & Centre de Lutte contre le Cancer Georges François Leclerc, Dijon, France
| | - Alain Lortholary
- Service d'Oncologie Médicale, Centre Catherine de Sienne, Nantes, France
| | | | - Olivier Caron
- Gustave Roussy, Université Paris-Saclay, Département de Médecine Oncologique, Villejuif, France
| | | | - Isabelle Coupier
- Service de Génétique Médicale et Oncogénétique, Hôpital Arnaud de Villeneuve, CHU Montpellier & INSERM 896, CRCM Val d'Aurelle, Montpellier, France
| | - Nicolas Servant
- Inserm, Paris, France.,Institut Curie, Paris, France.,Mines ParisTech, Fontainebleau, France.,PSL Research University, Paris, France
| | - Anne Boland
- Centre National de Recherche en Génomique Humaine, Institut de Biologie François Jacob, CEA, Evry, France
| | - Sylvie Mazoyer
- Inserm, U1028, CNRS UMR5292, Centre de Recherche en Neurosciences de Lyon, France
| | - Jean-François Deleuze
- Centre National de Recherche en Génomique Humaine, Institut de Biologie François Jacob, CEA, Evry, France
| | - Dominique Stoppa-Lyonnet
- Service de Génétique, Institut Curie, Paris, France.,Inserm, U830, Institut Curie, Paris, France.,Université Paris Descartes, Paris, France
| | - Nadine Andrieu
- Inserm, Paris, France.,Institut Curie, Paris, France.,Mines ParisTech, Fontainebleau, France.,PSL Research University, Paris, France
| | - Fabienne Lesueur
- Inserm, Paris, France.,Institut Curie, Paris, France.,Mines ParisTech, Fontainebleau, France.,PSL Research University, Paris, France
| |
Collapse
|