دورية أكاديمية

Genetic regulation of OAS1 nonsense-mediated decay underlies association with COVID-19 hospitalization in patients of European and African ancestries.

التفاصيل البيبلوغرافية
العنوان: Genetic regulation of OAS1 nonsense-mediated decay underlies association with COVID-19 hospitalization in patients of European and African ancestries.
المؤلفون: Banday AR; Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA., Stanifer ML; Department of Infectious Diseases, Molecular Virology, University Hospital Heidelberg, Heidelberg, Germany.; Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, FL, USA., Florez-Vargas O; Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA., Onabajo OO; Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA., Papenberg BW; Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA., Zahoor MA; Toronto Centre for Liver Disease, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada., Mirabello L; Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA., Ring TJ; Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA., Lee CH; Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA., Albert PS; Biostatistics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA., Andreakos E; Laboratory of Immunobiology, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece., Arons E; Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA., Barsh G; HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA., Biesecker LG; Center for Precision Health Research, National Human Genome Research Institute, Bethesda, MD, USA., Boyle DL; Altman Clinical & Translational Research Institute, UC San Diego Health Sciences, San Diego, CA, USA., Brahier MS; Georgetown University School of Medicine, Washington, DC, USA., Burnett-Hartman A; Institute for Health Research, Kaiser Permanente Colorado, Aurora, CO, USA., Carrington M; Basic Science Program, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD, USA.; Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.; Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA., Chang E; Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea., Choe PG; Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea., Chisholm RL; Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA., Colli LM; Department of Medical Imaging, Hematology, and Oncology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil., Dalgard CL; Uniformed Services University of the Health Sciences, Bethesda, MD, USA., Dude CM; Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, GA, USA., Edberg J; Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA., Erdmann N; Department of Medicine, Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham, AL, USA., Feigelson HS; Institute for Health Research, Kaiser Permanente Colorado, Aurora, CO, USA., Fonseca BA; Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil., Firestein GS; Altman Clinical & Translational Research Institute, UC San Diego Health Sciences, San Diego, CA, USA., Gehring AJ; Toronto Centre for Liver Disease, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.; Department of Immunology, University of Toronto, Toronto, Ontario, Canada., Guo C; Division of Cellular Polarity and Viral Infection, German Cancer Research Center (DKFZ), Heidelberg, Germany., Ho M; Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA., Holland S; Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA., Hutchinson AA; Cancer Genomics Research Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD, USA., Im H; Genome Opinion, Inc., Seoul, Republic of Korea., Irby L; Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, GA, USA., Ison MG; Divisions of Infectious Diseases and Organ Transplantation, Northwestern University Feinberg School of Medicine, Chicago, IL, USA., Joseph NT; Department of Obstetrics & Gynecology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA., Kim HB; Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea.; Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea., Kreitman RJ; Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA., Korf BR; Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, USA., Lipkin SM; Department of Medicine and Program in Mendelian Genetics, Weill Cornell Medicine, New York, NY, USA., Mahgoub SM; Department of Medicine, Infectious Diseases Division, Howard University Hospital, Howard University College of Medicine, Washington, DC, USA., Mohammed I; Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA., Paschoalini GL; Department of Medical Imaging, Hematology, and Oncology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil., Pacheco JA; Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA., Peluso MJ; Division of HIV, Infectious Diseases and Global Medicine, University of California, San Francisco, CA, USA., Rader DJ; Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA., Redden DT; Department of Biostatistics, University of Alabama at Birmingham, Birmingham, AL, USA., Ritchie MD; Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA., Rosenblum B; Center for Precision Health Research, National Human Genome Research Institute, Bethesda, MD, USA., Ross ME; Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA., Anna HPS; Laboratory of Genetic Susceptibility, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA., Savage SA; Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA., Sharma S; Department of Biochemistry and Molecular Biology, National Human Genome Center, Howard University College of Medicine, Washington, DC, USA., Siouti E; Laboratory of Immunobiology, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece., Smith AK; Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, GA, USA., Triantafyllia V; Laboratory of Immunobiology, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece., Vargas JM; Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA., Vargas JD; Veterans Affairs Medical Center, Washington, DC, USA., Verma A; Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA., Vij V; Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA., Wesemann DR; Department of Medicine, Division of Allergy and Immunology, Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA., Yeager M; Cancer Genomics Research Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD, USA., Yu X; Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA., Zhang Y; Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA., Boulant S; Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, FL, USA.; Division of Cellular Polarity and Viral Infection, German Cancer Research Center (DKFZ), Heidelberg, Germany.; Department of Infectious Diseases, Virology, University Hospital Heidelberg, Heidelberg, Germany., Chanock SJ; Laboratory of Genetic Susceptibility, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA., Feld JJ; Toronto Centre for Liver Disease, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.; Department of Immunology, University of Toronto, Toronto, Ontario, Canada., Prokunina-Olsson L; Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA. prokuninal@mail.nih.gov.
المصدر: Nature genetics [Nat Genet] 2022 Aug; Vol. 54 (8), pp. 1103-1116. Date of Electronic Publication: 2022 Jul 14.
نوع المنشور: Journal Article; Research Support, Non-U.S. Gov't; Research Support, N.I.H., Intramural; Research Support, N.I.H., Extramural; Research Support, U.S. Gov't, Non-P.H.S.
اللغة: English
بيانات الدورية: Publisher: Nature Pub. Co Country of Publication: United States NLM ID: 9216904 Publication Model: Print-Electronic Cited Medium: Internet ISSN: 1546-1718 (Electronic) Linking ISSN: 10614036 NLM ISO Abbreviation: Nat Genet Subsets: MEDLINE
أسماء مطبوعة: Original Publication: New York, NY : Nature Pub. Co., c1992-
مواضيع طبية MeSH: COVID-19*/genetics, 2',5'-Oligoadenylate Synthetase/genetics ; 2',5'-Oligoadenylate Synthetase/metabolism ; Alleles ; Hospitalization ; Humans ; SARS-CoV-2/genetics
مستخلص: The chr12q24.13 locus encoding OAS1-OAS3 antiviral proteins has been associated with coronavirus disease 2019 (COVID-19) susceptibility. Here, we report genetic, functional and clinical insights into this locus in relation to COVID-19 severity. In our analysis of patients of European (n = 2,249) and African (n = 835) ancestries with hospitalized versus nonhospitalized COVID-19, the risk of hospitalized disease was associated with a common OAS1 haplotype, which was also associated with reduced severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) clearance in a clinical trial with pegIFN-λ1. Bioinformatic analyses and in vitro studies reveal the functional contribution of two associated OAS1 exonic variants comprising the risk haplotype. Derived human-specific alleles rs10774671-A and rs1131454 -A decrease OAS1 protein abundance through allele-specific regulation of splicing and nonsense-mediated decay (NMD). We conclude that decreased OAS1 expression due to a common haplotype contributes to COVID-19 severity. Our results provide insight into molecular mechanisms through which early treatment with interferons could accelerate SARS-CoV-2 clearance and mitigate against severe COVID-19.
(© 2022. This is a U.S. Government work and not under copyright protection in the US; foreign copyright protection may apply.)
التعليقات: Update of: medRxiv. 2021 Jul 13:2021.07.09.21260221. doi: 10.1101/2021.07.09.21260221. (PMID: 34282422)
References: Pairo-Castineira, E. et al. Genetic mechanisms of critical illness in Covid-19. Nature 591, 92–98 (2020). (PMID: 3330754610.1038/s41586-020-03065-y)
COVID-19 Host Genetics Initiative. Mapping the human genetic architecture of COVID-19. Nature 600, 472–477 (2021). (PMID: 10.1038/s41586-021-03767-x)
Sadler, A. J. & Williams, B. R. Interferon-inducible antiviral effectors. Nat. Rev. Immunol. 8, 559–568 (2008). (PMID: 18575461252226810.1038/nri2314)
Kerr, I. M., Brown, R. E. & Hovanessian, A. G. Nature of inhibitor of cell-free protein synthesis formed in response to interferon and double-stranded RNA. Nature 268, 540–542 (1977). (PMID: 19621710.1038/268540a0)
Duggal, P., Gillanders, E. M., Holmes, T. N. & Bailey-Wilson, J. E. Establishing an adjusted p-value threshold to control the family-wide type 1 error in genome wide association studies. BMC Genomics 9, 516 (2008). (PMID: 18976480262121210.1186/1471-2164-9-516)
Huffman, J. E. et al. Multi-ancestry fine mapping implicates OAS1 splicing in risk of severe COVID-19. Nat. Genet. 54, 125–127 (2022). (PMID: 35027740883753710.1038/s41588-021-00996-8)
Mendez, F. L., Watkins, J. C. & Hammer, M. F. Global genetic variation at OAS1 provides evidence of archaic admixture in Melanesian populations. Mol. Biol. Evol. 29, 1513–1520 (2012). (PMID: 2231915710.1093/molbev/msr301)
Mendez, F. L., Watkins, J. C. & Hammer, M. F. Neandertal origin of genetic variation at the cluster of OAS immunity genes. Mol. Biol. Evol. 30, 798–801 (2013). (PMID: 2331595710.1093/molbev/mst004)
Sams, A. J. et al. Adaptively introgressed Neandertal haplotype at the OAS locus functionally impacts innate immune responses in humans. Genome Biol 17, 246 (2016). (PMID: 27899133512924910.1186/s13059-016-1098-6)
Zeberg, H. & Paabo, S. A genomic region associated with protection against severe COVID-19 is inherited from Neandertals. Proc. Natl Acad. Sci. U S A 118, e2026309118 (2021). (PMID: 33593941793628210.1073/pnas.2026309118)
Bonnevie-Nielsen, V. et al. Variation in antiviral 2’,5’-oligoadenylate synthetase (2'5’AS) enzyme activity is controlled by a single-nucleotide polymorphism at a splice-acceptor site in the OAS1 gene. Am. J. Hum. Genet. 76, 623–633 (2005). (PMID: 10.1086/429391)
Wickenhagen, A. et al. A prenylated dsRNA sensor protects against severe COVID-19. Science 374, eabj3624 (2021). (PMID: 34581622761283410.1126/science.abj3624)
Soveg, F. W. et al. Endomembrane targeting of human OAS1 p46 augments antiviral activity. Elife 10, e71047 (2021). (PMID: 34342578835741610.7554/eLife.71047)
Zhou, S. et al. A Neanderthal OAS1 isoform protects individuals of European ancestry against COVID-19 susceptibility and severity. Nat. Med. 27, 659–667 (2021). (PMID: 3363340810.1038/s41591-021-01281-1)
Sun, B. B. et al. Genomic atlas of the human plasma proteome. Nature 558, 73–79 (2018). (PMID: 29875488669754110.1038/s41586-018-0175-2)
Colombo, M., Karousis, E. D., Bourquin, J., Bruggmann, R. & Muhlemann, O. Transcriptome-wide identification of NMD-targeted human mRNAs reveals extensive redundancy between SMG6- and SMG7-mediated degradation pathways. RNA 23, 189–201 (2017). (PMID: 27864472523879410.1261/rna.059055.116)
Samuel, C. E. Antiviral actions of interferons. Clin. Microbiol. Rev. 14, 778–809 (2001). (PMID: 115857858900310.1128/CMR.14.4.778-809.2001)
Galani, I. E. et al. Untuned antiviral immunity in COVID-19 revealed by temporal type I/III interferon patterns and flu comparison. Nat. Immunol. 22, 32–40 (2021). (PMID: 3327763810.1038/s41590-020-00840-x)
Hadjadj, J. et al. Impaired type I interferon activity and inflammatory responses in severe COVID-19 patients. Science 369, 718–724 (2020). (PMID: 32661059740263210.1126/science.abc6027)
Lei, X. et al. Activation and evasion of type I interferon responses by SARS-CoV-2. Nat. Commun. 11, 3810 (2020). (PMID: 32733001739289810.1038/s41467-020-17665-9)
Blanco-Melo, D. et al. Imbalanced host response to SARS-CoV-2 drives development of COVID-19. Cell 181, 1036–1045.e9 (2020). (PMID: 32416070722758610.1016/j.cell.2020.04.026)
Prokunina-Olsson, L. et al. COVID-19 and emerging viral infections: the case for interferon lambda. J. Exp. Med. 217, e20200653 (2020). (PMID: 32289152715580710.1084/jem.20200653)
O’Brien, T. R. et al. Weak induction of interferon expression by severe acute respiratory syndrome coronavirus 2 supports clinical trials of interferon-lambda to treat early Coronavirus Disease 2019. Clin. Infect. Dis. 71, 1410–1412 (2020). (PMID: 32301957718435710.1093/cid/ciaa453)
Stanifer, M. L. et al. Critical role of type III interferon in controlling SARS-CoV-2 infection in human intestinal epithelial cells. Cell Rep 32, 107863 (2020). (PMID: 32610043730363710.1016/j.celrep.2020.107863)
Onabajo, O. O. et al. Interferons and viruses induce a novel truncated ACE2 isoform and not the full-length SARS-CoV-2 receptor. Nat. Genet. 52, 1283–1293 (2020). (PMID: 3307791610.1038/s41588-020-00731-9)
Feld, J. J. et al. Peginterferon lambda for the treatment of outpatients with COVID-19: a phase 2, placebo-controlled randomised trial. Lancet Respir. Med. 9, 498–510 (2021). (PMID: 33556319790670710.1016/S2213-2600(20)30566-X)
Lim, J. K. et al. Genetic variation in OAS1 is a risk factor for initial infection with West Nile virus in man. PLoS Pathog. 5, e1000321 (2009). (PMID: 19247438264268010.1371/journal.ppat.1000321)
Zhao, Y., Kang, H., Ji, Y. & Chen, X. Evaluate the relationship between polymorphisms of OAS1 gene and susceptibility to chronic hepatitis C with high resolution melting analysis. Clin. Exp. Med. 13, 171–176 (2013). (PMID: 2271094210.1007/s10238-012-0193-6)
He, J. et al. Association of SARS susceptibility with single nucleic acid polymorphisms of OAS1 and MxA genes: a case-control study. BMC Infect. Dis. 6, 106 (2006). (PMID: 16824203155040710.1186/1471-2334-6-106)
Chebath, J., Benech, P., Hovanessian, A., Galabru, J. & Revel, M. Four different forms of interferon-induced 2′,5′-oligo(A) synthetase identified by immunoblotting in human cells. J. Biol. Chem. 262, 3852–3857 (1987). (PMID: 243450510.1016/S0021-9258(18)61434-6)
O’Brien, M. et al. OAS1: a multiple sclerosis susceptibility gene that influences disease severity. Neurology 75, 411–418 (2010). (PMID: 2067963410.1212/WNL.0b013e3181ebdd2b)
Li, H. et al. Identification of a Sjogren’s syndrome susceptibility locus at OAS1 that influences isoform switching, protein expression, and responsiveness to type I interferons. PLoS Genet. 13, e1006820 (2017). (PMID: 28640813550166010.1371/journal.pgen.1006820)
Wen, L. et al. Exome-wide association study identifies four novel loci for systemic lupus erythematosus in Han Chinese population. Ann. Rheum. Dis. 77, 417 (2018). (PMID: 2923383210.1136/annrheumdis-2017-211823)
Yin, X. et al. Meta-analysis of 208370 East Asians identifies 113 susceptibility loci for systemic lupus erythematosus. Ann. Rheum. Dis. 80, 632–640 (2021). (PMID: 3327296210.1136/annrheumdis-2020-219209)
Lin, R. J. et al. Distinct antiviral roles for human 2’,5’-oligoadenylate synthetase family members against dengue virus infection. J. Immunol. 183, 8035–8043 (2009). (PMID: 1992345010.4049/jimmunol.0902728)
Carey, C. M. et al. Recurrent loss-of-function mutations reveal costs to OAS1 antiviral activity in primates. Cell Host Microbe 25, 336–343.e4 (2019). (PMID: 30713099660916110.1016/j.chom.2019.01.001)
Romero-Brey, I. & Bartenschlager, R. Membranous replication factories induced by plus-strand RNA viruses. Viruses 6, 2826–2857 (2014). (PMID: 25054883411379510.3390/v6072826)
Kjaer, K. H. et al. Mitochondrial localization of the OAS1 p46 isoform associated with a common single nucleotide polymorphism. BMC Cell Biol 15, 33 (2014). (PMID: 25205466416562110.1186/1471-2121-15-33)
Muir, A. J. et al. Phase 1b study of pegylated interferon lambda 1 with or without ribavirin in patients with chronic genotype 1 hepatitis C virus infection. Hepatology 52, 822–832 (2010). (PMID: 2056435210.1002/hep.23743)
Eiger BioPharmaceuticals Inc. Eiger’s single-dose Peginterferon Lambda for COVID-19 reduced risk of hospitalization or ER visits by 50% in a predominantly vaccinated population in Phase 3 TOGETHER study (Eiger BioPharmaceuticals Inc., 2022). https://ir.eigerbio.com/news-releases/news-release-details/eigers-single-dose-peginterferon-lambda-covid-19-reduced-risk.
Monk, P. D. et al. Safety and efficacy of inhaled nebulised interferon beta-1a (SNG001) for treatment of SARS-CoV-2 infection: a randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Respir. Med. 9, 196–206 (2021). (PMID: 3318916110.1016/S2213-2600(20)30511-7)
Zhou, Q. et al. Interferon-alpha2b treatment for COVID-19. Front. Immunol. 11, 1061 (2020). (PMID: 32574262724274610.3389/fimmu.2020.01061)
Zhou, Q. et al. Interferon-alpha2b treatment for COVID-19 is associated with improvements in lung abnormalities. Viruses 13, 44 (2020). (PMID: 782425210.3390/v13010044)
Purcell, S. et al. PLINK: a tool set for whole-genome association and population-based linkage analyses. Am. J. Hum. Genet. 81, 559–575 (2007). (PMID: 17701901195083810.1086/519795)
Laird, N. M. & Ware, J. H. Random-effects models for longitudinal data. Biometrics 38, 963–974 (1982). (PMID: 716879810.2307/2529876)
Ferguson, W., Dvora, S., Fikes, R. W., Stone, A. C. & Boissinot, S. Long-term balancing selection at the antiviral gene OAS1 in Central African chimpanzees. Mol. Biol. Evol. 29, 1093–1103 (2012). (PMID: 2210421210.1093/molbev/msr247)
Dunn, K. W., Kamocka, M. M. & McDonald, J. H. A practical guide to evaluating colocalization in biological microscopy. Am. J. Physiol. Cell Physiol. 300, C723–C742 (2011). (PMID: 21209361307462410.1152/ajpcell.00462.2010)
معلومات مُعتمدة: UL1 TR003096 United States TR NCATS NIH HHS; RC2 AG036607 United States AG NIA NIH HHS; R01 AI139538 United States AI NIAID NIH HHS; K23 AI157875 United States AI NIAID NIH HHS; R01 NS105477 United States NS NINDS NIH HHS; HHSN261200800001C United States CA NCI NIH HHS; P01 AI165072 United States AI NIAID NIH HHS; SC1 GM093999 United States GM NIGMS NIH HHS; L30 AI147159 United States AI NIAID NIH HHS; HHSN261200800001E United States CA NCI NIH HHS
المشرفين على المادة: EC 2.7.7.- (OAS1 protein, human)
EC 2.7.7.84 (2',5'-Oligoadenylate Synthetase)
تواريخ الأحداث: Date Created: 20220714 Date Completed: 20220809 Latest Revision: 20240615
رمز التحديث: 20240615
مُعرف محوري في PubMed: PMC9355882
DOI: 10.1038/s41588-022-01113-z
PMID: 35835913
قاعدة البيانات: MEDLINE
الوصف
تدمد:1546-1718
DOI:10.1038/s41588-022-01113-z