1
|
Gasmi A, Kassym L, Menzel A, Anzar W, Dadar M, Semenova Y, Arshad M, Bihunyak T, Meguid NA, Peana M, Bekbergenova Z, Bjørklund G. Genetic and Epigenetic Determinants of COVID-19 Susceptibility: A Systematic Review. Curr Med Chem 2025; 32:753-770. [PMID: 38251695 DOI: 10.2174/0109298673267890231221100659] [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: 06/18/2023] [Revised: 08/04/2023] [Accepted: 11/14/2023] [Indexed: 01/23/2024]
Abstract
BACKGROUND The molecular mechanisms regulating coronavirus pathogenesis are complex, including virus-host interactions associated with replication and innate immune control. However, some genetic and epigenetic conditions associated with comorbidities increase the risk of hospitalization and can prove fatal in infected patients. This systematic review will provide insight into host genetic and epigenetic factors that interfere with COVID-19 expression in light of available evidence. METHODS This study conducted a systematic review to examine the genetic and epigenetic susceptibility to COVID-19 using a comprehensive approach. Through systematic searches and applying relevant keywords across prominent online databases, including Scopus, PubMed, Web of Science, and Science Direct, we compiled all pertinent papers and reports published in English between December 2019 and June 2023. RESULTS The findings reveal that the host's HLA genotype plays a substantial role in determining how viral protein antigens are showcased and the subsequent immune system reaction to these antigens. Within females, genes responsible for immune system regulation are found on the X chromosome, resulting in reduced viral load and inflammation levels when contrasted with males. Possessing blood group A may contribute to an increased susceptibility to contracting COVID-19 as well as a heightened risk of mortality associated with the disease. The capacity of SARS-CoV-2 involves inhibiting the antiviral interferon (IFN) reactions, resulting in uncontrolled viral multiplication. CONCLUSION There is a notable absence of research into the gender-related predisposition to infection, necessitating a thorough examination. According to the available literature, a significant portion of individuals affected by the ailment or displaying severe ramifications already had suppressed immune systems, categorizing them as a group with elevated risk.
Collapse
Affiliation(s)
- Amin Gasmi
- Department of Research, Société Francophone de Nutrithérapie et de Nutrigénétique Appliquée, Villeurbanne, France
| | - Laura Kassym
- Department of Research, Astana Medical University, Astana, Kazakhstan
| | - Alain Menzel
- Department of Research, Laboratoires Réunis, Junglinster, Luxembourg
| | - Wajiha Anzar
- Department of Research, Dow University of Health Sciences, Karachi, Pakistan
| | - Maryam Dadar
- Department of Research, CONEM Iran Microbiology Research Group, Tehran, Iran
| | - Yuliya Semenova
- Department of Research, Nazarbayev University School of Medicine, Astana, Kazakhstan
| | - Mehreen Arshad
- Department of Research, National University of Sciences and Technology, Islamabad, Pakistan
| | - Tetyana Bihunyak
- Department of Research, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Nagwa Abdel Meguid
- Research on Children with Special Needs Department, National Research Centre, Giza, Egypt
- CONEM Egypt Child Brain Research Group, National Research Center, Giza, Egypt
| | - Massimiliano Peana
- Department of Chemical, Physical, Mathematical and Natural Sciences, University of Sassari, Sassari, Italy
| | | | - Geir Bjørklund
- Department of Research, Council for Nutritional and Environmental Medicine (CONEM), Mo i Rana, Norway
| |
Collapse
|
2
|
DE Oliveira Sales L, DA Silva JBS, DE Pinho Pessoa FMC, Dias Nogueira BM, DE Oliveira LLB, Khayat AS, DE Moraes Filho MO, DE Moraes MEA, Montenegro RC, Moreira-Nunes CA. Hyperexpression of PTAFR and PF4 as Possible Platelet Risk Biomarkers in Patients With COVID-19. In Vivo 2024; 38:2853-2863. [PMID: 39477442 PMCID: PMC11535951 DOI: 10.21873/invivo.13766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 07/18/2024] [Accepted: 07/19/2024] [Indexed: 11/07/2024]
Abstract
BACKGROUND/AIM SARS-CoV-2 infection presents different severity levels that suggest the influence of genetic factors on the clinical outcome of the disease. In cases of severe COVID-19, the presence of elevated coagulation markers, increased platelet activation and aggregation and the risk of thrombotic complications are described. Given the participation of these cells in several serious viral infections and their negative role when associated with a prothrombotic response, it is important to understand the mechanistic role of SARS-CoV-2 in platelet physiology. This study evaluated the hyperexpression of platelet-activating factor receptor (PTAFR) and platelet factor 4 (PF4) in unvaccinated and hospitalized patients with COVID-19. PATIENTS AND METHODS The study included 43 COVID-19 patients stratified according to WHO guidelines. Subsequently, the expression of the PTAFR and PF4 genes were evaluated using the real-time quantitative PCR and their possible correlation with the severity of the disease and clinical variables including hospitalization, outcome, sex, age and laboratory parameters (platelet count, INR and D-dimer). RESULTS The analysis demonstrated a significant (p<0.05) hyperexpression of these genes COVID-19 patients (n=43) compared to healthy controls. Expression of these genes in patients was not statistically significant (p>0.05) different between patients stratified according to clinical variables. CONCLUSION The expression of PTAFR and PF4 suggests an important molecular pathway in the pathophysiology of the disease and may be valuable platelet biomarkers to indicate increased risk in patients with COVID-19 who require hospital care, contributing to personalized intervention strategies and improving their clinical management.
Collapse
Affiliation(s)
- Lívia DE Oliveira Sales
- Department of Medicine, Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE, Brazil
| | | | - Flávia Melo Cunha DE Pinho Pessoa
- Department of Medicine, Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE, Brazil
| | - Beatriz Maria Dias Nogueira
- Department of Medicine, Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE, Brazil
| | - Lais Lacerda Brasil DE Oliveira
- Department of Medicine, Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE, Brazil
| | - André Salim Khayat
- Department of Biological Sciences, Oncology Research Center, Federal University of Pará, Belém, PA, Brazil
| | - Manoel Odorico DE Moraes Filho
- Department of Medicine, Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE, Brazil
| | - Maria Elisabete Amaral DE Moraes
- Department of Medicine, Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE, Brazil
| | - Raquel Carvalho Montenegro
- Department of Medicine, Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE, Brazil
| | - Caroline Aquino Moreira-Nunes
- Department of Medicine, Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE, Brazil;
- Clementino Fraga Group, Central Unity, Molecular Biology Laboratory, Fortaleza, CE, Brazil
- Department of Biological Sciences, Oncology Research Center, Federal University of Pará, Belém, PA, Brazil
| |
Collapse
|
3
|
Zheng K, Chong AY, Mentzer AJ. How could our genetics impact COVID-19 vaccine response? Expert Rev Clin Immunol 2024; 20:1027-1039. [PMID: 38676712 DOI: 10.1080/1744666x.2024.2346584] [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: 12/22/2023] [Accepted: 04/19/2024] [Indexed: 04/29/2024]
Abstract
INTRODUCTION The COVID-19 pandemic, caused by the SARS-CoV-2 virus, has posed unprecedented global health challenges since its emergence in December 2019. The rapid availability of vaccines has been estimated to save millions of lives, but there is variation in how individuals respond to vaccines, influencing their effectiveness at an individual, and population level. AREAS COVERED This review focuses on human genetic factors influencing the immune response and effectiveness of vaccines, highlighting the importance of associations across the HLA locus. Genome-Wide Association Studies (GWAS) and other genetic association analyses have identified statistically significant associations between specific HLA alleles including HLA-DRB1*13, DBQ1*06, and A*03 impacting antibody responses and the risk of breakthrough infections post-vaccination. Relationships between these associations and potential mechanisms and links with risks of natural infection or disease are explored, and this review concludes by emphasizing how understanding the mechanisms of these genetic determinants may inform the development of tailored vaccination strategies. EXPERT OPINION Although complex, we believe these findings from the SARS-CoV2 pandemic offer a unique opportunity to understand the relationships between HLA and infection and vaccine response, with a goal of optimizing individual protection against COVID-19 in the ongoing pandemic, and possibly influencing wider vaccine development in the future.
Collapse
Affiliation(s)
- Keyi Zheng
- Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Amanda Y Chong
- Centre for Human Genetics, University of Oxford, Oxford, UK
| | | |
Collapse
|
4
|
Wang L, Jackson VE, Fearnley LG, Bahlo M. UKB.COVID19: an R package for UK Biobank COVID-19 data processing and analysis. F1000Res 2024; 10:830. [PMID: 39193262 PMCID: PMC11347911 DOI: 10.12688/f1000research.55370.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/23/2024] [Indexed: 08/29/2024] Open
Abstract
COVID-19 caused by SARS-CoV-2 has resulted in a global pandemic with a rapidly developing global health and economic crisis. Variations in the disease have been observed and have been associated with the genomic sequence of either the human host or the pathogen. Worldwide scientists scrambled initially to recruit patient cohorts to try and identify risk factors. A resource that presented itself early on was the UK Biobank (UKBB), which is investigating the respective contributions of genetic predisposition and environmental exposure to the development of disease. To enable COVID-19 studies, UKBB is now receiving COVID-19 test data for their participants every two weeks. In addition, UKBB is delivering more frequent updates of death and hospital inpatient data (including critical care admissions) on the UKBB Data Portal. This frequently changing dataset requires a tool that can rapidly process and analyse up-to-date data. We developed an R package specifically for the UKBB COVID-19 data, which summarises COVID-19 test results, performs association tests between COVID-19 susceptibility/severity and potential risk factors such as age, sex, blood type, comorbidities and generates input files for genome-wide association studies (GWAS). By applying the R package to data released in April 2021, we found that age, body mass index, socioeconomic status and smoking are positively associated with COVID-19 susceptibility, severity, and mortality. Males are at a higher risk of COVID-19 infection than females. People staying in aged care homes have a higher chance of being exposed to SARS-CoV-2. By performing GWAS, we replicated the 3p21.31 genetic finding for COVID-19 susceptibility and severity. The ability to iteratively perform such analyses is highly relevant since the UKBB data is updated frequently. As a caveat, users must arrange their own access to the UKBB data to use the R package.
Collapse
Affiliation(s)
- Longfei Wang
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
| | - Victoria E Jackson
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
| | - Liam G Fearnley
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
| | - Melanie Bahlo
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
| |
Collapse
|
5
|
Kovanda A, Lukežič T, Maver A, Vokač Križaj H, Čižek Sajko M, Šelb J, Rijavec M, Bidovec-Stojković U, Bitežnik B, Rituper B, Korošec P, Peterlin B. Genomic Landscape of Susceptibility to Severe COVID-19 in the Slovenian Population. Int J Mol Sci 2024; 25:7674. [PMID: 39062917 PMCID: PMC11277002 DOI: 10.3390/ijms25147674] [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: 06/03/2024] [Revised: 07/05/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Determining the genetic contribution of susceptibility to severe SARS-CoV-2 infection outcomes is important for public health measures and individualized treatment. Through intense research on this topic, several hundred genes have been implicated as possibly contributing to the severe infection phenotype(s); however, the findings are complex and appear to be population-dependent. We aimed to determine the contribution of human rare genetic variants associated with a severe outcome of SARS-CoV-2 infections and their burden in the Slovenian population. A panel of 517 genes associated with severe SARS-CoV-2 infection were obtained by combining an extensive review of the literature, target genes identified by the COVID-19 Host Genetic Initiative, and the curated Research COVID-19 associated genes from PanelApp, England Genomics. Whole genome sequencing was performed using PCR-free WGS on DNA from 60 patients hospitalized due to severe COVID-19 disease, and the identified rare genomic variants were analyzed and classified according to the ACMG criteria. Background prevalence in the general Slovenian population was determined by comparison with sequencing data from 8025 individuals included in the Slovenian genomic database (SGDB). Results show that several rare pathogenic/likely pathogenic genomic variants in genes CFTR, MASP2, MEFV, TNFRSF13B, and RNASEL likely contribute to the severe infection outcomes in our patient cohort. These results represent an insight into the Slovenian genomic diversity associated with a severe COVID-19 outcome.
Collapse
Affiliation(s)
- Anja Kovanda
- Clinical Institute of Genomic Medicine, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Tadeja Lukežič
- Clinical Institute of Genomic Medicine, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
| | - Aleš Maver
- Clinical Institute of Genomic Medicine, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Hana Vokač Križaj
- Clinical Institute of Genomic Medicine, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
| | - Mojca Čižek Sajko
- Clinical Institute of Genomic Medicine, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
| | - Julij Šelb
- University Clinic of Respiratory and Allergic Diseases Golnik, 4204 Golnik, Slovenia
| | - Matija Rijavec
- University Clinic of Respiratory and Allergic Diseases Golnik, 4204 Golnik, Slovenia
| | | | - Barbara Bitežnik
- University Clinic of Respiratory and Allergic Diseases Golnik, 4204 Golnik, Slovenia
| | - Boštjan Rituper
- University Clinic of Respiratory and Allergic Diseases Golnik, 4204 Golnik, Slovenia
| | - Peter Korošec
- University Clinic of Respiratory and Allergic Diseases Golnik, 4204 Golnik, Slovenia
| | - Borut Peterlin
- Clinical Institute of Genomic Medicine, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| |
Collapse
|
6
|
SINGH MAIREMBAMSTELIN, YELLABOINA SAILU, ANSARI MAIRAJAHMED. A COMPREHENSIVE REVIEW ON THE MULTIFACETED INTERACTIONS BETWEEN HOST IMMUNITY AND VIRAL PATHOGENESIS IN COVID-19. INTERNATIONAL JOURNAL OF APPLIED PHARMACEUTICS 2024:37-45. [DOI: 10.22159/ijap.2024v16i4.50576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
The Corona Virus Disease (COVID-19) pandemic has presented unparalleled challenges, marked by a wide array of clinical presentations spanning from asymptomatic carriage to severe respiratory compromise and multi-organ dysfunction. It is crucial to comprehend the intricate interplay between host immunity and viral pathogenesis to elucidate disease mechanisms and guide therapeutic strategies. This review delves into the multifaceted interactions between host immunity and viral pathogenesis in COVID-19, with a particular focus on the impact of host factors such as age, sex, comorbidities, and genetic predisposition on disease severity. Utilizing state-of-the-art methodologies, including multiomics approaches, has yielded an expansive molecular portrayal of COVID-19, furnishing innovative perspectives on host immune reactions, viral pathogenicity, and disease advancement. Establishing standardized methodologies for data analysis and interpretation while concurrently addressing ethical considerations and promoting interdisciplinary collaboration are crucial steps in advancing our comprehension of COVID-19 pathogenesis. Despite obstacles like complexities in data integration, this review highlights the imperative of persistent endeavors in deciphering the complex interactions between hosts and pathogens to alleviate the global health ramifications of COVID-19.
Collapse
|
7
|
Saadat A, Gouttenoire J, Ripellino P, Semela D, Amar S, Frey BM, Fontana S, Mdawar-Bailly E, Moradpour D, Fellay J, Fraga M. Inborn errors of type I interferon immunity in patients with symptomatic acute hepatitis E. Hepatology 2024; 79:1421-1431. [PMID: 38079352 PMCID: PMC11095861 DOI: 10.1097/hep.0000000000000701] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 11/06/2023] [Indexed: 03/13/2024]
Abstract
BACKGROUND AND AIMS The clinical spectrum of human infection by HEV ranges from asymptomatic to severe acute hepatitis. Furthermore, HEV can cause diverse neurological manifestations, especially Parsonage-Turner syndrome. Here, we used a large-scale human genomic approach to search for genetic determinants of severe clinical presentations of HEV infection. APPROACH AND RESULTS We performed whole genome sequencing in 3 groups of study participants with PCR-proven acute HEV infection: (1) 24 patients with symptomatic acute hepatitis E; (2) 12 patients with HEV-associated Parsonage-Turner syndrome; and (3) 16 asymptomatic blood donors (controls). For variant calling and annotation, we used GATK4 best practices followed by Variant Effect Predictor (VEP) and Annovar. For variant classification, we implemented the American College of Medical Genetics and Genomics/Association for Molecular Pathology Bayesian classification framework in R. Variants with a probability of pathogenicity >0.9 were considered damaging. We used all genes with at least 1 damaging variant as input for pathway enrichment analyses.We observed a significant enrichment of type I interferon response pathways in the symptomatic hepatitis group: 10 out of 24 patients carried a damaging variant in one of 9 genes encoding either intracellular viral sensors ( IFIH1 , DDX58 , TLR3 , POLR3B , POLR3C ) or other molecules involved in type I interferon response [interferon regulatory factor 7 ( IRF7 ), MYD88 , OAS3 , GAPDH ]. We did not find any enriched pathway in the Parsonage-Turner syndrome group or in the controls. CONCLUSIONS Our results highlight the essential role of type I interferon in preventing symptomatic acute hepatitis E.
Collapse
Affiliation(s)
- Ali Saadat
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Jérôme Gouttenoire
- Division of Gastroenterology and Hepatology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Paolo Ripellino
- Department of Neurology, Neurocenter of Southern Switzerland, EOC, Lugano, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - David Semela
- Division of Gastroenterology and Hepatology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Soraya Amar
- Swiss Transfusion, Swiss Red Cross, Bern, Switzerland
| | - Beat M. Frey
- Blood Transfusion Service SRC, Schlieren/Zurich, Switzerland
| | | | | | - Elise Mdawar-Bailly
- Division of Gastroenterology and Hepatology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Darius Moradpour
- Division of Gastroenterology and Hepatology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Jacques Fellay
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Precision Medicine Unit, Biomedical Data Science Center, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Montserrat Fraga
- Division of Gastroenterology and Hepatology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
8
|
Deng X, Tang K, Wang Z, He S, Luo Z. Impacts of Inflammatory Cytokines Variants on Systemic Inflammatory Profile and COVID-19 Severity. J Epidemiol Glob Health 2024; 14:363-378. [PMID: 38376765 PMCID: PMC11176143 DOI: 10.1007/s44197-024-00204-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 01/30/2024] [Indexed: 02/21/2024] Open
Abstract
BACKGROUND Cytokine storm is known to impact the prognosis of coronavirus disease 2019 (COVID-19), since pro-inflammatory cytokine variants are associated with cytokine storm. It is tempting to speculate that pro-inflammatory cytokines variants may impact COVID-19 outcomes by modulating cytokine storm. Here, we verified this hypothesis via a comprehensive analysis. METHODS PubMed, Cochrane Library, Central, CINAHL, and ClinicalTrials.gov were searched until December 15, 2023. Case-control or cohort studies that investigated the impacts of rs1800795 or rs1800629 on COVID-19 susceptibility, severity, mortality, IL-6, TNF-α, or CRP levels were included after an anonymous review by two independent reviewers and consultations of disagreement by a third independent reviewer. RESULTS 47 studies (8305 COVID-19 individuals and 17,846 non-COVID-19 individuals) were analyzed. The rs1800629 A allele (adenine at the -308 position of the promoter was encoded by the A allele) was associated with higher levels of tumor necrosis factor-α (TNF-α) and C-reactive protein (CRP). In contrast, the rs1800795 C allele (cytosine at the -174 position of the promoter was encoded by the C allele) was linked to higher levels of interleukin-6 (IL-6) and CRP. In addition, the A allele of rs1800629 increased the severity and mortality of COVID-19. However, the C allele of rs1800795 only increased COVID-19 susceptibility. CONCLUSIONS rs1800629 and rs1800795 variants of pro-inflammatory cytokines have significant impacts on systemic inflammatory profile and COVID-19 clinical outcomes. rs1800629 may serve as a genetic marker for severe COVID-19.
Collapse
Affiliation(s)
- XueJun Deng
- Department of Cardiology, Suining Central Hospital, Suining, 629000, Sichuan, China
| | - Kai Tang
- Department of Cardiology, Suining Central Hospital, Suining, 629000, Sichuan, China
| | - Zhiqiang Wang
- Orthopedic Center 1 Department of Orthopedic Trauma, Suining Central Hospital, Suining, Sichuan, China.
| | - Suyu He
- The Fourth Department of Digestive Disease Center, Suining Central Hospital, Suining, 629000, Sichuan, China.
| | - Zhi Luo
- Department of Cardiology, Suining Central Hospital, Suining, 629000, Sichuan, China.
| |
Collapse
|
9
|
Montenegro AFL, Clementino MAF, Yaochite JNU. Type I interferon pathway genetic variants in severe COVID-19. Virus Res 2024; 342:199339. [PMID: 38354910 PMCID: PMC10901847 DOI: 10.1016/j.virusres.2024.199339] [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: 11/21/2023] [Revised: 02/09/2024] [Accepted: 02/11/2024] [Indexed: 02/16/2024]
Abstract
Coronavirus Disease 2019 (COVID-19) is an infectious disease caused by SARS-CoV-2. According to the World Health Organization (WHO), there have been over 760 million reported cases and over 6 million deaths caused by this disease worldwide. The severity of COVID-19 is based on symptoms presented by the patient and is divided as asymptomatic, mild, moderate, severe, and critical. The manifestations are interconnected with genetic variations. The innate immunity is the quickest response mechanism of an organism against viruses. Type I interferon pathway plays a key role in antiviral responses due to viral replication inhibition in infected cells and adaptive immunity stimulation induced by interferon molecules. Thus, variants in type I interferon pathway's genes are being studied in different COVID-19 manifestations. This review summarizes the role of variants in type I interferon pathway's genes on prognosis and severity progression of COVID-19.
Collapse
Affiliation(s)
- A F L Montenegro
- Laboratório de Imunologia Celular e Molecular, Departamento de Análises Clínicas e Toxicológicas da Faculdade de Farmácia, Odontologia e Enfermagem, Universidade Federal do Ceará - UFC, Rua Pastor Samuel Munguba, 1210 - Rodolfo Teófilo, Fortaleza, Ceará, Brasil
| | - M A F Clementino
- Laboratório de Toxinologia Molecular, NUBIMED - Núcleo de Biomedicina, Universidade Federal do Ceará - UFC. Fortaleza, Ceará, Brasil
| | - J N U Yaochite
- Laboratório de Imunologia Celular e Molecular, Departamento de Análises Clínicas e Toxicológicas da Faculdade de Farmácia, Odontologia e Enfermagem, Universidade Federal do Ceará - UFC, Rua Pastor Samuel Munguba, 1210 - Rodolfo Teófilo, Fortaleza, Ceará, Brasil.
| |
Collapse
|
10
|
Gutiérrez-Pérez IA, Buendía-Roldán I, Zaragoza-García O, Pérez-Rubio G, Villafan-Bernal JR, Chávez-Galán L, Parra-Rojas I, Hernández-Zenteno RDJ, Fricke-Galindo I, Castro-Alarcón N, Bautista-Becerril B, Falfán-Valencia R, Guzmán-Guzmán IP. Association of PADI2 and PADI4 polymorphisms in COVID-19 host severity and non-survival. Heliyon 2024; 10:e27997. [PMID: 38524554 PMCID: PMC10958703 DOI: 10.1016/j.heliyon.2024.e27997] [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: 11/15/2023] [Revised: 02/27/2024] [Accepted: 03/10/2024] [Indexed: 03/26/2024] Open
Abstract
Background Enzymes of the peptidylarginine deiminase family (PADs) play a relevant role in the pathogenesis of COVID-19. However, the association of single nucleotide polymorphisms (SNPs) in their genes with COVID-19 severity and death is unknown. Methodology We included 1045 patients who were diagnosed with COVID-19 between October 2020 and December 2021. All subjects were genotyped for PADI2 (rs1005753 and rs2235926) and PADI4 (rs11203366, rs11203367, and rs874881) SNPs by TaqMan assays and their associations with disease severity, death, and inflammatory biomarkers were evaluated. Results 291 patients presented had severe COVID-19 according to PaO2/FiO2, and 393 had a non-survival outcome. Carriers of the rs1005753 G/G genotype in the PADI2 gene presented susceptibility for severe COVID-19, while the heterozygous carriers in rs11203366, rs11203367, and rs874881 of the PADI4 gene showed risk of death. The GTACC haplotype in PADI2-PADI4 was associated with susceptibility to severe COVID-19, while the GCACC haplotype was a protective factor. The GCGTG haplotype was associated with severe COVID-19 but as a protective haplotype for death. Finally, the GTACC haplotype was associated with platelet-to-lymphocyte ratio (PLR), the GCACC haplotype with neutrophil-to-hemoglobin and lymphocyte and the GCGTG haplotype as a protective factor for the elevation of procalcitonin, D-dimer, CRP, LCRP, NHL, SII, NLR, and PLR. Conclusions Our results suggest that the haplotypic combination of GTACC and some individual genotypes of PADI2 and PADI4 contribute to the subjects' susceptibility for severity and death by COVID-19.
Collapse
Affiliation(s)
- Ilse Adriana Gutiérrez-Pérez
- Faculty of Chemical-Biological Sciences, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, 39000, Mexico
| | - Ivette Buendía-Roldán
- Translational Research Laboratory on Aging and Pulmonary Fibrosis, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, 14080, Mexico
| | - Oscar Zaragoza-García
- Faculty of Chemical-Biological Sciences, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, 39000, Mexico
| | - Gloria Pérez-Rubio
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, 14080, Mexico
| | - José Rafael Villafan-Bernal
- Investigador por Mexico, Laboratory of Immunogenomics and Metabolic Disease, Mexican National Institute of Genomic Medicine (INMEGEN), Mexico City, 14610, Mexico
| | - Leslie Chávez-Galán
- Laboratory of Integrative Immunology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, 14080, Mexico
| | - Isela Parra-Rojas
- Faculty of Chemical-Biological Sciences, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, 39000, Mexico
| | | | - Ingrid Fricke-Galindo
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, 14080, Mexico
| | - Natividad Castro-Alarcón
- Faculty of Chemical-Biological Sciences, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, 39000, Mexico
| | - Brandon Bautista-Becerril
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, 14080, Mexico
| | - Ramcés Falfán-Valencia
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, 14080, Mexico
| | - Iris Paola Guzmán-Guzmán
- Faculty of Chemical-Biological Sciences, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, 39000, Mexico
| |
Collapse
|
11
|
Jabeen M, Shoukat S, Shireen H, Bao Y, Khan A, Abbasi AA. Unraveling the genetic variations underlying virulence disparities among SARS-CoV-2 strains across global regions: insights from Pakistan. Virol J 2024; 21:55. [PMID: 38449001 PMCID: PMC10916261 DOI: 10.1186/s12985-024-02328-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 02/26/2024] [Indexed: 03/08/2024] Open
Abstract
Over the course of the COVID-19 pandemic, several SARS-CoV-2 variants have emerged that may exhibit different etiological effects such as enhanced transmissibility and infectivity. However, genetic variations that reduce virulence and deteriorate viral fitness have not yet been thoroughly investigated. The present study sought to evaluate the effects of viral genetic makeup on COVID-19 epidemiology in Pakistan, where the infectivity and mortality rate was comparatively lower than other countries during the first pandemic wave. For this purpose, we focused on the comparative analyses of 7096 amino-acid long polyprotein pp1ab. Comparative sequence analysis of 203 SARS-CoV-2 genomes, sampled from Pakistan during the first wave of the pandemic revealed 179 amino acid substitutions in pp1ab. Within this set, 38 substitutions were identified within the Nsp3 region of the pp1ab polyprotein. Structural and biophysical analysis of proteins revealed that amino acid variations within Nsp3's macrodomains induced conformational changes and modified protein-ligand interactions, consequently diminishing the virulence and fitness of SARS-CoV-2. Additionally, the epistatic effects resulting from evolutionary substitutions in SARS-CoV-2 proteins may have unnoticed implications for reducing disease burden. In light of these findings, further characterization of such deleterious SARS-CoV-2 mutations will not only aid in identifying potential therapeutic targets but will also provide a roadmap for maintaining vigilance against the genetic variability of diverse SARS-CoV-2 strains circulating globally. Furthermore, these insights empower us to more effectively manage and respond to potential viral-based pandemic outbreaks of a similar nature in the future.
Collapse
Affiliation(s)
- Momina Jabeen
- National Center for Bioinformatics, Program of Comparative and Evolutionary Genomics, Faculty of Biological Sciences, Quaid-i-Azam University, 45320, Islamabad, Pakistan
| | - Shifa Shoukat
- National Center for Bioinformatics, Program of Comparative and Evolutionary Genomics, Faculty of Biological Sciences, Quaid-i-Azam University, 45320, Islamabad, Pakistan
| | - Huma Shireen
- National Center for Bioinformatics, Program of Comparative and Evolutionary Genomics, Faculty of Biological Sciences, Quaid-i-Azam University, 45320, Islamabad, Pakistan
| | - Yiming Bao
- National Genomics Data Center & CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, China National Center for Bioinformation, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100101, Beijing, China
| | - Abbas Khan
- Department of Bioinformatics and Biological Statistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 200240, Shanghai, China
- School of Medical and Life Sciences, Sunway University, Sunway City, Malaysia
| | - Amir Ali Abbasi
- National Center for Bioinformatics, Program of Comparative and Evolutionary Genomics, Faculty of Biological Sciences, Quaid-i-Azam University, 45320, Islamabad, Pakistan.
| |
Collapse
|
12
|
Yadav V, Ravichandran S. Significance of understanding the genomics of host-pathogen interaction in limiting antibiotic resistance development: lessons from COVID-19 pandemic. Brief Funct Genomics 2024; 23:69-74. [PMID: 36722037 DOI: 10.1093/bfgp/elad001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 12/15/2022] [Accepted: 01/09/2023] [Indexed: 02/02/2023] Open
Abstract
The entire world is facing the stiff challenge of COVID-19 pandemic. To overcome the spread of this highly infectious disease, several short-sighted strategies were adopted such as the use of broad-spectrum antibiotics and antifungals. However, the misuse and/or overuse of antibiotics have accentuated the emergence of the next pandemic: antimicrobial resistance (AMR). It is believed that pathogens while transferring between humans and the environment carry virulence and antibiotic-resistant factors from varied species. It is presumed that all such genetic factors are quantifiable and predictable, a better understanding of which could be a limiting step for the progression of AMR. Herein, we have reviewed how genomics-based understanding of host-pathogen interactions during COVID-19 could reduce the non-judicial use of antibiotics and prevent the eruption of an AMR-based pandemic in future.
Collapse
Affiliation(s)
- Vikas Yadav
- Department of Translational Medicine, Clinical Research Centre, Skaone University Hospital, Lund University, Malmo SE-20213, Sweden
| | | |
Collapse
|
13
|
Bastin A, Abbasi F, Roustaei N, Abdesheikhi J, Karami H, Gholamnezhad M, Eftekhari M, Doustimotlagh A. Severity of oxidative stress as a hallmark in COVID-19 patients. Eur J Med Res 2023; 28:558. [PMID: 38049886 PMCID: PMC10696844 DOI: 10.1186/s40001-023-01401-2] [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: 08/07/2022] [Accepted: 09/27/2023] [Indexed: 12/06/2023] Open
Abstract
INTRODUCTION Understanding the mechanisms and identifying effective treatments for the COVID-19 outbreak are imperative. Therefore, this study aimed to assess the antioxidant status and oxidative stress parameters as potential pivotal mechanisms in asymptomatic, non-severe, and severe COVID-19 patients. METHODS This study is a case-control study that was performed on patients referred to the Persian Gulf Martyrs Hospital of Bushehr University of Medical Sciences, Bushehr, Iran, from May 2021 to September 2021. A total of 600 COVID-19 patients (non-severe and severe group) and 150 healthy volunteers of the same age and sex were selected during the same period. On the first day of hospitalization, 10 ml of venous blood was taken from subjects. Then, hematological, biochemical, serological, antioxidant and oxidative stress parameters were determined. RESULTS Our results indicated that ESR, CRP, AST, ALT, and LDH significantly augmented in the severe group as compared to the non-severe and normal groups (P ≤ 0.05). It was observed that the levels of FRAP, G6PD activity, and SOD activity significantly reduced in the non-severe patients in comparison with the severe and normal groups (P ≤ 0.05). We found that MDA content and NO metabolite markedly increased in severe patients as compared to the non-severe group. CONCLUSIONS Taken together, it seems that the balance between antioxidants and oxidants was disturbed in COVID-19 patients in favor of oxidant markers. In addition, this situation caused more aggravation in severe patients as compared to the non-severe group.
Collapse
Affiliation(s)
- Alireza Bastin
- Clinical Research Development Center, "The Persian Gulf Martyrs" Hospital, Bushehr University of Medical Sciences, Bushehr, Iran
- Department of Clinical Biochemistry, Faculty of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Fatemeh Abbasi
- Department of Infectious Disease, Faculty of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Narges Roustaei
- Department of Biostatistics and Epidemiology, School of Health, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Jahangir Abdesheikhi
- Department of Clinical Immunology, School of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Hossein Karami
- Clinical Research Development Center, "The Persian Gulf Martyrs" Hospital, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Mohammad Gholamnezhad
- Department of Infectious Disease, Faculty of Medicine, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Mahdieh Eftekhari
- Pharmaceutical Sciences Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran.
- Department of Pharmacognosy and Pharmaceutical Biotechnology, Faculty of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Amirhossein Doustimotlagh
- Medicinal Plants Research Center, Yasuj University of Medical Sciences, Yasuj, Iran.
- Department of Clinical Biochemistry, Faculty of Medicine, Yasuj University of Medical Sciences, Yasuj, Iran.
| |
Collapse
|
14
|
Nosik M, Berezhnya E, Bystritskaya E, Kiseleva I, Lobach O, Kireev D, Svitich O. Female Sex Hormones Upregulate the Replication Activity of HIV-1 Sub-Subtype A6 and CRF02_AG but Not HIV-1 Subtype B. Pathogens 2023; 12:880. [PMID: 37513727 PMCID: PMC10383583 DOI: 10.3390/pathogens12070880] [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: 04/11/2023] [Revised: 06/18/2023] [Accepted: 06/22/2023] [Indexed: 07/30/2023] Open
Abstract
More than 50% of all people living with HIV worldwide are women. Globally, HIV/AIDS is the leading cause of death among women aged 15 to 44. The safe and effective methods of hormonal contraception are an essential component of preventive medical care in order to reduce maternal and infant mortality. However, there is limited knowledge regarding the effect of hormones on the rate of viral replication in HIV infection, especially non-B subtypes. The goal of the present work was to study in vitro how the female hormones β-estradiol and progesterone affect the replication of the HIV-1 subtypes A6, CRF02_AG, and B. The findings show that high doses of hormones enhanced the replication of HIV-1 sub-subtype A6 by an average of 1.75 times and the recombinant variant CRF02_AG by 1.4 times but did not affect the replication of HIV-1 subtype B. No difference was detected in the expression of CCR5 and CXCR4 co-receptors on the cell surface, either in the presence or absence of hormones. However, one of the reasons for the increased viral replication could be the modulated TLRs secretion, as it was found that high doses of estradiol and progesterone upregulated, to varying degrees, the expression of TLR2 and TLR9 genes in the PBMCs of female donors infected with HIV-1 sub-subtype A6.
Collapse
Affiliation(s)
- Marina Nosik
- I.I. Mechnikov Institute of Vaccines and Sera, 105064 Moscow, Russia
| | - Elena Berezhnya
- I.I. Mechnikov Institute of Vaccines and Sera, 105064 Moscow, Russia
| | | | - Irina Kiseleva
- I.I. Mechnikov Institute of Vaccines and Sera, 105064 Moscow, Russia
| | - Olga Lobach
- I.I. Mechnikov Institute of Vaccines and Sera, 105064 Moscow, Russia
| | - Dmitry Kireev
- Central Research Institute of Epidemiology, 111123 Moscow, Russia
| | - Oxana Svitich
- I.I. Mechnikov Institute of Vaccines and Sera, 105064 Moscow, Russia
| |
Collapse
|
15
|
Shawi HRS, Anvari E, Fateh A. Role of FokI rs2228570 and Tru9I rs757343 Polymorphisms in the Mortality of Patients Infected with Different Variants of SARS-CoV-2. Arch Med Res 2023; 54:310-318. [PMID: 37032232 PMCID: PMC10063569 DOI: 10.1016/j.arcmed.2023.03.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/18/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023]
Abstract
BACKGROUND AND AIM Low vitamin D levels are associated with the severity of coronavirus disease 19 (COVID-19). Vitamin D receptor gene polymorphisms, such as Tru9I rs757343 and FokI rs2228570, have been suggested to be potential risk factors for severe COVID-19 outcomes. This study investigated how Tru9I rs757343 and FokI rs2228570 polymorphisms influenced the mortality rate of COVID-19 in relation to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants. METHODS The polymerase chain reaction-restriction fragment length polymorphism assay was used to genotype Tru9I rs757343 and FokI rs2228570 genotypes in 1,734 recovered and 1,450 deceased patients. RESULTS Our results demonstrated that the high mortality rate was correlated with FokI rs2228570 TT genotype in all three variants but was much higher in the Omicron BA.5 variant than in the Alpha and Delta variants. Furthermore, in patients infected with the Delta variant, FokI rs2228570 CT genotype was more highly correlated with the mortality rate compared to other variants. Thus, a high mortality rate was correlated with the Tru9I rs757343 AA genotype in the Omicron BA.5 variant, whereas this relationship was not observed in the other two variants. The T-A haplotype was related to COVID-19 mortality in all three variants, but its effect was more pronounced in the Alpha variant. Moreover, the T-G haplotype was significantly associated with all three variants. CONCLUSION Our findings showed that the effects of Tru9I rs757343 and FokI rs2228570 polymorphisms were related to SARS-CoV-2 variants. However, further studies are still required to validate our findings.
Collapse
Affiliation(s)
| | - Enayat Anvari
- Clinical Research Development Unit, Shahid Mostafa Khomeini Hospital, Ilam University of Medical Science, Ilam, Iran
| | - Abolfazl Fateh
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran; Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
16
|
Alotaibi BA, Aldali JA, Aldali HJ, Alasiri GA, Elsokkary EM, Al Mugairi A, Almuqrin AM. Risk Factors for Glucose 6-Phosphate Dehydrogenase and COVID-19 Disease-A Retrospective Study at a Major Saudi Tertiary Center. Viruses 2023; 15:1224. [PMID: 37376524 DOI: 10.3390/v15061224] [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: 05/08/2023] [Revised: 05/19/2023] [Accepted: 05/21/2023] [Indexed: 06/29/2023] Open
Abstract
Glucose-6-phosphate dehydrogenase (G6PD) insufficiency is a common enzymatic defect worldwide; it affects over 400 million people and is associated with various disorders. Recent research suggests that G6PD-deficient cells are susceptible to infection by human coronaviruses, as the G6PD enzyme is involved in the metabolism of oxidative stress, which may enhance COVID-19 mortality. This retrospective study aimed to examine the effect of COVID-19 on patients with G6PD deficiency by comparing the laboratory parameters of patients with G6PD enzyme deficiency alone, COVID-19 alone, and those with both COVID-19 and G6PD enzyme deficiency treated at a major Saudi tertiary center. The results indicated significant differences in hematological and biochemical parameters between the three patient groups, indicating that COVID-19 may influence these parameters, and that they could be used to measure the severity of COVID-19 disease. Moreover, this study suggests that patients with G6PD enzyme deficiency may be at higher risk for severe COVID-19 outcomes. Although the study is limited by the lack of a random selection method for group membership, the Kruskal-Wallis H-test was used to statistical assess the data. The study's findings can enhance the understanding of the relation between COVID-19 infected and G6PD-deficiency patients and inform clinical decision making for an improved patient outcome.
Collapse
Affiliation(s)
- Badi A Alotaibi
- Department of Clinical Laboratory Sciences, Collage of Applied Medical Sciences, King Saud Bin Abdulaziz University for Health Sciences, Riyadh 11481, Saudi Arabia
- King Abdullah International Medical Research Center, P.O. Box 3660, Riyadh 11481, Saudi Arabia
| | - Jehad A Aldali
- Department of Pathology, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 13317, Saudi Arabia
| | - Hamzah J Aldali
- Cellular and Molecular Medicine, College of Biomedical Science, University of Bristol, Bristol BS8 1QU, UK
| | - Glowi A Alasiri
- Department of Biochemistry, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 13317, Saudi Arabia
| | - Emadeldin M Elsokkary
- Psychology, Organisation, Imam Mohammed Ibn Saud Islamic University (IMSIU), Riyadh 13317, Saudi Arabia
| | - Areej Al Mugairi
- Hematopathology Division, Department of Pathology and Laboratory Medicine, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh 11426, Saudi Arabia
| | - Abdulaziz M Almuqrin
- Department of Clinical Laboratory Sciences, Collage of Applied Medical Sciences, King Saud University, Riyadh 11433, Saudi Arabia
| |
Collapse
|
17
|
Araújo A, Sgorlon G, Aguiar LE, Cidrão MHMC, Teixeira KS, Villalobos Salcedo JM, Passos-Silva AM, Vieira D. Influence of polymorphic variations of IFNL, HLA, and IL-6 genes in severe cases of COVID-19. Exp Biol Med (Maywood) 2023; 248:787-797. [PMID: 37452704 PMCID: PMC10350587 DOI: 10.1177/15353702231181343] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2023] Open
Abstract
The administration of vaccination doses to the global population has led to a decrease in the incidence of COVID-19. However, the clinical picture developed by infected individuals remains extremely concerning due to the great variability in the severity of cases even in vaccinated individuals. The clinical progression of the pathology is characterized by various influential factors such as sex, age group, comorbidities, and the genetics of the individual. The immune response to viral infections can be strongly influenced by the genetics of individuals; nucleotide variations called single-nucleotide polymorphisms (SNPs) in structures involved in the innate and adaptive immune response such as interferon (IFN)-λ, human leukocyte antigen (HLA), and interleukin (IL)-6 are frequently associated with pathological progression. In this study, we conducted a review of the main SNPs of these structures that are associated with severity in COVID-19. Searches were conducted on some platforms of the National Center for Biotechnology and Information (NCBI), and 102 studies were selected for full reading according to the inclusion criteria. IFNs showed a strong association with antiviral function, specifically, IFN-λ3 (IL-28B) demonstrated genetic variants commonly related to clinical progression in various pathologies. For COVID-19, rs12979860 and rs1298275 presented frequently described unfavorable genotypes for pathological conditions of hepatitis C and hepatocellular carcinoma. The high genetic variability of HLA was reported in the studies as a crucial factor relevant to the late immune response, mainly due to its ability to recognize antigens, with the HLA-B*46:01 SNP being associated with susceptibility to COVID-19. For IL-6, rs1554606 showed a strong relationship with the clinical progression of COVID-19. In addition, rs2069837 was identified with possible host protection relationships when linked to this infection.
Collapse
Affiliation(s)
- Adrhyan Araújo
- Laboratório de Virologia Molecular, Fundação Oswaldo Cruz Rondônia (FIOCRUZ/RO), Porto Velho 76812-329, Brazil
- Centro de Pesquisa em Medicina Tropical (CEPEM), Porto Velho 76812-329, Brazil
| | - Gabriella Sgorlon
- Laboratório de Virologia Molecular, Fundação Oswaldo Cruz Rondônia (FIOCRUZ/RO), Porto Velho 76812-329, Brazil
- Centro de Pesquisa em Medicina Tropical (CEPEM), Porto Velho 76812-329, Brazil
- Programa de Pós-Graduação em Biologia Experimental, Universidade Federal de Rondônia (UNIR), Porto Velho 76801-059, Brazil
| | | | | | - Karolaine Santos Teixeira
- Laboratório de Virologia Molecular, Fundação Oswaldo Cruz Rondônia (FIOCRUZ/RO), Porto Velho 76812-329, Brazil
- Centro de Pesquisa em Medicina Tropical (CEPEM), Porto Velho 76812-329, Brazil
| | - Juan Miguel Villalobos Salcedo
- Laboratório de Virologia Molecular, Fundação Oswaldo Cruz Rondônia (FIOCRUZ/RO), Porto Velho 76812-329, Brazil
- Universidade Federal de Rondônia (UNIR), Porto Velho 76801-059, Brazil
| | - Ana Maísa Passos-Silva
- Laboratório de Virologia Molecular, Fundação Oswaldo Cruz Rondônia (FIOCRUZ/RO), Porto Velho 76812-329, Brazil
- Centro de Pesquisa em Medicina Tropical (CEPEM), Porto Velho 76812-329, Brazil
- Programa de Pós-Graduação em Biologia Experimental, Universidade Federal de Rondônia (UNIR), Porto Velho 76801-059, Brazil
| | - Deusilene Vieira
- Laboratório de Virologia Molecular, Fundação Oswaldo Cruz Rondônia (FIOCRUZ/RO), Porto Velho 76812-329, Brazil
- Centro de Pesquisa em Medicina Tropical (CEPEM), Porto Velho 76812-329, Brazil
- Programa de Pós-Graduação em Biologia Experimental, Universidade Federal de Rondônia (UNIR), Porto Velho 76801-059, Brazil
| |
Collapse
|
18
|
Słomian D, Szyda J, Dobosz P, Stojak J, Michalska-Foryszewska A, Sypniewski M, Liu J, Kotlarz K, Suchocki T, Mroczek M, Stępień M, Sztromwasser P, Król ZJ. Better safe than sorry-Whole-genome sequencing indicates that missense variants are significant in susceptibility to COVID-19. PLoS One 2023; 18:e0279356. [PMID: 36662838 PMCID: PMC9858061 DOI: 10.1371/journal.pone.0279356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 12/06/2022] [Indexed: 01/22/2023] Open
Abstract
Undoubtedly, genetic factors play an important role in susceptibility and resistance to COVID-19. In this study, we conducted the GWAS analysis. Out of 15,489,173 SNPs, we identified 18,191 significant SNPs for severe and 11,799 SNPs for resistant phenotype, showing that a great number of loci were significant in different COVID-19 representations. The majority of variants were synonymous (60.56% for severe, 58.46% for resistant phenotype) or located in introns (55.77% for severe, 59.83% for resistant phenotype). We identified the most significant SNPs for a severe outcome (in AJAP1 intron) and for COVID resistance (in FIG4 intron). We found no missense variants with a potential causal function on resistance to COVID-19; however, two missense variants were determined as significant a severe phenotype (in PM20D1 and LRP4 exons). None of the aforementioned SNPs and missense variants found in this study have been previously associated with COVID-19.
Collapse
Affiliation(s)
- Dawid Słomian
- National Research Institute of Animal Production, Balice, Poland
| | - Joanna Szyda
- National Research Institute of Animal Production, Balice, Poland
- Department of Genetics, Biostatistics Group, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | - Paula Dobosz
- Central Clinical Hospital of Ministry of the Interior and Administration in Warsaw, Warsaw, Poland
- Department of Haematology, Transplantation and Internal Medicine, University Clinical Centre of the Medical University of Warsaw, Warsaw, Poland
| | - Joanna Stojak
- Central Clinical Hospital of Ministry of the Interior and Administration in Warsaw, Warsaw, Poland
- Department of Experimental Embryology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Magdalenka, Poland
| | | | - Mateusz Sypniewski
- Central Clinical Hospital of Ministry of the Interior and Administration in Warsaw, Warsaw, Poland
- Department of Genetics and Animal Breedings, Poznan University of Life Sciences, Poznan, Poland
| | - Jakub Liu
- Department of Genetics, Biostatistics Group, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | - Krzysztof Kotlarz
- Department of Genetics, Biostatistics Group, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | - Tomasz Suchocki
- National Research Institute of Animal Production, Balice, Poland
- Department of Genetics, Biostatistics Group, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | - Magdalena Mroczek
- Center for Cardiovascular Genetics & Gene Diagnostics, Foundation for People with Rare Diseases, Schlieren-Zurich, Switzerland
| | - Maria Stępień
- Department of Infectious Diseases, Doctoral School, Medical University of Lublin, Lublin, Poland
| | | | - Zbigniew J. Król
- Central Clinical Hospital of Ministry of the Interior and Administration in Warsaw, Warsaw, Poland
| |
Collapse
|
19
|
Karami Z, Moradi S, Eidi A, Soleimani M, Jafarian A. Induced pluripotent stem cells: Generation methods and a new perspective in COVID-19 research. Front Cell Dev Biol 2023; 10:1050856. [PMID: 36733338 PMCID: PMC9887183 DOI: 10.3389/fcell.2022.1050856] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 11/22/2022] [Indexed: 01/18/2023] Open
Abstract
Induced pluripotent stem cells (iPSCs) exhibit an unlimited ability to self-renew and produce various differentiated cell types, thereby creating high hopes for both scientists and patients as a great tool for basic research as well as for regenerative medicine purposes. The availability and safety of iPSCs for therapeutic purposes require safe and highly efficient methods for production of these cells. Different methods have been used to produce iPSCs, each of which has advantages and disadvantages. Studying these methods would be very helpful in developing an easy, safe, and efficient method for the generation of iPSCs. Since iPSCs can be generated from somatic cells, they can be considered as valuable cellular resources available for important research needs and various therapeutic purposes. Coronavirus disease 2019 (COVID-19) is a disease that has endangered numerous human lives worldwide and currently has no definitive cure. Therefore, researchers have been rigorously studying and examining all aspects of COVID-19 and potential treatment modalities and various drugs in order to enable the treatment, control, and prevention of COVID-19. iPSCs have become one of the most attractive and promising tools in this field by providing the ability to study COVID-19 and the effectiveness of drugs on this disease outside the human body. In this study, we discuss the different methods of generation of iPSCs as well as their respective advantages and disadvantages. We also present recent applications of iPSCs in the study and treatment of COVID-19.
Collapse
Affiliation(s)
- Zahra Karami
- 1Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Sharif Moradi
- 2Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Akram Eidi
- 1Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Masoud Soleimani
- 3Hematology and Cell Therapy Department, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran,4Department of Tissue Engineering and Applied Cell Science, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arefeh Jafarian
- 5Iranian Tissue Bank and Research Center, Tehran University of Medical Sciences, Tehran, Iran,*Correspondence: Arefeh Jafarian,
| |
Collapse
|
20
|
Garmendia JV, García AH, De Sanctis CV, Hajdúch M, De Sanctis JB. Autoimmunity and Immunodeficiency in Severe SARS-CoV-2 Infection and Prolonged COVID-19. Curr Issues Mol Biol 2022; 45:33-50. [PMID: 36661489 PMCID: PMC9857622 DOI: 10.3390/cimb45010003] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/08/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022] Open
Abstract
SARS-CoV-2 causes the complex and heterogeneous illness known as COVID-19. The disease primarily affects the respiratory system but can quickly become systemic, harming multiple organs and leading to long-lasting sequelae in some patients. Most infected individuals are asymptomatic or present mild symptoms. Antibodies, complement, and immune cells can efficiently eliminate the virus. However, 20% of individuals develop severe respiratory illness and multiple organ failure. Virus replication has been described in several organs in patients who died from COVID-19, suggesting a compromised immune response. Immunodeficiency and autoimmunity are responsible for this impairment and facilitate viral escape. Mutations in IFN signal transduction and T cell activation are responsible for the inadequate response in young individuals. Autoantibodies are accountable for secondary immunodeficiency in patients with severe infection or prolonged COVID-19. Antibodies against cytokines (interferons α, γ and ω, IL1β, IL6, IL10, IL-17, IL21), chemokines, complement, nuclear proteins and DNA, anticardiolipin, and several extracellular proteins have been reported. The type and titer of autoantibodies depend on age and gender. Organ-specific autoantibodies have been described in prolonged COVID-19. Their role in the disease is under study. Autoimmunity and immunodeficiency should be screened as risk factors for severe or prolonged COVID-19.
Collapse
Affiliation(s)
- Jenny Valentina Garmendia
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, 779 00 Olomouc, Czech Republic
| | - Alexis Hipólito García
- Institute of Immunology, Faculty of Medicine, Universidad Central de Venezuela, Caracas 1040, Venezuela
| | - Claudia Valentina De Sanctis
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, 779 00 Olomouc, Czech Republic
| | - Marián Hajdúch
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, 779 00 Olomouc, Czech Republic
- Czech Institute of Advanced Technology in Research [Catrin], Palacky University, 779 00 Olomouc, Czech Republic
| | - Juan Bautista De Sanctis
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, 779 00 Olomouc, Czech Republic
- Czech Institute of Advanced Technology in Research [Catrin], Palacky University, 779 00 Olomouc, Czech Republic
- Correspondence:
| |
Collapse
|
21
|
Pal M, Muinao T, Parihar A, Roy DK, Boruah HPD, Mahindroo N, Khan R. Biosensors based detection of novel biomarkers associated with COVID-19: Current progress and future promise. BIOSENSORS & BIOELECTRONICS: X 2022; 12:100281. [PMID: 36405494 PMCID: PMC9661549 DOI: 10.1016/j.biosx.2022.100281] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 11/02/2022] [Accepted: 11/03/2022] [Indexed: 11/16/2022]
Abstract
The pandemic situation of COVID-19 has caused global alarm in health care, devastating loss of lives, strangled economy, and paralysis of normal livelihood. The high inter-individual transmission rate created havoc in the global community. Although tremendous efforts are pitching in from across the globe to understand this disease, the clinical features seemed to have a wide range including fever, cough, and fatigue are the prominent features. Congestion, rhinorrhea, sore throat, and diarrhea are other less common features observed. The challenge of this disease lies in the difficulty in maneuvering the clinical course causing severe complications. One of the major causative factors for multi-organ failure in patients with severe COVID-19 complications is systemic vasculitis and cytokine-mediated coagulation disorders. Hence, effective markers trailing the disease severity and disease prognosis are urgently required for prompt medical treatment. In this review article, we have emphasized currently identified inflammatory, hematological, immunological, and biochemical biomarkers of COVID-19. We also discussed currently available biosensors for the detection of COVID-19-associated biomarkers & risk factors and the detection methods as well as their performances. These could be effective tools for rapid and more promising diagnoses in the current pandemic situation. Effective biomarkers and their rapid, scalable, & sensitive detection might be beneficial for the prevention of serious complications and the clinical management of the disease.
Collapse
Affiliation(s)
- Mintu Pal
- Biotechnology Group, Biological Sciences and Technology Division, CSIR-North East Institute of Science and Technology (NEIST), Academy of Scientific & Innovative Research (AcSIR), CSIR-NEIST Campus, Jorhat, Assam, 785006, India
- Department of Pharmacology, All India Institute of Medical Sciences (AIIMS), Bathinda, Punjab, 151001, India
| | - Thingreila Muinao
- Biotechnology Group, Biological Sciences and Technology Division, CSIR-North East Institute of Science and Technology (NEIST), Academy of Scientific & Innovative Research (AcSIR), CSIR-NEIST Campus, Jorhat, Assam, 785006, India
| | - Arpana Parihar
- CSIR-Advanced Materials and Processes Research Institute (AMPRI), Hoshangabad Road, Bhopal, 462026, MP, India
| | - Dilip Kumar Roy
- Department of Pharmaceutical Technology, JIS University, Kolkata, 700109, India
| | - Hari Prasanna Deka Boruah
- Biotechnology Group, Biological Sciences and Technology Division, CSIR-North East Institute of Science and Technology (NEIST), Academy of Scientific & Innovative Research (AcSIR), CSIR-NEIST Campus, Jorhat, Assam, 785006, India
- Government Model College, Kaziranga, Golaghat, Assam, 785609, India
| | - Neeraj Mahindroo
- School of Pharmacy, Dr. Vishwanath Karad MIT World Peace University, Pune, Maharashtra, 411038, India
| | - Raju Khan
- CSIR-Advanced Materials and Processes Research Institute (AMPRI), Hoshangabad Road, Bhopal, 462026, MP, India
| |
Collapse
|
22
|
Ali HN, Niranji SS, Al-Jaf SMA. Association of Toll-like receptor-4 polymorphism with SARS CoV-2 infection in Kurdish Population. HUMAN GENE (AMSTERDAM, NETHERLANDS) 2022; 34:201115. [PMID: 37521442 PMCID: PMC9529343 DOI: 10.1016/j.humgen.2022.201115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 09/17/2022] [Accepted: 09/28/2022] [Indexed: 11/07/2022]
Abstract
Genetic variations are critical for understanding clinical outcomes of infections including server acute respiratory syndrome coronavirus 2 (SARS CoV-2). The immunological reactions of human immune genes with SARS CoV-2 have been under investigation. Toll-like receptors (TLRs), a group of proteins, are important for microbial detections including bacteria and viruses. TLR4 can sense both bacterial lipopolysaccharides (LPS) and endogenous oxidized phospholipids triggered by Covid-19 infection. Two TLR4 single nucleotide polymorphisms (SNPs), Asp299Gly and Thr399Ile have been linked to infectious diseases. No studies have focused on these SNPs in association with Covid-19. This study aims to reveal the association between Covid-19 infection with these SNPs by comparing a group of patients and a general population. Restriction fragment length polymorphisms (RFLP) were used to identify the TLR4 SNPs in both the general population (n = 114) and Covid-19 patient groups (n = 125). The results found no association between the TLR4 polymorphisms and Covid-19 infections as the data showed no statistically significant difference between the compared groups. This suggested that these TLR4 SNPs may not be associated with Covid-19 infections.
Collapse
Key Words
- ACE2, Angiotensin converting enzyme 2
- ARSD, Acute respiratory distress syndrome
- Covid-19
- Covid-19, Coronavirus disease 2019
- IL, Interleukin
- LPS, Lipopolysaccharides (LPS)
- MyD88, Myeloid differentiation factor
- PCR-RFLP, Polymerase chain reaction-restriction fragment polymorphism
- SARS CoV-2
- SARS CoV-2, Server acute respiratory syndrome coronavirus 2
- SNP
- SNPs, Single nucleotide polymorphisms
- TLR4
- TLRs, Toll-like receptors
- TNF-α, Tumour necrosis factor alpha;
- TRIF, TIR-domain-containing adapter-inducing interferon-β.
- polymorphism
Collapse
Affiliation(s)
- Hussein N Ali
- College of Medicine, University of Garmian, Kurdistan region, Iraq
| | - Sherko S Niranji
- College of Medicine, University of Garmian, Kurdistan region, Iraq
- Coronavirus Research and Identification Lab., University of Garmian, Kurdistan region, Iraq
- Department of Biology, College of Education, University of Garmian, Kurdistan region, Iraq
| | - Sirwan M A Al-Jaf
- College of Medicine, University of Garmian, Kurdistan region, Iraq
- Coronavirus Research and Identification Lab., University of Garmian, Kurdistan region, Iraq
- Department of Biology, College of Education, University of Garmian, Kurdistan region, Iraq
| |
Collapse
|
23
|
Devarashetty SP, Ponna PK, Devarakonda V, Ramadas P. COVID-19 infection unmasking glucose-6-phosphate dehydrogenase deficiency. QJM 2022; 115:771-772. [PMID: 35904760 PMCID: PMC9384615 DOI: 10.1093/qjmed/hcac184] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 06/28/2022] [Indexed: 11/30/2022] Open
Affiliation(s)
- S P Devarashetty
- Address correspondence to S.P. Devarashetty, Department of Hematology-Oncology, Louisiana State University Health Shreveport, Shreveport, LA, USA.
| | - P K Ponna
- Department of Internal Medicine, Louisiana State University Health Shreveport, 1501 Kings Highway, Shreveport, LA, 71103, USA
| | - V Devarakonda
- Department of Internal Medicine, Louisiana State University Health Shreveport, 1501 Kings Highway, Shreveport, LA, 71103, USA
| | - Poornima Ramadas
- Department of Hematology-Oncology, Louisiana State University Health Shreveport, 1501 Kings Highway, Shreveport, LA, 71103, USA
| |
Collapse
|
24
|
Genetic variants determine intrafamilial variability of SARS-CoV-2 clinical outcomes in 19 Italian families. PLoS One 2022; 17:e0275988. [PMID: 36228008 PMCID: PMC9560599 DOI: 10.1371/journal.pone.0275988] [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: 03/09/2022] [Accepted: 09/27/2022] [Indexed: 11/07/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection results in a wide range of outcomes characterized by a high heterogeneity in both symptomatology and susceptibility to the disease. In such a perspective, COVID-19 may be considered as a multifactorial disease featured by the interaction between the environment, which is the virus itself, and the genetic profile of the host. Our analysis aimed at investigating the transmission dynamics of SARS-CoV-2 within families whose members responded in different ways to the infection, although the exposure was common to the entire group and occurred before the availability of any vaccine. The goal was to understand how the genetic background of each subject can influence the viral infection outcome and hence the above-mentioned clinical variability. We performed a segregation analysis in 19 Italian families with a designed custom panel of 42 genes involved in immunity and virus entry and which have also been shown to be related to SARS-CoV-2 host response. We carried out both a familial segregation analysis and a global statistical analysis. In the former we identified eighteen risk variants co-segregating with a COVID-positive status and six variants with a possible protective effect. In addition, sixteen variants showed a trend of association to a severe phenotype. Together with common SNPs, we detected private rare variants that may also provide insight into the observed clinical COVID-19 heterogeneity. The global statistical analysis confirmed statistically significant positive associations between SARS-CoV-2 individual response and some specific gene variants identified in familial analysis. In conclusion our data confirm that the clinical expression of COVID-19 is markedly influenced by the host genetic profile both with a mendelian transmission pattern and a polygenic architecture.
Collapse
|
25
|
Rizvi S, Rizvi SM, Raza ST, Abbas M, Fatima K, Zaidi ZH, Mahdi F. Implication of single nucleotide polymorphisms in Interleukin-10 gene ( rs1800896 and rs1800872) with severity of COVID-19. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2022; 23:145. [PMID: 37521849 PMCID: PMC9526386 DOI: 10.1186/s43042-022-00344-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 08/18/2022] [Indexed: 11/21/2022] Open
Abstract
Background Coronavirus disease 2019 (COVID-19) is an ongoing pandemic which has emerged as a new challenge for the medical sciences. Severity of COVID-19 is mostly determined with overexpressed proinflammatory cytokines eventually leading to endothelial dysfunction causing vital organ injury, especially in the lungs. It has been postulated that various genetic mutations might be associated with an increased risk of disease severity in COVID-19. This study was thus carried out to determine the association of rs1800896 and rs1800872 genetic polymorphism in IL-10 gene in determining COVID-19 severity. Methods The study included 160 RT-PCR confirmed COVID-19 patients with mild (n = 85) and severe (n = 75) conditions. All subjects were genotyped for Interleukin-10 (rs1800896 and rs1800872) gene polymorphisms using PCR-RFLP technique followed by statistical analysis. Results This study found a significant gender and age-based discrepancy in COVID-19 severity with 1.85-and 3.81-fold increased risk of COVID-19 in males of mild and severe groups as compared to females (p = 0.046 and p < 0.001) and 4.35-fold high risk in subjects ≥ 50 (p < 0.001). Genotyping analysis showed that IL-10 (rs1800872) gene polymorphism was strongly associated with COVID-19 severity (p = 0.01) whereas, IL-10 rs1800896 polymorphism was not found to confer the risk of COVID-19 severity in our population. Conclusion In this regard, the present study provided an evidence that IL-10 (rs1800872) gene polymorphism is strongly associated with COVID-19 severity and CC genotype confer a protective role in preventing severe disease progression. More detailed studies with a larger sample size on the genetic variations are required to establish the role of studied IL-10 gene polymorphisms with COVID-19 severity.
Collapse
Affiliation(s)
- Saliha Rizvi
- Department of Biotechnology, Era’s Lucknow Medical College and Hospital, Era University, Lucknow, Uttar Pradesh 226003 India
- Department of Personalized and Molecular Medicine, Era University, Lucknow, Uttar Pradesh 226003 India
| | - S. Mohd.Shiraz Rizvi
- Department of Biochemistry, Era’s Lucknow Medical College and Hospital, Lucknow, Uttar Pradesh 226003 India
| | - Syed Tasleem Raza
- Department of Biotechnology, Era’s Lucknow Medical College and Hospital, Era University, Lucknow, Uttar Pradesh 226003 India
- Department of Biochemistry, Era’s Lucknow Medical College and Hospital, Lucknow, Uttar Pradesh 226003 India
| | - Mohd. Abbas
- Department of Personalized and Molecular Medicine, Era University, Lucknow, Uttar Pradesh 226003 India
| | - Kaynat Fatima
- Department of Biotechnology, Era’s Lucknow Medical College and Hospital, Era University, Lucknow, Uttar Pradesh 226003 India
| | - Zeashan H. Zaidi
- Department of Community Medicine, Era’s Lucknow Medical College and Hospital, Lucknow, Uttar Pradesh 226003 India
| | - Farzana Mahdi
- Department of Biotechnology, Era’s Lucknow Medical College and Hospital, Era University, Lucknow, Uttar Pradesh 226003 India
- Department of Personalized and Molecular Medicine, Era University, Lucknow, Uttar Pradesh 226003 India
| |
Collapse
|
26
|
Bencze D, Fekete T, Pázmándi K. Correlation between Type I Interferon Associated Factors and COVID-19 Severity. Int J Mol Sci 2022; 23:ijms231810968. [PMID: 36142877 PMCID: PMC9506204 DOI: 10.3390/ijms231810968] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/11/2022] [Accepted: 09/13/2022] [Indexed: 11/18/2022] Open
Abstract
Antiviral type I interferons (IFN) produced in the early phase of viral infections effectively inhibit viral replication, prevent virus-mediated tissue damages and promote innate and adaptive immune responses that are all essential to the successful elimination of viruses. As professional type I IFN producing cells, plasmacytoid dendritic cells (pDC) have the ability to rapidly produce waste amounts of type I IFNs. Therefore, their low frequency, dysfunction or decreased capacity to produce type I IFNs might increase the risk of severe viral infections. In accordance with that, declined pDC numbers and delayed or inadequate type I IFN responses could be observed in patients with severe coronavirus disease (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), as compared to individuals with mild or no symptoms. Thus, besides chronic diseases, all those conditions, which negatively affect the antiviral IFN responses lengthen the list of risk factors for severe COVID-19. In the current review, we would like to briefly discuss the role and dysregulation of pDC/type I IFN axis in COVID-19, and introduce those type I IFN-dependent factors, which account for an increased risk of COVID-19 severity and thus are responsible for the different magnitude of individual immune responses to SARS-CoV-2.
Collapse
Affiliation(s)
- Dóra Bencze
- Department of Immunology, Faculty of Medicine, University of Debrecen, 1 Egyetem Square, H-4032 Debrecen, Hungary
- Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, 1 Egyetem Square, H-4032 Debrecen, Hungary
| | - Tünde Fekete
- Department of Immunology, Faculty of Medicine, University of Debrecen, 1 Egyetem Square, H-4032 Debrecen, Hungary
| | - Kitti Pázmándi
- Department of Immunology, Faculty of Medicine, University of Debrecen, 1 Egyetem Square, H-4032 Debrecen, Hungary
- Correspondence: ; Tel./Fax: +36-52-417-159
| |
Collapse
|
27
|
Association of the Transmembrane Serine Protease-2 (TMPRSS2) Polymorphisms with COVID-19. Viruses 2022; 14:v14091976. [PMID: 36146782 PMCID: PMC9505830 DOI: 10.3390/v14091976] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/29/2022] [Accepted: 09/01/2022] [Indexed: 12/23/2022] Open
Abstract
SARS-CoV-2 uses the ACE2 receptor and the cellular protease TMPRSS2 for entry into target cells. The present study aimed to establish if the TMPRSS2 polymorphisms are associated with COVID-19 disease. The study included 609 patients with COVID-19 confirmed by RT-PCR test and 291 individuals negative for the SARS-CoV-2 infection confirmed by RT-PCR test and without antibodies anti-SARS-CoV-2. Four TMPRSS2 polymorphisms (rs12329760, rs2298659, rs456298, and rs462574) were determined using the 5′exonuclease TaqMan assays. Under different inheritance models, the rs2298659 (pcodominant2 = 0.018, precessive = 0.006, padditive = 0.019), rs456298 (pcodominant1 = 0.014, pcodominant2 = 0.004; pdominant = 0.009, precessive = 0.004, padditive = 0.0009), and rs462574 (pcodominant1 = 0.017, pcodominant2 = 0.004, pdominant = 0.041, precessive = 0.002, padditive = 0.003) polymorphisms were associated with high risk of developing COVID-19. Two risks (ATGC and GAAC) and two protectives (GAGC and GAGT) haplotypes were detected. High levels of lactic acid dehydrogenase (LDH) were observed in patients with the rs462574AA and rs456298TT genotypes (p = 0.005 and p = 0.020, respectively), whereas, high heart rate was present in patients with the rs462574AA genotype (p = 0.028). Our data suggest that the rs2298659, rs456298, and rs462574 polymorphisms independently and as haplotypes are associated with the risk of COVID-19. The rs456298 and rs462574 genotypes are related to high levels of LDH and heart rate.
Collapse
|
28
|
Inflammasome Genetic Variants Are Associated with Protection to Clinical Severity of COVID-19 among Patients from Rio de Janeiro, Brazil. BIOMED RESEARCH INTERNATIONAL 2022; 2022:9082455. [PMID: 36105941 PMCID: PMC9467712 DOI: 10.1155/2022/9082455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 08/18/2022] [Accepted: 08/24/2022] [Indexed: 11/18/2022]
Abstract
COVID-19 has a broad spectrum of clinical manifestations, from asymptomatic or mild/moderate symptoms to severe symptoms and death. The mechanisms underlying its clinical evolution are still unclear. Upon SARS-CoV-2 infection, host factors, such as the inflammasome system, are activated by the presence of the virus inside host cells. The search for COVID-19 risk factors is of relevance for clinical management. In this study, we investigated the impact of inflammasome single-nucleotide polymorphisms (SNPs) in SARS-CoV-2-infected individuals with distinct severity profiles at clinical presentation. Patients were divided into two groups according to disease severity at clinical presentation based on the WHO Clinical Progression Scale. Group 1 included patients with mild/moderate disease (WHO < 6; n = 76), and group 2 included patients with severe/critical COVID-19 (WHO ≥ 6; n = 357). Inpatients with moderate to severe/critical profiles were recruited and followed-up at Hospital Center for COVID-19 Pandemic – National Institute of Infectology (INI)/FIOCRUZ, RJ, Brazil, from June 2020 to March 2021. Patients with mild disease were recruited at Oswaldo Cruz Institute (IOC)/FIOCRUZ, RJ, Brazil, in August 2020. Genotyping of 11 inflammasome SNPs was determined by real-time PCR. Protection and risk estimation were performed using unconditional logistic regression models. Significant differences in NLRP3 rs1539019 and CARD8 rs2043211 were observed between the two groups. Protection against disease severity was associated with the A/A genotype (ORadj = 0.36; P = 0.032), allele A (ORadj = 0.93; P = 0.010), or carrier-A (ORadj = 0.45; P = 0.027) in the NLRP3 rs1539019 polymorphism; A/T genotype (ORadj = 0.5; P = 0.045), allele T (ORadj = 0.93; P = 0.018), or carrier-T (ORadj = 0.48; P = 0.029) in the CARD8 rs2043211 polymorphism; and the A-C-G-C-C (ORadj = 0.11; P = 0.018), A-C-G-C-G (ORadj = 0.23; P = 0.003), C-C-G-C-C (ORadj = 0.37; P = 0.021), and C-T-G-A-C (ORadj = 0.04; P = 0.0473) in NLRP3 genetic haplotype variants. No significant associations were observed for the other polymorphisms. To the best of our knowledge, this is the first study demonstrating an association between CARD8 and NLRP3 inflammasome genetic variants and protection against COVID-19 severity, contributing to the discussion of the impact of inflammasomes on COVID-19 outcomes.
Collapse
|
29
|
Fan J, Long QX, Ren JH, Chen H, Li MM, Cheng Z, Chen J, Zhou L, Huang AL. Genome-wide association study of SARS-CoV-2 infection in Chinese population. Eur J Clin Microbiol Infect Dis 2022; 41:1155-1163. [PMID: 35927536 PMCID: PMC9362144 DOI: 10.1007/s10096-022-04478-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 07/18/2022] [Indexed: 11/28/2022]
Abstract
Coronavirus disease 2019 (COVID-19) is a global public health concern. The purpose of this study was to investigate the association between genetic variants and SARS-CoV-2 infection and the COVID-19 severity in Chinese population. A total of 256 individuals including 87 symptomatic patients (tested positive for SARS-CoV-2), 84 asymptomatic cases, and 85 close contacts of confirmed patients (tested negative for SARS-CoV-2) were recruited from February 2020 to May 2020. We carried out the whole exome genome sequencing between the individuals and conducted a genetic association study for SARS-CoV-2 infection and the COVID-19 severity. In total, we analyzed more than 100,000 single-nucleotide polymorphisms. The genome-wide association study suggested potential correlation between genetic variability in POLR2A, ANKRD27, MAN1A2, and ERAP1 genes and SARS-CoV-2 infection susceptibility. The most significant gene locus associated with SARS-CoV-2 infection was located in POLR2A (p = 5.71 × 10-6). Furthermore, genetic variants in PCNX2, CD200R1L, ZMAT3, PLCL2, NEIL3, and LINC00700 genes (p < 1 × 10-5) were closely associated with the COVID-19 severity in Chinese population. Our study confirmed that new genetic variant loci had significant association with SARS-CoV-2 infection and the COVID-19 severity in Chinese population, which provided new clues for the studies on the susceptibility of SARS-CoV-2 infection and the COVID-19 severity. These findings may give a better understanding on the molecular pathogenesis of COVID-19 and genetic basis of heterogeneous susceptibility, with potential impact on new therapeutic options.
Collapse
Affiliation(s)
- Jie Fan
- Department of Epidemiology, School of Public Health and Management, Research Center for Medicine and Social Development, Innovation Center for Social Risk Governance in Health, Chongqing Medical University, Chongqing, 400016, China.,Nanan District Center for Disease Control and Prevention, Chongqing, China
| | - Quan-Xin Long
- Key Laboratory of Molecular Biology On Infectious Diseases, Chinese Ministry of Education, Chongqing Medical University, Room 617, College of Life Sciences Building, 1 YixueYuan Road, YuZhong District, Chongqing, 400016, China
| | - Ji-Hua Ren
- Key Laboratory of Molecular Biology On Infectious Diseases, Chinese Ministry of Education, Chongqing Medical University, Room 617, College of Life Sciences Building, 1 YixueYuan Road, YuZhong District, Chongqing, 400016, China
| | - Hao Chen
- Department of Epidemiology, School of Public Health and Management, Research Center for Medicine and Social Development, Innovation Center for Social Risk Governance in Health, Chongqing Medical University, Chongqing, 400016, China
| | - Meng-Meng Li
- Department of Epidemiology, School of Public Health and Management, Research Center for Medicine and Social Development, Innovation Center for Social Risk Governance in Health, Chongqing Medical University, Chongqing, 400016, China
| | - Zheng Cheng
- Department of Epidemiology, School of Public Health and Management, Research Center for Medicine and Social Development, Innovation Center for Social Risk Governance in Health, Chongqing Medical University, Chongqing, 400016, China
| | - Juan Chen
- Key Laboratory of Molecular Biology On Infectious Diseases, Chinese Ministry of Education, Chongqing Medical University, Room 617, College of Life Sciences Building, 1 YixueYuan Road, YuZhong District, Chongqing, 400016, China. .,Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, Chongqing, China.
| | - Li Zhou
- Department of Epidemiology, School of Public Health and Management, Research Center for Medicine and Social Development, Innovation Center for Social Risk Governance in Health, Chongqing Medical University, Chongqing, 400016, China.
| | - Ai-Long Huang
- Key Laboratory of Molecular Biology On Infectious Diseases, Chinese Ministry of Education, Chongqing Medical University, Room 617, College of Life Sciences Building, 1 YixueYuan Road, YuZhong District, Chongqing, 400016, China.
| |
Collapse
|
30
|
David S, Dorado G, Duarte EL, David-Bosne S, Trigueiro-Louro J, Rebelo-de-Andrade H. COVID-19: impact on Public Health and hypothesis-driven investigations on genetic susceptibility and severity. Immunogenetics 2022; 74:381-407. [PMID: 35348847 PMCID: PMC8961091 DOI: 10.1007/s00251-022-01261-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 03/14/2022] [Indexed: 12/12/2022]
Abstract
COVID-19 is a new complex multisystem disease caused by the novel coronavirus SARS-CoV-2. In slightly over 2 years, it infected nearly 500 million and killed 6 million human beings worldwide, causing an unprecedented coronavirus pandemic. Currently, the international scientific community is engaged in elucidating the molecular mechanisms of the pathophysiology of SARS-CoV-2 infection as a basis of scientific developments for the future control of COVID-19. Global exome and genome analysis efforts work to define the human genetics of protective immunity to SARS-CoV-2 infection. Here, we review the current knowledge regarding the SARS-CoV-2 infection, the implications of COVID-19 to Public Health and discuss genotype to phenotype association approaches that could be exploited through the selection of candidate genes to identify the genetic determinants of severe COVID-19.
Collapse
Affiliation(s)
- Susana David
- Departamento de Genética Humana, Instituto Nacional de Saúde Doutor Ricardo Jorge (INSA,IP), Lisboa, Portugal.
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Lisboa, Portugal.
| | - Guillermo Dorado
- Atlántida Centro de Investigación y Desarrollo de Estudios Profesionales (CIDEP), Granada, Spain
| | - Elsa L Duarte
- MED-Instituto Mediterrâneo para a Agricultura, Ambiente e Desenvolvimento, Escola de Ciências e Tecnologia, Universidade de Évora, Évora, Portugal
| | | | - João Trigueiro-Louro
- Departamento de Doenças Infeciosas, INSA, IP, Lisboa, Portugal
- Host-Pathogen Interaction Unit, Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Lisboa, Portugal
- Hospital Egas Moniz, Centro Hospitalar Lisboa Ocidental, Lisboa, Portugal
| | - Helena Rebelo-de-Andrade
- Departamento de Doenças Infeciosas, INSA, IP, Lisboa, Portugal
- Host-Pathogen Interaction Unit, Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
31
|
García-Carmona S, Falfán-Valencia R, Verónica-Aguilar A, Buendía-Roldán I, Chávez-Galán L, Hernández-Zenteno RDJ, Martínez-Morales A, Fricke-Galindo I, Alanis-Ponce J, Valencia-Pérez Rea D, Gutiérrez-Pérez IA, Zaragoza-García O, Nava-Quiroz KJ, Camarena A, Mejía M, Guzmán-Guzmán IP, Pérez-Rubio G. COVID-19 Survivor Patients Carrying the Rs35705950 Risk Allele in MUC5B Have Higher Plasma Levels of Mucin 5B. Curr Issues Mol Biol 2022; 44:3283-3290. [PMID: 35892712 PMCID: PMC9329856 DOI: 10.3390/cimb44080226] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/13/2022] [Accepted: 07/15/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Genetic susceptibility to infectious diseases is partly due to the variation in the human genome, and COVID-19 is not the exception. This study aimed to identify whether risk alleles of known genes linked with emphysema (SERPINA1) and pulmonary fibrosis (MUC5B) are associated with severe COVID-19, and whether plasma mucin 5B differs according to patients' outcomes. MATERIALS AND METHODS We included 1258 Mexican subjects diagnosed with COVID-19. We genotyped rs2892474 and rs17580 of the SERPINA1 gene and rs35705950 of MUC5B. Based on the rs35705950 genotypes, mucin 5B plasma protein levels were quantified. RESULTS Homozygous for the risk alleles of the three polymorphisms were found in less than 5% of the study population, but no statistically significant difference in the genotype or allele association analysis. At the protein level, non-survivors carrying one or two copies of the risk allele rs35705950 in MUC5B (GT + TT) had lower levels of mucin 5B compared to the survivors (0.0 vs. 0.17 ng/mL, p = 0.0013). CONCLUSION The polymorphisms rs28929474 and rs17580 of SERPINA1 and rs35705950 of MUC5B are not associated with the risk of severe COVID-19 in the Mexican population. COVID-19 survivor patients bearing one or two copies of the rs35705950 risk allele have higher plasma levels of mucin 5B.
Collapse
Affiliation(s)
- Salvador García-Carmona
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (S.G.-C.); (R.F.-V.); (A.V.-A.); (A.M.-M.); (I.F.-G.); (J.A.-P.); (D.V.-P.R.); (I.A.G.-P.); (K.J.N.-Q.); (A.C.)
| | - Ramcés Falfán-Valencia
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (S.G.-C.); (R.F.-V.); (A.V.-A.); (A.M.-M.); (I.F.-G.); (J.A.-P.); (D.V.-P.R.); (I.A.G.-P.); (K.J.N.-Q.); (A.C.)
| | - Abigail Verónica-Aguilar
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (S.G.-C.); (R.F.-V.); (A.V.-A.); (A.M.-M.); (I.F.-G.); (J.A.-P.); (D.V.-P.R.); (I.A.G.-P.); (K.J.N.-Q.); (A.C.)
| | - Ivette Buendía-Roldán
- Translational Research Laboratory on Aging and Pulmonary Fibrosis, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico;
| | - Leslie Chávez-Galán
- Laboratory of Integrative Immunology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico;
| | | | - Alfonso Martínez-Morales
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (S.G.-C.); (R.F.-V.); (A.V.-A.); (A.M.-M.); (I.F.-G.); (J.A.-P.); (D.V.-P.R.); (I.A.G.-P.); (K.J.N.-Q.); (A.C.)
| | - Ingrid Fricke-Galindo
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (S.G.-C.); (R.F.-V.); (A.V.-A.); (A.M.-M.); (I.F.-G.); (J.A.-P.); (D.V.-P.R.); (I.A.G.-P.); (K.J.N.-Q.); (A.C.)
| | - Jesús Alanis-Ponce
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (S.G.-C.); (R.F.-V.); (A.V.-A.); (A.M.-M.); (I.F.-G.); (J.A.-P.); (D.V.-P.R.); (I.A.G.-P.); (K.J.N.-Q.); (A.C.)
| | - Daniela Valencia-Pérez Rea
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (S.G.-C.); (R.F.-V.); (A.V.-A.); (A.M.-M.); (I.F.-G.); (J.A.-P.); (D.V.-P.R.); (I.A.G.-P.); (K.J.N.-Q.); (A.C.)
| | - Ilse Adriana Gutiérrez-Pérez
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (S.G.-C.); (R.F.-V.); (A.V.-A.); (A.M.-M.); (I.F.-G.); (J.A.-P.); (D.V.-P.R.); (I.A.G.-P.); (K.J.N.-Q.); (A.C.)
- Faculty of Chemical-Biological Sciences, Universidad Autónoma de Guerrero, Chilpancingo 39087, Mexico; (O.Z.-G.); (I.P.G.-G.)
| | - Oscar Zaragoza-García
- Faculty of Chemical-Biological Sciences, Universidad Autónoma de Guerrero, Chilpancingo 39087, Mexico; (O.Z.-G.); (I.P.G.-G.)
| | - Karol J. Nava-Quiroz
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (S.G.-C.); (R.F.-V.); (A.V.-A.); (A.M.-M.); (I.F.-G.); (J.A.-P.); (D.V.-P.R.); (I.A.G.-P.); (K.J.N.-Q.); (A.C.)
| | - Angel Camarena
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (S.G.-C.); (R.F.-V.); (A.V.-A.); (A.M.-M.); (I.F.-G.); (J.A.-P.); (D.V.-P.R.); (I.A.G.-P.); (K.J.N.-Q.); (A.C.)
| | - Mayra Mejía
- Interstitial Pulmonary Diseases and Rheumatology Unit, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico;
| | - Iris Paola Guzmán-Guzmán
- Faculty of Chemical-Biological Sciences, Universidad Autónoma de Guerrero, Chilpancingo 39087, Mexico; (O.Z.-G.); (I.P.G.-G.)
| | - Gloria Pérez-Rubio
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (S.G.-C.); (R.F.-V.); (A.V.-A.); (A.M.-M.); (I.F.-G.); (J.A.-P.); (D.V.-P.R.); (I.A.G.-P.); (K.J.N.-Q.); (A.C.)
| |
Collapse
|
32
|
Dieter C, Brondani LDA, Leitão CB, Gerchman F, Lemos NE, Crispim D. Genetic polymorphisms associated with susceptibility to COVID-19 disease and severity: A systematic review and meta-analysis. PLoS One 2022; 17:e0270627. [PMID: 35793369 PMCID: PMC9258831 DOI: 10.1371/journal.pone.0270627] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 06/15/2022] [Indexed: 12/17/2022] Open
Abstract
Although advanced age and presence of comorbidities significantly impact the variation observed in the clinical symptoms of COVID-19, it has been suggested that genetic variants may also be involved in the disease. Thus, the aim of this study was to perform a systematic review with meta-analysis of the literature to identify genetic polymorphisms that are likely to contribute to COVID-19 pathogenesis. Pubmed, Embase and GWAS Catalog repositories were systematically searched to retrieve articles that investigated associations between polymorphisms and COVID-19. For polymorphisms analyzed in 3 or more studies, pooled OR with 95% CI were calculated using random or fixed effect models in the Stata Software. Sixty-four eligible articles were included in this review. In total, 8 polymorphisms in 7 candidate genes and 74 alleles of the HLA loci were analyzed in 3 or more studies. The HLA-A*30 and CCR5 rs333Del alleles were associated with protection against COVID-19 infection, while the APOE rs429358C allele was associated with risk for this disease. Regarding COVID-19 severity, the HLA-A*33, ACE1 Ins, and TMPRSS2 rs12329760T alleles were associated with protection against severe forms, while the HLA-B*38, HLA-C*6, and ApoE rs429358C alleles were associated with risk for severe forms of COVID-19. In conclusion, polymorphisms in the ApoE, ACE1, TMPRSS2, CCR5, and HLA loci appear to be involved in the susceptibility to and/or severity of COVID-19.
Collapse
Affiliation(s)
- Cristine Dieter
- Endocrine and Metabolism Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
- Postgraduate Program in Medical Sciences: Endocrinology, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Letícia de Almeida Brondani
- Endocrine and Metabolism Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
- Postgraduate Program in Medical Sciences: Endocrinology, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Cristiane Bauermann Leitão
- Endocrine and Metabolism Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
- Postgraduate Program in Medical Sciences: Endocrinology, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Fernando Gerchman
- Endocrine and Metabolism Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
- Postgraduate Program in Medical Sciences: Endocrinology, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Natália Emerim Lemos
- Endocrine and Metabolism Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
- Postgraduate Program in Medical Sciences: Endocrinology, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Daisy Crispim
- Endocrine and Metabolism Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
- Postgraduate Program in Medical Sciences: Endocrinology, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
33
|
Pandit R, Singh I, Ansari A, Raval J, Patel Z, Dixit R, Shah P, Upadhyay K, Chauhan N, Desai K, Shah M, Modi B, Joshi M, Joshi C. First report on genome wide association study in western Indian population reveals host genetic factors for COVID-19 severity and outcome. Genomics 2022; 114:110399. [PMID: 35680011 PMCID: PMC9169419 DOI: 10.1016/j.ygeno.2022.110399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 05/18/2022] [Accepted: 06/01/2022] [Indexed: 02/07/2023]
Abstract
Different human races across the globe responded in a different way to the SARS-CoV-2 infection leading to different disease severity. Therefore, it is anticipated that host genetic factors have a straight association with the COVID-19. We identified a total 6, 7, and 6 genomic loci for deceased-recovered, asymptomatic-recovered, and deceased-asymptomatic group comparison, respectively. Unfavourable alleles of the markers nearby the genes which are associated with lung and heart diseases such as Tumor necrosis factor superfamily (TNFSF4&18), showed noteworthy association with the disease severity and outcome for the COVID-19 patients in the western Indian population. The markers found with significant association with disease prognosis or recovery are of value in determining the individual's response to SARS-CoV-2 infection and can be used for the risk prediction in COVID-19. Besides, GWAS study in other populations from India may help to strengthen the outcome of this study.
Collapse
Affiliation(s)
- Ramesh Pandit
- Gujarat Biotechnology Research Centre (GBRC), Department of Science and Technology (Government of Gujarat), Gandhinagar, Gujarat 382011, India
| | - Indra Singh
- Gujarat Biotechnology Research Centre (GBRC), Department of Science and Technology (Government of Gujarat), Gandhinagar, Gujarat 382011, India
| | - Afzal Ansari
- Gujarat Biotechnology Research Centre (GBRC), Department of Science and Technology (Government of Gujarat), Gandhinagar, Gujarat 382011, India
| | - Janvi Raval
- Gujarat Biotechnology Research Centre (GBRC), Department of Science and Technology (Government of Gujarat), Gandhinagar, Gujarat 382011, India
| | - Zarna Patel
- Gujarat Biotechnology Research Centre (GBRC), Department of Science and Technology (Government of Gujarat), Gandhinagar, Gujarat 382011, India
| | - Raghav Dixit
- Commissionerate of Health Medical Services and Medical Education Gandhinagar, Gujarat 382010, India
| | - Pranay Shah
- Department of Microbiology, B.J. Medical College and Civil hospital, Institute of Medical Post-Graduate Studies and Research, Ahmedabad, Gujarat 380016, India
| | - Kamlesh Upadhyay
- Department of Medicine, B.J. Medical College and Civil hospital, Institute of Medical Post-Graduate Studies and Research, Ahmedabad, Gujarat 380016, India
| | - Naresh Chauhan
- Department of Community Medicine, Government Medical College, Surat, Gujarat 395001, India
| | - Kairavi Desai
- Department of Microbiology, Government Medical College, Bhavnagar, Gujarat 364001, India
| | - Meenakshi Shah
- Department of General Medicine, GMERS Medical College & Hospital, Gotri, Vadodara, Gujarat 390021, India
| | - Bhavesh Modi
- Department of Community Medicine, GMERS Medical College, Gandhinagar, Gujarat 382012, India
| | - Madhvi Joshi
- Gujarat Biotechnology Research Centre (GBRC), Department of Science and Technology (Government of Gujarat), Gandhinagar, Gujarat 382011, India.
| | - Chaitanya Joshi
- Gujarat Biotechnology Research Centre (GBRC), Department of Science and Technology (Government of Gujarat), Gandhinagar, Gujarat 382011, India.
| |
Collapse
|
34
|
Rabaan AA, Bakhrebah MA, Mutair AA, Alhumaid S, Al-Jishi JM, AlSihati J, Albayat H, Alsheheri A, Aljeldah M, Garout M, Alfouzan WA, Alhashem YN, AlBahrani S, Alshamrani SA, Alotaibi S, AlRamadhan AA, Albasha HN, Hajissa K, Temsah MH. Systematic Review on Pathophysiological Complications in Severe COVID-19 among the Non-Vaccinated and Vaccinated Population. Vaccines (Basel) 2022; 10:985. [PMID: 35891149 PMCID: PMC9318201 DOI: 10.3390/vaccines10070985] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/13/2022] [Accepted: 06/17/2022] [Indexed: 02/06/2023] Open
Abstract
COVID-19, caused by SARS-CoV-2, is one of the longest viral pandemics in the history of mankind, which have caused millions of deaths globally and induced severe deformities in the survivals. For instance, fibrosis and cavities in the infected lungs of COVID-19 are some of the complications observed in infected patients post COVID-19 recovery. These health abnormalities, including is multiple organ failure-the most striking pathological features of COVID-19-have been linked with diverse distribution of ACE2 receptor. Additionally, several health complications reports were reported after administration of COVID-19 vaccines in healthy individuals, but clinical or molecular pathways causing such complications are not yet studied in detail. Thus, the present systematic review established the comparison of health complication noted in vaccinated and non-vaccinated individuals (COVID-19 infected patients) to identify the association between vaccination and the multiorgan failure based on the data obtained from case studies, research articles, clinical trials/Cohort based studies and review articles published between 2020-2022. This review also includes the biological rationale behind the COVID-19 infection and its subsequent symptoms and effects including multiorgan failure. In addition, multisystem inflammatory syndrome (MIS) has been informed in individuals post vaccination that resulted in multiorgan failure but, no direct correlation of vaccination with MIS has been established. Similarly, hemophagocytic lymphohistiocytosis (HLH) also noted to cause multiorgan failure in some individuals following full vaccination. Furthermore, severe complications were recorded in elderly patients (+40 years of age), indicates that older age individuals are higher risk by COVID-19 and post vaccination, but available literature is not sufficient to comply with any conclusive statements on relationship between vaccination and multiorgan failure.
Collapse
Affiliation(s)
- Ali A. Rabaan
- Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran 31311, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
- Department of Public Health and Nutrition, The University of Haripur, Haripur 22610, Pakistan
| | - Muhammed A. Bakhrebah
- Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia;
| | - Abbas Al Mutair
- Research Center, Almoosa Specialist Hospital, Al-Ahsa 36342, Saudi Arabia;
- College of Nursing, Princess Norah Bint Abdulrahman University, Riyadh 11564, Saudi Arabia
- School of Nursing, Wollongong University, Wollongong, NSW 2522, Australia
- Nursing Department, Prince Sultan Military College of Health Sciences, Dhahran 33048, Saudi Arabia
| | - Saad Alhumaid
- Administration of Pharmaceutical Care, Al-Ahsa Health Cluster, Ministry of Health, Al-Ahsa 31982, Saudi Arabia;
| | - Jumana M. Al-Jishi
- Internal Medicine Department, Qatif Central Hospital, Qatif 32654, Saudi Arabia;
| | - Jehad AlSihati
- Internal Medicine Department, Gastroenterology Section, King Fahad Specialist Hospital, Dammam 31311, Saudi Arabia;
| | - Hawra Albayat
- Infectious Disease Department, King Saud Medical City, Riyadh 7790, Saudi Arabia; (H.A.); (A.A.)
| | - Ahmed Alsheheri
- Infectious Disease Department, King Saud Medical City, Riyadh 7790, Saudi Arabia; (H.A.); (A.A.)
| | - Mohammed Aljeldah
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, University of Hafr Al Batin, Hafr Al Batin 39524, Saudi Arabia;
| | - Mohammed Garout
- Department of Community Medicine and Health Care for Pilgrims, Faculty of Medicine, Umm Al-Qura University, Makkah 21955, Saudi Arabia;
| | - Wadha A. Alfouzan
- Department of Microbiology, Faculty of Medicine, Kuwait University, Safat 13110, Kuwait;
- Microbiology Unit, Department of Laboratories, Farwania Hospital, Farwania 85000, Kuwait
| | - Yousef N. Alhashem
- Department of Clinical Laboratory Sciences, Mohammed AlMana College of Health Sciences, Dammam 34222, Saudi Arabia;
| | - Salma AlBahrani
- Infectious Disease Unit, Specialty Internal Medicine, King Fahd Military Medical Complex, Dhahran 31932, Saudi Arabia;
| | - Saleh A. Alshamrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Najran University, Najran 61441, Saudi Arabia;
| | - Sultan Alotaibi
- Molecular Microbiology Department, King Fahad Medical City, Riyadh 11525, Saudi Arabia;
| | - Abdullah A. AlRamadhan
- Laboratory and Toxicology Department, Security Forces Specialized Comprehensive Clinics, Al-Ahsa 36441, Saudi Arabia;
| | - Hanadi N. Albasha
- Department of Infection Prevention and Control, Obeid Specialized Hospital, Riyadh 12627, Saudi Arabia;
| | - Khalid Hajissa
- Department of Medical Microbiology & Parasitology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia;
| | - Mohamad-Hani Temsah
- Pediatric Department, College of Medicine, King Saud University, Riyadh 11451, Saudi Arabia;
| |
Collapse
|
35
|
The Possible Role of Glucose-6-Phosphate Dehydrogenase in the SARS-CoV-2 Infection. Cells 2022; 11:cells11131982. [PMID: 35805067 PMCID: PMC9265820 DOI: 10.3390/cells11131982] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/18/2022] [Accepted: 06/17/2022] [Indexed: 12/15/2022] Open
Abstract
Glucose-6-phosphate dehydrogenase (G6PD) is the second rate-limiting enzyme of the pentose phosphate pathway. This enzyme is present in the cytoplasm of all mammalian cells, and its activity is essential for an adequate functioning of the antioxidant system and for the response of innate immunity. It is responsible for the production of nicotinamide adenine dinucleotide phosphate (NADPH), the first redox equivalent, in the pentose phosphate pathway. Viral infections such as SARS-CoV-2 may induce the Warburg effect with an increase in anaerobic glycolysis and production of lactate. This condition ensures the success of viral replication and production of the virion. Therefore, the activity of G6PD may be increased in COVID-19 patients raising the level of the NADPH, which is needed for the enzymatic and non-enzymatic antioxidant systems that counteract the oxidative stress caused by the cytokine storm. G6PD deficiency affects approximately 350–400 million people worldwide; therefore, it is one of the most prevalent diseases related to enzymatic deficiency worldwide. In G6PD-deficient patients exposed to SARS-CoV-2, the amount of NADPH is reduced, increasing the susceptibility for viral infection. There is loss of the redox homeostasis in them, resulting in severe pneumonia and fatal outcomes.
Collapse
|
36
|
Dobrijevic Z, Robajac D, Gligorijevic N, Šunderic M, Penezic A, Miljuš G, Nedic O. The association of ACE1, ACE2, TMPRSS2, IFITM3 and VDR polymorphisms with COVID-19 severity: A systematic review and meta-analysis. EXCLI JOURNAL 2022; 21:818-839. [PMID: 35949487 PMCID: PMC9360474 DOI: 10.17179/excli2022-4976] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/14/2022] [Indexed: 11/14/2022]
Abstract
Genes involved in the regulation of viral recognition and its entry into a host cell have been identified as candidates for genetic association studies on COVID-19 severity. Published findings on the effects of polymorphisms within ACE1, ACE2, TMPRSS2, IFITM3 and VDR genes remained inconclusive, so we conducted a systematic review and meta-analysis in order to elucidate their potential involvement in the genetic basis underlying the severity of COVID-19 and/or an outcome of SARS-CoV-2 infection. Identification of potentially eligible studies was based on PubMed, Scopus and Web of Science database search. Relevant studies (n=29) with a total number of 8247 SARS-CoV-2-positive participants were included in qualitative synthesis, while results of 21 studies involving 5939 were pooled in meta-analysis. Minor allele I of rs1799752 located within ACE1 was identified as a protective variant against severe COVID-19, while its effect on mortality rate was opposite. Similarly, minor allele A of ACE2 polymorphism, rs2285666, was found to associate with a decreased risk of severe COVID-19 (P = 0.003, OR = 0.512, 95 % CI = 0.331-0.793). Statistical significance was also seen for the association between COVID-19 severity and rs12329760 located within TMPRSS2. Our results did not support the supposed association of rs12252 in IFITM3 and polymorphisms within VDR with disease severity. We conclude that genetic variants within ACE1, ACE2 and TMPRSS2 may be potential biomarkers of COVID-19 severity, which needs to be further confirmed in a larger set of studies.
Collapse
Affiliation(s)
- Zorana Dobrijevic
- University of Belgrade - Institute for the Application of Nuclear Energy (INEP), Belgrade, Serbia
| | - Dragana Robajac
- University of Belgrade - Institute for the Application of Nuclear Energy (INEP), Belgrade, Serbia
| | - Nikola Gligorijevic
- University of Belgrade - Institute for the Application of Nuclear Energy (INEP), Belgrade, Serbia
| | - Miloš Šunderic
- University of Belgrade - Institute for the Application of Nuclear Energy (INEP), Belgrade, Serbia
| | - Ana Penezic
- University of Belgrade - Institute for the Application of Nuclear Energy (INEP), Belgrade, Serbia
| | - Goran Miljuš
- University of Belgrade - Institute for the Application of Nuclear Energy (INEP), Belgrade, Serbia
| | - Olgica Nedic
- University of Belgrade - Institute for the Application of Nuclear Energy (INEP), Belgrade, Serbia
| |
Collapse
|
37
|
Van Goethem N, Vandromme M, Van Oyen H, Haarhuis F, Brondeel R, Catteau L, André E, Cuypers L, Blot K, Serrien B. Severity of infection with the SARS-CoV-2 B.1.1.7 lineage among hospitalized COVID-19 patients in Belgium. PLoS One 2022; 17:e0269138. [PMID: 35657787 PMCID: PMC9165825 DOI: 10.1371/journal.pone.0269138] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 05/13/2022] [Indexed: 12/17/2022] Open
Abstract
INTRODUCTION The pathogenesis of COVID-19 depends on the interplay between host characteristics, viral characteristics and contextual factors. Here, we compare COVID-19 disease severity between hospitalized patients in Belgium infected with the SARS-CoV-2 variant B.1.1.7 and those infected with previously circulating strains. METHODS The study is conducted within a causal framework to study the severity of SARS-CoV-2 variants by merging surveillance registries in Belgium. Infection with SARS-CoV-2 B.1.1.7 ('exposed') was compared to infection with previously circulating strains ('unexposed') in terms of the manifestation of severe COVID-19, intensive care unit (ICU) admission, or in-hospital mortality. The exposed and unexposed group were matched based on the hospital and the mean ICU occupancy rate during the patient's hospital stay. Other variables identified as confounders in a Directed Acyclic Graph (DAG) were adjusted for using regression analysis. Sensitivity analyses were performed to assess the influence of selection bias, vaccination rollout, and unmeasured confounding. RESULTS We observed no difference between the exposed and unexposed group in severe COVID-19 disease or in-hospital mortality (RR = 1.15, 95% CI [0.93-1.38] and RR = 0.92, 95% CI [0.62-1.23], respectively). The estimated standardized risk to be admitted in ICU was significantly higher (RR = 1.36, 95% CI [1.03-1.68]) when infected with the B.1.1.7 variant. An age-stratified analysis showed that among the younger age group (≤65 years), the SARS-CoV-2 variant B.1.1.7 was significantly associated with both severe COVID-19 progression and ICU admission. CONCLUSION This matched observational cohort study did not find an overall increased risk of severe COVID-19 or death associated with B.1.1.7 infection among patients already hospitalized. There was a significant increased risk to be transferred to ICU when infected with the B.1.1.7 variant, especially among the younger age group. However, potential selection biases advocate for more systematic sequencing of samples from hospitalized COVID-19 patients.
Collapse
Affiliation(s)
- Nina Van Goethem
- Scientific Directorate of Epidemiology and Public Health, Sciensano, Brussels, Belgium
- Department of Epidemiology and Biostatistics, Institut de recherche expérimentale et clinique, Faculty of Public Health, Université catholique de Louvain, Woluwe-Saint-Lambert, Belgium
| | - Mathil Vandromme
- Scientific Directorate of Epidemiology and Public Health, Sciensano, Brussels, Belgium
| | - Herman Van Oyen
- Scientific Directorate of Epidemiology and Public Health, Sciensano, Brussels, Belgium
| | - Freek Haarhuis
- Scientific Directorate of Epidemiology and Public Health, Sciensano, Brussels, Belgium
| | - Ruben Brondeel
- Scientific Directorate of Epidemiology and Public Health, Sciensano, Brussels, Belgium
| | - Lucy Catteau
- Scientific Directorate of Epidemiology and Public Health, Sciensano, Brussels, Belgium
| | - Emmanuel André
- University Hospitals Leuven, Department of Laboratory Medicine, National Reference Centre for Respiratory Pathogens, Leuven, Belgium
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Laboratory Clinical Bacteriology and Mycology, Rega Institute for Medical Research, Leuven, Belgium
| | - Lize Cuypers
- University Hospitals Leuven, Department of Laboratory Medicine, National Reference Centre for Respiratory Pathogens, Leuven, Belgium
| | | | | | - Koen Blot
- Scientific Directorate of Epidemiology and Public Health, Sciensano, Brussels, Belgium
| | - Ben Serrien
- Scientific Directorate of Epidemiology and Public Health, Sciensano, Brussels, Belgium
| |
Collapse
|
38
|
Castelo-Branco L, Tsourti Z, Gennatas S, Rogado J, Sekacheva M, Viñal D, Lee R, Croitoru A, Vitorino M, Khallaf S, Šušnjar S, Soewoto W, Cardeña A, Djerouni M, Rossi M, Alonso-Gordoa T, Ngelangel C, Whisenant JG, Choueiri TK, Dimopoulou G, Pradervand S, Arnold D, Harrington K, Michielin O, Dafni U, Pentheroudakis G, Peters S, Romano E. COVID-19 in patients with cancer: first report of the ESMO international, registry-based, cohort study (ESMO-CoCARE). ESMO Open 2022; 7:100499. [PMID: 35644101 PMCID: PMC9080222 DOI: 10.1016/j.esmoop.2022.100499] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/22/2022] [Accepted: 04/23/2022] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND ESMO COVID-19 and CAncer REgistry (ESMO-CoCARE) is an international collaborative registry-based, cohort study gathering real-world data from Europe, Asia/Oceania and Africa on the natural history, management and outcomes of patients with cancer infected with severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). PATIENTS AND METHODS ESMO-CoCARE captures information on patients with solid/haematological malignancies, diagnosed with coronavirus disease 2019 (COVID-19). Data collected since June 2020 include demographics, comorbidities, laboratory measurements, cancer characteristics, COVID-19 clinical features, management and outcome. Parameters influencing COVID-19 severity/recovery were investigated as well as factors associated with overall survival (OS) upon SARS-CoV-2 infection. RESULTS This analysis includes 1626 patients from 20 countries (87% from 24 European, 7% from 5 North African, 6% from 8 Asian/Oceanian centres), with COVID-19 diagnosis from January 2020 to May 2021. Median age was 64 years, with 52% of female, 57% of cancer stage III/IV and 65% receiving active cancer treatment. Nearly 64% patients required hospitalization due to COVID-19 diagnosis, with 11% receiving intensive care. In multivariable analysis, male sex, older age, Eastern Cooperative Oncology Group (ECOG) performance status ≥2, body mass index (BMI) <25 kg/m2, presence of comorbidities, symptomatic disease, as well as haematological malignancies, active/progressive cancer, neutrophil-to-lymphocyte ratio (NLR) ≥6 and OnCovid Inflammatory Score ≤40 were associated with COVID-19 severity (i.e. severe/moderate disease requiring hospitalization). About 98% of patients with mild COVID-19 recovered, as opposed to 71% with severe/moderate disease. Advanced cancer stage was an additional adverse prognostic factor for recovery. At data cut-off, and with median follow-up of 3 months, the COVID-19-related death rate was 24.5% (297/1212), with 380 deaths recorded in total. Almost all factors associated with COVID-19 severity, except for BMI and NLR, were also predictive of inferior OS, along with smoking and non-Asian ethnicity. CONCLUSIONS Selected patient and cancer characteristics related to sex, ethnicity, poor fitness, comorbidities, inflammation and active malignancy predict for severe/moderate disease and adverse outcomes from COVID-19 in patients with cancer.
Collapse
Affiliation(s)
- L Castelo-Branco
- Scientific and Medical Division, ESMO (European Society for Medical Oncology), Lugano, Switzerland; NOVA National School of Public Health, NOVA University, Lisbon, Portugal
| | - Z Tsourti
- Frontier Science Foundation-Hellas, Athens, Greece
| | - S Gennatas
- Medical Oncology Department, The Royal Marsden Hospital - NHS Foundation Trust, London, UK
| | - J Rogado
- Medical Oncology Department, Hospital Universitario Infanta Leonor, Madrid, Spain
| | - M Sekacheva
- World-Class Research Center 'Digital Biodesign and Personalized Healthcare', Sechenov First Moscow State Medical University, Moscow, Russia
| | - D Viñal
- Medical Oncology Department, Hospital Universitario La Paz, Madrid, Spain
| | - R Lee
- Medical Oncology Department, The University of Manchester, Manchester, UK; The Christie NHS Foundation Trust, Manchester, UK
| | - A Croitoru
- Medical Oncology Department, Fundeni Clinical Institute, Bucharest, Romania
| | - M Vitorino
- Servico Oncologia, Hospital Prof. Dr Fernando Fonseca EPE (Hospital Amadora/Sintra), Amadora, Portugal
| | - S Khallaf
- Medical Oncology Department, South Egypt Cancer Institute (SECI), Assiut University, Assiut, Egypt
| | - S Šušnjar
- Department of Medical Oncology, Institute for Oncology and Radiology of Serbia, Belgrade, Serbia
| | - W Soewoto
- Department of Surgery, Oncology Division, Sebelas Maret University, Surakarta, Indonesia
| | - A Cardeña
- Medical Oncology Department, Hospital Universitario Fundación Alcorcón, Madrid, Spain
| | - M Djerouni
- Oncology Department, Dr Saadane Hospital, Biskra, Algeria
| | - M Rossi
- Oncology Deparment, ASO 'SS. Antonio, Biagio e Cesare Arrigo', Alessandria, Italy
| | - T Alonso-Gordoa
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - C Ngelangel
- Asian Cancer Institute - Asian Hospital and Medical Center, Metro Manila, the Philippines
| | - J G Whisenant
- Vanderbilt University Medical Center, Nashville, USA
| | - T K Choueiri
- The Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, USA; Harvard Medical School, Boston, USA
| | - G Dimopoulou
- Frontier Science Foundation-Hellas, Athens, Greece
| | - S Pradervand
- Oncology Department, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - D Arnold
- Oncology, Haematology, Palliative Care Department, Asklepios Klinik Altona - Asklepios Kliniken, Hamburg, Germany
| | - K Harrington
- Division of Radiotherapy and Imaging, The Royal Marsden/The Institute of Cancer Research NIHR Biomedical Research Centre, London, UK
| | - O Michielin
- Oncology Department, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - U Dafni
- Laboratory of Biostatistics, School of Health Sciences, National and Kapodistrian University of Athens, Athens Frontier Science Foundation-Hellas, Athens, Greece
| | - G Pentheroudakis
- Scientific and Medical Division, ESMO (European Society for Medical Oncology), Lugano, Switzerland
| | - S Peters
- Department, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - E Romano
- Center for Cancer Immunotherapy, Department of Oncology, PSL Research University, Institut Curie, Paris, France.
| |
Collapse
|
39
|
HLA Allele Frequencies and Association with Severity of COVID-19 Infection in Northern Italian Patients. Cells 2022; 11:cells11111792. [PMID: 35681490 PMCID: PMC9179900 DOI: 10.3390/cells11111792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 12/03/2022] Open
Abstract
HLA allelic distribution was analysed in a cohort of 96 Northern Italian subjects (53M/43F) (mean age 59.9 ± 13.3 years) from Lombardy who developed COVID-19 during the first two pandemic waves to investigate possible correlations between HLA molecules and disease severity. An important role of HLA- B and HLA-C loci in modulating the clinical severity of COVID-19 disease was identified. In particular, the HLA-B07 supertype was observed to be associated with a significant risk for severe disease; conversely, the HLA-B27 supertype and C*12:02 allele played a protective role as they were associated with milder disease. These associations were confirmed after applying a multinomial regression analysis to adjust the correlation for age, gender and comorbidities with COVID-19 severity. Though the power of results is limited by the small sample size, data herein contribute to shedding light on the role played by genetic background in COVID-19 infection.
Collapse
|
40
|
Liu P, Fang M, Luo Y, Zheng F, Jin Y, Cheng F, Zhu H, Jin X. Rare Variants in Inborn Errors of Immunity Genes Associated With Covid-19 Severity. Front Cell Infect Microbiol 2022; 12:888582. [PMID: 35694544 PMCID: PMC9184678 DOI: 10.3389/fcimb.2022.888582] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 04/21/2022] [Indexed: 01/08/2023] Open
Abstract
Host genetic factors have been shown to play an important role in SARS-CoV-2 infection and the course of Covid-19 disease. The genetic contributions of common variants influencing Covid-19 susceptibility and severity have been extensively studied in diverse populations. However, the studies of rare genetic defects arising from inborn errors of immunity (IEI) are relatively few, especially in the Chinese population. To fill this gap, we used a deeply sequenced dataset of nearly 500 patients, all of Chinese descent, to investigate putative functional rare variants. Specifically, we annotated rare variants in our call set and selected likely deleterious missense (LDM) and high-confidence predicted loss-of-function (HC-pLoF) variants. Further, we analyzed LDM and HC-pLoF variants between non-severe and severe Covid-19 patients by (a) performing gene- and pathway-level association analyses, (b) testing the number of mutations in previously reported genes mapped from LDM and HC-pLoF variants, and (c) uncovering candidate genes via protein-protein interaction (PPI) network analysis of Covid-19-related genes and genes defined from LDM and HC-pLoF variants. From our analyses, we found that (a) pathways Tuberculosis (hsa:05152), Primary Immunodeficiency (hsa:05340), and Influenza A (hsa:05164) showed significant enrichment in severe patients compared to the non-severe ones, (b) HC-pLoF mutations were enriched in Covid-19-related genes in severe patients, and (c) several candidate genes, such as IL12RB1, TBK1, TLR3, and IFNGR2, are uncovered by PPI network analysis and worth further investigation. These regions generally play an essential role in regulating antiviral innate immunity responses to foreign pathogens and in responding to many inflammatory diseases. We believe that our identified candidate genes/pathways can be potentially used as Covid-19 diagnostic markers and help distinguish patients at higher risk.
Collapse
Affiliation(s)
- Panhong Liu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- Beijing Genomeics Institute At Shenzhen, BGI-Shenzhen, Shenzhen, China
| | - Mingyan Fang
- Beijing Genomeics Institute At Shenzhen, BGI-Shenzhen, Shenzhen, China
- Beijing Genomeics Institute In Singapore, BGI-Singapore, Singapore, Singapore
| | - Yuxue Luo
- Beijing Genomeics Institute At Shenzhen, BGI-Shenzhen, Shenzhen, China
| | - Fang Zheng
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Jin
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fanjun Cheng
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huanhuan Zhu
- Beijing Genomeics Institute At Shenzhen, BGI-Shenzhen, Shenzhen, China
- *Correspondence: Xin Jin, ; Huanhuan Zhu,
| | - Xin Jin
- Beijing Genomeics Institute At Shenzhen, BGI-Shenzhen, Shenzhen, China
- Beijing Genomeics Institute In Singapore, BGI-Singapore, Singapore, Singapore
- School of Medicine, South China University of Technology, Guangzhou, China
- *Correspondence: Xin Jin, ; Huanhuan Zhu,
| |
Collapse
|
41
|
Pastana LF, Silva TA, Gellen LPA, Vieira GM, de Assunção LA, Leitão LPC, da Silva NM, Coelho RDCC, de Alcântara AL, Vinagre LWMS, Rodrigues JCG, Borges Leal DFDV, Fernandes MR, de Souza SJ, Kroll JE, Ribeiro-dos-Santos AM, Burbano RMR, Guerreiro JF, de Assumpção PP, Ribeiro-dos-Santos ÂC, dos Santos SEB, dos Santos NPC. The Genomic Profile Associated with Risk of Severe Forms of COVID-19 in Amazonian Native American Populations. J Pers Med 2022; 12:jpm12040554. [PMID: 35455670 PMCID: PMC9027999 DOI: 10.3390/jpm12040554] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/11/2022] [Accepted: 02/15/2022] [Indexed: 12/12/2022] Open
Abstract
Genetic factors associated with COVID-19 disease outcomes are poorly understood. This study aimed to associate genetic variants in the SLC6A20, LZTFL1, CCR9, FYCO1, CXCR6, XCR1, and ABO genes with the risk of severe forms of COVID-19 in Amazonian Native Americans, and to compare the frequencies with continental populations. The study population was composed of 64 Amerindians from the Amazon region of northern Brazil. The difference in frequencies between the populations was analyzed using Fisher’s exact test, and the results were significant when p ≤ 0.05. We investigated 64 polymorphisms in 7 genes; we studied 47 genetic variants that were new or had impact predictions of high, moderate, or modifier. We identified 15 polymorphisms with moderate impact prediction in 4 genes (ABO, CXCR6, FYCO1, and SLC6A20). Among the variants analyzed, 18 showed significant differences in allele frequency in the NAM population when compared to others. We reported two new genetic variants with modifier impact in the Amazonian population that could be studied to validate the possible associations with COVID-19 outcomes. The genomic profile of Amazonian Native Americans may be associated with protection from severe forms of COVID-19. This work provides genomic data that may help forthcoming studies to improve COVID-19 outcomes.
Collapse
Affiliation(s)
- Lucas Favacho Pastana
- Laboratório do Núcleo de Pesquisa em Oncologia, Universidade Federal do Pará, Belém 66073-000, Brazil; (L.F.P.); (T.A.S.); (L.P.A.G.); (G.M.V.); (L.A.d.A.); (L.P.C.L.); (N.M.d.S.); (R.d.C.C.C.); (A.L.d.A.); (L.W.M.S.V.); (J.C.G.R.); (D.F.d.V.B.L.); (M.R.F.); (R.M.R.B.); (P.P.d.A.); (S.E.B.d.S.)
| | - Thays Amâncio Silva
- Laboratório do Núcleo de Pesquisa em Oncologia, Universidade Federal do Pará, Belém 66073-000, Brazil; (L.F.P.); (T.A.S.); (L.P.A.G.); (G.M.V.); (L.A.d.A.); (L.P.C.L.); (N.M.d.S.); (R.d.C.C.C.); (A.L.d.A.); (L.W.M.S.V.); (J.C.G.R.); (D.F.d.V.B.L.); (M.R.F.); (R.M.R.B.); (P.P.d.A.); (S.E.B.d.S.)
| | - Laura Patrícia Albarello Gellen
- Laboratório do Núcleo de Pesquisa em Oncologia, Universidade Federal do Pará, Belém 66073-000, Brazil; (L.F.P.); (T.A.S.); (L.P.A.G.); (G.M.V.); (L.A.d.A.); (L.P.C.L.); (N.M.d.S.); (R.d.C.C.C.); (A.L.d.A.); (L.W.M.S.V.); (J.C.G.R.); (D.F.d.V.B.L.); (M.R.F.); (R.M.R.B.); (P.P.d.A.); (S.E.B.d.S.)
| | - Giovana Miranda Vieira
- Laboratório do Núcleo de Pesquisa em Oncologia, Universidade Federal do Pará, Belém 66073-000, Brazil; (L.F.P.); (T.A.S.); (L.P.A.G.); (G.M.V.); (L.A.d.A.); (L.P.C.L.); (N.M.d.S.); (R.d.C.C.C.); (A.L.d.A.); (L.W.M.S.V.); (J.C.G.R.); (D.F.d.V.B.L.); (M.R.F.); (R.M.R.B.); (P.P.d.A.); (S.E.B.d.S.)
| | - Letícia Almeida de Assunção
- Laboratório do Núcleo de Pesquisa em Oncologia, Universidade Federal do Pará, Belém 66073-000, Brazil; (L.F.P.); (T.A.S.); (L.P.A.G.); (G.M.V.); (L.A.d.A.); (L.P.C.L.); (N.M.d.S.); (R.d.C.C.C.); (A.L.d.A.); (L.W.M.S.V.); (J.C.G.R.); (D.F.d.V.B.L.); (M.R.F.); (R.M.R.B.); (P.P.d.A.); (S.E.B.d.S.)
| | - Luciana Pereira Colares Leitão
- Laboratório do Núcleo de Pesquisa em Oncologia, Universidade Federal do Pará, Belém 66073-000, Brazil; (L.F.P.); (T.A.S.); (L.P.A.G.); (G.M.V.); (L.A.d.A.); (L.P.C.L.); (N.M.d.S.); (R.d.C.C.C.); (A.L.d.A.); (L.W.M.S.V.); (J.C.G.R.); (D.F.d.V.B.L.); (M.R.F.); (R.M.R.B.); (P.P.d.A.); (S.E.B.d.S.)
| | - Natasha Monte da Silva
- Laboratório do Núcleo de Pesquisa em Oncologia, Universidade Federal do Pará, Belém 66073-000, Brazil; (L.F.P.); (T.A.S.); (L.P.A.G.); (G.M.V.); (L.A.d.A.); (L.P.C.L.); (N.M.d.S.); (R.d.C.C.C.); (A.L.d.A.); (L.W.M.S.V.); (J.C.G.R.); (D.F.d.V.B.L.); (M.R.F.); (R.M.R.B.); (P.P.d.A.); (S.E.B.d.S.)
| | - Rita de Cássia Calderaro Coelho
- Laboratório do Núcleo de Pesquisa em Oncologia, Universidade Federal do Pará, Belém 66073-000, Brazil; (L.F.P.); (T.A.S.); (L.P.A.G.); (G.M.V.); (L.A.d.A.); (L.P.C.L.); (N.M.d.S.); (R.d.C.C.C.); (A.L.d.A.); (L.W.M.S.V.); (J.C.G.R.); (D.F.d.V.B.L.); (M.R.F.); (R.M.R.B.); (P.P.d.A.); (S.E.B.d.S.)
| | - Angélica Leite de Alcântara
- Laboratório do Núcleo de Pesquisa em Oncologia, Universidade Federal do Pará, Belém 66073-000, Brazil; (L.F.P.); (T.A.S.); (L.P.A.G.); (G.M.V.); (L.A.d.A.); (L.P.C.L.); (N.M.d.S.); (R.d.C.C.C.); (A.L.d.A.); (L.W.M.S.V.); (J.C.G.R.); (D.F.d.V.B.L.); (M.R.F.); (R.M.R.B.); (P.P.d.A.); (S.E.B.d.S.)
| | - Lui Wallacy Morikawa Souza Vinagre
- Laboratório do Núcleo de Pesquisa em Oncologia, Universidade Federal do Pará, Belém 66073-000, Brazil; (L.F.P.); (T.A.S.); (L.P.A.G.); (G.M.V.); (L.A.d.A.); (L.P.C.L.); (N.M.d.S.); (R.d.C.C.C.); (A.L.d.A.); (L.W.M.S.V.); (J.C.G.R.); (D.F.d.V.B.L.); (M.R.F.); (R.M.R.B.); (P.P.d.A.); (S.E.B.d.S.)
| | - Juliana Carla Gomes Rodrigues
- Laboratório do Núcleo de Pesquisa em Oncologia, Universidade Federal do Pará, Belém 66073-000, Brazil; (L.F.P.); (T.A.S.); (L.P.A.G.); (G.M.V.); (L.A.d.A.); (L.P.C.L.); (N.M.d.S.); (R.d.C.C.C.); (A.L.d.A.); (L.W.M.S.V.); (J.C.G.R.); (D.F.d.V.B.L.); (M.R.F.); (R.M.R.B.); (P.P.d.A.); (S.E.B.d.S.)
| | - Diana Feio da Veiga Borges Leal
- Laboratório do Núcleo de Pesquisa em Oncologia, Universidade Federal do Pará, Belém 66073-000, Brazil; (L.F.P.); (T.A.S.); (L.P.A.G.); (G.M.V.); (L.A.d.A.); (L.P.C.L.); (N.M.d.S.); (R.d.C.C.C.); (A.L.d.A.); (L.W.M.S.V.); (J.C.G.R.); (D.F.d.V.B.L.); (M.R.F.); (R.M.R.B.); (P.P.d.A.); (S.E.B.d.S.)
| | - Marianne Rodrigues Fernandes
- Laboratório do Núcleo de Pesquisa em Oncologia, Universidade Federal do Pará, Belém 66073-000, Brazil; (L.F.P.); (T.A.S.); (L.P.A.G.); (G.M.V.); (L.A.d.A.); (L.P.C.L.); (N.M.d.S.); (R.d.C.C.C.); (A.L.d.A.); (L.W.M.S.V.); (J.C.G.R.); (D.F.d.V.B.L.); (M.R.F.); (R.M.R.B.); (P.P.d.A.); (S.E.B.d.S.)
| | - Sandro José de Souza
- Instituto do Cérebro, Universidade Federal do Rio Grande do Norte, Natal 59076-550, Brazil; (S.J.d.S.); (J.E.K.)
- BioME, Universidade Federal do Rio Grande do Norte, Natal 59078-400, Brazil
- Institute of Systems Genetics, West China Hospital, University of Sichuan, Chengdu 610041, China
| | - José Eduardo Kroll
- Instituto do Cérebro, Universidade Federal do Rio Grande do Norte, Natal 59076-550, Brazil; (S.J.d.S.); (J.E.K.)
| | - André Mauricio Ribeiro-dos-Santos
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, Belém 66075-110, Brazil; (A.M.R.-d.-S.); (J.F.G.); (Â.C.R.-d.-S.)
| | - Rommel Mario Rodríguez Burbano
- Laboratório do Núcleo de Pesquisa em Oncologia, Universidade Federal do Pará, Belém 66073-000, Brazil; (L.F.P.); (T.A.S.); (L.P.A.G.); (G.M.V.); (L.A.d.A.); (L.P.C.L.); (N.M.d.S.); (R.d.C.C.C.); (A.L.d.A.); (L.W.M.S.V.); (J.C.G.R.); (D.F.d.V.B.L.); (M.R.F.); (R.M.R.B.); (P.P.d.A.); (S.E.B.d.S.)
| | - João Farias Guerreiro
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, Belém 66075-110, Brazil; (A.M.R.-d.-S.); (J.F.G.); (Â.C.R.-d.-S.)
| | - Paulo Pimentel de Assumpção
- Laboratório do Núcleo de Pesquisa em Oncologia, Universidade Federal do Pará, Belém 66073-000, Brazil; (L.F.P.); (T.A.S.); (L.P.A.G.); (G.M.V.); (L.A.d.A.); (L.P.C.L.); (N.M.d.S.); (R.d.C.C.C.); (A.L.d.A.); (L.W.M.S.V.); (J.C.G.R.); (D.F.d.V.B.L.); (M.R.F.); (R.M.R.B.); (P.P.d.A.); (S.E.B.d.S.)
| | - Ândrea Campos Ribeiro-dos-Santos
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, Belém 66075-110, Brazil; (A.M.R.-d.-S.); (J.F.G.); (Â.C.R.-d.-S.)
| | - Sidney Emanuel Batista dos Santos
- Laboratório do Núcleo de Pesquisa em Oncologia, Universidade Federal do Pará, Belém 66073-000, Brazil; (L.F.P.); (T.A.S.); (L.P.A.G.); (G.M.V.); (L.A.d.A.); (L.P.C.L.); (N.M.d.S.); (R.d.C.C.C.); (A.L.d.A.); (L.W.M.S.V.); (J.C.G.R.); (D.F.d.V.B.L.); (M.R.F.); (R.M.R.B.); (P.P.d.A.); (S.E.B.d.S.)
| | - Ney Pereira Carneiro dos Santos
- Laboratório do Núcleo de Pesquisa em Oncologia, Universidade Federal do Pará, Belém 66073-000, Brazil; (L.F.P.); (T.A.S.); (L.P.A.G.); (G.M.V.); (L.A.d.A.); (L.P.C.L.); (N.M.d.S.); (R.d.C.C.C.); (A.L.d.A.); (L.W.M.S.V.); (J.C.G.R.); (D.F.d.V.B.L.); (M.R.F.); (R.M.R.B.); (P.P.d.A.); (S.E.B.d.S.)
- Correspondence: ; Tel.: +55-(91)-98107-0850
| |
Collapse
|
42
|
Bellanti F, Lo Buglio A, Vendemiale G. Redox Homeostasis and Immune Alterations in Coronavirus Disease-19. BIOLOGY 2022; 11:159. [PMID: 35205026 PMCID: PMC8869285 DOI: 10.3390/biology11020159] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/14/2022] [Accepted: 01/18/2022] [Indexed: 02/05/2023]
Abstract
The global Coronavirus Disease 2019 (COVID-19) pandemic is characterized by a wide variety of clinical features, from no or moderate symptoms to severe illness. COVID-19 is caused by the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) that first affects the respiratory tract. Other than being limited to lungs, SARS-CoV-2 may lead to a multisystem disease that can even be durable (long COVID). The clinical spectrum of COVID-19 depends on variability in the immune regulation. Indeed, disease progression is consequent to failure in the immune regulation, characterized by an intensification of the pro-inflammatory response. Disturbance of systemic and organ-related redox balance may be a further mechanism underlying variability in COVID-19 severity. Other than being determinant for SARS-CoV-2 entry and fusion to the host cell, reactive species and redox signaling are deeply involved in the immune response. This review sums up the present knowledge on the role of redox balance in the regulation of susceptibility to SARS-CoV-2 infection and related immune response, debating the effectiveness of antioxidant compounds in the management of COVID-19.
Collapse
Affiliation(s)
- Francesco Bellanti
- Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto 1, 71122 Foggia, Italy; (A.L.B.); (G.V.)
| | | | | |
Collapse
|
43
|
COVID-19 Pandemic and Periodontal Practice: The Immunological, Clinical, and Economic Points of View. BIOMED RESEARCH INTERNATIONAL 2022; 2022:3918980. [PMID: 35047633 PMCID: PMC8763038 DOI: 10.1155/2022/3918980] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 12/24/2021] [Indexed: 11/17/2022]
Abstract
The recent global health problem, COVID-19, has had far-reaching impacts on lifestyles. Although many effective WHO-approved vaccines have been produced that have reduced the spread and severity of the disease, it appears to persist in humans for a long time and possibly forever as everyday it turns out to have new mutations. COVID-19 involves the lungs and other organs primarily through cytokine storms, which have been implicated in many other inflammatory disorders, including periodontal diseases. COVID-19 is in a close association with dental and periodontal practice from two respects: first, repeated mandatory lockdowns have reduced patient referrals to dentists and limited the dental and periodontal procedures to emergency treatments, whereas it is important to recognize the oral manifestations of COVID-19 as well as the influence of oral and periodontal disease on the severity of COVID-19. Second, dentistry is one of the high-risk professions in terms of close contact with unmasked individuals, necessitating redefining the principles of infection control. The pressures of the economic recession on patients as well as dentists add to the difficulty of resuming elective dental services. Therefore, this study is divided into two parts corresponding to what mentioned above: the first part examines the clinical and immunological associations between COVID-19 and periodontal and oral diseases, and the second part delineates the measures needed to control the disease transmission in dental clinics as well as the economic impact of the pandemic era on dental services.
Collapse
|
44
|
Fallerini C, Picchiotti N, Baldassarri M, Zguro K, Daga S, Fava F, Benetti E, Amitrano S, Bruttini M, Palmieri M, Croci S, Lista M, Beligni G, Valentino F, Meloni I, Tanfoni M, Minnai F, Colombo F, Cabri E, Fratelli M, Gabbi C, Mantovani S, Frullanti E, Gori M, Crawley FP, Butler-Laporte G, Richards B, Zeberg H, Lipcsey M, Hultström M, Ludwig KU, Schulte EC, Pairo-Castineira E, Baillie JK, Schmidt A, Frithiof R, Mari F, Renieri A, Furini S. Common, low-frequency, rare, and ultra-rare coding variants contribute to COVID-19 severity. Hum Genet 2022; 141:147-173. [PMID: 34889978 PMCID: PMC8661833 DOI: 10.1007/s00439-021-02397-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 10/26/2021] [Indexed: 12/13/2022]
Abstract
The combined impact of common and rare exonic variants in COVID-19 host genetics is currently insufficiently understood. Here, common and rare variants from whole-exome sequencing data of about 4000 SARS-CoV-2-positive individuals were used to define an interpretable machine-learning model for predicting COVID-19 severity. First, variants were converted into separate sets of Boolean features, depending on the absence or the presence of variants in each gene. An ensemble of LASSO logistic regression models was used to identify the most informative Boolean features with respect to the genetic bases of severity. The Boolean features selected by these logistic models were combined into an Integrated PolyGenic Score that offers a synthetic and interpretable index for describing the contribution of host genetics in COVID-19 severity, as demonstrated through testing in several independent cohorts. Selected features belong to ultra-rare, rare, low-frequency, and common variants, including those in linkage disequilibrium with known GWAS loci. Noteworthily, around one quarter of the selected genes are sex-specific. Pathway analysis of the selected genes associated with COVID-19 severity reflected the multi-organ nature of the disease. The proposed model might provide useful information for developing diagnostics and therapeutics, while also being able to guide bedside disease management.
Collapse
Affiliation(s)
- Chiara Fallerini
- grid.9024.f0000 0004 1757 4641Department of Medical Biotechnologies, Med Biotech Hub and Competence Center, University of Siena, Siena, Italy ,grid.9024.f0000 0004 1757 4641Medical Genetics, University of Siena, Siena, Italy
| | - Nicola Picchiotti
- grid.9024.f0000 0004 1757 4641University of Siena, DIISM-SAILAB, Siena, Italy ,grid.8982.b0000 0004 1762 5736Department of Mathematics, University of Pavia, Pavia, Italy
| | - Margherita Baldassarri
- grid.9024.f0000 0004 1757 4641Department of Medical Biotechnologies, Med Biotech Hub and Competence Center, University of Siena, Siena, Italy ,grid.9024.f0000 0004 1757 4641Medical Genetics, University of Siena, Siena, Italy
| | - Kristina Zguro
- grid.9024.f0000 0004 1757 4641Department of Medical Biotechnologies, Med Biotech Hub and Competence Center, University of Siena, Siena, Italy
| | - Sergio Daga
- grid.9024.f0000 0004 1757 4641Department of Medical Biotechnologies, Med Biotech Hub and Competence Center, University of Siena, Siena, Italy ,grid.9024.f0000 0004 1757 4641Medical Genetics, University of Siena, Siena, Italy
| | - Francesca Fava
- grid.9024.f0000 0004 1757 4641Department of Medical Biotechnologies, Med Biotech Hub and Competence Center, University of Siena, Siena, Italy ,grid.9024.f0000 0004 1757 4641Medical Genetics, University of Siena, Siena, Italy ,grid.411477.00000 0004 1759 0844Genetica Medica, Azienda Ospedaliero-Universitaria Senese, Siena, Italy
| | - Elisa Benetti
- grid.9024.f0000 0004 1757 4641Department of Medical Biotechnologies, Med Biotech Hub and Competence Center, University of Siena, Siena, Italy
| | - Sara Amitrano
- grid.411477.00000 0004 1759 0844Genetica Medica, Azienda Ospedaliero-Universitaria Senese, Siena, Italy
| | - Mirella Bruttini
- grid.9024.f0000 0004 1757 4641Department of Medical Biotechnologies, Med Biotech Hub and Competence Center, University of Siena, Siena, Italy ,grid.9024.f0000 0004 1757 4641Medical Genetics, University of Siena, Siena, Italy ,grid.411477.00000 0004 1759 0844Genetica Medica, Azienda Ospedaliero-Universitaria Senese, Siena, Italy
| | - Maria Palmieri
- grid.9024.f0000 0004 1757 4641Department of Medical Biotechnologies, Med Biotech Hub and Competence Center, University of Siena, Siena, Italy ,grid.9024.f0000 0004 1757 4641Medical Genetics, University of Siena, Siena, Italy
| | - Susanna Croci
- grid.9024.f0000 0004 1757 4641Department of Medical Biotechnologies, Med Biotech Hub and Competence Center, University of Siena, Siena, Italy ,grid.9024.f0000 0004 1757 4641Medical Genetics, University of Siena, Siena, Italy
| | - Mirjam Lista
- grid.9024.f0000 0004 1757 4641Department of Medical Biotechnologies, Med Biotech Hub and Competence Center, University of Siena, Siena, Italy ,grid.9024.f0000 0004 1757 4641Medical Genetics, University of Siena, Siena, Italy
| | - Giada Beligni
- grid.9024.f0000 0004 1757 4641Department of Medical Biotechnologies, Med Biotech Hub and Competence Center, University of Siena, Siena, Italy ,grid.9024.f0000 0004 1757 4641Medical Genetics, University of Siena, Siena, Italy
| | - Floriana Valentino
- grid.9024.f0000 0004 1757 4641Department of Medical Biotechnologies, Med Biotech Hub and Competence Center, University of Siena, Siena, Italy ,grid.9024.f0000 0004 1757 4641Medical Genetics, University of Siena, Siena, Italy
| | - Ilaria Meloni
- grid.9024.f0000 0004 1757 4641Department of Medical Biotechnologies, Med Biotech Hub and Competence Center, University of Siena, Siena, Italy ,grid.9024.f0000 0004 1757 4641Medical Genetics, University of Siena, Siena, Italy
| | - Marco Tanfoni
- grid.9024.f0000 0004 1757 4641University of Siena, DIISM-SAILAB, Siena, Italy
| | - Francesca Minnai
- grid.429135.80000 0004 1756 2536Istituto di Tecnologie Biomediche-Consiglio Nazionale delle Ricerche, Segrate, MI Italy
| | - Francesca Colombo
- grid.429135.80000 0004 1756 2536Istituto di Tecnologie Biomediche-Consiglio Nazionale delle Ricerche, Segrate, MI Italy
| | - Enrico Cabri
- grid.4527.40000000106678902Pharmacogenomics Unit, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Maddalena Fratelli
- grid.4527.40000000106678902Pharmacogenomics Unit, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Chiara Gabbi
- grid.4714.60000 0004 1937 0626Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - Stefania Mantovani
- grid.419425.f0000 0004 1760 3027Department of Medicine, Clinical Immunology and Infectious Diseases, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Elisa Frullanti
- grid.9024.f0000 0004 1757 4641Department of Medical Biotechnologies, Med Biotech Hub and Competence Center, University of Siena, Siena, Italy ,grid.9024.f0000 0004 1757 4641Medical Genetics, University of Siena, Siena, Italy
| | - Marco Gori
- grid.9024.f0000 0004 1757 4641University of Siena, DIISM-SAILAB, Siena, Italy ,grid.503321.60000 0001 0561 3840Models and Algorithms for Artificial Intelligence (MAASAI) Research Group, Université Côte d’Azur, Inria, CNRS, I3S, Biot, France
| | - Francis P. Crawley
- Good Clinical Practice Alliance-Europe (GCPA) and Strategic Initiative for Developing Capacity in Ethical Review (SIDCER), Leuven, Belgium
| | - Guillaume Butler-Laporte
- grid.14709.3b0000 0004 1936 8649Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, QC Canada
| | - Brent Richards
- grid.14709.3b0000 0004 1936 8649Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Department of Human Genetics, McGill University, Montreal, QC Canada ,grid.13097.3c0000 0001 2322 6764Department of Twin Research, King’s College London, London, UK
| | - Hugo Zeberg
- grid.4714.60000 0004 1937 0626Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Miklos Lipcsey
- grid.8993.b0000 0004 1936 9457Anaesthesiology and Intensive Care Medicine, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden ,grid.8993.b0000 0004 1936 9457Hedenstierna Laboratory, CIRRUS, Anaesthesiology and Intensive Care Medicine, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Michael Hultström
- grid.8993.b0000 0004 1936 9457Anaesthesiology and Intensive Care Medicine, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden ,grid.8993.b0000 0004 1936 9457Integrative Physiology, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Kerstin U. Ludwig
- grid.10388.320000 0001 2240 3300Institute of Human Genetics, School of Medicine and University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Eva C. Schulte
- grid.411095.80000 0004 0477 2585Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, 80336 Munich, Germany ,grid.5252.00000 0004 1936 973XDepartment of Psychiatry and Psychotherapy, University Hospital, LMU Munich, 80336 Munich, Germany ,grid.6936.a0000000123222966Institute of Virology, Technical University Munich/Helmholtz Zentrum München, Munich, Germany
| | - Erola Pairo-Castineira
- grid.4305.20000 0004 1936 7988MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Crewe Road, Edinburgh, EH4 2XU UK ,grid.4305.20000 0004 1936 7988Roslin Institute, University of Edinburgh, Easter Bush, Edinburgh, EH25 9RG UK
| | - John Kenneth Baillie
- grid.4305.20000 0004 1936 7988MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Crewe Road, Edinburgh, EH4 2XU UK ,grid.4305.20000 0004 1936 7988Roslin Institute, University of Edinburgh, Easter Bush, Edinburgh, EH25 9RG UK ,grid.418716.d0000 0001 0709 1919Intensive Care Unit, Royal Infirmary of Edinburgh, 54 Little France Drive, Edinburgh, H16 5SA UK
| | - Axel Schmidt
- grid.10388.320000 0001 2240 3300Institute of Human Genetics, School of Medicine and University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Robert Frithiof
- grid.8993.b0000 0004 1936 9457Anaesthesiology and Intensive Care Medicine, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | | | | | | | - Francesca Mari
- grid.9024.f0000 0004 1757 4641Department of Medical Biotechnologies, Med Biotech Hub and Competence Center, University of Siena, Siena, Italy ,grid.9024.f0000 0004 1757 4641Medical Genetics, University of Siena, Siena, Italy ,grid.411477.00000 0004 1759 0844Genetica Medica, Azienda Ospedaliero-Universitaria Senese, Siena, Italy
| | - Alessandra Renieri
- Department of Medical Biotechnologies, Med Biotech Hub and Competence Center, University of Siena, Siena, Italy. .,Medical Genetics, University of Siena, Siena, Italy. .,Genetica Medica, Azienda Ospedaliero-Universitaria Senese, Siena, Italy. .,Medical Genetics Unit, University of Siena, Policlinico Le Scotte, Viale Bracci, 2, 53100, Siena, Italy.
| | - Simone Furini
- grid.9024.f0000 0004 1757 4641Department of Medical Biotechnologies, Med Biotech Hub and Competence Center, University of Siena, Siena, Italy
| |
Collapse
|
45
|
Van Goethem N, Danwang C, Bossuyt N, Van Oyen H, Roosens NHC, Robert A. A systematic review and meta-analysis of host genetic factors associated with influenza severity. BMC Genomics 2021; 22:912. [PMID: 34930124 PMCID: PMC8686082 DOI: 10.1186/s12864-021-08240-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 12/07/2021] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The severity of influenza disease can range from mild symptoms to severe respiratory failure and can partly be explained by host genetic factors that predisposes the host to severe influenza. Here, we aimed to summarize the current state of evidence that host genetic variants play a role in the susceptibility to severe influenza infection by conducting a systematic review and performing a meta-analysis for all markers with at least three or more data entries. RESULTS A total of 34 primary human genetic association studies were identified that investigated a total of 20 different genes. The only significant pooled ORs were retrieved for the rs12252 polymorphism: an overall OR of 1.52 (95% CI [1.06-2.17]) for the rs12252-C allele compared to the rs12252-T allele. A stratified analysis by ethnicity revealed opposite effects in different populations. CONCLUSION With exception for the rs12252 polymorphism, we could not identify specific genetic polymorphisms to be associated with severe influenza infection in a pooled meta-analysis. This advocates for the use of large, hypothesis-free, genome-wide association studies that account for the polygenic nature and the interactions with other host, pathogen and environmental factors.
Collapse
Affiliation(s)
- Nina Van Goethem
- Scientific Directorate of Epidemiology and Public Health, Sciensano, J. Wytsmanstraat 14, 1050 Brussels, Belgium
- Department of Epidemiology and Biostatistics, Institut de Recherche Expérimentale et Clinique, Faculty of Public Health, Université Catholique de Louvain, Clos Chapelle-aux-champs 30, 1200 Brussels, Belgium
| | - Célestin Danwang
- Department of Epidemiology and Biostatistics, Institut de Recherche Expérimentale et Clinique, Faculty of Public Health, Université Catholique de Louvain, Clos Chapelle-aux-champs 30, 1200 Brussels, Belgium
| | - Nathalie Bossuyt
- Scientific Directorate of Epidemiology and Public Health, Sciensano, J. Wytsmanstraat 14, 1050 Brussels, Belgium
| | - Herman Van Oyen
- Scientific Directorate of Epidemiology and Public Health, Sciensano, J. Wytsmanstraat 14, 1050 Brussels, Belgium
- Department of Public Health and Primary Care, Ghent University, De Pintelaan 185, 9000 Ghent, Belgium
| | - Nancy H. C. Roosens
- Transversal Activities in Applied Genomics, Sciensano, J. Wytsmanstraat 14, 1050 Brussels, Belgium
| | - Annie Robert
- Department of Epidemiology and Biostatistics, Institut de Recherche Expérimentale et Clinique, Faculty of Public Health, Université Catholique de Louvain, Clos Chapelle-aux-champs 30, 1200 Brussels, Belgium
| |
Collapse
|
46
|
Martínez-Vega RA, Jing W, Ortega-Villa AM, Delgado-Cuellar OM, Hernández-Hernández VA, Jan-Gómez JC, Rincón-León HA, Constantino-Santiesteban P, García-Guerra MP, Cetina-Díaz JH, Pérez-Tirado JM, Gómez-Cruz O, Amaya-Larios IY, Ramos-Castañeda J, Jesús SD. Risk Prediction of Death in Inpatient Adults With COVID-19 from Mexico. RESEARCH SQUARE 2021:rs.3.rs-996535. [PMID: 34751267 PMCID: PMC8575141 DOI: 10.21203/rs.3.rs-996535/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Background There is substantial variation in COVID-19 lethality across countries. In addition, in countries with populations with extreme economic inequalities, such as Mexico, there are regional and local differences in risk factors for COVID-19 death. The goal of this study was to test the hypothesis that the risk of death in Mexican COVID-19 patients was associated with the time between symptom onset and hospitalization and/or with the healthcare site. Also, death prognostic models were developed. Methods The study included two COVID-19 inpatient cohorts, one prospective and one retrospective from Chiapas, Mexico. Demographic, clinical and laboratory variables were collected, and the diagnosis of SARS-CoV-2 infection was performed using RT-qPCR in samples collected seven days since symptom onset. The 30-day mortality, since symptom onset, was the outcome, and clinical variables at the first 48 hours of hospitalization were independent factors. Multivariate logistic regression analyses were conducted. Results Of the 392 patients included, 233 died (59.4%). The time between symptom onset and hospitalization, the healthcare site and sex were not related to the 30-day mortality. Three death prognostic models were developed (AUC between 0.726 and 0.807). Age, LDH, AST, and lymphocyte count were included in all models, OSI-WHO Classification (Non-invasive ventilation or high-flow oxygen, and mechanical ventilation with or without organ support/ECMO) and leukocyte count in two models, and diabetes and diarrhea in one model. Conclusion The population evaluated had underlying deteriorated health before COVID-19 compared with regional and country population. The factors that determine the COVID-19 mortality risk in a relatively healthy population are sex, age and comorbidities. However, as this study shows, when populations have underlying poor health, some of these factors lose their associations with mortality risk, and others become more important.
Collapse
Affiliation(s)
| | - Wang Jing
- Frederick National Laboratory for Cancer Research
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Van Goethem N, Serrien B, Vandromme M, Wyndham-Thomas C, Catteau L, Brondeel R, Klamer S, Meurisse M, Cuypers L, André E, Blot K, Van Oyen H. Conceptual causal framework to assess the effect of SARS-CoV-2 variants on COVID-19 disease severity among hospitalized patients. Arch Public Health 2021; 79:185. [PMID: 34696806 PMCID: PMC8543112 DOI: 10.1186/s13690-021-00709-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 10/02/2021] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND SARS-CoV-2 strains evolve continuously and accumulate mutations in their genomes over the course of the pandemic. The severity of a SARS-CoV-2 infection could partly depend on these viral genetic characteristics. Here, we present a general conceptual framework that allows to study the effect of SARS-CoV-2 variants on COVID-19 disease severity among hospitalized patients. METHODS A causal model is defined and visualized using a Directed Acyclic Graph (DAG), in which assumptions on the relationship between (confounding) variables are made explicit. Various DAGs are presented to explore specific study design options and the risk for selection bias. Next, the data infrastructure specific to the COVID-19 surveillance in Belgium is described, along with its strengths and weaknesses for the study of clinical impact of variants. DISCUSSION A well-established framework that provides a complete view on COVID-19 disease severity among hospitalized patients by combining information from different sources on host factors, viral factors, and healthcare-related factors, will enable to assess the clinical impact of emerging SARS-CoV-2 variants and answer questions that will be raised in the future. The framework shows the complexity related to causal research, the corresponding data requirements, and it underlines important limitations, such as unmeasured confounders or selection bias, inherent to repurposing existing routine COVID-19 data registries. TRIAL REGISTRATION Each individual research project within the current conceptual framework will be prospectively registered in Open Science Framework (OSF identifier: https://doi.org/10.17605/OSF.IO/UEF29 ). OSF project created on 18 May 2021.
Collapse
Affiliation(s)
- Nina Van Goethem
- Scientific Directorate of Epidemiology and public health, Sciensano, J. Wytsmanstraat 14, 1050, Brussels, Belgium.
- Department of Epidemiology and Biostatistics, Institut de recherche expérimentale et clinique, Faculty of Public Health, Université catholique de Louvain, Clos Chapelle-aux-champs 30, 1200, Woluwe-Saint-Lambert, Belgium.
| | - Ben Serrien
- Scientific Directorate of Epidemiology and public health, Sciensano, J. Wytsmanstraat 14, 1050, Brussels, Belgium
| | - Mathil Vandromme
- Scientific Directorate of Epidemiology and public health, Sciensano, J. Wytsmanstraat 14, 1050, Brussels, Belgium
| | - Chloé Wyndham-Thomas
- Scientific Directorate of Epidemiology and public health, Sciensano, J. Wytsmanstraat 14, 1050, Brussels, Belgium
| | - Lucy Catteau
- Scientific Directorate of Epidemiology and public health, Sciensano, J. Wytsmanstraat 14, 1050, Brussels, Belgium
| | - Ruben Brondeel
- Scientific Directorate of Epidemiology and public health, Sciensano, J. Wytsmanstraat 14, 1050, Brussels, Belgium
| | - Sofieke Klamer
- Scientific Directorate of Epidemiology and public health, Sciensano, J. Wytsmanstraat 14, 1050, Brussels, Belgium
| | - Marjan Meurisse
- Scientific Directorate of Epidemiology and public health, Sciensano, J. Wytsmanstraat 14, 1050, Brussels, Belgium
| | - Lize Cuypers
- Department of Laboratory Medicine, National Reference Centre for Respiratory Pathogens, University Hospitals Leuven, Herestraat 49, BE-3000, Leuven, Belgium
| | - Emmanuel André
- Department of Laboratory Medicine, National Reference Centre for Respiratory Pathogens, University Hospitals Leuven, Herestraat 49, BE-3000, Leuven, Belgium
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory Clinical Bacteriology and Mycology, Herestraat 49, box 1040, BE-3000, Leuven, Belgium
| | - Koen Blot
- Scientific Directorate of Epidemiology and public health, Sciensano, J. Wytsmanstraat 14, 1050, Brussels, Belgium
| | - Herman Van Oyen
- Scientific Directorate of Epidemiology and public health, Sciensano, J. Wytsmanstraat 14, 1050, Brussels, Belgium
| |
Collapse
|
48
|
Anyfantakis D, Mantadaki AE, Mastronikolis S, Spandidos DA, Symvoulakis EK. COVID-19 pandemic and reasons to prioritize the needs of the health care system to ensure its sustainability: A scoping review from January to October 2020 (Review). Exp Ther Med 2021; 22:1039. [PMID: 34373725 PMCID: PMC8343896 DOI: 10.3892/etm.2021.10471] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 06/29/2021] [Indexed: 12/23/2022] Open
Abstract
The worldwide spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) led the World Health Organization to characterize the pandemic as a public health emergency of international concern. National health care systems in countries during the initial surge of the pandemic were unable to handle the sanitarian crisis that had emerged. Thus, the prevention and control of future global health emergencies must be a priority. The present scoping review aimed to retrieve articles that summarize the current experience on issues related to historical knowledge, and epidemiology, clinical features and overall burden of SARS-CoV-2 on health care services. In summary, a comprehensive overview of the information that has been learnt during this period is presented in the current review. Furthermore, taking into account the global experience, the need for planning cohesive and functional health services before similar pandemic events occur in the future is highlighted. The next public health issue should be prevented rather than treated. In spite of the vaccination benefits, a number of sporadic cases of SARS-CoV-2infections will persist. Information collected remains relevant for appraising how similar threats can be faced in the future. Overall, collaborative health care plans need to be rethought to increase preparedness.
Collapse
Affiliation(s)
| | - Aikaterini E. Mantadaki
- Department of Social Medicine, Faculty of Medicine, University of Crete, 71500 Heraklion, Greece
| | - Stylianos Mastronikolis
- Clinic of Social and Family Medicine, Faculty of Medicine, University of Crete, 71500 Heraklion, Greece
| | - Demetrios A. Spandidos
- Laboratory of Clinical Virology, School of Medicine, University of Crete, 71500 Heraklion, Greece
| | - Emmanouil K. Symvoulakis
- Clinic of Social and Family Medicine, Faculty of Medicine, University of Crete, 71500 Heraklion, Greece
| |
Collapse
|
49
|
Colona VL, Vasiliou V, Watt J, Novelli G, Reichardt JKV. Update on human genetic susceptibility to COVID-19: susceptibility to virus and response. Hum Genomics 2021; 15:57. [PMID: 34429158 PMCID: PMC8384585 DOI: 10.1186/s40246-021-00356-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Vito Luigi Colona
- Department of Biomedicine and Prevention, "Tor Vergata" University of Rome, 00133, Rome, Italy
| | - Vasilis Vasiliou
- Department of Environmental Health Sciences, School of Public Health, Yale University, New Haven, USA
| | - Jessica Watt
- College of Public Health, Medical and Veterinary Sciences, James Cook University, Smithfield, QLD, Australia
| | - Giuseppe Novelli
- Department of Biomedicine and Prevention, "Tor Vergata" University of Rome, 00133, Rome, Italy
- IRCCS Neuromed, Pozzilli, IS, Italy
- Department of Pharmacology, School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Juergen K V Reichardt
- Australian Institute of Tropical Health and Medicine, James Cook University, Smithfield, QLD, 4878, Australia.
| |
Collapse
|
50
|
Kaidashev I, Shlykova O, Izmailova O, Torubara O, Yushchenko Y, Tyshkovska T, Kyslyi V, Belyaeva A, Maryniak D. Host gene variability and SARS-CoV-2 infection: A review article. Heliyon 2021; 7:e07863. [PMID: 34458641 PMCID: PMC8382593 DOI: 10.1016/j.heliyon.2021.e07863] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 06/15/2021] [Accepted: 08/19/2021] [Indexed: 02/07/2023] Open
Abstract
SARS-CoV-2 is a global threat that influenced healthcare systems around the world. This virus caused an infection in humans with different clinical signs and syndromes, severity, and mortality. The key components of the COVID-19 molecular pathogenesis are coronavirus entry and replication, antigen presentation, humoral and cellular immunity, cytokine storm, coronavirus immune evasion. The analysis of recent literature displayed possible molecular targets in the key components of the COVID-19 pathogenesis. Some of these targets might have gene polymorphisms that influenced the COVID-19 course. Unfortunately, several findings are still putative or extrapolated from SARS and MERS experimental investigations or clinical trials. We systematised original data about gene polymorphisms of possible molecular targets and associations with the COVID-19 course. Most data were obtained for angiotensin-converting enzymes 1 and 2, TMPRSS2 gene polymorphisms. Only a few results were found for gene polymorphisms of adhesion molecules, interferon system components, cytokines, and transcriptional factors, oxidative stress and metabolic molecules, as well as haemocoagulation. Understanding the host gene variability and its associations with COVID-19 can provide insights into the disease pathogenesis, individual susceptibility to SARS-CoV-2 infection, severity, complications, and mortality prognosis for the disease. Besides, these data might help in the identification of appropriate targets for intervention.
Collapse
Affiliation(s)
- I. Kaidashev
- Poltava State Medical University, Poltava, Ukraine
| | - O. Shlykova
- Poltava State Medical University, Poltava, Ukraine
| | - O. Izmailova
- Poltava State Medical University, Poltava, Ukraine
| | - O. Torubara
- Poltava State Medical University, Poltava, Ukraine
| | | | | | - V. Kyslyi
- Poltava State Medical University, Poltava, Ukraine
| | - A. Belyaeva
- Poltava State Medical University, Poltava, Ukraine
| | - D. Maryniak
- Poltava State Medical University, Poltava, Ukraine
| |
Collapse
|