1
|
Azcarate D, Olasagasti Arsuaga F, Granizo Rodriguez E, Arana-Arri E, España PP, Intxausti M, Sancho C, García de Vicuña Meléndez A, Ibarrondo O, M de Pancorbo M. Human-genetic variants associated with susceptibility to SARS-CoV-2 infection. Gene 2025; 953:149423. [PMID: 40120867 DOI: 10.1016/j.gene.2025.149423] [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: 11/19/2024] [Revised: 03/13/2025] [Accepted: 03/15/2025] [Indexed: 03/25/2025]
Abstract
SARS-CoV-2, the third major coronavirus of the 21st century, causing COVID-19 disease, profoundly impacts public health and workforces worldwide. Identifying individuals at heightened risk of SARS-CoV-2 infection is crucial for targeted interventions and preparedness. This study investigated 35 SNVs within viral infection-associated genes in SARS-CoV-2 patients and uninfected controls from the Basque Country (March 2020-July 2021). Its primary aim was to uncover genetic markers indicative of SARS-CoV-2 susceptibility and explore genetic predispositions to infection. Association analyses revealed previously unreported associations between SNVs and susceptibility. Haplotype analyses uncovered novel links between haplotypes and susceptibility, surpassing individual SNV associations. Descriptive modelling identified key susceptibility factors, with rs11246068-CC (IFITM3), rs5742933-GG (ORMDL1), rs35337543-CG (IFIH1), and GGGCT (rs2070788, rs2298659, rs17854725, rs12329760, rs3787950) variation in TMPRSS2 emerging as main infection-susceptibility indicators for a COVID-19 pandemic situation. These findings underscore the importance of integrated SNV and haplotype analyses in delineating susceptibility to SARS-CoV-2 and informing proactive prevention strategies. The genetic markers profiled in this study offer valuable insights for future pandemic preparedness.
Collapse
Affiliation(s)
- Daniel Azcarate
- BIOMICs Research Group (BIOMICS and Microfluidics cluster), Zoology and animal cellular biology department, Faculty of Science and Technology (UPV/EHU), 48940 Leioa, Biscay (Basque Country), Spain
| | - Felix Olasagasti Arsuaga
- BIOMICs Research Group (BIOMICS and Microfluidics cluster), Biochemistry and molecular biology department, Faculty of Pharmacy (UPV/EHU), 01006 Vitoria-Gasteiz, Alava (Basque Country), Spain.
| | - Eva Granizo Rodriguez
- BIOMICs Research Group (BIOMICS and Microfluidics cluster), Zoology and animal cellular biology department, Faculty of Science and Technology (UPV/EHU), 48940 Leioa, Biscay (Basque Country), Spain
| | - Eunate Arana-Arri
- Clinical Epidemiology Unit, Cruces University Hospital, 48903 Barakaldo, Biscay (Basque Country), Spain
| | - Pedro Pablo España
- Pulmonology Service, Galdakao-Usansolo University Hospital, 48960 Galdakao, Biscay (Basque Country), Spain
| | - Maider Intxausti
- Pulmonology Service, Alava University Hospital - Txagorritxu, 01009 Vitoria-Gasteiz, Álava (Basque Country), Spain
| | - Cristina Sancho
- Department of Pneumology, Basurto University Hospital, 48013 Bilbao, Biscay (Basque Country), Spain
| | | | - Oliver Ibarrondo
- Consultant in Statistics and Health Economics Research, Debagoiena AP-OSI Research Unit, 20500 Arrasate, Gipuzkoa (Basque Country), Spain
| | - Marian M de Pancorbo
- BIOMICs Research Group (BIOMICS and Microfluidics cluster), Zoology and animal cellular biology department, Faculty of Science and Technology (UPV/EHU), 48940 Leioa, Biscay (Basque Country), Spain.
| |
Collapse
|
2
|
Martin Perez S, Sanchez Jimenez F, Fuentes Cantero S, Jímenez Barragan M, Sanchez Mora C, Borreguero Leon JM, Arrobas Velilla T, Valido Morales A, Delgado Torralbo JA, León-Justel A. Uso de inteligencia artificial en la predisposición genética a enfermedad crítica por COVID-19: evaluación comparativa de modelos de aprendizaje automático. ADVANCES IN LABORATORY MEDICINE 2025; 6:190-198. [PMID: 40438555 PMCID: PMC12107414 DOI: 10.1515/almed-2024-0129] [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] [Received: 08/30/2024] [Accepted: 02/20/2025] [Indexed: 06/01/2025]
Abstract
Objetivos La predicción temprana de enfermedad crítica por COVID-19 es crucial para optimizar el manejo clínico. Este estudio tiene como objetivo optimizar la predicción de enfermedad crítica por COVID-19 mediante la integración de datos clínicos, de laboratorio y polimorfismos genéticos en modelos de inteligencia artificial, evaluando y comparando el rendimiento de distintos algoritmos de aprendizaje automático. Métodos Se analizaron 155 pacientes hospitalizados, 23 de los cuales desarrollaron enfermedad crítica. Se realizó un análisis univariante para evaluar la asociación entre siete SNPs y 9 variables clínicas y 10 parámetros de laboratorio en la analítica al ingreso. Resultados De los 7 SNPS, solo tres SNPs se asociaron significativamente con enfermedad crítica: rs77534576, rs10774671 y rs10490770. Los modelos de ensemble consiguieron el mejor rendimiento: Random Forest (AUC=0,989), XGBoost (AUC=0,954) y AdaBoost (AUC=0,927). La importancia de las variables varió entre los modelos, destacando la edad, proteína C reactiva, cardiopatías y los tres SNPs en la mayoría de ellos. La incorporación de los SNPs mejoró el poder predictivo en comparación con estudios previos sin datos genéticos. La validación interna confirmó la superioridad y estabilidad de los modelos de ensemble. Conclusiones Los modelos de aprendizaje automático pueden ayudar en la predicción por enfermedad crítica por Covid-19. La incorporación de SNPs asociados a gravedad a los datos clínicos y de laboratorio mejora el poder predictivo. Se requieren estudios adicionales con cohortes más grandes y diversas para validar y generalizar estos hallazgos antes de su aplicación clínica.
Collapse
Affiliation(s)
- Salomon Martin Perez
- Unidad de Bioquímica clínica, Hospital Universitario Virgen Macarena, Sevilla, España
| | - Flora Sanchez Jimenez
- Unidad de Bioquímica clínica, Hospital Universitario Virgen Macarena, Sevilla, España
| | | | | | - Catalina Sanchez Mora
- Unidad de Bioquímica clínica, Hospital Universitario Virgen Macarena, Sevilla, España
| | | | | | | | | | - Antonio León-Justel
- Unidad de Bioquímica clínica, Hospital Universitario Virgen Macarena, Sevilla, España
| |
Collapse
|
3
|
Martín Pérez S, Sanchez Jimenez F, Fuentes Cantero S, Jímenez Barragan M, Sanchez Mora C, Borreguero Leon JM, Teresa AV, Valido Morales A, Delgado Torralbo JA, León Justel A. Use of artificial intelligence to assess genetic predisposition to develop critical COVID-19 disease: a comparative study of machine learning models. ADVANCES IN LABORATORY MEDICINE 2025; 6:181-189. [PMID: 40438551 PMCID: PMC12107411 DOI: 10.1515/almed-2025-0073] [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] [Received: 08/30/2024] [Accepted: 02/20/2025] [Indexed: 06/01/2025]
Abstract
Objectives Early prediction of critical COVID-19 disease is crucial for an optimal clinical management. The objective of this study was to optimize predictive models for critical COVID-19 disease. Clinical data, laboratory data and genetic polymorphisms were integrated into AI models to compare the performance of different machine learning algorithms. Methods Data from 155 inpatients were analyzed, 23 of whom developed critical disease. A univariate analysis was performed to assess potential correlations between seven SNPs, nine clinical variables and 10 laboratory parameters at admission. Results Of the 7 SNPs, only three SNPs demonstrated a significant association with critical disase, namely: rs77534576, rs10774671 and rs10490770. The ensemble models exhibited the best performance: Random Forest (AUC=0.989), XGBoost (AUC=0.954) and AdaBoost (AUC=0.927). Variable importance varied across models, with age, C-reactive protein, heart diseases and the three SNPs being the most influential features. The predictive power of models improved with the integration of the three SNPs, as compared to previous studies where genetic data were not included. Internal validation confirmed the superiority and stability of the ensemble models. Conclusions Machine learning models may help predict progression into critical COVID-19-disease. The predictive power of models improves when SNPs associated with COVID-19 severity are integrated with laboratory and clinical data. Prior to implementation in clinical practice, larger studies in different populations are needed to validate and support the generalization of these results.
Collapse
Affiliation(s)
- Salomón Martín Pérez
- Service of Clinical Biochemistry Virgen Macarena University Hospital Seville, Seville, Spain
| | - Flora Sanchez Jimenez
- Service of Clinical Biochemistry Virgen Macarena University Hospital Seville, Seville, Spain
| | - Sandra Fuentes Cantero
- Department of Clinical Laboratory Chemistry Rio Tinto General Hospital Huelva, Huelva, Spain
| | - Marta Jímenez Barragan
- Service of Clinical Biochemistry Virgen Macarena University Hospital Seville, Seville, Spain
| | - Catalina Sanchez Mora
- Service of Clinical Biochemistry Virgen Macarena University Hospital Seville, Seville, Spain
| | - Juan M. Borreguero Leon
- Service of Clinical Biochemistry Virgen Macarena University Hospital Seville, Seville, Spain
| | - Arrobas Velilla Teresa
- Service of Clinical Biochemistry Virgen Macarena University Hospital Seville, Seville, Spain
| | | | | | - Antonio León Justel
- Service of Clinical Biochemistry Virgen Macarena University Hospital Seville, Seville, Spain
| |
Collapse
|
4
|
Lammi V, Nakanishi T, Jones SE, Andrews SJ, Karjalainen J, Cortés B, O'Brien HE, Ochoa-Guzman A, Fulton-Howard BE, Broberg M, Haapaniemi HH, Kanai M, Pirinen M, Schmidt A, Mitchell RE, Mousas A, Mangino M, Huerta-Chagoya A, Sinnott-Armstrong N, Cirulli ET, Vaudel M, Kwong ASF, Maiti AK, Marttila MM, Posner DC, Rodriguez AA, Batini C, Minnai F, Dearman AR, Warmerdam CAR, Sequeros CB, Winkler TW, Jordan DM, Rešcenko R, Miano L, Lane JM, Chung RK, Guillen-Guio B, Leavy OC, Carvajal-Silva L, Aguilar-Valdés K, Frangione E, Guare L, Vergasova E, Marouli E, Striano P, Zainulabid UA, Kumar A, Ahmad HF, Edahiro R, Azekawa S, Long COVID Host Genetics Initiative, FinnGen, VA Million Veteran Program, MexGen-COVID Initiative, DBDS Genomic Consortium, GEN-COVID Multicenter Study, PHOSP-COVID Collaborative Group, GENCOV Study, Estonian Biobank Research Team, Luoh SW, Erikstrup C, Pedersen OBV, Lerner-Ellis J, Colombo A, Grzymski JJ, Ishii M, Okada Y, Beckmann ND, Kumari M, Wagner R, Heid IM, John C, Short PJ, Magnus P, Ansone L, Valenti LVC, Lee SA, Wain LV, Verdugo RA, Banasik K, Geller F, Franke LH, Rakitko A, Duncan EL, Renieri A, Tsilidis KK, de Cid R, Niavarani A, Abner E, Tusié-Luna T, Verma SS, Smith GD, Timpson NJ, Madduri RK, Cho K, Daly MJ, Ganna A, Schulte EC, Richards JB, et alLammi V, Nakanishi T, Jones SE, Andrews SJ, Karjalainen J, Cortés B, O'Brien HE, Ochoa-Guzman A, Fulton-Howard BE, Broberg M, Haapaniemi HH, Kanai M, Pirinen M, Schmidt A, Mitchell RE, Mousas A, Mangino M, Huerta-Chagoya A, Sinnott-Armstrong N, Cirulli ET, Vaudel M, Kwong ASF, Maiti AK, Marttila MM, Posner DC, Rodriguez AA, Batini C, Minnai F, Dearman AR, Warmerdam CAR, Sequeros CB, Winkler TW, Jordan DM, Rešcenko R, Miano L, Lane JM, Chung RK, Guillen-Guio B, Leavy OC, Carvajal-Silva L, Aguilar-Valdés K, Frangione E, Guare L, Vergasova E, Marouli E, Striano P, Zainulabid UA, Kumar A, Ahmad HF, Edahiro R, Azekawa S, Long COVID Host Genetics Initiative, FinnGen, VA Million Veteran Program, MexGen-COVID Initiative, DBDS Genomic Consortium, GEN-COVID Multicenter Study, PHOSP-COVID Collaborative Group, GENCOV Study, Estonian Biobank Research Team, Luoh SW, Erikstrup C, Pedersen OBV, Lerner-Ellis J, Colombo A, Grzymski JJ, Ishii M, Okada Y, Beckmann ND, Kumari M, Wagner R, Heid IM, John C, Short PJ, Magnus P, Ansone L, Valenti LVC, Lee SA, Wain LV, Verdugo RA, Banasik K, Geller F, Franke LH, Rakitko A, Duncan EL, Renieri A, Tsilidis KK, de Cid R, Niavarani A, Abner E, Tusié-Luna T, Verma SS, Smith GD, Timpson NJ, Madduri RK, Cho K, Daly MJ, Ganna A, Schulte EC, Richards JB, Ludwig KU, Marks-Hultström M, Zeberg H, Ollila HM. Genome-wide association study of long COVID. Nat Genet 2025; 57:1402-1417. [PMID: 40399555 PMCID: PMC12165857 DOI: 10.1038/s41588-025-02100-w] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Collaborators] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 01/27/2025] [Indexed: 06/16/2025]
Abstract
Infections can lead to persistent symptoms and diseases such as shingles after varicella zoster or rheumatic fever after streptococcal infections. Similarly, severe acute respiratory syndrome coronavirus 2 (SARS‑CoV‑2) infection can result in long coronavirus disease (COVID), typically manifesting as fatigue, pulmonary symptoms and cognitive dysfunction. The biological mechanisms behind long COVID remain unclear. We performed a genome-wide association study for long COVID including up to 6,450 long COVID cases and 1,093,995 population controls from 24 studies across 16 countries. We discovered an association of FOXP4 with long COVID, independent of its previously identified association with severe COVID-19. The signal was replicated in 9,500 long COVID cases and 798,835 population controls. Given the transcription factor FOXP4's role in lung physiology and pathology, our findings highlight the importance of lung function in the pathophysiology of long COVID.
Collapse
Grants
- S10 OD026880 NIH HHS
- UL1 TR004419 NCATS NIH HHS
- Wellcome Trust
- S10 OD030463 NIH HHS
- R01 AI170850 NIAID NIH HHS
- We are grateful for the support from the Academy of Finland (#353812), the NIH (R01AI170850), and the EU. This project has received funding from the European Union’s Horizon Europe research and innovation programme under grant agreement No 101057553. This work was supported by the Swiss State Secretariat for Education, Research and Innovation (SERI) under contract number 22.00094.
- Beatriz Cortés is supported by national grants PI18/01512.
- This work was supported in part through the computational resources and staff expertise provided by Scientific Computing at the Icahn School of Medicine at Mount Sinai and supported by the Clinical and Translational Science Awards (CTSA) grant UL1TR004419 from the National Center for Advancing Translational Sciences. Research reported in this paper was supported by the Office of Research Infrastructure of the National Institutes of Health under award number S10OD026880 and S10OD030463. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.
- REM worked within the MRC Integrative Epidemiology Unit at the University of Bristol, which is supported by the Medical Research Council (MC_UU_00032/01).
- TwinsUK is funded by the Wellcome Trust, Medical Research Council, Versus Arthritis, European Union Horizon 2020, Chronic Disease Research Foundation (CDRF), Zoe Ltd, the National Institute for Health and Care Research (NIHR) Clinical Research Network (CRN) and Biomedical Research Centre based at Guy’s and St Thomas’ NHS Foundation Trust in partnership with King’s College London.
- A.H.-C. is supported by the American Diabetes Association grant 11-23-PDF-35.
- Funding was provided to Desert Research Institute (DRI) by the Nevada Governor's Office of Economic Development. Funding was provided to the Renown Institute for Health Innovation by Renown Health and the Renown Health Foundation.
- The UK Medical Research Council and Wellcome (Grant ref: 217065/Z/19/Z) and the University of Bristol provide core support for ALSPAC.
- EXCEED has been supported by the University of Leicester, the National Institute for Health and Care Research Leicester Respiratory Biomedical Research Centre, the Wellcome Trust (WT 202849), and Medical Research Council grants G0902313 and UKRI_PC_19004, the latter through the UK Research and Innovation Industrial Strategy Challenge Fund, delivered through Health Data Research UK. EXCEED has also been supported by Cohort Access fees from studies funded by the Medical Research Council (MRC), Biotechnology and Biological Sciences Research Council, National Institute for Health and Care Research, the UK Space Agency, and GlaxoSmithKline. The views expressed are those of the author(s) and not necessarily those of the NIHR or the Department of Health and Social Care. This study used the ALICE High Performance Computing Facilities at the University of Leicester. The EXCEED study gratefully acknowledges the support of all participants and staff who have contributed to the study.
- Lude Franke and Robert Warmerdam are supported by grants from the Dutch Research Council (ZonMW-VICI 09150182010019 to L.F. ZonMW LongCOVID grant 10430302110002), European Union’s Horizon Europe Research and Innovation Program grant 101057553 (LongCovid) and through a Senior Investigator Grant from the Oncode Institute.
- Research supported by PNRR-MUR-M4C2 PE0000006 Research Program “MNESYS”—A multiscale integrated approach to the study of the nervous system in health and disease. IRCCS ‘G. Gaslini’ is a member of ERN-Epicare.
- This research was supported by AMED under Grant Number JP22wm0325031.
- The UK Household Longitudinal Study (Understanding Society) is led by the Institute for Social and Economic Research at the University of Essex. The UK Household Longitudinal Study is funded by the Economic and Social Research Council (ESRC: ES/H029745/1). The survey was conducted by Nat Cen and the genome-wide scan data were analyzed and deposited by the Wellcome Trust Sanger Institute (WT098051). The Understanding Society COVID-19 study is funded by the ESRC (ES/K005146/1) and the Health Foundation (2076161). Fieldwork for the survey is carried out by Ipsos MORI and Kantar. AD is partially funded by the ESRC (ES/S012486/1). BH is an ESRC/Biotechnology & Biological Sciences Research Council funded Soc-B Ph.D student (2604276).
- This work was supported by the Bavarian States Ministry of Science and Arts (StMWK; grant to Ralf Wagner and Klaus Überla) as well as by the National Research Network of the University Medicine (NUM; applied surveillance and testing; B-FAST) to Ralf Wagner and Klaus Überla. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
- C.J. was supported by a Medical Research Council Clinical Research Training Fellowship (MR/P00167X/1).
- InnovateUK (grant numbers 87774 and 60502)
- We want to acknowledge the participants and investigators of the Epirus Health Study (EHS). The EHS project is co-financed by the European Regional Development Fund of the European Union and Greek national funds through 1. the Operational Program Competitiveness, Entrepreneurship and Innovation (EPAnEK), NSRF 2014-2020 (Project code MIS: OΠΣ 5047228), and 2. the Operational Programme Epirus 2014–2020 of the Prefecture of Epirus (Project code MIS: HΠ1AB-0028180). The work in the Epirus Health Study was also supported by the National Public Investment Program of the Ministry of Development and Investment/General Secretariat for Research and Technology, in the framework of the Flagship Initiative to address SARS-CoV-2 (2020ΣE01300001).
- This study makes use of data generated by the GCAT - Genomes for Life and COVICAT study - Cohort COVID in Catalonia. GCAT is a cohort study of the Genomes of Catalonia, Fundacio IGTP. IGTP is part of the CERCA Program / Generalitat de Catalunya. GCAT have additional support by Spanish National Grant PI18/01512, TED2021-130626B-I00, La MaratoTV3 167/C/2021 and European Union under grant agreement no. 101046314 (END-VOC), and with partial support of MENARINI. Additional data included in this study was obtained in part by the COVICAT Study Group (Cohort Covid de Catalunya) supported by ISGlobal and IGTP, EIT COVID-19 Rapid Response activity 20873A and SR20-01024 La Caixa Foundation. This study was carried out using anonymized data provided by the Catalan Agency for Quality and Health Assessment, within the framework of the PADRIS Program. The authors of this study would like to acknowledge all GCAT project investigators who contributed to the generation of the GCAT data. A full list of the investigators is available from www.genomesforlife.com/. We thank the Blood and Tissue Bank from Catalonia (BST) and all the GCAT volunteers that participated in the study.
- Financial support grants CONACYT 312688 and PAPIIT/UNAM IG20042.
- GDS works within the MRC Integrative Epidemiology Unit at the University of Bristol, which is supported by the Medical Research Council (MC_UU_00032/01).
- NJT is a Wellcome Trust Investigator (202802/Z/16/Z), is the PI of the Avon Longitudinal Study of Parents and Children (MRC & WT 217065/Z/19/Z), is supported by the University of Bristol NIHR Biomedical Research Centre (BRC-1215-2001), the MRC Integrative Epidemiology Unit (MC_UU_00011/1) and works within the CRUK Integrative Cancer Epidemiology Programme (C18281/A29019). For the purpose of Open Access, the author has applied a CC BY public copyright licence to any Author Accepted Manuscript version arising from this submission.
- This project has received funding from the RIA HORIZON Research and Innovation Actions under GA No. 101057775. (Funded by the European Union. Views and opinions expressed are however those of the author(s) only and do not necessarily reflect those of the European Union or European Health and Digital Agency (HADEA). Neither the European Union nor the granting authority can be held responsible for them.)
- This research is supported by the Million Veteran Program (awards MVP#000 and MVP#067) and the Centralized Interactive Phenomics Resource (CIPHER) Program funded by the Office of Research and Development, Veterans Health Administration. This research used resources of the Knowledge Discovery Infrastructure at Oak Ridge National Laboratory, which is supported by the Office of Science of the US Department of Energy under Contract No. DE-AC05-00OR22725. This publication does not represent the views of the Department of Veteran Affairs or the United States Government.
- BGG is supported by Wellcome Trust grant 221680/Z/20/Z. For the purpose of open access, the author has applied a CC BY public copyright license to any Author Accepted Manuscript version arising from this submission.
- LVW holds a GlaxoSmithKline Asthma + Lung UK Chair in Respiratory Research (C17-1). This research was partially supported by the National Institute for Health Research (NIHR) Leicester Biomedical Research Centre.
- The C19-GenoNet project is funded by grants ANID COVID0961, COVID0789, COVID1005, ESR UTA2295 and ACT210085.
- European Regional Development Fund (ERDF), Measure 1.1.1.1 “Support for applied research”, project „Identification of molecular determinants associated with the risk for various COVID-19 long term effects: a comprehensive cohort-based study in Latvian population. (POST-COVID-TRACK)” financially supported sample analysis for Latvian cohort.
- Fondazione IRCCS Ca' Granda 5permille, "Fondazione COVID-19 Genomic Study (FOGS)".
- This work was supported by awards 2160 from Fast Grants.
- The work of E.A. was funded by the European Union through Horizon 2020 and Horizon Europe research and innovation programs under grants no. 894987, 101137201 and 101137154, and Estonian Research Council Grant PRG1291.
- The Danish analysis was funded by 101057129 — REACT — HORIZON-HLTH-2021-DISEASE-04 and DFF 0214-00127B.
- This work was supported by the Bavarian States Ministry of Science and Arts (StMWK; grant to Ralf Wagner and Klaus Überla) as well as by the National Research Network of the University Medicine (NUM; applied surveillance and testing; B-FAST) to Ralf Wagner and Klaus Überla.
- The SweCOVID was funded by the SciLifeLab/Knut and Alice Wallenberg national COVID-19 research program (M.H.: KAW 2020.0182, KAW 2020.0241), the Swedish Heart-Lung Foundation (M.H.: 20210089, 20190639, 20190637), the Swedish Research Council (R.F.: 2014-02569, 2014-07606, and H.Z.: 2021-03050), Swedish Society of Medicine (M.H.:SLS-938101), the and Swedish Kidney Foundation (R.F.: F2020-0054).
- We acknowledge funding from the Novo Nordisk Foundation (grants NNF17OC0027594 and NNF14CC0001) and Sygesikring Danmark (2020-0178).
- >PHOSP-COVID is jointly funded by a grant from the MRC-UK Research and Innovation and the Department of Health and Social Care through the National Institute for Health Research (NIHR) rapid response panel to tackle COVID-19 (grant references: MR/V027859/1 and COV0319). The views expressed in the publication are those of the author(s) and not necessarily those of the National Health Service (NHS), the NIHR or the Department of Health and Social Care. This study would not be possible without all the participants who have given their time and support. We thank all the participants and their families. We thank the many research administrators, health-care and social-care professionals who contributed to setting up and delivering the study at all of the 65 NHS trusts/Health boards and 25 research institutions across the UK, as well as all the supporting staff at the NIHR Clinical Research Network, Health Research Authority, Research Ethics Committee, Department of Health and Social Care, Public Health Scotland, and Public Health England, and support from the ISARIC Coronavirus Clinical Characterisation Consortium. We thank Kate Holmes at the NIHR Office for Clinical Research Infrastructure (NOCRI) for her support in coordinating the charities group. The PHOSP-COVID industry framework was formed to provide advice and support in commercial discussions, and we thank the Association of the British Pharmaceutical Industry as well NOCRI for coordinating this. We are very grateful to all the charities that have provided insight to the study: Action Pulmonary Fibrosis, Alzheimer’s Research UK, Asthma + Lung UK, British Heart Foundation, Diabetes UK, Cystic Fibrosis Trust, Kidney Research UK, MQ Mental Health, Muscular Dystrophy UK, Stroke Association Blood Cancer UK, McPin Foundations, and Versus Arthritis. We thank the NIHR Leicester Biomedical Research Centre patient and public involvement group and Long Covid Support.
- GENCOV: This work was supported by the Canadian Institutes of Health Research (Funding Reference Number VR4-172753).
- Project Tuscany Health Ecosystem spoke 7 translational medicine for rare, oncological and infectious diseases financed by European Union- Next generation EU, Mission 4 Component 2 Inv. 15 CUP:2266-2022-RA-PROFCMUR_PNRR_PC_THE_AFFSPOK; and INTERVENE EU H2020-SC1-FA-DTS-2018–2020, International consortium for integrative genomics prediction - Grant Agreement No. 101016775.
- We thank the Veterans, staff, and investigators of VA Million Veteran Program. The VA Million Veteran Program is funded by the Office of Research and Development, Veterans Health Administration.
- The FinnGen project is funded by two grants from Business Finland (HUS 4685/31/2016 and UH 4386/31/2016) and the following industry partners: AbbVie Inc., AstraZeneca UK Ltd, Biogen MA Inc., Bristol Myers Squibb (and Celgene Corporation & Celgene International II Sàrl), Genentech Inc., Merck Sharp & Dohme LCC, Pfizer Inc., GlaxoSmithKline Intellectual Property Development Ltd., Sanofi US Services Inc., Maze Therapeutics Inc., Janssen Biotech Inc, Novartis AG, and Boehringer Ingelheim International GmbH.
Collapse
Affiliation(s)
- Vilma Lammi
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Tomoko Nakanishi
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
- Centre for Clinical Epidemiology, Department of Medicine, Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, Quebec, Canada
- Kyoto-McGill International Collaborative Program in Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Department of Genome Informatics, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
- Research Fellow, Japan Society for the Promotion of Science, Tokyo, Japan
| | - Samuel E Jones
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Shea J Andrews
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Juha Karjalainen
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Beatriz Cortés
- Genomes for Life-GCAT Lab, CORE Program, Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
- Grup de REcerca en Impacte de les Malalties Cròniques i les seves Trajectòries (GRIMTra), Barcelona, Spain
| | | | - Ana Ochoa-Guzman
- Unidad de Biología Molecular y Medicina Genómica, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Brian E Fulton-Howard
- Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Martin Broberg
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Hele H Haapaniemi
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Masahiro Kanai
- Broad Institute, Cambridge, MA, USA
- Analytical and Translational Genetics Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Matti Pirinen
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- Department of Mathematics and Statistics, University of Helsinki, Helsinki, Finland
- Department of Public Health, University of Helsinki, Helsinki, Finland
| | - Axel Schmidt
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | - Ruth E Mitchell
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Abdou Mousas
- Department of Hygiene and Epidemiology, University of Ioannina School of Medicine, Ioannina, Greece
| | - Massimo Mangino
- Department of Twin Research, King's College London, London, UK
| | - Alicia Huerta-Chagoya
- Program in Metabolism and Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Genomic Medicine and Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Unidad de Biología Molecular y Medicina Genómica, Instituto Nacional de Ciencias Médicas y Nutrición, Mexico City, Mexico
| | - Nasa Sinnott-Armstrong
- Herbold Computational Biology Program, Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Finnish Institute of Molecular Medicine, University of Helsinki, Helsinki, Finland
| | | | - Marc Vaudel
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Genetics and Bioinformatics, Health Data and Digitalization, Norwegian Institute of Public Health, Oslo, Norway
- Computational Biology Unit, Department of Informatics, University of Bergen, Bergen, Norway
| | - Alex S F Kwong
- Centre for Clinical Brain Sciences, Division of Psychiatry, University of Edinburgh, Edinburgh, UK
| | - Amit K Maiti
- Department of Genetics and Genomics, Mydnavar, Southfield, MI, USA
| | - Minttu M Marttila
- University of Helsinki, Helsinki, Finland
- Helsinki University Central Hospital, Helsinki, Finland
| | | | | | - Chiara Batini
- Department of Population Health Sciences, University of Leicester, Leicester, UK
- University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Francesca Minnai
- Institute for Biomedical Technologies-National Research Council, Segrate, Italy
- Department of Medical Biotechnology and Translational Medicine (BioMeTra), Università degli Studi di Milano, Milan, Italy
| | - Anna R Dearman
- Institute for Social and Economic Research, University of Essex, Colchester, UK
| | - C A Robert Warmerdam
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- Oncode Investigator, Utrecht, the Netherlands
| | - Celia B Sequeros
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thomas W Winkler
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany
| | - Daniel M Jordan
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | | | | | - Jacqueline M Lane
- Brigham and Women's Hospital Division of Sleep and Circadian Disorders, Boston, MA, USA
- Massachusetts General Hospital, Center for Genomic Medicine, Boston, MA, USA
- Broad Institute, Molecular and Population Genetics Program, Cambridge, MA, USA
| | - Ryan K Chung
- Center for Computational Biology, University of California Berkeley, Berkeley, CA, USA
| | - Beatriz Guillen-Guio
- Department of Population Health Sciences, University of Leicester, Leicester, UK
- The Institute for Lung Health, NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| | - Olivia C Leavy
- Department of Population Health Sciences, University of Leicester, Leicester, UK
- The Institute for Lung Health, NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| | - Laura Carvajal-Silva
- Departamento de Oncología Básico Clínica, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Kevin Aguilar-Valdés
- Departamento de Oncología Básico Clínica, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Erika Frangione
- Mount Sinai Hospital, Sinai Health, Toronto, Ontario, Canada
| | - Lindsay Guare
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Eirini Marouli
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | | | - Ummu Afeera Zainulabid
- Department of Internal Medicine, Kulliyyah of Medicine, International Islamic University Malaysia, Pahang, Malaysia
| | - Ashutosh Kumar
- Department of Anatomy, All India Institute of Medical Sciences-Patna, Patna, India
| | - Hajar Fauzan Ahmad
- Faculty of Industrial Sciences and Technology, Universiti Malaysia Pahang Al Sultan Abdullah, Pahang, Malaysia
| | - Ryuya Edahiro
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Shuhei Azekawa
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | | | | | | | | | | | | | | | | | - Shiuh-Wen Luoh
- VA Portland Health Care System, Portland, Portland, OR, USA
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Christian Erikstrup
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
| | - Ole B V Pedersen
- Department of Clinical Immunology, Zealand University Hospital-Køge, Køge, Denmark
| | - Jordan Lerner-Ellis
- Mount Sinai Hospital, Sinai Health, Toronto, Ontario, Canada
- University of Toronto, Toronto, Ontario, Canada
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
| | - Alicia Colombo
- Departamento de Oncología Básico Clínica, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Departamento de Anatomía Patológica, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Servicio de Anatomía Patológica, Hospital Clínico de la Universidad de Chile, Santiago, Chile
| | - Joseph J Grzymski
- Department of Internal Medicine, University of Nevada Reno, School of Medicine, Reno, NV, USA
| | - Makoto Ishii
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yukinori Okada
- Department of Genome Informatics, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, Japan
| | - Noam D Beckmann
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
- Division of Data Driven Medicine, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Meena Kumari
- Institute for Social and Economic Research, University of Essex, Colchester, UK
| | - Ralf Wagner
- Institute of Medical Microbiology and Hygiene, Molecular Microbiology (Virology), University of Regensburg, Regensburg, Germany
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - Iris M Heid
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany
| | - Catherine John
- Department of Population Health Sciences, University of Leicester, Leicester, UK
- Leicester National Institute for Health and Care Research, Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | | | - Per Magnus
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Laura Ansone
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Luca V C Valenti
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
- Biological Resource Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Sulggi A Lee
- Department of Medicine, Division of HIV, Infectious Diseases and Global Medicine, University of California, San Francisco, CA, USA
| | - Louise V Wain
- Department of Population Health Sciences, University of Leicester, Leicester, UK
- The Institute for Lung Health, NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| | - Ricardo A Verdugo
- Departamento de Oncología Básico Clínica, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Instituto de Investigación Interdisciplinaria y Facultad de Medicina, Universidad de Talca, Talca, Chile
| | - Karina Banasik
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Lude H Franke
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- Oncode Investigator, Utrecht, the Netherlands
| | | | - Emma L Duncan
- Department of Endocrinology, Guy's and St Thomas' NHS Foundation Trust, London, UK
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Alessandra Renieri
- Medical Genetics, University of Siena, Siena, Italy
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy
- Genetica Medica, Azienda Ospedaliero-Universitaria Senese, Siena, Italy
| | - Konstantinos K Tsilidis
- Department of Hygiene and Epidemiology, University of Ioannina School of Medicine, Ioannina, Greece
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Rafael de Cid
- Genomes for Life-GCAT Lab, CORE Program, Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
- Grup de REcerca en Impacte de les Malalties Cròniques i les seves Trajectòries (GRIMTra), Barcelona, Spain
| | - Ahmadreza Niavarani
- Digestive Oncology Research Center, Digestive Disease Research Institute, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Erik Abner
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Teresa Tusié-Luna
- Instituto de Investigaciones Biomédicas, UNAM, Mexico City, Mexico
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Shefali S Verma
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - George Davey Smith
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Nicholas J Timpson
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Ravi K Madduri
- Data Science and Learning Division, Argonne National Laboratory, Lemont, IL, USA
| | - Kelly Cho
- Department of Medicine, Harvard Medical School and Mass General Brigham, Boston, MA, USA
| | - Mark J Daly
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- Broad Institute, Cambridge, MA, USA
- Analytical and Translational Genetics Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Andrea Ganna
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- Broad Institute, Cambridge, MA, USA
- Analytical and Translational Genetics Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Eva C Schulte
- Department of Psychiatry, University of Munich, Munich, Germany
- Institute of Human Genetics, University Hospital, Faculty of Medicine, University of Bonn, Bonn, Germany
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
- Institute of Psychiatric Phenomics and Genomics, University of Munich, Munich, Germany
- Department of Psychiatry, University Hospital, Faculty of Medicine, University of Bonn, Bonn, Germany
| | - J Brent Richards
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
- Centre for Clinical Epidemiology, Department of Medicine, Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, Quebec, Canada
- Department of Twin Research, King's College London, London, UK
- 5 Prime Sciences Inc, Montreal, Quebec, Canada
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Quebec, Canada
| | - Kerstin U Ludwig
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | - Michael Marks-Hultström
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Quebec, Canada
- Lady Davis Institute of Medical Research, Jewish General Hospital, McGill University, Montreal, Quebec, Canada
- Anaesthesiology and Intensive Care Medicine, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Hugo Zeberg
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany.
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
| | - Hanna M Ollila
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland.
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
Collaborators
Mari E K Niemi, Janick St-Cyr, Darin Adra, Madeleine Durand, David Bujold, Guillaume Bourque, Ariane Boisclair, Mylene Bertrand, Daniel Auld, Laetitia Laurent, Solomia Yanishevsky, G Mark Lathrop, Fangyi Shi, Simon Rousseau, Jiannis Ragoussis, Danielle Perley, Vincent Mooser, David R Morrison, Daniella Balla, Julia Heggemann, Sonja Schultz, Pari Behzad, Markus M Nöthen, Abigail Miller, Max C Pensel, Carlo Maj,
Collapse
|
5
|
Letovsky SI, Cao X, Hollenbach JA, Mack SJ, Maiers M. Association between HLA genetics and SARS-CoV-2 infection in a large real-world cohort. Genes Immun 2025; 26:213-221. [PMID: 40275118 DOI: 10.1038/s41435-025-00328-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 03/31/2025] [Accepted: 04/03/2025] [Indexed: 04/26/2025]
Abstract
Genetic variation in the human leukocyte antigen (HLA) region is thought to influence susceptibility to and severity of a variety of infectious diseases. Several studies have explored a possible relationship between HLA genetics and SARS-CoV-2 infection, although mixed results, small sample sizes, and difficulty controlling for exposure risk have made it difficult to draw firm conclusions. Here, a dataset of 419,234 subjects with HLA genotype data and COVID-19 PCR test results was studied. A baseline analysis was performed to examine the association of non-HLA factors on COVID-19 positivity. Then, multivariate logistic regressions, incorporating single and paired HLA alleles, were performed and then corrected for significant factors from the baseline analysis. Proxies for socioeconomic status and exposure risk were significantly associated with COVID-19 positivity across all ancestry groups studied. Forty-one single HLA alleles displayed significant association with COVID-19 positivity; after controlling for socioeconomic status and exposure risk, only eight significant associations remained. Additionally, two HLA allele pairs were associated with test positivity after correction. Of all variables, socioeconomic status showed the greatest effect size. The results from this study suggest that many, if not all, of the reported associations between HLA alleles and SARS-CoV-2 infection may be spurious, owing to confounding factors.
Collapse
Affiliation(s)
- Stanley I Letovsky
- Department of Data Science, AI, and Bioinformatics, Laboratory Corporation of America, Burlington, NC, USA.
| | - Xia Cao
- Department of Data Science, AI, and Bioinformatics, Laboratory Corporation of America, Burlington, NC, USA
| | - Jill A Hollenbach
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics; University of California, San Francisco, San Francisco, CA, USA
| | - Steven J Mack
- Department of Pediatrics, University of California, San Francisco, Oakland, CA, USA
| | - Martin Maiers
- CIBMTR® (Center for International Blood and Marrow Transplant Research), NMDP, Minneapolis, MN, USA
| |
Collapse
|
6
|
Rosa-Baez C, Borrego-Yaniz G, Rodriguez-Martin I, Kerick M, Acosta-Herrera M, Martín J, Ortiz-Fernández L. Cross-trait GWAS in COVID-19 and systemic sclerosis reveals novel genes implicated in fibrotic and inflammation pathways. Rheumatology (Oxford) 2025; 64:4022-4031. [PMID: 39878951 DOI: 10.1093/rheumatology/keaf028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 12/17/2024] [Accepted: 01/06/2025] [Indexed: 01/31/2025] Open
Abstract
OBJECTIVES Coronavirus disease 2019 (COVID-19) and SSc share multiple similarities in their clinical manifestations, alterations in immune response and therapeutic options. These resemblances have also been identified in other immune-mediated inflammatory diseases where a common genetic component has been found. Thus, we decided to evaluate for the first time this shared genetic architecture with SSc. METHODS For this study, we retrieved genomic data from two European-ancestry cohorts: 2 597 856 individuals from The COVID-19 Host Genetics Initiative consortium, and 26 679 individuals from the largest genomic scan in SSc. We performed a cross-trait meta-analyses including >9.3 million single nucleotide polymorphisms. Finally, we conducted functional annotation to prioritize potential causal genes and performed drug repurposing analysis. RESULTS Our results revealed a total of 19 non-HLA pleiotropic loci, including 2 novel associations for both conditions (BMP1 and PPARG) and 12 emerging as new shared loci. Functional annotation of these regions underscored their potential regulatory role and identified potential causal genes, many of which are implicated in fibrotic and inflammatory pathways. Remarkably, we observed an antagonistic pleiotropy model of the IFN signalling between COVID-19 and SSc, including the well-known TYK2 P1104A missense variant, showing a protective effect for SSc while being a risk factor for COVID-19, along with two additional novel pleiotropic associations (IRF8 and SENP7). Finally, our findings provide new therapeutic options that could potentially benefit both conditions. CONCLUSION Our study confirms the genetic resemblance between susceptibility to and severity of COVID-19 and SSc, revealing a novel common genetic contribution affecting fibrotic and immune pathways.
Collapse
Affiliation(s)
- Carlos Rosa-Baez
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine López-Neyra, CSIC, Granada, Spain
| | - Gonzalo Borrego-Yaniz
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine López-Neyra, CSIC, Granada, Spain
| | - Inmaculada Rodriguez-Martin
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine López-Neyra, CSIC, Granada, Spain
| | - Martin Kerick
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine López-Neyra, CSIC, Granada, Spain
| | - Marialbert Acosta-Herrera
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine López-Neyra, CSIC, Granada, Spain
- Systemic Autoimmune Disease Unit, Hospital Clínico San Cecilio, Instituto de Investigación Biosanitaria Ibs. GRANADA, Granada, Spain
| | - Javier Martín
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine López-Neyra, CSIC, Granada, Spain
| | - Lourdes Ortiz-Fernández
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine López-Neyra, CSIC, Granada, Spain
| |
Collapse
|
7
|
Antolí A, Vargas-Parra G, Sierra-Fortuny A, Gomez-Vazquez JL, Rofes P, Munté E, Viana-Errasti J, Marín-Montes R, López-Doriga A, Feliubadaló L, Del Valle J, Pérez-González A, Poveda E, Solanich X, Lázaro C. From Rare to Common: Genetic Insights into TLR7 Variants in a Multicentric Spanish Study on COVID-19 Severity. J Clin Immunol 2025; 45:100. [PMID: 40423910 PMCID: PMC12116960 DOI: 10.1007/s10875-025-01892-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Accepted: 05/10/2025] [Indexed: 05/28/2025]
Abstract
TLR7, which encodes a key receptor for single-stranded RNA (ssRNA) virus of the innate immune system, was recently associated with X-linked immunodeficiency and COVID-19 susceptibility. This study investigates the association between TLR7 variants and susceptibility to severe COVID-19 in a multicentric Spanish cohort. The TLR7 gene was sequenced in a cohort of 365 COVID-19 patients, stratified into two groups: one comprising mild and asymptomatic patients, considered as controls (n = 87), and the other consisting of moderate to severely affected patients hospitalized due to COVID-19 pneumonia, considered as cases (n = 278). A total of 152 unique TLR7 variants were identified, of note, six rare variants were identified in 11 cases (3.96%), all of whom belonged to the case group. The functional impact of rare TLR7 variants was assessed using a luciferase reporter assay and revealed that N215S is a loss-of-function (LOF) variant, while D332G exhibits an hypomorphic behavior. Conversely, H90Y, V219I, A448V, and R902K maintained normal signaling. No skewed X-inactivation was observed in female carriers of N215S or D332G. In addition, the common variants Q11L (rs179008), c.4-151A>G (rs179009) and c.*881C>G (rs3853839) were associated with severe pneumonia, while c.4-151A>G (rs179009) was specifically linked to Intensive Care Unit (ICU) admission. These findings highlight the role of TLR7 in antiviral immune response and its association with severe COVID-19 in men. The luciferase assay proves to be a reliable tool for evaluating TLR7 signaling, effectively distinguishing between neutral, LOF, and gain-of-function (GOF) variants. Further research is needed to better understand TLR7 variants and its implications in immunodeficiency and immune dysregulation.
Collapse
Affiliation(s)
- Arnau Antolí
- Internal Medicine Department, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain
- Adult Primary Immunodeficiency Unit (UFIPA), Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain
- The Systemic, Vascular Diseases and Ageing Group. Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
- Clinical Sciences Department, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Gardenia Vargas-Parra
- Hereditary Cancer Program, Catalan Institute of Oncology, L'Hospitalet de Llobregat, Barcelona, Spain
- Molecular Mechanisms and Experimental Therapy in Oncology Program, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Angels Sierra-Fortuny
- Internal Medicine Department, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain
- The Systemic, Vascular Diseases and Ageing Group. Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Jose Luis Gomez-Vazquez
- Internal Medicine Department, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain
- The Systemic, Vascular Diseases and Ageing Group. Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
- Clinical Sciences Department, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Paula Rofes
- Hereditary Cancer Program, Catalan Institute of Oncology, L'Hospitalet de Llobregat, Barcelona, Spain
- Molecular Mechanisms and Experimental Therapy in Oncology Program, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Elisabet Munté
- Hereditary Cancer Program, Catalan Institute of Oncology, L'Hospitalet de Llobregat, Barcelona, Spain
- Molecular Mechanisms and Experimental Therapy in Oncology Program, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Julen Viana-Errasti
- Molecular Mechanisms and Experimental Therapy in Oncology Program, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Raúl Marín-Montes
- Molecular Mechanisms and Experimental Therapy in Oncology Program, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
- Unit of Bioinformatics for Precision Oncology, Catalan Institute of Oncology, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Adriana López-Doriga
- Unit of Bioinformatics for Precision Oncology, Catalan Institute of Oncology, L'Hospitalet de Llobregat, Barcelona, Spain
- Nennisiwok AI Lab, Barcelona, Spain
| | - Lidia Feliubadaló
- Hereditary Cancer Program, Catalan Institute of Oncology, L'Hospitalet de Llobregat, Barcelona, Spain
- Molecular Mechanisms and Experimental Therapy in Oncology Program, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Jesús Del Valle
- Hereditary Cancer Program, Catalan Institute of Oncology, L'Hospitalet de Llobregat, Barcelona, Spain
- Molecular Mechanisms and Experimental Therapy in Oncology Program, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Alexandre Pérez-González
- Internal Medicine Department, Complexo Hospitalario Universitario de Vigo (CHUVI), SERGAS, Vigo, Pontevedra, Spain
- Virology and Pathogenesis, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Pontevedra, Spain
| | - Eva Poveda
- Virology and Pathogenesis, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Pontevedra, Spain
| | - Xavier Solanich
- Internal Medicine Department, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain
- Adult Primary Immunodeficiency Unit (UFIPA), Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain
- The Systemic, Vascular Diseases and Ageing Group. Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
- Clinical Sciences Department, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Conxi Lázaro
- Clinical Sciences Department, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain.
- Hereditary Cancer Program, Catalan Institute of Oncology, L'Hospitalet de Llobregat, Barcelona, Spain.
- Molecular Mechanisms and Experimental Therapy in Oncology Program, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
8
|
Boakye AO, Obirikorang C, Afum-Adjei Awuah A, Adu EA, Winter D, Boham EE, Alani H, Newton SK, Almoustapha NST, Deke J, Dzadey WO, Adu-Amoah L, Kroduah SA, Grant MA, Asare G, Amoako-Adusei A, Loag W, Kettenbeil J, Sarkodie YA, Oduro-Mensah E, Yawson AE, Apanga S, Odotei Adjei R, Adobasom-Anane AG, Lorenz E, Souares A, Maiga-Ascofaré O, May J, Struck NS, Amuasi JH. Genetic association of ACE2 rs2285666 (C>T) and rs2106809 (A>G) and susceptibility to SARS-CoV-2 infection among the Ghanaian population. Front Genet 2025; 16:1555515. [PMID: 40491571 PMCID: PMC12146278 DOI: 10.3389/fgene.2025.1555515] [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: 01/04/2025] [Accepted: 05/07/2025] [Indexed: 06/11/2025] Open
Abstract
Background Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), enters human cells using the angiotensin-converting enzyme 2 (ACE-2) receptor. ACE2 single nucleotide polymorphisms (SNPs) can influence susceptibility by affecting viral binding or gene expression. This study investigated the association between ACE2 SNPs, rs2285666 and rs2106809, and the SARS-CoV-2 infection susceptibility in a Ghanaian population. Methods Genomic DNA was extracted, using a magnetic bead-based method, from blood samples of a random-subset of 1,334 participants drawn from a two-stage cluster, population-based household cross-sectional SARS-CoV-2 IgG seroprevalence survey. Data collected included, socio-demographic characteristics, medical history, vaccination, and smoking status. Genotyping of the ACE2 SNPs was performed using Allele-Specific Oligonucleotide Polymerase Chain Reaction (ASO-PCR) combined with melting curve analysis. Logistic regression models were utilized to assess the association between the ACE2 SNPs and the susceptibility to SARS-CoV-2 infection. Results The median age of participants was 33 [Interquartile range (IQR) = 24-46] years. Females accounted for the majority of the sampled population, 64.3%. SARS-CoV-2-IgG seropositivity was (58.4%, 95%CI: 52.6%-64.2%) among the male population and (54.1%, 95%CI: 49.54%-58.61%) in the female population. There were no significant differences in overall allele or genotype frequencies of ACE2 SNPs between SARS-CoV-2 IgG seropositive and seronegative individuals for both females and males. Among females, those with the T allele of ACE2 rs2285666 had a 38% decreased susceptibility to SARS-CoV-2 infection under the dominant [adjusted odds ratio (aOR) = 0.62; 95%CI = 0.45-0.85, P = 0.003] and heterozygous advantage models (aOR = 0.62; 95%CI = 0.45-0.86, P = 0.004), after adjusting for confounders, but not thee recessive model (aOR = 0.41; 95%CI = 0.03-5.22, P = 0.490). No significant association was observed among males. Overall, the ACE2 rs2106809 was not associated with the susceptibility to SARS-CoV-2 infection in both males and females. Conclusion This study found no association between ACE2 rs2106809 genetic variant and susceptibility to SARS-CoV-2 infection, whilst the rs2285666 T-allele was associated with a decreased frequency for SARS-CoV-2 infection among Ghanaian females. These findings enhance our understanding of genetic factors influencing SARS-CoV-2 susceptibility, which could help identify at-risk populations and inform more targeted public health interventions in future outbreaks.
Collapse
Affiliation(s)
- Alexander Owusu Boakye
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Department of Molecular Medicine, School of Medicine and Dentistry, College of Health Sciences, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
| | - Christian Obirikorang
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Department of Molecular Medicine, School of Medicine and Dentistry, College of Health Sciences, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
| | - Anthony Afum-Adjei Awuah
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Department of Molecular Medicine, School of Medicine and Dentistry, College of Health Sciences, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
| | - Evans Asamoah Adu
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Department of Molecular Medicine, School of Medicine and Dentistry, College of Health Sciences, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
| | - Doris Winter
- Infectious Disease Epidemiology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Eric Ebenezer Boham
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Department of Molecular Medicine, School of Medicine and Dentistry, College of Health Sciences, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
| | - Hakim Alani
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Sylvester Kofi Newton
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Nana Safi Toure Almoustapha
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - James Deke
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Department of Clinical Microbiology, School of Medicine and Dentistry, College of Health Sciences, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
| | - Welbeck Odame Dzadey
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Louis Adu-Amoah
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Department of Molecular Medicine, School of Medicine and Dentistry, College of Health Sciences, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
| | - Sally-Ann Kroduah
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Department of Global and International Health, School of Public Health, College of Health Sciences, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
| | - Mary Ama Grant
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Gracelyn Asare
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Amos Amoako-Adusei
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Wibke Loag
- Infectious Disease Epidemiology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Jenny Kettenbeil
- Infectious Disease Epidemiology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Yaw Adu Sarkodie
- Department of Clinical Microbiology, School of Medicine and Dentistry, College of Health Sciences, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
| | | | - Alfred Edwin Yawson
- Department of Community Health, Medical School, University of Ghana, Accra, Ghana
| | - Stephen Apanga
- Department of Community Health and Preventive Medicine, School of Medicine, University for Development Studies, Tamale, Ghana
| | - Rose Odotei Adjei
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Department of Global and International Health, School of Public Health, College of Health Sciences, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
| | - Austin Gideon Adobasom-Anane
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Department of Global and International Health, School of Public Health, College of Health Sciences, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
| | - Eva Lorenz
- Infectious Disease Epidemiology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Borstel-Lübeck-Riems, Hamburg, Germany
| | - Aurélia Souares
- German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
- Heidelberg Institute of Global Health (HIGH), Heidelberg University Hospital, Heidelberg, Germany
| | - Oumou Maiga-Ascofaré
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Infectious Disease Epidemiology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Borstel-Lübeck-Riems, Hamburg, Germany
| | - Jürgen May
- Infectious Disease Epidemiology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Borstel-Lübeck-Riems, Hamburg, Germany
- Department of Tropical Medicine I, University Medical Centre Hamburg Eppendorf (UKE), Hamburg, Germany
| | - Nicole S. Struck
- Infectious Disease Epidemiology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Borstel-Lübeck-Riems, Hamburg, Germany
| | - John Humphery Amuasi
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Infectious Disease Epidemiology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- Department of Global and International Health, School of Public Health, College of Health Sciences, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
- Department of Tropical Medicine I, University Medical Centre Hamburg Eppendorf (UKE), Hamburg, Germany
| |
Collapse
|
9
|
Ruß AK, Schreiber S, Lieb W, Vehreschild JJ, Heuschmann PU, Illig T, Appel KS, Vehreschild MJGT, Krefting D, Reinke L, Viebke A, Poick S, Störk S, Reese JP, Zoller T, Krist L, Ellinghaus D, Foesel BU, Gieger C, Lorenz-Depiereux B, Witzenrath M, Anton G, Krawczak M, Heyckendorf J, Bahmer T. Genome-wide association study of post COVID-19 syndrome in a population-based cohort in Germany. Sci Rep 2025; 15:15791. [PMID: 40328884 PMCID: PMC12056214 DOI: 10.1038/s41598-025-00945-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 05/02/2025] [Indexed: 05/08/2025] Open
Abstract
If health impairments due to coronavirus disease 2019 (COVID-19) persist for 12 weeks or longer, patients are diagnosed with Post-COVID Syndrome (PCS), or Long-COVID. Although the COVID-19 pandemic has largely subsided in 2024, PCS is still a major health burden worldwide, and identifying potential genetic modifiers of PCS remains of great clinical and scientific interest. We therefore performed a case-control type genome-wide association study (GWAS) of three recently developed PCS (severity) scores in 2,247 participants of COVIDOM, a prospective, multi-centre, population-based cohort study of SARS-CoV-2-infected individuals in Germany. Each PCS score originally represented the weighted sum of the binary indicators of all, or a subset, of 12 PCS symptom complexes, assessed six months or later after the PCR test-confirmed SARS-CoV-2 infection of a participant. For various methodical reasons, however, the PCS scores were dichotomized along their respective median values in the present study, prior to the GWAS. Of the 6,383,167 single nucleotide polymorphisms included, various variants were found to be associated with at least one of the PCS scores, although not at the stringent genome-wide statistical significance level of 5 × 10- 8. With p = 6.6 × 10- 8, however, the genotype-phenotype association of SNP rs9792535 at position chr9:127,166,653 narrowly missed this threshold. The SNP is located in a region including the NEK6, PSMB7 and ADGRD2 genes which, however, does not immediately suggest an etiological connection to PCS. As regards functional plausibility, variants of a possible effect mapped to the olfactory receptor gene region (lead SNP rs10893121 at position chr11:123,854,744; p = 2.5 × 10- 6). Impairment of smell and taste is a pathognomonic feature of both, acute COVID-19 and PCS, and our results suggest that this connection may have a genetic basis. Three other genotype-phenotype associations pointed towards a possible etiological role in PCS of cellular virus repression (CHD6 gene region), activation of macrophages (SLC7A2) and the release of virus particles from infected cells (ARHGAP44). All other gene regions highlighted by our GWAS did not relate to pathophysiological processes currently discussed for PCS. Therefore, and because the genotype-phenotype associations observed in our GWAS were generally not very strong, the complexity of the genetic background of PCS appears to be as high as that of most other multifactorial traits in humans.
Collapse
Affiliation(s)
- Anne-Kathrin Ruß
- Institute of Medical Informatics and Statistics, University Medical Center Schleswig-Holstein, Kiel University, Brunswiker Straße 10, 24113, Kiel, Germany
- Institute of Epidemiology, University Medical Center Schleswig-Holstein, Kiel University, Kiel, Germany
| | - Stefan Schreiber
- Department of Internal Medicine I, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Wolfgang Lieb
- Institute of Epidemiology, University Medical Center Schleswig-Holstein, Kiel University, Kiel, Germany
| | - J Janne Vehreschild
- Institute of Digital Medicine and Clinical Data Science, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
- Department I of Internal Medicine, Faculty of Medicine, University Hospital CologneUniversity of Cologne, Cologne, Germany
| | - Peter U Heuschmann
- Institute of Clinical Epidemiology and Biometry, University of Würzburg, Würzburg, Germany
- Institute of Medical Data Science, University Hospital Würzburg, Würzburg, Germany
- Clinical Trial Center, University Hospital Würzburg, Würzburg, Germany
| | - Thomas Illig
- Hannover Unified Biobank, Hannover Medical School, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover, German Center for Lung Research, Hannover, Germany
| | - Katharina S Appel
- Institute of Digital Medicine and Clinical Data Science, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
| | - Maria J G T Vehreschild
- Medical Department 2, Center for Internal Medicine, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt, Germany
| | - Dagmar Krefting
- Department of Medical Informatics, University Medical Center Göttingen, Göttingen, Germany
- Campus Institute Data Sciences, Göttingen, Germany
| | - Lennart Reinke
- Department of Internal Medicine I, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Alin Viebke
- Department of Internal Medicine I, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Susanne Poick
- Institute of Epidemiology, University Medical Center Schleswig-Holstein, Kiel University, Kiel, Germany
- Department of Internal Medicine I, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Stefan Störk
- Department of Clinical Research and Epidemiology, Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
- Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany
| | - Jens-Peter Reese
- Institute of Clinical Epidemiology and Biometry, University of Würzburg, Würzburg, Germany
- Institute of Medical Data Science, University Hospital Würzburg, Würzburg, Germany
- Faculty of Health Sciences, THM University of Applied Sciences, Gießen, Germany
| | - Thomas Zoller
- Department of Infectious Diseases, Respiratory and Critical Care Medicine, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Lilian Krist
- Institute of Social Medicine, Epidemiology and Health Economics, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - David Ellinghaus
- Institute of Clinical Molecular Biology, University Medical Center Schleswig-Holstein, Kiel University, Kiel, Germany
| | - Bärbel U Foesel
- Institute of Epidemiology, Research Unit of Molecular Epidemiology, Helmholtz Munich - German Research Center for Environmental Health, Neuherberg, Germany
| | - Christian Gieger
- Institute of Epidemiology, Research Unit of Molecular Epidemiology, Helmholtz Munich - German Research Center for Environmental Health, Neuherberg, Germany
| | - Bettina Lorenz-Depiereux
- Institute of Epidemiology, Research Unit of Molecular Epidemiology, Helmholtz Munich - German Research Center for Environmental Health, Neuherberg, Germany
| | - Martin Witzenrath
- Department of Infectious Diseases and Critical Care Medicine, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- CAPNETZ Stiftung, Hannover, Germany
| | - Gabriele Anton
- Medical School OWL, Bielefeld University, Bielefeld, Germany
| | - Michael Krawczak
- Institute of Medical Informatics and Statistics, University Medical Center Schleswig-Holstein, Kiel University, Brunswiker Straße 10, 24113, Kiel, Germany.
| | - Jan Heyckendorf
- Department of Internal Medicine I, University Medical Center Schleswig-Holstein, Kiel, Germany
- Airway Research Center North (ARCN), German Center for Lung Research (DZL), Großhansdorf, Germany
- Leibniz Lung Clinic, Kiel, Germany
| | - Thomas Bahmer
- Department of Internal Medicine I, University Medical Center Schleswig-Holstein, Kiel, Germany
- Airway Research Center North (ARCN), German Center for Lung Research (DZL), Großhansdorf, Germany
| |
Collapse
|
10
|
Mayerhöfer T, Joannidis M, Klein S, Franke A, Margarita S, Ronzoni L, Pertler E, Wagner S, Sahanic S, Tancevski I, Haschka D, Hochhold C, Treml B, Valenti L, Tilg H, Schaefer B, Zoller H. The common genetic variant rs1278960 determining expression of Interferon-lambda predicts inflammatory response in critically ill COVID-19 patients. Sci Rep 2025; 15:15802. [PMID: 40328868 PMCID: PMC12056047 DOI: 10.1038/s41598-025-91628-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 02/21/2025] [Indexed: 05/08/2025] Open
Abstract
The single nucleotide polymorphism rs12979860 is associated with the production of IFNλ4, a type III interferon, which offers protection from viral infection via its proinflammatory properties. We investigated if a genetically determined increase in IFNλ4 affects disease progression in SARS-CoV-2. This prospective, single-center study involved critically ill SARS-CoV-2 patients admitted to the intensive care unit. We performed genotyping for rs12979860 and analyzed daily laboratory data. Genotype frequencies were compared with an external validation cohort. Critically ill individuals with COVID-19 (n = 184; 29.3% women) were included. Median age was 63 years. The TT genotype was present in 11%, CT in 48% and CC in 41%. At baseline, CRP, ferritin, transferrin and neopterin did not differ significantly between groups. Longitudinal analysis revealed significant genotype-dependent differences in CRP, ferritin and neopterin with the highest peak in TT patients after 10-15 days. A higher need for renal replacement therapy (31.6% vs. 11.7%, p = 0.044) and mechanical ventilation (22 days vs. 15 days, p = 0.018) was observed in the TT group. The SNP rs12979860 near IFNL4 is associated with distinct inflammatory trajectories in critically ill COVID-19 patients. Genetic determinants of the immune response influence the severity of inflammation and clinical outcomes in severe COVID-19.
Collapse
Affiliation(s)
- Timo Mayerhöfer
- Department of Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
- Division of Intensive Care and Emergency Medicine, Department of Internal Medicine, Medical University Innsbruck, Innsbruck, Austria
| | - Michael Joannidis
- Division of Intensive Care and Emergency Medicine, Department of Internal Medicine, Medical University Innsbruck, Innsbruck, Austria
| | - Sebastian Klein
- Division of Intensive Care and Emergency Medicine, Department of Internal Medicine, Medical University Innsbruck, Innsbruck, Austria
- Karl Landsteiner University of Health Sciences, Krems, Austria
- Department of Internal Medicine 2, University Hospital St. Pölten, St. Pölten, Austria
| | - Andre Franke
- Institute of Clinical Molecular Biology, Kiel University and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Sara Margarita
- Precision Medicine Lab, Biological Resource Center - Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Luisa Ronzoni
- Precision Medicine Lab, Biological Resource Center - Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Elke Pertler
- Department of Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
- Christian Doppler Laboratory for Iron and Phosphate Biology, Medical University of Innsbruck, Innsbruck, Austria
| | - Sonja Wagner
- Department of Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
- Christian Doppler Laboratory for Iron and Phosphate Biology, Medical University of Innsbruck, Innsbruck, Austria
| | - Sabina Sahanic
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Ivan Tancevski
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - David Haschka
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Christoph Hochhold
- Department of Anesthesiology and Intensive Care Medicine, Medical University Innsbruck, Innsbruck, Austria
| | - Benedikt Treml
- Department of Anesthesiology and Intensive Care Medicine, Medical University Innsbruck, Innsbruck, Austria
| | - Luca Valenti
- Precision Medicine Lab, Biological Resource Center - Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Herbert Tilg
- Department of Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Benedikt Schaefer
- Department of Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria.
| | - Heinz Zoller
- Department of Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
- Christian Doppler Laboratory for Iron and Phosphate Biology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
11
|
Chen S, Liang Y, Mo JMY, Li QHY, He B, Luo S, Burgess S, Au Yeung SL. Challenges in interpreting Mendelian randomization studies with a disease as the exposure: Using COVID-19 liability studies as an exemplar. Eur J Hum Genet 2025; 33:658-665. [PMID: 40164729 PMCID: PMC12048694 DOI: 10.1038/s41431-025-01840-x] [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: 07/20/2024] [Revised: 02/14/2025] [Accepted: 03/19/2025] [Indexed: 04/02/2025] Open
Abstract
Mendelian randomization (MR) studies using diseases as exposures are increasingly prevalent although any observed associations do not necessarily imply effect of diseases. To illustrate this challenge, we conducted a systematic review of MR studies focusing on COVID-19 consequence. We hypothesized if outcome genome-wide association studies (GWAS) were conducted before COVID-19 pandemic in late 2019, any observed associations in these studies were unlikely to be driven by COVID-19. We systematically searched PubMed, EMBASE, and MEDLINE for all MR studies published between 1 January 2019 and 20 May 2023. Inclusion criteria included MR studies which used COVID-19 as the exposure and designed to assess COVID-19's impact on health outcomes. We extracted relevant information, such as result interpretation and relevance assumption assessment. This review was registered at PROSPERO (CRD42023421079). Amongst 57 included studies, 45 studies used outcome GWAS published prior to 2019 whilst the remaining studies likely used outcome GWAS containing data collected before 2019. Relevance assumption was assessed mainly by p values. A total of 35 studies showed an association of COVID-19 liability with health outcomes. Regardless of the results, 45 studies attributed these as evidence (or lack of evidence) of COVID-19 consequence. In MR studies using disease liability as exposure, relevance assumption should consider the prevalence of the disease in the outcome GWAS in the context of 2 sample Mendelian randomization study rather than p values/F-statistic alone. Even when these are verified, these studies likely suffered from pleiotropy, making corresponding interpretation as effect of disease challenging.
Collapse
Affiliation(s)
- Siyu Chen
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong, China
| | - Ying Liang
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong, China
| | - Jacky Man Yuen Mo
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong, China
| | - Queenie Ho Yi Li
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong, China
| | - Baoting He
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong, China
| | - Shan Luo
- Department of Family Medicine and Primary Care, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong, China
| | - Stephen Burgess
- MRC Biostatistics Unit, Cambridge Institute of Public Health, University of Cambridge, Cambridge, United Kingdom
| | - Shiu Lun Au Yeung
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong, China.
| |
Collapse
|
12
|
Tsampasian V, Bäck M, Bernardi M, Cavarretta E, Dębski M, Gati S, Hansen D, Kränkel N, Koskinas KC, Niebauer J, Spadafora L, Frias Vargas M, Biondi-Zoccai G, Vassiliou VS. Cardiovascular disease as part of Long COVID: a systematic review. Eur J Prev Cardiol 2025; 32:485-498. [PMID: 38381595 DOI: 10.1093/eurjpc/zwae070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 02/01/2024] [Accepted: 02/17/2024] [Indexed: 02/23/2024]
Abstract
AIMS Long COVID syndrome has had a major impact on million patients' lives worldwide. The cardiovascular system is an important aspect of this multifaceted disease that may manifest in many ways. We have hereby performed a narrative review in order to identify the extent of the cardiovascular manifestations of the Long COVID syndrome. METHODS AND RESULTS An in-depth systematic search of the literature has been conducted for this narrative review. The systematic search of PubMed and Cochrane databases yielded 3993 articles, of which 629 underwent full-text screening. A total of 78 studies were included in the final qualitative synthesis and data evaluation. The pathophysiology of the cardiovascular sequelae of Long COVID syndrome and the cardiac manifestations and complications of Long COVID syndrome are critically evaluated. In addition, potential cardiovascular risk factors are assessed, and preventive methods and treatment options are examined in this review. CONCLUSION This systematic review poignantly summarizes the evidence from the available literature regarding the cardiovascular manifestations of Long COVID syndrome and reviews potential mechanistic pathways, diagnostic approaches, preventive measures, and treatment options.
Collapse
Affiliation(s)
| | - Maria Bäck
- Department of Molecular and Clinical Medicine, Sahlgrenska Academy, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
- Department of Medical and Health Sciences, Division of Physiotherapy, Linköping University, Linköping, Sweden
| | - Marco Bernardi
- Department of Clinical, Anesthesiology and Cardiovascular Sciences, Internal Medicine, Sapienza University of Rome, Rome, Italy
| | - Elena Cavarretta
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
- Mediterranea Cardiocentro, Naples, Italy
| | - Maciej Dębski
- Norwich Medical School, University of East Anglia, Norwich NR4 7TJ, UK
| | - Sabiha Gati
- Royal Brompton Hospital, UK and Imperial College London, London, UK
| | - Dominique Hansen
- Heart Centre Hasselt, Jessa Hospital, Hasselt, Belgium
- REVAL/BIOMED (Rehabilitation Research Centre), Hasselt University, Hasselt, Belgium
| | - Nicolle Kränkel
- DZHK (German Centre for Cardiovascular Research), Partner site Berlin, Germany
- Friede Springer, Centre of Cardiovascular Prevention at Charité, Charité, University Medicine Berlin, Berlin, Germany
- Deutsches Herzzentrum der Charité, Klinik für Kardiologie, Angiologie und Intensivmedizin, Campus Benjamin-Franklin (CBF), Charité University Medicine Berlin, 12203 Berlin, Germany
| | - Konstantinos C Koskinas
- Department of Cardiology, Bern University Hospital-INSELSPITAL, University of Bern, Bern, Switzerland
| | - Josef Niebauer
- University Institute of Sports Medicine, Prevention and Rehabilitation and Research Institute of Molecular Sports Medicine and Rehabilitation, Paracelsus Medical University, Salzburg, Austria
| | - Luigi Spadafora
- Department of Clinical, Anesthesiology and Cardiovascular Sciences, Internal Medicine, Sapienza University of Rome, Rome, Italy
| | - Manuel Frias Vargas
- Department of Medicine, Faculty of Medicine, Complutense University of Madrid, Madrid, Spain
- San Andres Primary Care Health Centre, Madrid, Spain
| | - Giuseppe Biondi-Zoccai
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
- Mediterranea Cardiocentro, Naples, Italy
| | - Vassilios S Vassiliou
- Norwich Medical School, University of East Anglia, Norwich NR4 7TJ, UK
- Department of Cardiology, Norfolk and Norwich University Hospital, Colney Lane, Norwich NR4 7UY, UK
| |
Collapse
|
13
|
Kleist AB, Szpakowska M, Talbot LJ, Slodkowicz G, Malinverni D, Thomas MA, Crawford KS, McGrail DJ, Dishman AF, Wedemeyer MJ, Sluter M, Yi SS, Sahni N, Peterson FC, Chevigné A, Volkman BF, Babu MM. Encoding and decoding selectivity and promiscuity in the human chemokine-GPCR interaction network. Cell 2025:S0092-8674(25)00398-8. [PMID: 40273912 DOI: 10.1016/j.cell.2025.03.046] [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: 05/26/2020] [Revised: 07/23/2024] [Accepted: 03/28/2025] [Indexed: 04/26/2025]
Abstract
In humans, selective and promiscuous interactions between 46 secreted chemokine ligands and 23 cell surface chemokine receptors of the G-protein-coupled receptor (GPCR) family form a complex network to coordinate cell migration. While chemokines and their GPCRs each share common structural scaffolds, the molecular principles driving selectivity and promiscuity remain elusive. Here, we identify conserved, semi-conserved, and variable determinants (i.e., recognition elements) that are encoded and decoded by chemokines and their receptors to mediate interactions. Selectivity and promiscuity emerge from an ensemble of generalized ("public/conserved") and specific ("private/variable") determinants distributed among structured and unstructured protein regions, with ligands and receptors recognizing these determinants combinatorially. We employ these principles to engineer a viral chemokine with altered GPCR coupling preferences and provide a web resource to facilitate sequence-structure-function studies and protein design efforts for developing immuno-therapeutics and cell therapies.
Collapse
Affiliation(s)
- Andrew B Kleist
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA; Medical Scientist Training Program, Medical College of Wisconsin, Milwaukee, WI, USA; MRC Laboratory of Molecular Biology, Cambridge, UK.
| | - Martyna Szpakowska
- Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Lindsay J Talbot
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN, USA; Department of Surgery, St. Jude Children's Research Hospital, Memphis, TN, USA
| | | | - Duccio Malinverni
- MRC Laboratory of Molecular Biology, Cambridge, UK; Center of Excellence for Data-Driven Discovery, Department of Structural Biology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Monica A Thomas
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA; Medical Scientist Training Program, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Kyler S Crawford
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA; Medical Scientist Training Program, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Daniel J McGrail
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Acacia F Dishman
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA; Medical Scientist Training Program, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Michael J Wedemeyer
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Madison Sluter
- Center of Excellence for Data-Driven Discovery, Department of Structural Biology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - S Stephen Yi
- Department of Oncology, Dell Medical School, University of Texas at Austin, Austin, TX, USA; Department of Biomedical Engineering, Cockrell School of Engineering, University of Texas at Austin, Austin, TX, USA
| | - Nidhi Sahni
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA; Program in Quantitative and Computational Biosciences, Baylor College of Medicine, Houston, TX, USA; Department of Epigenetics & Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Francis C Peterson
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA; Protein Foundry, LLC, West Allis, WI, USA; Program in Chemical Biology, Medical College of Wisconsin, Milwaukee, WI, USA; Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Andy Chevigné
- Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Brian F Volkman
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA; Protein Foundry, LLC, West Allis, WI, USA; Program in Chemical Biology, Medical College of Wisconsin, Milwaukee, WI, USA; Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, USA.
| | - M Madan Babu
- MRC Laboratory of Molecular Biology, Cambridge, UK; Center of Excellence for Data-Driven Discovery, Department of Structural Biology, St Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
14
|
Dalapati T, Wang L, Jones AG, Cardwell J, Konigsberg IR, Bossé Y, Sin DD, Timens W, Hao K, Yang I, Ko DC. Context-specific eQTLs provide deeper insight into causal genes underlying shared genetic architecture of COVID-19 and idiopathic pulmonary fibrosis. HGG ADVANCES 2025; 6:100410. [PMID: 39876559 PMCID: PMC11872446 DOI: 10.1016/j.xhgg.2025.100410] [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: 07/15/2024] [Revised: 01/22/2025] [Accepted: 01/22/2025] [Indexed: 01/30/2025] Open
Abstract
Most genetic variants identified through genome-wide association studies (GWASs) are suspected to be regulatory in nature, but only a small fraction colocalize with expression quantitative trait loci (eQTLs, variants associated with expression of a gene). Therefore, it is hypothesized but largely untested that integration of disease GWAS with context-specific eQTLs will reveal the underlying genes driving disease associations. We used colocalization and transcriptomic analyses to identify shared genetic variants and likely causal genes associated with critically ill COVID-19 and idiopathic pulmonary fibrosis. We first identified five genome-wide significant variants associated with both diseases. Four of the variants did not demonstrate clear colocalization between GWAS and healthy lung eQTL signals. Instead, two of the four variants colocalized only in cell type- and disease-specific eQTL datasets. These analyses pointed to higher ATP11A expression from the C allele of rs12585036, in monocytes and in lung tissue from primarily smokers, which increased risk of idiopathic pulmonary fibrosis (IPF) and decreased risk of critically ill COVID-19. We also found lower DPP9 expression (and higher methylation at a specific CpG) from the G allele of rs12610495, acting in fibroblasts and in IPF lungs, and increased risk of IPF and critically ill COVID-19. We further found differential expression of the identified causal genes in diseased lungs when compared to non-diseased lungs, specifically in epithelial and immune cell types. These findings highlight the power of integrating GWASs, context-specific eQTLs, and transcriptomics of diseased tissue to harness human genetic variation to identify causal genes and where they function during multiple diseases.
Collapse
Affiliation(s)
- Trisha Dalapati
- Medical Scientist Training Program, Duke University School of Medicine, Durham, NC, USA; Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
| | - Liuyang Wang
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
| | - Angela G Jones
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA; University Program in Genetics and Genomics, Duke University, Durham, NC, USA
| | - Jonathan Cardwell
- Department of Biomedical Informatics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Iain R Konigsberg
- Department of Biomedical Informatics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Yohan Bossé
- Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval, Department of Molecular Medicine, Québec City, QC, Canada
| | - Don D Sin
- Center for Heart Lung Innovation, University of British Columbia and St. Paul's Hospital, Vancouver, BC, Canada
| | - Wim Timens
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Ke Hao
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Ivana Yang
- Department of Biomedical Informatics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Dennis C Ko
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA; University Program in Genetics and Genomics, Duke University, Durham, NC, USA; Division of Infectious Diseases, Department of Medicine, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
15
|
Stukas S, Goshua G, Conway EM, Lee AYY, Hoiland RL, Sekhon MS, Y. C. Chen L. ABO blood group and COVID-19 severity: Associations with endothelial and adipocyte activation in critically ill patients. PLoS One 2025; 20:e0320251. [PMID: 40173171 PMCID: PMC11964209 DOI: 10.1371/journal.pone.0320251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 02/14/2025] [Indexed: 04/04/2025] Open
Abstract
BACKGROUND ABO blood group has been implicated both in susceptibility to, and severity of, SARS-CoV-2 infection. The aim of this study was to explore a potential association between ABO blood group and severity of COVID-19 infection in critically ill patients and the following biological mechanisms: inflammatory cytokines, endothelial injury, and adipokines. METHODS We conducted a retrospective study of 128 critically ill COVID-19 patients admitted to Vancouver General Hospital from March 2020-March 2021. Outcomes including 28-day mortality, need for mechanical ventilation and length of intensive care unit (ICU) stay were compared between patients with A & AB blood type vs. B & O blood type. Likewise, serum inflammatory markers, markers of endothelial activation, and adipokines were compared. RESULTS The association between ABO and severity of disease was confirmed. Patients with A&AB blood group had more frequent ventilation requirements compared to patients with blood group B&O (N(%): 35 (71%) vs 41 (52%), p = 0.041), higher total ICU mortality (14 (29%) vs 9 (11%), p = 0.018), longer median ICU stay (days, median [interquartile range]: 10 [6-19], vs 7 [3-14], p = 0.016) and longer median hospital stay (26 [14-36] vs. 17 [10-30] p = 0.034). No association was found between ABO blood group and serum inflammatory cytokines or their receptors [IL-6, IL-1b, IL-10, TNF, sIL-6R, sgp130] measured within the first 10 days of ICU stay. No association was found between ABO and plasma markers of endothelial injury [Thrombomodulin, ADAMTS13, sP-Selectin, Factor IX, Protein C, Protein S, vWF]. Among the plasma adipokines, there were no differences between lipocalin-2, PAI-1 or resistin. Notably, however, median adipsin was higher in patients with A&AB blood group compared to O&B (16.3 [4.2-38.5] x106 pg/mL vs. 9.61 [3.0-20.8] x 106 pg/mL, p = 0.048). CONCLUSIONS This retrospective single-center study confirms an association between A and AB blood type with more severe COVID-19. While an underlying mechanism was not identified, the finding of higher adipsin levels in patients with type A/AB blood warrants further investigation in larger prospective studies.
Collapse
Affiliation(s)
- Sophie Stukas
- Department of Pathology and Laboratory Medicine, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - George Goshua
- Department of Internal Medicine, Section of Medical Oncology & Hematology, Yale School of Medicine and Yale Cancer Center, New Haven, Connecticut, United States of America
- Center for Outcomes Research and Evaluation, Yale New Haven Hospital, New Haven, Connecticut, United States of America
| | - Edward M. Conway
- Department of Medicine, Centre for Blood Research, University of British Columbia, Vancouver, British Columbia, Canada
| | - Agnes Y. Y. Lee
- Division of Hematology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ryan L. Hoiland
- Department of Cellular and Physiological Sciences, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Centre for Chronic Disease Prevention and Management, University of British Columbia, Kelowna, British Columbia, Canada
| | - Mypinder S. Sekhon
- Division of Critical Care, University of British Columbia, Vancouver, British Columbia, Canada
- Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Luke Y. C. Chen
- Division of Hematology, Dalhousie University, Halifax, Nova Scotia, Canada
- Division of Hematology, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
16
|
Jacob IB, Lawal AO, Mahmoud SS, Kopsack EM, Reynolds ES, Meng Q, Fan H, Massa PT, Thangamani S, Jia H, Wang G. Differential immunoregulation by human surfactant protein A variants determines severity of SARS-CoV-2-induced lung disease. Front Immunol 2025; 16:1462278. [PMID: 40242753 PMCID: PMC12000003 DOI: 10.3389/fimmu.2025.1462278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 03/10/2025] [Indexed: 04/18/2025] Open
Abstract
Introduction COVID-19 remains a significant threat to public health globally. Infection in some susceptible individuals causes life-threatening acute lung injury (ALI/ARDS) and/or death. Human surfactant protein A (SP-A) is a C-type lectin expressed in the lung and other mucosal tissues, and it plays a critical role in host defense against various pathogens. The human SP-A genes (SFTPA1 and SFTPA2) are highly polymorphic and comprise several common genetic variants, i.e., SP-A1 (variants 6A2, 6A4) and SP-A2 (variants 1A0, 1A3). Here, we elucidated the differential antiviral and immunoregulatory roles of SP-A variants in response to SARS-CoV-2 infection in vivo. Methods Six genetically-modified mouse lines, expressing both hACE2 (SARS-CoV-2 receptor) and individual SP-A variants: (hACE2/6A2 (6A2), hACE2/6A4 (6A4), hACE2/1A0 (1A0), and hACE2/1A3 (1A3), one SP-A knockout (hACE2/SP-A KO (KO) and one hACE2/mouse SP-A (K18) mice, were challenged intranasally with 103 PFU SARS-CoV-2 or MEM medium (Sham). Results Infected KO and 1A0 mice had more weight loss and mortality compared to other mouse lines. Relative to other infected mouse lines, a more severe ALI was observed in KO, 1A0, and 6A2 mice. Reduced viral titers were generally observed in the lungs of infected SP-A mice relative to KO mice. Transcriptomic analysis revealed an upregulation in genes that play central roles in immune responses such as MyD88, Stat3, IL-18, and Jak2 in the lungs of KO and 1A0 mice. However, Mapk1 was significantly downregulated in 6A2 versus 1A0 mice. Analysis of biological pathways identified those involved in lung host defense and innate immunity, including pathogen-induced cytokine, NOD1/2, and Trem1 signaling pathways. Consistent with the transcriptomic data, levels of cytokines and chemokines such as G-CSF, IL-6, and IL-1β were comparatively higher in the lungs and sera of KO and 1A0 mice with the highest mortality rate. Furthermore, we observed the complexity of COVID-19, such as the difference between lung and systemic immune response to viral infection and of viral load and mortality among SP-A variants in this model. Conclusion These findings demonstrate that human SP-A variants differentially modulate SARS-CoV-2-induced lung injury and disease severity by differentially inhibiting viral infectivity and regulating immune-related gene expressions.
Collapse
Affiliation(s)
- Ikechukwu B. Jacob
- Department of Surgery, The State University of New York (SUNY) Upstate Medical University, Syracuse, NY, United States
- Department of Microbiology and Immunology, The State University of New York (SUNY) Upstate Medical University, Syracuse, NY, United States
| | - Akinkunmi O. Lawal
- Department of Surgery, The State University of New York (SUNY) Upstate Medical University, Syracuse, NY, United States
- Department of Microbiology and Immunology, The State University of New York (SUNY) Upstate Medical University, Syracuse, NY, United States
| | - Salma S. Mahmoud
- Department of Surgery, The State University of New York (SUNY) Upstate Medical University, Syracuse, NY, United States
| | - Emerson M. Kopsack
- Department of Surgery, The State University of New York (SUNY) Upstate Medical University, Syracuse, NY, United States
| | - Erin S. Reynolds
- Department of Microbiology and Immunology, The State University of New York (SUNY) Upstate Medical University, Syracuse, NY, United States
| | - Qinghe Meng
- Department of Surgery, The State University of New York (SUNY) Upstate Medical University, Syracuse, NY, United States
| | - Hongkuan Fan
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Paul T. Massa
- Department of Microbiology and Immunology, The State University of New York (SUNY) Upstate Medical University, Syracuse, NY, United States
- Department of Neurology, The State University of New York (SUNY) Upstate Medical University, Syracuse, NY, United States
| | - Saravanan Thangamani
- Department of Microbiology and Immunology, The State University of New York (SUNY) Upstate Medical University, Syracuse, NY, United States
| | - Hongpeng Jia
- Department of Surgery, Johns-Hopkins University, Baltimore, MD, United States
| | - Guirong Wang
- Department of Surgery, The State University of New York (SUNY) Upstate Medical University, Syracuse, NY, United States
- Department of Microbiology and Immunology, The State University of New York (SUNY) Upstate Medical University, Syracuse, NY, United States
| |
Collapse
|
17
|
Leyva H, Hamade N, Krishnathasan D, Goldhaber SZ, Welker J, Carroll B, Bonaca M, Lan Z, Bikdeli B, Piazza G. Non-O Blood Groups and Cardiovascular Outcomes in COVID-19: Findings From a Large Multicenter Study. JACC. ADVANCES 2025; 4:101641. [PMID: 40080920 PMCID: PMC11953960 DOI: 10.1016/j.jacadv.2025.101641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 01/31/2025] [Accepted: 02/11/2025] [Indexed: 03/15/2025]
Affiliation(s)
- Hannah Leyva
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Thrombosis Research Group, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Nada Hamade
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Thrombosis Research Group, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Darsiya Krishnathasan
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Thrombosis Research Group, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Samuel Z Goldhaber
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Thrombosis Research Group, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - James Welker
- Anne Arundel Research Institute, Annapolis, Mississippi, USA
| | - Brett Carroll
- Division of Cardiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Marc Bonaca
- CPC Clinical Research, Aurora, Colorado, USA; Department of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Zhou Lan
- Thrombosis Research Group, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Center for Clinical Investigation, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Behnood Bikdeli
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Thrombosis Research Group, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; YNHH/Yale Center for Outcomes Research and Evaluation (CORE), New Haven, Connecticut, USA
| | - Gregory Piazza
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Thrombosis Research Group, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
18
|
Odoom PN, Okoh OS, Asare YY, Mac-Arthur CO, Azumah JD, Mensah A, Yalley AK, Sagoe KW, Nii-Trebi NI. Blood type susceptibility to SARS-CoV-2 at a tertiary hospital in Accra, Ghana. Microbiol Spectr 2025; 13:e0110824. [PMID: 40130860 PMCID: PMC12053907 DOI: 10.1128/spectrum.01108-24] [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: 05/02/2024] [Accepted: 02/24/2025] [Indexed: 03/26/2025] Open
Abstract
Pandemics from viral outbreaks, such as that caused by SARS-CoV-2, have significant impacts worldwide. The factors that underlie differential susceptibility to severe COVID-19 outcomes are not fully understood. The role of the ABO blood group in the outcome of SARS-CoV-2 infections remains to be clarified in different populations. This study described the SARS-CoV-2 seroprevalence and examined the association of the ABO blood group with COVID-19 disease among apparently healthy and COVID-19 patients at the Korle Bu Teaching Hospital, Accra. The study involved 277 participants comprising 200 healthy individuals and 77 PCR-confirmed COVID-19 patients with mild or severe symptoms. Anti-SARS-CoV-2 antibody assay (IgM/IgG) was performed, and ABO blood grouping was done on plasma samples using the reverse blood grouping method. Statistical analyses were performed in R for the association of socio-demographic parameters and ABO blood groupings of participants with SARS-CoV-2 infection status. The total SARS-CoV-2 seropositivity was 61.4% (157/277). Most of the participants (245/277, 88.4%) were unvaccinated. Of the 245 unvaccinated individuals, 127 (51.8%) were IgG reactive. A significant association was observed between ABO blood group and COVID-19 disease status. Antigen A participants had a higher probability of symptomatic infection than non-antigen A individuals. Blood group O appeared more protective than other blood types among the participants. Seropositivity was high among the participants studied-vaccinated and unvaccinated. Blood group A is associated with an increased risk of COVID-19, whereas blood group O appears protective. Further studies involving larger sample sizes are required to confirm these findings. IMPORTANCE The transmissibility and virulence of SARS-CoV-2 and the severity of COVID-19 disease appeared to vary across nations and among populations. However, the factors that account for the differential susceptibility and COVID-19 outcomes are not well understood. The roles of host immune defense mechanisms and genetic makeup have been implicated. This study investigated the seroprevalence of anti-SARS-CoV-2 antibodies (IgM and IgG) in apparently healthy individuals and COVID-19 patients; using a reliable but inexpensive blood group typing based on direct hemagglutination technique and rigorous statistical analyses, we determined the association of ABO blood groups with COVID-19 disease. We found appreciably high seropositivity among the participants studied-both vaccinated and unvaccinated-and showed that blood type significantly influences SARS-CoV-2 infection and COVID-19 severity, with blood group A associated with severe COVID-19 disease, whereas blood group O appears protective. Further studies involving a larger sample size are required to confirm these findings.
Collapse
Affiliation(s)
- Prince N. Odoom
- Department of Medical Microbiology, University of Ghana Medical School, Accra, Ghana
| | - Olayinka S. Okoh
- Department of Chemical Sciences, Anchor University, Lagos, Nigeria
- Anchor University Centre for Global Health, Lagos, Nigeria
| | - Yaa Y. Asare
- Department of Biochemistry and Biotechnology, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Clara O. Mac-Arthur
- Department of Medical Laboratory Sciences, School of Biomedical and Allied Health Sciences, University of Ghana, Accra, Ghana
| | - Judith D. Azumah
- Department of Medical Microbiology, University of Ghana Medical School, Accra, Ghana
| | - Albert Mensah
- Department of Medical Microbiology, University of Ghana Medical School, Accra, Ghana
- Holy Child Catholic Hospital, Sekondi-Takoradi, Ghana
| | - Akua K. Yalley
- Department of Medical Laboratory Sciences, School of Biomedical and Allied Health Sciences, University of Ghana, Accra, Ghana
| | - Kwamena W. Sagoe
- Department of Medical Microbiology, University of Ghana Medical School, Accra, Ghana
| | - Nicholas I. Nii-Trebi
- Department of Medical Laboratory Sciences, School of Biomedical and Allied Health Sciences, University of Ghana, Accra, Ghana
| |
Collapse
|
19
|
Kalantar Neyestanaki MH, Gholizadeh O, Yasamineh S, Tarahomi M, Pooya P, Eslami M, Dadashpour M, Ghaffari H. Investigating the relationship between cycle threshold of SARS-CoV-2 RT-PCR, clinical features, and laboratory data in hospitalized COVID-19 patients in Semnan, Iran. Front Cell Infect Microbiol 2025; 15:1522375. [PMID: 40196044 PMCID: PMC11973279 DOI: 10.3389/fcimb.2025.1522375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 02/28/2025] [Indexed: 04/09/2025] Open
Abstract
Introduction COVID-19, caused by the SARS-CoV-2 virus, has emerged as a global public health crisis. Understanding the factors associated with disease severity and outcomes is crucial for effective patient management. This study aimed to investigate the association between cycle threshold (CT) values, demographic data, medical history, clinical manifestations, and laboratory findings in hospitalized COVID-19 patients in Semnan, Iran. Methods A cross-sectional study was conducted on 86 patients with confirmed COVID-19 admitted to two hospitals in Semnan, Iran, between December 2022 and March 2023. Respiratory swab samples were collected RT-PCR was performed, CT values were obtained, and data were collected from medical records, including demographic information, medical history, clinical manifestations, and laboratory results. Statistical analysis was performed using SPSS software. Results The study included 86 COVID-19 patients, with a slightly higher representation of females (55.8%) and a mean age of 67.43 years. Pre-existing conditions like hypertension, diabetes mellitus, and ischemic heart disease were prevalent among hospitalized patients. A majority of patients (59.3%) had severe COVID-19, as indicated by lower CT values, while 31.4% exhibited oxygen saturation levels below 90%. Significant differences were observed in FBS, CRP, WBC, Hb, Cr, and SPo2 levels between severe and non-severe patients. Correlation analysis revealed associations between age, CRP, Cr, BUN, FBS, Vitamin D, TG, LDL, HDL, AST, ALP, and SPo2. Reflecting complex interactions between inflammatory markers, organ function, and lipid metabolism in COVID-19 patients. Conclusion This study provides valuable insights into the association between CT values, clinical characteristics, and laboratory findings in hospitalized COVID-19 patients. The findings underscore the importance of CT values in assessing disease severity and potential prognostication. Further research is warranted to validate these findings in larger and more diverse patient populations.
Collapse
Affiliation(s)
| | - Omid Gholizadeh
- Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran, Iran
| | - Saman Yasamineh
- Young Researchers and Elite Club, Islamic Azad University, Tabriz, Iran
| | - Mahdieh Tarahomi
- Student Research Committee, Semnan University of Medical Sciences, Semnan, Iran
| | - Pegah Pooya
- Department of Molecular Virology, Semnan Health Reference Laboratory, Semnan University of Medical Sciences, Semnan, Iran
| | - Majid Eslami
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Department of Bacteriology and Virology, Semnan University of Medical Sciences, Semnan, Iran
| | - Mehdi Dadashpour
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Hadi Ghaffari
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Department of Bacteriology and Virology, Semnan University of Medical Sciences, Semnan, Iran
| |
Collapse
|
20
|
Narin Çopur E, Ergün D, Ergün R, Atik S, Türk Dağı H, Körez MK. Risk Factors Affecting the Severity, Mortality, and Intensive Care Unit Admission of COVID-19 Patients: A Series of 1075 Cases. Viruses 2025; 17:429. [PMID: 40143356 PMCID: PMC11946003 DOI: 10.3390/v17030429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 03/11/2025] [Accepted: 03/15/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND The clinical spectrum of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is broad; it can range from asymptomatic cases to mild upper respiratory tract illness, respiratory failure, and severe multiorgan failure resulting in death. Therefore, it is important to identify the clinical course of the disease and the factors associated with mortality. OBJECTIVE The aim of this study is to identify the risk factors associated with the severity of the disease, intensive care unit admission, and mortality in COVID-19 patients. METHODS A total of 1075 patients with clinical and radiological findings compatible with COVID-19 pneumonia and positive SARS-CoV-2 PCR were selected and retrospectively screened. All included patients were classified according to the 7th edition of the 2019 Coronavirus Disease Guidelines published by the National Health Commission of China. RESULTS It was observed that elevated white blood count (WBC) increased the severity of COVID-19 by 3.26 times and the risk of intensive care unit (ICU) admission by 3.47 times. Patients with high D-dimer levels had a 91% increased risk, and those with high fibrinogen levels had a 2.08 times higher risk of severe disease. High C-reactive protein (CRP) values were found to increase disease severity by 6.89 times, mortality by 12.84 times, and ICU admission by 3.37 times. CONCLUSIONS Identifying the factors associated with disease severity, ICU admission, and mortality in COVID-19 patients could help reduce disability and mortality rates in pandemics.
Collapse
Affiliation(s)
- Ecem Narin Çopur
- Department of Pulmonary Medicine, Dr. Yaşar Eryılmaz Doğubeyazıt State Hospital, Ağrı 04402, Turkey
| | - Dilek Ergün
- Department of Pulmonary Medicine, Faculty of Medicine, Selcuk University, Konya 42130, Turkey;
| | - Recai Ergün
- Department of Pulmonary Medicine, Faculty of Medicine, Selcuk University, Konya 42130, Turkey;
| | - Serap Atik
- Department of Pulmonary Medicine, Iğdır Dr. Nevruz Erez State Hospital, Iğdır 76000, Turkey;
| | - Hatice Türk Dağı
- Department of Medical Microbiology, Faculty of Medicine, Selcuk University, Konya 42130, Turkey;
| | - Muslu Kazım Körez
- Department of Biostatistics, Faculty of Medicine, Selcuk University, Konya 42130, Turkey;
| |
Collapse
|
21
|
İdikut A, Değer İ, Göktaş G, Karahan S, Sarınç S, Köksal D, Babaoğlu MO, Babaoğlu E. Association of Endothelial Nitric Oxide Synthase Polymorphisms with Clinical Severity in Patients with COVID-19. J Clin Med 2025; 14:1931. [PMID: 40142738 PMCID: PMC11943162 DOI: 10.3390/jcm14061931] [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: 01/13/2025] [Revised: 02/17/2025] [Accepted: 03/10/2025] [Indexed: 03/28/2025] Open
Abstract
Background/Objectives: To elucidate the factors that contribute to individual variability in the progression of COVID-19, experiments on endothelial nitric oxide synthase polymorphisms have been reported. Nitric oxide synthase (NOS3) is located in the endothelium and is involved in the regulation of inflammation and vascular homeostasis. In this study, we investigated the association between COVID-19 severity and NOS3 G894T and NOS3 27-bp VNTR 4b/a genetic polymorphisms. Methods: Patients with COVID-19 (n = 178) were divided into Group 1 (mild disease) and Group 2 (severe disease) based on oxygen saturation levels in room air (Group 1, SpO2 ≥ 93%, n = 107; and Group 2, SpO2 < 93%, n = 73) and hospitalization requirements. Genotyping was performed using polymerase chain reaction-restriction fragment length polymorphism analysis. Results: Overall, genotype and allele frequencies of the NOS3 genetic polymorphisms were similar across the two study groups (p > 0.05). However, the subgroup analysis showed a notable trend for the 4b/4a allele distribution between Groups 1 and 2. In the younger subgroup of patients (≤50 years old) without chronic obstructive pulmonary disease, Group 2 tended to have a higher frequency of the 4b allele than Group 1 (97.4% vs. 85.4% p = 0.06) and a higher occurrence of 4b/4b genotype (94.7% vs. 74.0%, p = 0.05). Additionally, a rarely observed 4c allele was detected only in two subjects within Group 2 but not in Group 1. Conclusions: These findings suggest a trend of association between COVID-19 severity and NOS3 27-bp VNTR 4b/a genetic polymorphism. Genetic analysis may reveal patient susceptibility to disease, prognosis risk factors, and drug responsiveness.
Collapse
Affiliation(s)
- Aytekin İdikut
- Department of Chest Diseases, Faculty of Medicine, Hacettepe University, Ankara 06230, Türkiye (S.S.); (D.K.)
| | - İlter Değer
- Department of Pharmacology, Faculty of Medicine, Hacettepe University, Ankara 06230, Türkiye; (İ.D.); (M.O.B.)
| | - Gamze Göktaş
- Department of Chest Diseases, Faculty of Medicine, Hacettepe University, Ankara 06230, Türkiye (S.S.); (D.K.)
| | - Sevilay Karahan
- Department of Bioistatistics, Faculty of Medicine, Hacettepe University, Ankara 06230, Türkiye;
| | - Sevinç Sarınç
- Department of Chest Diseases, Faculty of Medicine, Hacettepe University, Ankara 06230, Türkiye (S.S.); (D.K.)
| | - Deniz Köksal
- Department of Chest Diseases, Faculty of Medicine, Hacettepe University, Ankara 06230, Türkiye (S.S.); (D.K.)
| | - Melih O. Babaoğlu
- Department of Pharmacology, Faculty of Medicine, Hacettepe University, Ankara 06230, Türkiye; (İ.D.); (M.O.B.)
| | - Elif Babaoğlu
- Department of Chest Diseases, Faculty of Medicine, Hacettepe University, Ankara 06230, Türkiye (S.S.); (D.K.)
| |
Collapse
|
22
|
Alloza-Moral I, Aldekoa-Etxabe A, Tulloch-Navarro R, Fiat-Arriola A, Mar C, Urrechaga E, Ponga C, Artiga-Folch I, Garcia-Bediaga N, Aspichueta P, Martin C, Zarandona-Garai A, Pérez-Fernández S, Arana-Arri E, Triviño JC, Uranga A, España PP, Vandenbroeck-van-Caeckenbergh K. Genetic Analysis and Predictive Modeling of COVID-19 Severity in a Hospital-Based Patient Cohort. Biomolecules 2025; 15:393. [PMID: 40149929 PMCID: PMC11940120 DOI: 10.3390/biom15030393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/27/2025] [Accepted: 02/28/2025] [Indexed: 03/29/2025] Open
Abstract
The COVID-19 pandemic has had a devastating impact, with more than 7 million deaths worldwide. Advanced age and comorbidities partially explain severe cases of the disease, but genetic factors also play a significant role. Genome-wide association studies (GWASs) have been instrumental in identifying loci associated with SARS-CoV-2 infection. Here, we report the results from a >820 K variant GWAS in a COVID-19 patient cohort from the hospitals associated with IIS Biobizkaia. We compared intensive care unit (ICU)-hospitalized patients with non-ICU-hospitalized patients. The GWAS was complemented with an integrated phenotype and genetic modeling analysis using HLA genotypes, a previously identified COVID-19 polygenic risk score (PRS) and clinical data. We identified four variants associated with COVID-19 severity with genome-wide significance (rs58027632 in KIF19; rs736962 in HTRA1; rs77927946 in DMBT1; and rs115020813 in LINC01283). In addition, we designed a multivariate predictive model including HLA, PRS and clinical data which displayed an area under the curve (AUC) value of 0.79. Our results combining human genetic information with clinical data may help to improve risk assessment for the development of a severe outcome of COVID-19.
Collapse
Affiliation(s)
- Iraide Alloza-Moral
- Inflammation & Biomarkers Group, Biobizkaia Health Research Institute, 48903 Barakaldo, Spain; (I.A.-M.); (A.A.-E.); (R.T.-N.); (A.F.-A.); (C.M.)
- Physiology Department, Faculty of Medicine and Nursery, Basque Country University (UPV/EHU), 48940 Leioa, Spain;
- Red de Enfermedades Inflamatorias (REI), Redes de Investigación Cooperativa Orientada a Resultados en Salud (RICORS), Carlos IIII Health Research Institute, 28029 Madrid, Spain
| | - Ane Aldekoa-Etxabe
- Inflammation & Biomarkers Group, Biobizkaia Health Research Institute, 48903 Barakaldo, Spain; (I.A.-M.); (A.A.-E.); (R.T.-N.); (A.F.-A.); (C.M.)
- Red de Enfermedades Inflamatorias (REI), Redes de Investigación Cooperativa Orientada a Resultados en Salud (RICORS), Carlos IIII Health Research Institute, 28029 Madrid, Spain
| | - Raquel Tulloch-Navarro
- Inflammation & Biomarkers Group, Biobizkaia Health Research Institute, 48903 Barakaldo, Spain; (I.A.-M.); (A.A.-E.); (R.T.-N.); (A.F.-A.); (C.M.)
- Red de Enfermedades Inflamatorias (REI), Redes de Investigación Cooperativa Orientada a Resultados en Salud (RICORS), Carlos IIII Health Research Institute, 28029 Madrid, Spain
| | - Ainhoa Fiat-Arriola
- Inflammation & Biomarkers Group, Biobizkaia Health Research Institute, 48903 Barakaldo, Spain; (I.A.-M.); (A.A.-E.); (R.T.-N.); (A.F.-A.); (C.M.)
- Red de Enfermedades Inflamatorias (REI), Redes de Investigación Cooperativa Orientada a Resultados en Salud (RICORS), Carlos IIII Health Research Institute, 28029 Madrid, Spain
| | - Carmen Mar
- Pneumology Department, Galdakao-Usansolo University Hospital, Biobizkaia Health Research Institute, 48960 Galdakao, Spain; (C.M.); (E.U.); (C.P.); (A.U.); (P.-P.E.)
| | - Eloisa Urrechaga
- Pneumology Department, Galdakao-Usansolo University Hospital, Biobizkaia Health Research Institute, 48960 Galdakao, Spain; (C.M.); (E.U.); (C.P.); (A.U.); (P.-P.E.)
| | - Cristina Ponga
- Pneumology Department, Galdakao-Usansolo University Hospital, Biobizkaia Health Research Institute, 48960 Galdakao, Spain; (C.M.); (E.U.); (C.P.); (A.U.); (P.-P.E.)
| | - Isabel Artiga-Folch
- Inflammation & Biomarkers Group, Biobizkaia Health Research Institute, 48903 Barakaldo, Spain; (I.A.-M.); (A.A.-E.); (R.T.-N.); (A.F.-A.); (C.M.)
| | - Naiara Garcia-Bediaga
- Bioinformatic Unit, Biobizkaia Health Research Institute, 48903 Barakaldo, Spain; (N.G.-B.); (A.Z.-G.); (S.P.-F.)
| | - Patricia Aspichueta
- Physiology Department, Faculty of Medicine and Nursery, Basque Country University (UPV/EHU), 48940 Leioa, Spain;
- Research Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), 28029 Madrid, Spain
- Biobizkaia Health Research Institute, Cruces University Hospital, 48903 Barakaldo, Spain
| | - Cesar Martin
- Inflammation & Biomarkers Group, Biobizkaia Health Research Institute, 48903 Barakaldo, Spain; (I.A.-M.); (A.A.-E.); (R.T.-N.); (A.F.-A.); (C.M.)
- Biochemistry and Molecular Biology Department, Science and Technology School, Basque Country University (UPV/EHU), 48940 Leioa, Spain
- Biofisika Institute (UPV/EHU, CSIC), 48940 Leioa, Spain
| | - Aitor Zarandona-Garai
- Bioinformatic Unit, Biobizkaia Health Research Institute, 48903 Barakaldo, Spain; (N.G.-B.); (A.Z.-G.); (S.P.-F.)
| | - Silvia Pérez-Fernández
- Bioinformatic Unit, Biobizkaia Health Research Institute, 48903 Barakaldo, Spain; (N.G.-B.); (A.Z.-G.); (S.P.-F.)
| | - Eunate Arana-Arri
- Clinical Epidemiology Unit, Biobizkaia Health Research Institute, Cruces University Hospital, Plaza de Cruces s/n, 48903 Barakaldo, Spain;
| | | | - Ane Uranga
- Pneumology Department, Galdakao-Usansolo University Hospital, Biobizkaia Health Research Institute, 48960 Galdakao, Spain; (C.M.); (E.U.); (C.P.); (A.U.); (P.-P.E.)
| | - Pedro-Pablo España
- Pneumology Department, Galdakao-Usansolo University Hospital, Biobizkaia Health Research Institute, 48960 Galdakao, Spain; (C.M.); (E.U.); (C.P.); (A.U.); (P.-P.E.)
| | - Koen Vandenbroeck-van-Caeckenbergh
- Inflammation & Biomarkers Group, Biobizkaia Health Research Institute, 48903 Barakaldo, Spain; (I.A.-M.); (A.A.-E.); (R.T.-N.); (A.F.-A.); (C.M.)
- Red de Enfermedades Inflamatorias (REI), Redes de Investigación Cooperativa Orientada a Resultados en Salud (RICORS), Carlos IIII Health Research Institute, 28029 Madrid, Spain
- Biochemistry and Molecular Biology Department, Science and Technology School, Basque Country University (UPV/EHU), 48940 Leioa, Spain
- Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Spain
| |
Collapse
|
23
|
Adilović M. COVID-19 related complications. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2025; 213:259-314. [PMID: 40246346 DOI: 10.1016/bs.pmbts.2025.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
The COVID-19 pandemic has significantly impacted global healthcare systems, revealed vulnerabilities and prompted a re-evaluation of medical practices. Acute complications from the virus, including cardiovascular and neurological issues, have underscored the necessity for timely medical interventions. Advances in diagnostic methods and personalized therapies have been pivotal in mitigating severe outcomes. Additionally, Long COVID has emerged as a complex challenge, affecting various body systems and leading to respiratory, cardiovascular, neurological, psychological, and musculoskeletal problems. This broad spectrum of complications highlights the importance of multidisciplinary management approaches that prioritize therapy, rehabilitation, and patient-centered care. Vulnerable populations such as paediatric patients, pregnant women, and immunocompromised individuals face unique risks and complications, necessitating continuous monitoring and tailored management strategies to reduce morbidity and mortality associated with COVID-19.
Collapse
Affiliation(s)
- Muhamed Adilović
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnička cesta, Sarajevo, Bosnia and Herzegovina.
| |
Collapse
|
24
|
Torres-Poveda K, Bahena-Román M, Contreras-Ochoa CO, Lagunas-Martínez A, Bermúdez-Morales VH, Pando-Robles V, Ortiz-Flores E, Cortés-Pedroza F, Santana-Román ME, Martínez-Campos C, Sánchez-Alemán M, Manzo-Merino J, Morales-Ortega A, Madrid-González DA, Cantú-Cuevas MA, Barón-Olivares H, Madrid-Marina V. High nasopharyngeal and serum IL-6 levels and the - 573G > C polymorphism (rs1800796) are linked with the risk of severe COVID-19 in a Mexican population: a case‒control study. BMC Infect Dis 2025; 25:315. [PMID: 40045221 PMCID: PMC11884130 DOI: 10.1186/s12879-025-10695-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 02/19/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND COVID-19 was the leading cause of death in Mexico between 2020 and 2021. SARS-CoV-2 infection varies widely among individuals and populations. Since variations in genes related to the immune response may play a role in the susceptibility to and outcome of COVID-19, the associations of gene polymorphisms (SNPs) of IL-6 (- 573G > C, rs1800796), TNF-α (- 308G > A, rs1800629), and IFN-γ (- 1615 C > T, rs2069705) with the expression levels of these proteins in the nasopharynx and serum were evaluated in a Mexican population with mild, severe, or critical COVID-19. METHODS A total of 560 COVID-19 patients (309 mild, 163 severe, and 88 critical cases) and 560 age- and sex-matched COVID-19-negative controls were recruited for this case‒control study. The selected SNPs were genotyped via allelic discrimination. Logistic regression analysis was conducted considering four models of inheritance, and ORs were determined for each genotypic variant, adjusting for associated comorbidities in the multivariate model. The nasopharyngeal mRNA expression levels of IL-6, IFN-γ and TNF-α were determined. The levels of IL-6, IFN-γ, IFN-α2, and TNF-α in the serum were quantified. Significant differences were assessed via the Wilcoxon Mann‒Whitney U test. RESULTS The C allele of the IL-6 - 573 SNP was associated with a greater risk of mild and severe COVID-19 (OR: 2.3, CI: 1.897-2.838, p = 0.0001; and OR: 1.5, CI: 1.167-1.949, p = 0.002, respectively), whereas the A allele of the TNF-α - 308 SNP and the T allele of the IFN-γ - 1615 SNP were shown protective roles against severe COVID-19 (OR: 0.3, CI: 0.189-0.537, p = 0.0001; and OR: 0.7, CI: 0.563-1.006, p = 0.05) and against critical COVID-19 (OR: 0.3, CI: 0.158-0.640, p = 0.001; and OR: 0.4, CI: 0.290-0.678, p = 0.0001), adjusting for diabetes and hypertension. Nasopharyngeal IL-6 expression levels were lower in mild COVID-19 patients (p = 0.001) than in critical patients (p = 0.005). Serum IL-6 levels were significantly elevated in the critical cases (p = 0.01). CONCLUSIONS Our results revealed that the IL-6 - 573 G > C SNP and increased IL-6 nasopharyngeal and serum levels are associated with the risk of severe COVID-19 in a Mexican population.
Collapse
Affiliation(s)
- Kirvis Torres-Poveda
- Center for Research on Infectious Diseases, Instituto Nacional de Salud Pública (INSP), Cuernavaca, Mexico
- Secretaria de Ciencia, Humanidades, Tecnología e Innovación (SECIHTI)-Instituto Nacional de Salud Pública, Cuernavaca, Mexico
| | - Margarita Bahena-Román
- Center for Research on Infectious Diseases, Instituto Nacional de Salud Pública (INSP), Cuernavaca, Mexico
| | - Carla O Contreras-Ochoa
- Center for Research on Infectious Diseases, Instituto Nacional de Salud Pública (INSP), Cuernavaca, Mexico
| | - Alfredo Lagunas-Martínez
- Center for Research on Infectious Diseases, Instituto Nacional de Salud Pública (INSP), Cuernavaca, Mexico
| | | | - Victoria Pando-Robles
- Center for Research on Infectious Diseases, Instituto Nacional de Salud Pública (INSP), Cuernavaca, Mexico
| | - Esmeralda Ortiz-Flores
- Center for Research on Infectious Diseases, Instituto Nacional de Salud Pública (INSP), Cuernavaca, Mexico
| | - Fabiola Cortés-Pedroza
- Center for Research on Infectious Diseases, Instituto Nacional de Salud Pública (INSP), Cuernavaca, Mexico
| | - María E Santana-Román
- Center for Research on Infectious Diseases, Instituto Nacional de Salud Pública (INSP), Cuernavaca, Mexico
| | - Cecilia Martínez-Campos
- Center for Research on Infectious Diseases, Instituto Nacional de Salud Pública (INSP), Cuernavaca, Mexico
- Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - Miguel Sánchez-Alemán
- Center for Research on Infectious Diseases, Instituto Nacional de Salud Pública (INSP), Cuernavaca, Mexico
| | - Joaquin Manzo-Merino
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Ausencio Morales-Ortega
- Laboratorio Estatal de Salud Pública. Health Services of the State of Morelos, Jiutepec, Mexico
| | | | | | - Héctor Barón-Olivares
- Dirección General de Coordinación y Supervisión. Health Services of the State of Morelos, Cuernavaca, Mexico
| | - Vicente Madrid-Marina
- Center for Research on Infectious Diseases, Instituto Nacional de Salud Pública (INSP), Cuernavaca, Mexico.
| |
Collapse
|
25
|
Górska A, Canziani LM, Rinaldi E, Pana ZD, Beale S, Bai F, Boxma-de Klerk BM, de Bruijn S, Donà D, Ekkelenkamp MB, Incardona F, Mallon P, Marchetti GC, Puhan M, Riva A, Simensen VC, Vaillant M, van der Zalm MM, van Kuijk SMJ, Wingerden SV, Judd A, Tacconelli E, Peñalvo JL. Learning from post-COVID-19 condition for epidemic preparedness: a variable catalogue for future post-acute infection syndromes. Clin Microbiol Infect 2025; 31:380-388. [PMID: 39662824 DOI: 10.1016/j.cmi.2024.12.001] [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: 09/12/2024] [Revised: 11/13/2024] [Accepted: 12/03/2024] [Indexed: 12/13/2024]
Abstract
SCOPE The emergence of post-COVID-19 condition (PCC) after SARS-CoV-2 infection underscores the critical need for preparedness in addressing future post-acute infection syndromes (PAIS), particularly those linked to epidemic outbreaks. The lack of standardized clinical and epidemiological data during the COVID-19 pandemic has significantly hindered timely diagnosis and effective treatment of PCC, highlighting the necessity of pre-emptively standardizing data collection in clinical studies to better define and manage future PAIS. In response, the Cohort Coordination Board, a consortium of European-funded COVID-19 research projects, has reviewed data from PCC studies conducted by its members. This paper leverages the Cohort Coordination Board's expertise to propose a standardized catalogue of variables, informed by the lessons learned during the pandemic, intended for immediate use in the design of future observational studies and clinical trials for emerging infections of epidemic potential. RECOMMENDATIONS The early implementation of standardized data collection, facilitated by the PAIS data catalogue, is essential for accelerating the identification and management of PAIS in future epidemics. This approach will enable more precise syndrome definitions, expedite diagnostic processes, and optimize treatment strategies, while also supporting long-term follow-up of affected individuals. The availability of harmonized data collection protocols will enhance preparedness across European and international cohort studies, and trials enabling a prompt and coordinated response, as well as more efficient resource allocation, in the event of emerging infections and associated PAIS.
Collapse
Affiliation(s)
- Anna Górska
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | | | - Eugenia Rinaldi
- Berlin Institute of Health at Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Zoi D Pana
- Medical School, Basic and Clinical Studies Department, University of Nicosia, Nicosia, Cyprus
| | - Sarah Beale
- UCL Institute of Health Informatics, University College London, London, United Kingdom
| | - Francesca Bai
- Clinic of Infectious Diseases, San Paolo Hospital, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Bianca M Boxma-de Klerk
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Simeon de Bruijn
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Daniele Donà
- Department of Women's and Children's Health, Università Degli Studi di Padova, Padova, Italy
| | - Miquel B Ekkelenkamp
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Patrick Mallon
- Centre for Experimental Pathogen Host Research, University College Dublin, Dublin, Ireland
| | - Giulia C Marchetti
- Clinic of Infectious Diseases, San Paolo Hospital, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy; Department of Health Science, University of Milan, Milan, Italy
| | - Milo Puhan
- Epidemiology, Biostatistics and Prevention Institute, University of Zurich, Zurich, Switzerland
| | - Agostino Riva
- Department of Biomedical and Clinical Sciences, Università Degli Studi di Milano, Milan, Italy
| | - Victoria C Simensen
- Department of Vaccines and Immunisation, Division of Infectious Disease Control, Norwegian Institute of Public Health, Oslo, Norway
| | - Michel Vaillant
- Competence Centre for Methodology and Statistics, Department of Medical Informatics, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Marieke M van der Zalm
- Department of Paediatrics and Child Health, Stellenbosch University, Cape Town, South Africa
| | | | - Sophie van Wingerden
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Ali Judd
- MRC Clinical Trials Unit, University College London, London, United Kingdom; Fondazione Penta ETS, Padova, Italy
| | - Evelina Tacconelli
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - José L Peñalvo
- National Center for Epidemiology, Carlos III Health Institute (ISCIII), Madrid, Spain.
| |
Collapse
|
26
|
Yao C, Dong Y, Zhou H, Zou X, Alhaskawi A, Ezzi SHA, Wang Z, Lai J, Kota VG, Abdulla MHAH, Liu Z, Abdalbary SA, Alenikova O, Lu H. COVID-19 and acute limb ischemia: latest hypotheses of pathophysiology and molecular mechanisms. J Zhejiang Univ Sci B 2025; 26:333-352. [PMID: 40274383 PMCID: PMC12021539 DOI: 10.1631/jzus.b2300512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 01/01/2024] [Indexed: 04/26/2025]
Abstract
Coronavirus disease 2019 (COVID-19) is a multi-system disease that can lead to various severe complications. Acute limb ischemia (ALI) has been increasingly recognized as a COVID-19-associated complication that often predicts a poor prognosis. However, the pathophysiology and molecular mechanisms underlying COVID-19-associated ALI remain poorly understood. Hypercoagulability and thrombosis are considered important mechanisms, but we also emphasize the roles of vasospasm, hypoxia, and acidosis in the pathogenesis of the disease. The angiotensin-converting enzyme 2 (ACE2) pathway, inflammation, and platelet activation may be important molecular mechanisms underlying these pathological changes induced by COVID-19. Furthermore, we discuss the hypotheses of risk factors for COVID-19-associated ALI from genetic, age, and gender perspectives based on our analysis of molecular mechanisms. Additionally, we summarize therapeutic approaches such as use of the interleukin-6 (IL-6) blocker tocilizumab, calcium channel blockers, and angiotensin-converting enzyme inhibitors, providing insights for the future treatment of coronavirus-associated limb ischemic diseases.
Collapse
Affiliation(s)
- Chengjun Yao
- Department of Orthopaedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yanzhao Dong
- Department of Orthopaedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Haiying Zhou
- Department of Orthopaedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Xiaodi Zou
- Department of Orthopaedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310005, China
| | - Ahmad Alhaskawi
- Department of Orthopaedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Sohaib Hasan Abdullah Ezzi
- Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Department of Orthopaedics, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Zewei Wang
- Department of Orthopaedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Jingtian Lai
- Department of Orthopaedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Vishnu Goutham Kota
- Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | | | - Zhenfeng Liu
- Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Sahar Ahmed Abdalbary
- Department of Orthopaedic Physical Therapy, Faculty of Physical Therapy, Nahda University, Beni Suef 2711860, Egypt
| | - Olga Alenikova
- Republic Scientific Practical Center of Neurology and Neurosurgery, Ministry of Health of the Republic of Belarus, Minsk 220004, Belarus
| | - Hui Lu
- Department of Orthopaedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.
| |
Collapse
|
27
|
Martin-Castaño B, Diez-Echave P, García-García J, Hidalgo-García L, Ruiz-Malagon AJ, Molina-Tijeras JA, Rodríguez-Sojo MJ, Redruello-Romero A, Martínez-Zaldívar M, Mota E, Cobo F, Díaz-Villamarin X, Alvarez-Estevez M, García F, Morales-García C, Merlos S, Garcia-Flores P, Colmenero-Ruiz M, Hernández-Quero J, Nuñez M, Rodriguez-Cabezas ME, Carazo A, Martin J, Moron R, Rodríguez Nogales A, Galvez J. The relationship between gut and nasopharyngeal microbiome composition can predict the severity of COVID-19. eLife 2025; 13:RP95292. [PMID: 39963971 PMCID: PMC11835386 DOI: 10.7554/elife.95292] [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] [Indexed: 02/20/2025] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a respiratory illness caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that displays great variability in clinical phenotype. Many factors have been described to be correlated with its severity, and microbiota could play a key role in the infection, progression, and outcome of the disease. SARS-CoV-2 infection has been associated with nasopharyngeal and gut dysbiosis and higher abundance of opportunistic pathogens. To identify new prognostic markers for the disease, a multicentre prospective observational cohort study was carried out in COVID-19 patients divided into three cohorts based on symptomatology: mild (n = 24), moderate (n = 51), and severe/critical (n = 31). Faecal and nasopharyngeal samples were taken, and the microbiota was analysed. Linear discriminant analysis identified Mycoplasma salivarium, Prevotella dentalis, and Haemophilus parainfluenzae as biomarkers of severe COVID-19 in nasopharyngeal microbiota, while Prevotella bivia and Prevotella timonensis were defined in faecal microbiota. Additionally, a connection between faecal and nasopharyngeal microbiota was identified, with a significant ratio between P. timonensis (faeces) and P. dentalis and M. salivarium (nasopharyngeal) abundances found in critically ill patients. This ratio could serve as a novel prognostic tool for identifying severe COVID-19 cases.
Collapse
Affiliation(s)
- Benita Martin-Castaño
- Centro de Salud Las Gabias, Distrito Granada-MetropolitanoGranadaSpain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA)GranadaSpain
| | - Patricia Diez-Echave
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA)GranadaSpain
- Department of Pharmacology, Center for Biomedical Research (CIBM), University of GranadaGranadaSpain
| | - Jorge García-García
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA)GranadaSpain
- Department of Pharmacology, Center for Biomedical Research (CIBM), University of GranadaGranadaSpain
- Servicio Microbiología, Hospital Universitario Clínico San CecilioGranadaSpain
| | - Laura Hidalgo-García
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA)GranadaSpain
- Department of Pharmacology, Center for Biomedical Research (CIBM), University of GranadaGranadaSpain
| | - Antonio Jesús Ruiz-Malagon
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA)GranadaSpain
- Department of Pharmacology, Center for Biomedical Research (CIBM), University of GranadaGranadaSpain
| | - José Alberto Molina-Tijeras
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA)GranadaSpain
- Department of Pharmacology, Center for Biomedical Research (CIBM), University of GranadaGranadaSpain
| | - María Jesús Rodríguez-Sojo
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA)GranadaSpain
- Department of Pharmacology, Center for Biomedical Research (CIBM), University of GranadaGranadaSpain
| | | | - Margarita Martínez-Zaldívar
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA)GranadaSpain
- Centro de Salud “Salvador Caballero”, Distrito Granada-MetropolitanoGranadaSpain
| | - Emilio Mota
- Centro de Salud “Salvador Caballero”, Distrito Granada-MetropolitanoGranadaSpain
| | - Fernando Cobo
- Servicio Microbiología, Hospital Universitario Virgen de las NievesGranadaSpain
| | | | - Marta Alvarez-Estevez
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA)GranadaSpain
- Servicio Microbiología, Hospital Universitario Clínico San CecilioGranadaSpain
- CIBER de Enfermedades Infecciosas (CIBER-Infecc), Instituto de Salud Carlos IIIMadridSpain
| | - Federico García
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA)GranadaSpain
- Servicio Microbiología, Hospital Universitario Clínico San CecilioGranadaSpain
- CIBER de Enfermedades Infecciosas (CIBER-Infecc), Instituto de Salud Carlos IIIMadridSpain
| | | | - Silvia Merlos
- Respiratory Medicine Department, Hospital Universitario Virgen de las NievesGranadaSpain
| | - Paula Garcia-Flores
- Respiratory Medicine Department, Hospital Universitario Virgen de las NievesGranadaSpain
| | - Manuel Colmenero-Ruiz
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA)GranadaSpain
- Servicio de Medicina Intensiva, Hospital Universitario Clínico San CecilioGranadaSpain
| | - José Hernández-Quero
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA)GranadaSpain
- Servicio de Enfermedades Infecciosas, Hospital Universitario Clínico San CecilioGranadaSpain
| | - Maria Nuñez
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA)GranadaSpain
- Servicio Farmacia Hospitalaria, Hospital Universitario Clínico San CecilioGranadaSpain
- CIBER de Epidemiología y Salud Pública (CIBER-ESP), Instituto de Salud Carlos IIIMadridSpain
| | - Maria Elena Rodriguez-Cabezas
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA)GranadaSpain
- Department of Pharmacology, Center for Biomedical Research (CIBM), University of GranadaGranadaSpain
| | - Angel Carazo
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA)GranadaSpain
- Servicio Microbiología, Hospital Universitario Clínico San CecilioGranadaSpain
| | - Javier Martin
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine López-Neyra, CSICGranadaSpain
| | - Rocio Moron
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA)GranadaSpain
- Servicio Farmacia Hospitalaria, Hospital Universitario Clínico San CecilioGranadaSpain
| | - Alba Rodríguez Nogales
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA)GranadaSpain
- Department of Pharmacology, Center for Biomedical Research (CIBM), University of GranadaGranadaSpain
| | - Julio Galvez
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA)GranadaSpain
- Department of Pharmacology, Center for Biomedical Research (CIBM), University of GranadaGranadaSpain
- CIBER de Enfermedades Hepáticas y Digestivas (CIBER-EHD), Instituto de Salud Carlos IIIMadridSpain
| |
Collapse
|
28
|
Naiditch H, Betts MR, Larman HB, Levi M, Rosenberg AZ. Immunologic and inflammatory consequences of SARS-CoV-2 infection and its implications in renal disease. Front Immunol 2025; 15:1376654. [PMID: 40012912 PMCID: PMC11861071 DOI: 10.3389/fimmu.2024.1376654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 12/23/2024] [Indexed: 02/28/2025] Open
Abstract
The emergence of the COVID-19 pandemic made it critical to understand the immune and inflammatory responses to the SARS-CoV-2 virus. It became increasingly recognized that the immune response was a key mediator of illness severity and that its mechanisms needed to be better understood. Early infection of both tissue and immune cells, such as macrophages, leading to pyroptosis-mediated inflammasome production in an organ system critical for systemic oxygenation likely plays a central role in the morbidity wrought by SARS-CoV-2. Delayed transcription of Type I and Type III interferons by SARS-CoV-2 may lead to early disinhibition of viral replication. Cytokines such as interleukin-1 (IL-1), IL-6, IL-12, and tumor necrosis factor α (TNFα), some of which may be produced through mechanisms involving nuclear factor kappa B (NF-κB), likely contribute to the hyperinflammatory state in patients with severe COVID-19. Lymphopenia, more apparent among natural killer (NK) cells, CD8+ T-cells, and B-cells, can contribute to disease severity and may reflect direct cytopathic effects of SARS-CoV-2 or end-organ sequestration. Direct infection and immune activation of endothelial cells by SARS-CoV-2 may be a critical mechanism through which end-organ systems are impacted. In this context, endovascular neutrophil extracellular trap (NET) formation and microthrombi development can be seen in the lungs and other critical organs throughout the body, such as the heart, gut, and brain. The kidney may be among the most impacted extrapulmonary organ by SARS-CoV-2 infection owing to a high concentration of ACE2 and exposure to systemic SARS-CoV-2. In the kidney, acute tubular injury, early myofibroblast activation, and collapsing glomerulopathy in select populations likely account for COVID-19-related AKI and CKD development. The development of COVID-19-associated nephropathy (COVAN), in particular, may be mediated through IL-6 and signal transducer and activator of transcription 3 (STAT3) signaling, suggesting a direct connection between the COVID-19-related immune response and the development of chronic disease. Chronic manifestations of COVID-19 also include systemic conditions like Multisystem Inflammatory Syndrome in Children (MIS-C) and Adults (MIS-A) and post-acute sequelae of COVID-19 (PASC), which may reflect a spectrum of clinical presentations of persistent immune dysregulation. The lessons learned and those undergoing continued study likely have broad implications for understanding viral infections' immunologic and inflammatory consequences beyond coronaviruses.
Collapse
Affiliation(s)
- Hiam Naiditch
- Department of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Michael R. Betts
- Department of Microbiology and Institute of Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - H. Benjamin Larman
- Institute for Cell Engineering, Division of Immunology, Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
| | - Moshe Levi
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC, United States
| | - Avi Z. Rosenberg
- Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
29
|
Mallol R, Rombauts A, Abelenda-Alonso G, Gudiol C, Balsalobre M, Carratalà J. Metabolomic profile of severe COVID-19 and a signature predictive of progression towards severe disease status: a prospective cohort study (METCOVID). Sci Rep 2025; 15:4963. [PMID: 39929875 PMCID: PMC11811168 DOI: 10.1038/s41598-025-87288-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 01/17/2025] [Indexed: 02/13/2025] Open
Abstract
Profound metabolomic alterations occur during COVID-19. Early identification of the subset of hospitalised COVID-19 patients at risk of developing severe disease is critical for optimal resource utilization and prompt treatment. This work explores the metabolomic profile of hospitalised adult COVID-19 patients with severe disease, and establishes a predictive signature for disease progression. Within 48 hours of admission, serum samples were collected from 148 hospitalised patients for nuclear magnetic resonance (NMR) spectroscopy. Lipoprotein profiling was performed using the 1H-NMR-based Liposcale test, while low molecular weight metabolites were analysed using one-dimensional Carr-Purcell-Meiboom-Gill pulse spectroscopy and an adaptation of the Dolphin method for lipophilic extracts. Severe COVID-19, per WHO's Clinical Progression Scale, was characterized by altered lipoprotein distribution, elevated signals of glyc-A and glyc-B, a shift towards a catabolic state with elevated levels of branched-chain amino acids, and accumulation of ketone bodies. Furthermore, COVID-19 patients initially presenting with moderate disease but progressing to severe stages exhibited a distinct metabolic signature. Our multivariate model demonstrated a cross-validated AUC of 0.82 and 72% predictive accuracy for severity progression. NMR spectroscopy-based metabolomic profiling enables the identification of moderate COVID-19 patients at risk of disease progression, aiding in resource allocation and early intervention.
Collapse
Affiliation(s)
- Roger Mallol
- Fundació Institut Universitari per a la recerca a l'Atenció Primària de Salut Jordi Gol i Gurina (IDIAPJGol), 08007, Barcelona, Spain
| | - Alexander Rombauts
- Department of Infectious Diseases, Hospital Universitari de Bellvitge-IDIBELL, 08907, Barcelona, Spain.
| | - Gabriela Abelenda-Alonso
- Department of Infectious Diseases, Hospital Universitari de Bellvitge-IDIBELL, 08907, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Carlota Gudiol
- Department of Infectious Diseases, Hospital Universitari de Bellvitge-IDIBELL, 08907, Barcelona, Spain
- Department of Medicine, Universitat de Barcelona, 08007, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029, Madrid, Spain
- Institut Català d'Oncologia (ICO), Hospital Duran i Reynals, 08908, Barcelona, Spain
| | - Marc Balsalobre
- Human Environment Research, La Salle-Universitat Ramon Llull, 08022, Barcelona, Spain
| | - Jordi Carratalà
- Department of Infectious Diseases, Hospital Universitari de Bellvitge-IDIBELL, 08907, Barcelona, Spain
- Department of Medicine, Universitat de Barcelona, 08007, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029, Madrid, Spain
| |
Collapse
|
30
|
Vogi V, Haschka D, Forer L, Schwendinger S, Petzer V, Coassin S, Tancevski I, Sonnweber T, Löffler-Ragg J, Puchhammer-Stöckl E, Graninger M, Wolf D, Kronenberg F, Zschocke J, Jukic E, Weiss G. Severe COVID-19 disease is associated with genetic factors affecting plasma ACE2 receptor and CRP concentrations. Sci Rep 2025; 15:4708. [PMID: 39922945 PMCID: PMC11807156 DOI: 10.1038/s41598-025-89306-4] [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: 12/18/2023] [Accepted: 02/04/2025] [Indexed: 02/10/2025] Open
Abstract
A hyperinflammatory state with highly elevated concentrations of inflammatory biomarkers such as C-reactive protein (CRP) is a characteristic feature of severe coronavirus disease 2019 (COVID-19). To examine a potential role of common genetic factors that may influence COVID-19 outcomes, we investigated whether individuals with a polygenic predisposition for a pro-inflammatory response (in the form of Polygenic Scores) are more likely to develop severe COVID-19. The innovative approach of polygenic scores to investigate genetic factors in COVID-19 severity should provide a comprehensive approach beyond single-gene studies. In our cohort of 156 patients of European ancestry, two overlapping Polygenic Scores (PGS) predicting a genetic predisposition to basal CRP concentrations were significantly different between non-severe and severe COVID-19 cases and were associated with less severe COVID-19 outcomes. Furthermore, specific single nucleotide polymorphisms (SNPs) that contribute to either of the two Polygenic Scores predicting basal CRP levels are associated with different traits that represent risk factors for COVID-19 disease initiation (ACE2 receptor, viral replication) and progression (CRP). We suggest that genetically determined enforced CRP formation may contribute to strengthening of innate immune responses and better initial pathogen control thereby reducing the risk of subsequent hyperinflammation and adverse course of COVID-19.
Collapse
Affiliation(s)
- Verena Vogi
- Institute of Human Genetics, Medical University Innsbruck, Innsbruck, 6020, Austria
| | - David Haschka
- Department of Internal Medicine II (Infectious Diseases, Immunology, Pneumology and Rheumatology), Medical University Innsbruck, Innsbruck, 6020, Austria
| | - Lukas Forer
- Institute of Genetic Epidemiology, Medical University Innsbruck, Innsbruck, 6020, Austria
| | - Simon Schwendinger
- Institute of Human Genetics, Medical University Innsbruck, Innsbruck, 6020, Austria
| | - Verena Petzer
- Department of Internal Medicine V (Hematology and Internistic Oncology), Medical University Innsbruck, Innsbruck, 6020, Austria
| | - Stefan Coassin
- Institute of Genetic Epidemiology, Medical University Innsbruck, Innsbruck, 6020, Austria
| | - Ivan Tancevski
- Department of Internal Medicine II (Infectious Diseases, Immunology, Pneumology and Rheumatology), Medical University Innsbruck, Innsbruck, 6020, Austria
| | - Thomas Sonnweber
- Department of Internal Medicine II (Infectious Diseases, Immunology, Pneumology and Rheumatology), Medical University Innsbruck, Innsbruck, 6020, Austria
| | - Judith Löffler-Ragg
- Department of Internal Medicine II (Infectious Diseases, Immunology, Pneumology and Rheumatology), Medical University Innsbruck, Innsbruck, 6020, Austria
| | | | - Marianne Graninger
- Department of Virology, Medical University Vienna, Vienna, 1090, Austria
| | - Dominik Wolf
- Department of Internal Medicine V (Hematology and Internistic Oncology), Medical University Innsbruck, Innsbruck, 6020, Austria
| | - Florian Kronenberg
- Institute of Genetic Epidemiology, Medical University Innsbruck, Innsbruck, 6020, Austria
| | - Johannes Zschocke
- Institute of Human Genetics, Medical University Innsbruck, Innsbruck, 6020, Austria
| | - Emina Jukic
- Institute of Human Genetics, Medical University Innsbruck, Innsbruck, 6020, Austria.
| | - Günter Weiss
- Department of Internal Medicine II (Infectious Diseases, Immunology, Pneumology and Rheumatology), Medical University Innsbruck, Innsbruck, 6020, Austria.
| |
Collapse
|
31
|
Kunvar S, Czarnomska S, Pertoldi C, Stronen AV, Ruczyńska I, Tokarska M. A lethal disease of the European bison - posthitis is conditionally determined by its genomics. Sci Rep 2025; 15:4483. [PMID: 39915645 PMCID: PMC11802879 DOI: 10.1038/s41598-025-88702-0] [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: 02/16/2024] [Accepted: 01/30/2025] [Indexed: 02/09/2025] Open
Abstract
Posthitis is an incurable lethal disease of males of the European bison (Bison bonasus), regarded to be one of the major threats to the survival of the iconic species. Multiple attempts have been undertaken over the last 30 years to identify a source of infection and a primary pathogen. Studies indicated the disease could have a genetic background after tools developed for cattle (Bos taurus) revealed genomic regions that could be associated with its occurrence. In this study, we applied deep coverage targeted sequencing to 74 regions on 10 of the bison's chromosomes (1, 9, 12, 13, 15, 23, 25, 26, 29, and X) in search for species-specific single nucleotide polymorphism (SNP) markers that could help to explain the mechanism of the disease and be used to test for posthitis susceptibility. The association results were ranked based on p-values lower than 0.005 and odds ratios (OR) higher than 1. We obtained 30 SNP markers that met these requirements, all located on chromosome 25. However, none of the SNPs found in the study was significantly associated with posthitis occurrence after Bonferroni correction. The conditional nature of posthitis and 'false negative' sampling represent major difficulties in investigating this disease, and we recommend a complex genomic and environmental factors association assay that could eventually explain the puzzling etiology of posthitis and help mitigate this threat to the European bison.
Collapse
Affiliation(s)
- Sazia Kunvar
- Mammal Research Institute PAS, Stoczek 1, 17-230, Białowieża, Poland.
| | - Sylwia Czarnomska
- Museum and Institute of Zoology PAS, Twarda 51/55, 00-818, Warsaw, Poland
| | - Cino Pertoldi
- Department of Chemistry and Bioscience, Section of Bioscience and Engineering, Aalborg University, Fredrik Bajers Vej 7H, 9220, Aalborg, Denmark
- Aalborg Zoo, Mølleparkvej 63, 9000, Aalborg, Denmark
| | - Astrid V Stronen
- Department of Biology, Chair of Ecology and Environment Conservation, University of Ljubliana, Večna Pot 111, Ljubljana, Slovenia
| | - Iwona Ruczyńska
- Mammal Research Institute PAS, Stoczek 1, 17-230, Białowieża, Poland
| | | |
Collapse
|
32
|
Strunz B, Momayyezi P, Bilev E, Muvva JR, Chen P, Bister J, Schaffer M, Akber M, Cornillet M, Horowitz A, Malmberg KJ, Rooyackers O, Aleman S, Ljunggren HG, Björkström NK, Strålin K, Hammer Q. The HLA-B -21 M/T dimorphism associates with disease severity in COVID-19. Genes Immun 2025; 26:70-74. [PMID: 39487235 PMCID: PMC11832411 DOI: 10.1038/s41435-024-00302-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 10/15/2024] [Accepted: 10/18/2024] [Indexed: 11/04/2024]
Abstract
Host genetics shape immune responses and influence severity of infectious diseases. The HLA-B -21 M/T dimorphism tunes the functionality of natural killer (NK) cells expressing the inhibitory receptor NKG2A. NKG2A+ NK cells have been reported to recognize SARS-CoV-2-infected cells, but it remains unclear whether the HLA-B -21 M/T dimorphism associates with COVID-19 severity. Here, we investigated the influence of the HLA-B -21 M/T dimorphism in a cohort of 230 unvaccinated patients hospitalized with COVID-19 and requiring respiratory support. We found that HLA-B -21 M/M genotypes were more prevalent in patients with moderate compared to severe COVID-19 (6.0% vs. 0.9%). Comparison of age- and sex-matched sub-groups revealed that patients with M/M genotypes required mechanical respiratory support less frequently (OR = 0.13, 95% CI = 0.01-0.76, P = 0.013). Furthermore, patients with M/M genotypes showed a coordinately shifted signature of clinical laboratory parameters, coinciding with elevated serum levels of the anti-viral cytokine IFN-γ. These findings demonstrate that HLA-B variants associate with COVID-19 severity and suggest that the robust functionality of NKG2A+ NK cells in patients carrying the M/M genotype may contribute to protection from severe disease.
Collapse
Grants
- Åke Wiberg Stiftelse (Åke Wiberg Foundation)
- This work received funding from the Swedish Research Council (2020-06250 to C.M. and 2020-02286 to K.-J.M.), Swedish Foundation for Strategic Research (to K.-J.M.), Sweden’s Innovation Agency (to K.-J.M.), and Knut and Alice Wallenberg Foundation (2018.0106 to K.-J.M.). This work was further supported by Region Stockholm (2020-0733 to P.M.), Åke Wibergs Stiftelse (M22-0013), KI Foundations (2022-01606), KI Foundation for Virus Research (2021-00069, 2022-00245, and 2023-00155), Petrus och Augusta Hedlunds Stiftelse (M2021-1533 and M2022-1821), Stiftelsen Clas Groschinskys Minnesfold (M21120 and M2233), Stiftelsen Lars Hiertas Minne (FO2021-0263 and FO2023-0167), Stiftelsen Tornspiran, and Jonas Söderquist Stiftelse (all to Q.H.).
Collapse
Affiliation(s)
- Benedikt Strunz
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Pouria Momayyezi
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Eleni Bilev
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Jagadeeswara Rao Muvva
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Puran Chen
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Jonna Bister
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Marie Schaffer
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Mira Akber
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Martin Cornillet
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Amir Horowitz
- Department of Oncological Sciences, Precision Immunology Institute, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Karl-Johan Malmberg
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Olav Rooyackers
- Division of Anesthesiology and Intensive Care, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Soo Aleman
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Hans-Gustaf Ljunggren
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Niklas K Björkström
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Kristoffer Strålin
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
- Division of Infectious Diseases and Dermatology, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Quirin Hammer
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
33
|
Tscharntke LT, Jung N, Hanses F, Koll CEM, Pilgram L, Rieg S, Borgmann S, de Miranda SMN, Scherer M, Spinner CD, Rüthrich M, Vehreschild MJGT, von Bergwelt-Baildon M, Wille K, Merle U, Hower M, Rothfuss K, Nadalin S, Klinker H, Fürst J, Greiffendorf I, Raichle C, Friedrichs A, Rauschning D, de With K, Eberwein L, Riedel C, Milovanovic M, Worm M, Schultheis B, Schubert J, Bota M, Beutel G, Glück T, Schmid M, Wintermantel T, Peetz H, Steiner S, Ribel E, Schäfer H, Vehreschild JJ, Stecher M. Role and benefits of infectious diseases specialists in the COVID-19 pandemic: Multilevel analysis of care provision in German hospitals using data from the Lean European Open Survey on SARS-CoV-2 infected patients (LEOSS) cohort. Infection 2025; 53:259-269. [PMID: 39150640 PMCID: PMC11825572 DOI: 10.1007/s15010-024-02362-2] [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: 01/09/2024] [Accepted: 07/23/2024] [Indexed: 08/17/2024]
Abstract
PURPOSE This study investigates the care provision and the role of infectious disease (ID) specialists during the coronavirus disease-2019 (COVID-19) pandemic. METHODS A survey was conducted at German study sites participating in the Lean European Open Survey on SARS-CoV-2 infected patients (LEOSS). Hospitals certified by the German Society of Infectious diseases (DGI) were identified as ID centers. We compared care provision and the involvement of ID specialists between ID and non-ID hospitals. Then we applied a multivariable regression model to analyse how clinical ID care influenced the mortality of COVID-19 patients in the LEOSS cohort. RESULTS Of the 40 participating hospitals in the study, 35% (14/40) were identified as ID centers. Among those, clinical ID care structures were more commonly established, and ID specialists were always involved in pandemic management and the care of COVID-19 patients. Overall, 68% (27/40) of the hospitals involved ID specialists in the crisis management team, 78% (31/40) in normal inpatient care, and 80% (28/35) in intensive care. Multivariable analysis revealed that COVID-19 patients in ID centers had a lower mortality risk compared to those in non-ID centers (odds ratio: 0.61 (95% CI 0.40-0.93), p = 0.021). CONCLUSION ID specialists played a crucial role in pandemic management and inpatient care.
Collapse
Affiliation(s)
- Lene T Tscharntke
- Klinik I für Innere Medizin, Centrum für Integrierte Onkologie Aachen Bonn Köln Düsseldorf, Universität zu Köln, Medizinische Fakultät und Uniklinik Köln, Köln, Germany
| | - Norma Jung
- Klinik I für Innere Medizin, Centrum für Integrierte Onkologie Aachen Bonn Köln Düsseldorf, Universität zu Köln, Medizinische Fakultät und Uniklinik Köln, Köln, Germany
| | - Frank Hanses
- Universitätsklinikum Regensburg, Zentrale Notaufnahme, Regensburg, Germany
| | - Carolin E M Koll
- Klinik I Für Innere Medizin, Universität Zu Köln, Medizinische Fakultät Und Uniklinik Köln, Köln, Germany
- Deutsches Zentrum Für Infektionsforschung (DZIF), Partnerstandort Bonn-Köln, Cologne, Germany
| | - Lisa Pilgram
- Klinik Für Innere Medizin, Hämatologie Und Onkologie, Universitätsklinikum Frankfurt, Goethe Universität Frankfurt, Frankfurt am Main, Germany
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Siegbert Rieg
- Abteilung Infektiologie, Klinik Für Innere Medizin II, Universitätsklinikum Freiburg, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| | - Stefan Borgmann
- Abteilung Klinische Infektiologie Und Hygiene, Klinikum Ingolstadt, Ingolstadt, Germany
| | - Susana M Nunes de Miranda
- Klinik I Für Innere Medizin, Universität Zu Köln, Medizinische Fakultät Und Uniklinik Köln, Köln, Germany
| | - Margarete Scherer
- Klinik I Für Innere Medizin, Universität Zu Köln, Medizinische Fakultät Und Uniklinik Köln, Köln, Germany
| | - Christoph D Spinner
- Technische Universität München, Fakultät Für Medizin, Klinikum Rechts Der Isar, Klinik Und Poliklinik Für Innere Medizin II, Munich, Germany
| | - Maria Rüthrich
- Klinik für interdisziplinäre Intensivmedizin, Vivantes Humboldt-Klinikum Berlin, Berlin, Germany
| | - Maria J G T Vehreschild
- Deutsches Zentrum Für Infektionsforschung (DZIF), Partnerstandort Bonn-Köln, Cologne, Germany
- Klinik für Innere Medizin, Infektiologie, Universitätsklinikum Frankfurt, Goethe Universität Frankfurt, Frankfurt am Main, Germany
| | | | - Kai Wille
- Universität Bochum, Universitätsklinik für Hämatologie, Onkologie, Hämostaseologie und Palliativmedizin, Minden, Germany
| | - Uta Merle
- Abteilung für Gastroenterologie und Infektiologie, Universitätsklinikum Heidelberg, Heidelberg, Germany
| | - Martin Hower
- Klinikum Dortmund gGmbH, Klinik für Pneumologie, Infektiologie und internistische Intensivmedizin, Klinikum der Universität Witten/Herdecke, Dortmund, Germany
| | - Katja Rothfuss
- Abteilung für Gastroenterologie, Hepatologie und Endokrinologie, Robert-Bosch-Krankenhaus Stuttgart, Stuttgart, Germany
| | - Silvio Nadalin
- Universitätsklinik für Allgemein-, Viszeral- und Transplantationschirurgie, Universitätsklinikum Tübingen, Tübingen, Germany
| | - Hartwig Klinker
- Universitätsklinikum Würzburg, Medizinische Klinik und Poliklinik II, Infektiologie, Würzburg, Germany
| | - Julia Fürst
- Medizinische Klinik I, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Ingo Greiffendorf
- Kliniken Maria Hilf Mönchengladbach GmbH, Innere Medizin I, Klinik für Hämatologie, Onkologie und Gastroenterologie, Mönchengladbach, Germany
| | - Claudia Raichle
- Tropenklinik Paul-Lechler Krankenhaus Tübingen, Tübingen, Germany
| | - Anette Friedrichs
- Universitätsklinikum Schleswig-Holstein, Campus Kiel, Klinik für Innere Medizin I, Kiel, Germany
| | - Dominic Rauschning
- Klinik IB für Innere Medizin, Bundeswehrkrankenhaus Koblenz, Koblenz, Germany
| | - Katja de With
- Universitätsklinikum Carl Gustav Carus Dresden an der TU Dresden, Klinische Infektiologie, Dresden, Germany
| | - Lukas Eberwein
- Klinikum Leverkusen, Medizinische Klinik IV, Leverkusen, Germany
| | | | - Milena Milovanovic
- Malteser Krankenhaus St. Franziskus Hospital Flensburg, Flensburg, Germany
| | - Maximilian Worm
- Oberlausitz-Kliniken gGmbH/Krankenhäuser Bautzen und Bischofswerda, Bischofswerda, Germany
| | - Beate Schultheis
- Marien Hospital Herne, Universitätsklinikum der Ruhr-Universität Bochum, Bochum, Germany
| | | | - Marc Bota
- Agaplesion Bethesda Krankenhaus Bergedorf, Hamburg, Germany
| | - Gernot Beutel
- Medizinische Hochschule Hannover Klinik für Hämatologie, Hämostaseologie, Onkologie und Stammzelltransplantation, Hannover, Germany
| | - Thomas Glück
- Kliniken Südostbayern AG Trostberg, Trostberg, Germany
| | - Michael Schmid
- Medizinische Klinik I Hegau-Bodensee-Klinikum Singen, Singen, Germany
| | | | | | | | - Elena Ribel
- Hunsrück Klinik Kreuznacher Diakonie, Simmern, Germany
| | - Harald Schäfer
- SHG Kliniken Völklingen, Med. Klinik II, Pneumologie, Thorakale Onkologie, Infektiologie, Völklingen, Germany
| | - Jörg Janne Vehreschild
- Klinik I für Innere Medizin, Centrum für Integrierte Onkologie Aachen Bonn Köln Düsseldorf, Universität zu Köln, Medizinische Fakultät und Uniklinik Köln, Köln, Germany.
- Deutsches Zentrum Für Infektionsforschung (DZIF), Partnerstandort Bonn-Köln, Cologne, Germany.
- Klinik Für Innere Medizin, Hämatologie Und Onkologie, Universitätsklinikum Frankfurt, Goethe Universität Frankfurt, Frankfurt am Main, Germany.
| | - Melanie Stecher
- Klinik I Für Innere Medizin, Universität Zu Köln, Medizinische Fakultät Und Uniklinik Köln, Köln, Germany
- Deutsches Zentrum Für Infektionsforschung (DZIF), Partnerstandort Bonn-Köln, Cologne, Germany
| |
Collapse
|
34
|
Zendehdel A, Asoodeh A, Ansari M, JamaliMoghaddamsiyahkali S. The Investigation of the Distribution of ABO/Rh Blood Group in Hospitalized COVID-19 Patients and Its Association With Disease Severity, Clinical Outcomes, Lab Tests, and Radiologic Findings. Health Sci Rep 2025; 8:e70250. [PMID: 39931255 PMCID: PMC11808318 DOI: 10.1002/hsr2.70250] [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: 07/20/2023] [Revised: 09/24/2024] [Accepted: 11/17/2024] [Indexed: 02/13/2025] Open
Abstract
Background and Aims it is important to identify patients at higher risk for severity and poor outcomes of COVID-19 infection, to have better disease management and pandemic control. In this study, we aimed to assess the distribution of ABO and Rh blood groups in hospitalized COVID-19 infected patients and demonstrate its association with severity and outcomes of the disease. Methods This is a cross-sectional study at Ziaeian Specialist Hospital, in Tehran, Iran. Of all confirmed COVID-19 infected patients who were admitted to this hospital, 273 patients were enrolled in this study and categorized based on their disease severity or clinical outcomes including intensive care unit (ICU) admission, need for mechanical ventilation and mortality. The distribution of ABO and Rh blood groups was assessed and compared between different groups, to investigate the association of blood group types with disease severity or outcomes. Also, the study population was categorized based on their blood group types to demonstrate the association of laboratory parameters, radiologic findings, and length of hospitalization with blood groups. Sex, age and underlying disease were adjusted in the final model by multivariate regression analysis. Results This study showed that Blood group A (35.9%) was the most prevalent among hospitalized COVID-19 patients followed by O (34.8%), B (21.6%), and AB (7.7%) (A > O > B > AB). ABO and Rh blood group was not associated with disease severity, need for mechanical ventilation, or ICU admission, while blood group B was associated with an increased risk of death in comparison with type O, in hospitalized COVID-19 patients (p = 0.02). The number of patients with severe levels of C-reactive protein (CRP) test results was lower in O blood group patients in comparison with non-O blood groups (p = 0.01). Conclusion No significant association was found between blood groups and other lab tests, radiologic findings, and length of hospitalization.
Collapse
Affiliation(s)
- Abolfazl Zendehdel
- Internal Medicine Department, Ziaeian HospitalTehran University of Medical SciencesTehranIran
- Family Medicine Department, Ziaeian HospitalTehran University of Medical SciencesTehranIran
| | - Azadeh Asoodeh
- Department Family Medicine, Comprehensive Health CenterTehran University of Medical SciencesTehranIran
| | - Mohsen Ansari
- Radiology Department, Amir al‐Momenin HospitalIslamic Azad University of Medical SciencesTehranIran
| | | |
Collapse
|
35
|
Kebriaei A, Besharati R, Namdar Ahmad Abad H, Havakhah S, Khosrojerdi M, Azimian A. The relationship between microRNAs and COVID-19 complications. Noncoding RNA Res 2025; 10:16-24. [PMID: 39296641 PMCID: PMC11406673 DOI: 10.1016/j.ncrna.2024.08.007] [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: 04/02/2024] [Revised: 08/20/2024] [Accepted: 08/21/2024] [Indexed: 09/21/2024] Open
Abstract
Over the past three years, since the onset of COVID-19, several scientific studies have concentrated on understanding susceptibility to the virus, the progression of the illness, and possible long-term complexity. COVID-19 is broadly recognized with effects on multiple systems in the body, and various factors related to society, medicine, and genetics/epigenetics may contribute to the intensity and results of the disease. Additionally, a SARS-CoV-2 infection can activate pathological activities and expedite the emergence of existing health issues into clinical problems. Forming easily accessible, distinctive, and permeable biomarkers is essential for categorizing patients, preventing the disease, predicting its course, and tailoring treatments for COVID-19 individually. One promising candidate for such biomarkers is microRNAs, which could serve various purposes in understanding diverse forms of COVID-19, including susceptibility, intensity, disease progression, outcomes, and potential therapeutic options. This review provides an overview of the most significant findings related to the involvement of microRNAs in COVID-19 pathogenesis. Furthermore, it explores the function of microRNAs in a broad span of effects that may arise from accompanying or underlying health status. It underscores the value of comprehending how diverse conditions, such as neurological disorders, diabetes, cardiovascular diseases, and obesity, interact with COVID-19.
Collapse
Affiliation(s)
- Abdollah Kebriaei
- Department of Pathobiology and Laboratory Sciences, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Reza Besharati
- Department of Pathobiology and Laboratory Sciences, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Hasan Namdar Ahmad Abad
- Department of Pathobiology and Laboratory Sciences, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Shahrzad Havakhah
- Department of Physiology, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Mahsa Khosrojerdi
- Department of Immunology and Allergy, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Azimian
- Department of Pathobiology and Laboratory Sciences, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| |
Collapse
|
36
|
Malaspina P, Jodice C, Ciminelli BM, Biancolella M, Colona VL, Latini A, Leonardis F, Rogliani P, Novelli A, Novelli G, Novelletto A. Genetic diversity of the immunoglobulin heavy chain locus in cohorts of patients affected with SARS-CoV-2. Hum Genomics 2025; 19:7. [PMID: 39885568 PMCID: PMC11780896 DOI: 10.1186/s40246-025-00719-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 01/17/2025] [Indexed: 02/01/2025] Open
Abstract
BACKGROUND The Immunoglobulin Heavy Chain (IGH) genomic region is responsible for the production of circulating antibodies and warrants careful investigation for its association with COVID-19 characteristics. Multiple allelic variants within and across different IGH gene segments form a limited set of haplotypes. Previous studies have shown associations between some of these haplotypes and clinical outcomes of COVID-19. We typed 445 individuals of European ancestry, stratified for gender, age, and clinical status for 4 SNPs, two of which result in amino acid substitutions in IGHA2 and IGHG4, respectively. We analyzed associations at the single-locus level and for 4-loci haplotypes, inferred by phasing, after stratifying the overall cohort by gender, age, and disease severity. RESULTS Only weak evidence of significant differences between subgroups was obtained at the level of a single SNP. However, when the haplotypic data were analyzed for the young and old subgroups separately, uneven partitioning was observed regarding the occurrence of severe cases and Resistors. We then examined the cross-tabulation of disease severity in males and females, based on the presence of each haplotype in the genotype. Two haplotypes were underrepresented in young severe cases compared to old severe ones. The same two haplotypes were overrepresented among young Resistors. These findings provide stronger support for, the weak associations observed at the single locus level. CONCLUSIONS Two haplotypes seem to act as protective factors specifically in young individuals, counteracting the general increase in vulnerability with age. This observation aligns with stronger genetic effects seen in young patients for other susceptibility genes. Our findings complement previous research identifying specific genetic variants that influence COVID-19 susceptibility and severity, emphasizing the complex interplay between host genetics and viral infection outcomes. Our results are consistent with a potential causative role of IGH regulatory regions (e.g. HS1.2), which are flanked by the SNP set here analyzed.
Collapse
Affiliation(s)
- Patrizia Malaspina
- Department of Biology, Tor Vergata University of Rome, Via della Ricerca Scientifica 1, 00133, Rome, Italy.
| | - Carla Jodice
- Department of Biology, Tor Vergata University of Rome, Via della Ricerca Scientifica 1, 00133, Rome, Italy
| | - Bianca Maria Ciminelli
- Department of Biology, Tor Vergata University of Rome, Via della Ricerca Scientifica 1, 00133, Rome, Italy
| | - Michela Biancolella
- Department of Biology, Tor Vergata University of Rome, Via della Ricerca Scientifica 1, 00133, Rome, Italy
| | - Vito Luigi Colona
- Research Unit of Neurorehabilitation, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Andrea Latini
- Department of Biomedicine and Prevention, Tor Vergata University of Rome, Rome, Italy
| | | | | | - Antonio Novelli
- Laboratory of Medical Genetics, Translational Cytogenomics Research Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Giuseppe Novelli
- Department of Biomedicine and Prevention, Tor Vergata University of Rome, Rome, Italy
- Tor Vergata University Hospital, Rome, Italy
| | - Andrea Novelletto
- Department of Biology, Tor Vergata University of Rome, Via della Ricerca Scientifica 1, 00133, Rome, Italy
| |
Collapse
|
37
|
Chaddock NJM, Crossfield SSR, Pujades-Rodriguez M, Iles MM, Morgan AW. Genetic proxies for clinical traits are associated with increased risk of severe COVID-19. Sci Rep 2025; 15:2083. [PMID: 39814935 PMCID: PMC11736077 DOI: 10.1038/s41598-025-86260-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 01/09/2025] [Indexed: 01/18/2025] Open
Abstract
Routine use of genetic data in healthcare is much-discussed, yet little is known about its performance in epidemiological models including traditional risk factors. Using severe COVID-19 as an exemplar, we explore the integration of polygenic risk scores (PRS) into disease models alongside sociodemographic and clinical variables. PRS were optimized for 23 clinical variables and related traits previously-associated with severe COVID-19 in up to 450,449 UK Biobank participants, and tested in 9,560 individuals diagnosed in the pre-vaccination era. Associations were further adjusted for (i) sociodemographic and (ii) clinical variables. Pathway analyses of PRS were performed to improve biological understanding of disease. In univariate analyses, 17 PRS were associated with increased risk of severe COVID-19 and, of these, four remained associated with COVID-19 outcomes following adjustment for sociodemographic/clinical variables: hypertension PRS (OR = 1.1, 95%CI 1.03-1.18), atrial fibrillation PRS (OR = 1.12, 95%CI 1.03-1.22), peripheral vascular disease PRS (OR = 0.9, 95%CI 0.82-0.99), and Alzheimer's disease PRS (OR = 1.14, 95%CI 1.05-1.25). Pathway analyses revealed enrichment of genetic variants in pathways for cardiac muscle contraction (genes N = 5; beta[SE] = 3.48[0.60]; adjusted-P = 1.86 × 10-5). These findings underscore the potential for integrating genetic data into epidemiological models and highlight the advantages of utilizing multiple trait PRS rather than a single PRS for a specific outcome of interest.
Collapse
Affiliation(s)
- N J M Chaddock
- University of Leeds (School of Medicine and Leeds Institute for Data Analytics), Leeds, UK
| | - S S R Crossfield
- University of Leeds (School of Medicine and Leeds Institute for Data Analytics), Leeds, UK
| | - M Pujades-Rodriguez
- University of Leeds (School of Medicine and Leeds Institute for Data Analytics), Leeds, UK
| | - M M Iles
- University of Leeds (School of Medicine and Leeds Institute for Data Analytics), Leeds, UK
- Leeds Teaching Hospitals NHS Trust (NIHR Leeds Biomedical Research Centre and NIHR Leeds Medtech and In vitro Diagnostics Co-operative), Leeds, UK
| | - A W Morgan
- University of Leeds (School of Medicine and Leeds Institute for Data Analytics), Leeds, UK.
- Leeds Teaching Hospitals NHS Trust (NIHR Leeds Biomedical Research Centre and NIHR Leeds Medtech and In vitro Diagnostics Co-operative), Leeds, UK.
| |
Collapse
|
38
|
Laffranchi M, Paraboschi EM, Bianchetto-Aguilera F, Tamassia N, Gasperini S, Gardiman E, Piserà A, Del Prete A, Invernizzi P, Gismondi A, Mantovani A, Cassatella MA, Asselta R, Sozzani S. Neutrophils restricted contribution of CCRL2 genetic variants to COVID-19 severity. Heliyon 2025; 11:e41267. [PMID: 39811276 PMCID: PMC11731188 DOI: 10.1016/j.heliyon.2024.e41267] [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: 07/23/2024] [Revised: 11/12/2024] [Accepted: 12/14/2024] [Indexed: 01/02/2025] Open
Abstract
The 3p21.31 locus is the most robust genomic region associated with COVID-19 severity. This locus contains a main chemokine receptor (CKR) cluster. We tested expression quantitative trait loci (eQTL) targeting the 3p21.31 CKR cluster linked to COVID-19 hospitalization in Europeans from the COVID-19 HGI meta-analysis. Among these, CCRL2, a key regulator of neutrophil trafficking, was targeted by neutrophil-restricted eQTLs. We confirmed these eQTLs in an Italian COVID-19 cohort. Haplotype analysis revealed a link between an increased CCRL2 expression and COVID-19 severity and hospitalization. By the exposure of neutrophils to a TLR8 ligand, reflecting a viral infection, we revealed specific chromatin domains within the 3p21.31 locus exclusive to neutrophils. In addition, the identified variants mapped within these regions altered the binding motif of neutrophils-expressed transcription factors. These results support that CCRL2 eQTL variants contribute to the risk of severe COVID-19 by selectively affecting neutrophil functions.
Collapse
Affiliation(s)
- Mattia Laffranchi
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- Istituto Pasteur-Fondazione Cenci Bolognetti, Rome, Italy
| | - Elvezia Maria Paraboschi
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | | | - Nicola Tamassia
- Department of Medicine, Section of General Pathology, University of Verona, 37134, Verona, Italy
| | - Sara Gasperini
- Department of Medicine, Section of General Pathology, University of Verona, 37134, Verona, Italy
| | - Elisa Gardiman
- Department of Medicine, Section of General Pathology, University of Verona, 37134, Verona, Italy
| | - Arianna Piserà
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Annalisa Del Prete
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Pietro Invernizzi
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, European Reference Network on Hepatological Diseases (ERN RARE-LIVER), IRCCS Fondazione San Gerardo Dei Tintori, Monza, Italy
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Angela Gismondi
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- Istituto Pasteur-Fondazione Cenci Bolognetti, Rome, Italy
| | - Alberto Mantovani
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
- The William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Marco A. Cassatella
- Department of Medicine, Section of General Pathology, University of Verona, 37134, Verona, Italy
| | - Rosanna Asselta
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Silvano Sozzani
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- Istituto Pasteur-Fondazione Cenci Bolognetti, Rome, Italy
| |
Collapse
|
39
|
Halligan NLN, Hanks SC, Matsuo K, Martins T, Zöllner S, Quasney MW, Scott LJ, Dahmer MK. Variants in the β-globin locus are associated with pneumonia in African American children. HGG ADVANCES 2025; 6:100374. [PMID: 39444160 PMCID: PMC11664401 DOI: 10.1016/j.xhgg.2024.100374] [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: 03/29/2024] [Revised: 10/14/2024] [Accepted: 10/14/2024] [Indexed: 10/25/2024] Open
Abstract
In African American adults, the strongest genetic predictor of pneumonia appears to be the A allele of rs334, a variant in the β-globin gene, which in homozygous form causes sickle cell disease (SCD). No comparable studies have been done in African American children. We performed genome-wide association analyses of 482 African American children with documented pneumonia and 2,048 African American control individuals using genotypes imputed from two reference panels: 1000 Genomes (1KG) (which contains rs334) and TOPMed (does not contain rs334). Using 1KG imputed genotypes, the most significant variant was rs334 (A allele; odds ratio [OR] = 2.76; 95% CI, 2.21-3.74; p = 5.9 × 10-19); using TOPMed imputed genotypes the most significant variant was rs2226952, found in the β-globin locus control region (G allele; OR = 2.14; 95% CI, 1.78-2.57; p = 5.1 × 10-16). After conditioning on rs334, the most strongly associated variant in the β-globin locus, rs33930165 (T allele, 1KG: OR = 4.09; 95% CI, 2.29-7.29; p = 1.7 × 10-6; TOPMed: OR = 3.58; 95% CI, 2.18-5.90; p = 4.7 × 10-7), which as a compound heterozygote with rs334 A allele, can cause SCD. To compare the power of different sample sets we developed a way to estimate the power of sample sets with different sample sizes, genotype arrays, and imputation platforms. Our results suggest that, in African American children, the strongest genetic determinants of pneumonia are those that increase the risk of SCD.
Collapse
Affiliation(s)
- Nadine L N Halligan
- Division of Critical Care Medicine, Department of Pediatrics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sarah C Hanks
- Department of Biostatistics and Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - Karen Matsuo
- Department of Biostatistics and Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - Taylor Martins
- Department of Biostatistics and Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sebastian Zöllner
- Department of Biostatistics and Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA; Department of Psychiatry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Michael W Quasney
- Division of Critical Care Medicine, Department of Pediatrics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Laura J Scott
- Department of Biostatistics and Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Mary K Dahmer
- Division of Critical Care Medicine, Department of Pediatrics, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
40
|
Zhou X, Wu Y, Zhu Z, Lu C, Zhang C, Zeng L, Xie F, Zhang L, Zhou F. Mucosal immune response in biology, disease prevention and treatment. Signal Transduct Target Ther 2025; 10:7. [PMID: 39774607 PMCID: PMC11707400 DOI: 10.1038/s41392-024-02043-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 09/05/2024] [Accepted: 10/27/2024] [Indexed: 01/11/2025] Open
Abstract
The mucosal immune system, as the most extensive peripheral immune network, serves as the frontline defense against a myriad of microbial and dietary antigens. It is crucial in preventing pathogen invasion and establishing immune tolerance. A comprehensive understanding of mucosal immunity is essential for developing treatments that can effectively target diseases at their entry points, thereby minimizing the overall impact on the body. Despite its importance, our knowledge of mucosal immunity remains incomplete, necessitating further research. The outbreak of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has underscored the critical role of mucosal immunity in disease prevention and treatment. This systematic review focuses on the dynamic interactions between mucosa-associated lymphoid structures and related diseases. We delve into the basic structures and functions of these lymphoid tissues during disease processes and explore the intricate regulatory networks and mechanisms involved. Additionally, we summarize novel therapies and clinical research advances in the prevention of mucosal immunity-related diseases. The review also addresses the challenges in developing mucosal vaccines, which aim to induce specific immune responses while maintaining tolerance to non-pathogenic microbes. Innovative therapies, such as nanoparticle vaccines and inhalable antibodies, show promise in enhancing mucosal immunity and offer potential for improved disease prevention and treatment.
Collapse
Affiliation(s)
- Xiaoxue Zhou
- School of Medicine, Hangzhou City University, Hangzhou, China
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Yuchen Wu
- The First School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhipeng Zhu
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Chu Lu
- The First Affiliated Hospital, the Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Chunwu Zhang
- The First School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Linghui Zeng
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Feng Xie
- The First Affiliated Hospital, the Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China.
| | - Long Zhang
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China.
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Fangfang Zhou
- The First Affiliated Hospital, the Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
41
|
Gonzalez Aleman G, Vavougios GD, Tartaglia C, Uvais NA, Guekht A, Hosseini AA, Lo Re V, Ferreccio C, D'Avossa G, Zamponi HP, Figueredo Aguiar M, Yecora A, Ul Haq Katshu MZ, Stavrou VT, Boutlas S, Gourgoulianis KI, Botero C, González Insúa F, Perez-Lloret S, Zinchuk M, Gersamija A, Popova S, Bryzgalova Y, Sviatskaya E, Russelli G, Avorio F, Wang S, Edison P, Niimi Y, Sohrabi HR, Mukaetova Ladinska EB, Neidre D, de Erausquin GA. Age-dependent phenotypes of cognitive impairment as sequelae of SARS-CoV-2 infection. Front Aging Neurosci 2025; 16:1432357. [PMID: 39839305 PMCID: PMC11747492 DOI: 10.3389/fnagi.2024.1432357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 10/28/2024] [Indexed: 01/23/2025] Open
Abstract
Cognitive changes associated with PASC may not be uniform across populations. We conducted individual-level pooled analyses and meta-analyses of cognitive assessments from eight prospective cohorts, comprising 2,105 patients and 1,432 controls from Argentina, Canada, Chile, Greece, India, Italy, Russia, and the UK. The meta-analysis found no differences by country of origin. The profile and severity of cognitive impairment varied by age, with mild attentional impairment observed in young and middle-aged adults, but memory, language, and executive function impairment in older adults. The risk of moderate to severe impairment doubled in older adults. Moderately severe or severe impairment was significantly associated with infection diagnoses (chi-square = 26.57, p ≤ 0.0001) and the severity of anosmia (chi-square = 31.81, p ≤ 0.0001). We found distinct age-related phenotypes of cognitive impairment in patients recovering from COVID-19. We identified the severity of acute illness and the presence of olfactory dysfunction as the primary predictors of dementia-like impairment in older adults.
Collapse
Affiliation(s)
- Gabriela Gonzalez Aleman
- Department of Psychology, School of Psychology and Psychopedagogy, Universidad Catolica Argentina, Buenos Aires, Argentina
| | - George D. Vavougios
- Department of Neurology, Medical School, University of Cyprus, Nicosia, Cyprus
- Department of Respiratory Medicine, University of Thessaly, Larissa, Greece
| | - Carmela Tartaglia
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
- Memory Clinic, Department of Neurology, Toronto Western Hospital, Toronto, ON, Canada
| | - Nalakath A. Uvais
- Department of Psychiatry, Iqraa International Hospital and Research Centre, Calicut, India
| | - Alla Guekht
- Department of Neurology, Moscow Research and Clinical Centre for Neuropsychiatry, Moscow, Russia
- Department of Neurology, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Akram A. Hosseini
- Nottingham University Hospitals NHS Trust, Nottingham, United Kingdom
- Nottingham Sir Peter Mansfield Imaging Centre, University of Nottingham, Nottingham, United Kingdom
| | - Vincenzina Lo Re
- Neurology Service, Department of Diagnostic and Therapeutic Services, IRCCS ISMETT, Palermo, Italy
- Department of Experimental Medicine and Clinical Neuroscience, University of Pittsburgh Medical Center (UPMC), Palermo, Italy
| | - Catterina Ferreccio
- Department of Public Health School of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases, ACCDiS, Santiago, Chile
| | - Giovanni D'Avossa
- School of Psychology and Sports Sciences, Bangor University, Bangor, United Kingdom
| | - Hernan P. Zamponi
- Secretariat for Mental Health and Addictions, Ministry of Health, Government of Jujuy, San Salvador de Jujuy, Argentina
| | - Mariana Figueredo Aguiar
- Instituto San Lazaro de Neurociencias, Fundacion de Lucha contra los Trastornos Neurologicos y Psiquiatricos en Minorias, FULTRA, San Salvador de Jujuy, Argentina
| | - Agustin Yecora
- Secretariat for Mental Health and Addictions, Ministry of Health, Government of Jujuy, San Salvador de Jujuy, Argentina
- Instituto San Lazaro de Neurociencias, Fundacion de Lucha contra los Trastornos Neurologicos y Psiquiatricos en Minorias, FULTRA, San Salvador de Jujuy, Argentina
| | - Mohammad Zia Ul Haq Katshu
- Institute of Mental Health, University of Nottingham, Nottinghamshire Healthcare NHS Foundation Trust, Nottingham, United Kingdom
| | - Vasileios T. Stavrou
- Department of Neurology, Medical School, University of Cyprus, Nicosia, Cyprus
- Department of Respiratory Medicine, University of Thessaly, Larissa, Greece
| | - Stylianos Boutlas
- Department of Neurology, Medical School, University of Cyprus, Nicosia, Cyprus
| | | | - Camila Botero
- Department of Psychology, School of Psychology and Psychopedagogy, Universidad Catolica Argentina, Buenos Aires, Argentina
| | - Francisco González Insúa
- Department of Psychology, School of Psychology and Psychopedagogy, Universidad Catolica Argentina, Buenos Aires, Argentina
| | - Santiago Perez-Lloret
- Health Observatory, Vice Rectorate for Research, Universidad Catolica Argentina, Buenos Aires, Argentina
| | - Mikhail Zinchuk
- Department of Neurology, Moscow Research and Clinical Centre for Neuropsychiatry, Moscow, Russia
| | - Anna Gersamija
- Department of Neurology, Moscow Research and Clinical Centre for Neuropsychiatry, Moscow, Russia
| | - Sofya Popova
- Department of Neurology, Moscow Research and Clinical Centre for Neuropsychiatry, Moscow, Russia
| | - Yulia Bryzgalova
- Department of Neurology, Moscow Research and Clinical Centre for Neuropsychiatry, Moscow, Russia
| | - Ekaterina Sviatskaya
- Department of Neurology, Moscow Research and Clinical Centre for Neuropsychiatry, Moscow, Russia
| | - Giovanna Russelli
- Neurology Service, Department of Diagnostic and Therapeutic Services, IRCCS ISMETT, Palermo, Italy
- Department of Experimental Medicine and Clinical Neuroscience, University of Pittsburgh Medical Center (UPMC), Palermo, Italy
| | - Federica Avorio
- Neurology Service, Department of Diagnostic and Therapeutic Services, IRCCS ISMETT, Palermo, Italy
- Department of Experimental Medicine and Clinical Neuroscience, University of Pittsburgh Medical Center (UPMC), Palermo, Italy
| | - Sophia Wang
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States
- Indiana Alzheimer's Disease Research Center, Indianapolis, IN, United States
| | - Paul Edison
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom
- Cardiff University, Cardiff, United Kingdom
| | - Yoshiki Niimi
- Faculty of Medicine, University of Tokyo, Tokyo, Japan
| | - Hamid R. Sohrabi
- Murdoch University Centre for Healthy Ageing, School of Psychology, Murdoch University, Murdoch, WA, Australia
| | - Elizabeta B. Mukaetova Ladinska
- Department of Psychology and Visual Sciences, University of Leicester, Leicester, United Kingdom
- The Evington Centre, Leicester General Hospital, Leicester, United Kingdom
| | - Daria Neidre
- Laboratory for Brain Development, Modulation and Repair, Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health San Antonio, San Antonio, TX, United States
| | - Gabriel A. de Erausquin
- Laboratory for Brain Development, Modulation and Repair, Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health San Antonio, San Antonio, TX, United States
- Laboratory of Electrophysiology Imaging, Radiology Research Institute, University of Texas Health San Antonio, San Antonio, TX, United States
- Department of Neurology, Joe & Teresa Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX, United States
| |
Collapse
|
42
|
Zailani H, Satyanarayanan SK, Liao WC, Su KP, Chang JPC. Omega-3 Polyunsaturated Fatty Acids in Chronic Obstructive Pulmonary Disease Patients with COVID-19: A Review. Curr Nutr Rep 2025; 14:12. [PMID: 39760917 DOI: 10.1007/s13668-024-00599-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/06/2024] [Indexed: 01/07/2025]
Abstract
PURPOSE OF THE REVIEW Mounting evidence indicates that individuals with chronic obstructive pulmonary disease (COPD) face a heightened risk of severe outcomes upon contracting coronavirus disease 2019 (COVID-19). Current medications for COVID-19 often carry side effects, necessitating alternative therapies with improved tolerance. This review explores the biological mechanisms rendering COPD patients more susceptible to severe COVID-19 and investigates the potential of omega-3 polyunsaturated fatty acids (n-3 PUFAs) in mitigating the severity of COVID-19 in COPD patients. RECENT FINDINGS Current evidence indicates that COPD patients are at an increased risk of severe COVID-19 due to factors including compromised pulmonary function, dysregulated inflammation, weakened immune response, increased oxidative stress, elevated expression of angiotensin-converting enzyme (ACE2) receptors in the lungs, and genetic predispositions. Remarkably, n-3 PUFAs exhibit the potential in ameliorating the clinical outcomes of COPD patients with COVID-19 by modulating inflammation, reinforcing the body's antioxidant defenses, reducing viral entry and replication, and enhancing immunity. N-3 PUFAs hold potential for improving COVID-19 outcomes in patients with COPD. However, there has been limited investigation into the therapeutic effects of n-3 PUFAs in enhancing clinical outcomes for COPD patients. Rigorous clinical studies are essential to evaluate the impact of n-3 PUFAs on COPD patients with concurrent COVID-19 infection.
Collapse
Grants
- MOST 109-2320-B-038-057- MY3, 110-2321-B-006-004, 110-2811-B-039-507, 110-2320-B-039-048-MY2,110-2320-B-039- 047-MY3, 110-2813-C-039-327-B, 110-2314-B-039-029-MY3, 111-2321-B-006-008, and NSTC 111-2314-B-039-041-MY3 Ministry of Science and Technology, Taiwan
- MOST 109-2320-B-038-057- MY3, 110-2321-B-006-004, 110-2811-B-039-507, 110-2320-B-039-048-MY2,110-2320-B-039- 047-MY3, 110-2813-C-039-327-B, 110-2314-B-039-029-MY3, 111-2321-B-006-008, and NSTC 111-2314-B-039-041-MY3 Ministry of Science and Technology, Taiwan
- ANHRF 109-31, 109-40, 110-13, 110-26, 110-44, 110-45, 111-27, 111-28, 111-47, 111-48, and 111-52 An-Nan Hospital, China Medical University, Tainan, Taiwan
- CMRC-CMA-2 Higher Education Sprout Project by the Ministry of Education, Taiwan
- CMRC-CMA-2 Higher Education Sprout Project by the Ministry of Education, Taiwan
- CMU 110- AWARD-02, 110-N-17, 1110-SR-73 China Medical University, Taiwan
- CMU 110- AWARD-02, 110-N-17, 1110-SR-73 China Medical University, Taiwan
- DMR-106-101, 106-227, 109-102, 109-244, 110-124, 111-245, 112-097, 112-086, 112-109 and DMR-HHC-109-11, HHC-109-12, HHC-110-10, and HHC-111-8 China Medical University Hospital
- DMR-106-101, 106-227, 109-102, 109-244, 110-124, 111-245, 112-097, 112-086, 112-109 and DMR-HHC-109-11, HHC-109-12, HHC-110-10, and HHC-111-8 China Medical University Hospital
Collapse
Affiliation(s)
- Halliru Zailani
- Mind-Body Interface Research Center (MBI-Lab), China Medical University Hospital, Taichung, Taiwan
- Graduate Institute of Nutrition, China Medical University, Taichung, Taiwan
- Department of Biochemistry, Ahmadu Bello University, Zaria, Nigeria
| | - Senthil Kumaran Satyanarayanan
- Mind-Body Interface Research Center (MBI-Lab), China Medical University Hospital, Taichung, Taiwan
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong Science Park, Hong Kong, China
| | - Wei-Chih Liao
- Division of Pulmonary and Critical Medicine, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan.
- College of Medicine, China Medical University, Taichung, Taiwan.
| | - Kuan-Pin Su
- Mind-Body Interface Research Center (MBI-Lab), China Medical University Hospital, Taichung, Taiwan
- College of Medicine, China Medical University, Taichung, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Jane Pei-Chen Chang
- Mind-Body Interface Research Center (MBI-Lab), China Medical University Hospital, Taichung, Taiwan.
- College of Medicine, China Medical University, Taichung, Taiwan.
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.
- Child and Adolescent Psychiatry Division, Department of Psychiatry, China Medical University Hospital, No. 2 Yu-Der Rd, North District, Taichung, 404, Taiwan.
| |
Collapse
|
43
|
Mózner O, Szabó E, Kulin A, Várady G, Moldvay J, Vass V, Szentesi A, Jánosi Á, Hegyi P, Sarkadi B. Potential associations of selected polymorphic genetic variants with COVID-19 disease susceptibility and severity. PLoS One 2025; 20:e0316396. [PMID: 39752416 PMCID: PMC11698323 DOI: 10.1371/journal.pone.0316396] [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: 08/08/2024] [Accepted: 12/10/2024] [Indexed: 01/06/2025] Open
Abstract
In this study, we analyzed the potential associations of selected laboratory and anamnestic parameters, as well as 12 genetic polymorphisms (SNPs), with clinical COVID-19 occurrence and severity in 869 hospitalized patients. The SNPs analyzed by qPCR were selected based on population-wide genetic (GWAS) data previously indicating association with the severity of COVID-19, and additional SNPs that have been shown to be important in cellular processes were also examined. We confirmed the associations of COVID-19 with pre-existing diabetes and found an unexpected association between less severe disease and the loss of smell and taste. Regarding the genetic polymorphisms, a higher allele frequency of the LZTFL1 and IFNAR2 minor variants significantly correlated with greater COVID-19 disease susceptibility (hospitalization) and severity, and a similar tendency was observed for the RAVER1 and the MUC5B variants. Interestingly, the ATP2B4 minor haplotype, protecting against malaria, correlated with an increased disease susceptibility, while in diabetic patients disease susceptibility was lower in the presence of a reduced-function ABCG2 transporter variant. Our current results, which should be reinforced by larger studies, indicate that together with laboratory and anamnestic parameters, genetic polymorphisms may have predictive value for the clinical occurrence and severity of COVID-19.
Collapse
Affiliation(s)
- Orsolya Mózner
- Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
- Doctoral School, Semmelweis University, Budapest, Hungary
| | - Edit Szabó
- Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
| | - Anna Kulin
- Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
- Doctoral School, Semmelweis University, Budapest, Hungary
| | - György Várady
- Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
| | - Judit Moldvay
- 1 Department of Pulmonology, National Korányi Institute of Pulmonology
- Department of Pulmonology, University of Szeged Albert Szent-Györgyi Medical School
| | - Vivien Vass
- Institute for Translational Medicine, University of Pécs, Medical School, Pécs, Hungary
- Institute of Pancreatic Diseases and Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Andrea Szentesi
- Institute for Translational Medicine, University of Pécs, Medical School, Pécs, Hungary
| | - Ágoston Jánosi
- Institute for Translational Medicine, University of Pécs, Medical School, Pécs, Hungary
- Heim Pál National Pediatric Institute, Budapest, Hungary
| | - Péter Hegyi
- Institute for Translational Medicine, University of Pécs, Medical School, Pécs, Hungary
- Institute of Pancreatic Diseases and Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Balázs Sarkadi
- Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
- Doctoral School, Semmelweis University, Budapest, Hungary
| |
Collapse
|
44
|
Spalinger MR, Sanati G, Chatterjee P, Hai R, Li J, Santos AN, Nordgren TM, Tremblay ML, Eckmann L, Hanson E, Scharl M, Wu X, Boland BS, McCole DF. Tofacitinib Mitigates the Increased SARS-CoV-2 Infection Susceptibility Caused by an IBD Risk Variant in the PTPN2 Gene. Cell Mol Gastroenterol Hepatol 2025; 19:101447. [PMID: 39756517 PMCID: PMC11953972 DOI: 10.1016/j.jcmgh.2024.101447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 01/07/2025]
Abstract
BACKGROUND & AIMS Coronavirus disease (COVID-19), caused by severe acquired respiratory syndrome-Coronavirus-2 (SARS-CoV-2), triggered a global pandemic with severe medical and socioeconomic consequences. Although fatality rates are higher among the elderly and those with underlying comorbidities, host factors that promote susceptibility to SARS-CoV-2 infection and severe disease are poorly understood. Although individuals with certain autoimmune/inflammatory disorders show increased susceptibility to viral infections, there is incomplete knowledge of SARS-CoV-2 susceptibility in these diseases. The aim of our study was to investigate whether the autoimmunity risk gene, PTPN2, which also confers elevated risk to develop inflammatory bowel disease, affects susceptibility to SARS-CoV-2 viral uptake. METHODS Using samples from PTPN2 genotyped patients with inflammatory bowel disease, PTPN2-deficient mice, and human intestinal and lung epithelial cell lines, we investigated how PTPN2 affects expression of the SARS-CoV-2 receptor angiotensin converting enzyme 2 (ACE2), and uptake of virus-like particles expressing the SARS-CoV2 spike protein and live SARS-CoV-2 virus. RESULTS We report that the autoimmune PTPN2 loss-of-function risk variant rs1893217 promotes expression of the SARS-CoV-2 receptor, ACE2, and increases cellular entry of SARS-CoV-2 spike protein and live virus. Elevated ACE2 expression and viral entry were mediated by increased Janus kinase-signal transducers and activators of transcription signaling and were reversed by the Janus kinase inhibitor, tofacitinib. CONCLUSION Collectively, our findings uncover a novel risk biomarker for increased expression of the SARS-CoV-2 receptor and viral entry, and identify a clinically approved therapeutic agent to mitigate this risk.
Collapse
Affiliation(s)
- Marianne R Spalinger
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California; Department of Gastroenterology and Hepatology, University Hospital Zurich, and University of Zurich, Zurich, Switzerland
| | - Golshid Sanati
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California
| | - Pritha Chatterjee
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California
| | - Rong Hai
- Department of Microbiology and Plant Pathology, University of California Riverside, Riverside, California
| | - Jiang Li
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California
| | - Alina N Santos
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California
| | - Tara M Nordgren
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California; Current position: College of Veterinary Medicine, Colorado State University, Fort Collins, Colorado
| | - Michel L Tremblay
- Department of Biochemistry and Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
| | - Lars Eckmann
- Division of Gastroenterology, University of California San Diego, La Jolla, California
| | - Elaine Hanson
- Division of Gastroenterology, University of California San Diego, La Jolla, California
| | - Michael Scharl
- Department of Gastroenterology and Hepatology, University Hospital Zurich, and University of Zurich, Zurich, Switzerland
| | - Xiwei Wu
- Integrative Genomics Core, Beckman Research Institute of City of Hope, Monrovia, California
| | - Brigid S Boland
- Division of Gastroenterology, University of California San Diego, La Jolla, California
| | - Declan F McCole
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California.
| |
Collapse
|
45
|
Chen S, Zhang Q, Zhang X, Xie P, Guo H, Lu F, Zhou C, Dong F. Casual correlation between overweight, obesity, and severe COVID-19 infection with respiratory failure: A two-sample Mendelian randomization. Medicine (Baltimore) 2025; 104:e41006. [PMID: 40184103 PMCID: PMC11709160 DOI: 10.1097/md.0000000000041006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 11/21/2024] [Accepted: 12/01/2024] [Indexed: 04/05/2025] Open
Abstract
This study aimed to detect the causal association of overweight and obesity on severe COVID-19 infection with respiratory failure through a two-sample Mendelian randomization (MR) method based on the genome-wide association studies datasets. All genome-wide association studies summary data of exposures and outcome used in this study were obtained from the IEU database derived from Europeans. The study mainly used the inverse variance weighted method to test causal relationship. Simultaneously, MR-PRESSO and MR-EGGER were used to detect the pleiotropy, and sensitivity analysis was performed using leave-one-out analysis. In the inverse variance weighted analyses, we found no causal association between obesity (e.g., OR = 1.15, 95% CIs = 0.96-1.37, P = .13 for obesity-ebi-a-GCST90000255), obesity subtypes (e.g., OR = 1.93, 95% CIs = 0.90-4.14, P = .10 for obesity and other hyperalimentation) as well as overweight (OR = 0.90, 95% CIs = 0.64-1.27, P = .54) and severe COVID-19 infection with respiratory failure. The findings showed no causal association between obesity or overweight and severe COVID-19 infection with respiratory failure. Further validation is needed regarding whether obesity or overweight is a risk factor for it.
Collapse
Affiliation(s)
- Shiqiang Chen
- Department of Emergency and Critical Care Medicine, Yuhuan People’s Hospital, Taizhou, China
| | - Qiang Zhang
- Department of Emergency and Critical Care Medicine, Yuhuan People’s Hospital, Taizhou, China
| | - Xiaobing Zhang
- Department of Emergency and Critical Care Medicine, Yuhuan People’s Hospital, Taizhou, China
| | - Peiyao Xie
- Department of Emergency and Critical Care Medicine, Yuhuan People’s Hospital, Taizhou, China
| | - Hua Guo
- Department of Emergency and Critical Care Medicine, Yuhuan People’s Hospital, Taizhou, China
| | - Fengling Lu
- Department of Emergency and Critical Care Medicine, Yuhuan People’s Hospital, Taizhou, China
| | - Chaoyang Zhou
- Department of Critical Care Medicine, Yuhuan People’s Hospital, Taizhou, China
| | - Fubo Dong
- Department of Critical Care Medicine, Yuhuan People’s Hospital, Taizhou, China
| |
Collapse
|
46
|
El Houdi M, Skhoun H, El Fessikh M, Benmansour R, El Yousfi FZ, Nebhani C, Tagajdid MR, Lahlou Amine I, El Annaz H, Ameziane El Hassani R, Ouzzif Z, Abouqal R, Ennibi K, Bouhouche A, El Baghdadi J. Association study of the JAK/STAT signaling pathway with susceptibility to COVID-19 in moroccan patient and in-silico analysis of rare variants. Virus Res 2025; 351:199509. [PMID: 39647533 PMCID: PMC11699608 DOI: 10.1016/j.virusres.2024.199509] [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: 09/10/2024] [Revised: 12/02/2024] [Accepted: 12/05/2024] [Indexed: 12/10/2024]
Abstract
The goal of our study was to explore the association of the polymorphisms in the JAK/STAT pathway among Moroccan COVID-19 patients, using a case-control approach. Next-generation sequencing was employed to investigate the IFNAR1, IFNAR2, JAK1, TYK2, STAT2, and IRF9 genes within the JAK/STAT pathway. We also performed an in silico study to examine the rare variants in this pathway. Statistical analyses were conducted using MedCalc software. Protein 3D structures were determined via the I-TASSER server, with variant structures generated using PyMOL. YASARA View allowed local 3D analysis comparing native and variant structures for pathogenic rare variants. The study encompassed 206 COVID-19 patients, averaging 45.70 ± 12.73 years and a control group (N=118). Among the examined genes, 15 common polymorphisms and 7 rare variants were identified. Adjustment for age and gender revealed a significant association between TYK2 p.Gly363Ser (p=0.036) and COVID-19 infection, where the GA variant exhibited protective effects (0.6361 [0.3405-1.1884], p=0.035). Additionally, STAT2 p.Met594Ile showed an association to COVID-19 risk (p=0.042), with heterozygous GC being linked to infection (p=0.037, OR=2.7135 [0.5684 -12.9532]). Notably, IFNAR1 p.Val168Leu mutated C allele was significantly associated with reduced susceptibility to COVID-19 severity (p=0.028, OR=0.5936 [0.3725 - 0.9461]), under the additive model (p=0.045, OR=0.626 [0.3958 - 0.9899]). Rare variants IFNAR1 p.Trp318Cys, p.Ser476Phe, and IFNAR2 p.Cys271Tyr were predicted deleterious, impacting protein structure via hydrogen bond and hydrophobic interaction alterations. Burden analysis of rare variants revealed a protective cumulative effect against COVID-19 severity for TYK2 (p=0.0013, OR=0.1438 [0.04237 - 0.4803]) under the dominant model. This study underscores the role of genetic factors in COVID-19 susceptibility and advocates further explorations regarding functional impacts of JAK/STAT pathway rare variants.
Collapse
Affiliation(s)
- Meriem El Houdi
- Genetics Unit, Military Hospital Mohammed V, Rabat, Morocco; Laboratory of Human Pathologies Biology and Genomic Center of Human Pathologies, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Morocco.
| | - Hanaa Skhoun
- Genetics Unit, Military Hospital Mohammed V, Rabat, Morocco; Laboratory of Human Pathologies Biology and Genomic Center of Human Pathologies, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Morocco.
| | - Meriem El Fessikh
- Genetics Unit, Military Hospital Mohammed V, Rabat, Morocco; Laboratory of Human Pathologies Biology and Genomic Center of Human Pathologies, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Morocco.
| | - Reda Benmansour
- Virology Laboratory, Center of Virology, Infectious and Tropical Diseases, Military Hospital Mohammed V, School of Medicine and Pharmacy, University Mohammed V, Rabat, Morocco.
| | - Fatima-Zahra El Yousfi
- Laboratory of Human Genetics, Medical School and Pharmacy, University Mohammed V in Rabat, Morocco.
| | - Chaimae Nebhani
- Laboratory of Human Genetics, Medical School and Pharmacy, University Mohammed V in Rabat, Morocco.
| | - Mohamed Rida Tagajdid
- Virology Laboratory, Center of Virology, Infectious and Tropical Diseases, Military Hospital Mohammed V, School of Medicine and Pharmacy, University Mohammed V, Rabat, Morocco.
| | - Idriss Lahlou Amine
- Virology Laboratory, Center of Virology, Infectious and Tropical Diseases, Military Hospital Mohammed V, School of Medicine and Pharmacy, University Mohammed V, Rabat, Morocco.
| | - Hicham El Annaz
- Virology Laboratory, Center of Virology, Infectious and Tropical Diseases, Military Hospital Mohammed V, School of Medicine and Pharmacy, University Mohammed V, Rabat, Morocco.
| | - Rabii Ameziane El Hassani
- Laboratory of Human Pathologies Biology and Genomic Center of Human Pathologies, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Morocco.
| | - Zohra Ouzzif
- Laboratories Pole, Military Hospital Mohammed V, Rabat, Morocco.
| | - Redouane Abouqal
- Laboratory of Biostatistics, Clinical and Epidemiological Research, Department of Public Health, Medical School and Pharmacy, University Mohammed V in Rabat, Morocco.
| | - Khalid Ennibi
- Center of Virology, Infectious and Tropical Diseases, Military Hospital Mohammed V, Rabat, Morocco.
| | - Ahmed Bouhouche
- Laboratory of Human Genetics, Medical School and Pharmacy, University Mohammed V in Rabat, Morocco; Genomic Center of the Cheikh Zaid Foundation, Abulcasis International University of Health Sciences, Rabat, Morocco.
| | - Jamila El Baghdadi
- Genetics Unit, Military Hospital Mohammed V, Rabat, Morocco; Laboratories Pole, Military Hospital Mohammed V, Rabat, Morocco.
| |
Collapse
|
47
|
Carvalho-Rassbach M, Ventura LHA, Assis LO, Fabrini S, Natividade MMP, Silva JL, Gomes KB, Faria AMC, Alvarez-Leite JI. The Apoprotein E4 isotype does not affect the severity of COVID-19 infection and other flu-like syndromes. J Med Microbiol 2025; 74. [PMID: 39841509 DOI: 10.1099/jmm.0.001951] [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] [Indexed: 01/23/2025] Open
Abstract
Introduction. Apolipoprotein E (ApoE), especially the ApoE4 isotype, is suggested to influence the severity of respiratory viral infections; however, this association is still unclear.Hypothesis. The presence of allele ε4 impacts the development of flu-like syndromes.Aim. This study aimed to evaluate the impact of the Apo E4 isoform on the severity and duration of flu-like syndromes, including the coronavirus disease COVID-19.Methodology. This study comprised 280 individuals presenting flu-like symptoms, all genotyped for ApoE isoforms. Data were collected on clinical course, comorbidities, nutritional status, biochemical and inflammatory markers, SARS-CoV-2 reverse transcription PCR results and disease severity (mild, moderate or severe) according to the World Health Organization criteria. The individuals were analysed as a whole and within subgroups based on the SARS-CoV-2-positive (COVID-19 group) or SARS-CoV-2-negative (flu-like syndrome group) test.Results. The frequency of the ε4 allele was similar across the whole population and in both the COVID-19 and flu-like syndrome subgroups (17 and 18%, respectively). No differences were seen in sex, age range, self-reported skin colour, body mass index (BMI), number of comorbidities, vaccination status, biochemical, cytokine and lipid profiles (except for total cholesterol) in the flu-like group when ε4 allele carriers and non-carriers were compared. In the COVID-19 group, the ε4 allele did not correlate with disease severity or duration, number of comorbidities or inflammatory biomarkers. While gender distribution was equal in the overall COVID-19 population, male gender strongly correlated with COVID-19 severity. Multivariate analysis showed that older individuals, male gender, higher BMI and the presence of comorbidities were linked to increased chances of developing moderate and severe disease. IL-4 was the only factor found to reduce the risk of severe COVID-19.Conclusion. The presence of one ɛ4 allele showed no association with the duration and severity of flu-like syndromes, including COVID-19. Nonetheless, SARS-CoV-2-positive individuals tend to be older men with a higher BMI and a tendency to be overweight or with obesity. Regarding COVID-19 severity, BMI, male sex and the number of associated comorbidities were the factors that increased the chance of developing a more severe form of COVID-19.
Collapse
Affiliation(s)
- Marina Carvalho-Rassbach
- Departamento de Bioqumica e Imunologia, Instituto de Cincias Biolgicas, Universidade Federal de Minas Gerais
| | - Lucas Haniel Araujo Ventura
- Departamento de Bioqumica e Imunologia, Instituto de Cincias Biolgicas, Universidade Federal de Minas Gerais
| | - Larissa Oliveira Assis
- Departamento de Bioqumica e Imunologia, Instituto de Cincias Biolgicas, Universidade Federal de Minas Gerais
| | - Sabrina Fabrini
- Departamento de Bioqumica e Imunologia, Instituto de Cincias Biolgicas, Universidade Federal de Minas Gerais
| | | | - Janayne Luihan Silva
- Departamento de Bioqumica e Imunologia, Instituto de Cincias Biolgicas, Universidade Federal de Minas Gerais
| | - Karina Braga Gomes
- Departamento de Anlises Clnicas e Toxicolgicas da Faculdade de Farmcia, Universidade Federal de Minas Gerais
| | - Ana Maria Caetano Faria
- Departamento de Bioqumica e Imunologia, Instituto de Cincias Biolgicas, Universidade Federal de Minas Gerais
| | - Jacqueline I Alvarez-Leite
- Departamento de Bioqumica e Imunologia, Instituto de Cincias Biolgicas, Universidade Federal de Minas Gerais
| |
Collapse
|
48
|
Liu N, Guan M, Ma B, Chu H, Tian G, Zhang Y, Li C, Zheng W, Wang X. Unraveling genetic mysteries: A comprehensive review of GWAS and DNA insights in animal and plant pathosystems. Int J Biol Macromol 2025; 285:138216. [PMID: 39631605 DOI: 10.1016/j.ijbiomac.2024.138216] [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: 08/19/2024] [Revised: 11/13/2024] [Accepted: 11/28/2024] [Indexed: 12/07/2024]
Abstract
DNA serves as the carrier of genetic information, with sequence variations playing a pivotal role in defining hereditary traits. Genome-Wide Association Studies (GWAS) facilitate the investigation of the links between genetic variations and phenotypes, significantly influencing biological research, particularly in animal and plant pathology. By identifying genetic markers associated with specific traits or diseases, GWAS enhances our understanding of host-pathogen interactions and improves disease-resistant breeding strategies. It has been vital in revealing the genetic basis of disease resistance, pinpointing key genes and DNA loci, which enrich genetic resources for breeding programs and deepen our knowledge of disease resistance mechanisms at the DNA level. Additionally, GWAS contributes to pathogen population genetics, facilitating a thorough exploration of pathogen virulence. Integrating GWAS with marker-assisted selection enhances breeding efficiency and precision in selecting for disease-resistant traits. While previous research has largely focused on host genetics, the genetic variation of pathogens is equally significant. Notably, reports integrating animal and plant pathosystems are still lacking. Given the importance of these systems, this review summarizes key advancements in this field, addresses current challenges, and proposes future directions, thereby offering a vital reference for ongoing research.
Collapse
Affiliation(s)
- Na Liu
- Collaborative Innovation Center of Henan Grain Crops/State Key Laboratory of Wheat and Maize Crop Science, College of Life Sciences, Henan Agricultural University, 450046 Zhengzhou, China
| | - Mengxin Guan
- Collaborative Innovation Center of Henan Grain Crops/State Key Laboratory of Wheat and Maize Crop Science, College of Life Sciences, Henan Agricultural University, 450046 Zhengzhou, China
| | - Baozhan Ma
- Collaborative Innovation Center of Henan Grain Crops/State Key Laboratory of Wheat and Maize Crop Science, College of Life Sciences, Henan Agricultural University, 450046 Zhengzhou, China
| | - Hao Chu
- Collaborative Innovation Center of Henan Grain Crops/State Key Laboratory of Wheat and Maize Crop Science, College of Life Sciences, Henan Agricultural University, 450046 Zhengzhou, China
| | - Guangxiang Tian
- Collaborative Innovation Center of Henan Grain Crops/State Key Laboratory of Wheat and Maize Crop Science, College of Life Sciences, Henan Agricultural University, 450046 Zhengzhou, China
| | - Yanyan Zhang
- Collaborative Innovation Center of Henan Grain Crops/State Key Laboratory of Wheat and Maize Crop Science, College of Life Sciences, Henan Agricultural University, 450046 Zhengzhou, China
| | - Chuang Li
- Collaborative Innovation Center of Henan Grain Crops/State Key Laboratory of Wheat and Maize Crop Science, College of Life Sciences, Henan Agricultural University, 450046 Zhengzhou, China; Center of Crop Genome Engineering, College of Agronomy, Henan Agricultural University, 450046 Zhengzhou, China.
| | - Wenming Zheng
- Collaborative Innovation Center of Henan Grain Crops/State Key Laboratory of Wheat and Maize Crop Science, College of Life Sciences, Henan Agricultural University, 450046 Zhengzhou, China.
| | - Xu Wang
- Collaborative Innovation Center of Henan Grain Crops/State Key Laboratory of Wheat and Maize Crop Science, College of Life Sciences, Henan Agricultural University, 450046 Zhengzhou, China.
| |
Collapse
|
49
|
Tang D, Liu X, He J. Association between single-nucleotide polymorphisms of ADAMTS13 and severe COVID-19: a case-control study in Chinese Han adults. Arch Med Sci 2024; 20:2067-2071. [PMID: 39967935 PMCID: PMC11831343 DOI: 10.5114/aoms/199514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 12/25/2024] [Indexed: 02/20/2025] Open
Affiliation(s)
- Daoyan Tang
- Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan Province, China
| | - Xiaoshu Liu
- Department of Respiratory and Critical Care Medicine, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Jianqing He
- Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan Province, China
| |
Collapse
|
50
|
Safo SE, Lu H. Scalable randomized kernel methods for multiview data integration and prediction with application to Coronavirus disease. Biostatistics 2024; 26:kxaf001. [PMID: 39973130 PMCID: PMC11839864 DOI: 10.1093/biostatistics/kxaf001] [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: 06/30/2023] [Revised: 12/17/2024] [Accepted: 12/18/2024] [Indexed: 02/21/2025] Open
Abstract
There is still more to learn about the pathobiology of coronavirus disease (COVID-19) despite 4 years of the pandemic. A multiomics approach offers a comprehensive view of the disease and has the potential to yield deeper insight into the pathogenesis of the disease. Previous multiomics integrative analysis and prediction studies for COVID-19 severity and status have assumed simple relationships (ie linear relationships) between omics data and between omics and COVID-19 outcomes. However, these linear methods do not account for the inherent underlying nonlinear structure associated with these different types of data. The motivation behind this work is to model nonlinear relationships in multiomics and COVID-19 outcomes, and to determine key multidimensional molecules associated with the disease. Toward this goal, we develop scalable randomized kernel methods for jointly associating data from multiple sources or views and simultaneously predicting an outcome or classifying a unit into one of 2 or more classes. We also determine variables or groups of variables that best contribute to the relationships among the views. We use the idea that random Fourier bases can approximate shift-invariant kernel functions to construct nonlinear mappings of each view and we use these mappings and the outcome variable to learn view-independent low-dimensional representations. We demonstrate the effectiveness of the proposed methods through extensive simulations. When the proposed methods were applied to gene expression, metabolomics, proteomics, and lipidomics data pertaining to COVID-19, we identified several molecular signatures for COVID-19 status and severity. Our results agree with previous findings and suggest potential avenues for future research. Our algorithms are implemented in Pytorch and interfaced in R and available at: https://github.com/lasandrall/RandMVLearn.
Collapse
Affiliation(s)
- Sandra E Safo
- Division of Biostatistics and Health Data Science, University of Minnesota, 2221 University Ave SE, Minneapolis, MN 55414, United States
| | - Han Lu
- Division of Biostatistics and Health Data Science, University of Minnesota, 2221 University Ave SE, Minneapolis, MN 55414, United States
| |
Collapse
|