1
|
Cekin N, Akin S, Pinarbasi E, Doğan OH. Impact of IL-6 rs1800795 and rs1800796 polymorphisms on clinical outcomes of COVID-19: a study on severity of disease in Turkish population. Mamm Genome 2025; 36:213-229. [PMID: 39567384 DOI: 10.1007/s00335-024-10085-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 11/09/2024] [Indexed: 11/22/2024]
Abstract
Coronavirus disease (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is exacerbated by cytokine storms, leading to severe inflammation. Interleukin-6 (IL-6) plays a critical role in this process, and variations in its promoter may influence disease severity. This study aims to investigate the relationship between IL6 promoter polymorphisms rs1800795 (G > C) and rs1800796 (G > C) and the severity of COVID-19 in the Turkish population. A total of 332 participants were included: 84 control, 80 with mild COVID-19, and 168 with severe COVID-19. IL6 polymorphisms were genotyped using the restriction fragment length polymorphism (RFLP) method. The genotypes rs1800795 GC (OR = 3.00, 95% CI: 1.669-5.398, p < 0.000), CC (OR = 7.44, 95% CI: 2.899-19.131, p < 0.000), and rs1800796 GC (OR = 2.76, 95% CI: 1.603-4.761, p < 0.000), as well as the alleles rs1800795 C (OR = 3.01, p < 0.000) and rs1800796 C (OR = 1.97, p = 0.002), may be associated with the severity of COVID-19. According to the Jonckheere-Terpstra (J-T) test, the most significant trends that vary linearly with disease severity were observed for D-dimer [J-T = 15.896, Effect size = 0.68 (0.61 to 0.76), p < 0.000] and CRP [J-T = 15.389, Effect size = 0.66 (0.59 to 0.73), p < 0.000]. The distribution of clinical parameters across genotype combinations (rs1800796/rs1800795*) showed that GC/GC* and GC/CC* were linked to a higher risk of severe inflammation, clotting, and organ damage. Additionally, it has been determined that the G-C and C-C haplotypes may be associated with increased severity of COVID-19. The rs1800795 and rs1800796 polymorphisms are linked to COVID-19 severity and could help guide future treatment strategies.
Collapse
Affiliation(s)
- Nilgun Cekin
- Department of Medical Biology, Faculty of Medicine, Sivas Cumhuriyet University, Sivas, Turkey.
- Faculty of Medicine, Department of Medical Biochemistry, Sivas Cumhuriyet University, Sivas, Turkey.
| | - Seyda Akin
- Department of Medical Biology, Faculty of Medicine, Sivas Cumhuriyet University, Sivas, Turkey
| | - Ergun Pinarbasi
- Department of Medical Biology, Faculty of Medicine, Sivas Cumhuriyet University, Sivas, Turkey
| | - Okan Halef Doğan
- Department of Medical Biology, Faculty of Medicine, Sivas Cumhuriyet University, Sivas, Turkey
| |
Collapse
|
2
|
Molino C, Bergantini L, Santucci S, Pitinca MT, d'Alessandro M, Cameli P, Taddei S, Bargagli E. SARS-CoV-2 and Dysphagia: A Retrospective Analysis of COVID-19 Patients with Swallowing Disorders. Dysphagia 2025; 40:162-168. [PMID: 38782803 PMCID: PMC11762431 DOI: 10.1007/s00455-024-10715-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 04/25/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND COVID-19 can lead to impairment of neural networks involved in swallowing, since the act of swallowing is coordinated and performed by a diffuse brain network involving peripheral nerves and muscles. Dysphagia has been identified as a risk and predictive factor for the severest form of SARS-CoV-2 infection. OBJECTIVES To investigate the association between swallowing disorders and COVID-19 in patients hospitalized for COVID-19. METHODS We collected demographic data, medical information specific to dysphagia and data on medical treatments of patients with COVID-19. RESULTS A total of 43 hospitalized COVID-19 patients were enrolled in the study. Twenty (46%) were evaluated positive for dysphagia and 23 (54%) were evaluated negative. Neurocognitive disorders and diabetes were mostly associated with patients who resulted positive for dysphagia. Respiratory impairment caused by COVID-19 seems to be a cause of dysphagia, since all patients who needed oxygen-therapy developed symptoms of dysphagia, unlike patients who did not. In the dysphagic group, alteration of the swallowing trigger resulted in the severest form of dysphagia. An association was found between the severest form of COVID-19 and dysphagia. This group consisted predominantly of males with longer hospitalization. CONCLUSIONS Identification of COVID-19 patients at risk for dysphagia is crucial for better patient management.
Collapse
Affiliation(s)
- Christopher Molino
- Department of Medical Sciences, Surgery and Neurosciences, Respiratory Disease and Lung Transplant Unit, University Hospital of Siena (Azienda Ospedaliera Universitaria Senese, AOUS), Siena University, Viale Bracci, Siena, 53100, Italy
| | - Laura Bergantini
- Department of Medical Sciences, Surgery and Neurosciences, Respiratory Disease and Lung Transplant Unit, University Hospital of Siena (Azienda Ospedaliera Universitaria Senese, AOUS), Siena University, Viale Bracci, Siena, 53100, Italy.
| | | | | | - Miriana d'Alessandro
- Department of Medical Sciences, Surgery and Neurosciences, Respiratory Disease and Lung Transplant Unit, University Hospital of Siena (Azienda Ospedaliera Universitaria Senese, AOUS), Siena University, Viale Bracci, Siena, 53100, Italy
| | - Paolo Cameli
- Department of Medical Sciences, Surgery and Neurosciences, Respiratory Disease and Lung Transplant Unit, University Hospital of Siena (Azienda Ospedaliera Universitaria Senese, AOUS), Siena University, Viale Bracci, Siena, 53100, Italy
| | | | - Elena Bargagli
- Department of Medical Sciences, Surgery and Neurosciences, Respiratory Disease and Lung Transplant Unit, University Hospital of Siena (Azienda Ospedaliera Universitaria Senese, AOUS), Siena University, Viale Bracci, Siena, 53100, Italy
| |
Collapse
|
3
|
Bergantini L, Gangi S, d'Alessandro M, Cameli P, Perea B, Meocci M, Fabbri G, Bianchi F, Bargagli E. Altered serum concentrations of IL-8, IL-32 and IL-10 in patients with lung impairment 6 months after COVID-19. Immunobiology 2024; 229:152813. [PMID: 38805808 DOI: 10.1016/j.imbio.2024.152813] [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: 12/13/2023] [Revised: 05/03/2024] [Accepted: 05/23/2024] [Indexed: 05/30/2024]
Abstract
Post-COVID symptoms are reported in 10-35 % of patients not requiring hospitalization, and in up to 80 % of hospitalized patients and patients with severe disease. The pathogenesis of post-COVID syndrome remains largely unknown. Some evidence suggests that prolonged inflammation has a key role in the pathogenesis of most post-COVID manifestations. We evaluated a panel of inflammatory and immune-mediated cytokines in individuals with altered HRCT features and in patients without any long-term COVID symptoms. Blood samples of 89 adult patients previously hospitalized with COVID-19 were collected and stratified as patients with and without HRCT evidence of fibrotic lung alterations. Serum analyte concentrations of IL-4, IL-2, CXCL10 (IP-10), IL-1β, TNF-α, CCL2 (MCP-1), IL-17A, IL-6, IL-10, IFN-γ, IL-12p70 and TGF-β1 (free active form) were quantified by bead-based multiplex assay. Clinical and functional data were recorded in a database. With the use of machine learning approach, IL-32, IL-8, and IL-10 proved to be associated with the development of HRCT evidence of lung sequelae at follow-up. Direct comparison of cytokine levels in the two groups showed increased levels of IL-32 and decreased levels of IL-8 in patients with lung impairment. After further stratification of patients by severity (severe versus mild/moderate) during hospitalization, IL-10 emerged as the only cytokine showing decreased levels in severe patients. These findings contribute to a better understanding of the immune response and potential prognostic markers in patients with lung sequelae after COVID-19.
Collapse
Affiliation(s)
- Laura Bergantini
- Department of Medical Science, Surgery and Neuroscience, Respiratory Disease and Lung Transplant Unit, University of Siena, Italy
| | - Sara Gangi
- Department of Medical Science, Surgery and Neuroscience, Respiratory Disease and Lung Transplant Unit, University of Siena, Italy
| | - Miriana d'Alessandro
- Department of Medical Science, Surgery and Neuroscience, Respiratory Disease and Lung Transplant Unit, University of Siena, Italy.
| | - Paolo Cameli
- Department of Medical Science, Surgery and Neuroscience, Respiratory Disease and Lung Transplant Unit, University of Siena, Italy
| | - Beatrice Perea
- Department of Medical Science, Surgery and Neuroscience, Respiratory Disease and Lung Transplant Unit, University of Siena, Italy
| | - Martina Meocci
- Department of Medical Science, Surgery and Neuroscience, Respiratory Disease and Lung Transplant Unit, University of Siena, Italy
| | - Gaia Fabbri
- Department of Medical Science, Surgery and Neuroscience, Respiratory Disease and Lung Transplant Unit, University of Siena, Italy
| | - Francesco Bianchi
- Department of Medical Science, Surgery and Neuroscience, Respiratory Disease and Lung Transplant Unit, University of Siena, Italy
| | - Elena Bargagli
- Department of Medical Science, Surgery and Neuroscience, Respiratory Disease and Lung Transplant Unit, University of Siena, Italy
| |
Collapse
|
4
|
Martelloni G, Turchi A, Fallerini C, Degl’Innocenti A, Baldassarri M, Olmi S, Furini S, Renieri A. Host genetics and COVID-19 severity: increasing the accuracy of latest severity scores by Boolean quantum features. Front Genet 2024; 15:1362469. [PMID: 38841724 PMCID: PMC11150643 DOI: 10.3389/fgene.2024.1362469] [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: 12/28/2023] [Accepted: 04/09/2024] [Indexed: 06/07/2024] Open
Abstract
The impact of common and rare variants in COVID-19 host genetics has been widely studied. In particular, in Fallerini et al. (Human genetics, 2022, 141, 147-173), common and rare variants were used to define an interpretable machine learning model for predicting COVID-19 severity. First, variants were converted into sets of Boolean features, depending on the absence or the presence of variants in each gene. An ensemble of LASSO logistic regression models was used to identify the most informative Boolean features with respect to the genetic bases of severity. After that, the Boolean features, selected by these logistic models, were combined into an Integrated PolyGenic Score (IPGS), which offers a very simple description of the contribution of host genetics in COVID-19 severity.. IPGS leads to an accuracy of 55%-60% on different cohorts, and, after a logistic regression with both IPGS and age as inputs, it leads to an accuracy of 75%. The goal of this paper is to improve the previous results, using not only the most informative Boolean features with respect to the genetic bases of severity but also the information on host organs involved in the disease. In this study, we generalize the IPGS adding a statistical weight for each organ, through the transformation of Boolean features into "Boolean quantum features," inspired by quantum mechanics. The organ coefficients were set via the application of the genetic algorithm PyGAD, and, after that, we defined two new integrated polygenic scores (IPGS p h 1 and IPGS p h 2 ). By applying a logistic regression with both IPGS, (IPGS p h 2 (or indifferently IPGS p h 1 ) and age as inputs, we reached an accuracy of 84%-86%, thus improving the results previously shown in Fallerini et al. (Human genetics, 2022, 141, 147-173) by a factor of 10%.
Collapse
Affiliation(s)
| | - Alessio Turchi
- INAF Osservatorio Astrofisico di Arcetri, Florence, Italy
| | - Chiara Fallerini
- Medical Genetics, University of Siena, Siena, Italy
- Department of Medical Biotechnologies, Med Biotech Hub and Competence Center, University of Siena, Siena, Italy
| | - Andrea Degl’Innocenti
- Department of Medical Biotechnologies, Med Biotech Hub and Competence Center, University of Siena, Siena, Italy
| | - Margherita Baldassarri
- Medical Genetics, University of Siena, Siena, Italy
- Department of Medical Biotechnologies, Med Biotech Hub and Competence Center, University of Siena, Siena, Italy
| | - Simona Olmi
- CNR-Consiglio Nazionale delle Ricerche—Istituto dei Sistemi Complessi, Sesto Fiorentino, Italy
| | - Simone Furini
- Department of Electrical, Electronic and Information Engineering “Guglielmo Marconi”, University of Bologna, Cesena, Italy
| | - Alessandra Renieri
- Medical Genetics, University of Siena, Siena, Italy
- Department of Medical Biotechnologies, Med Biotech Hub and Competence Center, University of Siena, Siena, Italy
- Genetica Medica, Azienda Ospedaliero-Universitaria Senese, Siena, Italy
| |
Collapse
|
5
|
Farias TD, Brugiapaglia S, Croci S, Magistroni P, Curcio C, Zguro K, Fallerini C, Fava F, Pettini F, Kichula KM, Pollock NR, Font-Porterias N, Palmer WH, Marin WM, Baldassarri M, Bruttini M, Hollenbach JA, Hendricks AE, Meloni I, Novelli F, GEN-COVID Multicenter Study Group, Renieri A, Furini S, Norman PJ, Amoroso A. HLA-DPB1*13:01 associates with enhanced, and KIR2DS4*001 with diminished protection from developing severe COVID-19. HLA 2024; 103:e15251. [PMID: 37850268 PMCID: PMC10873037 DOI: 10.1111/tan.15251] [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: 01/04/2023] [Revised: 08/22/2023] [Accepted: 09/26/2023] [Indexed: 10/19/2023]
Abstract
Extreme polymorphism of HLA and killer-cell immunoglobulin-like receptors (KIR) differentiates immune responses across individuals. Additional to T cell receptor interactions, subsets of HLA class I act as ligands for inhibitory and activating KIR, allowing natural killer (NK) cells to detect and kill infected cells. We investigated the impact of HLA and KIR polymorphism on the severity of COVID-19. High resolution HLA class I and II and KIR genotypes were determined from 403 non-hospitalized and 1575 hospitalized SARS-CoV-2 infected patients from Italy collected in 2020. We observed that possession of the activating KIR2DS4*001 allotype is associated with severe disease, requiring hospitalization (OR = 1.48, 95% CI 1.20-1.85, pc = 0.017), and this effect is greater in individuals homozygous for KIR2DS4*001 (OR = 3.74, 95% CI 1.75-9.29, pc = 0.003). We also observed the HLA class II allotype, HLA-DPB1*13:01 protects SARS-CoV-2 infected patients from severe disease (OR = 0.49, 95% CI 0.33-0.74, pc = 0.019). These association analyses were replicated using logistic regression with sex and age as covariates. Autoantibodies against IFN-α associated with COVID-19 severity were detected in 26% of 156 hospitalized patients tested. HLA-C*08:02 was more frequent in patients with IFN-α autoantibodies than those without, and KIR3DL1*01502 was only present in patients lacking IFN-α antibodies. These findings suggest that KIR and HLA polymorphism is integral in determining the clinical outcome following SARS-CoV-2 infection, by influencing the course both of innate and adaptive immunity.
Collapse
Affiliation(s)
- Ticiana D.J. Farias
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, Colorado, 80045, USA
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, 80045, USA
| | - Silvia Brugiapaglia
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, 10126, Italy
- Laboratory of Tumor Immunology Center for Experimental Research and Medical Studies, Città della Salute e della Scienza di Torino, Turin, 10126, Italy
| | - Susanna Croci
- Medical Genetics, University of Siena, Siena, 53100, Italy
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, 53100, Italy
| | - Paola Magistroni
- Immunogenetics and Transplant Biology, Azienda Ospedaliera Universitaria, Città della Salute e della Scienza di Torino, Turin, 10126, Italy
| | - Claudia Curcio
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, 10126, Italy
- Laboratory of Tumor Immunology Center for Experimental Research and Medical Studies, Città della Salute e della Scienza di Torino, Turin, 10126, Italy
| | - Kristina Zguro
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, 53100, Italy
| | - Chiara Fallerini
- Medical Genetics, University of Siena, Siena, 53100, Italy
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, 53100, Italy
| | - Francesca Fava
- Medical Genetics, University of Siena, Siena, 53100, Italy
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, 53100, Italy
- Genetica Medica, Azienda Ospedaliero-Universitaria Senese, Siena, 53100, Italy
| | - Francesco Pettini
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, 53100, Italy
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, 53100, Italy
| | - Katherine M. Kichula
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, Colorado, 80045, USA
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, 80045, USA
| | - Nicholas R. Pollock
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, Colorado, 80045, USA
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, 80045, USA
| | - Neus Font-Porterias
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, Colorado, 80045, USA
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, 80045, USA
| | - William H. Palmer
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, Colorado, 80045, USA
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, 80045, USA
| | - Wesley M. Marin
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Margherita Baldassarri
- Medical Genetics, University of Siena, Siena, 53100, Italy
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, 53100, Italy
| | - Mirella Bruttini
- Medical Genetics, University of Siena, Siena, 53100, Italy
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, 53100, Italy
- Genetica Medica, Azienda Ospedaliero-Universitaria Senese, Siena, 53100, Italy
| | - Jill A. Hollenbach
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Audrey E. Hendricks
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, Colorado, 80045, USA
- Department of Mathematical and Statistical Sciences, and Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, Colorado, 80045, USA
| | - Ilaria Meloni
- Medical Genetics, University of Siena, Siena, 53100, Italy
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, 53100, Italy
| | - Francesco Novelli
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, 10126, Italy
- Laboratory of Tumor Immunology Center for Experimental Research and Medical Studies, Città della Salute e della Scienza di Torino, Turin, 10126, Italy
- Molecular Biotechnology Center, University of Turin, Turin, 10126, Italy
| | | | - Alessandra Renieri
- Medical Genetics, University of Siena, Siena, 53100, Italy
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, 53100, Italy
- Genetica Medica, Azienda Ospedaliero-Universitaria Senese, Siena, 53100, Italy
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, 53100, Italy
| | - Simone Furini
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, 53100, Italy
| | - Paul J. Norman
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, Colorado, 80045, USA
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, 80045, USA
| | - Antonio Amoroso
- Immunogenetics and Transplant Biology, Azienda Ospedaliera Universitaria, Città della Salute e della Scienza di Torino, Turin, 10126, Italy
- Department of Medical Sciences, University of Turin, Turin, 10126, Italy
| |
Collapse
|
6
|
Stanelle-Bertram S, Beck S, Mounogou NK, Schaumburg B, Stoll F, Al Jawazneh A, Schmal Z, Bai T, Zickler M, Beythien G, Becker K, de la Roi M, Heinrich F, Schulz C, Sauter M, Krasemann S, Lange P, Heinemann A, van Riel D, Leijten L, Bauer L, van den Bosch TPP, Lopuhaä B, Busche T, Wibberg D, Schaudien D, Goldmann T, Lüttjohann A, Ruschinski J, Jania H, Müller Z, Pinho Dos Reis V, Krupp-Buzimkic V, Wolff M, Fallerini C, Baldassarri M, Furini S, Norwood K, Käufer C, Schützenmeister N, von Köckritz-Blickwede M, Schroeder M, Jarczak D, Nierhaus A, Welte T, Kluge S, McHardy AC, Sommer F, Kalinowski J, Krauss-Etschmann S, Richter F, von der Thüsen J, Baumgärtner W, Klingel K, Ondruschka B, Renieri A, Gabriel G. CYP19A1 mediates severe SARS-CoV-2 disease outcome in males. Cell Rep Med 2023; 4:101152. [PMID: 37572667 PMCID: PMC10518605 DOI: 10.1016/j.xcrm.2023.101152] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 05/10/2023] [Accepted: 07/18/2023] [Indexed: 08/14/2023]
Abstract
Male sex represents one of the major risk factors for severe COVID-19 outcome. However, underlying mechanisms that mediate sex-dependent disease outcome are as yet unknown. Here, we identify the CYP19A1 gene encoding for the testosterone-to-estradiol metabolizing enzyme CYP19A1 (also known as aromatase) as a host factor that contributes to worsened disease outcome in SARS-CoV-2-infected males. We analyzed exome sequencing data obtained from a human COVID-19 cohort (n = 2,866) using a machine-learning approach and identify a CYP19A1-activity-increasing mutation to be associated with the development of severe disease in men but not women. We further analyzed human autopsy-derived lungs (n = 86) and detect increased pulmonary CYP19A1 expression at the time point of death in men compared with women. In the golden hamster model, we show that SARS-CoV-2 infection causes increased CYP19A1 expression in the lung that is associated with dysregulated plasma sex hormone levels and reduced long-term pulmonary function in males but not females. Treatment of SARS-CoV-2-infected hamsters with a clinically approved CYP19A1 inhibitor (letrozole) improves impaired lung function and supports recovery of imbalanced sex hormones specifically in males. Our study identifies CYP19A1 as a contributor to sex-specific SARS-CoV-2 disease outcome in males. Furthermore, inhibition of CYP19A1 by the clinically approved drug letrozole may furnish a new therapeutic strategy for individualized patient management and treatment.
Collapse
Affiliation(s)
| | - Sebastian Beck
- Department for Viral Zoonoses - One Health, Leibniz Institute of Virology, Hamburg, Germany
| | - Nancy Kouassi Mounogou
- Department for Viral Zoonoses - One Health, Leibniz Institute of Virology, Hamburg, Germany
| | - Berfin Schaumburg
- Department for Viral Zoonoses - One Health, Leibniz Institute of Virology, Hamburg, Germany
| | - Fabian Stoll
- Department for Viral Zoonoses - One Health, Leibniz Institute of Virology, Hamburg, Germany
| | - Amirah Al Jawazneh
- Department for Viral Zoonoses - One Health, Leibniz Institute of Virology, Hamburg, Germany
| | - Zoé Schmal
- Department for Viral Zoonoses - One Health, Leibniz Institute of Virology, Hamburg, Germany
| | - Tian Bai
- Department for Viral Zoonoses - One Health, Leibniz Institute of Virology, Hamburg, Germany
| | - Martin Zickler
- Department for Viral Zoonoses - One Health, Leibniz Institute of Virology, Hamburg, Germany
| | - Georg Beythien
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Kathrin Becker
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Madeleine de la Roi
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Fabian Heinrich
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Claudia Schulz
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Martina Sauter
- Institute for Pathology and Neuropathology, University Hospital Tübingen, Tübingen, Germany
| | - Susanne Krasemann
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Core Facility Experimental Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Philine Lange
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Axel Heinemann
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Debby van Riel
- Department of Viroscience, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Lonneke Leijten
- Department of Viroscience, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Lisa Bauer
- Department of Viroscience, Erasmus Medical Center, Rotterdam, the Netherlands
| | | | - Boaz Lopuhaä
- Department of Pathology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Tobias Busche
- Medical School East Westphalia-Lippe & Center for Biotechnology (CeBiTec), Bielefeld University, Bielefeld, Germany
| | - Daniel Wibberg
- Microbial Genomics and Biotechnology, Center for Biotechnology (CeBiTec), Bielefeld University, Bielefeld, Germany
| | - Dirk Schaudien
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Hannover, Germany
| | - Torsten Goldmann
- Pathology of the University Medical Center Schleswig-Holstein, Campus Lübeck and the Research Center Borstel, Research Center Borstel, Leibniz Center for Medicine and Biosciences, German Center for Lung Research (DZL), Borstel, Germany
| | - Anna Lüttjohann
- Department for Viral Zoonoses - One Health, Leibniz Institute of Virology, Hamburg, Germany
| | - Jenny Ruschinski
- Department for Viral Zoonoses - One Health, Leibniz Institute of Virology, Hamburg, Germany
| | - Hanna Jania
- Department for Viral Zoonoses - One Health, Leibniz Institute of Virology, Hamburg, Germany
| | - Zacharias Müller
- Department for Viral Zoonoses - One Health, Leibniz Institute of Virology, Hamburg, Germany
| | | | - Vanessa Krupp-Buzimkic
- Department for Viral Zoonoses - One Health, Leibniz Institute of Virology, Hamburg, Germany; Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Martin Wolff
- Early Life Origins of Chronic Lung Disease, Research Center Borstel, Leibniz Lung Center, Member of the German Center for Lung Research (DZL), Borstel, Germany
| | - Chiara Fallerini
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy; Medical Genetics, University of Siena, Siena, Italy
| | - Margherita Baldassarri
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy; Medical Genetics, University of Siena, Siena, Italy
| | - Simone Furini
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Katrina Norwood
- Department for Computational Biology of Infection Research, Helmholtz Center for Infection Research, Braunschweig, Germany; Braunschweig Integrated Centre of Systems Biology (BRICS), Technische Universität Braunschweig, Braunschweig, Germany
| | - Christopher Käufer
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany
| | | | - Maren von Köckritz-Blickwede
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Hannover, Germany; Department of Biochemistry, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Maria Schroeder
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Dominik Jarczak
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Axel Nierhaus
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias Welte
- Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany; Biomedical Research in Endstage and Obstructive Lung Disease, Member of the German Center for Lung Research, Hannover, Germany
| | - Stefan Kluge
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Alice C McHardy
- German Center for Infection Research (DZIF), Braunschweig, Germany; Department for Computational Biology of Infection Research, Helmholtz Center for Infection Research, Braunschweig, Germany; Braunschweig Integrated Centre of Systems Biology (BRICS), Technische Universität Braunschweig, Braunschweig, Germany; Cluster of Excellence RESIST (EXC 2355), Hannover Medical School, Hannover, Germany
| | - Frank Sommer
- Division Men's Health, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jörn Kalinowski
- Microbial Genomics and Biotechnology, Center for Biotechnology (CeBiTec), Bielefeld University, Bielefeld, Germany
| | - Susanne Krauss-Etschmann
- Early Life Origins of Chronic Lung Disease, Research Center Borstel, Leibniz Lung Center, Member of the German Center for Lung Research (DZL), Borstel, Germany; Institute for Experimental Medicine, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Franziska Richter
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Jan von der Thüsen
- Department of Pathology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Wolfgang Baumgärtner
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Karin Klingel
- Institute for Pathology and Neuropathology, University Hospital Tübingen, Tübingen, Germany
| | - Benjamin Ondruschka
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Alessandra Renieri
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy; Medical Genetics, University of Siena, Siena, Italy; Genetica Medica, Azienda Ospedaliero-Universitaria Senese, Siena, Italy
| | - Gülsah Gabriel
- Department for Viral Zoonoses - One Health, Leibniz Institute of Virology, Hamburg, Germany; Institute of Virology, University of Veterinary Medicine Hannover, Hannover, Germany; German Center for Infection Research (DZIF), Braunschweig, Germany.
| |
Collapse
|
7
|
Bergantini L, d’Alessandro M, Gangi S, Bianchi F, Cameli P, Perea B, Meocci M, Fabbri G, Marrucci S, Ederbali M, Bargagli E. Predictive Role of Cytokine and Adipokine Panel in Hospitalized COVID-19 Patients: Evaluation of Disease Severity, Survival and Lung Sequelae. Int J Mol Sci 2023; 24:12994. [PMID: 37629176 PMCID: PMC10455616 DOI: 10.3390/ijms241612994] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/10/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) may determine a multisystemic chronic syndrome after resolution of SARS-CoV-2 infection in a significant percentage of patients. Persistent cytokine dysregulation can contribute to long-lasting inflammation and tissue damage, resulting in the diverse, often debilitating symptoms experienced by some patients (so-called long COVID syndrome). The aim of our study was to evaluate the value of a panel of serum biomarkers of severity and prognosis in patients hospitalized for COVID-19 and also as predictive factors for the development of post-COVID lung sequelae after discharge from the hospital. All blood sampling was performed in the first 24 h after admission to the hospital. Serum analyte concentrations of IL-4, IL-2, CXCL10 (IP-10), IL-1β, TNF-α, CCL2 (MCP-1), IL-17A, IL-6, IL-10, IFN-γ, IL-12p70 and TGF-β1 were quantified by bead-based multiplex LEGENDplex™ analysis and commercially available ELISA kits. A total of 108 COVID-19 patients were enrolled in the study. Comparative analysis of these proteins showed higher levels of TGF-β and IL-6 and lower levels of RBP-4 and IL-10 in the severe group. Age, adiponectin, IL-8 and IL-32 resulted as the best predictors for survival. Moreover, IL-1β, IL17A, TNF-α, TGF-β, IL-4 and IL-6 were significantly higher in patients who showed HRCT evidence of fibrotic interstitial alterations at follow-up than patients who did not. The initial inflammatory status of patients on admission to the hospital with COVID-19, as reflected by the present panel of adipose tissue-related biomarkers and cytokines, offered insights into medium-term prognosis.
Collapse
Affiliation(s)
- Laura Bergantini
- Respiratory Disease and Lung Transplant Unit, Department of Medical Science, Surgery and Neurosciences, Siena University, 53100 Siena, Italy; (L.B.); (M.d.); (S.G.); (E.B.)
| | - Miriana d’Alessandro
- Respiratory Disease and Lung Transplant Unit, Department of Medical Science, Surgery and Neurosciences, Siena University, 53100 Siena, Italy; (L.B.); (M.d.); (S.G.); (E.B.)
| | - Sara Gangi
- Respiratory Disease and Lung Transplant Unit, Department of Medical Science, Surgery and Neurosciences, Siena University, 53100 Siena, Italy; (L.B.); (M.d.); (S.G.); (E.B.)
| | - Francesco Bianchi
- Pneumology Department, Azienda USL Toscana Sud-Est, “Misericordia” Hospital, 58100 Grosseto, Italy
| | - Paolo Cameli
- Respiratory Disease and Lung Transplant Unit, Department of Medical Science, Surgery and Neurosciences, Siena University, 53100 Siena, Italy; (L.B.); (M.d.); (S.G.); (E.B.)
| | - Beatrice Perea
- Respiratory Disease and Lung Transplant Unit, Department of Medical Science, Surgery and Neurosciences, Siena University, 53100 Siena, Italy; (L.B.); (M.d.); (S.G.); (E.B.)
| | - Martina Meocci
- Respiratory Disease and Lung Transplant Unit, Department of Medical Science, Surgery and Neurosciences, Siena University, 53100 Siena, Italy; (L.B.); (M.d.); (S.G.); (E.B.)
| | - Gaia Fabbri
- Respiratory Disease and Lung Transplant Unit, Department of Medical Science, Surgery and Neurosciences, Siena University, 53100 Siena, Italy; (L.B.); (M.d.); (S.G.); (E.B.)
| | - Sofia Marrucci
- Respiratory Disease and Lung Transplant Unit, Department of Medical Science, Surgery and Neurosciences, Siena University, 53100 Siena, Italy; (L.B.); (M.d.); (S.G.); (E.B.)
| | - Moftah Ederbali
- Respiratory Disease and Lung Transplant Unit, Department of Medical Science, Surgery and Neurosciences, Siena University, 53100 Siena, Italy; (L.B.); (M.d.); (S.G.); (E.B.)
| | - Elena Bargagli
- Respiratory Disease and Lung Transplant Unit, Department of Medical Science, Surgery and Neurosciences, Siena University, 53100 Siena, Italy; (L.B.); (M.d.); (S.G.); (E.B.)
| |
Collapse
|
8
|
Bergantini L, Baldassarri M, d'Alessandro M, Brunelli G, Fabbri G, Zguro K, Degl'Innocenti A, Fallerini C, Bargagli E, Renieri A. Ultra-rare RTEL1 gene variants associate with acute severity of COVID-19 and evolution to pulmonary fibrosis as a specific long COVID disorder. Respir Res 2023; 24:158. [PMID: 37328761 DOI: 10.1186/s12931-023-02458-7] [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] [Received: 01/14/2023] [Accepted: 05/22/2023] [Indexed: 06/18/2023] Open
Abstract
BACKGROUND Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) is a novel coronavirus that caused an ongoing pandemic of a pathology termed Coronavirus Disease 19 (COVID-19). Several studies reported that both COVID-19 and RTEL1 variants are associated with shorter telomere length, but a direct association between the two is not generally acknowledged. Here we demonstrate that up to 8.6% of severe COVID-19 patients bear RTEL1 ultra-rare variants, and show how this subgroup can be recognized. METHODS A cohort of 2246 SARS-CoV-2-positive subjects, collected within the GEN-COVID Multicenter study, was used in this work. Whole exome sequencing analysis was performed using the NovaSeq6000 System, and machine learning methods were used for candidate gene selection of severity. A nested study, comparing severely affected patients bearing or not variants in the selected gene, was used for the characterisation of specific clinical features connected to variants in both acute and post-acute phases. RESULTS Our GEN-COVID cohort revealed a total of 151 patients carrying at least one RTEL1 ultra-rare variant, which was selected as a specific acute severity feature. From a clinical point of view, these patients showed higher liver function indices, as well as increased CRP and inflammatory markers, such as IL-6. Moreover, compared to control subjects, they present autoimmune disorders more frequently. Finally, their decreased diffusion lung capacity for carbon monoxide after six months of COVID-19 suggests that RTEL1 variants can contribute to the development of SARS-CoV-2-elicited lung fibrosis. CONCLUSION RTEL1 ultra-rare variants can be considered as a predictive marker of COVID-19 severity, as well as a marker of pathological evolution in pulmonary fibrosis in the post-COVID phase. This notion can be used for a rapid screening in hospitalized infected people, for vaccine prioritization, and appropriate follow-up assessment for subjects at risk. Trial Registration NCT04549831 ( www. CLINICALTRIAL org ).
Collapse
Affiliation(s)
- Laura Bergantini
- Respiratory Disease Unit, Department of Medical Sciences, University Hospital of Siena (Azienda Ospedaliera Universitaria Senese, AOUS), Policlinico Le Scotte, Viale Bracci, 2, 53100, Siena, Italy
| | - Margherita Baldassarri
- Medical Genetics Unit, University of Siena, Policlinico Le Scotte, Viale Bracci, 2, 53100, Siena, Italy
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100, Siena, Italy
| | - Miriana d'Alessandro
- Respiratory Disease Unit, Department of Medical Sciences, University Hospital of Siena (Azienda Ospedaliera Universitaria Senese, AOUS), Policlinico Le Scotte, Viale Bracci, 2, 53100, Siena, Italy
| | - Giulia Brunelli
- Medical Genetics Unit, University of Siena, Policlinico Le Scotte, Viale Bracci, 2, 53100, Siena, Italy
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100, Siena, Italy
| | - Gaia Fabbri
- Respiratory Disease Unit, Department of Medical Sciences, University Hospital of Siena (Azienda Ospedaliera Universitaria Senese, AOUS), Policlinico Le Scotte, Viale Bracci, 2, 53100, Siena, Italy
| | - Kristina Zguro
- Medical Genetics Unit, University of Siena, Policlinico Le Scotte, Viale Bracci, 2, 53100, Siena, Italy
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100, Siena, Italy
| | - Andrea Degl'Innocenti
- Medical Genetics Unit, University of Siena, Policlinico Le Scotte, Viale Bracci, 2, 53100, Siena, Italy
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100, Siena, Italy
| | - Chiara Fallerini
- Medical Genetics Unit, University of Siena, Policlinico Le Scotte, Viale Bracci, 2, 53100, Siena, Italy
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100, Siena, Italy
| | - Elena Bargagli
- Respiratory Disease Unit, Department of Medical Sciences, University Hospital of Siena (Azienda Ospedaliera Universitaria Senese, AOUS), Policlinico Le Scotte, Viale Bracci, 2, 53100, Siena, Italy.
| | - Alessandra Renieri
- Medical Genetics Unit, University of Siena, Policlinico Le Scotte, Viale Bracci, 2, 53100, Siena, Italy.
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100, Siena, Italy.
- Genetica Medica, Azienda Ospedaliero-Universitaria Senese, 53100, Siena, Italy.
| |
Collapse
|
9
|
Cameli P, Pordon E, d'Alessandro M, Marzi ML, Galasso L, Biuzzi C, Bergantini L, Bargagli E, Scolletta S, Franchi F. MR-proADM as Prognostic Factor of Outcome in COVID-19 Patients. Biomedicines 2023; 11:1680. [PMID: 37371775 DOI: 10.3390/biomedicines11061680] [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: 05/13/2023] [Revised: 06/07/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND Serum mid-regional proadrenomedullin (MR-proADM) has emerged as a marker of organ failure (mainly lungs and kidneys) and poor prognosis in patients admitted to intensive care (IC); some reports also suggest it and other markers, such as Krebs von den Lungen-6 (KL-6) and interleukin-6 (IL-6), as a prognostic biomarker of COVID-19. The aim of the study was to evaluate the performance MR-proADM in hospitalized COVID-19 patients for predicting in-hospital mortality and need for non-invasive or invasive respiratory support. METHODS We enrolled 74 patients hospitalized in the COVID Unit of Siena Hospital from March to May 2020, for whom serum samples were available on admission for assay of MR-proADM, KL-6 and IL-6. Demographic data, comorbidities, medical history and clinical laboratory data on days 1-3 of admission and Simplified Acute Physiology Score and Simplified Organ Failure Assessment scores calculated at day 1 were collected retrospectively, as well as mortality and IC admission data. RESULTS 12 patients died in hospital (16%) and 14 patients were admitted to IC (19%). Serum concentrations of MR-proADM on admission and on day 1 were higher among non-survivors than among survivors (p = 0.015 and p = 0.045, respectively), while those on day 3 were not significantly different. Patients needing respiratory support had higher MR-proADM concentrations on admission than the others (p = 0.046), and those requiring invasive mechanical ventilation had higher MR-proADM on day 1 (p = 0.017). Serum concentrations of KL-6 and IL-6 were significantly higher in non-survivors (p = 0.03 and p = 0.004, respectively). ROC curve analysis showed that serum MR-proADM on day 1 had the best accuracy in predicting death and/or IC admission (AUC = 0.9583, p = 0.0006); the combination of all three biomarkers further improved the accuracy of prediction of death or IC admission (AUC = 0.9793; p = 0.00004). CONCLUSIONS Our data sustain the potential of serum MR-proADM as a reliable prognostic biomarker of hospitalized COVID-19 patients and confirms the utility of the three markers in the management and risk stratification of hospitalized patients. The markers are collected mini-invasively and are quick to analyze and cost-effective.
Collapse
Affiliation(s)
- Paolo Cameli
- Respiratory Diseases Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy
| | - Elena Pordon
- Respiratory Diseases Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy
| | - Miriana d'Alessandro
- Respiratory Diseases Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy
| | - Maria Laura Marzi
- Anesthesia and Intensive Care Unit, Department of Medicine, Surgery and Neurosciences, University Hospital of Siena, 53100 Siena, Italy
| | - Lucrezia Galasso
- Clinical Pathology Unit, Innovation, Experimentation and Clinical and Translational Research Department, University Hospital of Siena, 53100 Siena, Italy
| | - Cesare Biuzzi
- Anesthesia and Intensive Care Unit, Department of Medicine, Surgery and Neurosciences, University Hospital of Siena, 53100 Siena, Italy
| | - Laura Bergantini
- Respiratory Diseases Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy
| | - Elena Bargagli
- Respiratory Diseases Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy
| | - Sabino Scolletta
- Anesthesia and Intensive Care Unit, Department of Medicine, Surgery and Neurosciences, University Hospital of Siena, 53100 Siena, Italy
| | - Federico Franchi
- Cardiothoracic and Vascular Anesthesia and Intensive Care Unit, Department of Medical Science, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy
| |
Collapse
|
10
|
Resnik R, Lopez Mingorance F, Rivera F, Mitchell F, Gonzalez CD, Vaccaro MI. Autophagy in Inflammatory Response against SARS-CoV-2. Int J Mol Sci 2023; 24:4928. [PMID: 36902354 PMCID: PMC10002778 DOI: 10.3390/ijms24054928] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/21/2023] [Accepted: 02/27/2023] [Indexed: 03/08/2023] Open
Abstract
The coronavirus disease pandemic, which profoundly reshaped the world in 2019 (COVID-19), and is currently ongoing, has affected over 200 countries, caused over 500 million cumulative cases, and claimed the lives of over 6.4 million people worldwide as of August 2022. The causative agent is severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Depicting this virus' life cycle and pathogenic mechanisms, as well as the cellular host factors and pathways involved during infection, has great relevance for the development of therapeutic strategies. Autophagy is a catabolic process that sequesters damaged cell organelles, proteins, and external invading microbes, and delivers them to the lysosomes for degradation. Autophagy would be involved in the entry, endo, and release, as well as the transcription and translation, of the viral particles in the host cell. Secretory autophagy would also be involved in developing the thrombotic immune-inflammatory syndrome seen in a significant number of COVID-19 patients that can lead to severe illness and even death. This review aims to review the main aspects that characterize the complex and not yet fully elucidated relationship between SARS-CoV-2 infection and autophagy. It briefly describes the key concepts regarding autophagy and mentions its pro- and antiviral roles, while also noting the reciprocal effect of viral infection in autophagic pathways and their clinical aspects.
Collapse
Affiliation(s)
- Roxana Resnik
- Instituto de Bioquimica y Medicina Molecular, Consejo Nacional de Investigaciones Cientificas y Tecnicas, Universidad de Buenos Aires, Buenos Aires C1425FQB, Argentina
| | - Fabiana Lopez Mingorance
- Instituto de Bioquimica y Medicina Molecular, Consejo Nacional de Investigaciones Cientificas y Tecnicas, Universidad de Buenos Aires, Buenos Aires C1425FQB, Argentina
| | - Francisco Rivera
- Centro de Educacion Medica e Investigaciones Clinicas, Medicina Traslacional, Buenos Aires C1430EFA, Argentina
| | - Florencia Mitchell
- Centro de Educacion Medica e Investigaciones Clinicas, Medicina Traslacional, Buenos Aires C1430EFA, Argentina
| | - Claudio D. Gonzalez
- Centro de Educacion Medica e Investigaciones Clinicas, Medicina Traslacional, Buenos Aires C1430EFA, Argentina
| | - Maria I. Vaccaro
- Instituto de Bioquimica y Medicina Molecular, Consejo Nacional de Investigaciones Cientificas y Tecnicas, Universidad de Buenos Aires, Buenos Aires C1425FQB, Argentina
| |
Collapse
|
11
|
Markers of Bronchiolitis Obliterans Syndrome after Lung Transplant: Between Old Knowledge and Future Perspective. Biomedicines 2022; 10:biomedicines10123277. [PMID: 36552035 PMCID: PMC9775233 DOI: 10.3390/biomedicines10123277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/02/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022] Open
Abstract
Bronchiolitis obliterans syndrome (BOS) is the most common form of CLAD and is characterized by airflow limitation and an obstructive spirometric pattern without high-resolution computed tomography (HRCT) evidence of parenchymal opacities. Computed tomography and microCT analysis show abundant small airway obstruction, starting from the fifth generation of airway branching and affecting up to 40-70% of airways. The pathogenesis of BOS remains unclear. It is a multifactorial syndrome that leads to pathological tissue changes and clinical manifestations. Because BOS is associated with the worst long-term survival in LTx patients, many studies are focused on the early identification of BOS. Markers may be useful for diagnosis and for understanding the molecular and immunological mechanisms involved in the onset of BOS. Diagnostic and predictive markers of BOS have also been investigated in various biological materials, such as blood, BAL, lung tissue and extracellular vesicles. The aim of this review was to evaluate the scientific literature on markers of BOS after lung transplant. We performed a systematic review to find all available data on potential prognostic and diagnostic markers of BOS.
Collapse
|
12
|
Baldassarri M, Zguro K, Tomati V, Pastorino C, Fava F, Croci S, Bruttini M, Picchiotti N, Furini S, GEN-COVID Multicenter Study, Pedemonte N, Gabbi C, Renieri A, Fallerini C. Gain- and Loss-of-Function CFTR Alleles Are Associated with COVID-19 Clinical Outcomes. Cells 2022; 11:4096. [PMID: 36552859 PMCID: PMC9776607 DOI: 10.3390/cells11244096] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 11/25/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022] Open
Abstract
Carriers of single pathogenic variants of the CFTR (cystic fibrosis transmembrane conductance regulator) gene have a higher risk of severe COVID-19 and 14-day death. The machine learning post-Mendelian model pinpointed CFTR as a bidirectional modulator of COVID-19 outcomes. Here, we demonstrate that the rare complex allele [G576V;R668C] is associated with a milder disease via a gain-of-function mechanism. Conversely, CFTR ultra-rare alleles with reduced function are associated with disease severity either alone (dominant disorder) or with another hypomorphic allele in the second chromosome (recessive disorder) with a global residual CFTR activity between 50 to 91%. Furthermore, we characterized novel CFTR complex alleles, including [A238V;F508del], [R74W;D1270N;V201M], [I1027T;F508del], [I506V;D1168G], and simple alleles, including R347C, F1052V, Y625N, I328V, K68E, A309D, A252T, G542*, V562I, R1066H, I506V, I807M, which lead to a reduced CFTR function and thus, to more severe COVID-19. In conclusion, CFTR genetic analysis is an important tool in identifying patients at risk of severe COVID-19.
Collapse
Affiliation(s)
- Margherita Baldassarri
- Medical Genetics, University of Siena, 53100 Siena, Italy
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Kristina Zguro
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Valeria Tomati
- UOC Genetica Medica, IRCCS Istituto Giannina Gaslini, 16148 Genova, Italy
| | - Cristina Pastorino
- UOC Genetica Medica, IRCCS Istituto Giannina Gaslini, 16148 Genova, Italy
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, 16126 Genoa, Italy
| | - Francesca Fava
- Medical Genetics, University of Siena, 53100 Siena, Italy
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
- Genetica Medica, Azienda Ospedaliero-Universitaria Senese, 53100 Siena, Italy
| | - Susanna Croci
- Medical Genetics, University of Siena, 53100 Siena, Italy
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Mirella Bruttini
- Medical Genetics, University of Siena, 53100 Siena, Italy
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
- Genetica Medica, Azienda Ospedaliero-Universitaria Senese, 53100 Siena, Italy
| | - Nicola Picchiotti
- Department of Mathematics, University of Pavia, 27100 Pavia, Italy
- University of Siena, DIISM-SAILAB, 53100 Siena, Italy
| | - Simone Furini
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | | | | | - Chiara Gabbi
- Department of Biosciences and Nutrition, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Alessandra Renieri
- Medical Genetics, University of Siena, 53100 Siena, Italy
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
- Genetica Medica, Azienda Ospedaliero-Universitaria Senese, 53100 Siena, Italy
| | - Chiara Fallerini
- Medical Genetics, University of Siena, 53100 Siena, Italy
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| |
Collapse
|
13
|
Onoja A, Picchiotti N, Fallerini C, Baldassarri M, Fava F, Colombo F, Chiaromonte F, Renieri A, Furini S, Raimondi F. An explainable model of host genetic interactions linked to COVID-19 severity. Commun Biol 2022; 5:1133. [PMID: 36289370 PMCID: PMC9606365 DOI: 10.1038/s42003-022-04073-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 10/05/2022] [Indexed: 11/25/2022] Open
Abstract
We employed a multifaceted computational strategy to identify the genetic factors contributing to increased risk of severe COVID-19 infection from a Whole Exome Sequencing (WES) dataset of a cohort of 2000 Italian patients. We coupled a stratified k-fold screening, to rank variants more associated with severity, with the training of multiple supervised classifiers, to predict severity based on screened features. Feature importance analysis from tree-based models allowed us to identify 16 variants with the highest support which, together with age and gender covariates, were found to be most predictive of COVID-19 severity. When tested on a follow-up cohort, our ensemble of models predicted severity with high accuracy (ACC = 81.88%; AUCROC = 96%; MCC = 61.55%). Our model recapitulated a vast literature of emerging molecular mechanisms and genetic factors linked to COVID-19 response and extends previous landmark Genome-Wide Association Studies (GWAS). It revealed a network of interplaying genetic signatures converging on established immune system and inflammatory processes linked to viral infection response. It also identified additional processes cross-talking with immune pathways, such as GPCR signaling, which might offer additional opportunities for therapeutic intervention and patient stratification. Publicly available PheWAS datasets revealed that several variants were significantly associated with phenotypic traits such as "Respiratory or thoracic disease", supporting their link with COVID-19 severity outcome.
Collapse
Affiliation(s)
- Anthony Onoja
- Laboratorio di Biologia Bio@SNS, Scuola Normale Superiore, Pisa, Italy
| | - Nicola Picchiotti
- University of Siena, DIISM-SAILAB, Siena, Italy
- Department of Mathematics, University of Pavia, Pavia, Italy
| | - Chiara Fallerini
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy
- Medical Genetics, University of Siena, Siena, Italy
| | - Margherita Baldassarri
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy
- Medical Genetics, University of Siena, Siena, Italy
| | - Francesca Fava
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy
- Medical Genetics, University of Siena, Siena, Italy
- Genetica Medica, Azienda Ospedaliero-Universitaria Senese, Siena, Italy
| | - Francesca Colombo
- Istituto di Tecnologie Biomediche-Consiglio Nazionale delle Ricerche, Segrate, MI, Italy
| | - Francesca Chiaromonte
- Dept. of Statistics and Huck Institutes of the Life Sciences, Penn State University, University Park, PA, 16802, USA
- Institute of Economics and EMbeDS, Sant'Anna School of Advanced Studies, 56127, Pisa, Italy
| | - Alessandra Renieri
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy.
- Medical Genetics, University of Siena, Siena, Italy.
- Genetica Medica, Azienda Ospedaliero-Universitaria Senese, Siena, Italy.
| | - Simone Furini
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | | |
Collapse
|
14
|
Renieri A. COVID-19: a challenge and an opportunity. Eur J Hum Genet 2022; 30:870-871. [PMID: 35835955 PMCID: PMC9283098 DOI: 10.1038/s41431-022-01142-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 06/23/2022] [Indexed: 11/30/2022] Open
Affiliation(s)
- Alessandra Renieri
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy. .,Medical Genetics, University of Siena, Siena, Italy. .,Genetica Medica, Azienda Ospedaliero-Universitaria Senese, Siena, Italy.
| |
Collapse
|
15
|
Zguro K, Baldassarri M, Fava F, Beligni G, Daga S, Leoncini R, Galasso L, Cirianni M, Rusconi S, Siano M, Francisci D, Schiaroli E, Luchi S, Morelli G, Martinelli E, Girardis M, Busani S, Parisi SG, Panese S, Piscopo C, Capasso M, Tacconi D, Spertilli Raffaelli C, Giliberti A, Gori G, Katsikis PD, Lorubbio M, Calzoni P, Ognibene A, Bocchia M, Tozzi M, Bucalossi A, Marotta G, Furini S, GEN-COVID Multicenter Study, Renieri A, Fallerini C. Carriers of ADAMTS13 Rare Variants Are at High Risk of Life-Threatening COVID-19. Viruses 2022; 14:1185. [PMID: 35746657 PMCID: PMC9227269 DOI: 10.3390/v14061185] [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: 03/21/2022] [Revised: 05/25/2022] [Accepted: 05/25/2022] [Indexed: 01/08/2023] Open
Abstract
Thrombosis of small and large vessels is reported as a key player in COVID-19 severity. However, host genetic determinants of this susceptibility are still unclear. Congenital Thrombotic Thrombocytopenic Purpura is a severe autosomal recessive disorder characterized by uncleaved ultra-large vWF and thrombotic microangiopathy, frequently triggered by infections. Carriers are reported to be asymptomatic. Exome analysis of about 3000 SARS-CoV-2 infected subjects of different severities, belonging to the GEN-COVID cohort, revealed the specific role of vWF cleaving enzyme ADAMTS13 (A disintegrin-like and metalloprotease with thrombospondin type 1 motif, 13). We report here that ultra-rare variants in a heterozygous state lead to a rare form of COVID-19 characterized by hyper-inflammation signs, which segregates in families as an autosomal dominant disorder conditioned by SARS-CoV-2 infection, sex, and age. This has clinical relevance due to the availability of drugs such as Caplacizumab, which inhibits vWF-platelet interaction, and Crizanlizumab, which, by inhibiting P-selectin binding to its ligands, prevents leukocyte recruitment and platelet aggregation at the site of vascular damage.
Collapse
Affiliation(s)
- Kristina Zguro
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (K.Z.); (M.B.); (F.F.); (G.B.); (S.D.); (S.F.); (C.F.)
| | - Margherita Baldassarri
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (K.Z.); (M.B.); (F.F.); (G.B.); (S.D.); (S.F.); (C.F.)
- Medical Genetics, University of Siena, 53100 Siena, Italy
| | - Francesca Fava
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (K.Z.); (M.B.); (F.F.); (G.B.); (S.D.); (S.F.); (C.F.)
- Medical Genetics, University of Siena, 53100 Siena, Italy
- Genetica Medica, Azienda Ospedaliero-Universitaria Senese, 53100 Siena, Italy
| | - Giada Beligni
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (K.Z.); (M.B.); (F.F.); (G.B.); (S.D.); (S.F.); (C.F.)
- Medical Genetics, University of Siena, 53100 Siena, Italy
| | - Sergio Daga
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (K.Z.); (M.B.); (F.F.); (G.B.); (S.D.); (S.F.); (C.F.)
- Medical Genetics, University of Siena, 53100 Siena, Italy
| | - Roberto Leoncini
- Laboratorio Patologia Clinica, Azienda Ospedaliero-Universitaria Senese, 53100 Siena, Italy; (R.L.); (L.G.); (M.C.); (P.C.)
| | - Lucrezia Galasso
- Laboratorio Patologia Clinica, Azienda Ospedaliero-Universitaria Senese, 53100 Siena, Italy; (R.L.); (L.G.); (M.C.); (P.C.)
| | - Michele Cirianni
- Laboratorio Patologia Clinica, Azienda Ospedaliero-Universitaria Senese, 53100 Siena, Italy; (R.L.); (L.G.); (M.C.); (P.C.)
| | - Stefano Rusconi
- Infectious Diseases Unit, ASST Ovest Milanese, 20025 Legnano, Italy;
- Department of Biomedical and Clinical Sciences Luigi Sacco, University of Milan, 20157 Milan, Italy;
| | - Matteo Siano
- Department of Biomedical and Clinical Sciences Luigi Sacco, University of Milan, 20157 Milan, Italy;
| | - Daniela Francisci
- Infectious Diseases Clinic, “Santa Maria della Misericordia” Hospital, University of Perugia, 06124 Perugia, Italy; (D.F.); (E.S.)
| | - Elisabetta Schiaroli
- Infectious Diseases Clinic, “Santa Maria della Misericordia” Hospital, University of Perugia, 06124 Perugia, Italy; (D.F.); (E.S.)
| | - Sauro Luchi
- Infectious Disease Unit, Hospital of Lucca, 55100 Lucca, Italy; (S.L.); (G.M.)
| | - Giovanna Morelli
- Infectious Disease Unit, Hospital of Lucca, 55100 Lucca, Italy; (S.L.); (G.M.)
| | - Enrico Martinelli
- Department of Respiratory Diseases, Azienda Ospedaliera di Cremona, 26100 Cremona, Italy;
| | - Massimo Girardis
- Department of Anesthesia and Intensive Care, University of Modena and Reggio Emilia, 41124 Modena, Italy; (M.G.); (S.B.)
| | - Stefano Busani
- Department of Anesthesia and Intensive Care, University of Modena and Reggio Emilia, 41124 Modena, Italy; (M.G.); (S.B.)
| | | | - Sandro Panese
- Clinical Infectious Diseases, Mestre Hospital, 30171 Venezia, Italy;
| | - Carmelo Piscopo
- Medical Genetics and Laboratory Genetics Unit, “Antonio Cardarelli” hospital, 80131 Naples, Italy;
| | - Mario Capasso
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80138 Naples, Italy;
- CEINGE Biotecnologie Avanzate, 80145 Naples, Italy
| | - Danilo Tacconi
- Department of Specialized and Internal Medicine, Infectious Diseases Unit, San Donato Hospital Arezzo, 52100 Arezzo, Italy; (D.T.); (C.S.R.)
| | - Chiara Spertilli Raffaelli
- Department of Specialized and Internal Medicine, Infectious Diseases Unit, San Donato Hospital Arezzo, 52100 Arezzo, Italy; (D.T.); (C.S.R.)
| | - Annarita Giliberti
- Medical Genetics Unit, Meyer Children’s University Hospital, 50134 Florence, Italy; (A.G.); (G.G.)
| | - Giulia Gori
- Medical Genetics Unit, Meyer Children’s University Hospital, 50134 Florence, Italy; (A.G.); (G.G.)
| | - Peter D. Katsikis
- Department of Immunology, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands;
| | - Maria Lorubbio
- UOC Laboratorio Analisi Chimico Cliniche, 52100 Arezzo, Italy; (M.L.); (A.O.)
| | - Paola Calzoni
- Laboratorio Patologia Clinica, Azienda Ospedaliero-Universitaria Senese, 53100 Siena, Italy; (R.L.); (L.G.); (M.C.); (P.C.)
| | - Agostino Ognibene
- UOC Laboratorio Analisi Chimico Cliniche, 52100 Arezzo, Italy; (M.L.); (A.O.)
| | - Monica Bocchia
- Hematology Unit, Department of Medical Science, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy;
| | - Monica Tozzi
- Stem Cell Transplant and Cellular Therapy Unit, University Hospital of Siena, 53100 Siena, Italy; (M.T.); (A.B.); (G.M.)
| | - Alessandro Bucalossi
- Stem Cell Transplant and Cellular Therapy Unit, University Hospital of Siena, 53100 Siena, Italy; (M.T.); (A.B.); (G.M.)
| | - Giuseppe Marotta
- Stem Cell Transplant and Cellular Therapy Unit, University Hospital of Siena, 53100 Siena, Italy; (M.T.); (A.B.); (G.M.)
| | - Simone Furini
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (K.Z.); (M.B.); (F.F.); (G.B.); (S.D.); (S.F.); (C.F.)
| | | | - Alessandra Renieri
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (K.Z.); (M.B.); (F.F.); (G.B.); (S.D.); (S.F.); (C.F.)
- Medical Genetics, University of Siena, 53100 Siena, Italy
- Genetica Medica, Azienda Ospedaliero-Universitaria Senese, 53100 Siena, Italy
| | - Chiara Fallerini
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (K.Z.); (M.B.); (F.F.); (G.B.); (S.D.); (S.F.); (C.F.)
- Medical Genetics, University of Siena, 53100 Siena, Italy
| |
Collapse
|
16
|
Host genetic basis of COVID-19: from methodologies to genes. Eur J Hum Genet 2022; 30:899-907. [PMID: 35618891 PMCID: PMC9135575 DOI: 10.1038/s41431-022-01121-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 04/04/2022] [Accepted: 05/09/2022] [Indexed: 01/03/2023] Open
Abstract
The COVID-19 pandemic caused by the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is having a massive impact on public health, societies, and economies worldwide. Despite the ongoing vaccination program, treating COVID-19 remains a high priority; thus, a better understanding of the disease is urgently needed. Initially, susceptibility was associated with age, sex, and other prior existing comorbidities. However, as these conditions alone could not explain the highly variable clinical manifestations of SARS-CoV-2 infection, the attention was shifted toward the identification of the genetic basis of COVID-19. Thanks to international collaborations like The COVID-19 Host Genetics Initiative, it became possible the elucidation of numerous genetic markers that are not only likely to help in explaining the varied clinical outcomes of COVID-19 patients but can also guide the development of novel diagnostics and therapeutics. Within this framework, this review delineates GWAS and Burden test as traditional methodologies employed so far for the discovery of the human genetic basis of COVID-19, with particular attention to recently emerged predictive models such as the post-Mendelian model. A summary table with the main genome-wide significant genomic loci is provided. Besides, various common and rare variants identified in genes like TLR7, CFTR, ACE2, TMPRSS2, TLR3, and SELP are further described in detail to illustrate their association with disease severity.
Collapse
|
17
|
Bergantini L, Mainardi A, d’Alessandro M, Cameli P, Bennett D, Bargagli E, Sestini P. Common Molecular Pathways Between Post-COVID19 Syndrome and Lung Fibrosis: A Scoping Review. Front Pharmacol 2022; 13:748931. [PMID: 35308222 PMCID: PMC8931519 DOI: 10.3389/fphar.2022.748931] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 02/14/2022] [Indexed: 01/18/2023] Open
Abstract
The pathogenetic mechanism of post-Covid-19 pulmonary fibrosis is currently a topic of intense research interest, but still largely unexplored. The aim of this work was to carry out a systematic exploratory search of the literature (Scoping review) to identify and systematize the main pathogenetic mechanisms that are believed to be involved in this phenomenon, in order to highlight the same molecular aspect of the lung. These aims could be essential in the future for therapeutic management. We identified all primary studies involving in post COVID19 syndrome with pulmonary fibrosis as a primary endpoint by performing data searches in various systematic review databases. Two reviewers independently reviewed all abstracts (398) and full text data. The quality of study has been assess through SANRA protocol. A total of 32 studies involving were included, included the possible involvement of inflammatory cytokines, concerned the renin-angiotensin system, the potential role of galectin-3, epithelial injuries in fibrosis, alveolar type 2 involvement, Neutrophil extracellular traps (NETs) and the others implied other specific aspects (relationship with clinical and mechanical factors, epithelial transition mesenchymal, TGF-β signaling pathway, midkine, caspase and macrophages, genetics). In most cases, these were narrative reviews or letters to the editor, except for 10 articles, which presented original data, albeit sometimes in experimental models. From the development of these researches, progress in the knowledge of the phenomenon and hopefully in its prevention and therapy may originate.
Collapse
Affiliation(s)
- Laura Bergantini
- Department of Medical Sciences, Surgery and Neurosciences, Respiratory Disease and Lung Transplant Unit, Respiratory Diseases and Transplant Unit, Siena University, Siena, Italy
| | | | | | | | | | | | | |
Collapse
|
18
|
Abstract
Systematic bio- and databanks are key prerequisites for modern radiation research to investigate radiation response mechanisms in the context of genetic, environmental and lifestyle-associated factors. This report presents the current status of the German Uranium Miners’ Biobank. In 2008, the bio- and databank was established at the Federal Office for Radiation Protection, and the sampling of biological materials from former uranium miners with and without lung cancer was initiated. For this purpose, various biological specimens, such as DNA and RNA, were isolated from blood samples as well as from formalin-fixed paraffin-embedded lung tissue. High-quality biomaterials suitable for OMICs research and the associated data on occupational radiation and dust exposure, and medical and lifestyle data from over 1000 individuals have been stored so far. Various experimental data, e.g., genome-wide SNPs, whole genome transcriptomic and miRNA data, as well as individual chromosomal aberration data from subgroups of biobank samples, are already available upon request for in-depth research on radiation-induced long-term effects, individual radiation susceptibility to lung cancer and radon-induced fingerprints in lung cancer. This biobank is the first systematic uranium miners´ biobank worldwide that is suitable for OMICs research on radiation-exposed workers. It offers the opportunity to link radiation-induced perturbations of biological pathways or processes and putative adverse outcome(s) by OMICs profiling at different biological organization levels.
Collapse
|
19
|
Fallerini C, Picchiotti N, Baldassarri M, Zguro K, Daga S, Fava F, Benetti E, Amitrano S, Bruttini M, Palmieri M, Croci S, Lista M, Beligni G, Valentino F, Meloni I, Tanfoni M, Minnai F, Colombo F, Cabri E, Fratelli M, Gabbi C, Mantovani S, Frullanti E, Gori M, Crawley FP, Butler-Laporte G, Richards B, Zeberg H, Lipcsey M, Hultström M, Ludwig KU, Schulte EC, Pairo-Castineira E, Baillie JK, Schmidt A, Frithiof R, WES/WGS Working Group Within the HGI, GenOMICC Consortium, Mari F, Renieri A, Furini S. Common, low-frequency, rare, and ultra-rare coding variants contribute to COVID-19 severity. Hum Genet 2022; 141:147-173. [PMID: 34889978 PMCID: PMC8661833 DOI: 10.1007/s00439-021-02397-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 10/26/2021] [Indexed: 12/13/2022]
Abstract
The combined impact of common and rare exonic variants in COVID-19 host genetics is currently insufficiently understood. Here, common and rare variants from whole-exome sequencing data of about 4000 SARS-CoV-2-positive individuals were used to define an interpretable machine-learning model for predicting COVID-19 severity. First, variants were converted into separate sets of Boolean features, depending on the absence or the presence of variants in each gene. An ensemble of LASSO logistic regression models was used to identify the most informative Boolean features with respect to the genetic bases of severity. The Boolean features selected by these logistic models were combined into an Integrated PolyGenic Score that offers a synthetic and interpretable index for describing the contribution of host genetics in COVID-19 severity, as demonstrated through testing in several independent cohorts. Selected features belong to ultra-rare, rare, low-frequency, and common variants, including those in linkage disequilibrium with known GWAS loci. Noteworthily, around one quarter of the selected genes are sex-specific. Pathway analysis of the selected genes associated with COVID-19 severity reflected the multi-organ nature of the disease. The proposed model might provide useful information for developing diagnostics and therapeutics, while also being able to guide bedside disease management.
Collapse
Affiliation(s)
- Chiara Fallerini
- grid.9024.f0000 0004 1757 4641Department of Medical Biotechnologies, Med Biotech Hub and Competence Center, University of Siena, Siena, Italy ,grid.9024.f0000 0004 1757 4641Medical Genetics, University of Siena, Siena, Italy
| | - Nicola Picchiotti
- grid.9024.f0000 0004 1757 4641University of Siena, DIISM-SAILAB, Siena, Italy ,grid.8982.b0000 0004 1762 5736Department of Mathematics, University of Pavia, Pavia, Italy
| | - Margherita Baldassarri
- grid.9024.f0000 0004 1757 4641Department of Medical Biotechnologies, Med Biotech Hub and Competence Center, University of Siena, Siena, Italy ,grid.9024.f0000 0004 1757 4641Medical Genetics, University of Siena, Siena, Italy
| | - Kristina Zguro
- grid.9024.f0000 0004 1757 4641Department of Medical Biotechnologies, Med Biotech Hub and Competence Center, University of Siena, Siena, Italy
| | - Sergio Daga
- grid.9024.f0000 0004 1757 4641Department of Medical Biotechnologies, Med Biotech Hub and Competence Center, University of Siena, Siena, Italy ,grid.9024.f0000 0004 1757 4641Medical Genetics, University of Siena, Siena, Italy
| | - Francesca Fava
- grid.9024.f0000 0004 1757 4641Department of Medical Biotechnologies, Med Biotech Hub and Competence Center, University of Siena, Siena, Italy ,grid.9024.f0000 0004 1757 4641Medical Genetics, University of Siena, Siena, Italy ,grid.411477.00000 0004 1759 0844Genetica Medica, Azienda Ospedaliero-Universitaria Senese, Siena, Italy
| | - Elisa Benetti
- grid.9024.f0000 0004 1757 4641Department of Medical Biotechnologies, Med Biotech Hub and Competence Center, University of Siena, Siena, Italy
| | - Sara Amitrano
- grid.411477.00000 0004 1759 0844Genetica Medica, Azienda Ospedaliero-Universitaria Senese, Siena, Italy
| | - Mirella Bruttini
- grid.9024.f0000 0004 1757 4641Department of Medical Biotechnologies, Med Biotech Hub and Competence Center, University of Siena, Siena, Italy ,grid.9024.f0000 0004 1757 4641Medical Genetics, University of Siena, Siena, Italy ,grid.411477.00000 0004 1759 0844Genetica Medica, Azienda Ospedaliero-Universitaria Senese, Siena, Italy
| | - Maria Palmieri
- grid.9024.f0000 0004 1757 4641Department of Medical Biotechnologies, Med Biotech Hub and Competence Center, University of Siena, Siena, Italy ,grid.9024.f0000 0004 1757 4641Medical Genetics, University of Siena, Siena, Italy
| | - Susanna Croci
- grid.9024.f0000 0004 1757 4641Department of Medical Biotechnologies, Med Biotech Hub and Competence Center, University of Siena, Siena, Italy ,grid.9024.f0000 0004 1757 4641Medical Genetics, University of Siena, Siena, Italy
| | - Mirjam Lista
- grid.9024.f0000 0004 1757 4641Department of Medical Biotechnologies, Med Biotech Hub and Competence Center, University of Siena, Siena, Italy ,grid.9024.f0000 0004 1757 4641Medical Genetics, University of Siena, Siena, Italy
| | - Giada Beligni
- grid.9024.f0000 0004 1757 4641Department of Medical Biotechnologies, Med Biotech Hub and Competence Center, University of Siena, Siena, Italy ,grid.9024.f0000 0004 1757 4641Medical Genetics, University of Siena, Siena, Italy
| | - Floriana Valentino
- grid.9024.f0000 0004 1757 4641Department of Medical Biotechnologies, Med Biotech Hub and Competence Center, University of Siena, Siena, Italy ,grid.9024.f0000 0004 1757 4641Medical Genetics, University of Siena, Siena, Italy
| | - Ilaria Meloni
- grid.9024.f0000 0004 1757 4641Department of Medical Biotechnologies, Med Biotech Hub and Competence Center, University of Siena, Siena, Italy ,grid.9024.f0000 0004 1757 4641Medical Genetics, University of Siena, Siena, Italy
| | - Marco Tanfoni
- grid.9024.f0000 0004 1757 4641University of Siena, DIISM-SAILAB, Siena, Italy
| | - Francesca Minnai
- grid.429135.80000 0004 1756 2536Istituto di Tecnologie Biomediche-Consiglio Nazionale delle Ricerche, Segrate, MI Italy
| | - Francesca Colombo
- grid.429135.80000 0004 1756 2536Istituto di Tecnologie Biomediche-Consiglio Nazionale delle Ricerche, Segrate, MI Italy
| | - Enrico Cabri
- grid.4527.40000000106678902Pharmacogenomics Unit, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Maddalena Fratelli
- grid.4527.40000000106678902Pharmacogenomics Unit, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Chiara Gabbi
- grid.4714.60000 0004 1937 0626Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - Stefania Mantovani
- grid.419425.f0000 0004 1760 3027Department of Medicine, Clinical Immunology and Infectious Diseases, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Elisa Frullanti
- grid.9024.f0000 0004 1757 4641Department of Medical Biotechnologies, Med Biotech Hub and Competence Center, University of Siena, Siena, Italy ,grid.9024.f0000 0004 1757 4641Medical Genetics, University of Siena, Siena, Italy
| | - Marco Gori
- grid.9024.f0000 0004 1757 4641University of Siena, DIISM-SAILAB, Siena, Italy ,grid.503321.60000 0001 0561 3840Models and Algorithms for Artificial Intelligence (MAASAI) Research Group, Université Côte d’Azur, Inria, CNRS, I3S, Biot, France
| | - Francis P. Crawley
- Good Clinical Practice Alliance-Europe (GCPA) and Strategic Initiative for Developing Capacity in Ethical Review (SIDCER), Leuven, Belgium
| | - Guillaume Butler-Laporte
- grid.14709.3b0000 0004 1936 8649Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, QC Canada
| | - Brent Richards
- grid.14709.3b0000 0004 1936 8649Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Department of Human Genetics, McGill University, Montreal, QC Canada ,grid.13097.3c0000 0001 2322 6764Department of Twin Research, King’s College London, London, UK
| | - Hugo Zeberg
- grid.4714.60000 0004 1937 0626Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Miklos Lipcsey
- grid.8993.b0000 0004 1936 9457Anaesthesiology and Intensive Care Medicine, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden ,grid.8993.b0000 0004 1936 9457Hedenstierna Laboratory, CIRRUS, Anaesthesiology and Intensive Care Medicine, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Michael Hultström
- grid.8993.b0000 0004 1936 9457Anaesthesiology and Intensive Care Medicine, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden ,grid.8993.b0000 0004 1936 9457Integrative Physiology, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Kerstin U. Ludwig
- grid.10388.320000 0001 2240 3300Institute of Human Genetics, School of Medicine and University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Eva C. Schulte
- grid.411095.80000 0004 0477 2585Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, 80336 Munich, Germany ,grid.5252.00000 0004 1936 973XDepartment of Psychiatry and Psychotherapy, University Hospital, LMU Munich, 80336 Munich, Germany ,grid.6936.a0000000123222966Institute of Virology, Technical University Munich/Helmholtz Zentrum München, Munich, Germany
| | - Erola Pairo-Castineira
- grid.4305.20000 0004 1936 7988MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Crewe Road, Edinburgh, EH4 2XU UK ,grid.4305.20000 0004 1936 7988Roslin Institute, University of Edinburgh, Easter Bush, Edinburgh, EH25 9RG UK
| | - John Kenneth Baillie
- grid.4305.20000 0004 1936 7988MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Crewe Road, Edinburgh, EH4 2XU UK ,grid.4305.20000 0004 1936 7988Roslin Institute, University of Edinburgh, Easter Bush, Edinburgh, EH25 9RG UK ,grid.418716.d0000 0001 0709 1919Intensive Care Unit, Royal Infirmary of Edinburgh, 54 Little France Drive, Edinburgh, H16 5SA UK
| | - Axel Schmidt
- grid.10388.320000 0001 2240 3300Institute of Human Genetics, School of Medicine and University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Robert Frithiof
- grid.8993.b0000 0004 1936 9457Anaesthesiology and Intensive Care Medicine, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | | | | | | | - Francesca Mari
- grid.9024.f0000 0004 1757 4641Department of Medical Biotechnologies, Med Biotech Hub and Competence Center, University of Siena, Siena, Italy ,grid.9024.f0000 0004 1757 4641Medical Genetics, University of Siena, Siena, Italy ,grid.411477.00000 0004 1759 0844Genetica Medica, Azienda Ospedaliero-Universitaria Senese, Siena, Italy
| | - Alessandra Renieri
- Department of Medical Biotechnologies, Med Biotech Hub and Competence Center, University of Siena, Siena, Italy. .,Medical Genetics, University of Siena, Siena, Italy. .,Genetica Medica, Azienda Ospedaliero-Universitaria Senese, Siena, Italy. .,Medical Genetics Unit, University of Siena, Policlinico Le Scotte, Viale Bracci, 2, 53100, Siena, Italy.
| | - Simone Furini
- grid.9024.f0000 0004 1757 4641Department of Medical Biotechnologies, Med Biotech Hub and Competence Center, University of Siena, Siena, Italy
| |
Collapse
|
20
|
Bergantini L, d’Alessandro M, Cameli P, Cavallaro D, Gangi S, Cekorja B, Sestini P, Bargagli E. NK and T Cell Immunological Signatures in Hospitalized Patients with COVID-19. Cells 2021; 10:3182. [PMID: 34831404 PMCID: PMC8618013 DOI: 10.3390/cells10113182] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/09/2021] [Accepted: 11/11/2021] [Indexed: 02/07/2023] Open
Abstract
Severe acute respiratory syndrome caused by coronavirus 2 emerged in Wuhan (China) in December 2019 and has severely challenged the human population. NK and T cells are involved in the progression of COVID-19 infection through the ability of NK cells to modulate T-cell responses, and by the stimulation of cytokine release. No detailed investigation of the NK cell landscape in clinical SARS-CoV-2 infection has yet been reported. A total of 35 COVID-19 hospitalised patients were stratified for clinical severity and 17 healthy subjects were enrolled. NK cell subsets and T cell subsets were analysed with flow cytometry. Serum cytokines were detected with a bead-based multiplex assay. Fewer CD56dimCD16brightNKG2A+NK cells and a parallel increase in the CD56+CD69+NK, CD56+PD-1+NK, CD56+NKp44+NK subset were reported in COVID-19 than HC. A significantly higher adaptive/memory-like NK cell frequency in patients with severe disease than in those with mild and moderate phenotypes were reported. Moreover, adaptive/memory-like NK cell frequencies were significantly higher in patients who died than in survivors. Severe COVID-19 patients showed higher serum concentrations of IL-6 than mild and control groups. Direct correlation emerged for IL-6 and adaptive/memory-like NK. All these findings provide new insights into the immune response of patients with COVID-19. In particular, they demonstrate activation of NK through overexpression of CD69 and CD25 and show that PD-1 inhibitory signalling maintains an exhausted phenotype in NK cells. These results suggest that adaptive/memory-like NK cells could be the basis of promising targeted therapy for future viral infections.
Collapse
|
21
|
Rivas AL, van Regenmortel MHV. COVID-19 related interdisciplinary methods: Preventing errors and detecting research opportunities. Methods 2021; 195:3-14. [PMID: 34029715 PMCID: PMC8545872 DOI: 10.1016/j.ymeth.2021.05.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 05/18/2021] [Accepted: 05/19/2021] [Indexed: 12/12/2022] Open
Abstract
More than 130,000 peer-reviewed studies have been published within one year after COVID-19 emerged in many countries. This large and rapidly growing field may overwhelm the synthesizing abilities of both researchers and policy-makers. To provide a sinopsis, prevent errors, and detect cognitive gaps that may require interdisciplinary research methods, the literature on COVID-19 is summarized, twice. The overall purpose of this study is to generate a dialogue meant to explain the genesis of and/or find remedies for omissions and contradictions. The first review starts in Biology and ends in Policy. Policy is chosen as a destination because it is the setting where cognitive integration must occur. The second review follows the opposite path: it begins with stated policies on COVID-19 and then their assumptions and disciplinary relationships are identified. The purpose of this interdisciplinary method on methods is to yield a relational and explanatory view of the field -one strategy likely to be incomplete but usable when large bodies of literature need to be rapidly summarized. These reviews identify nine inter-related problems, research needs, or omissions, namely: (1) nation-wide, geo-referenced, epidemiological data collection systems (open to and monitored by the public); (2) metrics meant to detect non-symptomatic cases -e.g., test positivity-; (3) cost-benefit oriented methods, which should demonstrate they detect silent viral spreaders even with limited testing; (4) new personalized tests that inform on biological functions and disease correlates, such as cell-mediated immunity, co-morbidities, and immuno-suppression; (5) factors that influence vaccine effectiveness; (6) economic predictions that consider the long-term consequences likely to follow epidemics that growth exponentially; (7) the errors induced by self-limiting and/or implausible paradigms, such as binary and reductionist approaches; (8) new governance models that emphasize problem-solving skills, social participation, and the use of scientific knowledge; and (9) new educational programs that utilize visual aids and audience-specific communication strategies. The analysis indicates that, to optimally address these problems, disciplinary and social integration is needed. By asking what is/are the potential cause(s) and consequence(s) of each issue, this methodology generates visualizations that reveal possible relationships as well as omissions and contradictions. While inherently limited in scope and likely to become obsolete, these shortcomings are avoided when this 'method on methods' is frequently practiced. Open-ended, inter-/trans-disciplinary perspectives and broad social participation may help researchers and citizens to construct, de-construct, and re-construct COVID-19 related research.
Collapse
Affiliation(s)
- Ariel L Rivas
- Center for Global Health, School of Medicine, University of New Mexico, Albuquerque, NM, United States.
| | - Marc H V van Regenmortel
- University of Vienna, Austria; and Higher School of Biotechnology, University of Strasbourg, and French National Research Center, France
| |
Collapse
|
22
|
Long-term health sequelae and quality of life at least 6 months after infection with SARS-CoV-2: design and rationale of the COVIDOM-study as part of the NAPKON population-based cohort platform (POP). Infection 2021; 49:1277-1287. [PMID: 34642875 PMCID: PMC8508400 DOI: 10.1007/s15010-021-01707-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 09/22/2021] [Indexed: 11/16/2022]
Abstract
Purpose Over the course of COVID-19 pandemic, evidence has accumulated that SARS-CoV-2 infections may affect multiple organs and have serious clinical sequelae, but on-site clinical examinations with non-hospitalized samples are rare. We, therefore, aimed to systematically assess the long-term health status of samples of hospitalized and non-hospitalized SARS-CoV-2 infected individuals from three regions in Germany. Methods The present paper describes the COVIDOM-study within the population-based cohort platform (POP) which has been established under the auspices of the NAPKON infrastructure (German National Pandemic Cohort Network) of the national Network University Medicine (NUM). Comprehensive health assessments among SARS-CoV-2 infected individuals are conducted at least 6 months after the acute infection at the study sites Kiel, Würzburg and Berlin. Potential participants were identified and contacted via the local public health authorities, irrespective of the severity of the initial infection. A harmonized examination protocol has been implemented, consisting of detailed assessments of medical history, physical examinations, and the collection of multiple biosamples (e.g., serum, plasma, saliva, urine) for future analyses. In addition, patient-reported perception of the impact of local pandemic-related measures and infection on quality-of-life are obtained. Results As of July 2021, in total 6813 individuals infected in 2020 have been invited into the COVIDOM-study. Of these, about 36% wished to participate and 1295 have already been examined at least once. Conclusion NAPKON-POP COVIDOM-study complements other Long COVID studies assessing the long-term consequences of an infection with SARS-CoV-2 by providing detailed health data of population-based samples, including individuals with various degrees of disease severity. Trial registration Registered at the German registry for clinical studies (DRKS00023742).
Collapse
|
23
|
C9orf72 Intermediate Repeats Confer Genetic Risk for Severe COVID-19 Pneumonia Independently of Age. Int J Mol Sci 2021; 22:ijms22136991. [PMID: 34209673 PMCID: PMC8268051 DOI: 10.3390/ijms22136991] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 06/24/2021] [Accepted: 06/24/2021] [Indexed: 12/13/2022] Open
Abstract
A cytokine storm, autoimmune features and dysfunctions of myeloid cells significantly contribute to severe coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Genetic background of the host seems to be partly responsible for severe phenotype and genes related to innate immune response seem critical host determinants. The C9orf72 gene has a role in vesicular trafficking, autophagy regulation and lysosome functions, is highly expressed in myeloid cells and is involved in immune functions, regulating the lysosomal degradation of mediators of innate immunity. A large non-coding hexanucleotide repeat expansion (HRE) in this gene is the main genetic cause of frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS), both characterized by neuroinflammation and high systemic levels of proinflammatory cytokines, while HREs of intermediate length, although rare, are more frequent in autoimmune disorders. C9orf72 full mutation results in haploinsufficiency and intermediate HREs seem to modulate gene expression as well and impair autophagy. Herein, we sought to explore whether intermediate HREs in C9orf72 may be a risk factor for severe COVID-19. Although we found intermediate HREs in only a small portion of 240 patients with severe COVID-19 pneumonia, the magnitude of risk for requiring non-invasive or mechanical ventilation conferred by harboring intermediate repeats >10 units in at least one C9orf72 allele was more than twice respect to having shorter expansions, when adjusted for age (odds ratio (OR) 2.36; 95% confidence interval (CI) 1.04-5.37, p = 0.040). The association between intermediate repeats >10 units and more severe clinical outcome (p = 0.025) was also validated in an independent cohort of 201 SARS-CoV-2 infected patients. These data suggest that C9orf72 HREs >10 units may influence the pathogenic process driving more severe COVID-19 phenotypes.
Collapse
|
24
|
Zhang S, Cooper-Knock J, Weimer AK, Harvey C, Julian TH, Wang C, Li J, Furini S, Frullanti E, Fava F, Renieri A, Pan C, Song J, Billing-Ross P, Gao P, Shen X, Timpanaro IS, Kenna KP, VA Million Veteran Program, GEN-COVID Network, Davis MM, Tsao PS, Snyder MP. Common and rare variant analyses combined with single-cell multiomics reveal cell-type-specific molecular mechanisms of COVID-19 severity. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.06.15.21258703. [PMID: 34189540 PMCID: PMC8240695 DOI: 10.1101/2021.06.15.21258703] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The determinants of severe COVID-19 in non-elderly adults are poorly understood, which limits opportunities for early intervention and treatment. Here we present novel machine learning frameworks for identifying common and rare disease-associated genetic variation, which outperform conventional approaches. By integrating single-cell multiomics profiling of human lungs to link genetic signals to cell-type-specific functions, we have discovered and validated over 1,000 risk genes underlying severe COVID-19 across 19 cell types. Identified risk genes are overexpressed in healthy lungs but relatively downregulated in severely diseased lungs. Genetic risk for severe COVID-19, within both common and rare variants, is particularly enriched in natural killer (NK) cells, which places these immune cells upstream in the pathogenesis of severe disease. Mendelian randomization indicates that failed NKG2D-mediated activation of NK cells leads to critical illness. Network analysis further links multiple pathways associated with NK cell activation, including type-I-interferon-mediated signalling, to severe COVID-19. Our rare variant model, PULSE, enables sensitive prediction of severe disease in non-elderly patients based on whole-exome sequencing; individualized predictions are accurate independent of age and sex, and are consistent across multiple populations and cohorts. Risk stratification based on exome sequencing has the potential to facilitate post-exposure prophylaxis in at-risk individuals, potentially based around augmentation of NK cell function. Overall, our study characterizes a comprehensive genetic landscape of COVID-19 severity and provides novel insights into the molecular mechanisms of severe disease, leading to new therapeutic targets and sensitive detection of at-risk individuals.
Collapse
|
25
|
Baldassarri M, Fava F, Fallerini C, Daga S, Benetti E, Zguro K, Amitrano S, Valentino F, Doddato G, Giliberti A, Di Sarno L, Palmieri M, Carriero ML, Alaverdian D, Beligni G, Iuso N, Castelli F, Quiros-Roldan E, Mondelli MU, Miceli R, Frullanti E, Furini S, Mari F, Renieri A, Gabbi C, on behalf of the GEN-COVID Multicenter Study. Severe COVID-19 in Hospitalized Carriers of Single CFTR Pathogenic Variants. J Pers Med 2021; 11:558. [PMID: 34203982 PMCID: PMC8232773 DOI: 10.3390/jpm11060558] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 06/10/2021] [Accepted: 06/11/2021] [Indexed: 12/19/2022] Open
Abstract
The clinical presentation of COVID-19 is extremely heterogeneous, ranging from asymptomatic to severely ill patients. Thus, host genetic factors may be involved in determining disease presentation and progression. Given that carriers of single cystic fibrosis (CF)-causing variants of the CFTR gene-CF-carriers-are more susceptible to respiratory tract infections, our aim was to determine their likelihood of undergoing severe COVID-19. We implemented a cohort study of 874 individuals diagnosed with COVID-19, during the first pandemic wave in Italy. Whole exome sequencing was performed and validated CF-causing variants were identified. Forty subjects (16 females and 24 males) were found to be CF-carriers. Among mechanically ventilated patients, CF-carriers were more represented (8.7%) and they were significantly (p < 0.05) younger (mean age 51 years) compared to noncarriers (mean age 61.42 years). Furthermore, in the whole cohort, the age of male CF-carriers was lower, compared to noncarriers (p < 0.05). CF-carriers had a relative risk of presenting an abnormal inflammatory response (CRP ≥ 20 mg/dL) of 1.69 (p < 0.05) and their hazard ratio of death at day 14 was 3.10 (p < 0.05) in a multivariate regression model, adjusted for age, sex and comorbidities. In conclusion, CF-carriers are more susceptible to the severe form of COVID-19, showing also higher risk of 14-day death.
Collapse
Affiliation(s)
- Margherita Baldassarri
- Medical Genetics, University of Siena, 53100 Siena, Italy; (M.B.); (F.F.); (C.F.); (S.D.); (F.V.); (G.D.); (A.G.); (L.D.S.); (M.P.); (M.L.C.); (D.A.); (G.B.); (N.I.); (E.F.); (F.M.)
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (K.Z.); (S.F.)
| | - Francesca Fava
- Medical Genetics, University of Siena, 53100 Siena, Italy; (M.B.); (F.F.); (C.F.); (S.D.); (F.V.); (G.D.); (A.G.); (L.D.S.); (M.P.); (M.L.C.); (D.A.); (G.B.); (N.I.); (E.F.); (F.M.)
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (K.Z.); (S.F.)
- Genetica Medica, Azienda Ospedaliero-Universitaria Senese, 53100 Siena, Italy;
| | - Chiara Fallerini
- Medical Genetics, University of Siena, 53100 Siena, Italy; (M.B.); (F.F.); (C.F.); (S.D.); (F.V.); (G.D.); (A.G.); (L.D.S.); (M.P.); (M.L.C.); (D.A.); (G.B.); (N.I.); (E.F.); (F.M.)
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (K.Z.); (S.F.)
| | - Sergio Daga
- Medical Genetics, University of Siena, 53100 Siena, Italy; (M.B.); (F.F.); (C.F.); (S.D.); (F.V.); (G.D.); (A.G.); (L.D.S.); (M.P.); (M.L.C.); (D.A.); (G.B.); (N.I.); (E.F.); (F.M.)
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (K.Z.); (S.F.)
| | - Elisa Benetti
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (K.Z.); (S.F.)
| | - Kristina Zguro
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (K.Z.); (S.F.)
| | - Sara Amitrano
- Genetica Medica, Azienda Ospedaliero-Universitaria Senese, 53100 Siena, Italy;
| | - Floriana Valentino
- Medical Genetics, University of Siena, 53100 Siena, Italy; (M.B.); (F.F.); (C.F.); (S.D.); (F.V.); (G.D.); (A.G.); (L.D.S.); (M.P.); (M.L.C.); (D.A.); (G.B.); (N.I.); (E.F.); (F.M.)
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (K.Z.); (S.F.)
| | - Gabriella Doddato
- Medical Genetics, University of Siena, 53100 Siena, Italy; (M.B.); (F.F.); (C.F.); (S.D.); (F.V.); (G.D.); (A.G.); (L.D.S.); (M.P.); (M.L.C.); (D.A.); (G.B.); (N.I.); (E.F.); (F.M.)
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (K.Z.); (S.F.)
| | - Annarita Giliberti
- Medical Genetics, University of Siena, 53100 Siena, Italy; (M.B.); (F.F.); (C.F.); (S.D.); (F.V.); (G.D.); (A.G.); (L.D.S.); (M.P.); (M.L.C.); (D.A.); (G.B.); (N.I.); (E.F.); (F.M.)
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (K.Z.); (S.F.)
| | - Laura Di Sarno
- Medical Genetics, University of Siena, 53100 Siena, Italy; (M.B.); (F.F.); (C.F.); (S.D.); (F.V.); (G.D.); (A.G.); (L.D.S.); (M.P.); (M.L.C.); (D.A.); (G.B.); (N.I.); (E.F.); (F.M.)
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (K.Z.); (S.F.)
| | - Maria Palmieri
- Medical Genetics, University of Siena, 53100 Siena, Italy; (M.B.); (F.F.); (C.F.); (S.D.); (F.V.); (G.D.); (A.G.); (L.D.S.); (M.P.); (M.L.C.); (D.A.); (G.B.); (N.I.); (E.F.); (F.M.)
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (K.Z.); (S.F.)
| | - Miriam Lucia Carriero
- Medical Genetics, University of Siena, 53100 Siena, Italy; (M.B.); (F.F.); (C.F.); (S.D.); (F.V.); (G.D.); (A.G.); (L.D.S.); (M.P.); (M.L.C.); (D.A.); (G.B.); (N.I.); (E.F.); (F.M.)
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (K.Z.); (S.F.)
| | - Diana Alaverdian
- Medical Genetics, University of Siena, 53100 Siena, Italy; (M.B.); (F.F.); (C.F.); (S.D.); (F.V.); (G.D.); (A.G.); (L.D.S.); (M.P.); (M.L.C.); (D.A.); (G.B.); (N.I.); (E.F.); (F.M.)
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (K.Z.); (S.F.)
| | - Giada Beligni
- Medical Genetics, University of Siena, 53100 Siena, Italy; (M.B.); (F.F.); (C.F.); (S.D.); (F.V.); (G.D.); (A.G.); (L.D.S.); (M.P.); (M.L.C.); (D.A.); (G.B.); (N.I.); (E.F.); (F.M.)
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (K.Z.); (S.F.)
| | - Nicola Iuso
- Medical Genetics, University of Siena, 53100 Siena, Italy; (M.B.); (F.F.); (C.F.); (S.D.); (F.V.); (G.D.); (A.G.); (L.D.S.); (M.P.); (M.L.C.); (D.A.); (G.B.); (N.I.); (E.F.); (F.M.)
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (K.Z.); (S.F.)
| | - Francesco Castelli
- Department of Infectious and Tropical Diseases, University of Brescia and ASST Spedali Civili Hospital, 25123 Brescia, Italy; (F.C.); (E.Q.-R.)
| | - Eugenia Quiros-Roldan
- Department of Infectious and Tropical Diseases, University of Brescia and ASST Spedali Civili Hospital, 25123 Brescia, Italy; (F.C.); (E.Q.-R.)
| | - Mario Umberto Mondelli
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100 Pavia, Italy;
- Division of Infectious Diseases and Immunology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Rosalba Miceli
- Clinical Epidemiology and Trial Organization, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale Tumori di Milano, 20133 Milan, Italy;
| | - Elisa Frullanti
- Medical Genetics, University of Siena, 53100 Siena, Italy; (M.B.); (F.F.); (C.F.); (S.D.); (F.V.); (G.D.); (A.G.); (L.D.S.); (M.P.); (M.L.C.); (D.A.); (G.B.); (N.I.); (E.F.); (F.M.)
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (K.Z.); (S.F.)
| | - Simone Furini
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (K.Z.); (S.F.)
| | - Francesca Mari
- Medical Genetics, University of Siena, 53100 Siena, Italy; (M.B.); (F.F.); (C.F.); (S.D.); (F.V.); (G.D.); (A.G.); (L.D.S.); (M.P.); (M.L.C.); (D.A.); (G.B.); (N.I.); (E.F.); (F.M.)
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (K.Z.); (S.F.)
- Genetica Medica, Azienda Ospedaliero-Universitaria Senese, 53100 Siena, Italy;
| | - Alessandra Renieri
- Medical Genetics, University of Siena, 53100 Siena, Italy; (M.B.); (F.F.); (C.F.); (S.D.); (F.V.); (G.D.); (A.G.); (L.D.S.); (M.P.); (M.L.C.); (D.A.); (G.B.); (N.I.); (E.F.); (F.M.)
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (K.Z.); (S.F.)
- Genetica Medica, Azienda Ospedaliero-Universitaria Senese, 53100 Siena, Italy;
| | | | | |
Collapse
|
26
|
Cantalupo S, Lasorsa VA, Russo R, Andolfo I, D’Alterio G, Rosato BE, Frisso G, Abete P, Cassese GM, Servillo G, Gentile I, Piscopo C, Della Monica M, Fiorentino G, Russo G, Cerino P, Buonerba C, Pierri B, Zollo M, Iolascon A, Capasso M. Regulatory Noncoding and Predicted Pathogenic Coding Variants of CCR5 Predispose to Severe COVID-19. Int J Mol Sci 2021; 22:5372. [PMID: 34065289 PMCID: PMC8161088 DOI: 10.3390/ijms22105372] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/12/2021] [Accepted: 05/18/2021] [Indexed: 11/17/2022] Open
Abstract
Genome-wide association studies (GWAS) found locus 3p21.31 associated with severe COVID-19. CCR5 resides at the same locus and, given its known biological role in other infection diseases, we investigated if common noncoding and rare coding variants, affecting CCR5, can predispose to severe COVID-19. We combined single nucleotide polymorphisms (SNPs) that met the suggestive significance level (P ≤ 1 × 10-5) at the 3p21.31 locus in public GWAS datasets (6406 COVID-19 hospitalized patients and 902,088 controls) with gene expression data from 208 lung tissues, Hi-C, and Chip-seq data. Through whole exome sequencing (WES), we explored rare coding variants in 147 severe COVID-19 patients. We identified three SNPs (rs9845542, rs12639314, and rs35951367) associated with severe COVID-19 whose risk alleles correlated with low CCR5 expression in lung tissues. The rs35951367 resided in a CTFC binding site that interacts with CCR5 gene in lung tissues and was confirmed to be associated with severe COVID-19 in two independent datasets. We also identified a rare coding variant (rs34418657) associated with the risk of developing severe COVID-19. Our results suggest a biological role of CCR5 in the progression of COVID-19 as common and rare genetic variants can increase the risk of developing severe COVID-19 by affecting the functions of CCR5.
Collapse
Affiliation(s)
- Sueva Cantalupo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80136 Napoli, Italy; (S.C.); (V.A.L.); (R.R.); (I.A.); (B.E.R.); (G.F.); (M.Z.); (A.I.)
- CEINGE Biotecnologie Avanzate, 80145 Napoli, Italy;
| | - Vito Alessandro Lasorsa
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80136 Napoli, Italy; (S.C.); (V.A.L.); (R.R.); (I.A.); (B.E.R.); (G.F.); (M.Z.); (A.I.)
- CEINGE Biotecnologie Avanzate, 80145 Napoli, Italy;
| | - Roberta Russo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80136 Napoli, Italy; (S.C.); (V.A.L.); (R.R.); (I.A.); (B.E.R.); (G.F.); (M.Z.); (A.I.)
- CEINGE Biotecnologie Avanzate, 80145 Napoli, Italy;
| | - Immacolata Andolfo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80136 Napoli, Italy; (S.C.); (V.A.L.); (R.R.); (I.A.); (B.E.R.); (G.F.); (M.Z.); (A.I.)
- CEINGE Biotecnologie Avanzate, 80145 Napoli, Italy;
| | | | - Barbara Eleni Rosato
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80136 Napoli, Italy; (S.C.); (V.A.L.); (R.R.); (I.A.); (B.E.R.); (G.F.); (M.Z.); (A.I.)
- CEINGE Biotecnologie Avanzate, 80145 Napoli, Italy;
| | - Giulia Frisso
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80136 Napoli, Italy; (S.C.); (V.A.L.); (R.R.); (I.A.); (B.E.R.); (G.F.); (M.Z.); (A.I.)
- CEINGE Biotecnologie Avanzate, 80145 Napoli, Italy;
| | - Pasquale Abete
- COVID Hospital, P.O.S. Anna e SS. Madonna della Neve di Boscotrecase, Ospedali Riuniti Area Vesuviana, 80042 Boscotrecase, Italy; (P.A.); (G.M.C.)
| | - Gian Marco Cassese
- COVID Hospital, P.O.S. Anna e SS. Madonna della Neve di Boscotrecase, Ospedali Riuniti Area Vesuviana, 80042 Boscotrecase, Italy; (P.A.); (G.M.C.)
| | - Giuseppe Servillo
- Dipartimento di Neuroscienze e Scienze Riproduttive ed Odontostomatologiche, Università degli Studi di Napoli Federico II, 80131 Napoli, Italy;
| | - Ivan Gentile
- Dipartimento di Medicina Clinica e Chirurgia, Università degli Studi di Napoli Federico II, 80131 Napoli, Italy;
| | - Carmelo Piscopo
- Medical and Laboratory Genetics Unit, A.O.R.N. ‘Antonio Cardarelli’, 80131 Napoli, Italy; (C.P.); (M.D.M.)
| | - Matteo Della Monica
- Medical and Laboratory Genetics Unit, A.O.R.N. ‘Antonio Cardarelli’, 80131 Napoli, Italy; (C.P.); (M.D.M.)
| | | | - Giuseppe Russo
- Unità di Radiologia, Casa di Cura Villa dei Fiori, 80011 Acerra, Italy;
| | - Pellegrino Cerino
- Istituto Zooprofilattico Sperimentale del Mezzogiorno, 80055 Portici, Italy; (P.C.); (C.B.); (B.P.)
| | - Carlo Buonerba
- Istituto Zooprofilattico Sperimentale del Mezzogiorno, 80055 Portici, Italy; (P.C.); (C.B.); (B.P.)
| | - Biancamaria Pierri
- Istituto Zooprofilattico Sperimentale del Mezzogiorno, 80055 Portici, Italy; (P.C.); (C.B.); (B.P.)
- Dipartimento di Medicina, Chirurgia e Odontoiatria “Scuola Medica Salernitana”, Università di Salerno, 84081 Baronissi, Italy
| | - Massimo Zollo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80136 Napoli, Italy; (S.C.); (V.A.L.); (R.R.); (I.A.); (B.E.R.); (G.F.); (M.Z.); (A.I.)
- CEINGE Biotecnologie Avanzate, 80145 Napoli, Italy;
| | - Achille Iolascon
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80136 Napoli, Italy; (S.C.); (V.A.L.); (R.R.); (I.A.); (B.E.R.); (G.F.); (M.Z.); (A.I.)
- CEINGE Biotecnologie Avanzate, 80145 Napoli, Italy;
| | - Mario Capasso
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80136 Napoli, Italy; (S.C.); (V.A.L.); (R.R.); (I.A.); (B.E.R.); (G.F.); (M.Z.); (A.I.)
- CEINGE Biotecnologie Avanzate, 80145 Napoli, Italy;
| |
Collapse
|
27
|
Out now in May's EJHG. Eur J Hum Genet 2021; 29:721. [PMID: 33972741 PMCID: PMC8110754 DOI: 10.1038/s41431-021-00896-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
|
28
|
Bergantini L, Bargagli E, d'Alessandro M, Refini RM, Cameli P, Galasso L, Scapellato C, Montagnani F, Scolletta S, Franchi F, Valente S, Bennett D, Sebastiani G, Frediani B, Dotta F. Prognostic bioindicators in severe COVID-19 patients. Cytokine 2021; 141:155455. [PMID: 33548798 PMCID: PMC7843114 DOI: 10.1016/j.cyto.2021.155455] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 01/17/2021] [Accepted: 01/18/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND Severe acute respiratory syndrome caused by novel coronavirus 2 (SARS-CoV-2) emerged in Wuhan (China) in December 2019. Here we evaluated a panel of biomarkers to phenotype patients and to define the role of immuno-inflammatory mediators as biomarkers of severity. MATERIALS AND METHODS Serum samples were obtained from 24 COVID-19 patients on admission to hospital, before any treatment or infusion of intravenous steroids or invasive ventilation. KL-6 IL-6 and C-peptide were measured by chemiluminescent enzyme immunoassay. IL-6 assay was validated for accuracy and precision. The validity of variables used to distinguish severe from mild-to-moderate patients was assessed by areas under curves (AUC) of the receiver operating characteristic (ROC) and logistic regression was performed to combine parameters of the two groups. RESULTS In the severe group, IL-6, CRP and KL-6 concentrations were significantly higher than in mild-to-moderate patients. KL-6, IL-6 and CRP concentrations were directly correlated with each other. ROC curve analysis of the logistic regression model including IL-6, KL-6 and CRP showed the best performance with an AUC of 0.95. CONCLUSIONS Besides corroborating previous reports of over-expression of IL-6 in severe COVID-19 patients requiring mechanical ventilation, analytical determination of other mediators showed that IL-6 concentrations were correlated with those of KL-6 and CRP. The combination of these three prognostic bioindicators made it possible to distinguish severe COVID-19 patients with poor prognosis from mild-to-moderate patients.
Collapse
Affiliation(s)
- L Bergantini
- Respiratory Diseases and Lung Transplant Unit, Department of Medical and Surgical Sciences & Neurosciences, Siena University Hospital, Siena, Italy.
| | - E Bargagli
- Respiratory Diseases and Lung Transplant Unit, Department of Medical and Surgical Sciences & Neurosciences, Siena University Hospital, Siena, Italy
| | - M d'Alessandro
- Respiratory Diseases and Lung Transplant Unit, Department of Medical and Surgical Sciences & Neurosciences, Siena University Hospital, Siena, Italy
| | - R M Refini
- Respiratory Diseases and Lung Transplant Unit, Department of Medical and Surgical Sciences & Neurosciences, Siena University Hospital, Siena, Italy
| | - P Cameli
- Respiratory Diseases and Lung Transplant Unit, Department of Medical and Surgical Sciences & Neurosciences, Siena University Hospital, Siena, Italy
| | - L Galasso
- Clinical Pathology Unit, Innovation, Experimentation and Clinical and Translational Research Department, University Hospital of Siena, Siena, Italy
| | - C Scapellato
- Clinical Pathology Unit, Innovation, Experimentation and Clinical and Translational Research Department, University Hospital of Siena, Siena, Italy
| | - F Montagnani
- Department of Medical Biotechnologies, University of Siena, Italy; Infectious and Tropical Diseases Unit, Department of Medical Science, Siena University Hospital, Italy
| | - S Scolletta
- Anaesthesia and Intensive Care Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - F Franchi
- Anaesthesia and Intensive Care Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - S Valente
- Department of Cardiovascular Diseases, University of Siena, Siena, Italy
| | - D Bennett
- Respiratory Diseases and Lung Transplant Unit, Department of Medical and Surgical Sciences & Neurosciences, Siena University Hospital, Siena, Italy
| | - G Sebastiani
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena and Fondazione Umberto Di Mario ONLUS-Toscana Life Science Park, Siena, Italy
| | - B Frediani
- Rheumatology Unit, Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - F Dotta
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena and Fondazione Umberto Di Mario ONLUS-Toscana Life Science Park, Siena, Italy
| |
Collapse
|
29
|
Andolfo I, Russo R, Lasorsa VA, Cantalupo S, Rosato BE, Bonfiglio F, Frisso G, Abete P, Cassese GM, Servillo G, Esposito G, Gentile I, Piscopo C, Villani R, Fiorentino G, Cerino P, Buonerba C, Pierri B, Zollo M, Iolascon A, Capasso M. Common variants at 21q22.3 locus influence MX1 and TMPRSS2 gene expression and susceptibility to severe COVID-19. iScience 2021; 24:102322. [PMID: 33748697 PMCID: PMC7968217 DOI: 10.1016/j.isci.2021.102322] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 02/15/2021] [Accepted: 03/15/2021] [Indexed: 12/15/2022] Open
Abstract
The established risk factors of coronavirus disease 2019 (COVID-19) are advanced age, male sex, and comorbidities, but they do not fully explain the wide spectrum of disease manifestations. Genetic factors implicated in the host antiviral response provide for novel insights into its pathogenesis. We performed an in-depth genetic analysis of chromosome 21 exploiting the genome-wide association study data, including 6,406 individuals hospitalized for COVID-19 and 902,088 controls with European genetic ancestry from the COVID-19 Host Genetics Initiative. We found that five single nucleotide polymorphisms within TMPRSS2 and near MX1 gene show associations with severe COVID-19. The minor alleles of the five single nucleotide polymorphisms (SNPs) correlated with a reduced risk of developing severe COVID-19 and high level of MX1 expression in blood. Our findings demonstrate that host genetic factors can influence the different clinical presentations of COVID-19 and that MX1 could be a potential therapeutic target.
Collapse
Affiliation(s)
- Immacolata Andolfo
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples, Federico II, 80145 Naples, Italy
- CEINGE Biotecnologie Avanzate, Via Gaetano Salvatore, 486, 80145 Napoli, Italy
| | - Roberta Russo
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples, Federico II, 80145 Naples, Italy
- CEINGE Biotecnologie Avanzate, Via Gaetano Salvatore, 486, 80145 Napoli, Italy
| | - Vito Alessandro Lasorsa
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples, Federico II, 80145 Naples, Italy
- CEINGE Biotecnologie Avanzate, Via Gaetano Salvatore, 486, 80145 Napoli, Italy
| | - Sueva Cantalupo
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples, Federico II, 80145 Naples, Italy
- CEINGE Biotecnologie Avanzate, Via Gaetano Salvatore, 486, 80145 Napoli, Italy
| | - Barbara Eleni Rosato
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples, Federico II, 80145 Naples, Italy
- CEINGE Biotecnologie Avanzate, Via Gaetano Salvatore, 486, 80145 Napoli, Italy
| | - Ferdinando Bonfiglio
- Dipartimento di Ingegneria chimica, dei Materiali e della Produzione industriale, Università degli Studi di Napoli Federico II, Napoli, Italy
| | - Giulia Frisso
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples, Federico II, 80145 Naples, Italy
- CEINGE Biotecnologie Avanzate, Via Gaetano Salvatore, 486, 80145 Napoli, Italy
| | - Pasquale Abete
- COVID Hospital, P.O.S. Anna e SS. Madonna della Neve di Boscotrecase, Ospedali Riuniti Area Vesuviana, Napoli, Italy
| | - Gian Marco Cassese
- COVID Hospital, P.O.S. Anna e SS. Madonna della Neve di Boscotrecase, Ospedali Riuniti Area Vesuviana, Napoli, Italy
| | - Giuseppe Servillo
- Dipartimento di Neuroscienze e Scienze riproduttive ed odontostomatologiche, Università degli Studi di Napoli Federico II, Napoli, Italy
| | - Gabriella Esposito
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples, Federico II, 80145 Naples, Italy
- CEINGE Biotecnologie Avanzate, Via Gaetano Salvatore, 486, 80145 Napoli, Italy
| | - Ivan Gentile
- Dipartimento di Medicina clinica e Chirurgia, Università degli Studi di Napoli Federico II, Napoli, Italy
| | - Carmelo Piscopo
- Medical and Laboratory Genetics Unit, A.O.R.N. ‘Antonio Cardarelli’, Napoli, Italy
| | - Romolo Villani
- Poison Centre, A.O.R.N. ‘Antonio Cardarelli’, Napoli, Italy
| | | | - Pellegrino Cerino
- Istituto Zooprofilattico Sperimentale del Mezzogiorno, Napoli, Italy
| | - Carlo Buonerba
- Istituto Zooprofilattico Sperimentale del Mezzogiorno, Napoli, Italy
| | - Biancamaria Pierri
- Istituto Zooprofilattico Sperimentale del Mezzogiorno, Napoli, Italy
- Dipartimento di Medicina, Chirurgia e Odontoiatria "Scuola Medica Salernitana", Università di Salerno, Baronissi, Italy
| | - Massimo Zollo
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples, Federico II, 80145 Naples, Italy
- CEINGE Biotecnologie Avanzate, Via Gaetano Salvatore, 486, 80145 Napoli, Italy
| | - Achille Iolascon
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples, Federico II, 80145 Naples, Italy
- CEINGE Biotecnologie Avanzate, Via Gaetano Salvatore, 486, 80145 Napoli, Italy
| | - Mario Capasso
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples, Federico II, 80145 Naples, Italy
- CEINGE Biotecnologie Avanzate, Via Gaetano Salvatore, 486, 80145 Napoli, Italy
| |
Collapse
|
30
|
Monticelli M, Hay Mele B, Benetti E, Fallerini C, Baldassarri M, Furini S, Frullanti E, Mari F, GEN-COVID Multicenter Study, Andreotti G, Cubellis MV, Renieri A. Protective Role of a TMPRSS2 Variant on Severe COVID-19 Outcome in Young Males and Elderly Women. Genes (Basel) 2021; 12:596. [PMID: 33921689 PMCID: PMC8073081 DOI: 10.3390/genes12040596] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 04/09/2021] [Indexed: 02/06/2023] Open
Abstract
The protease encoded by the TMPRSS2 gene facilitates viral infections and has been implicated in the pathogenesis of SARS-CoV-2. We analyzed the TMPRSS2 sequence and correlated the protein variants with the clinical features of a cohort of 1177 patients affected by COVID-19 in Italy. Nine relatively common variants (allele frequency > 0.01) and six missense variants which may affect the protease activity according to PolyPhen-2 in HumVar-trained mode were identified. Among them, p.V197M (p.Val197Met) (rs12329760) emerges as a common variant that has a deleterious effect on the protease and a protective effect on the patients. Its role appears particularly relevant in two subgroups of patients-young males and elderly women-and among those affected by co-morbidities, where the variant frequency is higher among individuals who were mildly affected by the disease and did not need hospitalization or oxygen therapy than among those more severely affected, who required oxygen therapy, ventilation or intubation. This study provides useful information for the identification of patients at risk of developing a severe form of COVID-19, and encourages the usage of drugs affecting the expression of TMPRSS2 or inhibiting protein activity.
Collapse
Affiliation(s)
- Maria Monticelli
- Department of Biology, Università Federico II, 80126 Napoli, Italy; (M.M.); (B.H.M.)
| | - Bruno Hay Mele
- Department of Biology, Università Federico II, 80126 Napoli, Italy; (M.M.); (B.H.M.)
- Integrative Marine Ecology Department, Stazione Zoologica Anton Dohrn, Villa Comunale, 80121 Napoli, Italy
| | - Elisa Benetti
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (C.F.); (M.B.); (S.F.); (E.F.); (F.M.); (A.R.)
| | - Chiara Fallerini
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (C.F.); (M.B.); (S.F.); (E.F.); (F.M.); (A.R.)
- Medical Genetics, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Margherita Baldassarri
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (C.F.); (M.B.); (S.F.); (E.F.); (F.M.); (A.R.)
- Medical Genetics, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Simone Furini
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (C.F.); (M.B.); (S.F.); (E.F.); (F.M.); (A.R.)
| | - Elisa Frullanti
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (C.F.); (M.B.); (S.F.); (E.F.); (F.M.); (A.R.)
- Medical Genetics, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Francesca Mari
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (C.F.); (M.B.); (S.F.); (E.F.); (F.M.); (A.R.)
- Medical Genetics, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
- Genetica Medica, Azienda Ospedaliero-Universitaria Senese, 53100 Siena, Italy
| | | | | | - Maria Vittoria Cubellis
- Department of Biology, Università Federico II, 80126 Napoli, Italy; (M.M.); (B.H.M.)
- Istituto di Chimica Biomolecolare—CNR, 80078 Pozzuoli, Italy
| | - Alessandra Renieri
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (C.F.); (M.B.); (S.F.); (E.F.); (F.M.); (A.R.)
- Medical Genetics, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
- Genetica Medica, Azienda Ospedaliero-Universitaria Senese, 53100 Siena, Italy
| |
Collapse
|
31
|
Fallerini C, Daga S, Mantovani S, Benetti E, Picchiotti N, Francisci D, Paciosi F, Schiaroli E, Baldassarri M, Fava F, Palmieri M, Ludovisi S, Castelli F, Quiros-Roldan E, Vaghi M, Rusconi S, Siano M, Bandini M, Spiga O, Capitani K, Furini S, Mari F, Renieri A, Mondelli MU, Frullanti E. Association of Toll-like receptor 7 variants with life-threatening COVID-19 disease in males: findings from a nested case-control study. eLife 2021; 10:67569. [PMID: 33650967 PMCID: PMC7987337 DOI: 10.7554/elife.67569] [Citation(s) in RCA: 151] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 02/24/2021] [Indexed: 12/26/2022] Open
Abstract
Background: Recently, loss-of-function variants in TLR7 were identified in two families in which COVID-19 segregates like an X-linked recessive disorder environmentally conditioned by SARS-CoV-2. We investigated whether the two families represent the tip of the iceberg of a subset of COVID-19 male patients. Methods: This is a nested case-control study in which we compared male participants with extreme phenotype selected from the Italian GEN-COVID cohort of SARS-CoV-2-infected participants (<60 y, 79 severe cases versus 77 control cases). We applied the LASSO Logistic Regression analysis, considering only rare variants on young male subsets with extreme phenotype, picking up TLR7 as the most important susceptibility gene. Results: Overall, we found TLR7 deleterious variants in 2.1% of severely affected males and in none of the asymptomatic participants. The functional gene expression profile analysis demonstrated a reduction in TLR7-related gene expression in patients compared with controls demonstrating an impairment in type I and II IFN responses. Conclusions: Young males with TLR7 loss-of-function variants and severe COVID-19 represent a subset of male patients contributing to disease susceptibility in up to 2% of severe COVID-19. Funding: Funded by private donors for the Host Genetics Research Project, the Intesa San Paolo for 2020 charity fund, and the Host Genetics Initiative. Clinical trial number: NCT04549831.
Collapse
Affiliation(s)
- Chiara Fallerini
- Medical Genetics, University of Siena, Siena, Italy.,Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Sergio Daga
- Medical Genetics, University of Siena, Siena, Italy.,Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Stefania Mantovani
- Division of Infectious Diseases and Immunology, Department of Medical Sciences and Infectious Diseases, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Elisa Benetti
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Nicola Picchiotti
- Department of Mathematics, University of Pavia, Pavia, Italy.,University of Siena, DIISM-SAILAB, Siena, Italy
| | - Daniela Francisci
- Infectious Diseases Clinic, Department of Medicine 2, Azienda Ospedaliera di Perugia and University of Perugia, Santa Maria Hospital, Perugia, Italy.,Infectious Diseases Clinic, "Santa Maria" Hospital, University of Perugia, Perugia, Italy
| | - Francesco Paciosi
- Infectious Diseases Clinic, Department of Medicine 2, Azienda Ospedaliera di Perugia and University of Perugia, Santa Maria Hospital, Perugia, Italy.,Infectious Diseases Clinic, "Santa Maria" Hospital, University of Perugia, Perugia, Italy
| | - Elisabetta Schiaroli
- Infectious Diseases Clinic, Department of Medicine 2, Azienda Ospedaliera di Perugia and University of Perugia, Santa Maria Hospital, Perugia, Italy
| | - Margherita Baldassarri
- Medical Genetics, University of Siena, Siena, Italy.,Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Francesca Fava
- 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
| | - Maria Palmieri
- Medical Genetics, University of Siena, Siena, Italy.,Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Serena Ludovisi
- Division of Infectious Diseases and Immunology, Department of Medical Sciences and Infectious Diseases, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy.,Department of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy
| | - Francesco Castelli
- Department of Infectious and Tropical Diseases, University of Brescia and ASST Spedali Civili Hospital, Brescia, Italy
| | - Eugenia Quiros-Roldan
- Department of Infectious and Tropical Diseases, University of Brescia and ASST Spedali Civili Hospital, Brescia, Italy
| | - Massimo Vaghi
- Chirurgia Vascolare, Ospedale Maggiore di Crema, Crema, Italy
| | - Stefano Rusconi
- Department of Biomedical and Clinical Sciences Luigi Sacco, University of Milan, Milan, Italy.,III Infectious Diseases Unit, ASST-FBF-Sacco, Milan, Italy
| | - Matteo Siano
- Department of Biomedical and Clinical Sciences Luigi Sacco, University of Milan, Milan, Italy
| | - Maria Bandini
- Department of Preventive Medicine, Azienda USL Toscana Sud Est, Siena, Italy
| | - Ottavia Spiga
- University of Siena, DIISM-SAILAB, Siena, Italy.,Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Katia Capitani
- Medical Genetics, University of Siena, Siena, Italy.,Molecular Mechanisms of Oncogenesis, ISPRO Core Research Laboratory (CRL), Firenze, Italy
| | - Simone Furini
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Francesca Mari
- 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
| | -
- Medical Genetics, University of Siena, Siena, Italy
| | - 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
| | - Mario U Mondelli
- Division of Infectious Diseases and Immunology, Department of Medical Sciences and Infectious Diseases, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy.,Department of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy
| | - Elisa Frullanti
- Medical Genetics, University of Siena, Siena, Italy.,Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| |
Collapse
|
32
|
Shorter androgen receptor polyQ alleles protect against life-threatening COVID-19 disease in European males. EBioMedicine 2021; 65:103246. [PMID: 33647767 PMCID: PMC7908850 DOI: 10.1016/j.ebiom.2021.103246] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/24/2021] [Accepted: 02/02/2021] [Indexed: 12/14/2022] Open
Abstract
Background While SARS-CoV-2 similarly infects men and women, COVID-19 outcome is less favorable in men. Variability in COVID-19 severity may be explained by differences in the host genome. Methods We compared poly-amino acids variability from WES data in severely affected COVID-19 patients versus SARS-CoV-2 PCR-positive oligo-asymptomatic subjects. Findings Shorter polyQ alleles (≤22) in the androgen receptor (AR) conferred protection against severe outcome in COVID-19 in the first tested cohort (both males and females) of 638 Italian subjects. The association between long polyQ alleles (≥23) and severe clinical outcome (p = 0.024) was also validated in an independent cohort of Spanish men <60 years of age (p = 0.014). Testosterone was higher in subjects with AR long-polyQ, possibly indicating receptor resistance (p = 0.042 Mann-Whitney U test). Inappropriately low serum testosterone level among carriers of the long-polyQ alleles (p = 0.0004 Mann-Whitney U test) predicted the need for intensive care in COVID-19 infected men. In agreement with the known anti-inflammatory action of testosterone, patients with long-polyQ and age ≥60 years had increased levels of CRP (p = 0.018, not accounting for multiple testing). Interpretation We identify the first genetic polymorphism that appears to predispose some men to develop more severe disease. Failure of the endocrine feedback to overcome AR signaling defects by increasing testosterone levels during the infection leads to the polyQ tract becoming dominant to serum testosterone levels for the clinical outcome. These results may contribute to designing reliable clinical and public health measures and provide a rationale to test testosterone as adjuvant therapy in men with COVID-19 expressing long AR polyQ repeats. Funding MIUR project “Dipartimenti di Eccellenza 2018-2020” to Department of Medical Biotechnologies University of Siena, Italy (Italian D.L. n.18 March 17, 2020) and “Bando Ricerca COVID-19 Toscana” project to Azienda Ospedaliero-Universitaria Senese. Private donors for COVID-19 research and charity funds from Intesa San Paolo.
Collapse
|