Hostname: page-component-76fb5796d-45l2p Total loading time: 0 Render date: 2024-04-26T09:56:29.872Z Has data issue: false hasContentIssue false

COVID-19 pandemic: insights into molecular mechanisms leading to sex-based differences in patient outcomes

Published online by Cambridge University Press:  03 August 2021

Ashutosh Kumar*
Affiliation:
Etiologically Elusive Disorders Research Network (EEDRN), New Delhi, India Department of Anatomy, All India Institute of Medical Sciences (AIIMS), Patna, India
Ravi K. Narayan
Affiliation:
Etiologically Elusive Disorders Research Network (EEDRN), New Delhi, India Department of Anatomy, All India Institute of Medical Sciences (AIIMS), Patna, India
Maheswari Kulandhasamy
Affiliation:
Etiologically Elusive Disorders Research Network (EEDRN), New Delhi, India Department of Biochemistry, Maulana Azad Medical College (MAMC), New Delhi, India
Pranav Prasoon
Affiliation:
Etiologically Elusive Disorders Research Network (EEDRN), New Delhi, India Pittsburgh Center for Pain Research, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
Chiman Kumari
Affiliation:
Etiologically Elusive Disorders Research Network (EEDRN), New Delhi, India Department of Anatomy, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
Sujeet Kumar
Affiliation:
Etiologically Elusive Disorders Research Network (EEDRN), New Delhi, India Centre for Proteomics and Drug Discovery, Amity Institute of Biotechnology, Amity University, Mumbai, Maharashtra, India
Vikas Pareek
Affiliation:
Etiologically Elusive Disorders Research Network (EEDRN), New Delhi, India Centre for Cognitive and Brain Sciences, Indian Institute of Technology, Gandhinagar, Gujarat, India
Kishore Sesham
Affiliation:
Etiologically Elusive Disorders Research Network (EEDRN), New Delhi, India Department of Anatomy, All India Institute of Medical Sciences (AIIMS), Mangalagiri, Andhra Pradesh, India
Prakash S. Shekhawat
Affiliation:
Etiologically Elusive Disorders Research Network (EEDRN), New Delhi, India Department of Hematology, Nil Ratan Sircar Medical College and Hospital (NRSMCH), Kolkata, India
Kamla Kant
Affiliation:
Etiologically Elusive Disorders Research Network (EEDRN), New Delhi, India Department of Microbiology, All India Institute of Medical Sciences (AIIMS), Bathinda, India
Santosh Kumar
Affiliation:
Etiologically Elusive Disorders Research Network (EEDRN), New Delhi, India Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, USA
*
Author for correspondence: Ashutosh Kumar, E-mail: drashutoshkumar@aiimspatna.org

Abstract

Recent epidemiological studies analysing sex-disaggregated patient data of coronavirus disease 2019 (COVID-19) across the world revealed a distinct sex bias in the disease morbidity as well as the mortality – both being higher for the men. Similar antecedents have been known for the previous viral infections, including from coronaviruses, such as severe acute respiratory syndrome (SARS) and middle-east respiratory syndrome (MERS). A sound understanding of molecular mechanisms leading to the biological sex bias in the survival outcomes of the patients in relation to COVID-19 will act as an essential requisite for developing a sex-differentiated approach for therapeutic management of this disease. Recent studies which have explored molecular mechanism(s) behind sex-based differences in COVID-19 pathogenesis are scarce; however, existing evidence, for other respiratory viral infections, viz. SARS, MERS and influenza, provides important clues in this regard. In attempt to consolidate the available knowledge on this issue, we conducted a systematic review of the existing empirical knowledge and recent experimental studies following Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. The qualitative analysis of the collected data unravelled multiple molecular mechanisms, such as evolutionary and genetic/epigenetic factors, sex-linkage of viral host cell entry receptor and immune response genes, sex hormone and gut microbiome-mediated immune-modulation, as the possible key reasons for the sex-based differences in patient outcomes in COVID-19.

Type
Review
Copyright
Copyright © The Author(s), 2021. Published by Cambridge University Press

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Takahashi, T, et al. (2020) Sex differences in immune responses that underlie COVID-19 disease outcomes. Nature 588, 315320.CrossRefGoogle ScholarPubMed
Channappanavar, R, et al. (2017) Sex-based differences in susceptibility to severe acute respiratory syndrome coronavirus infection. Journal of Immunology 198, 40464053.CrossRefGoogle ScholarPubMed
Scully, EP, et al. (2020) Considering how biological sex impacts immune responses and COVID-19 outcomes. Nature Reviews Immunology 20, 442447.CrossRefGoogle ScholarPubMed
Bunders, MJ and Altfeld, M (2020) Implications of sex differences in immunity for SARS-CoV-2 pathogenesis and design of therapeutic interventions. Immunity 53, 487495.CrossRefGoogle ScholarPubMed
Úbeda, F and Jansen, VAA (2016) The evolution of sex-specific virulence in infectious diseases. Nature Communications 7, 13849.CrossRefGoogle ScholarPubMed
Zuk, M and Stoehr, AM (2010) Sex differences in susceptibility to infection: An evolutionary perspective. In Klein, SL and Roberts, CW (eds), Sex Hormones and Immunity to Infection. Berlin, Heidelberg: Springer-Verlag Berlin Heidelberg, pp. 117.Google Scholar
Taneja, V (2018) Sex hormones determine immune response. Frontiers in Immunology 9, 1931.CrossRefGoogle ScholarPubMed
Dhar, D and Mohanty, A (2020) Gut microbiota and COVID-19- possible link and implications. Virus Research 285, 198018.CrossRefGoogle ScholarPubMed
Jin, J-M, et al. (2020) Gender differences in patients with COVID-19: focus on severity and mortality. Frontiers in Public Health 8, 152.CrossRefGoogle ScholarPubMed
Williamson, EJ, et al. (2020) Factors associated with COVID-19-related death using OpenSAFELY. Nature 584, 430436.CrossRefGoogle ScholarPubMed
Peckham, H, et al. (2020) Male sex identified by global COVID-19 meta-analysis as a risk factor for death and ITU admission. Nature Communications 11, 6317.CrossRefGoogle ScholarPubMed
Yanez, ND, et al. (2020) COVID-19 mortality risk for older men and women. BMC Public Health 20, 1742.CrossRefGoogle ScholarPubMed
Nasiri, MJ, et al. (2020) COVID-19 clinical characteristics, and sex-specific risk of mortality: systematic review and meta-analysis. Frontiers in Medicine 7, 459.CrossRefGoogle ScholarPubMed
Dataset Global Health 50/50. Available at https://globalhealth5050.org/the-sex-gender-and-covid-19-project/dataset/ (Accessed 13 October 2020).Google Scholar
Kumar, S, et al. (2020) Assessment of the prevalence of symptoms in patients under institutional isolation in COVID-19 pandemic in India. Indian Journal of Palliative Care 26, 89.Google ScholarPubMed
Dehingia, N and Raj, A (2020) Sex differences in COVID-19 case fatality: do we know enough? Lancet Global Health 9, e14e15.CrossRefGoogle Scholar
Hoffmann, M, et al. (2020) SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell 181, 271280, e8.CrossRefGoogle ScholarPubMed
Zipeto, D, et al. (2020) ACE2/ADAM17/TMPRSS2 interplay may be the main risk factor for COVID-19. Frontiers in immunology 11, 576745.CrossRefGoogle ScholarPubMed
Glowacka, I, et al. (2010) Differential downregulation of ACE2 by the spike proteins of severe acute respiratory syndrome coronavirus and human coronavirus NL63. Journal of Virology 84, 1198–205.CrossRefGoogle ScholarPubMed
Kumar, A, et al. (2020) SARS-CoV-2 cell entry receptor ACE2 mediated endothelial dysfunction leads to vascular thrombosis in COVID-19 patients. Medical Hypotheses 145, 110320.CrossRefGoogle ScholarPubMed
Wang, Z and Xu, X (2020) scRNA-seq profiling of human testes reveals the presence of the ACE2 receptor, a target for SARS-CoV-2 infection in spermatogonia, Leydig and Sertoli cells. Cells 9, 920.CrossRefGoogle ScholarPubMed
Shen, Q, et al. (2020) The ACE2 expression in Sertoli cells and germ cells may cause male reproductive disorder after SARS-CoV-2 infection. Journal of Cellular and Molecular Medicine 24, 94729477.CrossRefGoogle ScholarPubMed
Xu, J, et al. (2006) Orchitis: a complication of severe acute respiratory syndrome (SARS). Biology of Reproduction 74, 410416.CrossRefGoogle Scholar
Deshmukh, V, et al. (2021) Histopathological observations in COVID-19: a systematic review. Journal of Clinical Pathology 74, 7683.CrossRefGoogle ScholarPubMed
Cardona Maya, WD, Du Plessis, SS and Velilla, PA (2020) SARS-CoV-2 and the testis: similarity with other viruses and routes of infection. Reproductive Biomedicine Online 40, 763764.CrossRefGoogle ScholarPubMed
Zhao, J, et al. (2003) Clinical pathology and pathogenesis of severe acute respiratory syndrome. Zhonghua Shi Yan He Lin Chuang Bing Du Xue Za Zhi 17, 217–21 (in Chinese).Google ScholarPubMed
SARS-CoV-2 related proteins – The Human Protein Atlas. Available at https://www.proteinatlas.org/humanproteome/sars-cov-2 (Accessed 1 June 2020).Google Scholar
Montopoli, M, et al. (2020) Androgen-deprivation therapies for prostate cancer and risk of infection by SARS-CoV-2: a population-based study (N = 4532). Annals of Oncology 8, 10401045.CrossRefGoogle Scholar
Mohamed, MS, et al. (2021) Sex differences in COVID-19: the role of androgens in disease severity and progression. Endocrine 71, 38.CrossRefGoogle ScholarPubMed
Baratchian, M, et al. (2020) No evidence that androgen regulation of pulmonary TMPRSS2 explains sex-discordant COVID-19 outcomes. bioRxiv [Preprint] 2020.04.21.051201.Google Scholar
Samuel, RM, et al. (2020) Androgen signaling regulates SARS-CoV-2 receptor levels and is associated with severe COVID-19 symptoms in men. Cell Stem Cell 27, 876889, e12.CrossRefGoogle ScholarPubMed
Schurz, H, et al. (2019) The X chromosome and sex-specific effects in infectious disease susceptibility. Human Genomics 13, 2.CrossRefGoogle ScholarPubMed
Klein, SL and Flanagan, KL (2016) Sex differences in immune responses. Nature Review Immunology 16, 626638.CrossRefGoogle ScholarPubMed
Kucharzik, T, et al. (1998) IL-4, IL-10 and IL-13 down-regulate monocyte-chemoattracting protein-1 (MCP-1) production in activated intestinal epithelial cells. Clinical and Experimental Immunology 111, 152157.CrossRefGoogle ScholarPubMed
Rudensky, AY (2011) Regulatory T cells and Foxp3. Immunological Reviews 241, 260268.CrossRefGoogle ScholarPubMed
Tukiainen, T, et al. (2017) Landscape of X chromosome inactivation across human tissues. Nature 550, 244248.CrossRefGoogle ScholarPubMed
Ziegler, CGK, et al. (2020) SARS-CoV-2 receptor ACE2 is an interferon-stimulated gene in human airway epithelial cells and is detected in specific cell subsets across tissues. Cell 181, 10161035, e19.CrossRefGoogle ScholarPubMed
Stopsack, KH, et al. (2020) TMPRSS2 and COVID-19: serendipity or opportunity for intervention? Cancer Discovery 10, 779782.CrossRefGoogle ScholarPubMed
Benetti, E, et al. (2020) ACE2 gene variants may underlie interindividual variability and susceptibility to COVID-19 in the Italian population. European Journal of Human Genetics 28, 16021614.CrossRefGoogle ScholarPubMed
Cao, Y, et al. (2020) Comparative genetic analysis of the novel coronavirus (2019-nCoV/SARS-CoV-2) receptor ACE2 in different populations. Cell Discovery 6, 14.CrossRefGoogle ScholarPubMed
van der Made, CI, et al. (2020) Presence of genetic variants among young men with severe COVID-19. JAMA 324, 663673.CrossRefGoogle ScholarPubMed
Hou, Y, et al. (2020) New insights into genetic susceptibility of COVID-19: An ACE2 and TMPRSS2 polymorphism analysis. BMC Medicine 18, 216.CrossRefGoogle ScholarPubMed
Feng, Y, et al. (2017) Extracellular microRNAs induce potent innate immune responses via TLR7/MyD88-dependent mechanisms. Journal of Immunology 199, 21062117.CrossRefGoogle ScholarPubMed
Pontecorvi, G, et al. (2020) microRNAs as new possible actors in gender disparities of COVID-19 pandemic. Acta Physiologica 230, e13538.CrossRefGoogle ScholarPubMed
Fink, AL (2018) Biological sex affects vaccine efficacy and protection against influenza in mice. Proceedings of the National Academy of Sciences USA 115, 1247712482.CrossRefGoogle ScholarPubMed
Flanagan, KL, et al. (2017) Sex and gender differences in the outcomes of vaccination over the life course. Annual Review of Cell and Developmental Biology 33, 577599.CrossRefGoogle ScholarPubMed
Bukowska, A, et al. (2017) Protective regulation of the ACE2/ACE gene expression by estrogen in human atrial tissue from elderly men. Experimental Biology and Medicine 242, 14121423.CrossRefGoogle ScholarPubMed
Viveiros, A, et al. (2021) Sex differences in COVID-19: candidate pathways, genetics of ACE2, and sex hormones. American Journal of Physiology-Heart and Circulatory Physiology 320, H296304.CrossRefGoogle ScholarPubMed
Hall, OJ and Klein, SL (2017) Progesterone-based compounds affect immune responses and susceptibility to infections at diverse mucosal sites. Mucosal Immunology 10, 10971107.CrossRefGoogle ScholarPubMed
Ghazizadeh, Z, et al. (2020) Androgen regulates SARS-CoV-2 receptor levels and is associated with severe COVID-19 symptoms in men. BioRxiv. the preprint server for biology 2020.05.12.091082.Google ScholarPubMed
Gadi, N, et al. (2020) What's sex got to do with COVID-19? Gender-based differences in the host immune response to coronaviruses. Frontiers in Immunology 11, 2147.CrossRefGoogle Scholar
Furman, D, et al. (2014) Systems analysis of sex differences reveals an immunosuppressive role for testosterone in the response to influenza vaccination. Proceedings of the National Academy of Sciences USA 111, 869874.CrossRefGoogle ScholarPubMed
Mohamad, NV, et al. (2019) The relationship between circulating testosterone and inflammatory cytokines in men. The Aging Male 22, 129140.CrossRefGoogle ScholarPubMed
Çayan, S, et al. (2020) Effect of serum total testosterone and its relationship with other laboratory parameters on the prognosis of coronavirus disease 2019 (COVID-19) in SARS-CoV-2 infected male patients: a cohort study. The Aging Male 23, 111.CrossRefGoogle ScholarPubMed
Bhopal, SS and Bhopal, R (2020) Sex differential in COVID-19 mortality varies markedly by age. Lancet (London, England) 396, 532533.CrossRefGoogle ScholarPubMed
Kumar, A, et al. (2021) Pathogenesis guided therapeutic management of COVID-19: an immunological perspective. International Reviews in Immunology 40, 5471.CrossRefGoogle ScholarPubMed
Coperchini, F, et al. (2020) The cytokine storm in COVID-19: an overview of the involvement of the chemokine/chemokine-receptor system. Cytokine Growth Factor Review 53, 2532.CrossRefGoogle ScholarPubMed
Dhar, SK, et al. (2021) IL-6 and IL-10 as predictors of disease severity in COVID-19 patients: results from meta-analysis and regression. Heliyon 7, e06155.CrossRefGoogle ScholarPubMed
Zhang, J, et al. (2020) Serum interleukin-6 is an indicator for severity in 901 patients with SARS-CoV-2 infection: a cohort study. Journal of Translational Medicine 18, 406.CrossRefGoogle ScholarPubMed
Fuseini, H, et al. (2018) Testosterone decreases house dust mite-induced type 2 and IL-17A-mediated airway inflammation. Journal of Immunology 201, 18431854.CrossRefGoogle ScholarPubMed
Fuseini, H, et al. (2019) ERα signaling increased IL-17A production in Th17 cells by upregulating IL-23R expression, mitochondrial respiration, and proliferation. Frontiers in Immunology 10, 2740.CrossRefGoogle ScholarPubMed
Newcomb, DC, et al. (2015) Estrogen and progesterone decrease let-7f microRNA expression and increase IL-23/IL-23 receptor signaling and IL-17A production in patients with severe asthma. Journal of Allergy and Clinical Immunology 136, 10251034, e11.CrossRefGoogle ScholarPubMed
Lindemans, CA, et al. (2015) Interleukin-22 promotes intestinal-stem-cell-mediated epithelial regeneration. Nature 528, 560564.CrossRefGoogle ScholarPubMed
Pociask, DA, et al. (2013) IL-22 is essential for lung epithelial repair following influenza infection. American Journal of Pathology 182, 12861296.CrossRefGoogle ScholarPubMed
Ziegler, SM and Altfeld, M (2017) Human immunodeficiency virus 1 and type I interferons – where sex makes a difference. Frontiers in Immunology 8, 1224.CrossRefGoogle ScholarPubMed
Griesbeck, M, et al. (2015) Sex differences in plasmacytoid dendritic cell levels of IRF5 drive higher IFN-production in women. Journal of Immunology 195, 53275336.CrossRefGoogle Scholar
Karnam, G, et al. (2012) CD200 receptor controls sex-specific TLR7 responses to viral infection. PLoS Pathogen 8, e1002710.CrossRefGoogle ScholarPubMed
Scheuplein, VA, et al. (2015) High secretion of interferons by human plasmacytoid dendritic cells upon recognition of middle east respiratory syndrome coronavirus. Journal of Virology 89, 38593869.CrossRefGoogle ScholarPubMed
Li, SW, et al. (2016) SARS coronavirus papain-like protease inhibits the TLR7 signaling pathway through removing Lys63-linked polyubiquitination of TRAF3 and TRAF6. International Journal of Molecular Sciences 17, 678.CrossRefGoogle ScholarPubMed
Haagmans, BL, et al. (2004) Pegylated interferon-α protects type 1 pneumocytes against SARS coronavirus infection in macaques. Nature Medicine 10, 290293.CrossRefGoogle ScholarPubMed
Mantlo, E, et al. (2020) Antiviral activities of type I interferons to SARS-CoV-2 infection. Antiviral Research 179, 104811.CrossRefGoogle ScholarPubMed
Berghöfer, B, et al. (2006) TLR7 ligands induce higher IFN-α production in females. Journal of Immunology 177, 20882096.CrossRefGoogle ScholarPubMed
Chang, JJ, et al. (2013) Higher expression of several interferon-stimulated genes in HIV-1-infected females after adjusting for the level of viral replication. Journal of Infectious Diseases 208, 830838.CrossRefGoogle ScholarPubMed
Meier, A, et al. (2009) Sex differences in the Toll-like receptor-mediated response of plasmacytoid dendritic cells to HIV-1. Nature Medicine 15, 955959.CrossRefGoogle ScholarPubMed
Seillet, C, et al. (2012) The TLR-mediated response of plasmacytoid dendritic cells is positively regulated by estradiol in vivo through cell-intrinsic estrogen receptor α signaling. Blood 119, 454464.CrossRefGoogle ScholarPubMed
Blanco-Melo, D, et al. (2020) Imbalanced host response to SARS-CoV-2 drives development of COVID-19. Cell 181, 10361045, e9.CrossRefGoogle ScholarPubMed
Chu, H, et al. (2020) Comparative replication and immune activation profiles of SARS-CoV-2 and SARS-CoV in human lungs: an ex vivo study with implications for the pathogenesis of COVID-19. Clinical Infectious Diseases 71, 14001409.CrossRefGoogle Scholar
Zhou, Z, et al. (2020) Heightened innate immune responses in the respiratory tract of COVID-19 patients. Cell Host & Microbe 27, 883890, e2.CrossRefGoogle ScholarPubMed
Zheng, S, et al. (2020) Viral load dynamics and disease severity in patients infected with SARS-CoV-2 in Zhejiang province, China, January–March 2020: retrospective cohort study. BMJ 369, m1443.CrossRefGoogle ScholarPubMed
Xu, K, et al. (2020) Factors associated with prolonged viral RNA shedding in patients with coronavirus disease 2019 (COVID-19). Clinical Infectious Diseases 71, 799806.CrossRefGoogle Scholar
Gause, WC, Rothlin, C and Loke, P (2020) Heterogeneity in the initiation, development and function of type 2 immunity. Nature Reviews Immunology 20, 603614.CrossRefGoogle ScholarPubMed
Laffont, S, et al. (2017) Androgen signaling negatively controls group 2 innate lymphoid cells. Journal of Experimental Medicine 214, 15811592.CrossRefGoogle ScholarPubMed
Ricardo-Gonzalez, RR, et al. (2018) Tissue signals imprint ILC2 identity with anticipatory function. Nature Immunology 19, 10931099.CrossRefGoogle ScholarPubMed
Peters, MC, et al. (2020) COVID-19-related genes in sputum cells in asthma: relationship to demographic features and corticosteroids. American Journal of Respiratory and Critical Care Medicine 202, 8390.CrossRefGoogle ScholarPubMed
Bastard, P, et al. (2020) Auto-antibodies against type I IFNs in patients with life-threatening COVID-19. Science (New York, N.Y.) 65, eabd4585.CrossRefGoogle Scholar
Tadount, F, et al. (2020) Is there a difference in the immune response, efficacy, effectiveness and safety of seasonal influenza vaccine in males and females? – A systematic review. Vaccine 38, 444459.CrossRefGoogle Scholar
van Zyl-Smit, RN, Richards, G and Leone, FT (2020) Tobacco smoking and COVID-19 infection. Lancet Respiratory Medicine 8, 664665.CrossRefGoogle ScholarPubMed
Alqahtani, JS, et al. (2020) Prevalence, severity and mortality associated with COPD and smoking in patients with COVID-19: a rapid systematic review and meta-analysis. PLoS One 15, e0233147.CrossRefGoogle ScholarPubMed
WHO (2020) Smoking and COVID-19. Available at https://www.who.int/news-room/commentaries/detail/smoking-and-covid-19 (Accessed 5 October 5 2020).Google Scholar
Sankaran-Walters, S, et al. (2013) Sex differences matter in the gut: effect on mucosal immune activation and inflammation. Biology of Sex Differences 4, 10.CrossRefGoogle ScholarPubMed
Shin, JH, et al. (2019) Serum level of sex steroid hormone is associated with diversity and profiles of human gut microbiome. Respiratory Microbiology 170, 192201.Google ScholarPubMed
Haitao, T, et al. (2020) COVID-19 and sex differences: mechanisms and biomarkers. Mayo Clinic Proceedings 95, 21892203.CrossRefGoogle ScholarPubMed
Baker, JM, Al-Nakkash, L and Herbst-Kralovetz, MM (2017) Estrogen–gut microbiome axis: physiological and clinical implications. Maturitas 103, 4553.CrossRefGoogle ScholarPubMed
Kumar, A, et al. (2020) Relevance of SARS-CoV-2 related factors ACE2 and TMPRSS2 expressions in gastrointestinal tissue with pathogenesis of digestive symptoms, diabetes-associated mortality, and disease recurrence in COVID-19 patients. Medical Hypotheses 144, 110271.CrossRefGoogle ScholarPubMed
Supplementary material: PDF

Kumar et al. supplementary material

Kumar et al. supplementary material

Download Kumar et al. supplementary material(PDF)
PDF 178.4 KB