A dynamic mucin mRNA signature associates with COVID-19 disease presentation and severity

BACKGROUND SARS-CoV-2 infection induces mucin overexpression, further promoting disease. Given that mucins are critical components of innate immunity, unraveling their expression profiles that dictate the course of disease could greatly enhance our understanding and management of COVID-19. METHODS Using validated RT-PCR assays, we assessed mucin mRNA expression in the blood of patients with symptomatic COVID-19 compared with symptomatic patients without COVID-19 and healthy controls and correlated the data with clinical outcome parameters. Additionally, we analyzed mucin expression in mucus and lung tissue from patients with COVID-19 and investigated the effect of drugs for COVID-19 treatment on SARS-CoV-2–induced mucin expression in pulmonary epithelial cells. RESULTS We identified a dynamic blood mucin mRNA signature that clearly distinguished patients with symptomatic COVID-19 from patients without COVID-19 based on expression of MUC1, MUC2, MUC4, MUC6, MUC13, MUC16, and MUC20 (AUCROC of 91.8%; sensitivity and specificity of 90.6% and 93.3%, respectively) and that discriminated between mild and critical COVID-19 based on the expression of MUC16, MUC20, and MUC21 (AUCROC of 89.1%; sensitivity and specificity of 90.0% and 85.7%, respectively). Differences in the transcriptional landscape of mucins in critical cases compared with mild cases identified associations with COVID-19 symptoms, respiratory support, organ failure, secondary infections, and mortality. Furthermore, we identified different mucins in the mucus and lung tissue of critically ill COVID-19 patients and showed the ability of baricitinib, tocilizumab, favipiravir, and remdesivir to suppress expression of SARS-CoV-2–induced mucins. CONCLUSION This multifaceted blood mucin mRNA signature showed the potential role of mucin profiling in diagnosing, estimating severity, and guiding treatment options in patients with COVID-19. FUNDING The Antwerp University Research and the Research Foundation Flanders COVID-19 funds.

[1]  D. Bennett,et al.  Serial KL-6 measurements in COVID-19 patients , 2021, Internal and Emergency Medicine.

[2]  P. Brodin Immune determinants of COVID-19 disease presentation and severity , 2021, Nature Medicine.

[3]  P. Mukherjee,et al.  Mucin signature as a potential tool to predict susceptibility to COVID‐19 , 2020, Physiological reports.

[4]  S. Cosgrove,et al.  Prevalence of Co-infection at the Time of Hospital Admission in COVID-19 Patients, A Multicenter Study , 2020, Open forum infectious diseases.

[5]  Cameron R. Wolfe,et al.  Baricitinib plus Remdesivir for Hospitalized Adults with Covid-19 , 2020, The New England journal of medicine.

[6]  P. Verweij,et al.  Fungal infections should be part of the core outcome set for COVID-19 , 2020, The Lancet Infectious Diseases.

[7]  Y. Dou,et al.  Transcriptomic signatures and repurposing drugs for COVID-19 patients: findings of bioinformatics analyses , 2020, Computational and Structural Biotechnology Journal.

[8]  A. Farcomeni,et al.  JAK inhibition reduces SARS-CoV-2 liver infectivity and modulates inflammatory responses to reduce morbidity and mortality , 2020, Science Advances.

[9]  C. Qin,et al.  Mucus production stimulated by IFN-AhR signaling triggers hypoxia of COVID-19 , 2020, Cell Research.

[10]  Eric A. Meyerowitz,et al.  Efficacy of Tocilizumab in Patients Hospitalized with Covid-19 , 2020, The New England journal of medicine.

[11]  B. Weynand,et al.  Favipiravir at high doses has potent antiviral activity in SARS-CoV-2−infected hamsters, whereas hydroxychloroquine lacks activity , 2020, Proceedings of the National Academy of Sciences.

[12]  S. Farhadian,et al.  Sex differences in immune responses that underlie COVID-19 disease outcomes , 2020, Nature.

[13]  I. Bassett,et al.  Pediatric Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2): Clinical Presentation, Infectivity, and Immune Responses , 2020, The Journal of Pediatrics.

[14]  J. Mason,et al.  A dynamic COVID-19 immune signature includes associations with poor prognosis , 2020, Nature Medicine.

[15]  Fei Liu,et al.  Elevated MUC1 and MUC5AC mucin protein levels in airway mucus of critical ill COVID‐19 patients , 2020, Journal of medical virology.

[16]  Shu-Yan Liu,et al.  Mucin mutations and aberrant expression are associated with the pathogenesis of immune thrombocytopenia. , 2020, Thrombosis research.

[17]  Xi He,et al.  Single-cell analysis reveals bronchoalveolar epithelial dysfunction in COVID-19 patients , 2020, Protein & Cell.

[18]  C. von Kalle,et al.  COVID-19 severity correlates with airway epithelium–immune cell interactions identified by single-cell analysis , 2020, Nature Biotechnology.

[19]  Adam C. Labonte,et al.  Comprehensive transcriptomic analysis of COVID-19 blood, lung, and airway , 2020, Scientific Reports.

[20]  S. Rose-John,et al.  Tocilizumab does not block interleukin-6 (IL-6) signaling in murine cells , 2020, PloS one.

[21]  S. Ruan Likelihood of survival of coronavirus disease 2019 , 2020, The Lancet Infectious Diseases.

[22]  Xavier Duval,et al.  Clinical and virological data of the first cases of COVID-19 in Europe: a case series , 2020, The Lancet Infectious Diseases.

[23]  W. Triampo,et al.  The Blood Circulating Rare Cell Population. What Is It and What Is It Good for? , 2020, Cells.

[24]  G. Herrler,et al.  SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor , 2020, Cell.

[25]  Wu Zhong,et al.  Remdesivir and chloroquine effectively inhibit the recently emerged novel coronavirus (2019-nCoV) in vitro , 2020, Cell Research.

[26]  T. Breugelmans,et al.  In depth study of transmembrane mucins in association with intestinal barrier dysfunction during the course of T cell transfer and DSS-induced colitis. , 2020, Journal of Crohn's & colitis.

[27]  Y. Hu,et al.  Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China , 2020, The Lancet.

[28]  E. Mao,et al.  Paclitaxel alleviated sepsis-induced acute lung injury by activating MUC1 and suppressing TLR-4/NF-κB pathway , 2019, Drug design, development and therapy.

[29]  Yoshiya Tanaka,et al.  Janus Kinase Inhibitor Baricitinib Modulates Human Innate and Adaptive Immune System , 2018, Front. Immunol..

[30]  Markus M. Rinschen,et al.  Autosomal Tubulointerstitial Kidney Disease—MUC1 Type: Differential Proteomics Suggests that Mutated MUC1 (insC) Affects Vesicular Transport in Renal Epithelial Cells , 2018, Proteomics.

[31]  E. Lillehoj,et al.  MUC1: The First Respiratory Mucin with an Anti-Inflammatory Function , 2017, Journal of clinical medicine.

[32]  N. Udagawa,et al.  A Jak1/2 inhibitor, baricitinib, inhibits osteoclastogenesis by suppressing RANKL expression in osteoblasts in vitro , 2017, PloS one.

[33]  C. Liu,et al.  Gastric De Novo Muc13 Expression and Spasmolytic Polypeptide-Expressing Metaplasia during Helicobacter heilmannii Infection , 2014, Infection and Immunity.

[34]  E. Hachulla,et al.  Effect of In Vitro and In Vivo Anakinra on Cytokines Production in Schnitzler Syndrome , 2013, PloS one.

[35]  S. Pereson,et al.  Cellular ageing, increased mortality and FTLD‐TDP‐associated neuropathology in progranulin knockout mice , 2012, The Journal of pathology.

[36]  K. Pantel,et al.  Circulating epithelial cells in patients with benign colon diseases. , 2012, Clinical chemistry.

[37]  D. Kufe,et al.  Mucins in cancer: function, prognosis and therapy , 2009, Nature Reviews Cancer.

[38]  Mark M Davis,et al.  Isolating highly enriched populations of circulating epithelial cells and other rare cells from blood using a magnetic sweeper device , 2009, Proceedings of the National Academy of Sciences.

[39]  P. Friedman,et al.  Low-level expression of HER2 and CK19 in normal peripheral blood mononuclear cells: relevance for detection of circulating tumor cells , 2008, Journal of hematology & oncology.

[40]  N. Kohno,et al.  Circulating KL‐6/MUC1 as an independent predictor for disseminated intravascular coagulation in acute respiratory distress syndrome , 2008, Journal of internal medicine.

[41]  V. Korolik,et al.  Mucins in the mucosal barrier to infection , 2008, Mucosal Immunology.

[42]  S. Pocock,et al.  The Strengthening the Reporting of Observational Studies in Epidemiology (STROBE) statement: guidelines for reporting observational studies. , 2007, Bulletin of the World Health Organization.

[43]  S. Pocock,et al.  Strengthening the reporting of observational studies in epidemiology (STROBE) statement: guidelines for reporting observational studies , 2007, BMJ : British Medical Journal.

[44]  Gerd Schmitz,et al.  Aberrant intestinal expression and allelic variants of mucin genes associated with inflammatory bowel disease , 2006, Journal of Molecular Medicine.

[45]  Stephen Tomlinson,et al.  The effect of dexamethasone on human mucin 1 expression and antibody‐dependent complement sensitivity in a prostate cancer cell line in vitro and in vivo , 2004, Immunology.

[46]  J. Taylor‐Papadimitriou,et al.  Form and pattern of MUC1 expression on T cells activated in vivo or in vitro suggests a function in T‐cell migration , 2003, Immunology.

[47]  G. Sutherland,et al.  MUC13, a Novel Human Cell Surface Mucin Expressed by Epithelial and Hemopoietic Cells* , 2001, The Journal of Biological Chemistry.

[48]  J. Vincent,et al.  The SOFA (Sepsis-related Organ Failure Assessment) score to describe organ dysfunction/failure , 1996, Intensive Care Medicine.

[49]  Arthur S Slutsky,et al.  Acute Respiratory Distress Syndrome The Berlin Definition , 2012 .

[50]  M. Conrad,et al.  A role for mucin in the absorption of inorganic iron and other metal cations. A study in rats. , 1991, Gastroenterology.