Lasting Changes to Circulating Leukocytes in People with Mild SARS-CoV-2 Infections

Survivors of severe SARS-CoV-2 infections frequently suffer from a range of post-infection sequelae. Whether survivors of mild or asymptomatic infections can expect any long-term health consequences is not yet known. Herein we investigated lasting changes to soluble inflammatory factors and cellular immune phenotype and function in individuals who had recovered from mild SARS-CoV-2 infections (n=22) compared to those that had recovered from other mild respiratory infections (n=11). Individuals who had mild SARS-CoV-2 infections had elevated levels of C-reactive protein 1-3 months after symptom onset, and changes in phenotype and function of circulating T cells that were not apparent in individuals 6-9 months post-symptom onset. Markers of monocyte activation and expression of adherence and chemokine receptors indicative of altered migratory capacity were also higher at 1-3 months post-infection in individuals who had mild SARS-CoV-2, but these were no longer elevated by 6-9 months post-infection. Perhaps most surprisingly, polyclonal activation of T cells was higher in individuals who had recently experienced a mild SARS-CoV-2 infection compared to individuals with other recent respiratory infections. These data are indicative of prolonged immune activation and systemic inflammation that persists for up to three months after mild or asymptomatic SARS-CoV-2 infections.

[1]  Konrad U. Förstner,et al.  6th European Congress of Immunology, 1-4 September 2021, Virtual meeting. , 2021, European journal of immunology.

[2]  X. Mariette,et al.  Systemic and organ-specific immune-related manifestations of COVID-19 , 2021, Nature Reviews Rheumatology.

[3]  A. Banerjee,et al.  Post-covid syndrome in individuals admitted to hospital with covid-19: retrospective cohort study , 2021, BMJ.

[4]  E. Poveda,et al.  Dendritic cell deficiencies persist seven months after SARS-CoV-2 infection , 2021, Cellular & Molecular Immunology.

[5]  Mario Sansone,et al.  Cytokine signature and COVID-19 prediction models in the two waves of pandemics , 2021, Scientific Reports.

[6]  Benjamin Bowe,et al.  High-dimensional characterization of post-acute sequelae of COVID-19 , 2021, Nature.

[7]  A. Banerjee,et al.  Epidemiology of post-COVID syndrome following hospitalisation with coronavirus: a retrospective cohort study , 2021, medRxiv.

[8]  J. Donnelly,et al.  Readmission and Death After Initial Hospital Discharge Among Patients With COVID-19 in a Large Multihospital System. , 2020, JAMA.

[9]  R. Alon,et al.  Leukocyte trafficking to the lungs and beyond: lessons from influenza for COVID-19 , 2020, Nature reviews. Immunology.

[10]  G. Matarese,et al.  T Cells: Warriors of SARS-CoV-2 Infection , 2020, Trends in Immunology.

[11]  V. Chopra,et al.  Sixty-Day Outcomes Among Patients Hospitalized With COVID-19 , 2020, Annals of Internal Medicine.

[12]  M. Honigsbaum,et al.  Taking pandemic sequelae seriously: from the Russian influenza to COVID-19 long-haulers , 2020, The Lancet.

[13]  Steven M. Holland,et al.  Autoantibodies against type I IFNs in patients with life-threatening COVID-19 , 2020, Science.

[14]  J. Greenbaum,et al.  Antigen-Specific Adaptive Immunity to SARS-CoV-2 in Acute COVID-19 and Associations with Age and Disease Severity , 2020, Cell.

[15]  Matthew S. Miller,et al.  Characteristics of Anti-SARS-CoV-2 Antibodies in Recovered COVID-19 Subjects , 2020, Viruses.

[16]  T. Greenhalgh,et al.  Management of post-acute covid-19 in primary care , 2020, BMJ.

[17]  H. Prescott,et al.  Recovery From Severe COVID-19: Leveraging the Lessons of Survival From Sepsis. , 2020, JAMA.

[18]  S. Mallal,et al.  Selective and cross-reactive SARS-CoV-2 T cell epitopes in unexposed humans , 2020, Science.

[19]  Zeyu Chen,et al.  T cell responses in patients with COVID-19 , 2020, Nature Reviews Immunology.

[20]  Elisabeth Mahase Covid-19: What do we know about “long covid”? , 2020, BMJ.

[21]  M. Evert,et al.  Coronavirus disease 2019 induces multi‐lineage, morphologic changes in peripheral blood cells , 2020, EJHaem.

[22]  Morten Nielsen,et al.  Robust T Cell Immunity in Convalescent Individuals with Asymptomatic or Mild COVID-19 , 2020, Cell.

[23]  M. Mann,et al.  Multilevel proteomics reveals host perturbations by SARS-CoV-2 and SARS-CoV , 2020, Nature.

[24]  R. Pranata,et al.  Lymphopenia in severe coronavirus disease-2019 (COVID-19): systematic review and meta-analysis , 2020, Journal of Intensive Care.

[25]  C. Blish,et al.  The Innate Immune System: Fighting on the Front Lines or Fanning the Flames of COVID-19? , 2020, Cell Host & Microbe.

[26]  Angelo Mazza,et al.  An outbreak of severe Kawasaki-like disease at the Italian epicentre of the SARS-CoV-2 epidemic: an observational cohort study , 2020, The Lancet.

[27]  Xiaohu Zheng,et al.  Effective treatment of severe COVID-19 patients with tocilizumab , 2020, Proceedings of the National Academy of Sciences.

[28]  Hong-juan Liu,et al.  COVID-19 infection induces readily detectable morphological and inflammation-related phenotypic changes in peripheral blood monocytes, the severity of which correlate with patient outcome , 2020, medRxiv.

[29]  Yu Cui,et al.  TGF-β signaling controls Foxp3 methylation and T reg cell differentiation by modulating Uhrf1 activity , 2019, The Journal of experimental medicine.

[30]  C. Verschoor,et al.  A comprehensive assessment of immunophenotyping performed in cryopreserved peripheral whole blood , 2018, Cytometry. Part B, Clinical cytometry.

[31]  S. Karampatos,et al.  Monocyte activation is elevated in women with knee-osteoarthritis and associated with inflammation, BMI and pain. , 2017, Osteoarthritis and cartilage.

[32]  C. Hedrick,et al.  Nonclassical patrolling monocyte function in the vasculature. , 2015, Arteriosclerosis, thrombosis, and vascular biology.

[33]  Smita Y. Patel,et al.  Establishment of a healthy human range for the whole blood “OX40” assay for the detection of antigen‐specific CD4+ T cells by flow cytometry , 2014, Cytometry. Part B, Clinical cytometry.

[34]  J. Ananworanich,et al.  Human antigen‐specific CD4+CD25+CD134+CD39+ T cells are enriched for regulatory T cells and comprise a substantial proportion of recall responses , 2014, European journal of immunology.

[35]  Xiaopei Huang,et al.  The Development and Function of Memory Regulatory T Cells after Acute Viral Infections , 2012, The Journal of Immunology.

[36]  O. Rotzschke,et al.  Influenza A Virus Infection Results in a Robust, Antigen-Responsive, and Widely Disseminated Foxp3+ Regulatory T Cell Response , 2011, Journal of Virology.

[37]  Rachel E. Owen,et al.  Tregs control the development of symptomatic West Nile virus infection in humans and mice. , 2009, The Journal of clinical investigation.

[38]  D. Cooper,et al.  High Levels of Human Antigen-Specific CD4+ T Cells in Peripheral Blood Revealed by Stimulated Coexpression of CD25 and CD134 (OX40)1 , 2009, The Journal of Immunology.

[39]  A. Cumano,et al.  Monitoring of Blood Vessels and Tissues by a Population of Monocytes with Patrolling Behavior , 2007, Science.

[40]  P. Greenberg,et al.  Activation-induced expression of CD137 permits detection, isolation, and expansion of the full repertoire of CD8+ T cells responding to antigen without requiring knowledge of epitope specificities. , 2007, Blood.

[41]  C. Geula,et al.  Ccr2 deficiency impairs microglial accumulation and accelerates progression of Alzheimer-like disease , 2007, Nature Medicine.

[42]  T. Springer,et al.  Stimulated mobilization of monocyte Mac-1 and p150,95 adhesion proteins from an intracellular vesicular compartment to the cell surface. , 1987, The Journal of clinical investigation.

[43]  Qin Ning,et al.  Clinical and immunological features of severe and moderate coronavirus disease 2019 , 2020 .

[44]  P. Kubes,et al.  Monocyte Conversion During Inflammation and Injury. , 2017, Arteriosclerosis, thrombosis, and vascular biology.