30
views
0
recommends
+1 Recommend
1 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

      Activation of cytotoxic T cell population and inversion of CD4:CD8 ratio as manifestations of cellular immune response in SARS-COV-2 infection

      letter

      Read this article at

      ScienceOpenPublisherPMC
      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          To the Editor, Amid the recent worldwide coronavirus disease 2019 (COVID-19) outbreak caused by the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), there has been increasing interest in the host-pathogen interaction and the resulting immune dysregulation. The role that the innate immune system plays in responding to SARS-CoV-2 is yet to be fully understood. The effect of this virus on the different lymphocyte populations is based on single-case reports and sporadic series [1–5]. Here, we report the findings from our evaluation of the cellular immune response in six patients infected with SARS-CoV-2 from New York City. We used flow cytometry to analyze helper and cytotoxic T cell populations, assess the antibody-secreting cells (ASCs) and examine the expression of activation markers. Briefly, we reviewed flow cytometry studies from six patients who were confirmed positive for the infection by SARS-CoV-2 using nucleic acid testing via RT-PCR of throat swab specimens using RT-PCR. All patients tested positive for SARS-CoV2 within 14 days of the flow cytometry study from our institution, a major tertiary academic center in New York City, which has been a hot spot of the COVID-19 pandemic. Originally, five of the flow cytometry studies were performed as a workup for hematological malignancies. Five peripheral blood and one bone marrow aspirate samples are included. Flow cytometry was performed by five-color analysis on the Navios Flow Cytometer (Beckman Coulter, Miami, FL), and data analysis was performed on the Kaluza Flow Cytometry Analysis Software (Beckman Coulter, Miami, FL). Leukocytes were stained with antibodies against CD3, CD4, CD8, CD19, CD38, and HLA-DR. Fluorochromes included fluorescein isothiocyanate (FITC), PE-cyanine 5 (PC5), PE-cyanine 7 (PC7), PE-Texas Red (ECD), and phycoerythrin (PE). Our initial measurements defined both CD4+ and CD8+ T cell populations. We calculated the CD4:CD8 ratio. Then, we assessed the expression of CD38 and HLA-DR on both populations as surrogate markers of activation. To screen for ASCs, we elected to use a simplified two-antibody gating strategy by evaluating the expression of CD38 on the CD19+ cells taking into account that this phenotype is expressed in most ASCs regardless of their type. Four out of the six patients included in this study showed varying degrees of lymphopenia (Table 1). Four patients showed characteristically low CD4:CD8 ratio; these four patients showed bright expression of CD38 with partial/dim HLA-DR on the CD8+ T cells indicative of cellular activation (Fig. 1). Analysis of the CD19+ cells for CD38 expression showed no increase in the number of ASCs in any of the patients. The remaining two patients with normal CD4:CD8 ratio showed no expression of CD38 or HLA-DR on the T cells and no evidence of increased ASCs. Although five of the cases had a history of hematologic malignancy, only one case (patient 1) was found to have hematologic neoplasm at the time of the study. For this case, the abnormal population was detected by flow cytometry, whereby this patient had a large B-lymphoblast population (first-time diagnostic study). None of the patients was on treatment for malignancy at the time of study. Table 1 Summary of the clinical and laboratory findings of the patients included in the study Patient Source Original diagnosis Lymphocytes count (normal 1.1–4.8 × 103 μL) CD4:CD8 ratio (normal 1.0–4.0) CD38 expression on cytotoxic T cells HLA-DR expression on cytotoxic T cells 1* PB B-ALL 5.3 0.76 + + 2 PB MDS 0.2 4.58 – – 3 PB CNL 0.3 5.82 – – 4 PB AITL 0.3 0.79 + (dim) + (dim) 5 BM PCN 0.9 0.28 + + 6 PB n/a 3.1 0.35 + + *Patient #1 presented with active disease B-ALL B-lymphoblastic leukemia, MDS myelodysplastic syndrome, CNL chronic neutrophilic leukemia, AITL angioimmunoblastic T-cell lymphoma, PCN plasma cell neoplasm, PB peripheral blood, BM bone marrow Fig. 1 a Flow cytometry analysis of the T cell population with gating on the CD3+ CD8+ T cells showing bright expression CD38 with dim/partial expression of HLA-DR. b Mean fluorescence intensity for CD4 and CD8 showing the high number of CD8+ cells and the low number of CD4+ cells In this study, we report some of the characteristics of the cellular immune response to the SARS-CoV-2 virus infection, as seen in six patients in New York City. Our results demonstrate that a subset of SARS-CoV-2-infected patients shows changes in the T cell population, evident by the decrease in the CD4:CD8 ratio and activation of the cytotoxic T cell population in the setting of lymphopenia. The decrease in the CD4:CD8 ratio is likely due to suppression in the CD4+ T cells. The current observation of lymphopenia is in accordance with the findings from previous reports studying SARS-CoV-2 [3, 4]. Lymphopenia is seen in infections by SARS, measles, and H5N1 influenza viruses and in contrast to viruses with lymphocytic proliferative responses like early human immunodeficiency virus (HIV-1), cytomegalovirus (CMV), and Epstein-Barr virus (EBV) [6]. Although the absolute total number of lymphocytes and the absolute lymphocyte subset numbers are decreased, an inverted CD4:CD8 ratio is likely due to the greater reduction of the helper CD4+ T cell population. Moreover, the cytotoxic CD8+ T cell population shows features of activation by expressing CD38 and, to a lesser extent, HLA-DR. These features are identical to what was described in patients with influenza, Ebola, HIV, and CMV infections. The activation of the CD8+ population suggests that similar to the aforementioned viruses, the cytotoxic T cell population probably plays an integral role in the immune response to SARS-CoV-2 infection [1, 3, 4, 7, 8]. We also speculate that the decrease in the CD4:CD8 ratio and the activation of the cytotoxic T cells happen synchronously. While proliferation of ASCs would be expected in SARS-CoV-2 patients, we did not detect such an increase in any of our patients. This could be attributed to the possibility that our results provide a snapshot in the natural history of the disease based on a single reading and we do not exclude the possibility of elevated ASC numbers at different time points. Additionally, a small minority of ASCs may not be detected by the simplified CD38 and CD19 panel that we used. There are several limitations to our study. First, this is a retrospective, single-center study, with a small study sample. Second, the absence of temporal follow-up evaluations restricts understanding the natural history of the disease and the changes seen in the different studied parameters. Lastly, the underlying hematologic malignancy acting as a confounder is possible as many hematologic malignancies affect the T cell population. However, given that only one case had an active neoplastic disease, and the fact that the changes we report here are not common in hematologic malignancies, it is unlikely that this is actually confounding the results. In fact, hematologic malignancies including, acute leukemia and plasma cell dyscrasia, are associated with a heterogeneous T cell reaction characterized by decreased levels of naive CD4+ and CD8+ T cells and increasing the numbers of the so-called CD4+/CD25+ regulatory T cells (Tregs) [9] . Despite these limitations, by using flow cytometry, we have confirmed and elaborated on the T cell compartment alterations found in the setting of SARS-CoV-2 infection [1–4, 10]. In conclusion, we document the presence of a distinctive T cell response in SARS-CoV-2 patients evident by alteration of CD4:CD8 ratio, likely due to suppression of the CD4 population and activation of the cytotoxic CD8+ T cell population. Although these findings should be interpreted in the context of the study limitations, they provide potential immune parameters for further detailed studies to elucidate the immune reaction to SARS-CoV-2 virus.

          Related collections

          Most cited references4

          • Record: found
          • Abstract: found
          • Article: not found

          Pathological findings of COVID-19 associated with acute respiratory distress syndrome

          Since late December, 2019, an outbreak of a novel coronavirus disease (COVID-19; previously known as 2019-nCoV)1, 2 was reported in Wuhan, China, 2 which has subsequently affected 26 countries worldwide. In general, COVID-19 is an acute resolved disease but it can also be deadly, with a 2% case fatality rate. Severe disease onset might result in death due to massive alveolar damage and progressive respiratory failure.2, 3 As of Feb 15, about 66 580 cases have been confirmed and over 1524 deaths. However, no pathology has been reported due to barely accessible autopsy or biopsy.2, 3 Here, we investigated the pathological characteristics of a patient who died from severe infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) by postmortem biopsies. This study is in accordance with regulations issued by the National Health Commission of China and the Helsinki Declaration. Our findings will facilitate understanding of the pathogenesis of COVID-19 and improve clinical strategies against the disease. A 50-year-old man was admitted to a fever clinic on Jan 21, 2020, with symptoms of fever, chills, cough, fatigue and shortness of breath. He reported a travel history to Wuhan Jan 8–12, and that he had initial symptoms of mild chills and dry cough on Jan 14 (day 1 of illness) but did not see a doctor and kept working until Jan 21 (figure 1 ). Chest x-ray showed multiple patchy shadows in both lungs (appendix p 2), and a throat swab sample was taken. On Jan 22 (day 9 of illness), the Beijing Centers for Disease Control (CDC) confirmed by reverse real-time PCR assay that the patient had COVID-19. Figure 1 Timeline of disease course according to days from initial presentation of illness and days from hospital admission, from Jan 8–27, 2020 SARS-CoV-2=severe acute respiratory syndrome coronavirus 2. He was immediately admitted to the isolation ward and received supplemental oxygen through a face mask. He was given interferon alfa-2b (5 million units twice daily, atomisation inhalation) and lopinavir plus ritonavir (500 mg twice daily, orally) as antiviral therapy, and moxifloxacin (0·4 g once daily, intravenously) to prevent secondary infection. Given the serious shortness of breath and hypoxaemia, methylprednisolone (80 mg twice daily, intravenously) was administered to attenuate lung inflammation. Laboratory tests results are listed in the appendix (p 4). After receiving medication, his body temperature reduced from 39·0 to 36·4 °C. However, his cough, dyspnoea, and fatigue did not improve. On day 12 of illness, after initial presentation, chest x-ray showed progressive infiltrate and diffuse gridding shadow in both lungs. He refused ventilator support in the intensive care unit repeatedly because he suffered from claustrophobia; therefore, he received high-flow nasal cannula (HFNC) oxygen therapy (60% concentration, flow rate 40 L/min). On day 13 of illness, the patient's symptoms had still not improved, but oxygen saturation remained above 95%. In the afternoon of day 14 of illness, his hypoxaemia and shortness of breath worsened. Despite receiving HFNC oxygen therapy (100% concentration, flow rate 40 L/min), oxygen saturation values decreased to 60%, and the patient had sudden cardiac arrest. He was immediately given invasive ventilation, chest compression, and adrenaline injection. Unfortunately, the rescue was not successful, and he died at 18:31 (Beijing time). Biopsy samples were taken from lung, liver, and heart tissue of the patient. Histological examination showed bilateral diffuse alveolar damage with cellular fibromyxoid exudates (figure 2A, B ). The right lung showed evident desquamation of pneumocytes and hyaline membrane formation, indicating acute respiratory distress syndrome (ARDS; figure 2A). The left lung tissue displayed pulmonary oedema with hyaline membrane formation, suggestive of early-phase ARDS (figure 2B). Interstitial mononuclear inflammatory infiltrates, dominated by lymphocytes, were seen in both lungs. Multinucleated syncytial cells with atypical enlarged pneumocytes characterised by large nuclei, amphophilic granular cytoplasm, and prominent nucleoli were identified in the intra-alveolar spaces, showing viral cytopathic-like changes. No obvious intranuclear or intracytoplasmic viral inclusions were identified. Figure 2 Pathological manifestations of right (A) and left (B) lung tissue, liver tissue (C), and heart tissue (D) in a patient with severe pneumonia caused by SARS-CoV-2 SARS-CoV-2=severe acute respiratory syndrome coronavirus 2. The pathological features of COVID-19 greatly resemble those seen in SARS and Middle Eastern respiratory syndrome (MERS) coronavirus infection.4, 5 In addition, the liver biopsy specimens of the patient with COVID-19 showed moderate microvesicular steatosis and mild lobular and portal activity (figure 2C), indicating the injury could have been caused by either SARS-CoV-2 infection or drug-induced liver injury. There were a few interstitial mononuclear inflammatory infiltrates, but no other substantial damage in the heart tissue (figure 2D). Peripheral blood was prepared for flow cytometric analysis. We found that the counts of peripheral CD4 and CD8 T cells were substantially reduced, while their status was hyperactivated, as evidenced by the high proportions of HLA-DR (CD4 3·47%) and CD38 (CD8 39·4%) double-positive fractions (appendix p 3). Moreover, there was an increased concentration of highly proinflammatory CCR6+ Th17 in CD4 T cells (appendix p 3). Additionally, CD8 T cells were found to harbour high concentrations of cytotoxic granules, in which 31·6% cells were perforin positive, 64·2% cells were granulysin positive, and 30·5% cells were granulysin and perforin double-positive (appendix p 3). Our results imply that overactivation of T cells, manifested by increase of Th17 and high cytotoxicity of CD8 T cells, accounts for, in part, the severe immune injury in this patient. X-ray images showed rapid progression of pneumonia and some differences between the left and right lung. In addition, the liver tissue showed moderate microvesicular steatosis and mild lobular activity, but there was no conclusive evidence to support SARS-CoV-2 infection or drug-induced liver injury as the cause. There were no obvious histological changes seen in heart tissue, suggesting that SARS-CoV-2 infection might not directly impair the heart. Although corticosteroid treatment is not routinely recommended to be used for SARS-CoV-2 pneumonia, 1 according to our pathological findings of pulmonary oedema and hyaline membrane formation, timely and appropriate use of corticosteroids together with ventilator support should be considered for the severe patients to prevent ARDS development. Lymphopenia is a common feature in the patients with COVID-19 and might be a critical factor associated with disease severity and mortality. 3 Our clinical and pathological findings in this severe case of COVID-19 can not only help to identify a cause of death, but also provide new insights into the pathogenesis of SARS-CoV-2-related pneumonia, which might help physicians to formulate a timely therapeutic strategy for similar severe patients and reduce mortality. This online publication has been corrected. The corrected version first appeared at thelancet.com/respiratory on February 25, 2020
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Breadth of concomitant immune responses prior to patient recovery: a case report of non-severe COVID-19

            To the Editor — We report the kinetics of immune responses in relation to clinical and virological features of a patient with mild-to-moderate coronavirus disease 2019 (COVID-19) that required hospitalization. Increased antibody-secreting cells (ASCs), follicular helper T cells (TFH cells), activated CD4+ T cells and CD8+ T cells and immunoglobulin M (IgM) and IgG antibodies that bound the COVID-19-causing coronavirus SARS-CoV-2 were detected in blood before symptomatic recovery. These immunological changes persisted for at least 7 d following full resolution of symptoms. A 47-year-old woman from Wuhan, Hubei province, China, presented to an emergency department in Melbourne, Australia. Her symptoms commenced 4 d earlier with lethargy, sore throat, dry cough, pleuritic chest pain, mild dyspnea and subjective fevers (Fig. 1a). She traveled from Wuhan to Australia 11 d before presentation. She had no contact with the Huanan seafood market or with known COVID-19 cases. She was otherwise healthy and was a non-smoker taking no medications. Clinical examination revealed a temperature of 38.5 °C, a pulse rate of 120 beats per minute, a blood pressure of 140/80 mm Hg, a respiratory rate of 22 breaths per minute, and oxygen saturation 98% while breathing ambient air. Lung auscultation revealed bi-basal rhonchi. At presentation on day 4, SARS-CoV-2 was detected in a nasopharyngeal swab specimen by real-time reverse-transcriptase PCR. SARS-CoV-2 was again detected at days 5–6 in nasopharyngeal, sputum and fecal samples, but was undetectable from day 7 (Fig. 1a). Blood C-reactive protein was elevated at 83.2, with normal counts of lymphocytes (4.3 × 109 cells per liter (range, 4.0 × 109 to 12.0 × 109 cells per liter)) and neutrophils (6.3 × 109 cells per liter (range, 2.0 × 109 to 8.0 × 109 × 109 cells per liter)). No other respiratory pathogens were detected. Her management was intravenous fluid rehydration without supplemental oxygenation. No antibiotics, steroids or antiviral agents were administered. Chest radiography demonstrated bi-basal infiltrates at day 5 that cleared on day 10 (Fig. 1b). She was discharged to home isolation on day 11. Her symptoms resolved completely by day 13, and she remained well at day 20, with progressive increases in plasma SARS-CoV-2-binding IgM and IgG antibodies from day 7 until day 20 (Fig. 1c and Extended Data Fig. 1). The patient was enrolled through the Sentinel Travelers Research Preparedness Platform for Emerging Infectious Diseases novel coronavirus substudy (SETREP-ID-coV) and provided written informed consent before the study. Patient care and research were conducted in compliance with the Case Report guidelines and the Declaration of Helsinki. Experiments were performed with ethics approvals HREC/17/MH/53, HREC/15/MonH/64/2016.196 and UoM#1442952.1/#1443389.4. Fig. 1 Emergence of immune responses during non-severe symptomatic COVID-19. a, Timeline of COVID-19, showing detection of SARS-CoV-2 in sputum, nasopharyngeal aspirates and feces but not urine, rectal swab or whole blood. SARS-CoV-2 was quantified by rRT-PCR; cycle threshold (Ct) is shown. A higher Ct value means lower viral load. Dashed horizontal line indicates limit of detection (LOD) threshold (Ct = 45). Open circles, undetectable SARS-CoV-2. b, Anteroposterior chest radiographs on days 5 and 10 following symptom onset, showing radiological improvement from hospital admission to discharge. c, Immunofluorescence antibody staining, repeated twice in duplicate, for detection of IgG and IgM bound to SARS-CoV-2-infected Vero cells, assessed with plasma (diluted 1:20) obtained at days 7–9 and 20 following symptom onset. d–f, Frequency (left set of plots) of CD27hiCD38hi ASCs (gated on CD3–CD19+ lymphocytes) and activated ICOS+PD-1+ TFH cells (gated on CD4+CXCR5+ lymphocytes) (d), activated CD38+HLA-DR+ CD8+ or CD4+ T cells (e), and CD14+CD16+ monocytes and activated HLA-DR+ natural killer (NK) cells (gated on CD3–CD14–CD56+ cells) (f), detected by flow cytometry of blood collected at days 7–9 and 20 following symptom onset in the patient and in healthy donors (n = 5; median with interquartile range); gating examples at right. Bottom right histograms and line graphs, staining of granzyme A (GZMA (A)), granzyme B (GZMB (B)), granzyme K (GZMK (K)), granzyme M (GZMM (M)) and perforin (Prf) in parent CD8+ and CD4+ T cells and activated CD38+HLA-DR+ CD8+ and CD4+ T cells. Gating and experimental details are in Extended Data Fig. 3. Source data We analyzed the kinetics and breadth of immune responses associated with clinical resolution of COVID-19. As ASCs are key for the rapid production of antibodies following infection with Ebola virus 1,2 and infection with and vaccination against influenza virus 2,3 , and activated circulating TFH cells (cTFH cells) are concomitantly induced following vaccination against influenza virus 3 , we defined the frequency of CD3–CD19+CD27hiCD38hi ASC and CD4+CXCR5+ICOS+PD-1+ cTFH cell responses before symptomatic recovery. ASCs appeared in the blood at the time of viral clearance (day 7; 1.48%) and peaked on day 8 (6.91%). The emergence of cTFH cells occurred concurrently in blood at day 7 (1.98%), increasing on day 8 (3.25%) and day 9 (4.46%) (Fig. 1d). The peak of both ASCs and cTFH cells was markedly higher in the patient with COVID-19 than in healthy control participants (0.61% ± 0.40% and 1.83% ± 0.77%, respectively (average ± s.d.); n = 5). Both ASCs and cTFH cells were prominently present during convalescence (day 20) (4.54% and 7.14%, respectively; Fig. 1d). Thus, our study provides evidence on the recruitment of both ASCs and cTFH cells in this patient’s blood while she was still unwell and 3 d before the resolution of symptoms. Since co-expression of CD38 and HLA-DR is the key phenotype of the activation of CD8+ T cells in response to viral infections, we analyzed co-expression of CD38 and HLA-DR. As per reports for Ebola and influenza 1,4 , co-expression of CD38 and HLA-DR on CD8+ T cells (assessed as the frequency of CD38+HLA-DR+ CD8+ T cells) rapidly increased in this patient from day 7 (3.57%) to day 8 (5.32%) and day 9 (11.8%), then decreased at day 20 (7.05%) (Fig. 1e). Furthermore, the frequency of CD38+HLA-DR+ CD8+ T cells was much higher in this patient than in healthy individuals (1.47% ± 0.50%; n = 5). CD38+HLA-DR+ T cells were also recently documented in a patient with COVID-19 at one time point 5 . Similarly, co-expression of CD38 and HLA-DR on CD4+ T cells (assessed as the frequency of CD38+HLA-DR+ CD4+ T cells) increased between day 7 (0.55%) and day 9 (3.33%) in this patient, relative to that of healthy donors (0.63% ± 0.28%; n = 5), although at lower levels than that of CD8+ T cells. CD38+HLA-DR+ T cells, especially CD8+ T cells, produced larger amounts of granzymes A and B and perforin (~34–54% higher) than did their parent cells (CD8+ or CD4+ populations; Fig. 1e). Thus, the emergence and rapid increase in activated CD38+HLA-DR+ T cells, especially CD8+ T cells, at days 7–9 preceded the resolution of symptoms. Details on data reproducibility are in the Life Sciences Reporting Summary. Analysis of CD16+CD14+ monocytes, which are related to immunopathology, showed lower frequencies of CD16+CD14+ monocytes in the blood of this patient at days 7, 8 and 9 (1.29%, 0.43% and 1.47%, respectively) than in that of healthy control donors (9.03% ± 4.39%; n = 5) (Fig. 1f), possibly indicative of the efflux of CD16+CD14+ monocytes from the blood to the site of infection. No differences in activated HLA-DR+CD3–CD56+ natural killer cells were found. As pro-inflammatory cytokines and chemokines are predictive of severe clinical outcomes for influenza 6 , we quantified 17 pro-inflammatory cytokines and chemokines in plasma. We found low levels of the chemokine MCP-1 (CCL2) in the patient’s plasma (Extended Data Fig. 2a), although this was comparable to results obtained for healthy donors (22.15 ± 13.81; n = 5), patients infected with influenza A virus or influenza B, assessed at days 7–9 (33.85 ± 30.12; n = 5), and a patient infected with the human coronavirus HCoV-229e (40.56). Thus, in contrast to severe avian H7N9 disease, which had elevated cytokines IL-6, IL-8, IL-10, MIP-1β and IFN-γ 6 , minimal pro-inflammatory cytokines and chemokines were found in this patient with COVID-19, even while she was symptomatic at days 7–9. As the single-nucleotide polymorphism rs12252-C/C in the gene IFITM3 (which encodes interferon-induced transmembrane protein 3) is linked to severe influenza 6,7 , we analyzed IFITM3-rs12252 in the patient with COVID-19 and found the ‘risk’ IFITM3-rs12252-C/C variant (Extended Data Fig. 2b). As the prevalence of IFITM3-rs12252-C/C in the Chinese population is 26.5% (the 1000 Genomes Project) 6 , further investigation of the IFITM3-rs12252-C/C allele in larger cohorts of people with COVID-19 is worth pursuing. Collectively, our study provides novel contributions to the understanding of the breadth and kinetics of immune responses during a non-severe case of COVID-19. This patient did not experience complications of respiratory failure or acute respiratory distress syndrome, did not require supplemental oxygenation, and was discharged within a week of hospitalization, consistent with non-severe but symptomatic disease. We have provided evidence on the recruitment of immune cell populations (ASCs, TFH cells and activated CD4+ and CD8+ T cells), together with IgM and IgG SARS-CoV-2-binding antibodies, in the patient’s blood before the resolution of symptoms. We propose that these immune parameters should be characterized in larger cohorts of people with COVID-19 with different disease severities to determine whether they could be used to predict disease outcome and evaluate new interventions that might minimize severity and/or to inform protective vaccine candidates. Furthermore, our study indicates that robust multi-factorial immune responses can be elicited to the newly emerged virus SARS-CoV-2 and, similar to the avian H7N9 disease 8 , early adaptive immune responses might correlate with better clinical outcomes. Reporting Summary Further information on research design is available in the Nature Research Reporting Summary linked to this article. Online content Any methods, additional references, Nature Research reporting summaries, source data, extended data, supplementary information, acknowledgements, peer review information; details of author contributions and competing interests; and statements of data and code availability are available at 10.1038/s41591-020-0819-2. Supplementary information Reporting Summary
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Increased expression of CD8 marker on T-cells in COVID-19 patients

              Background Cell-mediated immunity including T-cells (T helper and cytotoxic) plays an essential role in efficient antiviral responses against coronavirus disease-2019 (COVID-19). Therefore, in this study, we evaluated the ratio and expression of CD4 and CD8 markers in COVID-19 patients to clarify the immune characterizations of CD4 and CD8 T-cells in COVID-19 patients. Methods Peripheral blood samples of 25 COVID-19 patients and 25 normal individuals with similar age and sex as the control group were collected. White blood cells, platelets, and lymphocytes were counted and CD4 and CD8 T lymphocytes were evaluated by flow cytometry. Results The number of white blood cells, lymphocytes, and platelets were reduced significantly in COVID-19 patients (P   0.05); however, the CD8 MFI increased significantly in COVID-19 infected patients (P < 0.05). Conclusion Although, there is no significant difference in the ratio of CD4 to CD8 between two groups, the expression level of CD8 in COVID-19 patients was significantly higher than the normal individuals. This result suggested that the cellular immune responses triggered by COVID-19 infection were developed through overexpression of CD8 and hyperactivation of cytotoxic T lymphocytes.
                Bookmark

                Author and article information

                Contributors
                siraj.eljamal@mssm.edu
                Journal
                J Hematop
                J Hematop
                Journal of Hematopathology
                Springer Berlin Heidelberg (Berlin/Heidelberg )
                1868-9256
                1865-5785
                2 July 2020
                : 1-3
                Affiliations
                [1 ]GRID grid.59734.3c, ISNI 0000 0001 0670 2351, Department of Pathology, Molecular and Cell Based Medicine, , Icahn School of Medicine at Mount Sinai, ; 1 Gustave L. Levy Pl, New York, NY 10029 USA
                [2 ]GRID grid.59734.3c, ISNI 0000 0001 0670 2351, Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, , Tisch Cancer Institute, ; New York, NY USA
                [3 ]GRID grid.416167.3, Department of Genetics and Genomic Sciences, , Hasso Plattner Institute for Digital Health at Mount Sinai , ; New York, USA
                [4 ]GRID grid.59734.3c, ISNI 0000 0001 0670 2351, Department of Nephrology, , Icahn School of Medicine at Mount Sinai, ; New York, NY USA
                Author information
                http://orcid.org/0000-0001-6299-1071
                Article
                405
                10.1007/s12308-020-00405-9
                7332243
                f7d1a435-27ec-4ad8-899d-dd9cc92a5174
                © Springer-Verlag GmbH Germany, part of Springer Nature 2020

                This article is made available via the PMC Open Access Subset for unrestricted research re-use and secondary analysis in any form or by any means with acknowledgement of the original source. These permissions are granted for the duration of the World Health Organization (WHO) declaration of COVID-19 as a global pandemic.

                History
                : 5 June 2020
                : 23 June 2020
                Categories
                Letter to the Editor

                Pathology
                Pathology

                Comments

                Comment on this article