Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2022 Mar 11;7(1):83.
doi: 10.1038/s41392-022-00919-x.

ACE2-independent infection of T lymphocytes by SARS-CoV-2

Affiliations

ACE2-independent infection of T lymphocytes by SARS-CoV-2

Xu-Rui Shen et al. Signal Transduct Target Ther. .

Abstract

SARS-CoV-2 induced marked lymphopenia in severe patients with COVID-19. However, whether lymphocytes are targets of viral infection is yet to be determined, although SARS-CoV-2 RNA or antigen has been identified in T cells from patients. Here, we confirmed that SARS-CoV-2 viral antigen could be detected in patient peripheral blood cells (PBCs) or postmortem lung T cells, and the infectious virus could also be detected from viral antigen-positive PBCs. We next prove that SARS-CoV-2 infects T lymphocytes, preferably activated CD4 + T cells in vitro. Upon infection, viral RNA, subgenomic RNA, viral protein or viral particle can be detected in the T cells. Furthermore, we show that the infection is spike-ACE2/TMPRSS2-independent through using ACE2 knockdown or receptor blocking experiments. Next, we demonstrate that viral antigen-positive T cells from patient undergone pronounced apoptosis. In vitro infection of T cells induced cell death that is likely in mitochondria ROS-HIF-1a-dependent pathways. Finally, we demonstrated that LFA-1, the protein exclusively expresses in multiple leukocytes, is more likely the entry molecule that mediated SARS-CoV-2 infection in T cells, compared to a list of other known receptors. Collectively, this work confirmed a SARS-CoV-2 infection of T cells, in a spike-ACE2-independent manner, which shed novel insights into the underlying mechanisms of SARS-CoV-2-induced lymphopenia in COVID-19 patients.

PubMed Disclaimer

Conflict of interest statement

The authors declare no competing interests.

Figures

Fig. 1
Fig. 1
Peripheral blood lymphocytes are infected by SARS-CoV-2 in COVID-19 patients. a Percentage of different types of lymphocytes in the healthy donors (n = 15) or in COVID-19 patients (n = 22). (b) Percentage of CD4 + and CD8 + T lymphocytes in healthy donor (n = 8) or in COVID-19 patients (n = 9). c, d Immunofluorescent test of the presence of SARS-CoV-2 viral antigen in T cells. PBCs (c) or postmortem lung section (d) from COVID-19 patients were stained with T lymphocytes (CD3, green), SARS-CoV-2 (NP, red) and nuclei (DAPI, blue). In-house-made pAb against SARS-CoV-2 NP was used. White arrows indicate areas T lymphocytes that were infected by SARS-CoV-2. Pictures were taken under confocal microscopy with a bar = 50 μm (c) or 200 μm (d). Comparison of mean values (a, b) between two groups was analyzed by Student’s t test. *P < 0.05; **P < 0.01; ****P < 0.0001; NS no significance
Fig. 2
Fig. 2
SARS-CoV-2 in vitro infection of T cell lines or primary T cells. a Schedule of experiments. b Unactivated or activated Jurkat cells were infected with SARS-CoV-2 (MOI = 0.1) and samples were collected at 0, 24, 48, and 72 h post infection. Viral load in cells or cell supernatant was then quantified by qPCR detection of total viral RNA (RBD of spike gene as target) or subgenomic RNA (sgRNA, M gene as target). c Depth and coverage comparison for SARS-CoV-2 0 h or 24 h-infected activated Jurkat cells. For each sample, virus quantity was normalized with its total reads number of sequencing. Two replicates are shown for each time point. d Viral NP in the infected activated Jurkat cells and cell supernatant was analyzed by western blot at 0, 24, 48, and 72 h post infection. e Viral NP 72 h-infected cells from (d) were analyzed by flow cytometry and the number of replicates that are represented in the bar graph is three. f Viral particles in infected activated Jurkat or MT4 cells were observed by transmission electron microscope. bar = 1 μm or 500 nm, magnification: 3500-folds and 9600-folds for Jurkat cell, 5000-folds or 11500-folds for MT4 cell. g Unactivated or activated primary T cells were infected with SARS-CoV-2 (MOI = 0.01) and samples were collected at 0, 4, 8, and 12 h post infection. Viral load in cells was then quantified by qPCR. h Colon organoids were infected with SARS-CoV-2 (MOI = 0.01). Zero hour and 24 h samples were harvested and quantified by qPCR. The data were analyzed by Student’s t test and statistical significance is indicated by the asterisks (*P < 0.05; **P < 0.01; ****P < 0.0001; NS no significance)
Fig. 3
Fig. 3
SARS-CoV-2 infection of T cell is spike-ACE2/TMPRSS2-independent. a The ACE2 expression level of Scramble or ACE2-knockdown Caco2 or Jurkat cells was analyzed by qPCR or WB. b ACE2 stably knockdown Caco2 or activated Jurkat cells were infected with SARS-CoV-2 (MOI = 0.01). Viral RNA or viral NP in cells was analyzed by qPCR or WB at 24 h post infection. c The ACE2 expression level of control or ACE-knockout Caco2 or Jurkat cells were quantified by qPCR or detected by WB. d ACE2 stably knock-out Caco2 or activated Jurkat cells were infected with SARS-CoV-2 (MOI = 0.01). Viral RNA or viral NP was detected using qPCR or WB. e For ACE2 blocking, Caco2 or activated Jurkat cells were pre-incubated with anti-ACE2 Ab (3.33 ng/μl final) before infected with SARS-CoV-2. For virus blocking, ACE2-Fc protein (10 μg/μl final) or RD#4-anti-Spike Ab (160 ng/μl final) were incubated with SARS-CoV-2 at a volume of 1:1 at 37 °C for 30 min. Cells were infected at 0.01 MOI for 24 h before they were quantified for SARS-CoV-2 viral RNA or NP protein by qPCR or WB. f The TMPRSS2 expression level of Caco2, Jurkat and activated Jurkat cells was analyzed using qPCR. g Caco2 or activated Jurkat cells were pre-incubated with Camostat (2 μM or 20 μM) for 1 h and then infected with SARS-CoV-2 (MOI = 0.01). Viral RNA or NP proteins were quantified. The results were derived from three independent experiments. Statistical analyses were carried out using Student’s t test (*P < 0.05; **P < 0.01; ****P < 0.0001; NS no significance)
Fig. 4
Fig. 4
SARS-CoV-2 infection-induced apoptosis in T cells. a Detection of apoptotic T lymphocytes in human PBCs. PBCs were prepared from COVID-19 patients or from healthy donors. Apoptosis in virally infected T lymphocytes were determined using CD3, SARS-CoV-2 NP antibodies, and TUNEL assay. Detection results for two patients and one healthy donor, or the statistics of apoptotic cells or TUNEL/NP double-positive cells were shown for healthy donors (H, n = 3) or patients (P, n = 5). Comparison of mean values between two groups was analyzed by Student’s t test. *P < 0.05. b Unactivated or activated primary T cells were infected with SARS-CoV-2 (MOI = 0.01) for 8 h and cell apoptosis was analyzed with TUNEL assay. **P < 0.01. c Activated Jurkat cells were infected with SARS-CoV-2 (MOI = 0.01) for 0, 24, 48, and 72 h. The ratio of apoptotic cells in 72 h mock or SARS-CoV-2-infected cells were shown (flow chart). The ratio of apoptotic cells in different time points were also compared (plot). d Samples from C were subjected for RNA-seq analysis. The top ten upregulated GO pathways in 48 h compared to 24 h group are shown. Heatmap shows the normalized expression of genes that were enriched from PID HIF1 pathway. The data were analyzed by Student’s t test (*P < 0.05; **P < 0.01; NS no significance). e Compared to healthy donors, differentially expressed genes of severe COVID-19 patients were shown in the volcano plot, and the top ten differential expressed pathways were shown in the right panel
Fig. 5
Fig. 5
Exploration of potential receptors in T cells. a The expression of ACE2, TMPRSS2, and ITGB2 (LFA-1) in blood T cells from healthy donors and COVID-19 patients. The analysis is dependent on public single-cell NGS data. The expression of the three genes is indicated in SARS-CoV-2 viral RNA-positive T cells for patient penal. b BEAS-2B and activated Jurkat cells were incubated with different concentrations of AXL proteins (25, 50, or 100 μg/ml) at 37 °C for 30 min before infected by SARS-CoV-2 (MOI of 0.01). Intracellular viral RNA (RBD) at 24 h post infection was quantified using qPCR. c Knockdown or overexpression of AXL were performed on Jurkat cells and the expression level of AXL was detected using qPCR. Cell lines were infected by SARS-CoV-2 at an MOI of 0.01 for 24 h and viral RNA was quantified. d LFA-1 was stably overexpressed on ACE2 knockdown Caco2 (Caco2-ACE2-shRNA) or Jurkat cells, and the RNA level was quantified using qPCR. e Caco2, activated Jurkat and their respective LFA-1-overexpression cells were infected by SARS-CoV-2 at an MOI of 0.01 and harvested at 24 h post infection. Intracellular viral RNA was detected using qPCR. f, g Caco2, Caco2-ACE2-shRNA, and its LFA-1 overexpression cell line were infected by SARS-CoV-2 at an MOI = 5 for 8 h. The high content microscope was used to observe (f) and quantify (g) the viral NP-positive cells. h LFA-1 knockdown Jurkat cells were infected with SARS-CoV-2 (MOI = 0.01) for 24 h, and the expression of LFA-1 and intracellular viral RNA was analyzed using qPCR. i Activated Jurkat cells were pretreated with Lifitegrast, an inhibitor of LFA-1 at different concentrations (50, 100, or 200 nM) at 37 °C for 30 min before infection (MOI = 0.01). Intracellular viral RNA was quantified at 24 h post infection. The statistics was performed using Student’s t test (*P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001; NS no significance)

Similar articles

Cited by

References

    1. Hoffmann M, et al. SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell. 2020;181:271–280 e278. doi: 10.1016/j.cell.2020.02.052. - DOI - PMC - PubMed
    1. Li W, et al. Angiotensin-converting enzyme 2 is a functional receptor for the SARS coronavirus. Nature. 2003;426:450–454. doi: 10.1038/nature02145. - DOI - PMC - PubMed
    1. Monteil V, et al. Inhibition of SARS-CoV-2 infections in engineered human tissues using clinical-grade soluble human ACE2. Cell. 2020;181:905–913 e907. doi: 10.1016/j.cell.2020.04.004. - DOI - PMC - PubMed
    1. Wang Q, et al. Structural and functional basis of SARS-CoV-2 entry by using human ACE2. Cell. 2020;181:894–904 e899. doi: 10.1016/j.cell.2020.03.045. - DOI - PMC - PubMed
    1. Zhou, P. et al. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature579, 270–273 (2020). - PMC - PubMed

Publication types

Substances