Skip to main content
  • Letter to the Editor
  • Open access
  • Published:

SARS-CoV-2 interacts with platelets and megakaryocytes via ACE2-independent mechanism

Abstract

Evidence suggests that platelets may directly interact with SARS-CoV-2, raising the concern whether ACE2 receptor plays a role in this interaction. The current study showed that SARS-CoV-2 interacts with both platelets and megakaryocytes despite the limited efficiency. Abundance of the conventional receptor ACE2 and alternative receptors or co-factors for SARS-CoV-2 entry was characterized in platelets from COVID-19 patients and healthy persons as well as human megakaryocytes based on laboratory tests or previously reported RNA-seq data. The results suggest that SARS-CoV-2 interacts with platelets and megakaryocytes via ACE2-independent mechanism and may regulate alternative receptor expression associated with COVID-19 coagulation dysfunction.

To the editor

Associated with coagulative disorders, COVID-19 patients have increased platelet activation and aggregation, and platelet-monocyte aggregation [1,2,3], which highlights the critical role of platelets in SARS-CoV-2 infection and immunopathology [4]. Consistent with previous reports [1,2,3], our retrospective survey of plasma samples from a cohort of 62 cases (severe or fatal and moderate COVID-19 patients, Additional file 1: Table S1) showed that COVID-19 was associated with mild thrombocytopenia (platelet count < 150 × 109/L) and increased thrombosis (elevated D-dimer levels), and patients had increased platelet activation (elevated sP-selectin and sGPVI levels) and cytokine (PF4 and RANTES) release upon platelet activation (Fig. 1a). Direct interaction of SARS-CoV-2 with human platelets was suggested based on increased P-selectin translocation on platelet surface (Fig. 1b), and elevated levels of GPVI, PF4, and RANTES in platelet culture supernatants (Fig. 1c). However, the characteristics and mechanisms of the direct interaction between SARS-CoV-2 and platelets are not well elucidated, and the role of platelet receptors in the interaction remains to be clarified [4, 5].

Fig. 1
figure 1

Increased platelet activation in COVID-19 patients and that stimulated by SARS-CoV-2. a Platelet counts and D-dimer levels of patients with severe/fatal and moderate COVID-19 are shown as the medians and interquartile ranges. The normal range of platelet count (150–400 × 109/L) is shaded, and the upper limit value of D-dimer (0.5 mg/L) is indicated with dotted line. Soluble P-selectin levels (sP-selectin), soluble GPVI levels (sGPVI), PF4, and RANTES in plasma of patients with severe/fatal or moderate COVID-19, and healthy controls were measured through ELISA. b Platelet activation was investigated using platelets from healthy donors incubated with SARS-CoV-2, thrombin, or virus culture medium (Mock) for 3 h at 37 °C. P selectin surface translocation was measured using flow cytometry and results using platelets from two healthy donors are shown. c Levels of GPVI, PF4, and RANTES in the incubation supernatants were determined through ELISA. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001

SARS-CoV-2 infection in human platelets and its progenitor megakaryocyte cell line MEG-01 in vitro was subsequently characterized. SARS-CoV-2 N expression was observed in some platelets and MEG-01 cells (Fig. 2a). SARS-CoV-2 RNA was detected in both culture supernatant and MEG-01 cells after SARS-CoV-2 incubation and could be maintained with a slight increase until 48 h p.i. (Fig. 2b). This suggests that SARS-CoV-2 may infect and replicate in megakaryocytes despite insufficient efficiency. However, we failed to observe any viral particles in MEG-01 cells through electron microscopy, probably because of using insufficient dose of viruses (1 MOI) for incubation, or limited infection in MEG-01 cells, as indicated by IFA images. SARS-CoV-2 RNA copies were lower in platelets (10–102 copies/103 cells) and culture supernatant (103–104 copies/mL), which diminished after 12 h (data not shown). Therefore, we speculate that platelets may not support SARS-CoV-2 replication. This echoes recent studies which have shown that SARS-CoV-2 entry in platelets may not be common in COVID-19 patients: SARS-CoV-2 RNA was detected in platelets from a few severe (2/25, 8% [2]; 2/11, 18.2% [6]) and non-severe (9/38, 23.7% [6]) patients and was not detected in platelets from patients (0/24 [7]).

Fig. 2
figure 2

Characterization of SARS-CoV-2 interaction with human platelets and megakaryocytes. a IFA assays suggesting SARS-CoV-2 infection in platelets and megakaryocytes. Platelets from healthy donors and the megakaryocyte cell line MEG-01 were incubated with SARS-CoV-2 (1 MOI per test). SARS-CoV-2 N expression in platelets and MEG-01 cells were immunostained at 3 h p.i. and 24 h p.i., respectively. b Quantitative analysis of SARS-CoV-2 RNA copies in culture supernatants and in MEG-01 cells. c Immunofluorescence assay of ACE2 expression in MEG-01, platelets (PLT), Calu-3, Huh7, and 293 T cell lines. Bars, 10 μm. d Western blot analyses of ACE2 expression in cell lines including MEG-01, 293 T, HeLa, Huh7, and Calu-3, and platelets (PLT) from three healthy donors (D1, D2, and D3). Expression of β-actin in cells were blotted as inner control. e RNA transcripts of 14 receptors in human platelets and megakaryocytes were evaluated using bioinformatic methods using the RNA-seq data obtained from previous studies. f qRT-PCR detection of CD147, GRP78, KREMEN1, Cathepsin L, NRP1, ASGR1, and ACE2 in cell lines including Calu-3, Huh7, HeLa, 293 T, and MEG-01, and platelets from three healthy donors (D1, D2, and D3). The transcription levels were normalized to those of GAPDH in each of respective cell line or platelet samples and compared to MEG-01 or Calu-3 (shaded bars), as described in Additional file 1: Methods. g Comparison of CD147, KREMEN1, and NRP1 RNA levels in ICU and non-ICU COVID-19 patients with those in healthy persons using the RNA-seq data obtained from previous studies. h qRT-PCR detection of CD147, KREMEN1, and NRP1 transcription in MEG-01 cells after SARS-CoV-2 incubation

The evidence of direct interaction between SARS-CoV-2 and platelets or megakaryocytes raised the concern whether ACE2 plays a role in the process. The IFA and western blot assays showed a lack of ACE2 expression in both human platelets and megakaryocytes (Fig. 2c, d). The RNA abundance of 14 receptors or co-factors including ACE2 in human platelets and megakaryocytes was subsequently inspected based on RNA-seq data reported in previous studies [2, 8] (Additional file 1: Table S2 and S3). As summarized in Fig. 2e, the abundance order in platelets was: CD147 > GRP78 > KREMEN1 > ADAM17 > cathepsin L > NRP1 > ASGR1 > CD209L/L-SIGN > CD301 > CD26 > CD206, but CD209/DC-SIGN, ACE2, and TMPRSS2 were not identified. Human megakaryocytes had similar receptor profiles, coupled with the detection of CD209/DC-SIGN. We also verified receptor abundance in MEG-01 and human platelets using qRT-PCR. In MEG-01 cells, CD147, GRP78, KREMEN1, cathepsin L, NRP1, and ASGR1 were detected, while in platelets, CD147, GRP78, KREMEN1, and ASGR1 were detected. ACE2 was not detected in MEG-01 cells or platelets (Fig. 2f). These results indicate that SARS-CoV-2 may use receptors other than ACE2 to interact with platelets or megakaryocytes.

Further analysis using the RNA-seq data showed unchanged GRP78, ADAM1, cathepsin L, GRP1, and ASGR1 abundance in platelets between ICU and non-ICU COVID-19 patients and healthy persons and revealed elevated CD147 and KREMEN1 levels and reduced NRP1 levels in patients (Fig. 2g). This was also observed in MEG-01 cells with increased CD147 and KREMEN1 levels and slightly reduced NRP1 levels after SARS-CoV-2 incubation (Fig. 2h). These data suggest that SARS-CoV-2 infection may alter gene transcription in platelets and megakaryocytes, which is similar to DENV infection that markedly changes the platelet and megakaryocyte transcriptome [8].

Owing to their roles in binding to spike protein and facilitating virus entry [9,10,11], CD147, KREMEN1, and NRP1 triggering of SARS-CoV-2 entry in human platelets and megakaryocytes requires in-depth investigation. Moreover, based on the original functions of CD147 in signaling pathways via cell–cell interactions [9] and of NRP1 in cardiovascular, neuronal, and immune systems [10], SARS-CoV-2 interaction with platelets is suspected to regulate platelet-mediated immune response [12] and promote coagulation dysfunction in COVID-19 [10].

Availability of data and materials

All data generated or analyzed during this study are included in this published article (and its supplementary information files).

Abbreviations

COVID-19:

Coronavirus disease 2019

SARS-CoV-2:

Severe acute respiratory syndrome coronavirus

SARS-CoV-2 N:

SARS-CoV-2 nucleoprotein

ACE2:

Angiotensin-converting enzyme 2

sP-selectin:

Soluble P-selectin

sGPVI:

Soluble glycoprotein VI

PF4:

Platelet factor 4

RANTES:

C-C motif chemokine ligand 5, CXCL5

CD147:

Basigin

GRP78:

Glucose regulating protein 78

KREMEN1:

Kringle containing transmembrane protein 1

ADAM17:

A disintegrin and metalloproteinase 17

NRP1:

Neuroplin-1

ASGR1:

Asialoglycoprotein receptor 1

CD209L/L-SIGN:

C-type lectin domain family 4, member M, CLEC4M

CD301:

C-type lectin domain containing 10A, CLEC10A

CD26:

Dipeptidyl peptidase 4, DPP4

CD206:

Macrophage mannose receptor, MMR

CD209/DC-SIGN:

Dendritic cell (DC)-specific intracellular adhesion molecule 3 (ICAM-3)-grabbing non-integrin

TMPRSS2:

Transmembrane serine protease 2

DENV:

Dengue virus

IFAs:

Immunofluorescence assays

qRT-PCR:

Quantitative reverse transcription-polymerase chain reaction

MOI:

Multiplicity of infection

h p.i.:

Hours post-inoculation

References

  1. Hottz ED, Azevedo-Quintanilha IG, Palhinha L, Teixeira L, Barreto EA, Pão CRR, et al. Platelet activation and platelet-monocyte aggregates formation trigger tissue factor expression in patients with severe COVID-19. Blood. 2020;136:1330–41.

    Article  CAS  Google Scholar 

  2. Manne BK, Denorme F, Middleton EA, Portier I, Rowley JW, Stubben CJ, et al. Platelet gene expression and function in patients with COVID-19. Blood. 2020;136:1317–29.

    Article  CAS  Google Scholar 

  3. Middleton EA, He XY, Denorme F, Campbell RA, Ng D, Salvatore SP, et al. Neutrophil extracellulartraps (NETs) contribute to immunothrombosis in COVID-19 acute respiratory distress syndrome. Blood. 2020;136:1169–79.

    Article  CAS  Google Scholar 

  4. Koupenova M, Freedman JE. Platelets and COVID-19: Inflammation, hyperactivation and additionalquestions. Circ Res. 2020;127(11):1419–21.

    Article  CAS  Google Scholar 

  5. Campbell RA, Boilard E, Rondina MT. Is there a role for the ACE2 receptor in SARS-CoV-2 interactions with platelets? J Thromb Haemost. 2021;19(1):46–50.

    Article  CAS  Google Scholar 

  6. Zaid Y, Puhm F, Allaeys I, Naya A, Oudghiri M, Khalki L, et al. Platelets can associate with SARS-Cov-2 RNA and are hyperactivated in COVID-19. Circ Res. 2020;127(11):1404–18.

    Article  CAS  Google Scholar 

  7. Bury L, Camilloni B, Castronari R, Piselli E, Malvestiti M, Borghi M, et al. Search for SARS-CoV-2 RNA in platelets from COVID-19 patients. Platelets. 2021;32(2):284–7.

    Article  CAS  Google Scholar 

  8. Campbell RA, Schwertz H, Hottz ED, Rowley JW, Manne BK, Washington AV, et al. Human megakaryocytes possess intrinsic antiviral immunity through regulated induction of IFITM3. Blood. 2019;133(19):2013–26.

    Article  CAS  Google Scholar 

  9. Wang K, Chen W, Zhang Z, Deng Y, Lian JQ, Du P, et al. CD147-spike protein is a novel route for SARS-CoV-2 infection to host cells. Signal Transduct Target Ther. 2020;5(1):283.

    Article  CAS  Google Scholar 

  10. Mayi BS, Leibowitz JA, Woods AT, Ammon KA, Liu AE, Raja A. The role of Neuropilin-1 in COVID-19. PLOS Pathog. 2021;17(1):e1009153.

    Article  CAS  Google Scholar 

  11. Gu Y, Cao J, Zhang X, Gao H, Wang Y, Wang J, et al. Interaction network of SARS-CoV-2 with host receptome through spike protein. bioRxiv-Microbiol;2020. https://doi.org/10.1101/2020.09.09.287508

  12. Savla SR, Prabhavalkar KS, Bhatt LK. Cytokine storm associated coagulation complications in COVID-19 patients: Pathogenesis and Management. Expert Rev Anti Infect Ther. 2021. https://doi.org/10.1080/14787210.2021.1915129.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We acknowledge Mr. Ding Gao, Ms. Anna Du, Ms. Juan Min, Ms. Pei Zhang, and Ms. Bichao Xu from the Core Facility and Technical Support Facility of the Wuhan Institute of Virology for their technical assistance. We thank Mr. Jia Wu, Mr. Hao Tang, and Mr. Jun Liu from the team of BSL-3 Laboratory of Wuhan Institute of Virology for their critical support in experimental activities, and Ms. Min Zhou and Mr. Zhong Zhang for their help with cell culture.

Funding

This work was supported by the National Natural Science Foundation of China (U20A20135), the National Program on Key Research Project of China (2018YFE0200402, 2019YFC1200701, and 2020YFC0845801), and the Fundamental Research Funds for the Central Universities (2020kfyXGYJ016).

Author information

Authors and Affiliations

Authors

Contributions

SS and DF designed and conceived the project. SS, ZJY, and FYH performed the research, analyzed the data, and wrote the manuscript. LSH, WJ, and ZX collected and analyzed the clinical data. All authors have contributed to and approved the manuscript.

Corresponding authors

Correspondence to Xin Zheng or Fei Deng.

Ethics declarations

Ethics approval and consent to participate

The study protocol was approved by the Ethics Committee of Tongji Medical College, Huazhong University of Science and Technology (number: 2020/0042–02-02).

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1.

Detailed materials and methods, and supplementary tables and figure.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Shen, S., Zhang, J., Fang, Y. et al. SARS-CoV-2 interacts with platelets and megakaryocytes via ACE2-independent mechanism. J Hematol Oncol 14, 72 (2021). https://doi.org/10.1186/s13045-021-01082-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13045-021-01082-6

Keywords