Thromb Haemost 2021; 121(09): 1258-1262
DOI: 10.1055/a-1347-5555
Letter to the Editor

Platelets are Hyperactivated but Show Reduced Glycoprotein VI Reactivity in COVID-19 Patients

Valentine Léopold
1   Center of Experimental and Molecular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
2   Department of Anesthesiology and Critical Care, Paris University, Lariboisière Hospital, Paris, France
3   Inserm UMR-S 942, Cardiovascular Markers in Stress Conditions (MASCOT), University of Paris, Paris, France
,
Liza Pereverzeva
1   Center of Experimental and Molecular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
,
Alex R. Schuurman
1   Center of Experimental and Molecular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
,
Tom D. Y. Reijnders
1   Center of Experimental and Molecular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
,
Anno Saris
1   Center of Experimental and Molecular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
,
Justin de Brabander
1   Center of Experimental and Molecular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
,
Christine C. A. van Linge
1   Center of Experimental and Molecular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
,
Renée A. Douma
4   Department of Internal Medicine, Flevo Hospital, Almere, The Netherlands
,
Osoul Chouchane
1   Center of Experimental and Molecular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
,
Rienk Nieuwland
5   Laboratory of Experimental Clinical Chemistry, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
,
W. Joost Wiersinga
1   Center of Experimental and Molecular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
6   Division of Infectious Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
,
Cornelis van 't Veer
1   Center of Experimental and Molecular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
,
Tom van der Poll
1   Center of Experimental and Molecular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
6   Division of Infectious Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
› Author Affiliations
Funding V.L. is supported by Assistance Publique des Hôpitaux de Paris, Ecole de l'INSERM Liliane Bettencourt, Institut Servier; L.P. is supported by JPI-AMR/ZonMW # 547001008; T.D.Y.R. and A.S. are supported by NACTAR (# 16447) financed by the Dutch Research Council (NWO); J.B. is supported by European Commission (FAIR # 847786); O.C. is supported by: Landsteiner Foundation (LSBR # 1901).

Coronavirus disease-2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), has rapidly spread worldwide since December 2019. Disease severity is strongly associated with abnormal routine coagulation tests (particularly increased D-dimer levels) and thrombocytopenia.[1] These alterations are accompanied by a high prevalence of deep venous thrombosis, pulmonary embolism, and arterial thrombotic events at uncommon sites.[2] Platelets are at the crossroad of thrombosis and immunity and contribute to thrombus formation and disease severity during bacterial sepsis[3] [4] and viral infections.[5] [6] We here investigated platelet activation and responsiveness to relevant stimuli in hospitalized COVID-19 patients.

Patients were included in the ELDER-BIOME protocol (clinicaltrials.gov identifier NCT02928367). We prospectively enrolled suspected COVID-19 patients admitted to the ward, in two hospitals in The Netherlands (Academic Medical Center in Amsterdam and Flevo Hospital in Almere). COVID-19 infection was confirmed by polymerase chain reaction (PCR) of material obtained by nasopharyngeal swab. Controls were hospital employees (n = 8) or patients visiting the outpatient clinics without infectious disease symptoms (n = 18); the results obtained in these control subgroups did not differ and were combined. Informed consent was obtained from all participants. Citrated peripheral blood was collected within 48 hours after admission and processed within 4 hours. Platelet-rich plasma (PRP) was obtained by centrifugation (180 × g, 15 minutes, 21°C), mixed with a 1:5 ratio of acid citrate dextrose and subsequently centrifuged (200 × g, 2 minutes) to remove remaining erythrocytes and leukocytes. Platelets in buffer-diluted PRP were stimulated for 30 minutes with cross-linked collagen-related peptide (CRP-XL, CambCol-Laboratory, Cambridge, United Kingdom; 0.1 or 1 µg/mL) or thrombin receptor activator peptide-6 (TRAP-6, Bachem, Bubendorf, Switzerland; 15 µM). Platelet activation was assessed by flow cytometry using anti-CD61-AlexaFluor700 (clone VI-PL2), anti-CD62P-PerCP-Cy5.5 (clone AK4), anti-CD63-PE-Cy7 (clone H5C6), and anti-glycoprotein (GP) IIbIIIa-AlexaFluor647 (clone PAC-1) (Biolegend, San Diego, California, United States). Analysis was performed on FACSCanto II until 10,000 CD61+ events were recorded. Data were analyzed using FlowJo v10 (BD Biosciences, San Jose, California, United States). Platelet-neutrophil complexes were measured in whole blood (without ex vivo stimulation) as previously described,[7] using anti-CD66b-FITC, anti-CD11b-PE, anti-CD61-AlexaFluor700 (Biolegend), and Viability Dye-LFluor780 (eBioscience, San Diego, California, United States). Platelet numbers in PRP were established by flow cytometry, using Precision Count Beads (BD Bioscience). Soluble GPVI was measured in citrated plasma (1,500 × g, 15 minutes) by enzyme-linked immunosorbent assay (ELISA) (Invitrogen, Waltham, Massachusetts, United States). Soluble P-selectin GP ligand-1 (PSGL-1), CD40L, and D-dimer were measured using LEGENDplex Human Thrombosis Panel (Biolegend). Soluble P-selectin and platelet factor (PF) 4 were measured by ELISA (R&D Duoset, Minneapolis, Minnesota, United States). All analyses were done using GraphPad Prism v8.0 (San Diego, California, United States). Results were compared using paired Wilcoxon test or unpaired Mann–Whitney U tests. We applied linear regression to assess the correlation between neutrophil counts and soluble PSGL-1 levels. p-Values < 0.05 defined statistical significance.

We enrolled 35 patients and 26 controls between April 10 and May 15, 2020. Five patients had a negative SARS-CoV-2 PCR and were excluded from analyses. Baseline characteristics of patients and controls were comparable, except for body mass index and ethnicity ([Table 1]). Neutrophil counts were higher in COVID-19 patients compared with controls, whereas platelet counts were unaltered. Three patients (10.0%) were subsequently admitted to the intensive care unit and four (13.3%) died during hospital stay. Two patients (6.6%) were diagnosed with pulmonary embolism.

Table 1

Baseline characteristics of patients and control subjects

Controls

COVID-19

p-Value

n = 26

n = 30

Age

56.5 [45.5–71.7]

61.5 [50.2–68.5]

0.53

Gender (male)

14 (53.8)

13 (43.3)

0.61

Ethnicity

 African

2 (7.2)

10 (33.3)

< 0.01

 Caucasian

23 (88.5)

10 (33.3)

 Surinam

1 (3.8)

7 (23.3)

 Other

0 (0.0)

3 (10.0)

Body mass index

25.9 [22.3–28.5]

29.5 [27.0–32.7]

< 0.01

Comorbidities

 Respiratory

4 (15.4)

3 (10.0)

0.45

  Asthma

1 (3.8)

1 (3.3)

> 0.99

  COPD

2 (7.7)

1 (3.3)

0.90

 Cardiovascular

9 (34.6)

15 (50.0)

0.37

  Hypertension

9 (34.6)

13 (43.3)

0.70

  Ischemic cardiopathy

3 (11.5)

2 (6.7)

0.87

  Peripheral ischemic disease

1 (3.8)

1 (3.3)

> 0.99

  Congestive heart failure

3 (11.5)

1 (3.3)

0.50

  Stroke

2 (7.7)

2 (6.7)

> 0.99

 Diabetes

5 (19.2)

8 (26.7)

0.73

 Malignancy

6 (23.1)

1 (3.3)

0.07

 Chronic renal disease

0 (0.0)

2 (6.7)

0.54

Chronic medication

 Platelet antagonist

4 (15.4)[a]

5 (16.7)[b]

> 0.99

 NSAID

2 (7.7)[c]

0 (0.0)

0.41

Clinical characteristics

 Oxygen saturation

95.5 [90.5–97.8][d]

 Temperature

37.5 [36.6–38.2]

 Systolic blood pressure

128 [119–141]

 Diastolic blood pressure

80 [73–89]

 qSOFA score 0

10 (33.3)

  1

19 (63.3)

  2

1 (3.3)

Laboratory at admission

Hemoglobin (g/L)

138.6 [125.7–151.5]

128.9 [117.6–133.7]

0.19

Platelets (109/L)

236 [180–272]

240 [194–344]

0.48

Leukocytes (109/L)

5.85 [5.08–7.17]

7.00 [5.82–9.07]

0.13

 Neutrophils

3.71 [3.07–4.64]

5.00 [4.20–6.09]

0.01

 Lymphocytes

1.37 [1.12–1.91]

1.05 [0.72–1.40]

0.04

 Monocytes

0.49 [0.42–0.68]

0.38 [0.27–0.60]

0.07

D-dimer (mg/L)

0.031 [0.019 - 0.046]

0.045 [0.024–0.11]

0.10

 Outcome

 Thrombotic event

2 (6.6)[e]

 ICU admission

3 (10.0)

 In-hospital death

4 (13.3)

Abbreviations: COPD, chronic obstructive pulmonary disease; ICU, intensive care unit; IQR, interquartile range; NSAID, nonsteroidal anti-inflammatory drug; qSOFA, quick Sequential Organ Failure Assessment.


Note: Data are shown as median (%) or median [IQR].


a Aspirin (n = 2), clopidogrel (n = 1), aspirin + clopidogrel (n = 1).


b Aspirin (n = 3), clopidogrel (n = 1), aspirin + clopidogrel (n = 1).


c Diclofenac (n = 1), naproxen (n = 1).


d With oxygen therapy (median 2 L/min).


e Pulmonary embolism (n = 2).


To assess circulating platelet activation, we measured the expression of P-selectin, CD63 (reflecting α- and dense granules secretion, respectively), and the fibrinogen-binding conformation of GPIIbIIIa.[8] Platelets from COVID-19 patients showed increased expression of P-selectin and active GPIIbIIIa, while CD63 expression was comparable to that of control platelets ([Fig. 1A]). The plasma levels of α-granule proteins PF4 and soluble CD40L were higher in COVID-19 patients, further indicating activated platelets in vivo ([Fig. 1B]). While earlier studies also reported enhanced α granule release,[9] [10] our study adds to this that dense granules likely are not affected in COVID-19 patients not requiring intensive care.

Zoom Image
Fig. 1 Platelet activation and reactivity in COVID-19 patients and controls. (A) Platelet expression of P-selectin, CD63, and active glycoprotein (GP) IIbIIIa measured at baseline by flow cytometry. (B) Plasma levels soluble CD40L, platelet factor (PF)4, soluble P-selectin, and soluble P-selectin GP ligand-1 (PSGL-1). (C) Platelet-neutrophil complexes (defined as CD61+ neutrophils) measured in whole blood by flow cytometry. (D) CD11b median fluorescence intensity (MFI) analyzed for all neutrophils (CD66b + ), neutrophils complexed with platelets (CD66b+ CD61 + ), and non-complexed neutrophils (CD66b+ CD61). (E) Correlation plot between soluble PSGL-1 and neutrophil counts. (F) Platelets were stimulated ex vivo with cross-linked collagen-related peptide (CRP-XL) 0.1 µg/mL, CRP-XL 1 µg/mL, or thrombin receptor activator peptide (TRAP) 15 µM for 30 minutes; activation markers were studied by flow cytometry. (G) Plasma levels of soluble GPVI. Results are shown as dot plot with box and whiskers. *p < 0.05; **p < 0.01; ***p < 0.001; ****p < 0.0001.

Platelet-neutrophil complexes are linked to platelet hyperactivation in vivo.[3] COVID-19 patients had more platelet-neutrophil complexes compared with controls ([Fig. 1C]). Moreover, neutrophils from COVID-19 patients had higher CD11b expression, suggesting a higher activation status ([Fig. 1D]). Interestingly, neutrophils that were in complex with platelets had the highest CD11b expression, implying that platelet activation and interaction with neutrophils is associated with increased neutrophil activation. Likely, the enhanced platelet P-selectin expression in COVID-19 patients contributed to the interaction with neutrophils.[8] Plasma levels of soluble P-selectin and PSGL-1 (the neutrophil receptor mediating platelet-neutrophil complex formation) were not different between patients and controls ([Fig. 1B]). Soluble PSGL-1 was positively correlated with neutrophil counts in COVID-19 patients (r = 0.53, p = 0.0029) but not in controls (r = 0.35, p = 0.24) ([Fig. 1E]), suggesting that PSGL-1 shedding is associated with neutrophil activation and demargination in COVID-19 patients. Recent studies corroborate our findings of enhanced platelet P-selectin expression with normal plasma soluble P-selectin levels in COVID-19 patients not requiring intensive care.[11] [12] Together, these results suggest that circulating platelets are hyperactivated in COVID-19 patients, which is associated with enhanced neutrophil activation.

To assess platelet reactivity in COVID-19 patients, we stimulated platelets ex vivo with a GPVI agonist (CRP-XL) or protease-activated receptor-1 agonist (TRAP), mimicking collagen and thrombin stimulation, respectively. Remarkably, platelets from COVID-19 patients showed a decreased sensitivity to low dose CRP-XL as measured by P-selectin, CD63, and active GPIIbIIIa surface expression; this hyporeactivity was also observed at a higher dose of CRP-XL for CD63 and active GPIIbIIIa expression ([Fig. 1F]). In sharp contrast, platelet reactivity to TRAP was increased in COVID-19 patients when compared with controls with regard to P-selectin expression, in agreement with previous study,[12] while CD63 and active GPIIbIIIa were not different. Our result of platelet hyporeactivity to GPVI stimulation in COVID-19 patients, confirmed with all measured membrane markers of α- and dense-granule release and GPIIbIIIa activation, is corroborated by a recent study that reported unaltered P-Selectin expression but decreased GPIIbIIIa activation in response to a high dose of CRP, another specific GPVI agonist.[12] Of note, earlier studies reporting increased platelet aggregation in COVID-19 patients used collagen to stimulate platelets,[12] [13] which unlike CRP-XL used here is not selective for GPVI.

GPVI is an essential platelet receptor that binds collagen and fibrin. We hypothesized that platelets desensitization of GPVI to CRP-XL in COVID-19 could be due to GPVI shedding, following in vivo transient interaction with its ligands such as collagen, fibrin, D-dimer, or fibrinogen (as previously reviewed by Nurden[14]). GPVI shedding, estimated by measuring soluble GPVI in plasma, was higher in COVID-19 patients than in controls ([Fig. 1G]). Besides GPVI shedding, an additional mechanism of GPVI hyposensitivity could be the disruption of GPVI signaling, for example, due to FcγRIIA irreversible proteolysis,[15] in the context of a viral infection and circulating immune complexes.[16]

In conclusion, we report hyperactivated platelets in COVID-19 patients associated with increased activation of neutrophils and complex formation. Platelets from COVID-19 patients were desensitized to CRP-XL stimulation which was accompanied by increased shedding of the CRP-XL receptor GPVI. Although previous studies have implicated platelet GPVI in thromboembolism,[14] our data argue against a role of platelet GPVI in COVID-19 hypercoagulability.



Publication History

Received: 28 July 2020

Accepted: 06 January 2021

Accepted Manuscript online:
07 January 2021

Article published online:
14 April 2021

© 2021. Thieme. All rights reserved.

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

 
  • References

  • 1 Connors JM, Levy JH. COVID-19 and its implications for thrombosis and anticoagulation. Blood 2020; 135 (23) 2033-2040
  • 2 Rosen RJ. Thrombotic complications in critically ill patients with COVID 19. Thromb Res 2020; 191: 56
  • 3 Iba T, Levy JH. Inflammation and thrombosis: roles of neutrophils, platelets and endothelial cells and their interactions in thrombus formation during sepsis. J Thromb Haemost 2018; 16 (02) 231-241
  • 4 van der Poll T, Parker RI. Platelet activation and endothelial cell dysfunction. Crit Care Clin 2020; 36 (02) 233-253
  • 5 Hottz ED, Bozza FA, Bozza PT. Platelets in immune response to virus and immunopathology of viral infections. Front Med 2018; 5 (APR): 121
  • 6 Koupenova M, Corkrey HA, Vitseva O. et al. The role of platelets in mediating a response to human influenza infection. Nat Commun 2019; 10 (01) 1780
  • 7 Claushuis TAM, de Vos AF, Nieswandt B. et al. Platelet glycoprotein VI aids in local immunity during pneumonia-derived sepsis caused by gram-negative bacteria. Blood 2018; 131 (08) 864-876
  • 8 van der Meijden PEJ, Heemskerk JWM. Platelet biology and functions: new concepts and clinical perspectives. Nat Rev Cardiol 2019; 16 (03) 166-179
  • 9 Zaid Y, Puhm F, Allaeys I. et al. Platelets can associate with SARS-CoV-2 RNA and are hyperactivated in COVID-19. Circ Res 2020; 127 (11) 1404-1418
  • 10 Hottz ED, Azevedo-Quintanilha IG, Palhinha L. et al. Platelet activation and platelet-monocyte aggregate formation trigger tissue factor expression in patients with severe COVID-19. Blood 2020; 136 (11) 1330-1341
  • 11 Goshua G, Pine AB, Meizlish ML. et al. Articles endotheliopathy in COVID-19-associated coagulopathy: evidence from a single-centre, cross-sectional study. Lancet Haematol 2020; 3026 (20) 1-8
  • 12 Manne BK, Denorme F, Middleton EA. et al. Platelet gene expression and function in COVID-19 patients. Blood 2020; 136 (11) 1317-1329
  • 13 Zhang S, Liu Y, Wang X. et al. SARS-CoV-2 binds platelet ACE2 to enhance thrombosis in COVID-19. J Hematol Oncol 2020; 13 (01) 1-22
  • 14 Nurden AT. Clinical significance of altered collagen-receptor functioning in platelets with emphasis on glycoprotein VI. Blood Rev 2019; 38 DOI: 10.1016/j.blre.2019.100592.
  • 15 Gardiner EE, Karunakaran D, Arthur JF. et al. Dual ITAM-mediated proteolytic pathways for irreversible inactivation of platelet receptors: De-ITAM-izing FcγRIIa. Blood 2008; 111 (01) 165-174
  • 16 Nazi I, Smith JW, Arnold DM. et al. The association between platelet activation and FcγRIIa proteolysis. J Thromb Haemost 2011; 9 (04) 885-887